1
|
Wei H, Zhang Y, Gao M, Yang J, Wang S, Zhou X, Wei H, Xiao F. Loss of FAM172A gene prompts cell proliferation in liver regeneration. Mol Cell Biochem 2025; 480:1183-1195. [PMID: 38896202 DOI: 10.1007/s11010-024-05044-7] [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] [Received: 11/24/2023] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
The present study was designed to explore the function of FAM172A in liver regeneration and HCC. Mice were sacrificed after 70% partial hepatectomy (PH). RNA sequencing was performed on primary hepatocytes of WT and FAM172A-/- mice. We used HepG2 cells to construct cell lines with stably knockdown and overexpression of FAM172A. The expression of FAM172A in liver tissues was investigated by immunohistochemical staining, and we also used public database to perform survival analysis and prognostic model in HCC. Compared with WT mice after PH, normalized liver weight/body weight (LW/BW) ratio and the proliferating cell nuclear antigen (PCNA) protein level of FAM172A-/- mice elevated. The DEGs were mainly enriched in inflammatory response, tumor necrosis factor production, and wound healing. FAM172A knockdown enhanced the NFκB-TNFα and pERK-YAP1-Cyclin D1 axis. FAM172A peptide inhibited proliferation of primary hepatocytes. Moreover, the low expression of FAM172A in human HCC tissues implies a lower likelihood of survival and a valid diagnostic marker for HCC. Loss of FAM172A gene promotes cell proliferation by pERK-YAP1-Cyclin D1 and pNFκB-TNFα pathways during liver regeneration after PH. FAM172A may be a favorable diagnosis marker of HCC.
Collapse
Affiliation(s)
- Herui Wei
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Yifan Zhang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, 100044, China
| | - Meixin Gao
- Department of Gastroenterology, Beijing Tongren Hospital, Capital Medical University, Beijing, 100176, China
| | - Junru Yang
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Shiwei Wang
- Department of Gastroenterology, Peking University People's Hospital, Beijing, 100044, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, Beijing, 100044, China
| | - Xingang Zhou
- Department of Pathology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China
| | - Hongshan Wei
- Department of Gastroenterology, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| | - Fan Xiao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
- Beijing Institute of Infectious Diseases, Beijing, 100015, China.
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.
| |
Collapse
|
2
|
Lee G, Muir TW. Distinct phases of cellular signaling revealed by time-resolved protein synthesis. Nat Chem Biol 2024; 20:1353-1360. [PMID: 38977789 DOI: 10.1038/s41589-024-01677-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 06/12/2024] [Indexed: 07/10/2024]
Abstract
The post-translational regulation of protein function is involved in most cellular processes. As such, synthetic biology tools that operate at this level provide opportunities for manipulating cellular states. Here we deploy proximity-triggered protein trans-splicing technology to enable the time-resolved synthesis of target proteins from premade parts. The modularity of the strategy allows for the addition or removal of various control elements as a function of the splicing reaction, in the process permitting the cellular location and/or activity state of starting materials and products to be differentiated. The approach is applied to a diverse set of proteins, including the kinase oncofusions breakpoint cluster region-Abelson (BCR-ABL) and DNAJ-PKAc where dynamic cellular phosphorylation events are dissected, revealing distinct phases of signaling and identifying molecular players connecting the oncofusion to cancer transformation as new therapeutic targets of cancer cells. We envision that the tools and control strategies developed herein will allow the activity of both naturally occurring and designer proteins to be harnessed for basic and applied research.
Collapse
Affiliation(s)
- Gihoon Lee
- Department of Chemistry, Princeton University, Princeton, NJ, USA
| | - Tom W Muir
- Department of Chemistry, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
3
|
Hecker M, Fitzner B, Koczan D, Klehmet J, Grothe M, Schwab M, Winkelmann A, Meister S, Dudesek A, Ludwig-Portugall I, Eulitz K, Zettl UK. Differential gene expression in B cells and T helper cells following high-dose glucocorticoid therapy for multiple sclerosis relapse. Biomed Pharmacother 2024; 175:116721. [PMID: 38749180 DOI: 10.1016/j.biopha.2024.116721] [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: 03/05/2024] [Revised: 04/30/2024] [Accepted: 05/06/2024] [Indexed: 06/03/2024] Open
Abstract
BACKGROUND Despite remarkable advances in the therapy of multiple sclerosis (MS), patients with MS may still experience relapses. High-dose short-term methylprednisolone (MP) remains the standard treatment in the acute management of MS relapses due to its potent anti-inflammatory and immunosuppressive properties. However, there is a lack of studies on the cell type-specific transcriptome changes that are induced by this synthetic glucocorticoid (GC). Moreover, it is not well understood why some patients do not benefit adequately from MP therapy. METHODS We collected peripheral blood from MS patients in relapse immediately before and after ∼3-5 days of therapy with MP at 4 study centers. CD19+ B cells and CD4+ T cells were then isolated for profiling the transcriptome with high-density arrays. The patients' improvement of neurological symptoms was evaluated after ∼2 weeks by the treating physicians. We finally analyzed the data to identify genes that were differentially expressed in response to the therapy and whose expression differed between clinical responders and non-responders. RESULTS After MP treatment, a total of 33 genes in B cells and 55 genes in T helper cells were significantly up- or downregulated. The gene lists overlap in 10 genes and contain genes that have already been described as GC-responsive genes in the literature on other cell types and diseases. Their differential expression points to a rapid and coordinated modulation of multiple signaling pathways that influence transcription. Genes that were previously suggested as potential prognostic biomarkers of the clinical response to MP therapy could not be confirmed in our data. However, a greater increase in the expression of genes encoding proteins with antimicrobial activity was detected in CD4+ T cells from non-responders compared to responders. CONCLUSION Our study delved into the cell type-specific effects of MP at the transcriptional level. The data suggest a therapy-induced ectopic expression of some genes (e.g., AZU1, ELANE and MPO), especially in non-responders. The biological consequences of this remain to be explored in greater depth. A better understanding of the molecular mechanisms underlying clinical recovery from relapses in patients with MS will help to optimize future treatment decisions.
Collapse
Affiliation(s)
- Michael Hecker
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany.
| | - Brit Fitzner
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Dirk Koczan
- Institute of Immunology, Rostock University Medical Center, Rostock, Germany
| | - Juliane Klehmet
- Center for Multiple Sclerosis, Department of Neurology, Jüdisches Krankenhaus Berlin, Berlin, Germany
| | - Matthias Grothe
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Schwab
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Alexander Winkelmann
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Stefanie Meister
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | - Ales Dudesek
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| | | | | | - Uwe Klaus Zettl
- Division of Neuroimmunology, Department of Neurology, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
4
|
Kale R, Samant C, Bokare A, Verma M, Nandakumar K, Bhonde M. Inhibition of SGK1 potentiates the anticancer activity of PI3K inhibitor in NSCLC cells through modulation of mTORC1, p‑ERK and β‑catenin signaling. Biomed Rep 2023; 19:94. [PMID: 37901878 PMCID: PMC10603377 DOI: 10.3892/br.2023.1676] [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: 08/18/2023] [Accepted: 09/29/2023] [Indexed: 10/31/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the deadliest types of cancer with poor prognosis, accounting for 85% of all lung cancer cases. The phosphoinositide 3-kinase (PI3K) signaling pathway is most frequently altered in NSCLC; nonetheless, targeting this pathway yields limited success primarily because of drug-induced resistance. PI3K-independent activation of serum and glucocorticoid-induced kinase 1 (SGK1) is responsible for development of resistance to PI3K/AKT inhibitors in breast cancer. The present study investigated potential of inhibiting SGK1 activity for the potentiation of PI3K inhibitor activity in NSCLC cell lines using in vitro anti-proliferation assays, protein expression profiling using western blotting and cell cycle analysis. The findings revealed that combined inhibition of PI3K/AKT and SGK1 resulted in synergistic anticancer activity, with increased apoptosis, DNA damage and cell cycle arrest in G1 phase. Furthermore, high SGK1 protein expression in NSCLC cell lines was associated with increased resistance to PI3K inhibitors. Therefore, enhanced SGK1 expression may serve as a marker to predict therapeutic response to PI3K/AKT inhibitors. Profiling of downstream signaling proteins demonstrated that, at the molecular level SGK1-mediated sensitization of NSCLC cell lines to PI3K inhibitors was achieved via inhibition of mTORC1 signaling. Increased sensitivity of NSCLC cell lines was also mediated by other oncogenic pathways, such as Ras/MEK/ERK and Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Ramesh Kale
- Department of Pharmacology, Novel Drug Discovery and Development, Lupin Limited, Pune, Maharashtra 412115, India
| | - Charudatt Samant
- Department of Pharmacology, Novel Drug Discovery and Development, Lupin Limited, Pune, Maharashtra 412115, India
| | - Anand Bokare
- Department of Pharmacology, Novel Drug Discovery and Development, Lupin Limited, Pune, Maharashtra 412115, India
| | - Mahip Verma
- Department of Pharmacology, Novel Drug Discovery and Development, Lupin Limited, Pune, Maharashtra 412115, India
| | - Krishnadas Nandakumar
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Mandar Bhonde
- Department of Pharmacology, Novel Drug Discovery and Development, Lupin Limited, Pune, Maharashtra 412115, India
| |
Collapse
|
5
|
Lee G, Muir TW. Distinct phases of cellular signaling revealed by time-resolved protein synthesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.10.548208. [PMID: 37503273 PMCID: PMC10369872 DOI: 10.1101/2023.07.10.548208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
The post-translational regulation of protein function is involved in most cellular processes. As such, synthetic biology tools that operate at this level provide opportunities for manipulating cellular states. Here, we deploy a proximity-triggered protein trans-splicing technology to enable the time-resolved synthesis of target proteins from pre-made parts. The modularity of the strategy allows for the addition or removal of various control elements as a function of the splicing reaction, in the process permitting the cellular location and/or activity state of starting materials and products to be differentiated. The approach is applied to a diverse set of proteins, including the kinase oncofusions BCR/ABL and DNAJB1/PRKACA where dynamic cellular phosphorylation events are dissected, revealing distinct phases of signaling and identifying molecular players connecting the oncofusion to cancer transformation as novel therapeutic targets of cancer cells. We envision that the tools and control strategies developed herein will allow the activity of both naturally occurring and designer proteins to be harnessed for basic and applied research.
Collapse
Affiliation(s)
- Gihoon Lee
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Tom W. Muir
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
6
|
Li X, Wang Z, Jiao C, Zhang Y, Xia N, Yu W, Chen X, Wikana LP, Liu Y, Sun L, Chen M, Xiao Y, Shi Y, Han S, Pu L. Hepatocyte SGK1 activated by hepatic ischemia-reperfusion promotes the recurrence of liver metastasis via IL-6/STAT3. J Transl Med 2023; 21:121. [PMID: 36788538 PMCID: PMC9926712 DOI: 10.1186/s12967-023-03977-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/09/2023] [Indexed: 02/16/2023] Open
Abstract
BACKGROUND Liver metastasis is the leading cause of death in patients with colorectal cancer (CRC). Surgical resection of the liver metastases increases the incidence of long-term survival in patients with colorectal liver metastasis (CRLM). However, many patients experience CRLM recurrence after the initial liver resection. As an unavoidable pathophysiological process in liver surgery, liver ischemia-reperfusion (IR) injury increases the risk of tumor recurrence and metastasis. METHODS Colorectal liver metastasis (CRLM) mouse models and mouse liver partial warm ischemia models were constructed. The levels of lipid peroxidation were detected in cells or tissues. Western Blot, qPCR, elisa, immunofluorescence, immunohistochemistry, scanning electron microscope, flow cytometry analysis were conducted to evaluate the changes of multiple signaling pathways during CRLM recurrence under liver ischemia-reperfusion (IR) background, including SGK1/IL-6/STAT3, neutrophil extracellular traps (NETs) formation, polymorphonuclear myeloid-derived suppressor cell (PMN-MDSC) infiltration. RESULTS Hepatocyte serum/glucocorticoid regulated kinase 1 (SGK1) was activated in response to hepatic ischemia-reperfusion injury to pass hepatocyte STAT3 phosphorylation and serum amyloid A (SAA) hyperactivation signals in CRLM-IR mice, such regulation is dependent on SGK-activated IL-6 autocrine. Administration of the SGK1 inhibitor GSK-650394 further reduced ERK-related neutrophil extracellular traps (NETs) formation and polymorphonucler myeloid-derived suppressor cells (PMN-MDSC) infiltration compared with targeting hepatocyte SGK1 alone, thereby alleviating CRLM in the context of IR. CONCLUSIONS Our study demonstrates that hepatocyte and immune cell SGK1 synergistically promote postoperative CRLM recurrence in response to hepatic IR stress, and identifies SGK1 as a translational target that may improve postoperative CRLM recurrence.
Collapse
Affiliation(s)
- Xiangdong Li
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Ziyi Wang
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Chenyu Jiao
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Yu Zhang
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Nan Xia
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Wenjie Yu
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Xuejiao Chen
- grid.89957.3a0000 0000 9255 8984Department of General Surgery, Affiliated Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Likalamu Pascalia Wikana
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Yue Liu
- grid.89957.3a0000 0000 9255 8984Department of General Surgery, Affiliated Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China
| | - Linfeng Sun
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Minhao Chen
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Yuhao Xiao
- grid.412676.00000 0004 1799 0784Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China ,grid.477246.40000 0004 1803 0558Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China ,grid.89957.3a0000 0000 9255 8984NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China
| | - Yuhua Shi
- Department of General Surgery, Affiliated Yancheng School of Clinical Medicine of Nanjing Medical University, Yancheng, China.
| | - Sheng Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China. .,NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China. .,Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, China. .,NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, China.
| |
Collapse
|
7
|
Huang H, Yao H, Wei Y, Chen M, Sun J. Cellular senescence-related long noncoding ribonucleic acids: Predicting prognosis in hepatocellular carcinoma. Cancer Rep (Hoboken) 2023; 6:e1791. [PMID: 36726348 PMCID: PMC10075286 DOI: 10.1002/cnr2.1791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 01/10/2023] [Accepted: 01/21/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Due to their inherent role in cell function, long non-coding ribonucleic acids (lncRNAs) mediate changes in the microenvironment, and thereby participate in the development of cellular senescence. AIMS This study aimed to identify cellular senescence-related lncRNAs that could predict the prognosis of liver cancer. METHODS AND RESULTS Gene expression and clinical data were downloaded from the UCSC Xena platform, ICGC, and TCGA databases. Cox regression and LASSO regression were used to establish a cellular senescence-related lncRNA model. ROC curves and Kaplan-Meier survival curves were then constructed to predict patient prognosis. Cox regression analysis and clinical characteristics were used to evaluate the capability of the model. Tumor mutational burden and tumor-infiltrating immune cell analyses were subsequently performed in the risk subgroups and the samples in the entire cohort were reclustered. Finally, potential small molecule immune-targeted drugs were identified based on the model. The cellular senescence-related prognostic model that was constructed based on AGAP11 and FAM182B. Along with the results of Cox regression and Lasso regression, the risk score was found to be an independent factor for predicting overall survival in cohorts. In the subgroup analysis, the prognosis of the low-risk group in each cohort was significantly higher than that of the high-risk group; the area under temporal ROC curves and clinical ROC curves were all greater than 0.65, respectively. C-index shows that the risk scores are greater than 0.6, showing the stability of the model. The high-risk group demonstrated lower tumor microenvironment and higher tumor mutational burden scores, further verifying the reliability of the model grouping results. Analysis of tumor-infiltrating immune cells indicated that CD8+ and γδ T cells were more abundant among patients in the low-risk group; cluster reorganization indicated that the two groups had different prognoses and proportions of immune cells. The p value of potential drugs predicted based on the expression of model lncRNAs were all less than .05, demonstrating the potential of model lncRNAs as therapeutic targets to some extent. CONCLUSION A prognostic model based on cellular senescence-associated lncRNAs was established and this may be used as a potential biomarker for the prognosis assessment of liver cancer patients.
Collapse
Affiliation(s)
- Hao Huang
- Department of Hepatopancreatobiliary Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Hao Yao
- Department of Hepatopancreatobiliary Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yaqing Wei
- Department of Hepatopancreatobiliary Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Ming Chen
- Department of Hepatopancreatobiliary Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Jinjin Sun
- Department of Hepatopancreatobiliary Surgery, The Second Hospital of Tianjin Medical University, Tianjin, China
| |
Collapse
|
8
|
Lengel D, Romm ZL, Bostwick AL, Huh JW, Snyder NW, Smith G, Raghupathi R. Glucocorticoid Receptor Overexpression in the Dorsal Hippocampus Attenuates Spatial Learning and Synaptic Plasticity Deficits Following Pediatric Traumatic Brain Injury. J Neurotrauma 2022; 39:979-998. [PMID: 35293260 DOI: 10.1089/neu.2022.0012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Traumatic brain injury (TBI) in children younger than 4 years old leads to long-term deficits in cognitive and learning abilities that can persist or even worsen as children age into adolescence. In this study, the role of glucocorticoid receptor (GR) function in the dorsal hippocampus (DH) in hippocampal-dependent cognitive function and synaptic plasticity were assessed following injury to the 11-day-old rat. Brain injury produced significant impairments in spatial learning and memory in the Morris water maze in male and female rats at 1-month post-injury (adolescence) which was accompanied by impairments in induction and maintenance of long-term potentiation (LTP) in the CA1 region of the DH. Brain injury resulted in a significant decrease in the expression of the glucocorticoid-inducible gene, serum- and glucocorticoid-kinase 1 (sgk1), suggestive of an impairment in GR transcriptional activity within the hippocampus. Lentiviral transfection of the human GR (hGR) in the DH improved spatial learning and memory in the Morris water maze and attenuated LTP deficits following TBI. GR overexpression in the DH was also associated with a significant increase in the mRNA expression levels of sgk1, and the glutamate receptor subunits GluA1 and GluA2 within the hippocampus. Overall, these findings support an important role of dorsal hippocampal GR function in learning and memory deficits following pediatric TBI and suggest that these effects may be related to the regulation of glutamate receptor subunit expression in the DH.
Collapse
Affiliation(s)
- Dana Lengel
- Drexel University College of Medicine, 12312, Philadelphia, Pennsylvania, United States.,Mount Sinai School of Medicine, 5925, Neuroscience, New York, New York, United States;
| | - Zoe L Romm
- Drexel University College of Medicine, 12312, Neurobiology and Anatomy, Philadelphia, Pennsylvania, United States;
| | - Anna L Bostwick
- Temple University, 6558, Microbiology and Immunology, Philadelphia, Pennsylvania, United States;
| | - Jimmy W Huh
- Childrens Hospital of Philadelphia, Anesthesiology and Critical Care, Critical Care Office-7C26, 34th Street & Civic Center Blvd., Philadelphia, Pennsylvania, United States, 19104;
| | - Nathaniel W Snyder
- Temple University, 6558, Microbiology and Immunology, Philadelphia, Pennsylvania, United States;
| | - George Smith
- Temple University, 6558, Pediatric Research Center, Philadelphia, Pennsylvania, United States;
| | - Ramesh Raghupathi
- Drexel University, 6527, Neurobiology and Anatomy, 2900 Queen Lane, Philadelphia, Philadelphia, Pennsylvania, United States, 19104-2816;
| |
Collapse
|
9
|
Rango E, D'Antona L, Iovenitti G, Brai A, Mancini A, Zamperini C, Trivisani CI, Marianelli S, Fallacara AL, Molinari A, Cianciusi A, Schenone S, Perrotti N, Dreassi E, Botta M. Si113-prodrugs selectively activated by plasmin against hepatocellular and ovarian carcinoma. Eur J Med Chem 2021; 223:113653. [PMID: 34161866 DOI: 10.1016/j.ejmech.2021.113653] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/01/2021] [Accepted: 06/13/2021] [Indexed: 11/25/2022]
Abstract
Si113, a pyrazolo[3,4-d]pyrimidine derivative, gained more attention as an anticancer agent due to its potent anticancer activity on both in vitro and in vivo hepatocellular carcinomas (HCC) and ovarian carcinoma models. But the drawback is the low water solubility which prevents its further development. In this context, we successfully overcame this limitation by synthesizing two novel prodrugs introducing the amino acid sequence D-Ala-Leu-Lys (TP). Moreover, TP sequence has a high affinity with plasmin, a protease recognized as overexpressed in many solid cancers, including HCC and ovarian carcinoma. The prodrugs were synthesized and fully characterized in terms of in vitro ADME properties, plasma stability and plasmin-induced release of the parent drug. The inhibitory activity against Sgk1 was evaluated and in vitro growth inhibition was evaluated on ovarian carcinoma and HCC cell lines in the presence and absence of human plasmin. In vivo pharmacokinetic properties and preliminary tissue distribution confirmed a better profile highlighting the importance of the prodrug approach. Finally, the prodrug antitumor efficacy was evaluated in an HCC xenografted murine model, where a significant reduction (around 90%) in tumor growth was observed. Treatment with ProSi113-TP in combination with paclitaxel in a paclitaxel-resistant ovarian carcinoma xenografted murine model, resulted in an impressive reduction of tumor volume greater than 95%. Our results revealed a promising activity of Si113 prodrugs and pave the way for their further development against resistant cancer.
Collapse
Affiliation(s)
- Enrico Rango
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Lucia D'Antona
- Dipartimento di Scienze della Salute, Università"Magna Graecia" di Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Giulia Iovenitti
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Annalaura Brai
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Arianna Mancini
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Claudio Zamperini
- Lead Discovery Siena S.r.l., Via Vittorio Alfieri 31, 53019, Castelnuovo Berardenga, Siena, Italy
| | - Claudia Immacolata Trivisani
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Stefano Marianelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Anna Lucia Fallacara
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Alessio Molinari
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Annarita Cianciusi
- Dipartimento di Farmacia, Università degli Studi di Genova, Viale Benedetto XV 3, Genoa, 16132, Italy
| | - Silvia Schenone
- Dipartimento di Farmacia, Università degli Studi di Genova, Viale Benedetto XV 3, Genoa, 16132, Italy
| | - Nicola Perrotti
- Dipartimento di Scienze della Salute, Università"Magna Graecia" di Catanzaro, Viale Europa, 88100, Catanzaro, Italy.
| | - Elena Dreassi
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| | - Maurizio Botta
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100, Siena, Italy; Lead Discovery Siena S.r.l., Via Vittorio Alfieri 31, 53019, Castelnuovo Berardenga, Siena, Italy; Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology Temple University, BioLife Science Building, Suite 333, 1900 North 12th Street, Philadelphia, PA, 19122, United States
| |
Collapse
|
10
|
Su M, Zhan L, Zhang Y, Zhang J. Yes-activated protein promotes primary resistance of BRAF V600E mutant metastatic colorectal cancer cells to mitogen-activated protein kinase pathway inhibitors. J Gastrointest Oncol 2021; 12:953-963. [PMID: 34295548 DOI: 10.21037/jgo-21-258] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 06/11/2021] [Indexed: 01/09/2023] Open
Abstract
Background Most colorectal cancer (CRC) patients with the BRAF V600E mutation display resistance to chemotherapy and targeted medicinal treatments. Thus, exploring new drugs and drug resistance mechanisms for the BRAF V600E mutation has become an urgent clinical priority. Methods MTS experiment, cell cloning experiment, cell scratching experiment, Transwell experiment, chromatin immunoprecipitation (ChIP), quantitative polymerase chain reaction (qPCR) and flow cytometry are used. Detect the transcription and protein expression of YAP in colorectal cancer cell lines, establish a transient cell line with YAP gene overexpression and knockdown, and detect the effect of YAP gene expression on the biological functions of colorectal cancer cells RKO and HT-29. And further study the mechanism of YAP regulating the response of RAF and MEK targeted therapy. Results In this study, for the first time, we verified that the expression of transcription factor yes-associated protein (YAP) was upregulated in BRAF V600E mutant CRC cells. After knocking down YAP, we observed a reduction in the growth rate, proliferation, and invasion ability of colon cancer cells. We further verified that YAP knockdown increased sensitivity of BRAF V600E mutant CRC cells to mitogen-activated protein kinase (MAPK) pathway inhibitors. In addition, we clarified the mechanism underlying YAP regulation of RAF and MAPK/extracellular signal-regulated kinase (MEK)-targeted therapy response: YAP cooperates with RAF→MEK pathway inhibitors to regulate the cell cycle, increase cell G1/S phase arrest, and increase apoptosis. Conclusions These results suggest that YAP expression may be related to the primary resistance of MAPK inhibitors in metastatic CRC with the BRAF V600E mutation. Therefore, the combination of YAP and MAPK pathway inhibitors in BRAF V600E mutant metastatic CRC may present a promising treatment method.
Collapse
Affiliation(s)
- Meng Su
- Medical Oncology Department of Gastrointestinal Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Lei Zhan
- Medical Oncology Department of Gastrointestinal Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yong Zhang
- Department of Pathology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Jingdong Zhang
- Medical Oncology Department of Gastrointestinal Cancer, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
11
|
Comprehensive Analysis of ERK1/2 Substrates for Potential Combination Immunotherapies. Trends Pharmacol Sci 2019; 40:897-910. [DOI: 10.1016/j.tips.2019.09.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 12/25/2022]
|
12
|
Zhu L, Dai LM, Shen H, Gu PQ, Zheng K, Liu YJ, Zhang L, Cheng JF. Qing Chang Hua Shi granule ameliorate inflammation in experimental rats and cell model of ulcerative colitis through MEK/ERK signaling pathway. Biomed Pharmacother 2019; 116:108967. [PMID: 31102937 DOI: 10.1016/j.biopha.2019.108967] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/04/2019] [Accepted: 05/08/2019] [Indexed: 01/16/2023] Open
Abstract
Ulcerative colitis (UC), a bowel disease with significant morbidity, is associated with inflammation. In this study, the effect of Qingchang Huashi granule (QCHS) on UC and its underlying mechanisms were explored using both animal and cell culture experiments. A rat UC model was induced with trinitro-benzene-sulfonic acid (TNBS), concentrations of the cytokines IL-1α, IL-6, IL-8, IL-1β, and TNF-α were significantly up-regulated and the concentrations of IL-4, IL-10, and IL-13 were significantly down-regulated compared with the control group (P < 0.05). In contrast, the QCHS and salicylazosulfapyridine (SASP) groups reversed these modulations (P < 0.05). A UC cell model in HT-29 cells was generated using TNF-α combined with lipopolysaccharide treatment. Cells treated with QCHS were used to investigate the possible mechanisms. The expression of apoptosis-related proteins, including Bax/Bcl-2, caspase-3, caspase-9, Fas/Fas-L, and Rafl in the QCHS and SASP groups, were significantly lower than that in the control group in both animal and cell experiments (P < 0.05). In addition, the in vitro results indicate changes in these indicators mediate the MEK/ERK signaling pathways via SGK1. Our results suggested that QCHS could be beneficial in preventing UC progression as an alternative drug for UC treatment.
Collapse
Affiliation(s)
- Lei Zhu
- Affiliated Hospital of Nanjing University of TCM, JiangSu Province Hospital of TCM, Nan Jing, 210029, Jiangsu Province, China
| | - Lu-Ming Dai
- Affiliated Hospital of Nanjing University of TCM, JiangSu Province Hospital of TCM, Nan Jing, 210029, Jiangsu Province, China
| | - Hong Shen
- Affiliated Hospital of Nanjing University of TCM, JiangSu Province Hospital of TCM, Nan Jing, 210029, Jiangsu Province, China.
| | - Pei-Qing Gu
- Affiliated Hospital of Nanjing University of TCM, JiangSu Province Hospital of TCM, Nan Jing, 210029, Jiangsu Province, China
| | - Kai Zheng
- Affiliated Hospital of Nanjing University of TCM, JiangSu Province Hospital of TCM, Nan Jing, 210029, Jiangsu Province, China
| | - Ya-Jun Liu
- Affiliated Hospital of Nanjing University of TCM, JiangSu Province Hospital of TCM, Nan Jing, 210029, Jiangsu Province, China
| | - Lu Zhang
- Affiliated Hospital of Nanjing University of TCM, JiangSu Province Hospital of TCM, Nan Jing, 210029, Jiangsu Province, China
| | - Jia-Fei Cheng
- Affiliated Hospital of Nanjing University of TCM, JiangSu Province Hospital of TCM, Nan Jing, 210029, Jiangsu Province, China
| |
Collapse
|
13
|
Lee LYW, Woolley C, Starkey T, Biswas S, Mirshahi T, Bardella C, Segditsas S, Irshad S, Tomlinson I. Serum- and Glucocorticoid-induced Kinase Sgk1 Directly Promotes the Differentiation of Colorectal Cancer Cells and Restrains Metastasis. Clin Cancer Res 2019; 25:629-640. [PMID: 30322876 PMCID: PMC6339518 DOI: 10.1158/1078-0432.ccr-18-1033] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/22/2018] [Accepted: 10/10/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE The molecular events that determine intestinal cell differentiation are poorly understood and it is unclear whether it is primarily a passive event or an active process. It is clinically important to gain a greater understanding of the process, because in colorectal cancer, the degree of differentiation of a tumor is associated with patient survival. SGK1 has previously been identified as a gene that is principally expressed in differentiated intestinal cells. In colorectal cancer, there is marked downregulation of SGK1 compared with normal tissue.Experimental Design: An inducible SGK1 viral overexpression system was utilized to induce reexpression of SGK1 in colorectal cancer cell lines. Transcriptomic and phenotypic analyses of these colorectal cancer lines was performed and validation in mouse and human cohorts was performed. RESULTS We demonstrate that SGK1 is upregulated in response to, and an important controller of, intestinal cell differentiation. Reexpression of SGK1 in colorectal cancer cell lines results in features of differentiation, decreased migration rates, and inhibition of metastasis in an orthotopic xenograft model. These effects may be mediated, in part, by SGK1-induced PKP3 expression and increased degradation of MYC. CONCLUSIONS Our results suggest that SGK1 is an important mediator of differentiation of colorectal cells and may inhibit colorectal cancer metastasis.
Collapse
Affiliation(s)
- Lennard Y W Lee
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom.
| | - Connor Woolley
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Thomas Starkey
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Sujata Biswas
- Cancer Cell Biology Group, Oxford Centre for Cancer Gene Research, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Tia Mirshahi
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Chiara Bardella
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Stefania Segditsas
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Shazia Irshad
- Molecular Mechanisms of Colorectal Cancer Group, Nuffield Department of Medicine, Oxford, United Kingdom
| | - Ian Tomlinson
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
14
|
Steinberg SF. Post-translational modifications at the ATP-positioning G-loop that regulate protein kinase activity. Pharmacol Res 2018; 135:181-187. [PMID: 30048755 DOI: 10.1016/j.phrs.2018.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 07/10/2018] [Accepted: 07/11/2018] [Indexed: 12/27/2022]
Abstract
Protein kinases are a superfamily of enzymes that control a wide range of cellular functions. These enzymes share a highly conserved catalytic core that folds into a similar bilobar three-dimensional structure. One highly conserved region in the protein kinase core is the glycine-rich loop (or G-loop), a highly flexible loop that is characterized by a consensus GxGxxG sequence. The G-loop points toward the catalytic cleft and functions to bind and position ATP for phosphotransfer. Of note, in many protein kinases, the second and third glycine residues in the G-loop triad flank residues that can be targets for phosphorylation (Ser, Thr, or Tyr) or other post-translational modifications (ubiquitination, acetylation, O-GlcNAcylation, oxidation). There is considerable evidence that cyclin-dependent kinases are held inactive through inhibitory phosphorylation of the conserved Thr/Tyr residues in this position of the G-loop and that dephosphorylation by cellular phosphatases is required for CDK activation and progression through the cell cycle. This review summarizes literature that identifies residues in or adjacent to the G-loop in other protein kinases that are targets for functionally important post-translational modifications.
Collapse
Affiliation(s)
- Susan F Steinberg
- Department of Pharmacology, College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
15
|
Lang F, Pelzl L, Hauser S, Hermann A, Stournaras C, Schöls L. To die or not to die SGK1-sensitive ORAI/STIM in cell survival. Cell Calcium 2018; 74:29-34. [PMID: 29807219 DOI: 10.1016/j.ceca.2018.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 05/02/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022]
Abstract
The pore forming Ca2+ release activated Ca2+ channel (CRAC) isoforms ORAI1-3 and their regulators STIM1,2 accomplish store operated Ca2+ entry (SOCE). Activation of SOCE may lead to cytosolic Ca2+ oscillations, which in turn support cell proliferation and cell survival. ORAI/STIM and thus SOCE are upregulated by the serum and glucocorticoid inducible kinase SGK1, a kinase under powerful genomic regulation and activated by phosphorylation via the phosphoinositol-3-phosphate pathway. SGK1 enhances ORAI1 abundance partially by phosphorylation of Nedd4-2, an ubiquitin ligase priming the channel protein for degradation. The SGK1-phosphorylated Nedd4-2 binds to the protein 14-3-3 and is thus unable to ubiquinate ORAI1. SGK1 further increases the ORAI1 and STIM1 protein abundance by activating nuclear factor kappa B (NF-κB), a transcription factor upregulating the expression of STIM1 and ORAI1. SGK1-sensitive upregulation of ORAI/STIM and thus SOCE is triggered by a wide variety of hormones and growth factors, as well as several cell stressors including ischemia, radiation, and cell shrinkage. SGK1 dependent upregulation of ORAI/STIM confers survival of tumor cells and thus impacts on growth and therapy resistance of cancer. On the other hand, SGK1-dependent upregulation of ORAI1 and STIM1 may support survival of neurons and impairment of SGK1-dependent ORAI/STIM activity may foster neurodegeneration. Clearly, further experimental effort is needed to define the mechanisms linking SGK1-dependent upregulation of ORAI1 and STIM1 to cell survival and to define the impact of SGK1-dependent upregulation of ORAI1 and STIM1 on malignancy and neurodegenerative disease.
Collapse
Affiliation(s)
- Florian Lang
- Department of Vegetative Physiology, Eberhad Karls University, Wilhelmstr. 56, D-72074 Tübingen, Germany.
| | - Lisann Pelzl
- Department of Vegetative Physiology, Eberhad Karls University, Wilhelmstr. 56, D-72074 Tübingen, Germany
| | - Stefan Hauser
- German Center for Neurodegenerative Diseases, Research Site Tübingen, Germany; Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| | - Andreas Hermann
- Department of Neurology and Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden, Germany & DZNE, German Center for Neurodegenerative Diseases, Research Site Dresden, Germany
| | - Christos Stournaras
- Department of Biochemistry, University of Crete Medical School, Heraklion, Greece
| | - Ludger Schöls
- German Center for Neurodegenerative Diseases, Research Site Tübingen, Germany; Department of Neurology and Hertie Institute for Clinical Brain Research, University of Tübingen, Germany
| |
Collapse
|
16
|
Talarico C, Dattilo V, D'Antona L, Barone A, Amodio N, Belviso S, Musumeci F, Abbruzzese C, Bianco C, Trapasso F, Schenone S, Alcaro S, Ortuso F, Florio T, Paggi MG, Perrotti N, Amato R. SI113, a SGK1 inhibitor, potentiates the effects of radiotherapy, modulates the response to oxidative stress and induces cytotoxic autophagy in human glioblastoma multiforme cells. Oncotarget 2017; 7:15868-84. [PMID: 26908461 PMCID: PMC4941283 DOI: 10.18632/oncotarget.7520] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 02/08/2016] [Indexed: 12/31/2022] Open
Abstract
Glioblastoma multiforme (GBM) is the most aggressive CNS tumor and is characterized by a very high frequency of clinical relapse after therapy and thus by a dismal prognosis, which strongly compromises patients survival. We have recently identified the small molecule SI113, as a potent and selective inhibitor of SGK1, a serine/threonine protein kinase, that modulates several oncogenic signaling cascades. The SI113-dependent SGK1 inhibition induces cell death, blocks proliferation and perturbs cell cycle progression by modulating SGK1-related substrates. SI113 is also able to strongly and consistently block, in vitro and in vivo, growth and survival of human hepatocellular-carcinomas, either used as a single agent or in combination with ionizing radiations. In the present paper we aim to study the effect of SI113 on human GBM cell lines with variable p53 expression. Cell viability, cell death, caspase activation and cell cycle progression were then analyzed by FACS and WB-based assays, after exposure to SI113, with or without oxidative stress and ionizing radiations. Moreover, autophagy and related reticulum stress response were evaluated. We show here, that i) SGK1 is over-expressed in highly malignant gliomas and that the treatment with SI113 leads to ii) significant increase in caspase-mediated apoptotic cell death in GBM cell lines but not in normal fibroblasts; iii)enhancement of the effects of ionizing radiations; iv) modulation of the response to oxidative reticulum stress; v) induction of cytotoxic autophagy. Evidence reported here underlines the therapeutic potential of SI113 in GBM, suggesting a new therapeutic strategy either alone or in combination with radiotherapy.
Collapse
Affiliation(s)
- Cristina Talarico
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Vincenzo Dattilo
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Lucia D'Antona
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Agnese Barone
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Nicola Amodio
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Stefania Belviso
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | | | - Claudia Abbruzzese
- Experimental Oncology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Cataldo Bianco
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Francesco Trapasso
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | | | - Stefano Alcaro
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Francesco Ortuso
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Tullio Florio
- Department of Medicina Interna e Specialità Mediche e Center of Excellence per la Ricerca Biomedica (CEBR), University of Genova, Genova, Italy
| | - Marco G Paggi
- Experimental Oncology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Nicola Perrotti
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Rosario Amato
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| |
Collapse
|
17
|
Conza D, Mirra P, Calì G, Tortora T, Insabato L, Fiory F, Schenone S, Amato R, Beguinot F, Perrotti N, Ulianich L. The SGK1 inhibitor SI113 induces autophagy, apoptosis, and endoplasmic reticulum stress in endometrial cancer cells. J Cell Physiol 2017; 232:3735-3743. [PMID: 28177128 DOI: 10.1002/jcp.25850] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Accepted: 02/07/2017] [Indexed: 12/14/2022]
Abstract
Endometrial cancer is often characterized by PI3K/AKT pathway deregulation. Recently it has been suggested that SGK1, a serine/threonine protein kinase that shares structural and functional similarities with the AKT family, might play a role in cancer, since its expression and/or activity has been found to be deregulated in different human tumors. However, the role of SGK1 in endometrial cancer has been poorly investigated. Here, we show that SGK1 expression is increased in tissue specimens from neoplastic endometrium. The SGK1 inhibitor SI113 induced a significant reduction of endometrial cancer cells viability, measured by the (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. This effect was associated to the increase of autophagy, as revealed by the increase of the markers LC3B-II and beclin I, detected by both immunofluorescence and western blot analysis. SI113 treatment caused also apoptosis of endometrial cancer cells, evidenced by the cleavage of the apoptotic markers PARP and Caspase-9. Intriguingly, these effects were associated to the induction of endoplasmic reticulum stress markers GRP78 and CHOP evaluated by both Real-Time RT-PCR and Western Blot analysis. Increased expression of SGK1 in endometrial cancer tissues suggest a role for SGK1 in this type of cancer, as reported for other malignancies. Moreover, the efficacy of SI113 in affecting endometrial cancer cells viability, possibly via endoplasmic reticulum stress activation, identifies SGK1 as an attractive molecular target for new tailored therapeutic intervention for the treatment of endometrial cancer.
Collapse
Affiliation(s)
- Domenico Conza
- Department of Medical and Translational Sciences of the University of Naples "Federico II" & URT dell'Istituto di Endocrinologia e Oncologia Sperimentale 'Gaetano Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Paola Mirra
- Department of Medical and Translational Sciences of the University of Naples "Federico II" & URT dell'Istituto di Endocrinologia e Oncologia Sperimentale 'Gaetano Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Gaetano Calì
- Istituto di Endocrinologia e Oncologia Sperimentale 'Gaetano Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Teresa Tortora
- Department of Medical and Translational Sciences of the University of Naples "Federico II" & URT dell'Istituto di Endocrinologia e Oncologia Sperimentale 'Gaetano Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Luigi Insabato
- Department of Advanced Biomedical Sciences, University "Federico II", Naples, Italy
| | - Francesca Fiory
- Department of Medical and Translational Sciences of the University of Naples "Federico II" & URT dell'Istituto di Endocrinologia e Oncologia Sperimentale 'Gaetano Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | | | - Rosario Amato
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Francesco Beguinot
- Department of Medical and Translational Sciences of the University of Naples "Federico II" & URT dell'Istituto di Endocrinologia e Oncologia Sperimentale 'Gaetano Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Nicola Perrotti
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Catanzaro, Italy
| | - Luca Ulianich
- Department of Medical and Translational Sciences of the University of Naples "Federico II" & URT dell'Istituto di Endocrinologia e Oncologia Sperimentale 'Gaetano Salvatore', Consiglio Nazionale delle Ricerche, Naples, Italy
| |
Collapse
|
18
|
Ulmer TF, Fragoulis A, Dohmeier H, Kroh A, Andert A, Stoppe C, Alizai H, Klink C, Coburn M, Neumann UP. Argon Delays Initiation of Liver Regeneration after Partial Hepatectomy in Rats. Eur Surg Res 2017; 58:204-215. [PMID: 28433997 DOI: 10.1159/000466690] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 02/28/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND The liver can heal up to restitutio ad integrum following damage resulting from various causes. Different studies have demonstrated the protective effect of argon on various cells and organs. To the best of our knowledge, the organ-protective effects of the noble gas argon on the liver have not yet been investigated, although argon appears to influence signal paths that are well-known mediators of liver regeneration. We hypothesized that argon inhalation prior to partial hepatectomy (70%) has a positive effect on the initiation of liver regeneration in rats. METHODS Partial hepatectomy (70%) with or without inhaled argon (50 vol%) was performed for 1 h. Liver tissue was harvested after 3, 36, and 96 h to analyze the mRNA and protein expression of hepatocyte growth factor (HGF), interleukin-6 (IL-6), tumor necrosis factor-α, and extracellular signal-regulated kinase 1/2. Histological tissue samples were prepared for immunohistochemistry (bromodeoxyuridine [BrdU], Ki-67, and TUNEL) and blood was analyzed regarding the effects of argon on liver function. Statistical analyses were performed using 1-way ANOVA followed by the post hoc Tukey-Kramer test. RESULTS After 3 h, the primary outcome parameter of hepatocyte proliferation was significantly reduced with argon 50 vol% inhalation in comparison to nitrogen inhalation (BrdU: 15.7 ± 9.7 vs. 7.7 ± 3.1 positive cells/1,000 hepatocytes, p = 0.013; Ki-67: 17.6 ± 13.3 vs. 4.7 ± 5.4 positive cells/1,000 hepatocytes, p = 0.006). This was most likely mediated by significant downregulation of HGF (after 3 h: 5.2 ± 3.2 vs. 2.3 ± 1.0 fold, p = 0.032; after 96 h: 2.1 ± 0.5 vs. 1.3 ± 0.3 fold, p = 0.029) and IL-6 (after 3 h: 43.7 ± 39.6 vs. 8.5 ± 9.2 fold, p = 0.032). Nevertheless, we could detect no significant effect on the weight of the residual liver, liver-body weight ratio, or liver blood test results after argon inhalation. CONCLUSION Impairment of liver regeneration was apparent after argon 50 vol% inhalation that was most probably mediated by downregulation of HGF and IL-6 in the initial phase. However, the present study was not adequately powered to prove that argon has detrimental effects on the liver. Further studies are needed to evaluate the effects of argon on livers with preexisting conditions as well as on ischemia-reperfusion models.
Collapse
Affiliation(s)
- Tom Florian Ulmer
- Department of General, Visceral, and Transplantation Surgery, University Hospital of RWTH Aachen, Aachen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Le Gallo M, Lozy F, Bell DW. Next-Generation Sequencing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 943:119-148. [DOI: 10.1007/978-3-319-43139-0_5] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
20
|
Talarico C, D'Antona L, Scumaci D, Barone A, Gigliotti F, Fiumara CV, Dattilo V, Gallo E, Visca P, Ortuso F, Abbruzzese C, Botta L, Schenone S, Cuda G, Alcaro S, Bianco C, Lavia P, Paggi MG, Perrotti N, Amato R. Preclinical model in HCC: the SGK1 kinase inhibitor SI113 blocks tumor progression in vitro and in vivo and synergizes with radiotherapy. Oncotarget 2016; 6:37511-25. [PMID: 26462020 PMCID: PMC4741945 DOI: 10.18632/oncotarget.5527] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 09/28/2015] [Indexed: 12/20/2022] Open
Abstract
The SGK1 kinase is pivotal in signal transduction pathways operating in cell transformation and tumor progression. Here, we characterize in depth a novel potent and selective pyrazolo[3,4-d]pyrimidine-based SGK1 inhibitor. This compound, named SI113, active in vitro in the sub-micromolar range, inhibits SGK1-dependent signaling in cell lines in a dose- and time-dependent manner. We recently showed that SI113 slows down tumor growth and induces cell death in colon carcinoma cells, when used in monotherapy or in combination with paclitaxel. We now demonstrate for the first time that SI113 inhibits tumour growth in hepatocarcinoma models in vitro and in vivo. SI113-dependent tumor inhibition is dose- and time-dependent. In vitro and in vivo SI113-dependent SGK1 inhibition determined a dramatic increase in apoptosis/necrosis, inhibited cell proliferation and altered the cell cycle profile of treated cells. Proteome-wide biochemical studies confirmed that SI113 down-regulates the abundance of proteins downstream of SGK1 with established roles in neoplastic transformation, e.g. MDM2, NDRG1 and RAN network members. Consistent with knock-down and over-expressing cellular models for SGK1, SI113 potentiated and synergized with radiotherapy in tumor killing. No short-term toxicity was observed in treated animals during in vivo SI113 administration. These data show that direct SGK1 inhibition can be effective in hepatic cancer therapy, either alone or in combination with radiotherapy.
Collapse
Affiliation(s)
- Cristina Talarico
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Lucia D'Antona
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Domenica Scumaci
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Agnese Barone
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Francesco Gigliotti
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Claudia Vincenza Fiumara
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Vincenzo Dattilo
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Enzo Gallo
- Section of Pathology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Paolo Visca
- Section of Pathology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Francesco Ortuso
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Claudia Abbruzzese
- Experimental Oncology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Lorenzo Botta
- Department of Biotecnologie, Chimica e Farmacia, University of Siena, Siena, Italy
| | | | - Giovanni Cuda
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Stefano Alcaro
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Cataldo Bianco
- Department of "Medicina Sperimentale e Clinica", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Patrizia Lavia
- Institute of Molecular Biology and Pathology (IBPM), National Research Council of Italy (CNR), c/o University "La Sapienza", Rome, Italy
| | - Marco G Paggi
- Experimental Oncology, Regina Elena National Cancer Institute, IRCCS, Rome, Italy
| | - Nicola Perrotti
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| | - Rosario Amato
- Department of "Scienze della Salute", University "Magna Graecia" of Catanzaro, Viale Europa, Catanzaro, Italy
| |
Collapse
|
21
|
Bai JA, Xu GF, Yan LJ, Zeng WW, Ji QQ, Wu JD, Tang QY. SGK1 inhibits cellular apoptosis and promotes proliferation via the MEK/ERK/p53 pathway in colitis. World J Gastroenterol 2015; 21:6180-6193. [PMID: 26034353 PMCID: PMC4445095 DOI: 10.3748/wjg.v21.i20.6180] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2014] [Revised: 12/12/2014] [Accepted: 02/12/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the role of serum-and-glucocorticoid-inducible-kinase-1 (SGK1) in colitis and its potential pathological mechanisms.
METHODS: SGK1 expression in mucosal biopsies from patients with active Crohn’s disease (CD) and normal controls was detected by immunohistochemistry. We established an acute colitis model in mice induced by 2,4,6-trinitrobenzene sulfonicacid, and demonstrated the presence of colitis using the disease activity index, the histologic activity index and hematoxylin and eosin staining. The cellular events and potential mechanisms were implemented with small interference RNA and an inhibitor of signaling molecule (i.e., U0126) in intestinal epithelial cells (IECs). The interaction between SGK1 and the signaling molecule was assessed by co-immunoprecipitation.
RESULTS: SGK1 expression was significantly increased in the inflamed epithelia of patients with active CD and TNBS-induced colitis model (0.58 ± 0.055 vs 0.85 ± 0.06, P < 0.01). At the cellular level, silencing of SGK1 by small interference RNA (siSGK1) significantly inhibited the phosphorylation of mitogen-activated protein kinase kinase 1 (MEK1) and the downstream molecule extracellular signal regulated protein kinase (ERK) 1/2, which induced the upregulation of p53 and Bcl-2-associated X protein, mediating the subsequent cellular apoptosis and proliferation in IECs. Cells treated with MEK1 inhibitor (i.e., U0126) before siSGK1 transfection showed a reversal of the siSGK1-induced cellular apoptosis.
CONCLUSION: Our data suggested that SGK1 may protect IECs in colitis from tumor necrosis factor-α-induced apoptosis partly by triggering MEK/ERK activation.
Collapse
|
22
|
Vasudevan HN, Mazot P, He F, Soriano P. Receptor tyrosine kinases modulate distinct transcriptional programs by differential usage of intracellular pathways. eLife 2015; 4. [PMID: 25951516 PMCID: PMC4450512 DOI: 10.7554/elife.07186] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 05/06/2015] [Indexed: 11/27/2022] Open
Abstract
Receptor tyrosine kinases (RTKs) signal through shared intracellular pathways yet mediate distinct outcomes across many cell types. To investigate the mechanisms underlying RTK specificity in craniofacial development, we performed RNA-seq to delineate the transcriptional response to platelet-derived growth factor (PDGF) and fibroblast growth factor (FGF) signaling in mouse embryonic palatal mesenchyme cells. While the early gene expression profile induced by both growth factors is qualitatively similar, the late response is divergent. Comparing the effect of MEK (Mitogen/Extracellular signal-regulated kinase) and PI3K (phosphoinositide-3-kinase) inhibition, we find the FGF response is MEK dependent, while the PDGF response is PI3K dependent. Furthermore, FGF promotes proliferation but PDGF favors differentiation. Finally, we demonstrate overlapping domains of PDGF-PI3K signaling and osteoblast differentiation in the palate and increased osteogenesis in FGF mutants, indicating this differentiation circuit is conserved in vivo. Our results identify distinct responses to PDGF and FGF and provide insight into the mechanisms encoding RTK specificity. DOI:http://dx.doi.org/10.7554/eLife.07186.001 Cells produce many different proteins that play a variety of important roles. For example, proteins called receptor tyrosine kinases can detect particular molecules and send signals to other parts of the cell to regulate the activity (or “expression”) of genes involved in cell division, movement, and other processes. Humans have 58 receptor tyrosine kinases, and defects in these proteins have been linked to diseases such as cancer and diabetes. However, many different receptors regulate the activities of shared sets of genes, so it is not clear how an individual receptor can specifically control the genes involved in a particular process. Two receptor tyrosine kinases called PDGFR and FGFR are crucial for the development of the face, palate, and head in humans and other animals. Vasudevan et al. used a technique called RNA-sequencing to find out which genes are regulated by these receptors in mouse palate cells. The experiments show that there is a common set of genes whose activities change quickly—within 1 hour—in response to the activation of either PDGFR or FGFR. However, several hours later, cells in which PDGFR is activated have different patterns of gene expression compared to those with active FGFR. Vasudevan et al. also found that FGFR promotes cell division, while PDGFR promotes the changing of palate cells into different types with more specialized roles. These different outcomes arise because PDGFR and FGFR use different signaling pathways that involve distinct proteins. For example, a protein called PI3K is critical for changes in gene expression in response to PDGFR but not FGFR. These results suggest that PGDRF and FGFR control different cellular processes in the palate by sending distinct signals into the cell. Understanding the receptor tyrosine kinases and the networks of genes they activate will help us to identify the signals that are important for other processes, such as the development of the face. DOI:http://dx.doi.org/10.7554/eLife.07186.002
Collapse
Affiliation(s)
- Harish N Vasudevan
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Pierre Mazot
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Fenglei He
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| | - Philippe Soriano
- Department of Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, United States
| |
Collapse
|
23
|
Hepatic serum- and glucocorticoid-regulated protein kinase 1 (SGK1) regulates insulin sensitivity in mice via extracellular-signal-regulated kinase 1/2 (ERK1/2). Biochem J 2015; 464:281-9. [PMID: 25222560 DOI: 10.1042/bj20141005] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Insulin resistance is a major hallmark of metabolic syndromes, including Type 2 diabetes. Although numerous functions of SGK1 (serum- and glucocorticoid-regulated kinase 1) have been identified, a direct effect of SGK1 on insulin sensitivity has not been previously reported. In the present study, we generated liver-specific SGK1-knockout mice and found that these mice developed glucose intolerance and insulin resistance. We also found that insulin signalling is enhanced or impaired in Hep1-6 cells infected with adenoviruses expressing SGK1 (Ad-SGK1) or shRNA directed against the coding region of SGK1 (Ad-shSGK1) respectively. In addition, we determined that SGK1 inhibits ERK1/2 (extracellular-signal-regulated kinase 1/2) activity in liver and Ad-shERK1/2-mediated inhibition of ERK1/2 reverses the attenuated insulin sensitivity in Ad-shSGK1 mice. Finally, we found that SGK1 functions are compromised under insulin-resistant conditions and overexpression of SGK1 by Ad-SGK1 significantly ameliorates insulin resistance in both glucosamine-treated HepG2 cells and livers of db/db mice, a genetic model of insulin resistance.
Collapse
|
24
|
Smith KP, Gifford KM, Waitzman JS, Rice SE. Survey of phosphorylation near drug binding sites in the Protein Data Bank (PDB) and their effects. Proteins 2015; 83:25-36. [PMID: 24833420 PMCID: PMC4233198 DOI: 10.1002/prot.24605] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Revised: 04/28/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022]
Abstract
While it is currently estimated that 40 to 50% of eukaryotic proteins are phosphorylated, little is known about the frequency and local effects of phosphorylation near pharmaceutical inhibitor binding sites. In this study, we investigated how frequently phosphorylation may affect the binding of drug inhibitors to target proteins. We examined the 453 non-redundant structures of soluble mammalian drug target proteins bound to inhibitors currently available in the Protein Data Bank (PDB). We cross-referenced these structures with phosphorylation data available from the PhosphoSitePlus database. Three hundred twenty-two of 453 (71%) of drug targets have evidence of phosphorylation that has been validated by multiple methods or labs. For 132 of 453 (29%) of those, the phosphorylation site is within 12 Å of the small molecule-binding site, where it would likely alter small molecule binding affinity. We propose a framework for distinguishing between drug-phosphorylation site interactions that are likely to alter the efficacy of drugs versus those that are not. In addition we highlight examples of well-established drug targets, such as estrogen receptor alpha, for which phosphorylation may affect drug affinity and clinical efficacy. Our data suggest that phosphorylation may affect drug binding and efficacy for a significant fraction of drug target proteins.
Collapse
Affiliation(s)
- Kyle P Smith
- Department of Cell and Molecular Biology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611
| | | | | | | |
Collapse
|
25
|
Römer M, Eichner J, Metzger U, Templin MF, Plummer S, Ellinger-Ziegelbauer H, Zell A. Cross-platform toxicogenomics for the prediction of non-genotoxic hepatocarcinogenesis in rat. PLoS One 2014; 9:e97640. [PMID: 24830643 PMCID: PMC4022579 DOI: 10.1371/journal.pone.0097640] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 04/10/2014] [Indexed: 02/07/2023] Open
Abstract
In the area of omics profiling in toxicology, i.e. toxicogenomics, characteristic molecular profiles have previously been incorporated into prediction models for early assessment of a carcinogenic potential and mechanism-based classification of compounds. Traditionally, the biomarker signatures used for model construction were derived from individual high-throughput techniques, such as microarrays designed for monitoring global mRNA expression. In this study, we built predictive models by integrating omics data across complementary microarray platforms and introduced new concepts for modeling of pathway alterations and molecular interactions between multiple biological layers. We trained and evaluated diverse machine learning-based models, differing in the incorporated features and learning algorithms on a cross-omics dataset encompassing mRNA, miRNA, and protein expression profiles obtained from rat liver samples treated with a heterogeneous set of substances. Most of these compounds could be unambiguously classified as genotoxic carcinogens, non-genotoxic carcinogens, or non-hepatocarcinogens based on evidence from published studies. Since mixed characteristics were reported for the compounds Cyproterone acetate, Thioacetamide, and Wy-14643, we reclassified these compounds as either genotoxic or non-genotoxic carcinogens based on their molecular profiles. Evaluating our toxicogenomics models in a repeated external cross-validation procedure, we demonstrated that the prediction accuracy of our models could be increased by joining the biomarker signatures across multiple biological layers and by adding complex features derived from cross-platform integration of the omics data. Furthermore, we found that adding these features resulted in a better separation of the compound classes and a more confident reclassification of the three undefined compounds as non-genotoxic carcinogens.
Collapse
Affiliation(s)
- Michael Römer
- Center of Bioinformatics Tuebingen (ZBIT), University of Tuebingen, Tübingen, Germany
| | - Johannes Eichner
- Center of Bioinformatics Tuebingen (ZBIT), University of Tuebingen, Tübingen, Germany
| | - Ute Metzger
- Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Markus F. Templin
- Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Simon Plummer
- CXR Biosciences, James Lindsay Place, Dundee Technopole, Dundee, Scotland, United Kingdom
| | | | - Andreas Zell
- Center of Bioinformatics Tuebingen (ZBIT), University of Tuebingen, Tübingen, Germany
| |
Collapse
|
26
|
Lang F, Voelkl J. Therapeutic potential of serum and glucocorticoid inducible kinase inhibition. Expert Opin Investig Drugs 2013; 22:701-14. [PMID: 23506284 DOI: 10.1517/13543784.2013.778971] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Expression of serum-and-glucocorticoid-inducible kinase-1 (SGK1) is low in most cells, but dramatically increases under certain pathophysiological conditions, such as glucocorticoid or mineralocorticoid excess, inflammation with TGFβ release, hyperglycemia, cell shrinkage and ischemia. SGK1 is activated by insulin and growth factors via phosphatidylinositide-3-kinase, 3-phosphoinositide-dependent kinase and mammalian target of rapamycin. SGK1 sensitive functions include activation of ion channels (including epithelial Na(+) channel ENaC, voltage gated Na(+) channel SCN5A transient receptor potential channels TRPV4 - 6, Ca(2+) release activated Ca(2+) channel Orai1/STIM1, renal outer medullary K(+) channel ROMK, voltage gated K(+) channels KCNE1/KCNQ1, kainate receptor GluR6, cystic fibrosis transmembrane regulator CFTR), carriers (including Na(+),Cl(-) symport NCC, Na(+),K(+),2Cl(-) symport NKCC, Na(+)/H(+) exchangers NHE1 and NHE3, Na(+), glucose symport SGLT1, several amino acid transporters), and Na(+)/K(+)-ATPase. SGK1 regulates several enzymes (e.g., glycogen synthase kinase-3, ubiquitin-ligase Nedd4-2) and transcription factors (e.g., forkhead transcription factor 3a, β-catenin, nuclear factor kappa B). AREAS COVERED The phenotype of SGK1 knockout mice is mild and SGK1 is apparently dispensible for basic functions. Excessive SGK1 expression and activity, however, contributes to the pathophysiology of several disorders, including hypertension, obesity, diabetes, thrombosis, stroke, fibrosing disease, infertility and tumor growth. A SGK1 gene variant (prevalence ∼ 3 - 5% in Caucasians and ∼ 10% in Africans) is associated with hypertension, stroke, obesity and type 2 diabetes. SGK1 inhibitors have been developed and shown to reduce blood pressure of hyperinsulinemic mice and to counteract tumor cell survival. EXPERT OPINION Targeting SGK1 may be a therapeutic option in several clinical conditions, including metabolic syndrome and tumor growth.
Collapse
Affiliation(s)
- Florian Lang
- University of Tuebingen, Department of Physiology, Tuebingen, Germany.
| | | |
Collapse
|
27
|
Kuntzsch D, Bergann T, Dames P, Fromm A, Fromm M, Davis RA, Melzig MF, Schulzke JD. The plant-derived glucocorticoid receptor agonist Endiandrin A acts as co-stimulator of colonic epithelial sodium channels (ENaC) via SGK-1 and MAPKs. PLoS One 2012; 7:e49426. [PMID: 23152905 PMCID: PMC3496671 DOI: 10.1371/journal.pone.0049426] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Accepted: 10/07/2012] [Indexed: 11/23/2022] Open
Abstract
In a search for secondary plant compounds that bind to the glucocorticoid receptor (GR), the cyclobutane lignan endiandrin A was discovered from the rainforest tree Endiandra anthropophagorum Domin. Our present study aims to characterize the effect of endiandrin A on GR-dependent induction of colonic sodium transport. The effect of endiandrin A was analyzed in GR-expressing colonic HT-29/B6 cells (HT-29/B6-GR). GR transactivation and subcellular localization were investigated by reporter gene assay and immunofluorescence. Epithelial sodium channel (ENaC) was analyzed by qRT-PCR and by measuring amiloride-sensitive short-circuit current (Isc) in Ussing chambers. Endiandrin A (End A) has been identified as GR receptor binder. However, it did not cause significant GR transactivation as pGRE-luciferase activity was only 7% of that of the maximum effect of dexamethasone. Interestingly, endiandrin A had a significant impact on dexamethasone-dependent sodium absorption in cells co-exposed to tumor necrosis factor (TNF)-α. This was in part due to up-regulation of β- and γ-ENaC subunit expression. Endiandrin A potentiated GR-mediated transcription by increasing GR protein expression and phosphorylation. It inhibited c-Jun N-terminal kinase (JNK) activation induced by dexamethasone and/or TNF-α and increased levels of GR localized to the nucleus. Additionally, endiandrin A increased the serum- and glucocorticoid-induced kinase (sgk)-1 via activation of p38. Finally, the regulation of ENaC function by endiandrin A was confirmed in rat native colon. In conclusion, endiandrin A potentiates glucocorticoid-driven activation of colonic epithelial sodium channels via JNK inhibition and p38 activation due to transcriptional up-regulation of β- and γ-ENaC-subunits along with induction of sgk-1.
Collapse
Affiliation(s)
- Dana Kuntzsch
- Department of Gastroenterology, Division of Nutritional Medicine, Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Theresa Bergann
- Department of Gastroenterology, Division of Nutritional Medicine, Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Petra Dames
- Department of Gastroenterology, Division of Nutritional Medicine, Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Anja Fromm
- Institute of Clinical Physiology, Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Michael Fromm
- Institute of Clinical Physiology, Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Rohan A. Davis
- Eskitis Institute, Griffith University, Brisbane, Queensland, Australia
| | - Matthias F. Melzig
- Institute of Pharmacy, Pharmaceutical Biology, Freie Universität Berlin, Berlin, Germany
| | - Joerg D. Schulzke
- Department of Gastroenterology, Division of Nutritional Medicine, Charité, Campus Benjamin Franklin, Berlin, Germany
- * E-mail:
| |
Collapse
|
28
|
Hassan M, Selimovic D, El-Khattouti A, Ghozlan H, Haikel Y, Abdelkader O. Hepatitis C virus-host interactions: Etiopathogenesis and therapeutic strategies. World J Exp Med 2012; 2:7-25. [PMID: 24520529 PMCID: PMC3905577 DOI: 10.5493/wjem.v2.i2.7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2012] [Revised: 04/16/2012] [Accepted: 04/18/2012] [Indexed: 02/06/2023] Open
Abstract
Hepatitis C virus (HCV) is a significant health problem facing the world. This virus infects more than 170 million people worldwide and is considered the major cause of both acute and chronic hepatitis. Persons become infected mainly through parenteral exposure to infected material by blood transfusions or injections with nonsterile needles. Although the sexual behavior is considered as a high risk factor for HCV infection, the transmission of HCV infection through sexual means, is less frequently. Currently, the available treatment for patients with chronic HCV infection is interferon based therapies alone or in combination with ribavirin and protease inhibitors. Although a sustained virological response of patients to the applied therapy, a great portion of patients did not show any response. HCV infection is mostly associated with progressive liver diseases including fibrosis, cirrhosis and hepatocellular carcinoma. Although the focus of many patients and clinicians is sometimes limited to that problem, the natural history of HCV infection (HCV) is also associated with the development of several extrahepatic manifestations including dermatologic, rheumatologic, neurologic, and nephrologic complications, diabetes, arterial hypertension, autoantibodies and cryglobulins. Despite the notion that HCV-mediated extrahepatic manifestations are credible, the mechanism of their modulation is not fully described in detail. Therefore, the understanding of the molecular mechanisms of HCV-induced alteration of intracellular signal transduction pathways, during the course of HCV infection, may offer novel therapeutic targets for HCV-associated both hepatic and extrahepatic manifestations. This review will elaborate the etiopathogenesis of HCV-host interactions and summarize the current knowledge of HCV-associated diseases and their possible therapeutic strategies.
Collapse
Affiliation(s)
- Mohamed Hassan
- Mohamed Hassan, Denis Selimovic, Youssef Haikel, National Institute of Health and Medical Research, U 977, Faculty of Medicine, and Dental Faculty, 11 Rue Humann, 67085 Strasbourg Cedex, France
| | - Denis Selimovic
- Mohamed Hassan, Denis Selimovic, Youssef Haikel, National Institute of Health and Medical Research, U 977, Faculty of Medicine, and Dental Faculty, 11 Rue Humann, 67085 Strasbourg Cedex, France
| | - Abdelouahid El-Khattouti
- Mohamed Hassan, Denis Selimovic, Youssef Haikel, National Institute of Health and Medical Research, U 977, Faculty of Medicine, and Dental Faculty, 11 Rue Humann, 67085 Strasbourg Cedex, France
| | - Hanan Ghozlan
- Mohamed Hassan, Denis Selimovic, Youssef Haikel, National Institute of Health and Medical Research, U 977, Faculty of Medicine, and Dental Faculty, 11 Rue Humann, 67085 Strasbourg Cedex, France
| | - Youssef Haikel
- Mohamed Hassan, Denis Selimovic, Youssef Haikel, National Institute of Health and Medical Research, U 977, Faculty of Medicine, and Dental Faculty, 11 Rue Humann, 67085 Strasbourg Cedex, France
| | - Ola Abdelkader
- Mohamed Hassan, Denis Selimovic, Youssef Haikel, National Institute of Health and Medical Research, U 977, Faculty of Medicine, and Dental Faculty, 11 Rue Humann, 67085 Strasbourg Cedex, France
| |
Collapse
|
29
|
Abstract
The serum and glucocorticoid kinase (SGK) family of serine/threonine kinases consists of three isoforms, SGK-1, SGK-2 and SGK-3. This family of kinases is highly homologous to the AKT kinase family, sharing similar upstream activators and downstream targets. SGKs have been implicated in the regulation of cell growth, proliferation, survival and migration: cellular processes that are dysregulated in cancer. Furthermore, SGKs lie downstream of phosphoinositide-3-kinase (PI3Kinase) signalling and interact at various levels with RAS/RAF/ERK signalling, two pathways that are involved in promoting tumorigenesis. Recent evidence suggests that mutant PI3Kinase can induce tumorigenesis through an AKT-independent but SGK3-dependent mechanism, thus implicating SGKs as potential players in malignant transformation. Here, we will review the current state of knowledge on the regulation of the SGKs and their role in normal cell physiology and transformation with a particular focus on SGK3.
Collapse
Affiliation(s)
- Maressa A Bruhn
- Growth Control and Differentiation Program, Peter MacCallum Cancer Centre, Melbourne, 3002, Victoria, Australia
| | | | | | | |
Collapse
|
30
|
Colorectal carcinoma cells--regulation of survival and growth by SGK1. Int J Biochem Cell Biol 2010; 42:1571-5. [PMID: 20541034 DOI: 10.1016/j.biocel.2010.05.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 05/28/2010] [Accepted: 05/30/2010] [Indexed: 12/21/2022]
Abstract
Colorectal carcinoma is among the most common malignancies. The tumour cells may arise from mutations in genes encoding proteins involved in the regulation of cell survival and proliferation. Recent evidence disclosed the sensitivity of colon carcinoma to the expression of ubiquitous serum and glucocorticoid inducible kinase-1 (SGK1). The kinase is activated by insulin and growth factors via the phosphatidylinositide-3-kinase (PI3K) and the 3-phosphoinositide dependent kinase (PDK1). SGK1 regulates channels, carriers and Na(+)/K(+)-ATPase, enzymes such as glycogen-synthase-kinase-3 (GSK3) and ubiquitin-ligase Nedd4-2, as well as several transcription factors. SGK1 regulates transport, hormone release, neuroexcitability, inflammation, cell proliferation and apoptosis. SGK1 contributes to metabolic syndrome and the pathophysiology of neurodegeneration, allergy, peptic ulcer, fibrosing disease and response to ischemia. SGK1 is upregulated in some tumours but downregulated in others. SGK1-sensitive mechanisms fostering tumour growth include activation of K(+) channels and Ca(2+) channels, Na(+)/H(+) exchanger, amino acid transporters and glucose transporters, upregulation of the nuclear factor NFkappaB and beta-catenin as well as downregulation of the transcription factors Foxo3a/FKHRL1 and p53. SGK1 enhances survival, invasiveness, motility, epithelial to mesenchymal transition and adhesiveness of tumour cells. Following deficiency of APC (adenoma polyposis coli) or chemical cancerogenesis, SGK1 knockout mice develop less intestinal tumours than their wild-type littermates and pharmacological SGK1 inhibition counteracts growth of prostate cancer cells.
Collapse
|