1
|
Cheng W, Zhang BF, Chen N, Liu Q, Ma X, Fu X, Xu M. Molecular Mechanism of Yangshen Maidong Decoction in the Treatment of Chronic Heart Failure based on Network Pharmacology, Molecular Docking, and Molecular Dynamics Simulations. Cell Biochem Biophys 2024; 82:1433-1451. [PMID: 38753250 DOI: 10.1007/s12013-024-01297-7] [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] [Accepted: 04/25/2024] [Indexed: 08/25/2024]
Abstract
Chronic heart failure (CHF) is a complex multifactorial clinical syndrome leading to abnormal cardiac structure and function. The severe form of this ailment is characterized by high disability, high mortality, and morbidity. Worldwide, 2-17% of patients die at first admission, of which 17-45% die within 1 year of admission and >50% within 5 years. Yangshen Maidong Decoction (YSMDD) is frequently used to treat the deficiency and pain of the heart. The specific mechanism of action of YSMDD in treating CHF, however, remains unclear. Therefore, a network pharmacology-based strategy combined with molecular docking and molecular dynamics simulations was employed to investigate the potential molecular mechanism of YSMDD against CHF. The effective components and their targets of YSMDD and related targets of CHF were predicted and screened based on the public database. The network pharmacology was used to explore the potential targets and possible pathways that involved in YSMDD treated CHF. Molecular docking and molecular dynamics simulations were performed to elucidate the binding affinity between the YSMDD and CHF targets. Screen results, 10 main active ingredients, and 6 key targets were acquired through network pharmacology analysis. Pathway enrichment analysis showed that intersectional targets associated pathways were enriched in the Prostate cancer pathway, Hepatitis B pathway, and C-type lectin receptor signaling pathways. Molecular docking and molecular dynamics simulations analysis suggested 5 critical active ingredients have high binding affinity to the 5 key targets. This research shows the multiple active components and molecular mechanisms of YSMDD in the treatment of CHF and offers resources and suggestions for future studies.
Collapse
Affiliation(s)
- Wei Cheng
- Department of Pharmacy, Guang'anmen Hospital Jinan Hospital (Jinan Municipal Hospital of Traditional Chinese Medicine), Jinan, 250012, China
| | - Bo-Feng Zhang
- Department of Pharmacy, Guang'anmen Hospital Jinan Hospital (Jinan Municipal Hospital of Traditional Chinese Medicine), Jinan, 250012, China
| | - Na Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Qun Liu
- Department of Pharmacy, Guang'anmen Hospital Jinan Hospital (Jinan Municipal Hospital of Traditional Chinese Medicine), Jinan, 250012, China
| | - Xin Ma
- Department of Pharmacy, Guang'anmen Hospital Jinan Hospital (Jinan Municipal Hospital of Traditional Chinese Medicine), Jinan, 250012, China
| | - Xiao Fu
- Department of Pharmacy, Guang'anmen Hospital Jinan Hospital (Jinan Municipal Hospital of Traditional Chinese Medicine), Jinan, 250012, China
| | - Min Xu
- Department of Pharmacy, Guang'anmen Hospital Jinan Hospital (Jinan Municipal Hospital of Traditional Chinese Medicine), Jinan, 250012, China.
| |
Collapse
|
2
|
Aragoneses-Cazorla G, Alvarez-Fernandez Garcia R, Martinez-Lopez A, Gomez Gomez M, Vallet-Regí M, Castillo-Lluva S, González B, Luque-Garcia JL. Mechanistic insights into the antitumoral potential and in vivo antiproliferative efficacy of a silver-based core@shell nanosystem. Int J Pharm 2024; 655:124023. [PMID: 38513815 DOI: 10.1016/j.ijpharm.2024.124023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 03/23/2024]
Abstract
This study delves into the biomolecular mechanisms underlying the antitumoral efficacy of a hybrid nanosystem, comprised of a silver core@shell (Ag@MSNs) functionalized with transferrin (Tf). Employing a SILAC proteomics strategy, we identified over 150 de-regulated proteins following exposure to the nanosystem. These proteins play pivotal roles in diverse cellular processes, including mitochondrial fission, calcium homeostasis, endoplasmic reticulum (ER) stress, oxidative stress response, migration, invasion, protein synthesis, RNA maturation, chemoresistance, and cellular proliferation. Rigorous validation of key findings substantiates that the nanosystem elicits its antitumoral effects by activating mitochondrial fission, leading to disruptions in calcium homeostasis, as corroborated by RT-qPCR and flow cytometry analyses. Additionally, induction of ER stress was validated through western blotting of ER stress markers. The cytotoxic action of the nanosystem was further affirmed through the generation of cytosolic and mitochondrial reactive oxygen species (ROS). Finally, in vivo experiments using a chicken embryo model not only confirmed the antitumoral capacity of the nanosystem, but also demonstrated its efficacy in reducing cellular proliferation. These comprehensive findings endorse the potential of the designed Ag@MSNs-Tf nanosystem as a groundbreaking chemotherapeutic agent, shedding light on its multifaceted mechanisms and in vivo applicability.
Collapse
Affiliation(s)
- Guillermo Aragoneses-Cazorla
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain
| | | | - Angelica Martinez-Lopez
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain
| | - Milagros Gomez Gomez
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain
| | - Maria Vallet-Regí
- Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Sonia Castillo-Lluva
- Department of Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain
| | - Blanca González
- Department of Chemistry in Pharmaceutical Sciences, Faculty of Pharmacy, Complutense University of Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (i+12), 28040 Madrid, Spain; Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| | - Jose L Luque-Garcia
- Department of Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040 Madrid, Spain.
| |
Collapse
|
3
|
Crespo-Garcia S, Fournier F, Diaz-Marin R, Klier S, Ragusa D, Masaki L, Cagnone G, Blot G, Hafiane I, Dejda A, Rizk R, Juneau R, Buscarlet M, Chorfi S, Patel P, Beltran PJ, Joyal JS, Rezende FA, Hata M, Nguyen A, Sullivan L, Damiano J, Wilson AM, Mallette FA, David NE, Ghosh A, Tsuruda PR, Dananberg J, Sapieha P. Therapeutic targeting of cellular senescence in diabetic macular edema: preclinical and phase 1 trial results. Nat Med 2024; 30:443-454. [PMID: 38321220 DOI: 10.1038/s41591-024-02802-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 01/03/2024] [Indexed: 02/08/2024]
Abstract
Compromised vascular endothelial barrier function is a salient feature of diabetic complications such as sight-threatening diabetic macular edema (DME). Current standards of care for DME manage aspects of the disease, but require frequent intravitreal administration and are poorly effective in large subsets of patients. Here we provide evidence that an elevated burden of senescent cells in the retina triggers cardinal features of DME pathology and conduct an initial test of senolytic therapy in patients with DME. In cell culture models, sustained hyperglycemia provoked cellular senescence in subsets of vascular endothelial cells displaying perturbed transendothelial junctions associated with poor barrier function and leading to micro-inflammation. Pharmacological elimination of senescent cells in a mouse model of DME reduces diabetes-induced retinal vascular leakage and preserves retinal function. We then conducted a phase 1 single ascending dose safety study of UBX1325 (foselutoclax), a senolytic small-molecule inhibitor of BCL-xL, in patients with advanced DME for whom anti-vascular endothelial growth factor therapy was no longer considered beneficial. The primary objective of assessment of safety and tolerability of UBX1325 was achieved. Collectively, our data suggest that therapeutic targeting of senescent cells in the diabetic retina with a BCL-xL inhibitor may provide a long-lasting, disease-modifying intervention for DME. This hypothesis will need to be verified in larger clinical trials. ClinicalTrials.gov identifier: NCT04537884 .
Collapse
Affiliation(s)
- Sergio Crespo-Garcia
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
- École d'optométrie, University of Montreal, Montreal, Quebec, Canada
| | - Frédérik Fournier
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Roberto Diaz-Marin
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO-HMR) Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Sharon Klier
- UNITY Biotechnology, South San Francisco, CA, USA
| | - Derek Ragusa
- UNITY Biotechnology, South San Francisco, CA, USA
| | | | - Gael Cagnone
- Departments of Pediatrics Ophthalmology, and Pharmacology, Centre Hospitalier Universitaire Sainte Justine Research Center, Montreal, Quebec, Canada
| | - Guillaume Blot
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Ikhlas Hafiane
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO-HMR) Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Agnieszka Dejda
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO-HMR) Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Rana Rizk
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Rachel Juneau
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO-HMR) Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Manuel Buscarlet
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Sarah Chorfi
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO-HMR) Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | | | | | - Jean-Sebastien Joyal
- Departments of Pediatrics Ophthalmology, and Pharmacology, Centre Hospitalier Universitaire Sainte Justine Research Center, Montreal, Quebec, Canada
| | - Flavio A Rezende
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO-HMR) Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Masayuki Hata
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Alex Nguyen
- UNITY Biotechnology, South San Francisco, CA, USA
| | | | | | - Ariel M Wilson
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO-HMR) Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | - Frédérick A Mallette
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
- Department of Medicine, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada
| | | | | | | | | | - Przemyslaw Sapieha
- Department of Biochemistry, Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada.
- Department of Ophthalmology, Centre Universitaire d'Ophtalmologie (CUO-HMR) Maisonneuve-Rosemont Hospital Research Centre, University of Montreal, Montreal, Quebec, Canada.
- UNITY Biotechnology, South San Francisco, CA, USA.
| |
Collapse
|
4
|
Xie B, Olalekan S, Back R, Ashitey NA, Eckart H, Basu A. Exploring the tumor micro-environment in primary and metastatic tumors of different ovarian cancer histotypes. Front Cell Dev Biol 2024; 11:1297219. [PMID: 38328306 PMCID: PMC10847324 DOI: 10.3389/fcell.2023.1297219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/06/2023] [Indexed: 02/09/2024] Open
Abstract
Ovarian cancer is a highly heterogeneous disease consisting of at least five different histological subtypes with varying clinical features, cells of origin, molecular composition, risk factors, and treatments. While most single-cell studies have focused on High grade serous ovarian cancer, a comprehensive landscape of the constituent cell types and their interactions within the tumor microenvironment are yet to be established in the different ovarian cancer histotypes. Further characterization of tumor progression, metastasis, and various histotypes are also needed to connect molecular signatures to pathological grading for personalized diagnosis and tailored treatment. In this study, we leveraged high-resolution single-cell RNA sequencing technology to elucidate the cellular compositions on 21 solid tumor samples collected from 12 patients with six ovarian cancer histotypes and both primary (ovaries) and metastatic (omentum, rectum) sites. The diverse collection allowed us to deconstruct the histotypes and tumor site-specific expression patterns of cells in the tumor, and identify key marker genes and ligand-receptor pairs that are active in the ovarian tumor microenvironment. Our findings can be used in improving precision disease stratification and optimizing treatment options.
Collapse
Affiliation(s)
- Bingqing Xie
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, United States
| | | | | | | | | | - Anindita Basu
- Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, United States
| |
Collapse
|
5
|
Nimer RM, Alfaqih MA, Shehabat ER, Mujammami M, Abdel Rahman AM. Label-free quantitative proteomics analysis for type 2 diabetes mellitus early diagnostic marker discovery using data-independent acquisition mass spectrometry (DIA-MS). Sci Rep 2023; 13:20880. [PMID: 38012280 PMCID: PMC10682489 DOI: 10.1038/s41598-023-48185-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 11/23/2023] [Indexed: 11/29/2023] Open
Abstract
Type-2 diabetes mellitus (T2DM) therapy requires early diagnosis and complication avoidance. Unfortunately, current diagnostic markers do not meet these needs. Data-independent acquisition mass spectrometry (DIA-MS) offers a solution for clinical diagnosis, providing reliable and precise sample quantification. This study utilized DIA-MS to investigate proteomic differential expression in the serum of recently diagnosed T2DM patients. The study conducted a comparative protein expression analysis between healthy and recently diagnosed T2DM groups (discovery cohort). A candidate protein was then validated using enzyme-linked immune assay (ELISA) on serum samples collected from T2DM patients (n = 87) and healthy control (n = 60) (validation cohort). A total of 1074 proteins were identified, and 90 were significantly dysregulated between the two groups, including 32 newly associated with T2DM. Among these proteins, the expression of S100 calcium-binding protein A6 (S100A6) was validated by ELISA. It showed a significant increase in T2DM samples compared to the control group. It was evaluated as a biomarker using the receiver operating characteristic (ROC) curve, consistent with the DIA-MS results. Novel proteins are reported to be involved in the development and progression of T2DM. Further studies are required to investigate the differential expression of candidate marker proteins in a larger population of T2DM patients.
Collapse
Affiliation(s)
- Refat M Nimer
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan.
| | - Mahmoud A Alfaqih
- Department of Physiology and Biochemistry, Faculty of Medicine, Jordan University of Science and Technology, Irbid, 22110, Jordan
- Department of Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, 15503, Bahrain
| | - Eman R Shehabat
- Department of Medical Laboratory Sciences, Jordan University of Science and Technology, Irbid, 22110, Jordan
| | - Muhammad Mujammami
- Department of Medicine, College of Medicine, King Saud University, 12372, Riyadh, Saudi Arabia
- University Diabetes Center, King Saud University Medical City, King Saud University, 12372, Riyadh, Saudi Arabia
| | - Anas M Abdel Rahman
- Department of Chemistry, Memorial University of Newfoundland, St. John's, NL, A1B 3X7, Canada
| |
Collapse
|
6
|
Yang F, Ma J, Zhu D, Wang Z, Li Y, He X, Zhang G, Kang X. The Role of S100A6 in Human Diseases: Molecular Mechanisms and Therapeutic Potential. Biomolecules 2023; 13:1139. [PMID: 37509175 PMCID: PMC10377078 DOI: 10.3390/biom13071139] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Revised: 07/11/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
S100A6, also known as calcyclin, is a low-molecular-weight Ca2+-binding protein from the S100 family that contains two EF-hands. S100A6 is expressed in a variety of mammalian cells and tissues. It is also expressed in lung, colorectal, pancreatic, and liver cancers, as well as other cancers such as melanoma. S100A6 has many molecular functions related to cell proliferation, the cell cycle, cell differentiation, and the cytoskeleton. It is not only involved in tumor invasion, proliferation, and migration, but also the pathogenesis of other non-neoplastic diseases. In this review, we focus on the molecular mechanisms and potential therapeutic targets of S100A6 in tumors, nervous system diseases, leukemia, endometriosis, cardiovascular disease, osteoarthritis, and other related diseases.
Collapse
Affiliation(s)
- Fengguang Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (X.H.); (G.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Jinglin Ma
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (X.H.); (G.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
- School of Petrochemical Engineering, Lanzhou University of Technology, Lanzhou 730050, China
| | - Daxue Zhu
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (X.H.); (G.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Zhaoheng Wang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (X.H.); (G.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Yanhu Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (X.H.); (G.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Xuegang He
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (X.H.); (G.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (X.H.); (G.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China; (F.Y.); (X.H.); (G.Z.)
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730030, China
- Orthopaedics Key Laboratory of Gansu Province, Lanzhou University Second Hospital, Lanzhou 730030, China
| |
Collapse
|
7
|
Jiang Y, Lei G, Lin T, Zhou N, Wu J, Wang Z, Fan Y, Sheng H, Mao R. 1,6-Hexanediol regulates angiogenesis via suppression of cyclin A1-mediated endothelial function. BMC Biol 2023; 21:75. [PMID: 37024934 PMCID: PMC10080975 DOI: 10.1186/s12915-023-01580-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 03/28/2023] [Indexed: 04/08/2023] Open
Abstract
BACKGROUND Angiogenesis plays important roles in physiological and pathologic conditions, but the mechanisms underlying this complex process often remain to be elucidated. In recent years, liquid-liquid phase separation (LLPS) has emerged as a new concept to explain many cellular functions and diseases. However, whether LLPS is involved in angiogenesis has not been studied until now. Here, we investigated the potential role of LLPS in angiogenesis and endothelial function. RESULTS We found 1,6-hexanediol (1,6-HD), an inhibitor of LLPS, but not 2,5-hexanediol (2,5-HD) dramatically decreases neovascularization of Matrigel plug and angiogenesis response of murine corneal in vivo. Moreover, 1,6-HD but not 2,5-HD inhibits microvessel outgrowth of aortic ring and endothelial network formation. The endothelial function of migration, proliferation, and cell growth is suppressed by 1,6-HD. Global transcriptional analysis by RNA-sequencing reveals that 1,6-HD specifically blocks cell cycle and downregulates cell cycle-related genes including cyclin A1. Further experimental data show that 1,6-HD treatment greatly reduces the expression of cyclin A1 but with minimal effect on cyclin D1, cyclin E1, CDK2, and CDK4. The inhibitory effect of 1,6-HD on cyclin A1 is mainly through transcriptional regulation because proteasome inhibitors fail to rescue its expression. Furthermore, overexpression of cyclin A1 in HUVECs largely rescues the dysregulated tube formation upon 1,6-HD treatment. CONCLUSIONS Our data reveal a critical role of LLPS inhibitor 1,6-HD in angiogenesis and endothelial function, which specifically affects endothelial G1/S transition through transcriptional suppression of CCNA1, implying LLPS as a possible novel player to modulate angiogenesis, and thus, it might represent an interesting therapeutic target to be investigated in clinic angiogenesis-related diseases in future.
Collapse
Affiliation(s)
- Yongying Jiang
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, People's Republic of China
| | - Gongyun Lei
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, People's Republic of China
| | - Ting Lin
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, People's Republic of China
| | - Nan Zhou
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, People's Republic of China
| | - Jintao Wu
- Heart Center of Henan Provincial People's Hospital, Central China Fuwai Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, Henan, 450003, People's Republic of China
| | - Zhou Wang
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Yihui Fan
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, Jiangsu, 226001, People's Republic of China
| | - Hongzhuan Sheng
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, People's Republic of China.
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, 19 Qixiu Road, Nantong, Jiangsu, 226001, People's Republic of China.
| |
Collapse
|
8
|
Gong J, Zhao S, Heng N, Wang Y, Hu Z, Wang H, Zhu H. The Dynamic Transcription Profiles of Proliferating Bovine Ovarian Granulosa When Exposed to Increased Levels of β-Hydroxybutyric Acid. Front Vet Sci 2022; 9:915956. [PMID: 35990259 PMCID: PMC9389329 DOI: 10.3389/fvets.2022.915956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/09/2022] [Indexed: 11/13/2022] Open
Abstract
Ketosis is common in high-yield dairy cows. It is a condition that is characterized by the accumulation of serum β-hydroxybutyric acid (BHBA). Both subclinical ketosis and clinical ketosis can compromise the reproductive performance and cause long-lasting negative effects on reproductive efficiency by affecting the proliferation of follicular and granulosa cells. However, the regulatory mechanisms involved in the development of follicular cells and granulosa cells in cows experiencing subclinical ketosis and clinical ketosis remain largely unknown. To investigate the effect of a ketosis-triggered increase in BHBA on bovine follicular granulosa cell development, we detected a significant reduction in the proliferation of granulosa cells (P < 0.05) in the BHBA-1.2 mM and BHBA-2.4 mM groups and a significant increase in the number of granulosa cells in the G1 phase of the cell cycle (P < 0.05). RNA-seq and trend analysis were used to identify differentially expressed genes by comparing three clusters: low-concentration response to 1.2 mM BHBA, high-concentration response to 2.4 mM BHBA, and the similar trend (up or down) response following BHBA concentration increased. GO and KEGG enrichment analyses were performed separately for each cluster. Analysis showed that two novel down-regulated genes (G0S2 and S100A6), which are associated with cell proliferation and cycle progression, were enriched in the low-concentration response to 1.2 mM BHBA. Another differentially expressed gene (PARP), which plays a role in the apoptotic pathway, was enriched in the high-concentration response to 2.4 mM BHBA. We also found that CYP27B1 and CYP17A1, which are associated with Ca2+ homeostasis and estrogen synthesis, were enriched in a similar trend response. In conclusion, we describe the dynamic transcription profiles of granulosa cells under different levels of β-hydroxybutyric stress and report key regulators that may underlie the detrimental effects on the development of follicles and granulosa cells, thus representing potential therapeutic targets to improve fertility in dairy cows with subclinical ketosis or clinical ketosis.
Collapse
|
9
|
Leśniak W, Filipek A. S100A6 as a Constituent and Potential Marker of Adult and Cancer Stem Cells. Stem Cell Rev Rep 2022; 18:2699-2708. [PMID: 35796891 DOI: 10.1007/s12015-022-10403-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/24/2022] [Indexed: 10/17/2022]
Abstract
Adult or tissue stem cells are present in various tissues of the organism where they reside in a specific environment called the niche. Owing to their ability to generate a progeny that can proliferate and differentiate into specialized cell types, adult stem cells constitute a source of new cells necessary for tissue maintenance and/or regeneration. Under normal conditions they divide with a frequency matching the pace of tissue renewal but, following tissue damage, they can migrate to the site of injury and expand/divide intensively to facilitate tissue repair. For this reason much hope is being placed on the use of adult stem cells in regenerative therapies, including tissue engineering. Identification and characterization of tissue stem cells has been a laborious process due to their scarcity and lack of universal markers. Nonetheless, recent studies, employing various types of transcriptomic analyses, revealed some common trends in gene expression pattern among stem cells derived from different tissues, suggesting the importance of certain genes/proteins for the unique properties of these cells. S100A6, a small calcium binding protein, has been recognized as an important factor influencing cell proliferation and differentiation. Accumulating results show that S100A6 is a constituent of adult stem cells and, in some cases, may even be considered as their marker. Thus, in this review we summarize literature data concerning the presence of S100A6 in adult and cancer stem cells and speculate on its potential role and usefulness as a marker of these cells.
Collapse
Affiliation(s)
- Wiesława Leśniak
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02- 093, Warsaw, Poland.
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02- 093, Warsaw, Poland
| |
Collapse
|
10
|
The Functional Interaction of EGFR with AT1R or TP in Primary Vascular Smooth Muscle Cells Triggers a Synergistic Regulation of Gene Expression. Cells 2022; 11:cells11121936. [PMID: 35741065 PMCID: PMC9222111 DOI: 10.3390/cells11121936] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/11/2022] [Accepted: 06/15/2022] [Indexed: 02/01/2023] Open
Abstract
In vivo, cells are simultaneously exposed to multiple stimuli whose effects are difficult to distinguish. Therefore, they are often investigated in experimental cell culture conditions where stimuli are applied separately. However, it cannot be presumed that their individual effects simply add up. As a proof-of-principle to address the relevance of transcriptional signaling synergy, we investigated the interplay of the Epidermal Growth Factor Receptor (EGFR) with the Angiotensin-II (AT1R) or the Thromboxane-A2 (TP) receptors in murine primary aortic vascular smooth muscle cells. Transcriptome analysis revealed that EGFR-AT1R or EGFR-TP simultaneous activations led to different patterns of regulated genes compared to individual receptor activations (qualitative synergy). Combined EGFR-TP activation also caused a variation of amplitude regulation for a defined set of genes (quantitative synergy), including vascular injury-relevant ones (Klf15 and Spp1). Moreover, Gene Ontology enrichment suggested that EGFR and TP-induced gene expression changes altered processes critical for vascular integrity, such as cell cycle and senescence. These bioinformatics predictions regarding the functional relevance of signaling synergy were experimentally confirmed. Therefore, by showing that the activation of more than one receptor can trigger a synergistic regulation of gene expression, our results epitomize the necessity to perform comprehensive network investigations, as the study of individual receptors may not be sufficient to understand their physiological or pathological impact.
Collapse
|
11
|
Brady K, Talbot CC, Long JA, Welch G, French N, Nicholson D, Bakst MR. Transcriptome analysis of blastoderms exposed to prolonged egg storage and short periods of incubation during egg storage. BMC Genomics 2022; 23:262. [PMID: 35379173 PMCID: PMC8981843 DOI: 10.1186/s12864-022-08463-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 03/08/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Cool temperature egg storage prior to incubation is a common practice in the broiler industry; however, prolonged egg storage causes increased embryonic mortality and decreased hatchability and growth in surviving chicks. Exposing eggs to short periods of incubation during egg storage (SPIDES) reduces the adverse consequences of prolonged storage. SPIDES increases blastodermal cell viability by reducing apoptosis, though the counteracting mechanisms are unclear. To define the impact of prolonged storage and SPIDES, transcriptome analysis compared gene expression from blastoderms isolated from eggs exposed to the following treatments: control (CR, stored at 17 °C for 4 days), prolonged storage (NSR, stored at 17 °C for 21 days), SPIDES (SR, stored at 17 °C for 21 days with SPIDES), and incubated control (C2, stored at 17 °C for 4 days followed by incubation to HH (Hamburger-Hamilton) stage 2, used as the ideal standard development) (n = 3/group). Data analysis was performed using the CLC Genomics Workbench platform. Functional annotation was performed using DAVID and QIAGEN Ingenuity Pathway Analysis. RESULTS In total, 4726 DEGs (differentially expressed genes) were identified across all experimental group comparisons (q < 0.05, FPKM> 20, |fold change| > 1.5). DEGs common across experimental comparisons were involved in cellular homeostasis and cytoskeletal protein binding. The NSR group exhibited activation of ubiquitination, apoptotic, and cell senescence processes. The SR group showed activation of cell viability, division, and metabolic processes. Through comparison analysis, cellular respiration, tRNA charging, cell cycle control, and HMBG1 signaling pathways were significantly impacted by treatment and potential regulatory roles for ribosomal protein L23a (RPL23A) and MYC proto-oncogene, BHLH transcription factor (MYC) were identified. CONCLUSIONS Prolonged egg storage (NSR) resulted in enriched cell stress and death pathways; while SPIDES (SR) resulted in enriched basic cell and anti-apoptotic pathways. New insights into DNA repair mechanisms, RNA processing, shifts in metabolism, and chromatin dynamics in relation to egg storage treatment were obtained through this study. Although egg storage protocols have been examined through targeted gene expression approaches, this study provided a global view of the extensive molecular networks affected by prolonged storage and SPIDES and helped to identify potential upstream regulators for future experiments to optimize egg storage parameters.
Collapse
Affiliation(s)
- K Brady
- Animal Biosciences and Biotechnology Laboratory, BARC, ARS, USDA, 10300 Baltimore Ave. Bldg. 200, Rm. 103, Beltsville, MD, 20705, USA.
| | - C C Talbot
- Institute for Basic Biomedical Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - J A Long
- Animal Biosciences and Biotechnology Laboratory, BARC, ARS, USDA, 10300 Baltimore Ave. Bldg. 200, Rm. 103, Beltsville, MD, 20705, USA
| | - G Welch
- Animal Biosciences and Biotechnology Laboratory, BARC, ARS, USDA, 10300 Baltimore Ave. Bldg. 200, Rm. 103, Beltsville, MD, 20705, USA
| | - N French
- Aviagen Ltd., Newbridge, Midlothian, EH28 8SZ, UK
| | - D Nicholson
- Aviagen Ltd., Newbridge, Midlothian, EH28 8SZ, UK
| | - M R Bakst
- Animal Biosciences and Biotechnology Laboratory, BARC, ARS, USDA, 10300 Baltimore Ave. Bldg. 200, Rm. 103, Beltsville, MD, 20705, USA
| |
Collapse
|
12
|
Robaszkiewicz K, Jurewicz E, Moraczewska J, Filipek A. Ca 2+-dependent binding of S100A6 to cofilin-1 regulates actin filament polymerization-depolymerization dynamics. Cell Calcium 2021; 99:102457. [PMID: 34464867 DOI: 10.1016/j.ceca.2021.102457] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/10/2021] [Accepted: 08/11/2021] [Indexed: 12/15/2022]
Abstract
S100A6 is a Ca2+-binding protein belonging to the S100 family. Many reports indicate that S100A6 is involved in actin filament organization, however the mechanism of S100A6 action in this process is not fully understood. By screening S100A6 binding partners in NIH3T3 mouse fibroblasts, we have found that S100A6 binds cofilin-1, a protein required for the dynamics of actin polymerization and depolymerization. By applying various biochemical and cell biology assays, we have shown that S100A6 bound to cofilin-1 in a Ca2+-dependent manner and increased cofilin-1 affinity for F-actin. Microscopic analysis indicated that S100A6 significantly decreased severing of the actin filaments induced by cofilin-1. Moreover, in the presence of cofilin-1, S100A6 stabilized the filaments by inhibiting their depolymerization. When S100A6 was present at sub-stoichiometric concentrations in relation to actin, polymerization of G-actin accelerated by cofilin-1 was increased. At higher S100A6:actin ratios the polymerization rate was decreased. Altogether, these results show that S100A6 regulates actin filament dynamics by controlling activity of cofilin-1 and suggest that this regulation is Ca2+ -dependent.
Collapse
Affiliation(s)
- Katarzyna Robaszkiewicz
- Kazimierz Wielki University, Department of Biological Sciences, 12 Poniatowskiego Street, 85-671 Bydgoszcz, Poland
| | - Ewelina Jurewicz
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Joanna Moraczewska
- Kazimierz Wielki University, Department of Biological Sciences, 12 Poniatowskiego Street, 85-671 Bydgoszcz, Poland.
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warsaw, Poland.
| |
Collapse
|
13
|
Beltran-Camacho L, Jimenez-Palomares M, Rojas-Torres M, Sanchez-Gomar I, Rosal-Vela A, Eslava-Alcon S, Perez-Segura MC, Serrano A, Antequera-González B, Alonso-Piñero JA, González-Rovira A, Extremera-García MJ, Rodriguez-Piñero M, Moreno-Luna R, Larsen MR, Durán-Ruiz MC. Identification of the initial molecular changes in response to circulating angiogenic cells-mediated therapy in critical limb ischemia. Stem Cell Res Ther 2020; 11:106. [PMID: 32143690 PMCID: PMC7060566 DOI: 10.1186/s13287-020-01591-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/10/2020] [Accepted: 02/06/2020] [Indexed: 12/18/2022] Open
Abstract
Background Critical limb ischemia (CLI) constitutes the most aggressive form of peripheral arterial occlusive disease, characterized by the blockade of arteries supplying blood to the lower extremities, significantly diminishing oxygen and nutrient supply. CLI patients usually undergo amputation of fingers, feet, or extremities, with a high risk of mortality due to associated comorbidities. Circulating angiogenic cells (CACs), also known as early endothelial progenitor cells, constitute promising candidates for cell therapy in CLI due to their assigned vascular regenerative properties. Preclinical and clinical assays with CACs have shown promising results. A better understanding of how these cells participate in vascular regeneration would significantly help to potentiate their role in revascularization. Herein, we analyzed the initial molecular mechanisms triggered by human CACs after being administered to a murine model of CLI, in order to understand how these cells promote angiogenesis within the ischemic tissues. Methods Balb-c nude mice (n:24) were distributed in four different groups: healthy controls (C, n:4), shams (SH, n:4), and ischemic mice (after femoral ligation) that received either 50 μl physiological serum (SC, n:8) or 5 × 105 human CACs (SE, n:8). Ischemic mice were sacrificed on days 2 and 4 (n:4/group/day), and immunohistochemistry assays and qPCR amplification of Alu-human-specific sequences were carried out for cell detection and vascular density measurements. Additionally, a label-free MS-based quantitative approach was performed to identify protein changes related. Results Administration of CACs induced in the ischemic tissues an increase in the number of blood vessels as well as the diameter size compared to ischemic, non-treated mice, although the number of CACs decreased within time. The initial protein changes taking place in response to ischemia and more importantly, right after administration of CACs to CLI mice, are shown. Conclusions Our results indicate that CACs migrate to the injured area; moreover, they trigger protein changes correlated with cell migration, cell death, angiogenesis, and arteriogenesis in the host. These changes indicate that CACs promote from the beginning an increase in the number of vessels as well as the development of an appropriate vascular network. Graphical abstract ![]()
Collapse
Affiliation(s)
- Lucia Beltran-Camacho
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Margarita Jimenez-Palomares
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Marta Rojas-Torres
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Ismael Sanchez-Gomar
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Antonio Rosal-Vela
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Sara Eslava-Alcon
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | | | - Ana Serrano
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain
| | - Borja Antequera-González
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Jose Angel Alonso-Piñero
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Almudena González-Rovira
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | - Mª Jesús Extremera-García
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain.,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain
| | | | - Rafael Moreno-Luna
- Laboratory of Neuroinflammation, Hospital Nacional de Paraplejicos, SESCAM, Toledo, Spain
| | - Martin Røssel Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Mª Carmen Durán-Ruiz
- Biomedicine, Biotechnology and Public Health Department, Cádiz University, Cadiz, Spain. .,Institute of Biomedical Research Cadiz (INIBICA), Cadiz, Spain.
| |
Collapse
|
14
|
S100 proteins in atherosclerosis. Clin Chim Acta 2020; 502:293-304. [DOI: 10.1016/j.cca.2019.11.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 02/07/2023]
|
15
|
Haldar B, Hamilton CL, Solodushko V, Abney KA, Alexeyev M, Honkanen RE, Scammell JG, Cioffi DL. S100A6 is a positive regulator of PPP5C-FKBP51-dependent regulation of endothelial calcium signaling. FASEB J 2020; 34:3179-3196. [PMID: 31916625 DOI: 10.1096/fj.201901777r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/18/2019] [Accepted: 12/19/2019] [Indexed: 11/11/2022]
Abstract
ISOC is a cation current permeating the ISOC channel. In pulmonary endothelial cells, ISOC activation leads to formation of inter-endothelial cell gaps and barrier disruption. The immunophilin FK506-binding protein 51 (FKBP51), in conjunction with the serine/threonine protein phosphatase 5C (PPP5C), inhibits ISOC . Free PPP5C assumes an autoinhibitory state, which has low "basal" catalytic activity. Several S100 protein family members bind PPP5C increasing PPP5C catalytic activity in vitro. One of these family members, S100A6, exhibits a calcium-dependent translocation to the plasma membrane. The goal of this study was to determine whether S100A6 activates PPP5C in pulmonary endothelial cells and contributes to ISOC inhibition by the PPP5C-FKBP51 axis. We observed that S100A6 activates PPP5C to dephosphorylate tau T231. Following ISOC activation, cytosolic S100A6 translocates to the plasma membrane and interacts with the TRPC4 subunit of the ISOC channel. Global calcium entry and ISOC are decreased by S100A6 in a PPP5C-dependent manner and by FKBP51 in a S100A6-dependent manner. Further, calcium entry-induced endothelial barrier disruption is decreased by S100A6 dependent upon PPP5C, and by FKBP51 dependent upon S100A6. Overall, these data reveal that S100A6 plays a key role in the PPP5C-FKBP51 axis to inhibit ISOC and protect the endothelial barrier against calcium entry-induced disruption.
Collapse
Affiliation(s)
- Barnita Haldar
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| | - Caleb L Hamilton
- Department of Anatomy and Molecular Medicine, Alabama College of Osteopathic Medicine, Dothan, AL, USA
| | - Viktoriya Solodushko
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Kevin A Abney
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | - Mikhail Alexeyev
- Center for Lung Biology, University of South Alabama, Mobile, AL, USA.,Physiology and Cell Biology, University of South Alabama, Mobile, AL, USA
| | - Richard E Honkanen
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA
| | | | - Donna L Cioffi
- Departments of Biochemistry and Molecular Biology, University of South Alabama, Mobile, AL, USA.,Center for Lung Biology, University of South Alabama, Mobile, AL, USA
| |
Collapse
|
16
|
Filippini A, D'Amore A, D'Alessio A. Calcium Mobilization in Endothelial Cell Functions. Int J Mol Sci 2019; 20:ijms20184525. [PMID: 31547344 PMCID: PMC6769945 DOI: 10.3390/ijms20184525] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/02/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Endothelial cells (ECs) constitute the innermost layer that lines all blood vessels from the larger arteries and veins to the smallest capillaries, including the lymphatic vessels. Despite the histological classification of endothelium of a simple epithelium and its homogeneous morphological appearance throughout the vascular system, ECs, instead, are extremely heterogeneous both structurally and functionally. The different arrangement of cell junctions between ECs and the local organization of the basal membrane generate different type of endothelium with different permeability features and functions. Continuous, fenestrated and discontinuous endothelia are distributed based on the specific function carried out by the organs. It is thought that a large number ECs functions and their responses to extracellular cues depend on changes in intracellular concentrations of calcium ion ([Ca2+]i). The extremely complex calcium machinery includes plasma membrane bound channels as well as intracellular receptors distributed in distinct cytosolic compartments that act jointly to maintain a physiological [Ca2+]i, which is crucial for triggering many cellular mechanisms. Here, we first survey the overall notions related to intracellular Ca2+ mobilization and later highlight the involvement of this second messenger in crucial ECs functions with the aim at stimulating further investigation that link Ca2+ mobilization to ECs in health and disease.
Collapse
Affiliation(s)
- Antonio Filippini
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy.
| | - Antonella D'Amore
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy.
| | - Alessio D'Alessio
- Istituto di Istologia ed Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario "Agostino Gemelli", IRCCS, 00168 Rome, Italy.
| |
Collapse
|
17
|
Lucena S, Varela Coelho A, Anjo SI, Manadas B, Mrljak V, Capela E Silva F, Lamy E, Tvarijonaviciute A. Comparative proteomic analysis of saliva from dogs with and without obesity-related metabolic dysfuntion. J Proteomics 2019; 201:65-72. [PMID: 30991157 DOI: 10.1016/j.jprot.2019.04.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 03/20/2019] [Accepted: 04/05/2019] [Indexed: 12/19/2022]
Abstract
Dogs develop only some of the components of the human metabolic syndrome (MetS). Thus, in order to study possible MetS-related alterations in dogs, human MetS criteria were adapted to define canine MetS or so-called obesity-related metabolic dysfunction (ORMD). The main objective of this study was to identify changes in the salivary proteome of obese dogs with ORMD in comparison with obese dogs without ORMD which may constitute potential salivary biomarkers for assessing ORMD. In a first phase, 12 adult obese dogs with ORMD (N = 6) and without ORMD (N = 6) were included in the study. Subsequently, and with the aim of validating and strengthening the results, additional 12 obese dogs (6 with and 6 without ORMD) were tested in an independent experiment following the same protocol. Saliva samples were subjected to a quantitative proteomics analysis and the levels of nine salivary proteins were found to be significantly different between groups, among them those which had greatest fold-change were proteins involved in glycolysis and oxidative stress. In conclusion, despite metabolic syndrome to include different combinations of diseases, the observation of differences in salivary proteome suggests a potential of this fluid to understand the pathophysiology of the disease. SIGNIFICANCE: This is the first study evaluating proteomes of saliva in dogs, as a non invasive sample, in order to increase knowledge about the metabolic/physiopathological changes related to obesity-related metabolic dysfunction (ORMD) together with the identification of potential biomarkers for its diagnosis. As approximately 20% of dogs with naturally occurring obesity were described to suffer ORMD associated with insulin resistance and hypoadiponectinemia, the fact that indicate possible links between ORMD and associated diseases.
Collapse
Affiliation(s)
- Sónia Lucena
- Institute of Agrarian and Environmental Environmental Sciences (ICAAM), University of Évora, Portugal; Department of Veterinary Medicine, School of Science and Technology, University of Évora, Portugal
| | - Ana Varela Coelho
- Institute of Chemical and Biological Technology (ITQB), New University of Lisbon, Portugal
| | - Sandra I Anjo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal; Faculty of Medicine, University of Coimbra, Portugal
| | - Bruno Manadas
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | - Vladimir Mrljak
- Clinic for Internal Dieaases, Faculty of Veterinary Medicine, University of Zagreb, Heinzelova 55, 10 000 Zagreb, Croatia
| | - Fernando Capela E Silva
- Institute of Agrarian and Environmental Environmental Sciences (ICAAM), University of Évora, Portugal; Department of Biology, School of Science and Technology, University of Évora, Portugal
| | - Elsa Lamy
- Institute of Agrarian and Environmental Environmental Sciences (ICAAM), University of Évora, Portugal
| | - Asta Tvarijonaviciute
- Interlab-UMU, Regional Campus of International Excellence Campus Mare Nostrum, University of Murcia, Murcia 30100, Spain.
| |
Collapse
|
18
|
Graczyk-Jarzynka A, Sobiak B, Mlącki M, Wilanowski T, Leśniak W. S100A6 activates EGFR and its downstream signaling in HaCaT keratinocytes. J Cell Physiol 2019; 234:17561-17569. [PMID: 30805941 DOI: 10.1002/jcp.28379] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 01/13/2023]
Abstract
Epidermal growth factor receptor (EGFR) is a central transmitter of mitogenic signals in epithelial cells; enhanced EGFR activity is observed in many tumors of epithelial origin. S100A6 is a small calcium-binding protein, characteristic mainly of epithelial cells and fibroblasts, strongly implicated in cell proliferation and upregulated in tumors. In this study, using biochemical assays along with immunohistochemical and immunocytochemical analysis of organotypic and standard cultures of HaCaT keratinocytes with S100A6 overexpression or knock-down, we have examined the effect of S100A6 on EGFR activity and downstream signaling. We found that HaCaT cells overexpressing S100A6 had enhanced EGFR, phospho EGFR, and phospho extracellular signal-regulated kinase 1/2 (pERK1/2) staining intensity and level coupled to higher signal transducer and activator of transcription 3 (STAT3) activity. Conversely, S100A6 knockdown cells had impaired EGFR signaling that could be enhanced by addition of recombinant S100A6 to the culture media. Altogether the results show that S100A6 may exert its proproliferative effects through activating EGFR.
Collapse
Affiliation(s)
- Agnieszka Graczyk-Jarzynka
- Laboratory of Calcium Binding Proteins, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Barbara Sobiak
- Laboratory of Calcium Binding Proteins, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Michał Mlącki
- Laboratory of Signal Transduction, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Tomasz Wilanowski
- Laboratory of Signal Transduction, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Wiesława Leśniak
- Laboratory of Calcium Binding Proteins, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
19
|
Kasacka I, Piotrowska Ż, Filipek A, Lebkowski W. Comparative evaluation of cannabinoid receptors, apelin and S100A6 protein in the heart of women of different age groups. BMC Cardiovasc Disord 2018; 18:190. [PMID: 30286717 PMCID: PMC6172787 DOI: 10.1186/s12872-018-0923-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 09/18/2018] [Indexed: 12/19/2022] Open
Abstract
Background Recent studies have shown a significant role of the endocannabinoid system, apelin and S100A6 protein in the regulation of cardiovascular system functioning. The aim of the study was to compare and evaluate the distribution of cannabinoid receptors (CB1 and CB2), apelin and S100A6 protein in the heart of healthy women in different age groups. Methods The study was conducted on the hearts of 10 women (organ donors) without a history of cardiovascular disease, who were divided into two age groups: women older than 50 years and women under 50 years of age. Paraffin heart sections were processed by immunohistochemistry for detection of cannabinoids receptors (CB1 and CB2), apelin and S100A6 protein. Results CB1 and CB2 immunoreactivity in the cytoplasm of cardiomyocytes in the heart of women over 50 was weaker than in younger individuals. There was also strong immunoreactivity of CB1 in intercalated discs (ICDs) of the heart, only in women over 50. The presence of this receptor in this location was not found in women under 50. Apelin- and S100A6-immunoreactivity in the cardiomyocytes was stronger in older women compared to women under 50.The CB1, apelin and S100A6 immunostaining in the endothelium of myocardial vessels was weaker in women over 50 than in younger women, while intensity of CB2- immunoreaction in coronary endothelium was similar in both groups of women. The results of the study indicate the important role of endocannabinoids, apelin, and S100A6 protein in cardiac muscle function. Conclusion This report might contribute to a better understanding of the role of endocannabinoid system, apelin and S100 proteins in heart function as well as shed new light on processes involved in age-related cardiomyopathy.
Collapse
Affiliation(s)
- Irena Kasacka
- Department of Histology and Cytophysiology, Medical University of Bialystok, Mickiewicza 2C street, 15-222, Białystok, Poland.
| | - Żaneta Piotrowska
- Department of Histology and Cytophysiology, Medical University of Bialystok, Mickiewicza 2C street, 15-222, Białystok, Poland
| | - Anna Filipek
- Nencki Institute of Experimental Biology, Laboratory of Calcium Binding Proteins, Ludwika Pasteura 3 street, 02-093, Warszawa, Poland
| | - Wojciech Lebkowski
- Department of Neurosurgery, Medical University of Bialystok, Marii Skłodowskiej-Curie 24A street, 15-276, Białystok, Poland
| |
Collapse
|
20
|
EIF3C-enhanced exosome secretion promotes angiogenesis and tumorigenesis of human hepatocellular carcinoma. Oncotarget 2018; 9:13193-13205. [PMID: 29568350 PMCID: PMC5862571 DOI: 10.18632/oncotarget.24149] [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: 10/11/2017] [Accepted: 01/03/2018] [Indexed: 01/18/2023] Open
Abstract
Targeting tumor angiogenesis is a common strategy against human hepatocellular carcinoma (HCC). However, identification of molecular targets as biomarker for elevating therapeutic efficacy is critical to prolong HCC patient survival. Here, we showed that EIF3C (eukaryotic translation initiation factor 3 subunit C) is upregulated during HCC tumor progression and associated with poor patient survival. Expression of EIF3C did not alter proliferation and expression of other tumor progressive genes such as HIF1A, TGFβ1 and VEGF, but reduced cell migration in HCC cells. Nevertheless, expression of EIF3C in HCC cells significantly increase secretion of extracellular exosomes confirmed by increased exosomes labelling by PKH26 fluorescent dye, vesicles in exosome size detected by electronic microscopy and nanoparticle tracking analysis, and expression of divergent exosome markers. The EIF3C-increased exosomes were oncogenic to potentiate tumor angiogenesis via tube formation of HUVEC cells and growth of vessels by plugs assays on nude mice. Subcutaneous inoculation of EIF3C-exosomes mixed with Huh7 HCC cells not only promoted growth of vessels but also increased expression of EIF3C in tumors. Conversely, treatment of exosome inhibitor GW4869 reversed aforementioned oncogenic assays. We identified EIF3C activated expression of S100A11 involved in EIF3C-exosome increased tube formation in angiogenesis. Simultaneous high expression of EIF3C and S100A11 in human HCC tumors for RNA level in TCGA and protein level by IHC are associated with poor survival of HCC patients. Collectively, our results demonstrated that EIF3C overexpression is a potential target of angiogenesis for treatment with exosome inhibitor or S100A11 reduction to suppress HCC angiogenesis and tumorigenesis.
Collapse
|
21
|
Donato R, Sorci G, Giambanco I. S100A6 protein: functional roles. Cell Mol Life Sci 2017; 74:2749-2760. [PMID: 28417162 PMCID: PMC11107720 DOI: 10.1007/s00018-017-2526-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Revised: 02/24/2017] [Accepted: 02/27/2017] [Indexed: 12/20/2022]
Abstract
S100A6 protein belongs to the A group of the S100 protein family of Ca2+-binding proteins. It is expressed in a limited number of cell types in adult normal tissues and in several tumor cell types. As an intracellular protein, S100A6 has been implicated in the regulation of several cellular functions, such as proliferation, apoptosis, the cytoskeleton dynamics, and the cellular response to different stress factors. S100A6 can be secreted/released by certain cell types which points to extracellular effects of the protein. RAGE (receptor for advanced glycation endproducts) and integrin β1 transduce some extracellular S100A6's effects. Dosage of serum S100A6 might aid in diagnosis in oncology.
Collapse
Affiliation(s)
- Rosario Donato
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy.
- Department of Experimental Medicine, Istituto Interuniversitario di Miologia (Interuniversity Institute for Myology), Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy.
| | - Guglielmo Sorci
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
- Department of Experimental Medicine, Istituto Interuniversitario di Miologia (Interuniversity Institute for Myology), Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| | - Ileana Giambanco
- Department of Experimental Medicine, Centro Universitario per la Ricerca sulla Genomica Funzionale, Perugia Medical School, University of Perugia, Piazza Lucio Severi 1, 06132, Perugia, Italy
| |
Collapse
|
22
|
Liu L, Miao L, Liu Y, Qi A, Xie P, Chen J, Zhu H. S100A11 regulates renal carcinoma cell proliferation, invasion, and migration via the EGFR/Akt signaling pathway and E-cadherin. Tumour Biol 2017; 39:1010428317705337. [PMID: 28513300 DOI: 10.1177/1010428317705337] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
S100A11 is a S100 protein family member that contributes to cancer progression. Upregulated in human renal cancer tissues, S100A11 may be a prognostic marker for clear cell renal cell carcinoma, but how it functions in cancer is uncertain. Thus, we studied S100A11 and noted knockdown of S100A11 using short hairpin RNA, which inhibited proliferation, invasion, and migration of renal carcinoma cells as well as increased expression of E-cadherin and decreased expression of epidermal growth factor receptor/Akt in renal carcinoma cells. Therefore, S100A11 may be a key molecular target for treating renal carcinoma.
Collapse
Affiliation(s)
- Lin Liu
- 1 Xuzhou Medical University, Xuzhou, China.,2 Xinyi People's Hospital, Xinyi, China
| | - Long Miao
- 1 Xuzhou Medical University, Xuzhou, China
| | - Yang Liu
- 3 Xuzhou Medical University Affiliated Hospital, Xuzhou, China
| | - Aihua Qi
- 2 Xinyi People's Hospital, Xinyi, China
| | - Ping Xie
- 4 Huai'an Hospital Affiliated of Xuzhou Medical University and Huai'an Second People's Hospital, Huai'an, China
| | - Jiacun Chen
- 3 Xuzhou Medical University Affiliated Hospital, Xuzhou, China
| | - Haitao Zhu
- 3 Xuzhou Medical University Affiliated Hospital, Xuzhou, China
| |
Collapse
|
23
|
Wang XH, Du H, Li L, Shao DF, Zhong XY, Hu Y, Liu YQ, Xing XF, Cheng XJ, Guo T, Li S, Li ZY, Bu ZD, Wen XZ, Zhang LH, Ji JF. Increased expression of S100A6 promotes cell proliferation in gastric cancer cells. Oncol Lett 2016; 13:222-230. [PMID: 28123545 PMCID: PMC5245149 DOI: 10.3892/ol.2016.5419] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 10/26/2016] [Indexed: 11/26/2022] Open
Abstract
S100A6 is involved in regulating the progression of cancer. S100A6 can regulate the dynamics of cytoskeletal constituents, cell growth and differentiation by interacting with binding or target proteins. The present study investigated whether S100A6 affects cell proliferation in gastric cancer cells by stimulating several downstream factors. Firstly, the expression and localization of S100A6 were investigated using immunohistochemical staining, an immunoelectron microscopy and laser confocal scanning. A ChIP-Chip assay was performed to determine the downstream factors of S100A6 using promoter Chip analysis, including approximately the −800 to +200 regions around the transcription starting point. Polymerase chain reaction analysis was performed to confirm this. It was found that the intensity of S100A6 staining was markedly higher in the cytoplasm and nucleus, and its expression level correlated with that of the Ki67 protein. The overexpression of S100A6 also promoted cell proliferation in AGS and BGC823 cell lines, detected using a Cell Counting-Kit 8 assay. In cells overexpressing S100A6, the expression levels of interleukin (IL)-8, cyclin-dependent kinase (CDK)5, CDK4, minichromosome maintenance complex component 7 (MCM7) and B-cell lymphoma 2 (Bcl2) were noticeably increased. In conclusion, the increased expression of S100A6 promoted cell proliferation by regulating the expression levels of IL-8, CDK5, CDK4, MCM7 and Bcl2 in gastric cancer cells.
Collapse
Affiliation(s)
- Xiao-Hong Wang
- Department of Tissue Bank, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Hong Du
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Lin Li
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Duan-Fang Shao
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xi-Yao Zhong
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ying Hu
- Department of Tissue Bank, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Yi-Qiang Liu
- Department of Pathology, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xiao-Fang Xing
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xiao-Jing Cheng
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Ting Guo
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Shen Li
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Zi-Yu Li
- Department of Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Zhao-De Bu
- Department of Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Xian-Zi Wen
- Gastrointestinal Cancer Translational Research Laboratory, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Lian-Hai Zhang
- Department of Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| | - Jia-Fu Ji
- Department of Surgery, Peking University Cancer Hospital and Institute, Beijing 100142, P.R. China
| |
Collapse
|
24
|
Loosen SH, Benz F, Niedeggen J, Schmeding M, Schüller F, Koch A, Vucur M, Tacke F, Trautwein C, Roderburg C, Neumann UP, Luedde T. Serum levels of S100A6 are unaltered in patients with resectable cholangiocarcinoma. Clin Transl Med 2016; 5:39. [PMID: 27709523 PMCID: PMC5052241 DOI: 10.1186/s40169-016-0120-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/10/2016] [Indexed: 02/12/2023] Open
Abstract
Background Elevated expression levels of S100A6, a calcium-binding low-molecular-weight protein, were demonstrated in various malignancies. Moreover, increased serum levels of S100A6 were suggested as a novel biomarker for various inflammatory and malignant diseases including lung and gastric cancer. However, up to now, serum concentrations of S100A6 have not been analyzed in patients with cholangiocarcinoma (CCA). Methods S100A6 mRNA expression levels were analyzed in human and murine CCA tumor samples, using semi-quantitative reverse transcriptase PCR. S100A6 serum concentrations were measured using an enzyme-linked immunosorbent assay in 112 patients with CCA referred to surgery for curative resection and were compared to those of 42 healthy controls. Results were correlated with clinical data. Results S100A6 mRNA expression levels were significantly up-regulated in tumor samples of CCA patients and in tumor tissue of a CCA mouse model. In contrast, serum levels of S100A6 were not significantly altered in patients with CCA compared to healthy controls. Whereas no differences became apparent within the different clinical subgroups of CCA, patients with primary sclerosing cholangitis (PSC)-based CCA displayed higher levels of S100A6 compared to the other patients. Nevertheless, patients with higher S100A6 serum concentrations showed a trend towards an impaired prognosis compared to patients with lower levels. Finally, within our cohort of patients both the diagnostic and prognostic potentials of S100A6 were similar to those of the clinically established biomarkers CEA and CA19-9. Conclusion Although S100A6 was expressed at significantly higher levels in human and murine CCA tumor samples, S100A6 serum levels were not regulated in patients with CCA and are thus not suitable for diagnosis of CCA. However, CCA-patients with elevated S100A6 displayed a trend toward an impaired prognosis compared to patients with lower S100A6 levels, supporting its further evaluation as a prognostic biomarker in CCA. Electronic supplementary material The online version of this article (doi:10.1186/s40169-016-0120-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sven H Loosen
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Fabian Benz
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Jennifer Niedeggen
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Maximilian Schmeding
- Department of Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Florian Schüller
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Alexander Koch
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Mihael Vucur
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Frank Tacke
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Christian Trautwein
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Christoph Roderburg
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Ulf P Neumann
- Department of Surgery, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Tom Luedde
- Department of Medicine III, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany. .,Division of Gastroenterology, Hepatology and Hepatobiliary Oncology, University Hospital RWTH Aachen, Pauwelsstrasse 30, 52074, Aachen, Germany.
| |
Collapse
|
25
|
Lerchenmüller C, Heißenberg J, Damilano F, Bezzeridis VJ, Krämer I, Bochaton-Piallat ML, Hirschberg K, Busch M, Katus HA, Peppel K, Rosenzweig A, Busch H, Boerries M, Most P. S100A6 Regulates Endothelial Cell Cycle Progression by Attenuating Antiproliferative Signal Transducers and Activators of Transcription 1 Signaling. Arterioscler Thromb Vasc Biol 2016; 36:1854-67. [PMID: 27386938 DOI: 10.1161/atvbaha.115.306415] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Accepted: 06/27/2016] [Indexed: 12/11/2022]
Abstract
OBJECTIVE S100A6, a member of the S100 protein family, has been described as relevant for cell cycle entry and progression in endothelial cells. The molecular mechanism conferring S100A6's proliferative actions, however, remained elusive. APPROACH AND RESULTS Originating from the clinically relevant observation of enhanced S100A6 protein expression in proliferating endothelial cells in remodeling coronary and carotid arteries, our study unveiled S100A6 as a suppressor of antiproliferative signal transducers and activators of transcription 1 signaling. Discovery of the molecular liaison was enabled by combining gene expression time series analysis with bioinformatic pathway modeling in S100A6-silenced human endothelial cells stimulated with vascular endothelial growth factor A. This unbiased approach led to successful identification and experimental validation of interferon-inducible transmembrane protein 1 and protein inhibitors of activated signal transducers and activators of transcription as key components of the link between S100A6 and signal transducers and activators of transcription 1. CONCLUSIONS Given the important role of coordinated endothelial cell cycle activity for integrity and reconstitution of the inner lining of arterial blood vessels in health and disease, signal transducers and activators of transcription 1 suppression by S100A6 may represent a promising therapeutic target to facilitate reendothelialization in damaged vessels.
Collapse
Affiliation(s)
- Carolin Lerchenmüller
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries).
| | - Julian Heißenberg
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Federico Damilano
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Vassilios J Bezzeridis
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Isabel Krämer
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Marie-Luce Bochaton-Piallat
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Kristóf Hirschberg
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Martin Busch
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Hugo A Katus
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Karsten Peppel
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Anthony Rosenzweig
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Hauke Busch
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| | - Melanie Boerries
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries).
| | - Patrick Most
- From the Cardiovascular Research Center, Massachusetts General Hospital (C.L., F.D., A.R.), Cardiovascular Institute, Beth Israel Deaconess Medical Center (F.D.), and Boston Children's Hospital (V.J.B.), Harvard Medical School, Boston, MA; Molecular and Translational Cardiology (MTC), Department of Internal Medicine III, University Hospital Heidelberg, Germany (C.L., J.H., I.K., M. Busch, P.M.); Department of Pathology and Immunology, University of Geneva, Switzerland (M.-L.B.-P.); DZHK (German Center for Cardiovascular Research), Partner site Heidelberg/Mannheim, University of Heidelberg, Germany (K.H., M. Busch, H.A.K., P.M.); Center for Translational Medicine, Jefferson Medical College, Philadelphia, PA (K.P., P.M.); Systems Biology of the Cellular Microenvironment Group, Institute of Molecular Medicine and Cell Research, Albert-Ludwigs-University, Freiburg, Germany (H.B., M. Boerries); German Cancer Consortium (DKTK), Freiburg, Germany (H.B., M. Boerries); and German Cancer Research Center (DKFZ), Heidelberg, Germany (H.B., M. Boerries)
| |
Collapse
|
26
|
Miftakhova R, Hedblom A, Batkiewicz L, Anagnosaki L, Zhang Y, Sjölander A, Wingren AG, Wolgemuth DJ, Persson JL. Cyclin A1 regulates the interactions between mouse haematopoietic stem and progenitor cells and their niches. Cell Cycle 2016; 14:1948-60. [PMID: 25785996 DOI: 10.1080/15384101.2015.1026513] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
It remains poorly understood how the haematopoietic stem/progenitor cells (HSPC) are attracted to their niches and the functional consequences of such interaction. In the present study, we show that the cell cycle regulator cyclin A1 in association with vascular endothelial growth factor receptor 1 (VEGFR1), is required for HSPC and their niches to maintain their function and proper interaction. In the absence of cyclin A1, the HSPC in the BM are increased in their frequency and display an increased migratory and homing ability. Concomitantly, the ability of the endosteal and central BM niche zones to attract and home the wild-type HSPC is significantly reduced in cyclin A1-null mice as compared to the wild-type controls. The impaired proliferation and homing of HSPC in the BM of cyclin A1-null mice are attributed to the increased density of microvessels in the endosteal and central BM niche zones, which is associated with the increased VEGFR1 expression. Thus, modulation of cyclin A1 and VEGFR1 in HSPC and their niches may provide new insights into therapeutic approaches.
Collapse
Affiliation(s)
- Regina Miftakhova
- a Division of Experimental Cancer Research; Department of Translational Medicine; Clinical Research Center; Lund University ; Malmö , Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Elevated S100A6 (Calcyclin) enhances tumorigenesis and suppresses CXCL14-induced apoptosis in clear cell renal cell carcinoma. Oncotarget 2016; 6:6656-69. [PMID: 25760073 PMCID: PMC4466641 DOI: 10.18632/oncotarget.3169] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 01/18/2015] [Indexed: 01/17/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is often resistant to existing therapy. We found elevated S100A6 levels in ccRCC tissues, associated with higher grade pathological features and clinical stages in ccRCC patients. Knockdown of S100A6 inhibited cell proliferation in vitro and tumor growth in vivo. Gene expression profiling suggests a novel function of S100A6 in suppressing apoptosis, as well as a relationship between S100A6 and CXCL14, a pro-inflammatory chemokine. We suggest that the S100A6/CXCL14 signaling pathway is a potential therapeutic target in ccRCC.
Collapse
|
28
|
Nusshold C, Üllen A, Kogelnik N, Bernhart E, Reicher H, Plastira I, Glasnov T, Zangger K, Rechberger G, Kollroser M, Fauler G, Wolinski H, Weksler BB, Romero IA, Kohlwein SD, Couraud PO, Malle E, Sattler W. Assessment of electrophile damage in a human brain endothelial cell line utilizing a clickable alkyne analog of 2-chlorohexadecanal. Free Radic Biol Med 2016; 90:59-74. [PMID: 26577177 PMCID: PMC6392177 DOI: 10.1016/j.freeradbiomed.2015.11.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2015] [Revised: 11/05/2015] [Accepted: 11/07/2015] [Indexed: 11/19/2022]
Abstract
Peripheral leukocytes aggravate brain damage by releasing cytotoxic mediators that compromise blood-brain barrier function. One of the oxidants released by activated leukocytes is hypochlorous acid (HOCl) that is formed via the myeloperoxidase-H2O2-chloride system. The reaction of HOCl with the endogenous plasmalogen pool of brain endothelial cells results in the generation of 2-chlorohexadecanal (2-ClHDA), a toxic, lipid-derived electrophile that induces blood-brain barrier dysfunction in vivo. Here, we synthesized an alkynyl-analog of 2-ClHDA, 2-chlorohexadec-15-yn-1-al (2-ClHDyA) to identify potential protein targets in the human brain endothelial cell line hCMEC/D3. Similar to 2-ClHDA, 2-ClHDyA administration reduced cell viability/metabolic activity, induced processing of pro-caspase-3 and PARP, and led to endothelial barrier dysfunction at low micromolar concentrations. Protein-2-ClHDyA adducts were fluorescently labeled with tetramethylrhodamine azide (N3-TAMRA) by 1,3-dipolar cycloaddition in situ, which unveiled a preferential accumulation of 2-ClHDyA adducts in mitochondria, the Golgi, endoplasmic reticulum, and endosomes. Thirty-three proteins that are subject to 2-ClHDyA-modification in hCMEC/D3 cells were identified by mass spectrometry. Identified proteins include cytoskeletal components that are central to tight junction patterning, metabolic enzymes, induction of the oxidative stress response, and electrophile damage to the caveolar/endosomal Rab machinery. A subset of the targets was validated by a combination of N3-TAMRA click chemistry and specific antibodies by fluorescence microscopy. This novel alkyne analog is a valuable chemical tool to identify cellular organelles and protein targets of 2-ClHDA-mediated damage in settings where myeloperoxidase-derived oxidants may play a disease-propagating role.
Collapse
Affiliation(s)
- Christoph Nusshold
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria
| | - Andreas Üllen
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Nora Kogelnik
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Eva Bernhart
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Helga Reicher
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Ioanna Plastira
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Toma Glasnov
- Christian Doppler Laboratory for Flow Chemistry, Institute of Chemistry, University of Graz, Austria
| | | | - Gerald Rechberger
- BioTechMed Graz, Austria; Institute of Molecular Biosciences, NAWI-Graz, University of Graz, Austria; OMICS-Center Graz, BioTechMed Graz, Austria
| | | | - Günter Fauler
- Clinical Institute of Medical and Chemical Laboratory Diagnostics, Medical University of Graz, Austria
| | - Heimo Wolinski
- BioTechMed Graz, Austria; Institute of Molecular Biosciences, NAWI-Graz, University of Graz, Austria
| | - Babette B Weksler
- Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Ignacio A Romero
- Department of Biological Sciences, The Open University, Walton Hall, Milton Keynes MK7 6BJ, UK
| | - Sepp D Kohlwein
- BioTechMed Graz, Austria; Institute of Molecular Biosciences, NAWI-Graz, University of Graz, Austria
| | - Pierre-Olivier Couraud
- Institut Cochin, Inserm, U1016, CNRS UMR 8104, Paris Descartes University, Sorbonne Paris Cité, Paris, France
| | - Ernst Malle
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria
| | - Wolfgang Sattler
- Institute of Molecular Biology and Biochemistry, Medical University of Graz, Austria; BioTechMed Graz, Austria.
| |
Collapse
|
29
|
Huang J, Xie Y, Sun X, Zeh HJ, Kang R, Lotze MT, Tang D. DAMPs, ageing, and cancer: The 'DAMP Hypothesis'. Ageing Res Rev 2015; 24:3-16. [PMID: 25446804 DOI: 10.1016/j.arr.2014.10.004] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2014] [Revised: 09/24/2014] [Accepted: 10/22/2014] [Indexed: 12/25/2022]
Abstract
Ageing is a complex and multifactorial process characterized by the accumulation of many forms of damage at the molecular, cellular, and tissue level with advancing age. Ageing increases the risk of the onset of chronic inflammation-associated diseases such as cancer, diabetes, stroke, and neurodegenerative disease. In particular, ageing and cancer share some common origins and hallmarks such as genomic instability, epigenetic alteration, aberrant telomeres, inflammation and immune injury, reprogrammed metabolism, and degradation system impairment (including within the ubiquitin-proteasome system and the autophagic machinery). Recent advances indicate that damage-associated molecular pattern molecules (DAMPs) such as high mobility group box 1, histones, S100, and heat shock proteins play location-dependent roles inside and outside the cell. These provide interaction platforms at molecular levels linked to common hallmarks of ageing and cancer. They can act as inducers, sensors, and mediators of stress through individual plasma membrane receptors, intracellular recognition receptors (e.g., advanced glycosylation end product-specific receptors, AIM2-like receptors, RIG-I-like receptors, and NOD1-like receptors, and toll-like receptors), or following endocytic uptake. Thus, the DAMP Hypothesis is novel and complements other theories that explain the features of ageing. DAMPs represent ideal biomarkers of ageing and provide an attractive target for interventions in ageing and age-associated diseases.
Collapse
|
30
|
Ismail MF, El Boghdady NA, Shabayek MI, Awida HA, Abozeed H. Evaluation and screening of mRNA S100A genes as serological biomarkers in different stages of bladder cancer in Egypt. Tumour Biol 2015; 37:4621-31. [DOI: 10.1007/s13277-015-4264-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 10/15/2015] [Indexed: 12/18/2022] Open
|
31
|
Egeland EV, Boye K, Pettersen SJ, Haugen MH, Øyjord T, Malerød L, Flatmark K, Mælandsmo GM. Enrichment of nuclear S100A4 during G2/M in colorectal cancer cells: possible association with cyclin B1 and centrosomes. Clin Exp Metastasis 2015; 32:755-67. [PMID: 26349943 DOI: 10.1007/s10585-015-9742-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 09/03/2015] [Indexed: 01/01/2023]
Abstract
S100A4 promotes metastasis in several types of cancer, but the involved molecular mechanisms are still incompletely described. The protein is associated with a wide variety of biological functions and it locates to different subcellular compartments, including nuclei, cytoplasm and extracellular space. Nuclear expression of S100A4 has been associated with more advanced disease stage as well as poor outcome in colorectal cancer (CRC). The present study was initiated to investigate the nuclear function of S100A4 and thereby unravel potential biological mechanisms linking nuclear expression to a more aggressive phenotype. CRC cell lines show heterogeneity in nuclear S100A4 expression and preliminary experiments revealed cells in G2/M to have increased nuclear accumulation compared to G1 and S cells, respectively. Synchronization experiments validated nuclear S100A4 expression to be most prominent in the G2/M phase, but manipulating nuclear levels of S100A4 using lentiviral modified cells failed to induce changes in cell cycle distribution and proliferation. Proximity ligation assay did, however, demonstrate proximity between S100A4 and cyclin B1 in vitro, while confocal microscopy showed S100A4 to localize to areas corresponding to centrosomes in mitotic cells prior to chromosome segregation. This might indicate a novel and uncharacterized function of the metastasis-associated protein in CRC cells.
Collapse
Affiliation(s)
- Eivind Valen Egeland
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway.
| | - Kjetil Boye
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway.,Department of Oncology, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Solveig J Pettersen
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Mads H Haugen
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Tove Øyjord
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Lene Malerød
- Department of Molecular Cell Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway.,Department of Gastroenterological Surgery, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, 0318, Oslo, Norway
| | - Gunhild M Mælandsmo
- Department of Tumor Biology, Institute for Cancer Research, The Norwegian Radium Hospital, Oslo University Hospital, Montebello, 0310, Oslo, Norway. .,Department of Pharmacy, University of Tromsø, 9037, Tromsø, Norway.
| |
Collapse
|
32
|
Coumans JVF, Gau D, Poljak A, Wasinger V, Roy P, Moens PDJ. Profilin-1 overexpression in MDA-MB-231 breast cancer cells is associated with alterations in proteomics biomarkers of cell proliferation, survival, and motility as revealed by global proteomics analyses. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2015; 18:778-91. [PMID: 25454514 DOI: 10.1089/omi.2014.0075] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Despite early screening programs and new therapeutic strategies, metastatic breast cancer is still the leading cause of cancer death in women in industrialized countries and regions. There is a need for novel biomarkers of susceptibility, progression, and therapeutic response. Global analyses or systems science approaches with omics technologies offer concrete ways forward in biomarker discovery for breast cancer. Previous studies have shown that expression of profilin-1 (PFN1), a ubiquitously expressed actin-binding protein, is downregulated in invasive and metastatic breast cancer. It has also been reported that PFN1 overexpression can suppress tumorigenic ability and motility/invasiveness of breast cancer cells. To obtain insights into the underlying molecular mechanisms of how elevating PFN1 level induces these phenotypic changes in breast cancer cells, we investigated the alteration in global protein expression profiles of breast cancer cells upon stable overexpression of PFN1 by a combination of three different proteome analysis methods (2-DE, iTRAQ, label-free). Using MDA-MB-231 as a model breast cancer cell line, we provide evidence that PFN1 overexpression is associated with alterations in the expression of proteins that have been functionally linked to cell proliferation (FKPB1A, HDGF, MIF, PRDX1, TXNRD1, LGALS1, STMN1, LASP1, S100A11, S100A6), survival (HSPE1, HSPB1, HSPD1, HSPA5 and PPIA, YWHAZ, CFL1, NME1) and motility (CFL1, CORO1B, PFN2, PLS3, FLNA, FLNB, NME2, ARHGDIB). In view of the pleotropic effects of PFN1 overexpression in breast cancer cells as suggested by these new findings, we propose that PFN1-induced phenotypic changes in cancer cells involve multiple mechanisms. Our data reported here might also offer innovative strategies for identification and validation of novel therapeutic targets and companion diagnostics for persons with, or susceptibility to, breast cancer.
Collapse
Affiliation(s)
- Joëlle V F Coumans
- 1 School of Science and Technology, University of New England , Armidale, NSW, Australia
| | | | | | | | | | | |
Collapse
|
33
|
CD154-CD40 T-cell co-stimulation pathway is a key mechanism in kidney ischemia-reperfusion injury. Kidney Int 2015; 88:538-49. [PMID: 25993320 PMCID: PMC4558568 DOI: 10.1038/ki.2015.146] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 02/27/2015] [Accepted: 03/26/2015] [Indexed: 01/17/2023]
Abstract
Ischemia-reperfusion occurs in a great many clinical settings and contributes to organ failure or dysfunction. CD154-CD40 signaling in leukocyte–endothelial cell interactions or T-cell activation facilitates tissue inflammation and injury. Here we tested a siRNA anti-CD40 in rodent warm and cold ischemia models to check the therapeutic efficacy and anti-inflammatory outcome of in vivo gene silencing. In the warm ischemia model different doses were used, resulting in clear renal function improvement and a structural renoprotective effect. Renal ischemia activated the CD40 gene and protein expression, which was inhibited by intravenous siRNA administration. CD40 gene silencing improved renal inflammatory status, as seen by the reduction of CD68 and CD3 T-cell infiltrates, attenuated pro-inflammatory, and enhanced anti-inflammatory mediators. Furthermore, siRNA administration decreased a spleen pro-inflammatory monocyte subset and reduced TNFα secretion by splenic T cells. In the cold ischemia model with syngeneic and allogeneic renal transplantation, the most effective dose induced similar functional and structural renoprotective effects. Our data show the efficacy of our siRNA in modulating both the local and the systemic inflammatory milieu after an ischemic insult. Thus, CD40 silencing could emerge as a novel therapeutic strategy in solid organ transplantation.
Collapse
|
34
|
Li W, Jiang Z, Li T, Wei X, Zheng Y, Wu D, Yang L, Chen S, Xu B, Zhong M, Jiang J, Hu Y, Su H, Zhang M, Huang X, Geng S, Weng J, Du X, Liu P, Li Y, Liu H, Yao Y, Li P. Genome-wide analyses identify KLF4 as an important negative regulator in T-cell acute lymphoblastic leukemia through directly inhibiting T-cell associated genes. Mol Cancer 2015; 14:26. [PMID: 25644173 PMCID: PMC4350611 DOI: 10.1186/s12943-014-0285-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 12/29/2014] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Kruppel-like factor 4 (KLF4) induces tumorigenesis or suppresses tumor growth in a tissue-dependent manner. However, the roles of KLF4 in hematological malignancies and the mechanisms of action are not fully understood. METHODS Inducible KLF4-overexpression Jurkat cell line combined with mouse models bearing cell-derived xenografts and primary T-cell acute lymphoblastic leukemia (T-ALL) cells from four patients were used to assess the functional role of KLF4 in T-ALL cells in vitro and in vivo. A genome-wide RNA-seq analysis was conducted to identify genes regulated by KLF4 in T-ALL cells. Chromatin immunoprecipitation (ChIP) PCR was used to determine direct binding sites of KLF4 in T-ALL cells. RESULTS Here we reveal that KLF4 induced apoptosis through the BCL2/BCLXL pathway in human T-ALL cell lines and primary T-ALL specimens. In consistence, mice engrafted with KLF4-overexpressing T-ALL cells exhibited prolonged survival. Interestingly, the KLF4-induced apoptosis in T-ALL cells was compromised in xenografts but the invasion capacity of KLF4-expressing T-ALL cells to hosts was dramatically dampened. We found that KLF4 overexpression inhibited T cell-associated genes including NOTCH1, BCL11B, GATA3, and TCF7. Further mechanistic studies revealed that KLF4 directly bound to the promoters of NOTCH1, BCL2, and CXCR4 and suppressed their expression. Additionally, KLF4 induced SUMOylation and degradation of BCL11B. CONCLUSIONS These results suggest that KLF4 as a major transcription factor that suppresses the expression of T-cell associated genes, thus inhibiting T-ALL progression.
Collapse
Affiliation(s)
- Wei Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, 510530, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Zhiwu Jiang
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, 510530, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Tianzhong Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, 510530, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Xinru Wei
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, 510530, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Yi Zheng
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, 510530, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Donghai Wu
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, 510530, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| | - Lijian Yang
- Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China.
| | - Shaohua Chen
- Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China.
| | - Bing Xu
- Department of Hematology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, China.
| | - Mei Zhong
- Department of Obstetrics and Gynecology, Nan Fang Hospital of Southern Medical University, Guangzhou, 510515, China.
| | - Jue Jiang
- School of Pharmacy, Tongji Medical College, Huazhong Unviersity of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| | - Yufeng Hu
- School of Pharmacy, Tongji Medical College, Huazhong Unviersity of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| | - Hexiu Su
- School of Pharmacy, Tongji Medical College, Huazhong Unviersity of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| | - Minjie Zhang
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen University Town, Shenzhen, 518055, China.
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, No. 11 Xizhimen South St., Beijing, 100044, China.
| | - Suxia Geng
- Department of Hematology, Guangdong Provincial People's Hospital, Guangzhou, 510500, China.
| | - Jianyu Weng
- Department of Hematology, Guangdong Provincial People's Hospital, Guangzhou, 510500, China.
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital, Guangzhou, 510500, China.
| | - Pentao Liu
- Wellcome Trust Sanger Institute, Hinxton, Cambridge, CB10 1HH, England, UK.
| | - Yangqiu Li
- Institute of Hematology, Medical College, Jinan University, Guangzhou, 510632, China. .,Key Laboratory for Regenerative Medicine of Ministry of Education, Jinan University, Guangzhou, 510632, China.
| | - Hudan Liu
- School of Pharmacy, Tongji Medical College, Huazhong Unviersity of Science and Technology, 13 Hangkong Road, Wuhan, 430030, China.
| | - Yao Yao
- Drug Discovery Pipeline, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, 510530, China.
| | - Peng Li
- Key Laboratory of Regenerative Biology, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Science Park, Guangzhou, Guangdong, 510530, China. .,Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, South China Institute for Stem Cell Biology and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
| |
Collapse
|
35
|
Foertsch F, Teichmann N, Kob R, Hentschel J, Laubscher U, Melle C. S100A11 is involved in the regulation of the stability of cell cycle regulator p21(CIP1/WAF1) in human keratinocyte HaCaT cells. FEBS J 2013; 280:3840-53. [PMID: 23745637 DOI: 10.1111/febs.12378] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Revised: 06/03/2013] [Accepted: 06/04/2013] [Indexed: 01/13/2023]
Abstract
The cyclin-dependent kinase inhibitor p21(CIP1/WAF1) is a regulatory factor of the cell cycle. Its transcriptional activation and protein stability are tightly controlled by several distinct mechanisms. S100A11 is a member of the S100 family of Ca²⁺-binding proteins involved in several biological processes, including cell cycle progression and signal transduction. In the present study, we show that down-regulation of S100A11 results in the reduction of p21 protein in human HaCaT keratinocytes. It appears that a ubiquitin-independent proteasomal degradation process is involved in p21 degradation in S100A11 down-regulated cells. The application of a proteasome inhibitor stabilized p21 protein in these cells. Analysis of distinct signal transduction pathways revealed a disturbed phosphatidylinositol-3-kinase/Akt pathway after S100A11 knockdown. We determined that the glycogen synthase kinase-3, which is negatively regulated by phosphatidylinositol 3-kinase/Akt, was activated in cells possessing knocked-down S100A11 and appears to be involved in p21 protein destabilization. The application of a specific inhibitor of glycogen synthase kinase 3 resulted in an increase of the p21 protein level in S100A11 down-regulated HaCaT cells. Glycogen synthase kinase 3 is able to phosphorylate p21 at T57, which induces p21 proteasomal turnover. Mutation of the glycogen synthase kinase 3 site threonine 57 into alanine (T57A) stabilizes p21 in HaCaT cells lacking S100A11. Beside decreased p21 protein, down-regulation of S100A11 triggered the induction of apoptosis in HaCaT cells. These observations suggest that S100A11 is involved in the maintenance of p21 protein stability and appears to function as an inhibitor of apoptosis in human HaCaT keratinocyte cells. Thus, the data shed light on a novel pathway regulating p21 protein stability.
Collapse
|
36
|
Graczyk A, Słomnicki LP, Leśniak W. S100A6 competes with the TAZ2 domain of p300 for binding to p53 and attenuates p53 acetylation. J Mol Biol 2013; 425:3488-94. [PMID: 23796514 DOI: 10.1016/j.jmb.2013.06.007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 06/08/2013] [Accepted: 06/14/2013] [Indexed: 11/30/2022]
Abstract
S100A6 is a calcium binding protein that, like some other members of the S100 protein family, is able to bind p53. This interaction may be physiologically relevant considering the numerous connotations of S100 proteins and of S100A6, in particular, with cancer and metastasis. In this work, we show that the interaction with S100A6 is limited to unmodified or phosphorylated p53 and is inhibited by p53 acetylation. Using in vitro acetylation assay, we show that the presence of S100A6 attenuates p53 acetylation by p300. Furthermore, using ELISA, we show that S100A6 and the TAZ2 domain of p300 bind p53 with similar affinities and that S100A6 effectively competes with TAZ2 for binding to p53. Our results add another element to the complicated scheme of p53 activation.
Collapse
Affiliation(s)
- Agnieszka Graczyk
- Nencki Institute of Experimental Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | | | | |
Collapse
|
37
|
Kozhevnikova OS, Korbolina EE, Ershov NI, Kolosova NG. Rat retinal transcriptome: effects of aging and AMD-like retinopathy. Cell Cycle 2013; 12:1745-61. [PMID: 23656783 DOI: 10.4161/cc.24825] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Pathogenesis of age-related macular degeneration (AMD), the leading cause of vision loss in the elderly, remains poorly understood due to the paucity of animal models that fully replicate the human disease. Recently, we showed that senescence-accelerated OXYS rats develop a retinopathy similar to human AMD. To identify alterations in response to normal aging and progression of AMD-like retinopathy, we compared gene expression profiles of retina from 3- and 18-mo-old OXYS and control Wistar rats by means of high-throughput RNA sequencing (RNA-Seq). We identified 160 and 146 age-regulated genes in Wistar and OXYS retinas, respectively. The majority of them are related to the immune system and extracellular matrix turnover. Only 24 age-regulated genes were common for the two strains, suggestive of different rates and mechanisms of aging. Over 600 genes showed significant differences in expression between the two strains. These genes are involved in disease-associated pathways such as immune response, inflammation, apoptosis, Ca ( 2+) homeostasis and oxidative stress. The altered expression for selected genes was confirmed by qRT-PCR analysis. To our knowledge, this study represents the first analysis of retinal transcriptome from young and old rats with biologic replicates generated by RNA-Seq technology. We can conclude that the development of AMD-like retinopathy in OXYS rats is associated with an imbalance in immune and inflammatory responses. Aging alters the expression profile of numerous genes in the retina, and the genetic background of OXYS rats has a profound impact on the development of AMD-like retinopathy.
Collapse
Affiliation(s)
- Oyuna S Kozhevnikova
- Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (SB RAS), Novosibirsk, Russia
| | | | | | | |
Collapse
|