1
|
Yarmohammadi F, Karimi G. Serum and glucocorticoid-regulated kinase 1 (SGK1) as an emerging therapeutic target for cardiac diseases. Pharmacol Res 2024; 208:107369. [PMID: 39209082 DOI: 10.1016/j.phrs.2024.107369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 08/11/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Cardiac diseases encompass a wide range of conditions that affect the structure and function of the heart. These conditions are a leading cause of morbidity and mortality worldwide. The serum- and glucocorticoid-inducible kinase 1 (SGK1) is a serine/threonine kinase that plays a significant role in various cellular processes, including cell survival and stress response. Alterations in SGK1 activity can have significant impacts on health and disease. Multiple research findings have indicated that SGK1 is associated with heart disease due to its involvement in cardiac hypertrophy and fibrosis. This article reviews different signaling pathways associated with SGK1 activity in various heart conditions, including the SGK1/NF-κB and PI3K/SGK1 pathways.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Jamshed L, Jamshed S, Frank RA, Hewitt LM, Thomas PJ, Holloway AC. Assessing Receptor Activation in 2D and 3D Cultured Hepatocytes: Responses to a Single Compound and a Complex Mixture. TOXICS 2024; 12:631. [PMID: 39330559 PMCID: PMC11436198 DOI: 10.3390/toxics12090631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 08/07/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024]
Abstract
Responding to global standards and legislative updates in Canada, including Bill S-5 (2023), toxicity testing is shifting towards more ethical, in vitro methods. Traditional two-dimensional (2D) monolayer cell cultures, limited in replicating the complex in vivo environment, have prompted the development of more relevant three-dimensional (3D) spheroidal hepatocyte cultures. This study introduces the first 3D spheroid model for McA-RH7777 cells, assessing xenobiotic receptor activation, cellular signaling, and toxicity against dexamethasone and naphthenic acid (NA)-fraction components; NAFCs. Our findings reveal that 3D McA-RH7777 spheroids demonstrate enhanced sensitivity and more uniform dose-response patterns in gene expression related to xenobiotic metabolism (AhR and PPAR) for both single compounds and complex mixtures. Specifically, 3D cultures showed significant gene expression changes upon dexamethasone exposure and exhibited varying degrees of sensitivity and resistance to the apoptotic effects induced by NAFCs, in comparison to 2D cultures. The optimization of 3D culture conditions enhances the model's physiological relevance and enables the identification of genomic signatures under varied exposures. This study highlights the potential of 3D spheroid cultures in providing a more accurate representation of the liver's microenvironment and advancing our understanding of cellular mechanisms in toxicity testing.
Collapse
Affiliation(s)
- Laiba Jamshed
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4L8, Canada; (L.J.); (S.J.)
| | - Shanza Jamshed
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4L8, Canada; (L.J.); (S.J.)
| | - Richard A. Frank
- Water Science and Technology Directorate, Environment and Climate Change Canada, Burlington, ON L7S 1A1, Canada; (R.A.F.); (L.M.H.)
| | - L. Mark Hewitt
- Water Science and Technology Directorate, Environment and Climate Change Canada, Burlington, ON L7S 1A1, Canada; (R.A.F.); (L.M.H.)
| | - Philippe J. Thomas
- Environment and Climate Change Canada, National Wildlife Research Centre, Ottawa, ON K1S 5B6, Canada;
| | - Alison C. Holloway
- Department of Obstetrics and Gynecology, McMaster University, Hamilton, ON L8S 4L8, Canada; (L.J.); (S.J.)
| |
Collapse
|
3
|
Sun M, Zhang X, Tan B, Zhang Q, Zhao X, Dong D. Potential role of endoplasmic reticulum stress in doxorubicin-induced cardiotoxicity-an update. Front Pharmacol 2024; 15:1415108. [PMID: 39188945 PMCID: PMC11345228 DOI: 10.3389/fphar.2024.1415108] [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: 04/09/2024] [Accepted: 08/01/2024] [Indexed: 08/28/2024] Open
Abstract
As a chemotherapy agent, doxorubicin is used to combat cancer. However, cardiotoxicity has limited its use. The existing strategies fail to eliminate doxorubicin-induced cardiotoxicity, and an in-depth exploration of its pathogenesis is in urgent need to address the issue. Endoplasmic reticulum stress (ERS) occurs when Endoplasmic Reticulum (ER) dysfunction results in the accumulation of unfolded or misfolded proteins. Adaptive ERS helps regulate protein synthesis to maintain cellular homeostasis, while prolonged ERS stimulation may induce cell apoptosis, leading to dysfunction and damage to tissue and organs. Numerous studies on doxorubicin-induced cardiotoxicity strongly link excessive activation of the ERS to mechanisms including oxidative stress, calcium imbalance, autophagy, ubiquitination, and apoptosis. The researchers also found several clinical drugs, chemical compounds, phytochemicals, and miRNAs inhibited doxorubicin-induced cardiotoxicity by targeting ERS. The present review aims to outline the interactions between ERS and other mechanisms in doxorubicin-induced cardiotoxicity and summarize ERS's role in this type of cardiotoxicity. Additionally, the review enumerates several clinical drugs, phytochemicals, chemical compounds, and miRNAs targeting ERS for considering therapeutic regimens that address doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Mingli Sun
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Xin Zhang
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Boxuan Tan
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| | - Qingya Zhang
- Innovation Institute, China Medical University, Shenyang, Liaoning, China
| | - Xiaopeng Zhao
- College of Exercise and Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Dan Dong
- College of Basic Medical Science, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
4
|
Xu G, Li L, Lv M, Li C, Yu J, Zeng X, Meng X, Yu G, Liu K, Cheng S, Luo H, Xu B. Discovery of novel 4-trifluoromethyl-2-anilinoquinoline derivatives as potential anti-cancer agents targeting SGK1. Mol Divers 2024:10.1007/s11030-024-10951-4. [PMID: 39117890 DOI: 10.1007/s11030-024-10951-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 07/25/2024] [Indexed: 08/10/2024]
Abstract
Given the critical necessity for the development of more potent anti-cancer drugs, a series of novel compounds incorporating trifluoromethyl groups within the privileged 2-anilinoquinoline scaffold was designed, synthesized, and subjected to biological evaluation through a pharmacophore hybridization strategy. Upon evaluating the in vitro anti-cancer characteristics of the target compounds, it became clear that compound 8b, which contains a (4-(piperazin-1-yl)phenyl)amino substitution at the 2-position of the quinoline skeleton, displayed superior efficacy against four cancer cell lines by inducing apoptosis and cell cycle arrest. Following research conducted in a PC3 xenograft mouse model, it was found that compound 8b exhibited significant anti-cancer efficacy while demonstrating minimal toxicity. Additionally, the analysis of a 217-kinase panel pinpointed SGK1 as a potential target for this compound class with anti-cancer capabilities. This finding was further verified through molecular docking analysis and cellular thermal shift assays. To conclude, our results emphasize that compound 8b can be used as a lead compound for the development of anti-cancer drugs that target SGK1.
Collapse
Affiliation(s)
- Guangcan Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Lanlan Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Mengfan Lv
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Cheng Li
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
- Guizhou University of Traditional Chinese Medicine, Guiyang, 550025, China
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Xiaoping Zeng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Xueling Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Gang Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Kun Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China.
| | - Bixue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang, 550014, China.
- Natural Products Research Center of Guizhou Province/Guizhou Provincial Engineering Research Center for Natural Drugs, Guiyang, 550014, China.
| |
Collapse
|
5
|
Dong C, Lin JM, Wang Y, Zhu J, Lin L, Xu J, Du J. Exploring the Common Pathogenic Mechanisms of Psoriasis and Atopic Dermatitis: The Interaction between SGK1 and TIGIT Signaling Pathways. Inflammation 2024:10.1007/s10753-024-02115-1. [PMID: 39088121 DOI: 10.1007/s10753-024-02115-1] [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: 05/01/2024] [Revised: 07/17/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
This study aims to explore the common pathogenic mechanisms of psoriasis and atopic dermatitis, two T-cell-mediated autoimmune diseases. Utilizing single-cell transcriptomic sequencing data, we revealed that Treg cells primarily express TIGIT in both psoriasis and atopic dermatitis, and identified a subset of macrophages that highly express SGK1. These cells can interact with T cells via the NECTIN2-TIGIT signaling pathway, inhibiting the differentiation of T cells into a pro-inflammatory phenotype, thereby uncovering a common immunoregulatory mechanism in both diseases. Furthermore, we discovered that inhibition of SGK1 exacerbates the inflammatory response in disease models of both conditions. These findings not only provide a new perspective for a common therapeutic strategy for psoriasis and atopic dermatitis but also highlight the importance of considering these molecular interactions in future treatments. Validation of these observations through further qPCR, immunofluorescence, and animal studies has identified potential new targets for the treatment of psoriasis and atopic dermatitis.
Collapse
Affiliation(s)
- Canbin Dong
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jui-Ming Lin
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Yilun Wang
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Junhao Zhu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Lanmei Lin
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Jinhua Xu
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China
| | - Juan Du
- Department of Dermatology, Shanghai Institute of Dermatology, Huashan Hospital, Fudan University, Shanghai, 200040, People's Republic of China.
| |
Collapse
|
6
|
Lei Z, Pan C, Li F, Wei D, Ma Y. SGK1 promotes the lipid accumulation via regulating the transcriptional activity of FOXO1 in bovine. BMC Genomics 2024; 25:737. [PMID: 39080526 PMCID: PMC11290151 DOI: 10.1186/s12864-024-10644-0] [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: 03/11/2024] [Accepted: 07/22/2024] [Indexed: 08/02/2024] Open
Abstract
OBJECTIVES Serum/glucocorticoid-inducible kinase 1 (SGK1) gene encodes a serine/threonine protein kinase that plays an essential role in cellular stress response and regulation of multiple metabolic processes. However, its role in bovine adipogenesis remains unknown. In this study, we aimed to clarify the role of SGK1 in bovine lipid accumulation and improvement of meat quality. METHODS Preadipocytes were induced to differentiation to detect the temporal expression pattern of SGK1. Heart, liver, lung, spleen, kidney, muscle and fat tissues were collected to detect its tissue expression profile. Recombinant adenovirus and the lentivirus were packaged for overexpression and knockdown. Oil Red O staining, quantitative real-time PCR, Western blot analysis, Yeast two-hybrid assay, luciferase assay and RNA-seq were performed to study the regulatory mechanism of SGK1. RESULTS SGK1 showed significantly higher expression in adipose and significantly induced expression in differentiated adipocytes. Furthermore, overexpression of SGK1 greatly promoted adipogenesis and inhibited proliferation, which could be shown by the remarkable increasement of lipid droplet, and the expression levels of adipogenic marker genes and cell cycle-related genes. Inversely, its knockdown inhibited adipogenesis and facilitated proliferation. Mechanistically, SGK1 regulates the phosphorylation and expression of two critical proteins of FoxO family, FOXO1/FOXO3. Importantly, SGK1 attenuates the transcriptional repression role of FOXO1 for PPARγ via phosphorylating the site S256, then promoting the bovine fat deposition. CONCLUSIONS SGK1 is a required epigenetic regulatory factor for bovine preadipocyte proliferation and differentiation, which contributes to a better understanding of fat deposition and meat quality improvement in cattle.
Collapse
Affiliation(s)
- Zhaoxiong Lei
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Cuili Pan
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Fen Li
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China
| | - Dawei Wei
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China.
| | - Yun Ma
- College of Animal Science and Technology, Ningxia University, Yinchuan, 750021, China.
| |
Collapse
|
7
|
Jadhav SB, Vondrackova M, Potomova P, Sandoval-Acuña C, Smigova J, Klanicova K, Rosel D, Brabek J, Stursa J, Werner L, Truksa J. NDRG1 acts as an oncogene in triple-negative breast cancer and its loss sensitizes cells to mitochondrial iron chelation. Front Pharmacol 2024; 15:1422369. [PMID: 38983911 PMCID: PMC11231402 DOI: 10.3389/fphar.2024.1422369] [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: 04/23/2024] [Accepted: 06/06/2024] [Indexed: 07/11/2024] Open
Abstract
Multiple studies indicate that iron chelators enhance their anti-cancer properties by inducing NDRG1, a known tumor and metastasis suppressor. However, the exact role of NDRG1 remains controversial, as newer studies have shown that NDRG1 can also act as an oncogene. Our group recently introduced mitochondrially targeted iron chelators deferoxamine (mitoDFO) and deferasirox (mitoDFX) as effective anti-cancer agents. In this study, we evaluated the ability of these modified chelators to induce NDRG1 and the role of NDRG1 in breast cancer. We demonstrated that both compounds specifically increase NDRG1 without inducing other NDRG family members. We have documented that the effect of mitochondrially targeted chelators is at least partially mediated by GSK3α/β, leading to phosphorylation of NDRG1 at Thr346 and to a lesser extent on Ser330. Loss of NDRG1 increases cell death induced by mitoDFX. Notably, MDA-MB-231 cells lacking NDRG1 exhibit reduced extracellular acidification rate and grow slower than parental cells, while the opposite is true for ER+ MCF7 cells. Moreover, overexpression of full-length NDRG1 and the N-terminally truncated isoform (59112) significantly reduced sensitivity towards mitoDFX in ER+ cells. Furthermore, cells overexpressing full-length NDRG1 exhibited a significantly accelerated tumor formation, while its N-terminally truncated isoforms showed significantly impaired capacity to form tumors. Thus, overexpression of full-length NDRG1 promotes tumor growth in highly aggressive triple-negative breast cancer.
Collapse
Affiliation(s)
- Sukanya B. Jadhav
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
- Faculty of Sciences, Charles University, Prague, Czechia
| | - Michaela Vondrackova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
- Faculty of Sciences, Charles University, Prague, Czechia
- Faculty of Sciences, BIOCEV Research Centre, Charles University, Vestec, Czechia
| | - Petra Potomova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
- Faculty of Sciences, Charles University, Prague, Czechia
| | - Cristian Sandoval-Acuña
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| | - Jana Smigova
- Faculty of Sciences, BIOCEV Research Centre, Charles University, Vestec, Czechia
| | - Kristyna Klanicova
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| | - Daniel Rosel
- Faculty of Sciences, Charles University, Prague, Czechia
- Faculty of Sciences, BIOCEV Research Centre, Charles University, Vestec, Czechia
| | - Jan Brabek
- Faculty of Sciences, Charles University, Prague, Czechia
- Faculty of Sciences, BIOCEV Research Centre, Charles University, Vestec, Czechia
| | - Jan Stursa
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| | - Lukas Werner
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| | - Jaroslav Truksa
- Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV Research Centre, Vestec, Czechia
| |
Collapse
|
8
|
Tan Y, Huang Y, Xu C, Huang X, Li S, Yin Z. Long noncoding RNAs and mRNAs profiling in ovary during laying and broodiness in Taihe Black-Bone Silky Fowls (Gallus gallus Domesticus Brisson). BMC Genomics 2024; 25:357. [PMID: 38600449 PMCID: PMC11005167 DOI: 10.1186/s12864-024-10281-7] [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: 01/11/2024] [Accepted: 04/02/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND Broodiness significantly impacts poultry egg production, particularly notable in specific breeds such as the black-boned Silky, characterized by pronounced broodiness. An understanding of the alterations in ovarian signaling is essential for elucidating the mechanisms that influence broodiness. However, comparative research on the characteristics of long non-coding RNAs (lncRNAs) in the ovaries of broody chickens (BC) and high egg-laying chickens (GC) remains scant. In this investigation, we employed RNA sequencing to assess the ovarian transcriptomes, which include both lncRNAs and mRNAs, in eight Taihe Black-Bone Silky Fowls (TBsf), categorized into broody and high egg-laying groups. This study aims to provide a clearer understanding of the genetic underpinnings associated with broodiness and egg production. RESULTS We have identified a total of 16,444 mRNAs and 18,756 lncRNAs, of which 349 mRNAs and 651 lncRNAs exhibited significantly different expression (DE) between the BC and GC groups. Furthermore, we have identified the cis-regulated and trans-regulated target genes of differentially abundant lncRNA transcripts and have constructed an lncRNA-mRNA trans-regulated interaction network linked to ovarian follicle development. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) annotation analyses have revealed that DE mRNAs and the target genes of DE lncRNAs are associated with pathways including neuroactive ligand-receptor interaction, CCR6 chemokine receptor binding, G-protein coupled receptor binding, cytokine-cytokine receptor interaction, and ECM-receptor interaction. CONCLUSION Our research presents a comprehensive compilation of lncRNAs and mRNAs linked to ovarian development. Additionally, it establishes a predictive interaction network involving differentially abundant lncRNAs and differentially expressed genes (DEGs) within TBsf. This significantly contributes to our understanding of the intricate interactions between lncRNAs and genes governing brooding behavior.
Collapse
Affiliation(s)
- Yuting Tan
- Zijingang Campus, Animal Science College, Zhejiang University, Hangzhou, 310058, China
| | - Yunyan Huang
- Zijingang Campus, Animal Science College, Zhejiang University, Hangzhou, 310058, China
| | - Chunhui Xu
- Zijingang Campus, Animal Science College, Zhejiang University, Hangzhou, 310058, China
| | - Xuan Huang
- Zijingang Campus, Animal Science College, Zhejiang University, Hangzhou, 310058, China
| | - Shibao Li
- Zijingang Campus, Animal Science College, Zhejiang University, Hangzhou, 310058, China
| | - Zhaozheng Yin
- Zijingang Campus, Animal Science College, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
9
|
Xu Z, Hu J, Wei Z, Lei Y, Afewerky HK, Gao Y, Wan L, Li L, Lei L, Liu Y, Huang F, Yu T, Wang J, Li H, Liu R, Wang X. Dynamic changes in lysosome-related pathways in APP/PS1 mice with aging. MedComm (Beijing) 2024; 5:e540. [PMID: 38606360 PMCID: PMC11006716 DOI: 10.1002/mco2.540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 04/13/2024] Open
Abstract
Senile plaque, composed of amyloid β protein (Aβ) aggregates, is a critical pathological feature in Alzheimer's disease (AD), leading to cognitive dysfunction. However, how Aβ aggregates exert age-dependent toxicity and temporal cognitive dysfunction in APP/PS1 mice remains incompletely understood. In this study, we investigated AD pathogenesis and dynamic alterations in lysosomal pathways within the hippocampus of age-gradient male mice using transcriptome sequencing, molecular biology assays, and histopathological analyses. We observed high levels of β-amyloid precursor protein (APP) protein expression in the hippocampus at an early stage and age-dependent Aβ deposition. Transcriptome sequencing revealed the enrichment of differential genes related to the lysosome pathway. Furthermore, the protein expression of ATP6V0d2 and CTSD associated with lysosomal functions exhibited dynamic changes with age, increasing in the early stage and decreasing later. Similar age-dependent patterns were observed for the endosome function, autophagy pathway, and SGK1/FOXO3a pathway. Nissl and Golgi staining in the hippocampal region showed age-dependent neuronal loss and synaptic damage, respectively. These findings clearly define the age-gradient changes in the autophagy-lysosome system, the endosome/lysosome system, and the SGK1/FOXO3a pathway in the hippocampus of APP/PS1 mice, providing new perspectives and clues for understanding the possible mechanisms of AD, especially the transition from compensatory to decompensated state.
Collapse
Affiliation(s)
- Zhendong Xu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jichang Hu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Zhen Wei
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yu Lei
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Henok Kessete Afewerky
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yang Gao
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Lu Wan
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Longfei Li
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Ling Lei
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Yi Liu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Fang Huang
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Tong Yu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Jian‐Zhi Wang
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongChina
| | - Hong‐Lian Li
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Rong Liu
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
| | - Xiaochuan Wang
- Department of PathophysiologySchool of Basic MedicineKey Laboratory of Education Ministry/Hubei Province of China for Neurological DisordersTongji Medical College, Huazhong University of Science and TechnologyWuhanChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
10
|
Zhang Q, Tian Y, Fu Z, Wu S, Lan H, Zhou X, Shen W, Lou Y. The role of serum-glucocorticoid regulated kinase 1 in reproductive viability: implications from prenatal programming and senescence. Mol Biol Rep 2024; 51:376. [PMID: 38427115 PMCID: PMC10907440 DOI: 10.1007/s11033-024-09341-8] [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: 11/21/2023] [Accepted: 02/09/2024] [Indexed: 03/02/2024]
Abstract
OBJECTIVE Organisms and cellular viability are of paramount importance to living creatures. Disruption of the balance between cell survival and apoptosis results in compromised viability and even carcinogenesis. One molecule involved in keeping this homeostasis is serum-glucocorticoid regulated kinase (SGK) 1. Emerging evidence points to a significant role of SGK1 in cell growth and survival, cell metabolism, reproduction, and life span, particularly in prenatal programming and reproductive senescence by the same token. Whether the hormone inducible SGK1 kinase is a major driver in the pathophysiological processes of prenatal programming and reproductive senescence? METHOD The PubMed/Medline, Web of Science, Embase/Ovid, and Elsevier Science Direct literature databases were searched for articles in English focusing on SGK1 published up to July 2023 RESULT: Emerging evidence is accumulating pointing to a pathophysiological role of the ubiquitously expressed SGK1 in the cellular and organismal viability. Under the regulation of specific hormones, extracellular stimuli, and various signals, SGK1 is involved in several biological processes relevant to viability, including cell proliferation and survival, cell migration and differentiation. In line, SGK1 contributes to the development of germ cells, embryos, and fetuses, whereas SGK1 inhibition leads to abnormal gametogenesis, embryo loss, and truncated reproductive lifespan. CONCLUTION SGK1 integrates a broad spectrum of effects to maintain the homeostasis of cell survival and apoptosis, conferring viability to multiple cell types as well as both simple and complex organisms, and thus ensuring appropriate prenatal development and reproductive lifespan.
Collapse
Affiliation(s)
- Qiying Zhang
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Ye Tian
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Zhujing Fu
- Jinhua Municipal Central Hospital, Jinhua, 321001, China
| | - Shuangyu Wu
- Medical School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Huizhen Lan
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Xuanle Zhou
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Wendi Shen
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China
| | - Yiyun Lou
- Department of Gynaecology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, No. 453 Tiyuchang Road, Hangzhou, 310007, Zhejiang, China.
| |
Collapse
|
11
|
Gulzar M, Noor S, Hasan GM, Hassan MI. The role of serum and glucocorticoid-regulated kinase 1 in cellular signaling: Implications for drug development. Int J Biol Macromol 2024; 258:128725. [PMID: 38092114 DOI: 10.1016/j.ijbiomac.2023.128725] [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: 08/10/2023] [Revised: 11/23/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) is a ubiquitously expressed protein belonging to the Ser/Thr kinase family. It regulates diverse physiological processes, including epithelial sodium channel activity, hypertension, cell proliferation, and insulin sensitivity. Due to its significant role in the pathogenesis of numerous diseases, SGK1 can be exploited as a potential therapeutic target to address challenging health problems. SGK1 is associated with the development of obesity, and its overexpression enhances the sodium-glucose co-transporter 1 activity, which absorbs intestinal glucose. This review highlighted the detailed functional significance of SGK1 signaling and role in different diseases and subsequent therapeutic targeting. We aim to provide deeper mechanistic insights into understanding the pathogenesis and recent advancements in the SGK1 targeted drug development process. Small-molecule inhibitors are being developed with excellent binding affinity and improved SGK1 inhibition with desired selectivity. We have discussed small molecule inhibitors designed explicitly as potent SGK1 inhibitors and their therapeutic implications in various diseases. We further addressed the therapeutic potential and mechanism of action of these SGK1 inhibitors and provided a strong scientific foundation for developing effective therapeutics.
Collapse
Affiliation(s)
- Mehak Gulzar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India
| | - Gulam Mustafa Hasan
- Department of Basic Medical Science, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj 11942, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi 110025, India.
| |
Collapse
|
12
|
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
|
13
|
Huo R, Yang Y, Xu H, Zhao S, Song D, Weng J, Ma R, Sun Y, Wang J, Jiao Y, Zhang J, He Q, Wu R, Wang S, Zhao JZ, Zhang J, Wang J, Cao Y. Somatic GJA4 mutation in intracranial extra-axial cavernous hemangiomas. Stroke Vasc Neurol 2023; 8:453-462. [PMID: 37072338 PMCID: PMC10800255 DOI: 10.1136/svn-2022-002227] [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: 12/06/2022] [Accepted: 03/02/2023] [Indexed: 04/20/2023] Open
Abstract
OBJECTIVE Extra-axial cavernous hemangiomas (ECHs) are sporadic and rare intracranial occupational lesions that usually occur within the cavernous sinus. The aetiology of ECHs remains unknown. METHODS Whole-exome sequencing was performed on ECH lesions from 12 patients (discovery cohort) and droplet digital polymerase-chain-reaction (ddPCR) was used to confirm the identified mutation in 46 additional cases (validation cohort). Laser capture microdissection (LCM) was carried out to capture and characterise subgroups of tissue cells. Mechanistic and functional investigations were carried out in human umbilical vein endothelial cells and a newly established mouse model. RESULTS We detected somatic GJA4 mutation (c.121G>T, p.G41C) in 5/12 patients with ECH in the discovery cohort and confirmed the finding in the validation cohort (16/46). LCM followed by ddPCR revealed that the mutation was enriched in lesional endothelium. In vitro experiments in endothelial cells demonstrated that the GJA4 mutation activated SGK-1 signalling that in turn upregulated key genes involved in cell hyperproliferation and the loss of arterial specification. Compared with wild-type littermates, mice overexpressing the GJA4 mutation developed ECH-like pathological morphological characteristics (dilated venous lumen and elevated vascular density) in the retinal superficial vascular plexus at the postnatal 3 weeks, which were reversed by an SGK1 inhibitor, EMD638683. CONCLUSIONS We identified a somatic GJA4 mutation that presents in over one-third of ECH lesions and proposed that ECHs are vascular malformations due to GJA4-induced activation of the SGK1 signalling pathway in brain endothelial cells.
Collapse
Affiliation(s)
- Ran Huo
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yingxi Yang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Hongyuan Xu
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Shaozhi Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Dong Song
- Division of Life Science, Center for Systems Biology and Human Health and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Jiancong Weng
- Department of Neurosurgery, China-Japan Friendship Hospital, Beijing, China
| | - Ruochen Ma
- Division of Life Science, Center for Systems Biology and Human Health and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yingfan Sun
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jie Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yuming Jiao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junze Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ruolei Wu
- School of Medical Technology, Beijing Institute of Technology, Beijing, China
| | - Shuo Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Ji-Zong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Junting Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Jiguang Wang
- Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Hong Kong, China
- Division of Life Science, Center for Systems Biology and Human Health and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, InnoHK, Hong Kong SAR, China
| | - Yong Cao
- Department of Neurosurgery, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing, China
- Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Castillo Cabrera J, Dang H, Graves A, Zhang Z, Torres-Castillo J, Li K, King Z, Liu P, Aubé J, Bear JE, Damania B, Hagan RS, Baldwin AS. AGC kinase inhibitors regulate STING signaling through SGK-dependent and SGK-independent mechanisms. Cell Chem Biol 2023; 30:1601-1616.e6. [PMID: 37939709 PMCID: PMC10842197 DOI: 10.1016/j.chembiol.2023.10.008] [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: 08/06/2022] [Revised: 07/22/2023] [Accepted: 10/13/2023] [Indexed: 11/10/2023]
Abstract
Type 1 IFN expression is critical in the innate immune response, but aberrant expression is associated with autoimmunity and cancer. Here, we identify N-[4-(1H46 pyrazolo[3,4-b] pyrazin-6-yl)-phenyl]-sulfonamide (Sanofi-14h), a compound with preference for inhibition of the AGC family kinase SGK3, as an inhibitor of Ifnb1 gene expression in response to STING stimulation of macrophages. Sanofi-14h abrogated SGK activity and also impaired activation of the critical TBK1/IRF3 pathway downstream of STING activation, blocking interaction of STING with TBK1. Deletion of SGK1/3 in a macrophage cell line did not block TBK1/IRF3 activation but decreased expression of transcription factors, such as IRF7 and STAT1, required for the innate immune response. Other AGC kinase inhibitors blocked TBK1 and IRF3 activation suggesting common action on a critical regulatory node in the STING pathway. These studies reveal both SGK-dependent and SGK-independent mechanisms in the innate immune response and indicate an approach to block aberrant Ifnb1 expression.
Collapse
Affiliation(s)
- Johnny Castillo Cabrera
- Pathobiology and Translational Sciences Graduate Program, Department of Pathology and Laboratory Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Hong Dang
- Division of Pulmonary Diseases and Critical Care Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Marsico Lung Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Adam Graves
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zhigang Zhang
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jose Torres-Castillo
- Division of Pulmonary Diseases and Critical Care Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Marsico Lung Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Kelin Li
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Zayna King
- Department of Cell Biology and Physiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Pengda Liu
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Biochemistry and Biophysics, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jeff Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - James E Bear
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Cell Biology and Physiology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Robert S Hagan
- Division of Pulmonary Diseases and Critical Care Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Marsico Lung Institute, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| | - Albert S Baldwin
- Lineberger Comprehensive Cancer Center, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
15
|
Mozaffari MS. Serum Glucocorticoid-Regulated Kinase-1 in Ischemia-Reperfusion Injury: Blessing or Curse. J Pharmacol Exp Ther 2023; 387:277-287. [PMID: 37770199 DOI: 10.1124/jpet.123.001846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/05/2023] [Accepted: 09/11/2023] [Indexed: 10/03/2023] Open
Abstract
The family of serum-glucocorticoid-regulated kinase (SGK) consists of three paralogs, SGK-1, SGK-2, and SGK-3, with SGK-1 being the better studied. Indeed, recognition of the role of SGK-1 in regulation of cell survival and proliferation has led to introduction of a number of small-molecule inhibitors for some types of cancer. In addition, SGK-1 regulates major physiologic effects, such as renal solute transport, and contributes to the pathogenesis of non-neoplastic conditions involving major organs including the heart and the kidney. These observations raise the prospect for therapeutic modulation of SGK-1 to reduce the burden of such diseases as myocardial infarction and acute kidney injury. Following a brief description of the structure and function of SGK family of proteins, the present review is primarily focused on our current understanding of the role of SGK-1 in pathologies related to ischemia-reperfusion injury involving several organs (e.g., heart, kidney). The essential role of the mitochondrial permeability transition pore in cell death coupled with the pro-survival function of SGK-1 raise the prospect that its therapeutic modulation could beneficially impact conditions associated with ischemia-reperfusion injury. SIGNIFICANCE STATEMENT: Since the discovery of serum glucocorticoid-regulated kinase (SGK)-1, extensive research has unraveled its role in cancer biology and, thus, its therapeutic targeting. Increasingly, it is also becoming clear that SGK-1 is a major determinant of the outcome of ischemia-reperfusion injury to various organs. Thus, evaluation of existing information should help identify gaps in our current knowledge and also determine whether and how its therapeutic modulation could impact the outcome of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Mahmood S Mozaffari
- Department of Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
16
|
Razali NN, Raja Ali RA, Muhammad Nawawi KN, Yahaya A, Mohd Rathi ND, Mokhtar NM. Roles of phosphatidylinositol-3-kinases signaling pathway in inflammation-related cancer: Impact of rs10889677 variant and buparlisib in colitis-associated cancer. World J Gastroenterol 2023; 29:5543-5556. [PMID: 37970476 PMCID: PMC10642440 DOI: 10.3748/wjg.v29.i40.5543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/05/2023] [Accepted: 10/11/2023] [Indexed: 10/27/2023] Open
Abstract
BACKGROUND Phosphatidylinositol-3-kinases (PI3K) is a well-known route in inflammation-related cancer. Recent discovery on PI3K-related genes revealed a potential variant that links ulcerative colitis (UC) and colorectal cancer (CRC) with colitis-associated cancer (CAC). PI3K/AKT pathway has been recommended as a potential additional therapeutic option for CRC due to its substantial role in modifying cellular processes. Buparlisib is a pan-class I PI3K inhibitor previously shown to reduce tumor growth. AIM To investigate the regulation of rs10889677 and the role of buparlisib in the PI3K signaling pathway in CAC pathogenesis. METHODS Genomic DNA from 32 colonic samples, including CAC (n = 7), UC (n = 10) and CRC (n = 15), was sequenced for the rs10889677 mutation. The mutant and wildtype fragments were amplified and cloned in the pmirGLO vector. The luciferase activity of cloned vectors was assessed after transfection into the HT29 cell line. CAC mice were induced by a mixture of a single azoxymethane injection and three cycles of dextran sulphate sodium, then buparlisib was administered after 14 d. The excised colon was subjected to immunohistochemistry for Ki67 and Cleaved-caspase-3 markers and quantitative real-time polymerase chain reaction analysis for Pdk1 and Sgk2. RESULTS Luciferase activity decreased by 2.07-fold in the rs10889677 mutant, confirming the hypothesis that the variant disrupted miRNA binding sites, which led to an increase in IL23R expression and the activation of the PI3K signaling pathway. Furthermore, CAC-induced mice had a significantly higher disease activity index (P < 0.05). Buparlisib treatment significantly decreased mean weight loss in CAC-induced mice (P < 0.05), reduced the percentage of proliferating cells by 5%, and increased the number of apoptotic cells. The treatment also caused a downward trend of Pdk1 expression and significantly decreased Sgk2 expression. CONCLUSION Our findings suggested that the rs10889677 variant as a critical initiator of the PI3K signaling pathway, and buparlisib had the ability to prevent PI3K-non-AKT activation in the pathophysiology of CAC.
Collapse
Affiliation(s)
- Nurul Nadirah Razali
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
| | - Raja Affendi Raja Ali
- School of Medical and Life Sciences, Sunway University, Sunway City 47500, Malaysia
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
| | - Khairul Najmi Muhammad Nawawi
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
- Gastroenterology Unit, Department of Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
| | - Azyani Yahaya
- Department of Pathology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
| | - Norshafila Diana Mohd Rathi
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
| | - Norfilza Mohd Mokhtar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
- GUT Research Group, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia
| |
Collapse
|
17
|
Anderson G. Melatonin, BAG-1 and cortisol circadian interactions in tumor pathogenesis and patterned immune responses. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2023; 4:962-993. [PMID: 37970210 PMCID: PMC10645470 DOI: 10.37349/etat.2023.00176] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 08/07/2023] [Indexed: 11/17/2023] Open
Abstract
A dysregulated circadian rhythm is significantly associated with cancer risk, as is aging. Both aging and circadian dysregulation show suppressed pineal melatonin, which is indicated in many studies to be linked to cancer risk and progression. Another independently investigated aspect of the circadian rhythm is the cortisol awakening response (CAR), which is linked to stress-associated hypothalamus-pituitary-adrenal (HPA) axis activation. CAR and HPA axis activity are primarily mediated via activation of the glucocorticoid receptor (GR), which drives patterned gene expression via binding to the promotors of glucocorticoid response element (GRE)-expressing genes. Recent data shows that the GR can be prevented from nuclear translocation by the B cell lymphoma-2 (Bcl-2)-associated athanogene 1 (BAG-1), which translocates the GR to mitochondria, where it can have diverse effects. Melatonin also suppresses GR nuclear translocation by maintaining the GR in a complex with heat shock protein 90 (Hsp90). Melatonin, directly and/or epigenetically, can upregulate BAG-1, suggesting that the dramatic 10-fold decrease in pineal melatonin from adolescence to the ninth decade of life will attenuate the capacity of night-time melatonin to modulate the effects of the early morning CAR. The interactions of pineal melatonin/BAG-1/Hsp90 with the CAR are proposed to underpin how aging and circadian dysregulation are associated with cancer risk. This may be mediated via differential effects of melatonin/BAG-1/Hsp90/GR in different cells of microenvironments across the body, from which tumors emerge. This provides a model of cancer pathogenesis that better integrates previously disparate bodies of data, including how immune cells are regulated by cancer cells in the tumor microenvironment, at least partly via the cancer cell regulation of the tryptophan-melatonin pathway. This has a number of future research and treatment implications.
Collapse
|
18
|
Mozaffari MS, Abdelsayed R, Emami S, Kavuri S. Expression profiles of glucocorticoid-inducible proteins in human papilloma virus-related oropharyngeal squamous cell carcinoma. FRONTIERS IN ORAL HEALTH 2023; 4:1285139. [PMID: 37954869 PMCID: PMC10634427 DOI: 10.3389/froh.2023.1285139] [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: 08/29/2023] [Accepted: 10/11/2023] [Indexed: 11/14/2023] Open
Abstract
Introduction Human papillomavirus virus-related oropharyngeal squamous cell carcinoma (HPV-OPSCC) comprises a significant portion of head and neck cancers. Several glucocorticoid-inducible proteins play important roles in pathogenesis of some cancers but their status and roles in HPV-OPSCC remain elusive; these include the glucocorticoid-induced leucine zipper (GILZ), Annexin-A1 and serum glucocorticoid-regulated kinase-1 (SGK-1). Methods We determined expression profiles of these proteins, using immunohistochemistry, in archived biopsy samples of patients diagnosed with HPV-OPSCC; samples of non-cancer oral lesions (e.g., hyperkeratosis) were used as controls. Results GILZ staining was primarily confined to nuclei of all tissues but, in HPV-OPSCC specimens, neoplastic cells exhibiting mitosis displayed prominent cytoplasmic GILZ expression. On the other hand, nuclear, cytoplasmic and membranous Annexin-A1 staining was observed in suprabasal cell layers of control specimens. A noted feature of the HPV-OPSCC specimens was few clusters of matured and differentiated nonbasaloid cells that showed prominent nuclear and cytoplasmic Annexin-A1 staining while the remainder of the tumor mass was devoid of staining. Cytoplasmic and nuclear staining for SGK-1 was prominent for control than PV-OPSCC specimens while staining for phosphorylated SGK-1 (pSGK-1; active) was prominent for cell membrane and cytoplasm of control specimens but HPV-OPSCC specimens showed mild and patchy nuclear and cytoplasmic staining. Semi-quantitative analysis of GILZ immunostaining indicated increased staining area but similar normalized staining for HPV-OPSCC compared to control specimens. By contrast, staining area and normalized staining were reduced for other proteins in HPV-OPSCC than control specimens. Discussion Our collective observations suggest differential cellular localization and expression of glucocorticoid-inducible proteins in HPV-OPSCC suggestive of different functional roles in pathogenesis of this condition.
Collapse
Affiliation(s)
- Mahmood S. Mozaffari
- Departmentof Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Rafik Abdelsayed
- Departmentof Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, GA, United States
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Sahar Emami
- Departmentof Oral Biology and Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, GA, United States
| | - Sravan Kavuri
- Department of Pathology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
19
|
Leroux AE, Biondi RM. The choreography of protein kinase PDK1 and its diverse substrate dance partners. Biochem J 2023; 480:1503-1532. [PMID: 37792325 DOI: 10.1042/bcj20220396] [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: 06/06/2023] [Revised: 08/24/2023] [Accepted: 08/31/2023] [Indexed: 10/05/2023]
Abstract
The protein kinase PDK1 phosphorylates at least 24 distinct substrates, all of which belong to the AGC protein kinase group. Some substrates, such as conventional PKCs, undergo phosphorylation by PDK1 during their synthesis and subsequently get activated by DAG and Calcium. On the other hand, other substrates, including members of the Akt/PKB, S6K, SGK, and RSK families, undergo phosphorylation and activation downstream of PI3-kinase signaling. This review presents two accepted molecular mechanisms that determine the precise and timely phosphorylation of different substrates by PDK1. The first mechanism involves the colocalization of PDK1 with Akt/PKB in the presence of PIP3. The second mechanism involves the regulated docking interaction between the hydrophobic motif (HM) of substrates and the PIF-pocket of PDK1. This interaction, in trans, is equivalent to the molecular mechanism that governs the activity of AGC kinases through their HMs intramolecularly. PDK1 has been instrumental in illustrating the bi-directional allosteric communication between the PIF-pocket and the ATP-binding site and the potential of the system for drug discovery. PDK1's interaction with substrates is not solely regulated by the substrates themselves. Recent research indicates that full-length PDK1 can adopt various conformations based on the positioning of the PH domain relative to the catalytic domain. These distinct conformations of full-length PDK1 can influence the interaction and phosphorylation of substrates. Finally, we critically discuss recent findings proposing that PIP3 can directly regulate the activity of PDK1, which contradicts extensive in vitro and in vivo studies conducted over the years.
Collapse
Affiliation(s)
- Alejandro E Leroux
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| | - Ricardo M Biondi
- Instituto de Investigación en Biomedicina de Buenos Aires (IBioBA) - CONICET - Partner Institute of the Max Planck Society, Buenos Aires C1425FQD, Argentina
| |
Collapse
|
20
|
Pal A, Karanwal S, Chera JS, Batra V, Kumaresan A, Sarwalia P, Datta TK, Kumar R. Circulatory extracellular vesicle derived miR-195-5p promotes cellular apoptosis and suppresses cell proliferation in the buffalo endometrial primary cell culture. Sci Rep 2023; 13:16703. [PMID: 37794118 PMCID: PMC10551009 DOI: 10.1038/s41598-023-43530-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023] Open
Abstract
In pregnant animals, communication between the mother and conceptus occurs via extracellular vesicles (EVs) that carry several biomolecules such as nucleic acids (miRNAs, mRNAs), proteins, and lipids. At the time of implantation, the endometrium undergoes several morphological and physiological changes, such as angiogenesis, apoptosis, and cell proliferation regulation at the implantation site, to attain a receptive state. This study was conducted to detect pregnancy-specific miRNAs derived from extracellular vesicles in the systemic circulation of Bubalus bubalis (water buffalo) and to assess their functional significance in the modulation of endometrial primary cells. The extracellular vesicles were isolated from the blood plasma using a precipitation-based method and further characterized by various methods such as Differential light scattering, Nanoparticle tracking assay, Western blot, and transmission electron microscopy. The relative expression of the selected extracellular vesicles associated miRNAs (EV-miRNA) at different intervals (days 15, 19, 25, and 30) post artificial insemination (AI) was analyzed using RT-qPCR, and expression of miR-195-5p was found to be significantly higher (P < 0.01) in pregnant animals on day 19 post AI (implantation window) as compared to day 15 post AI. The elevated expression might indicate the involvement of this miRNA in the maternal-conceptus cross-talk occurring during the implantation period. The KEGG pathway enrichment and Gene Ontology analyses of the miR-195-5p target genes revealed that these were mostly involved in the PI3-Akt, MAPK, cell cycle, ubiquitin-mediated proteolysis, and mTOR signaling pathways, which are related to the regulation of cell proliferation. Transfecting the in vitro cultured cells with miR-195-5p mimic significantly suppressed (P < 0.05) the expression of its target genes such as YWHAQ, CDC27, AKT-3, FGF-7, MAPK8, SGK1, VEGFA, CACAND1, CUL2, MKNK1, and CACAN2D1. Furthermore, the downregulation of the miR-195-5p target genes was positively correlated with a significant increase in the apoptotic rate and a decrease in the proliferation. In conclusion, the current findings provide vital information on the presence of EV miR-195-5p in maternal circulation during the implantation window indicating its important role in the modulation of buffalo endometrium epithelial cells via promoting cell death. Altogether, the milieu of miR-195-5p may serve as a novel and potential molecular factor facilitating the implantation of the early embryo during the establishment of pregnancy in buffaloes. Thus, miR-195-5p may be identified as a unique circulatory EV biomarker related to establishing pregnancy in buffaloes as early as day 19 post-AI.
Collapse
Affiliation(s)
- Ankit Pal
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Seema Karanwal
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Jatinder Singh Chera
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Vipul Batra
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Arumugam Kumaresan
- Theriogenelogy Laboratory, SRS of National Dairy Research Institute, Bengaluru, India
| | - Parul Sarwalia
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Tirtha K Datta
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India
| | - Rakesh Kumar
- Animal Genomics Laboratory, Animal Biotechnology Centre, National Dairy Research Institute, Karnal, India.
| |
Collapse
|
21
|
Zhou X, Wang S, Li Y, Zhao H, Han X, Yu Y, Chen Y, Yang Y, Ma X, Huo H, Zhang M, Zhao Y, Ma N. Monocarboxylate transporter 4 promotes the migration of non‑cancerous L929 fibroblast cells by activating the IGF1/IGF1R/PIK3R3/SGK1 axis. Oncol Lett 2023; 26:460. [PMID: 37745980 PMCID: PMC10512108 DOI: 10.3892/ol.2023.14047] [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: 04/03/2023] [Accepted: 07/21/2023] [Indexed: 09/26/2023] Open
Abstract
The tumor microenvironment (TME) and Warburg effect are critical for the regulation of tumor metastasis. The monocarboxylate transporter (MCT) family members, particularly MCT4, which is encoded by the solute carrier family 16 member 3 gene, play an important role in the regulation of the TME and mediation of the Warburg effect by transporting lactate out of cancer cells. Migration and invasion are two key features of metastasis. Few studies have investigated the mechanism by which MCT4 promotes cell migration, and the suggested mechanisms by which MCT4 promotes migration vary in different tumor cell models. The purpose of the present study was to use non-cancerous cells as a research model to investigate the specific mechanism underlying the promotion of migration by MCT4. In a previous study, murine L929 cells overexpressing human MCT4 (MCT4-L929 cells) were generated and MCT4 was demonstrated to promote the migration and invasion of these non-cancerous cells. In the present study, MCT4-L929 cells and control-L929 cells were used to investigate the potential pathways and mechanisms through which MCT4 promotes cell migration. RNA sequencing analysis revealed 872 differentially expressed genes, comprising 337 and 535 upregulated and downregulated genes, respectively, in the MCT4-L929 cells. Reverse transcription-quantitative analysis and western blotting revealed that MCT4 overexpression increased the transcription and protein levels of insulin-like growth factor 1 (IGF1). In a wound healing assay, the migration of exogenous mouse IGF1-treated control-L929 cells was similar to that of MCT4-L929 cells. Additionally, the inhibition of IGF1 receptor (IGF1R) or serum/glucocorticoid regulated kinase 1 (SGK1), a downstream protein in the IGF1 and phosphoinositide 3-kinase PI3K regulatory subunit 3 (PIK3R3) pathways, in MCT4-L929 cells mitigated the cell migration-promoting effect of MCT4. These novel findings suggest that MCT4 may promote the migration of L929 fibroblast cells via activation of the IGF1/IGF1R/PIK3R3/SGK1 axis.
Collapse
Affiliation(s)
- Xiaoju Zhou
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Shuo Wang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yanyan Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - He Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Xue Han
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yue Yu
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yu Chen
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yu Yang
- Department of Biochemistry and Molecular Biology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Xiaonan Ma
- Department of Biochemistry and Molecular Biology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Hongjing Huo
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Manting Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Yongshan Zhao
- Department of Biochemistry and Molecular Biology, School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| | - Ningning Ma
- Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P.R. China
| |
Collapse
|
22
|
Li YH, Sun CC, Chen PM, Chen HH. SGK1 Target Genes Involved in Heart and Blood Vessel Functions in PC12 Cells. Cells 2023; 12:1641. [PMID: 37371111 DOI: 10.3390/cells12121641] [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: 03/28/2023] [Revised: 06/07/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) is expressed in neuronal cells and involved in the pathogenesis of hypertension and metabolic syndrome, regulation of neuronal function, and depression in the brain. This study aims to identify the cellular mechanisms and signaling pathways of SGK1 in neuronal cells. In this study, the SGK1 inhibitor GSK650394 is used to suppress SGK1 expression in PC12 cells using an in vitro neuroscience research platform. Comparative transcriptomic analysis was performed to investigate the effects of SGK1 inhibition in nervous cells using mRNA sequencing (RNA-seq), differentially expressed genes (DEGs), and gene enrichment analysis. In total, 12,627 genes were identified, including 675 and 2152 DEGs at 48 and 72 h after treatment with GSK650394 in PC12 cells, respectively. Gene enrichment analysis data indicated that SGK1 inhibition-induced DEGs were enriched in 94 and 173 genes associated with vascular development and functional regulation and were validated using real-time PCR, Western blotting, and GEPIA2. Therefore, this study uses RNA-seq, DEG analysis, and GEPIA2 correlation analysis to identify positive candidate genes and signaling pathways regulated by SGK1 in rat nervous cells, which will enable further exploration of the underlying molecular signaling mechanisms of SGK1 and provide new insights into neuromodulation in cardiovascular diseases.
Collapse
Affiliation(s)
- Yu-He Li
- Department of Laboratory Medicine, Zuoying Branch of Kaohsiung Armed Forces General Hospital, Kaohsiung 813, Taiwan
| | - Chia-Cheng Sun
- Physical Examination Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Po-Ming Chen
- Research Assistant Center, Show Chwan Memorial Hospital, Changhua 500, Taiwan
| | - Hsin-Hung Chen
- Department of Medical Education and Research, Kaohsiung Veterans General Hospital, Kaohsiung 813, Taiwan
| |
Collapse
|
23
|
Al-Alem U, Rauscher GH, Alem QA, Kajdacsy-Balla A, Mahmoud AM. Prognostic Value of SGK1 and Bcl-2 in Invasive Breast Cancer. Cancers (Basel) 2023; 15:3151. [PMID: 37370761 DOI: 10.3390/cancers15123151] [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: 03/22/2023] [Revised: 06/06/2023] [Accepted: 06/09/2023] [Indexed: 06/29/2023] Open
Abstract
It is crucial to understand molecular alterations in breast cancer and how they relate to clinicopathologic factors. We have previously shown that the glucocorticoid receptor (GCR) protein expression was reduced in invasive breast carcinoma compared to normal breast tissue. Glucocorticoids, signaling through the GCR, regulate several cellular processes via downstream targets such as serum/glucocorticoid-regulated kinase 1 (SGK1) and B-cell lymphoma 2 (Bcl-2). We measured the expression of SGK1 and Bcl-2, in respective breast cancer tissue arrays, from a multiracial cohort of breast cancer patients. Higher cytoplasmic SGK1 staining was stronger in breast cancer tissue compared to normal tissue, especially in hormone receptor-negative cases. Conversely, the expression of cytoplasmic Bcl-2 was reduced in breast cancer compared to normal tissue, especially in hormone receptor-negative cases. Bcl-2 staining was associated with the self-reported racial/ethnic category, an earlier clinical stage, a lower histological grade, and a higher survival rate. Bcl-2 expression was associated with longer survival in models adjusted for age and race (HR = 0.32, 95% CI: 0.15, 0.65), and Bcl-2 expression remained strongly positively associated with protection from breast cancer death, with additional adjustments for ER/PR status (HR = 0.41, 95% CI: 0.2, 0.85). SGK1 and Bcl-2 may play biological roles in breast cancer development and/or progression.
Collapse
Affiliation(s)
- Umaima Al-Alem
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Garth H Rauscher
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Qais Al Alem
- Division of Epidemiology and Biostatistics, School of Public Health, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Andre Kajdacsy-Balla
- Department of Pathology, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Abeer M Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, The University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, The University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
24
|
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: 0] [Impact Index Per Article: 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
|
25
|
Jang H, Park Y, Jang J. Serum and glucocorticoid-regulated kinase 1: Structure, biological functions, and its inhibitors. Front Pharmacol 2022; 13:1036844. [PMID: 36457711 PMCID: PMC9706101 DOI: 10.3389/fphar.2022.1036844] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/20/2022] [Indexed: 08/11/2023] Open
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) is a serine/threonine kinase belonging to the protein kinase A, G, and C (AGC) family. Upon initiation of the phosphoinositide 3-kinase (PI3K) signaling pathway, mammalian target of rapamycin complex 2 (mTORC2) and phosphoinositide-dependent protein kinase 1 (PDK1) phosphorylate the hydrophobic motif and kinase domain of SGK1, respectively, inducing SGK1 activation. SGK1 modulates essential cellular processes such as proliferation, survival, and apoptosis. Hence, dysregulated SGK1 expression can result in multiple diseases, including hypertension, cancer, autoimmunity, and neurodegenerative disorders. This review provides a current understanding of SGK1, particularly in sodium transport, cancer progression, and autoimmunity. In addition, we summarize the developmental status of SGK1 inhibitors, their structures, and respective potencies evaluated in pre-clinical experimental settings. Collectively, this review highlights the significance of SGK1 and proposes SGK1 inhibitors as potential drugs for treatment of clinically relevant diseases.
Collapse
Affiliation(s)
- Hyunsoo Jang
- College of Pharmacy, Korea University, Sejong, South Korea
| | - Youngjun Park
- Laboratory of Immune and Inflammatory Disease, College of Pharmacy, Jeju Research Institute of Pharmaceutical Sciences, Jeju National University, Jeju, South Korea
- Interdisciplinary Graduate Program in Advanced Convergence Technology and Science, Jeju National University, Jeju, South Korea
| | - Jaebong Jang
- College of Pharmacy, Korea University, Sejong, South Korea
| |
Collapse
|
26
|
Peng TF, Zhou YJ, Zhou J, Zhou Y, Li XC, Ouyang Q. Long non-coding RNA VPS9D1-AS1 enhances proliferation, invasion, and epithelial-mesenchymal transition in endometrial cancer via miR-377-3p/SGK1. Kaohsiung J Med Sci 2022; 38:1048-1059. [PMID: 36245426 DOI: 10.1002/kjm2.12606] [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: 06/01/2022] [Revised: 08/03/2022] [Accepted: 08/18/2022] [Indexed: 11/10/2022] Open
Abstract
Endometrial cancer (EC) is a kind of gynecologic malignancy with a rising incidence rate. This study aimed to explore the role of VPS9D1 antisense RNA1 (VPS9D1-AS1) in EC. The expression of VPS9D1-AS1, microRNA (miR)-377-3p, and serum and glucocorticoid-regulated kinase 1 (SGK1) was detected by Quantitative Real-Time PCR (qRT-PCR). Cell proliferation, invasion and epithelial-mesenchymal transition (EMT) were determined by cell counting kit-8 (CCK-8), 5-Ethynyl-2'-Deoxyuridine (EdU) transwell, and western bolt. VPS9D1-AS1 was predicted to sponge miR-377-3p via Starbase, and verified by luciferase reporter, RNA binding protein immunoprecipitation (RIP), and RNA pull-down experiments. The clinical characteristics of VPS9D1-AS1, miR-377-3p, and SGK1 were analyzed. The role of VPS9D1-AS1 on EC tumorigenesis was assessed in xenografted nude mice. VPS9D1-AS1 was upregulated in EC cells and tissues. Interference of VPS9D1-AS1 inhibited growth, invasion, and EMT of EC cells. Mechanically, VPS9D1-AS1 was a molecular sponge of miR-377-3p, and overexpression of miR-377-3p reversed VPS9D1-AS1-induced EC cells proliferation, invasion, and EMT. Moreover, SGK1 was confirmed to bind with miR-377-3p. Furthermore, overexpression of SGK1 alleviated sh-VPS9D1-AS1-caused effects on EC cells. High level of VPS9D1-AS1 and SGK1, or low miR-377-3p expression predicted a poor prognosis. The expression of the three genes was correlated with lymph node metastasis, pathological stage, and International Federation of Gynecology and Obstetrics (FIGO) stage, but not associated with age, ER, and PR expression. Interestingly, knockdown of VPS9D1-AS1 suppressed EC tumor growth in mice. VPS9D1-AS1 promoted cell invasion, proliferation, and EMT via modulating miR-377-3p/SGK1 axis, which provided new options for therapeutic strategies of EC.
Collapse
Affiliation(s)
- Tian-Fang Peng
- Department of Gynecological Oncology, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Yan-Jie Zhou
- Department of Gynecological Oncology, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Jian Zhou
- Department of Gynecological Oncology, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Yi Zhou
- Department of Gynecological Oncology, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Xin-Chun Li
- Department of Gynecological Oncology, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| | - Qiang Ouyang
- Department of Gynecological Oncology, Hunan Provincial Tumor Hospital, The Affiliated Tumor Hospital of Xiangya Medical School of Central South University, Changsha, China
| |
Collapse
|
27
|
Xiu M, Li L, Li Y, Gao Y. An update regarding the role of WNK kinases in cancer. Cell Death Dis 2022; 13:795. [PMID: 36123332 PMCID: PMC9485243 DOI: 10.1038/s41419-022-05249-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 01/23/2023]
Abstract
Mammalian WNK kinases (WNKs) are serine/threonine kinases that contain four members, WNK1-4. They function to maintain ion homeostasis and regulate blood pressure in mammals. Recent studies have revealed that the dysregulation of WNKs contributes to tumor growth, metastasis, and angiogenesis through complex mechanisms, especially through phosphorylating kinase substrates SPS1-related proline/alanine-rich kinase (SPAK) and oxidative stress-responsive kinase 1 (OSR1). Here, we review and discuss the relationships between WNKs and several key factors/biological processes in cancer, including ion channels, cation chloride cotransporters, sodium bicarbonate cotransporters, signaling pathways, angiogenesis, autophagy, and non-coding RNAs. In addition, the potential drugs for targeting WNK-SPAK/OSR1 signaling have also been discussed. This review summarizes and discusses knowledge of the roles of WNKs in cancer, which provides a comprehensive reference for future studies.
Collapse
Affiliation(s)
- Mengxi Xiu
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Li Li
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Yandong Li
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| | - Yong Gao
- grid.24516.340000000123704535Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, 200120 Shanghai, China
| |
Collapse
|
28
|
Díaz ECG, Lee AG, Sayles LC, Feria C, Sweet-Cordero EA, Yang F. A 3D Osteosarcoma Model with Bone-Mimicking Cues Reveals a Critical Role of Bone Mineral and Informs Drug Discovery. Adv Healthc Mater 2022; 11:e2200768. [PMID: 35767377 PMCID: PMC10162498 DOI: 10.1002/adhm.202200768] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/10/2022] [Indexed: 01/27/2023]
Abstract
Osteosarcoma (OS) is an aggressive bone cancer for which survival has not improved over three decades. While biomaterials have been widely used to engineer 3D soft-tissue tumor models, the potential of engineering 3D biomaterials-based OS models for comprehensive interrogation of OS pathology and drug discovery remains untapped. Bone is characterized by high mineral content, yet the role of bone mineral in OS progression and drug response remains unknown. Here, a microribbon-based OS model with bone-mimicking compositions is developed to elucidate the role of 3D culture and hydroxyapatite in OS signaling and drug response. The results reveal that hydroxyapatite in 3D is critical to support retention of OS signaling and drug resistance similar to patient tissues and mouse orthotopic tumors. The physiological relevance of this 3D model is validated using four established OS cell lines, seven patient-derived xenograft (PDX) cell lines and two animal models. Integrating 3D OS PDX models with RNA-sequencing identified 3D-specific druggable target, which predicts drug response in mouse orthotopic model. These results establish microribbon-based 3D OS models as a novel experimental tool to enable discovery of novel therapeutics that would be otherwise missed with 2D model and may serve as platforms to study patient-specific OS heterogeneity and drug resistance mechanisms.
Collapse
Affiliation(s)
| | - Alex G. Lee
- Division of Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California, 94143, USA
| | - Leanne C. Sayles
- Division of Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California, 94143, USA
| | - Criselle Feria
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - E. Alejandro Sweet-Cordero
- Division of Oncology, Department of Pediatrics, University of California San Francisco, San Francisco, California, 94143, USA
| | - Fan Yang
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Orthopaedic Surgery, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
29
|
Wu Y, Wang H, Li Y, Li Y, Liang Y, Zhong G, Zhang Q. Estrogen-increased SGK1 Promotes Endometrial Stromal Cell Invasion in Adenomyosis by Regulating with LPAR2. Reprod Sci 2022; 29:3026-3038. [PMID: 35799024 DOI: 10.1007/s43032-022-00990-3] [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: 04/21/2022] [Accepted: 05/24/2022] [Indexed: 12/01/2022]
Abstract
Adenomyosis is an estrogen-dependent gynecological disorder. The abnormal migration and invasion of the eutopic endometrium is thought to be the primary role in the pathogenesis of adenomyosis. However, the exact underlying mechanism remains unclear. This study investigated involvement of serum and glucocorticoid-regulated kinase 1 (SGK1) in the pathogenesis of adenomyosis. The SGK1 expression level was higher in the eutopic endometrium of adenomyosis. Upregulation of SGK1 can promote the migration, invasion of human stromal endometrial cells (HESC). Through RNA sequencing and other technical methods, we found that SGK1 regulates the expression of the important downstream molecule Lysophosphatidic acid receptor 2 (LPAR2), and ultimately regulates the expression level of functional proteins such as matrix metalloproteinase 2 and matrix metalloproteinase 9, which are related to migration and invasion. Then, we found that 17β-estradiol (E2) upregulated the expression of SGK1 in endometrial cells in a dose-dependent manner. Furthermore, SGK1 shRNA significantly suppressed the migration and invasion induced by E2 in endometrial cells, as well as the related factors. Our study revealed the possible role of SGK1 in the migration and invasion in the development of adenomyosis.
Collapse
Affiliation(s)
- Yingchen Wu
- Department of Gynecology and Obstetrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Hao Wang
- Department of Gynecology and Obstetrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yi Li
- Department of Gynecology and Obstetrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yangzhi Li
- Department of Gynecology and Obstetrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yihua Liang
- Department of Gynecology and Obstetrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Guangzheng Zhong
- Department of Urology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Qingxue Zhang
- Department of Gynecology and Obstetrics, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
30
|
Kong P, Zhang L, Zhang Z, Feng K, Sang Y, Duan X, Liu C, Sun T, Tao Z, Liu W. Emerging Proteins in CRPC: Functional Roles and Clinical Implications. Front Oncol 2022; 12:873876. [PMID: 35756667 PMCID: PMC9226405 DOI: 10.3389/fonc.2022.873876] [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: 02/11/2022] [Accepted: 03/30/2022] [Indexed: 11/13/2022] Open
Abstract
Prostate cancer (PCa) is the most common cancer in men in the western world, but the lack of specific and sensitive markers often leads to overtreatment of prostate cancer which eventually develops into castration-resistant prostate cancer (CRPC). Novel protein markers for diagnosis and management of CRPC will be promising. In this review, we systematically summarize and discuss the expression pattern of emerging proteins in tissue, cell lines, and serum when castration-sensitive prostate cancer (CSPC) progresses to CRPC; focus on the proteins involved in CRPC growth, invasion, metastasis, metabolism, and immune microenvironment; summarize the current understanding of the regulatory mechanisms of emerging proteins in CSPC progressed to CRPC at the molecular level; and finally summarize the clinical applications of emerging proteins as diagnostic marker, prognostic marker, predictive marker, and therapeutic marker.
Collapse
Affiliation(s)
- Piaoping Kong
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Lingyu Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhengliang Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Kangle Feng
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Yiwen Sang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuzhi Duan
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chunhua Liu
- Department of Blood Transfusion, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Tao Sun
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhihua Tao
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Weiwei Liu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
31
|
Song D, Zhou Z, Zhang D, Wu J, Hao Q, Zhao L, Ren H, Zhang B. Identification of an Endoplasmic Reticulum Stress-Related Gene Signature to Evaluate the Immune Status and Predict the Prognosis of Hepatocellular Carcinoma. Front Genet 2022; 13:850200. [PMID: 35711939 PMCID: PMC9197218 DOI: 10.3389/fgene.2022.850200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/15/2022] [Indexed: 12/21/2022] Open
Abstract
Liver cancer is the sixth most frequently diagnosed primary malignancy and ranks as the third leading cause of cancer-related death worldwide in 2020. ER stress also plays a vital role in the pathogenesis of malignancies. In the current study, we aimed to construct an endoplasmic reticulum stress-related genes (ERGs) signature to predict the overall survival (OS) of patients with HCC. Differentially expressed ERGs (DE-ERGs) were analyzed using The Cancer Genome Atlas (TCGA-LIHC cohort) and International Cancer Genome Consortium (ICGC-LIRI-JP cohort) databases. The prognostic gene signature was identified by the univariate Cox regression and Least Absolute Shrinkage and Selection Operator (LASSO)-penalized Cox proportional hazards regression analysis. The predictive ability of the model was evaluated by utilizing Kaplan-Meier curves and time-dependent receiver operating characteristic (ROC) curves. Gene set variant analysis (GSVA) was performed to explore the underlying biological processes and signaling pathways. CIBERPORT and single-sample Gene Set Enrichment Analysis (ssGSEA) were implemented to estimate the immune status between the different risk groups. A total of 113 DE-ERGs were identified between 50 normal samples and 365 HCC samples in the TCGA-LIHC cohort, and 48 DE-ERGs were associated with OS through the univariate Cox regression. A six DE-ERGs (PPARGC1A, SQSTM1, SGK1, PON1, CDK1, and G6PD) signature was constructed and classified patients into high-risk and low-risk groups. The risk score was an independent prognostic indicator for OS (HR > 1, p < 0.001). The function enrichment analysis indicated that cell cycle, RNA degradation, protein localization, and cell division were the main biological processes. The high-risk group had higher immune cell infiltration levels than those of the low-risk group. We predicted the response to targeted therapy in high- and low-risk patients with HCC and found that the high-risk patients were more sensitive to pazopanib. At last, we verified the expression of the six gene patterns in HCC tissues by qRT-PCR and immunohistochemistry. This signature may be a potential tool to provide a choice for prognosis prediction and personal management of patients with HCC.
Collapse
Affiliation(s)
- Dingli Song
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Zhenyu Zhou
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dai Zhang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, the Air Force Medical University, Xi'an, China
| | - Jie Wu
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qian Hao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lili Zhao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hong Ren
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Boxiang Zhang
- Department of Thoracic Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
32
|
Cicenas J, Meskinyte-Kausiliene E, Jukna V, Rimkus A, Simkus J, Soderholm D. SGK1 in Cancer: Biomarker and Drug Target. Cancers (Basel) 2022; 14:2385. [PMID: 35625991 PMCID: PMC9139822 DOI: 10.3390/cancers14102385] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/06/2022] [Accepted: 05/09/2022] [Indexed: 12/29/2022] Open
Abstract
Serum- and glucocorticoid-regulated kinases (SGKs) are members of the AGC family of serine/threonine kinases, consisting of three isoforms: SGK1, SGK2, and SGK3. SGK1 was initially cloned as a gene transcriptionally stimulated by serum and glucocorticoids in rat mammary tumor cells. It is upregulated in some cancers and downregulated in others. SGK1 increases tumor cell survival, adhesiveness, invasiveness, motility, and epithelial to mesenchymal transition. It stimulates tumor growth by mechanisms such as activation of K+ channels and Ca2+ channels, Na+/H+ exchanger, amino acid and glucose transporters, downregulation of Foxo3a and p53, and upregulation of β-catenin and NFκB. This chapter focuses on major aspects of SGK1 involvement in cancer, its use as biomarker as well as potential therapeutic target.
Collapse
Affiliation(s)
- Jonas Cicenas
- Proteomics Centre, Institute of Biochemistry, Vilnius University, Sauletekio al. 7, LT-10257 Vilnius, Lithuania
- MAP Kinase Resource, Bioinformatics, Melchiorstrasse 9, CH-3027 Bern, Switzerland; (A.R.); (J.S.)
- Center of Animal Husbandry Selections, Breeding Values and Dissemination, Agriculture Academy, Vytautas Magnus University, Studentų g. 11, LT-53361 Akademija, Lithuania; (E.M.-K.); (V.J.)
| | - Edita Meskinyte-Kausiliene
- Center of Animal Husbandry Selections, Breeding Values and Dissemination, Agriculture Academy, Vytautas Magnus University, Studentų g. 11, LT-53361 Akademija, Lithuania; (E.M.-K.); (V.J.)
| | - Vigilijus Jukna
- Center of Animal Husbandry Selections, Breeding Values and Dissemination, Agriculture Academy, Vytautas Magnus University, Studentų g. 11, LT-53361 Akademija, Lithuania; (E.M.-K.); (V.J.)
| | - Arnas Rimkus
- MAP Kinase Resource, Bioinformatics, Melchiorstrasse 9, CH-3027 Bern, Switzerland; (A.R.); (J.S.)
| | - Jokubas Simkus
- MAP Kinase Resource, Bioinformatics, Melchiorstrasse 9, CH-3027 Bern, Switzerland; (A.R.); (J.S.)
| | - Diana Soderholm
- Walker Art Center, 752 Vineland PI, Mineapolis, MN 55403, USA;
| |
Collapse
|
33
|
Ghani MJ, Gu W, Chen Z, Canessa CM. Lipid droplets and autophagosomes together with chaperones fine-tune expression of SGK1. J Cell Mol Med 2022; 26:2852-2865. [PMID: 35393773 PMCID: PMC9097849 DOI: 10.1111/jcmm.17300] [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: 08/23/2021] [Revised: 03/05/2022] [Accepted: 03/10/2022] [Indexed: 11/29/2022] Open
Abstract
Serum-glucocorticoid-induced kinase-1 (SGK1) regulates ion homeostasis and promotes survival under stress conditions. The expression of SGK1 is under transcriptional and post-translational regulations that are frequently altered in cancer and immune disorders. We report that an N-terminal amphipathic alpha-helix determines SGK1 expression levels through two distinct mechanisms. It tethers SGK1 to intracellular organelles generating a large pool of membrane-bound SGK1, which is differentially stabilized in lipid droplets (LD) in fed conditions or degraded in the endoplasmic reticulum by ER-phagy in starvation. Association of the α-helix to organelles does not depend on dedicated receptors or special phospholipids rather, it is intrinsic to its physicochemical properties and depends on the presence of bulky hydrophobic residues for attachment to LDs. The second mechanism is recruitment of protein-chaperones that recognize the α-helix as an unfolded protein promoting survival of the cytosolic SGK1 fraction. Together, the findings unveil an unexpected link between levels of energy storage and abundance of SGK1 and how changes in calorie intake could be used to modulate SGK1 expression, whereas the inhibition of molecular chaperones could serve as an additional enhancer in the treatment of malignancies and autoimmune disorders with high levels of SGK1 expression.
Collapse
Affiliation(s)
| | - Wenxue Gu
- School of Medicine, Tsinghua University, Beijing, China
| | - Zhuyuan Chen
- School of Medicine, Tsinghua University, Beijing, China
| | - Cecilia M Canessa
- School of Medicine, Tsinghua University, Beijing, China.,Yale School of Medicine, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
34
|
Mafi S, Mansoori B, Taeb S, Sadeghi H, Abbasi R, Cho WC, Rostamzadeh D. mTOR-Mediated Regulation of Immune Responses in Cancer and Tumor Microenvironment. Front Immunol 2022; 12:774103. [PMID: 35250965 PMCID: PMC8894239 DOI: 10.3389/fimmu.2021.774103] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 12/14/2021] [Indexed: 12/17/2022] Open
Abstract
The mechanistic/mammalian target of rapamycin (mTOR) is a downstream mediator in the phosphatidylinositol 3-kinase (PI3K)/Akt signaling pathways, which plays a pivotal role in regulating numerous cellular functions including cell growth, proliferation, survival, and metabolism by integrating a variety of extracellular and intracellular signals in the tumor microenvironment (TME). Dysregulation of the mTOR pathway is frequently reported in many types of human tumors, and targeting the PI3K/Akt/mTOR signaling pathway has been considered an attractive potential therapeutic target in cancer. The PI3K/Akt/mTOR signaling transduction pathway is important not only in the development and progression of cancers but also for its critical regulatory role in the tumor microenvironment. Immunologically, mTOR is emerging as a key regulator of immune responses. The mTOR signaling pathway plays an essential regulatory role in the differentiation and function of both innate and adaptive immune cells. Considering the central role of mTOR in metabolic and translational reprogramming, it can affect tumor-associated immune cells to undergo phenotypic and functional reprogramming in TME. The mTOR-mediated inflammatory response can also promote the recruitment of immune cells to TME, resulting in exerting the anti-tumor functions or promoting cancer cell growth, progression, and metastasis. Thus, deregulated mTOR signaling in cancer can modulate the TME, thereby affecting the tumor immune microenvironment. Here, we review the current knowledge regarding the crucial role of the PI3K/Akt/mTOR pathway in controlling and shaping the immune responses in TME.
Collapse
Affiliation(s)
- Sahar Mafi
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Behzad Mansoori
- The Wistar Institute, Molecular & Cellular Oncogenesis Program, Philadelphia, PA, United States
| | - Shahram Taeb
- Department of Radiology, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
- Medical Biotechnology Research Center, School of Paramedical Sciences, Guilan University of Medical Sciences, Rasht, Iran
| | - Hossein Sadeghi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - Reza Abbasi
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
| | - William C. Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Hong Kong, Hong Kong SAR, China
- *Correspondence: Davoud Rostamzadeh, ; ; William C. Cho, ;
| | - Davoud Rostamzadeh
- Department of Clinical Biochemistry, Yasuj University of Medical Sciences, Yasuj, Iran
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
- *Correspondence: Davoud Rostamzadeh, ; ; William C. Cho, ;
| |
Collapse
|
35
|
Wittmann C, Bacher F, Enyedy EA, Dömötör O, Spengler G, Madejski C, Reynisson J, Arion VB. Highly Antiproliferative Latonduine and Indolo[2,3- c]quinoline Derivatives: Complex Formation with Copper(II) Markedly Changes the Kinase Inhibitory Profile. J Med Chem 2022; 65:2238-2261. [PMID: 35104137 PMCID: PMC8842277 DOI: 10.1021/acs.jmedchem.1c01740] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
![]()
A series of latonduine
and indoloquinoline derivatives HL1–HL8 and their copper(II)
complexes (1–8) were synthesized and comprehensively
characterized. The structures of five compounds (HL6, [CuCl(L1)(DMF)]·DMF, [CuCl(L2)(CH3OH)], [CuCl(L3)]·0.5H2O, and [CuCl2(H2L5)]Cl·2DMF) were elucidated
by single crystal X-ray diffraction. The copper(II) complexes revealed
low micro- to sub-micromolar IC50 values with promising
selectivity toward human colon adenocarcinoma multidrug-resistant
Colo320 cancer cells as compared to the doxorubicin-sensitive Colo205
cell line. The lead compounds HL4 and 4 as well as HL8 and 8 induced apoptosis efficiently in Colo320 cells. In addition, the
copper(II) complexes had higher affinity to DNA than their metal-free
ligands. HL8 showed selective inhibition for
the PIM-1 enzyme, while 8 revealed strong inhibition
of five other enzymes, i.e., SGK-1, PKA, CaMK-1, GSK3β, and
MSK1, from a panel of 50 kinases. Furthermore, molecular modeling
of the ligands and complexes showed a good fit to the binding pockets
of these targets.
Collapse
Affiliation(s)
- Christopher Wittmann
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse, 42, Vienna A1090, Austria
| | - Felix Bacher
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse, 42, Vienna A1090, Austria
| | - Eva A Enyedy
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary.,MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Orsolya Dömötör
- Department of Inorganic and Analytical Chemistry, Interdisciplinary Excellence Centre, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary.,MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary
| | - Gabriella Spengler
- MTA-SZTE Lendület Functional Metal Complexes Research Group, University of Szeged, Dóm tér 7, Szeged H-6720, Hungary.,Department of Medical Microbiology, Albert Szent-Györgyi Health Center and Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, Szeged H-6725, Hungary
| | - Christian Madejski
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse, 42, Vienna A1090, Austria
| | - Jóhannes Reynisson
- School of Pharmacy and Bioengineering, Keele University, Hornbeam Building, Staffordshire ST5 5BG, United Kingdom
| | - Vladimir B Arion
- Institute of Inorganic Chemistry of the University of Vienna, Währinger Strasse, 42, Vienna A1090, Austria
| |
Collapse
|
36
|
Halland N, Schmidt F, Weiss T, Li Z, Czech J, Saas J, Ding-Pfennigdorff D, Dreyer MK, Strübing C, Nazare M. Rational Design of Highly Potent, Selective, and Bioavailable SGK1 Protein Kinase Inhibitors for the Treatment of Osteoarthritis. J Med Chem 2021; 65:1567-1584. [PMID: 34931844 DOI: 10.1021/acs.jmedchem.1c01601] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The serine/threonine kinase SGK1 is an activator of the β-catenin pathway and a powerful stimulator of cartilage degradation that is found to be upregulated under genomic control in diseased osteoarthritic cartilage. Today, no oral disease-modifying treatments are available and chronic treatment in this indication sets high requirements for the drug selectivity, pharmacokinetic, and safety profile. We describe the identification of a highly selective druglike 1H-pyrazolo[3,4-d]pyrimidine SGK1 inhibitor 17a that matches both safety and pharmacokinetic requirements for oral dosing. Rational compound design was facilitated by a novel hSGK1 co-crystal structure, and multiple ligand-based computer models were applied to guide the chemical optimization of the compound ADMET and selectivity profiles. Compounds were selected for subchronic proof of mechanism studies in the mouse femoral head cartilage explant model, and compound 17a emerged as a druglike SGK1 inhibitor, with a highly optimized profile suitable for oral dosing as a novel, potentially disease-modifying agent for osteoarthritis.
Collapse
Affiliation(s)
- Nis Halland
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Friedemann Schmidt
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Tilo Weiss
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Ziyu Li
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Jörg Czech
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Joachim Saas
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | | | - Matthias K Dreyer
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Carsten Strübing
- Integrated Drug Discovery, Sanofi R&D, Industriepark Höchst, D-65926 Frankfurt am Main, Germany
| | - Marc Nazare
- Leibniz-Institut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin-Buch, Germany
| |
Collapse
|
37
|
AlAjmi MF, Khan S, Choudhury A, Mohammad T, Noor S, Hussain A, Lu W, Eapen MS, Chimankar V, Hansbro PM, Sohal SS, Elasbali AM, Hassan MI. Impact of Deleterious Mutations on Structure, Function and Stability of Serum/Glucocorticoid Regulated Kinase 1: A Gene to Diseases Correlation. Front Mol Biosci 2021; 8:780284. [PMID: 34805284 PMCID: PMC8597711 DOI: 10.3389/fmolb.2021.780284] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Accepted: 10/19/2021] [Indexed: 11/22/2022] Open
Abstract
Serum and glucocorticoid-regulated kinase 1 (SGK1) is a Ser/Thr protein kinase involved in regulating cell survival, growth, proliferation, and migration. Its elevated expression and dysfunction are reported in breast, prostate, hepatocellular, lung adenoma, and renal carcinomas. We have analyzed the SGK1 mutations to explore their impact at the sequence and structure level by utilizing state-of-the-art computational approaches. Several pathogenic and destabilizing mutations were identified based on their impact on SGK1 and analyzed in detail. Three amino acid substitutions, K127M, T256A, and Y298A, in the kinase domain of SGK1 were identified and incorporated structurally into original coordinates of SGK1 to explore their time evolution impact using all-atom molecular dynamic (MD) simulations for 200 ns. MD results indicate substantial conformational alterations in SGK1, thus its functional loss, particularly upon T256A mutation. This study provides meaningful insights into SGK1 dysfunction upon mutation, leading to disease progression, including cancer, and neurodegeneration.
Collapse
Affiliation(s)
- Mohamed F. AlAjmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Shama Khan
- Drug Discovery and Development Centre (H3D), University of Cape Town, Cape Town, South Africa
| | - Arunabh Choudhury
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Saba Noor
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| | - Afzal Hussain
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Wenying Lu
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Mathew Suji Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Vrushali Chimankar
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, NSW, Australia
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs and Hunter Medical Research Institute, The University of Newcastle, Newcastle, NSW, Australia
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, TAS, Australia
| | - Abdelbaset Mohamed Elasbali
- Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Sakakah, Saudi Arabia
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, India
| |
Collapse
|
38
|
Abstract
BACKGROUND The serum and glucocorticoid-induced kinase-1 (SGK1) belonging to the AGC protein kinase family phosphorylates serine and threonine residues of target proteins. It regulates numerous ion channels and transporters and promotes survival under cellular stress. Unique to SGK1 is a tight control at transcriptional and post-transcriptional levels. SGK1 regulates multiple signal transduction pathways related to tumor development. Several studies have reported that SGK1 is upregulated in different types of human malignancies and induces resistance against inhibitors, drugs, and targeted therapies. RESULTS AND CONCLUSION This review highlights the cellular functions of SGK1, its crucial role in cancer development, and clinical insights for SGK1 targeted therapies. Furthermore, the role of SGK1-mediated autophagy as a potential therapeutic target for cancer has been discussed.
Collapse
|
39
|
Hyperstable SGK1 steps out of AKT's shadow. Blood 2021; 138:917-918. [PMID: 34529017 DOI: 10.1182/blood.2021012518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 05/17/2021] [Indexed: 11/20/2022] Open
|
40
|
Rellick SL, Hu G, Piktel D, Martin KH, Geldenhuys WJ, Nair RR, Gibson LF. Co-culture model of B-cell acute lymphoblastic leukemia recapitulates a transcription signature of chemotherapy-refractory minimal residual disease. Sci Rep 2021; 11:15840. [PMID: 34349149 PMCID: PMC8339057 DOI: 10.1038/s41598-021-95039-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 07/20/2021] [Indexed: 12/26/2022] Open
Abstract
B-cell acute lymphoblastic leukemia (ALL) is characterized by accumulation of immature hematopoietic cells in the bone marrow, a well-established sanctuary site for leukemic cell survival during treatment. While standard of care treatment results in remission in most patients, a small population of patients will relapse, due to the presence of minimal residual disease (MRD) consisting of dormant, chemotherapy-resistant tumor cells. To interrogate this clinically relevant population of treatment refractory cells, we developed an in vitro cell model in which human ALL cells are grown in co-culture with human derived bone marrow stromal cells or osteoblasts. Within this co-culture, tumor cells are found in suspension, lightly attached to the top of the adherent cells, or buried under the adherent cells in a population that is phase dim (PD) by light microscopy. PD cells are dormant and chemotherapy-resistant, consistent with the population of cells that underlies MRD. In the current study, we characterized the transcriptional signature of PD cells by RNA-Seq, and these data were compared to a published expression data set derived from human MRD B-cell ALL patients. Our comparative analyses revealed that the PD cell population is markedly similar to the MRD expression patterns from the primary cells isolated from patients. We further identified genes and key signaling pathways that are common between the PD tumor cells from co-culture and patient derived MRD cells as potential therapeutic targets for future studies.
Collapse
Affiliation(s)
- Stephanie L Rellick
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506, USA
- West Virginia University Cancer Institute, Morgantown, WV, 26506, USA
| | - Gangqing Hu
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506, USA
- Bioinformatics Core, West Virginia University, Morgantown, WV, 26506, USA
- West Virginia Clinical and Translational Science Institute, Morgantown, WV, 26506, USA
| | - Debra Piktel
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506, USA
- West Virginia University Cancer Institute, Morgantown, WV, 26506, USA
| | - Karen H Martin
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506, USA
- West Virginia University Cancer Institute, Morgantown, WV, 26506, USA
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, 26506, USA
| | - Rajesh R Nair
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506, USA
- West Virginia University Cancer Institute, Morgantown, WV, 26506, USA
| | - Laura F Gibson
- Department of Microbiology, Immunology and Cell Biology, West Virginia University School of Medicine, 1 Medical Center Drive, Morgantown, WV, 26506, USA.
- West Virginia University Cancer Institute, Morgantown, WV, 26506, USA.
| |
Collapse
|
41
|
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: 2.0] [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
|