1
|
Wamaitha SE, Rojas EJ, Monticolo F, Hsu FM, Sosa E, Mackie AM, Oyama K, Custer M, Murphy M, Laird DJ, Shu J, Hennebold JD, Clark AT. Defining the cell and molecular origins of the primate ovarian reserve. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.21.634052. [PMID: 39896577 PMCID: PMC11785033 DOI: 10.1101/2025.01.21.634052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The primate ovarian reserve is established during late fetal development and consists of quiescent primordial follicles in the ovarian cortex, each composed of granulosa cells surrounding an oocyte in dictate. As late stages of fetal development are not routinely accessible for study with human tissue, we exploited the evolutionary proximity of the rhesus macaque to investigate primate follicle formation. Similar to human prenatal ovaries, the rhesus also develops multiple types of pre-granulosa (PG) cells, with the majority of primordial follicles derived from PG2 with small variable contributions from PG1. We observed that activated medullary follicles recruit fetal theca cells to establish a two-cell system for sex-steroid hormone production prior to birth, providing a cell-based explanation for mini puberty.
Collapse
Affiliation(s)
- Sissy E. Wamaitha
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles; Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles; Los Angeles, CA 90095, USA
- Molecular Biology Institute; University of California Los Angeles; Los Angeles, CA 90095, USA
| | - Ernesto J. Rojas
- Department of Obstetrics, Gynecology and Reproductive Science, Center for Reproductive Sciences, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research; University of California, San Francisco; San Francisco, CA 94143, United States
| | - Francesco Monticolo
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School; Boston, MA 02114, USA
| | - Fei-man Hsu
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles; Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles; Los Angeles, CA 90095, USA
- Molecular Biology Institute; University of California Los Angeles; Los Angeles, CA 90095, USA
| | - Enrique Sosa
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles; Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles; Los Angeles, CA 90095, USA
- Molecular Biology Institute; University of California Los Angeles; Los Angeles, CA 90095, USA
| | - Amanda M. Mackie
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles; Los Angeles, CA 90095, USA
| | - Kiana Oyama
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center; Beaverton, OR 97006, USA
| | - Maggie Custer
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center; Beaverton, OR 97006, USA
| | - Melinda Murphy
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center; Beaverton, OR 97006, USA
| | - Diana J. Laird
- Department of Obstetrics, Gynecology and Reproductive Science, Center for Reproductive Sciences, Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research; University of California, San Francisco; San Francisco, CA 94143, United States
| | - Jian Shu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School; Boston, MA 02114, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard; Cambridge, MA 02142, USA
| | - Jon D. Hennebold
- Division of Reproductive & Developmental Sciences, Oregon National Primate Research Center; Beaverton, OR 97006, USA
- Department of Obstetrics and Gynecology, Oregon Health & Science University; Portland, OR 97239, USA
| | - Amander T. Clark
- Department of Molecular, Cell and Developmental Biology, University of California Los Angeles; Los Angeles, CA 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles; Los Angeles, CA 90095, USA
- Molecular Biology Institute; University of California Los Angeles; Los Angeles, CA 90095, USA
| |
Collapse
|
2
|
Filipovich E, Gorodkova E, Shcherbakova A, Asaad W, Popov S, Melnichenko G, Mokrysheva N, Utkina M. The role of cell cycle-related genes in the tumorigenesis of adrenal and thyroid neuroendocrine tumors. Heliyon 2025; 11:e41457. [PMID: 39834406 PMCID: PMC11742855 DOI: 10.1016/j.heliyon.2024.e41457] [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: 09/25/2024] [Revised: 12/16/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025] Open
Abstract
The molecular mechanisms underlying adrenal and thyroid neuroendocrine tumors, including their tumorigenesis, progression, and metastasis, involve unique pathways regulating cell cycle progression. To better understand these mechanisms and pathways, extensive in-depth research on cell cycle-related genes is necessary. This review aims to describe and interpret current single-cell RNA sequencing studies on neuroblastoma, medullary thyroid cancer, and pheochromocytoma tumors. Our review summarizes differentially expressed cell cycle-related genes with distinct functions, highlighting their potential as therapeutic targets and components of panels used to determine tumor type or aggressiveness. Although some insights have been gained, there is still limited information on these topics, and further research is required to explore the regulatory mechanisms of these tumors.
Collapse
Affiliation(s)
- Ekaterina Filipovich
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Ekaterina Gorodkova
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Anastasia Shcherbakova
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Walaa Asaad
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Sergey Popov
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Galina Melnichenko
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Natalya Mokrysheva
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| | - Marina Utkina
- Laboratory of General, Molecular and Population Genetics, Endocrinology Research Center, Moscow, 117292, Russia
| |
Collapse
|
3
|
Shen H, Yuan J, Tong D, Chen B, Yu E, Chen G, Peng C, Chang W, E J, Cao F. Regulator of G protein signaling 16 restrains apoptosis in colorectal cancer through disrupting TRAF6-TAB2-TAK1-JNK/p38 MAPK signaling. Cell Death Dis 2024; 15:438. [PMID: 38906869 PMCID: PMC11192724 DOI: 10.1038/s41419-024-06803-6] [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: 02/17/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/23/2024]
Abstract
Colorectal cancer (CRC) remains a major global cause of cancer-related mortality, lacking effective biomarkers and therapeutic targets. Revealing the critical pathogenic factors of CRC and the underlying mechanisms would offer potential therapeutic strategies for clinical application. G protein signaling (RGS) protein family modulators play essential role within regulating downstream signaling of GPCR receptors, with function in cancers unclear. Our study focused on the expression patterns of RGS proteins in CRC, identifying Regulator of G protein signaling 16 (RGS16) as a prospective diagnostic and therapeutic target. Analyzing 899 CRC tissues revealed elevated RGS16 levels, correlating with clinicopathological features and CRC prognosis by immunohistochemistry (IHC) combined with microarray. We confirmed the elevated RGS16 protein level in CRC, and found that patients with RGS16-high tumors exhibited decreased disease-specific survival (DSS) and disease-free survival (DFS) compared to those with low RGS16 expression. Functional assays demonstrated that RGS16 promoted the CRC progression, knockdown of RGS16 led to significantly increased apoptosis rates of CRC in vitro and in vivo. Notably, we also confirmed these phenotypes of RGS16 in organoids originated from resected primary human CRC tissues. Mechanistically, RGS16 restrained JNK/P38-mediated apoptosis in CRC cells through disrupting the recruitment of TAB2/TAK1 to TRAF6. This study provides insights into addressing the challenges posed by CRC, offering avenues for clinical translation.
Collapse
Affiliation(s)
- Hao Shen
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Environmental and Occupational Health, Naval Medical University, Shanghai, China
| | - Jie Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Dafeng Tong
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Bingchen Chen
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Enda Yu
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Guanglei Chen
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Cheng Peng
- Department of Health Management, Beidaihe Rest and Recuperation Center of PLA Joint Logistics Support Force, Qinhuangdao, China
| | - Wenjun Chang
- Department of Environmental and Occupational Health, Naval Medical University, Shanghai, China.
| | - Jifu E
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Fuao Cao
- Department of Colorectal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
4
|
Yang C, Zhang X, Yang X, Lian F, Sun Z, Huang Y, Shen W. Function and regulation of RGS family members in solid tumours: a comprehensive review. Cell Commun Signal 2023; 21:316. [PMID: 37924113 PMCID: PMC10623796 DOI: 10.1186/s12964-023-01334-7] [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: 05/10/2023] [Accepted: 09/25/2023] [Indexed: 11/06/2023] Open
Abstract
G protein-coupled receptors (GPCRs) play a key role in regulating the homeostasis of the internal environment and are closely associated with tumour progression as major mediators of cellular signalling. As a diverse and multifunctional group of proteins, the G protein signalling regulator (RGS) family was proven to be involved in the cellular transduction of GPCRs. Growing evidence has revealed dysregulation of RGS proteins as a common phenomenon and highlighted the key roles of these proteins in human cancers. Furthermore, their differential expression may be a potential biomarker for tumour diagnosis, treatment and prognosis. Most importantly, there are few systematic reviews on the functional/mechanistic characteristics and clinical application of RGS family members at present. In this review, we focus on the G-protein signalling regulator (RGS) family, which includes more than 20 family members. We analysed the classification, basic structure, and major functions of the RGS family members. Moreover, we summarize the expression changes of each RGS family member in various human cancers and their important roles in regulating cancer cell proliferation, stem cell maintenance, tumorigenesis and cancer metastasis. On this basis, we outline the molecular signalling pathways in which some RGS family members are involved in tumour progression. Finally, their potential application in the precise diagnosis, prognosis and treatment of different types of cancers and the main possible problems for clinical application at present are discussed. Our review provides a comprehensive understanding of the role and potential mechanisms of RGS in regulating tumour progression. Video Abstract.
Collapse
Affiliation(s)
- Chenglong Yang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Xiaoyuan Zhang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Xiaowen Yang
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Fuming Lian
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Zongrun Sun
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China
| | - Yongming Huang
- Department of General Surgery, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, 272067, China.
| | - Wenzhi Shen
- Key Laboratory of Precision Oncology in Universities of Shandong, Institute of Precision Medicine, Jining Medical University, Jining, 272067, China.
| |
Collapse
|
5
|
Xu Q, Yao M, Tang C. RGS2 and female common diseases: a guard of women's health. J Transl Med 2023; 21:583. [PMID: 37649067 PMCID: PMC10469436 DOI: 10.1186/s12967-023-04462-3] [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: 07/05/2023] [Accepted: 08/21/2023] [Indexed: 09/01/2023] Open
Abstract
Currently, women around the world are still suffering from various female common diseases with the high incidence, such as ovarian cancer, uterine fibroids and preeclampsia (PE), and some diseases are even with the high mortality rate. As a negative feedback regulator in G Protein-Coupled Receptor signaling (GPCR), the Regulator of G-protein Signaling (RGS) protein family participates in regulating kinds of cell biological functions by destabilizing the enzyme-substrate complex through the transformation of hydrolysis of G Guanosine Triphosphate (GTP). Recent work has indicated that, the Regulator of G-protein Signaling 2 (RGS2), a member belonging to the RGS protein family, is closely associated with the occurrence and development of certain female diseases, providing with the evidence that RGS2 functions in sustaining women's health. In this review paper, we summarize the current knowledge of RGS2 in female common diseases, and also tap and discuss its therapeutic potential by targeting multiple mechanisms.
Collapse
Affiliation(s)
- Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd, Hangzhou, 310052, People's Republic of China
| | - Mukun Yao
- Department of Gynecology, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd, Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
6
|
Li L, Xu Q, Tang C. RGS proteins and their roles in cancer: friend or foe? Cancer Cell Int 2023; 23:81. [PMID: 37118788 PMCID: PMC10148553 DOI: 10.1186/s12935-023-02932-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/21/2023] [Indexed: 04/30/2023] Open
Abstract
As negative modulators of G-protein-coupled receptors (GPCRs) signaling, regulators of G protein signaling (RGS) proteins facilitate various downstream cellular signalings through regulating kinds of heterotrimeric G proteins by stimulating the guanosine triphosphatase (GTPase) activity of G-protein α (Gα) subunits. The expression of RGS proteins is dynamically and precisely mediated by several different mechanisms including epigenetic regulation, transcriptional regulation -and post-translational regulation. Emerging evidence has shown that RGS proteins act as important mediators in controlling essential cellular processes including cell proliferation, survival -and death via regulating downstream cellular signaling activities, indicating that RGS proteins are fundamentally involved in sustaining normal physiological functions and dysregulation of RGS proteins (such as aberrant expression of RGS proteins) is closely associated with pathologies of many diseases such as cancer. In this review, we summarize the molecular mechanisms governing the expression of RGS proteins, and further discuss the relationship of RGS proteins and cancer.
Collapse
Affiliation(s)
- Lin Li
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China
- Department of Urology, Third Affiliated Hospital of the Second Military Medical University, Shanghai, 201805, China
| | - Qiang Xu
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China
| | - Chao Tang
- National Clinical Research Center for Child Health of the Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Rd., Hangzhou, 310052, People's Republic of China.
| |
Collapse
|
7
|
Du X, Pu X, Wang X, Zhang Y, Jiang T, Ge Y, Zhu H. A novel necroptosis-related lncRNA based signature predicts prognosis and response to treatment in cervical cancer. Front Genet 2022; 13:938250. [PMID: 36561319 PMCID: PMC9763697 DOI: 10.3389/fgene.2022.938250] [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: 05/07/2022] [Accepted: 11/24/2022] [Indexed: 12/12/2022] Open
Abstract
Background: Necroptosis has been demonstrated to play a crucial role in the prognosis prediction and assessment of treatment outcome in cancers, including cervical cancer. The purpose of this study was to explore the potential prognostic value of necroptosis-related lncRNAs and their relationship with immune microenvironment and response to treatment in cervical cancer. Methods: Data from The Cancer Genome Atlas (TCGA) were collected to obtain synthetic data matrices. Necroptosis-related lncRNAs were identified by Pearson Correlation analysis. Univariate Cox and multivariate Cox regression analysis and Lasso regression were used to construct a necroptosis-related LncRNAs signature. Kaplan-Meier analysis, univariate and multivariate Cox regression analyses, receiver operating characteristic (ROC) curve, nomogram, and calibration curves analysis were performed to validate this signature. Gene set enrichment analyses (GSEA), immunoassays, and the half-maximal inhibitory concentration (IC50) were also analyzed. Results: Initially, 119 necroptosis-related lncRNAs were identified based on necroptosis-related genes and differentially expressed lncRNAs between normal and cervical cancer samples. Then, a prognostic risk signature consisting of five necroptosis-related lncRNAs (DDN-AS1, DLEU1, RGS5, RUSC1-AS1, TMPO-AS1) was established by Cox regression analysis, and LASSO regression techniques. Based on this signature, patients with cervical cancer were classified into a low- or high-risk group. Cox regression confirmed this signature as an independent prognostic predictor with an AUC value of 0.789 for predicting 1-year OS. A nomogram including signature, age, and TNM stage grade was then established, and showed an AUC of 0.82 for predicting 1-year OS. Moreover, GSEA analysis showed that immune-related pathways were enriched in the low-risk group; immunoassays showed that most immune cells, ESTIMAT scores and immune scores were negatively correlated with risk score and that the expression of immune checkpoint-proteins (CD27, CD48, CD200, and TNFRSF14) were higher in the low-risk group. In addition, patients in the low-risk group were more sensitive to Rucaparib, Navitoclax and Crizotinib than those in the high-risk group. Conclusion: We established a novel necroptosis-related lncRNA based signature to predict prognosis, tumor microenvironment and response to treatment in cervical cancer. Our study provides clues to tailor prognosis prediction and individualized immunization/targeted therapy strategies.
Collapse
|
8
|
Wang F, Zhang L, Xu Y, Xie Y, Li S. Comprehensive Analysis and Identification of Key Driver Genes for Distinguishing Between Esophageal Adenocarcinoma and Squamous Cell Carcinoma. Front Cell Dev Biol 2021; 9:676156. [PMID: 34124063 PMCID: PMC8194272 DOI: 10.3389/fcell.2021.676156] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/09/2021] [Indexed: 01/01/2023] Open
Abstract
Background: Esophageal cancer (EC) is one of the deadliest cancers in the world. However, the mechanism that drives the evolution of EC is still unclear. On this basis, we identified the key genes and molecular pathways that may be related to the progression of esophageal adenocarcinoma and squamous cell carcinoma to find potential markers or therapeutic targets. Methods: GSE26886 were obtained from Gene Expression Omnibus (GEO) database. The differentially expressed genes (DEGs) among normal samples, EA, and squamous cell carcinoma were determined using R software. Then, potential functions of DEGs were determined using the Database for Annotation, Visualization and Integrated Discovery (DAVID). The STRING software was used to identify the most important modules in the protein-protein interaction (PPI) network. The expression levels of hub genes were confirmed using UALCAN database. Kaplan-Meier plotters were used to confirm the correlation between hub genes and outcomes in EC. Results: In this study, we identified 1,098 genes induced in esophageal adenocarcinoma (EA) and esophageal squamous cell carcinoma (ESCC), and 669 genes were reduced in EA and ESCC, suggesting that these genes may play an important role in the occurrence and development of EC tumors. Bioinformatics analysis showed that these genes were involved in cell cycle regulation and p53 and phosphoinositide 3-kinase (PI3K)/Akt signaling pathway. In addition, we identified 147 induced genes and 130 reduced genes differentially expressed in EA and ESCC. The expression of ESCC in the EA group was different from that in the control group. By PPI network analysis, we identified 10 hub genes, including GNAQ, RGS5, MAPK1, ATP1B1, HADHA, HSDL2, SLC25A20, ACOX1, SCP2, and NLN. TCGA validation showed that these genes were present in the dysfunctional samples between EC and normal samples and between EA and ESCC. Kaplan-Meier analysis showed that MAPK1, ACOX1, SCP2, and NLN were associated with overall survival in patients with ESCC and EA. Conclusions: In this study, we identified a series of DEGs between EC and normal samples and between EA and ESCC samples. We also identified 10 key genes involved in the EC process. We believe that this study may provide a new biomarker for the prognosis of EA and ESCC.
Collapse
Affiliation(s)
- Feng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lan Zhang
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yue Xu
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yilin Xie
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Shenglei Li
- Department of Pathology, The First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
9
|
Su S, Shahriyari L. RGS5 plays a significant role in renal cell carcinoma. ROYAL SOCIETY OPEN SCIENCE 2020; 7:191422. [PMID: 32431860 PMCID: PMC7211867 DOI: 10.1098/rsos.191422] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 03/31/2020] [Indexed: 06/11/2023]
Abstract
Recent advances in biotechnology led to generation of large complex biological and clinical datasets that can be used to infer the underlying mechanism of many diseases and arrive at personalized treatments. One of these datasets are the whole genome profiles, including a good collection of publicly available human gene expression datasets. In this project, we analysed gene expression profiles of patients with renal cell carcinoma (RCC). We found that the regulator of G-protein signalling 5 (RGS5) might play a crucial role in initiation and progression of RCC, and it might be prognostic. We observed that a high expression level of RGS5 is associated with better survival months. Importantly, when the grade of tumour increases, the RGS5 expression level significantly decreases. Although there is no difference between expression level of RGS5 in male and female patients with primary tumours in the right kidney, among patients with primary tumours in the left kidney, females have a significantly higher RGS5 expression than male patients. Interestingly, we also observed a significant association between the high expression level of RGS5 and low serum calcium level and elevated white blood cells level.
Collapse
Affiliation(s)
- Sumeyye Su
- Department of Mathematics, University of Texas Arlington, Arlington, TX 76019, USA
| | - Leili Shahriyari
- Department of Mathematics and Statistics, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
10
|
O'Brien JB, Wilkinson JC, Roman DL. Regulator of G-protein signaling (RGS) proteins as drug targets: Progress and future potentials. J Biol Chem 2019; 294:18571-18585. [PMID: 31636120 PMCID: PMC6901330 DOI: 10.1074/jbc.rev119.007060] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) play critical roles in regulating processes such as cellular homeostasis, responses to stimuli, and cell signaling. Accordingly, GPCRs have long served as extraordinarily successful drug targets. It is therefore not surprising that the discovery in the mid-1990s of a family of proteins that regulate processes downstream of GPCRs generated great excitement in the field. This finding enhanced the understanding of these critical signaling pathways and provided potentially new targets for pharmacological intervention. These regulators of G-protein signaling (RGS) proteins were viewed by many as nodes downstream of GPCRs that could be targeted with small molecules to tune signaling processes. In this review, we provide a brief overview of the discovery of RGS proteins and of the gradual and continuing discovery of their roles in disease states, focusing particularly on cancer and neurological disorders. We also discuss high-throughput screening efforts that have led to the discovery first of peptide-based and then of small-molecule inhibitors targeting a subset of the RGS proteins. We explore the unique mechanisms of RGS inhibition these chemical tools have revealed and highlight the most up-to-date studies using these tools in animal experiments. Finally, we discuss the future opportunities in the field, as there are clearly more avenues left to be explored and potentials to be realized.
Collapse
Affiliation(s)
- Joseph B O'Brien
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242
| | - Joshua C Wilkinson
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242
| | - David L Roman
- Department of Pharmaceutical Sciences and Experimental Therapeutics, University of Iowa, Iowa City, Iowa 52242; Iowa Neuroscience Institute, Iowa City, Iowa 52242; Holden Comprehensive Cancer Center, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242.
| |
Collapse
|
11
|
Long XH, Zhou YF, Lan M, Huang SH, Li Liu Z, Shu Y. Valosin-containing protein promotes metastasis of osteosarcoma through autophagy induction and anoikis inhibition via the ERK/NF-κβ/beclin-1 signaling pathway. Oncol Lett 2019; 18:3823-3829. [PMID: 31516594 DOI: 10.3892/ol.2019.10716] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 06/07/2019] [Indexed: 12/19/2022] Open
Abstract
Valosin-containing protein (VCP) promotes the development of metastasis in osteosarcoma (OS) via the PI3K/Akt signaling pathway. However, inhibition of the PI3K/Akt pathway does not completely reverse VCP-mediated invasion and migration of OS, suggesting that VCP-mediated OS invasion and migration involves additional mechanisms. In the present study, a positive correlation between the expression of VCP and cell autophagy was observed among OS tissues. Inhibiting VCP may decrease the survival of malignant cells; however, an autophagy stimulator may compensate for VCP inhibition and promote malignant cell survival. Altering the level of autophagy did not affect cell invasiveness or migration. ERK, NF-κβ and beclin-1 protein expression levels were markedly decreased following VCP inhibition. These findings indicated that VCP may induce autophagy and enhance anoikis resistance without affecting cell invasiveness or migration. Via anoikis resistance, VCP may promote metastasis in OS. Therefore, targeting of the ERK/NF-κβ/beclin-1 signaling pathway may be an effective therapeutic strategy for the management of OS.
Collapse
Affiliation(s)
- Xin Hua Long
- Department of Emergency Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yun Fei Zhou
- Department of Orthopedics, The First People's Hospital of Foshan, Foshan, Guangdong 528000, P.R. China
| | - Min Lan
- Department of Emergency Surgery, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Shan Hu Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Zhi Li Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yong Shu
- Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| |
Collapse
|