1
|
Machuca A, Peñalver GA, Garcia RAF, Martinez-Lopez A, Castillo-Lluva S, Garcia-Calvo E, Luque-Garcia JL. Advancing rhodium nanoparticle-based photodynamic cancer therapy: quantitative proteomics and in vivo assessment reveal mechanisms targeting tumor metabolism, progression and drug resistance. J Mater Chem B 2024. [PMID: 39453320 DOI: 10.1039/d4tb01631a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
Rhodium nanoparticles have been recently discovered as good photosensitizers with great potential in cancer photodynamic therapy by effectively inducing cytotoxicity in cancer cells under near-infrared laser. This study evaluates the molecular mechanisms underlying such antitumoral effect through quantitative proteomics. The results revealed that rhodium nanoparticle-based photodynamic therapy disrupts tumor metabolism by downregulating key proteins involved in ATP synthesis and mitochondrial function, leading to compromised energy production. The treatment also induces oxidative stress and apoptosis while targeting the invasion capacity of cancer cells. Additionally, key proteins involved in drug resistance are also affected, demonstrating the efficacy of the treatment in a multi-drug resistant cell line. In vivo evaluation using a chicken embryo model also confirmed the effectiveness of the proposed therapy in reducing tumor growth without affecting embryo viability.
Collapse
Affiliation(s)
- Andres Machuca
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Gabriel A Peñalver
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| | | | - Angelica Martinez-Lopez
- Department Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Sonia Castillo-Lluva
- Department Biochemistry and Molecular Biology, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain
| | - Estefania Garcia-Calvo
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| | - Jose L Luque-Garcia
- Department Analytical Chemistry, Faculty of Chemical Sciences, Complutense University of Madrid, 28040, Madrid, Spain.
| |
Collapse
|
2
|
de Castro Sampaio SS, Ramalho MCC, de Souza CS, de Almeida Rodrigues B, de Mendonça GRS, Lazarini M. RHO subfamily of small GTPases in the development and function of hematopoietic cells. J Cell Physiol 2024:e31469. [PMID: 39434451 DOI: 10.1002/jcp.31469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/16/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024]
Abstract
RHOA, RHOB, and RHOC comprise a subfamily of RHO GTPase proteins famed for controlling cytoskeletal dynamics. RHO proteins operate downstream of multiple signals emerging from the microenvironment, leading to diverse cell responses, such as proliferation, adhesion, and migration. Therefore, RHO signaling has been centrally placed in the regulation of blood cells. Despite their high homology, unique roles of RHOA, RHOB, and RHOC have been described in hematopoietic cells. In this article, we overview the contribution of RHO proteins in the development and function of each blood cell lineage. Additionally, we highlight the aberrations of the RHO signaling pathways found in hematological malignancies, providing clues for the identification of new therapeutic targets.
Collapse
Affiliation(s)
| | | | - Caroline Santos de Souza
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | - Mariana Lazarini
- Department of Clinical and Experimental Oncology, Federal University of São Paulo, São Paulo, Brazil
| |
Collapse
|
3
|
Guzman A, Kawase T, Devanny AJ, Efe G, Navaridas R, Yu K, Regunath K, Mercer IG, Avard RC, Muniz de Queiroz R, Rustgi AK, Kaufman LJ, Prives C. Mutant p53 regulates cancer cell invasion in complex three-dimensional environments through mevalonate pathway-dependent Rho/ROCK signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.13.618100. [PMID: 39464132 PMCID: PMC11507699 DOI: 10.1101/2024.10.13.618100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Certain mutations can confer neomorphic gain of function (GOF) activities to the p53 protein that affect cancer progression. Yet the concept of mutant p53 GOF has been challenged. Here, using various strategies to alter the status of mutant versions of p53 in different cell lines, we demonstrate that mutant p53 stimulates cancer cell invasion in three-dimensional environments. Mechanistically, mutant p53 enhances RhoA/ROCK-dependent cell contractility and cell-mediated extracellular matrix (ECM) re-organization via increasing mevalonate pathway-dependent RhoA localization to the membrane. In line with this, RhoA-dependent pro-invasive activity is also mediated by IDI-1, a mevalonate pathway product. Further, the invasion-enhancing effect of mutant p53 is dictated by the biomechanical properties of the surrounding ECM, thereby adding a cell-independent layer of regulation to mutant p53 GOF activity that is mediated by dynamic reciprocal cell-ECM interactions. Together our findings link mutant p53 metabolic GOF activity with an invasive cellular phenotype in physiologically relevant and context-dependent settings. Significance This study addresses the contribution of mutant p53 to the process of cancer cell dissemination in physiologically relevant three-dimensional environments - a key characteristic of metastatic disease. Several mutant p53 proteins display pro-oncogenic activity with respect to cancer cell invasion in 3D environments via mevalonate pathway-dependent Rho/ROCK signaling axis.
Collapse
|
4
|
Setayesh T, Hu Y, Vaziri F, Wei D, Wan YJY. The spatial impact of a Western diet in enriching Galectin-1-regulated Rho, ECM, and SASP signaling in a novel MASH-HCC mouse model. Biomark Res 2024; 12:122. [PMID: 39402682 PMCID: PMC11476289 DOI: 10.1186/s40364-024-00660-3] [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: 08/14/2024] [Accepted: 09/20/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) arising from metabolic dysfunction-associated steatohepatitis (MASH) presents a significant clinical challenge, particularly given the prevalence of the Western diet (WD). The influence of diet on the tumor microenvironment remains poorly understood. Galectin-1 (Gal-1) is a biomarker for HCC and has a crucial role in liver carcinogenesis. Our previous studies demonstrated that silencing Gal-1 effectively treats mouse HCC. However, the impacts of a WD on Gal-1 signaling on MASH to HCC progression are unknown, and this study addresses these knowledge gaps. METHODS We developed a novel MASH-HCC mouse model. Using spatial transcriptomics and multiplex immunohistochemistry (IHC), we studied the effects of a WD on the liver and tumor microenvironment. By modulating Gal-1 expression through silencing and overexpression, we explored the location-specific impacts of WD on Gal-1 signaling. RESULTS Pathways such as Rho signaling, extracellular matrix (ECM) remodeling, and senescence-associated secretory phenotypes (SASP) were prominently activated in WD-induced metabolic dysfunction-associated fatty liver disease (MAFLD) and MASH-HCC, compared to healthy livers controls. Furthermore, Rho GTPase effectors, ECM remodeling, neutrophil degranulation, cellular stress, and cell cycle pathways were consistently enriched in human and mouse MASH-HCC. Spatially, these pathways were enriched in the tumor and tumor margins of mouse MASH-HCC. Additionally, there was a notable increase in CD11c and PD-L1-positive cells from non-tumor tissues to the tumor margin and inside the tumor of MASH-HCC, suggesting compromised immune surveillance due to WD intake. Moreover, MASH-HCC exhibited significant Gal-1 induction in N-Cadherin-positive cells, indicating enhanced epithelial-to-mesenchymal transition (EMT). Modulating Gal-1 expression in MASH-HCC further established its specific roles in regulating Rho signaling and SASP in the tumor margin and non-tumor tissues in MASH-HCC. CONCLUSION WD intake significantly influences vital cellular processes involved in Gal-1-mediated signaling, including Rho signaling and ECM remodeling, in the tumor microenvironment, thereby contributing to the development of MASH-HCC.
Collapse
Affiliation(s)
- Tahereh Setayesh
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Ying Hu
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Farzam Vaziri
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Dongguang Wei
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA
| | - Yu-Jui Yvonne Wan
- Department of Medical Pathology and Laboratory Medicine, University of California, Davis, Room 3400B, Research Building III, 4645 2nd Ave, Sacramento, CA, 95817, USA.
| |
Collapse
|
5
|
Ning Y, Zheng M, Zhang Y, Jiao Y, Wang J, Zhang S. RhoA-ROCK2 signaling possesses complex pathophysiological functions in cancer progression and shows promising therapeutic potential. Cancer Cell Int 2024; 24:339. [PMID: 39402585 PMCID: PMC11475559 DOI: 10.1186/s12935-024-03519-7] [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: 01/12/2024] [Accepted: 09/29/2024] [Indexed: 10/19/2024] Open
Abstract
The Rho GTPase signaling pathway is responsible for cell-specific processes, including actin cytoskeleton organization, cell motility, cell division, and the transcription of specific genes. The implications of RhoA and the downstream effector ROCK2 in cancer epithelial-mesenchymal transition, migration, invasion, and therapy resistance associated with stem cells highlight the potential of targeting RhoA/ROCK2 signaling in therapy. Tumor relapse can occur due to cancer cells that do not fully respond to adjuvant chemoradiotherapy, targeted therapy, or immunotherapy. Rho signaling-mediated mitotic defects and cytokinesis failure lead to asymmetric cell division, allowing cells to form polyploids to escape cytotoxicity and promote tumor recurrence and metastasis. In this review, we elucidate the significance of RhoA/ROCK2 in the mechanisms of cancer progression and summarize their inhibitors that may improve treatment strategies.
Collapse
Affiliation(s)
- Yidi Ning
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, P.R. China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China
| | - Yue Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Yuqi Jiao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Jiangping Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, P.R. China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, 300121, P.R. China.
| |
Collapse
|
6
|
Zhang W, Bai X, Liu T, Mao Y, Zhang L, Wang W, Yu H. Rho GTPase activating protein 11A promotes tongue squamous cell carcinoma proliferation and is a transcriptional target of forkhead box M1. J Dent Sci 2024; 19:2268-2277. [PMID: 39347089 PMCID: PMC11437295 DOI: 10.1016/j.jds.2024.02.015] [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: 01/07/2024] [Revised: 02/16/2024] [Indexed: 10/01/2024] Open
Abstract
Background/purpose Rho GTPase activating protein 11A (ARHGAP11A) can facilitate GTP hydrolysis in RhoA. The functions of ARHGAP11A in oral squamous cell carcinoma (OSCC) have not yet been explored. This study aimed to investigate the expression profile of ARHGAP11A in OSCC, its correlation with patient prognosis, its effect on cell-cycle progression, and the mechanisms by which it is dysregulated. Materials and methods Bioinformatics analysis was conducted using data from The Cancer Genome Atlas-Head and Neck Squamous Cell Carcinoma (TCGA-HNSC). Lentiviruses carrying ARHGAP11A shRNAs were employed to determine the effects of ARHGAP11A knockdown on tumor cell proliferation and cell-cycle progression. Dual-luciferase reporter assays were utilized to examine how FOXM1 transcriptionally regulates ARHGAP11A expression. Results ARHGAP11A upregulation was associated with unfavorable overall survival (OS) in patients with TSCC (HR: 2.142, 95%CI: 1.224-3.749, P = 0.007), but not in patients with OSCC of sites other than the tongue. ARHGAP11A knockdown inhibited the proliferation of TSCC cells in vitro and in vivo, and induced G1 phase arrest. ARHGAP11A knockdown increased GTP-RhoA but decreased p-RB levels, while it did not affect the total expression of RhoA and RB. ARHGAP11A knockdown increased p27 and decreased cyclin E1 expression. ARHGAP11A is transcriptionally activated by FOXM1 via multiple FOXM1 binding sites in the promoter regions in TSCC cells. Conclusion This study revealed the oncogenic role of ARHGAP11A in TSCC, highlighting its impact on cell-cycle control and tumor proliferation. Furthermore, the regulatory relationship between FOXM1 and ARHGAP11A provides new insights into the transcriptional networks in TSCC.
Collapse
Affiliation(s)
- Weiwei Zhang
- Department of Orthodontics, Binzhou Medical University Hospital, Binzhou, China
| | - Xueyan Bai
- Department of Orthodontics, Binzhou Medical University Hospital, Binzhou, China
| | - Tingting Liu
- Department of Oral Medicine, Binzhou Medical University Hospital, Binzhou, China
| | - Yulong Mao
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Lingnan Zhang
- Department of Orthodontics, Binzhou Medical University Hospital, Binzhou, China
| | - Wenlong Wang
- Department of Oral and Maxillofacial Surgery, Binzhou Medical University Hospital, Binzhou, China
| | - Huanying Yu
- Department of Prosthodontics, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
7
|
Shouib R, Eitzen G. Inflammatory gene regulation by Cdc42 in airway epithelial cells. Cell Signal 2024; 122:111321. [PMID: 39067837 DOI: 10.1016/j.cellsig.2024.111321] [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: 02/04/2024] [Revised: 07/16/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
Cytokine release from airway epithelial cells is a key immunological process that coordinates an immune response in the lungs. We propose that the Rho GTPase, Cdc42, regulates both transcription and trafficking of cytokines, ultimately affecting the essential process of cytokine release and subsequent inflammation in the lungs. Here, we examined the pro-inflammatory transcriptional profile that occurs in bronchial epithelial cells (BEAS-2B) in response to TNF-α using RNA-Seq and differential gene expression analysis. To interrogate the role of Cdc42 in inflammatory gene expression, we used a pharmacological inhibitor of Cdc42, ML141, and determined changes in the transcriptomic profile induced by Cdc42 inhibition. Our results indicated that Cdc42 inhibition with ML141 resulted in a unique inflammatory phenotype concomitant with increased gene expression of ER stress genes, Golgi membrane and vesicle transport genes. To further interrogate the inflammatory pathways regulated by Cdc42, we made BEAS-2B knockdown strains for the signaling targets TRIB3, DUSP5, SESN2 and BMP4, which showed high differential expression in response to Cdc42 inhibition. Depletion of DUSP5 and TRIB3 reduced the pro-inflammatory response triggered by Cdc42 inhibition as shown by a reduction in cytokine transcript levels. Depletion of SESN2 and BMP4 did not affect cytokine transcript level, however, Golgi fragmentation was reduced. These results provide further evidence that in airway epithelial cells, Cdc42 is part of a signaling network that controls inflammatory gene expression and secretion by regulating Golgi integrity. Summary sentence:We define the Cdc42-regulated gene networks for inflammatory signaling in airway epithelial cells which includes regulation of ER stress response and vesicle trafficking pathways.
Collapse
Affiliation(s)
- Rowayna Shouib
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada
| | - Gary Eitzen
- Department of Cell Biology, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
8
|
Shen J, Su X, Wang S, Wang Z, Zhong C, Huang Y, Duan S. RhoJ: an emerging biomarker and target in cancer research and treatment. Cancer Gene Ther 2024; 31:1454-1464. [PMID: 38858534 DOI: 10.1038/s41417-024-00792-6] [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: 12/19/2023] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/12/2024]
Abstract
RhoJ is a Rho GTPase that belongs to the Cdc42 subfamily and has a molecular weight of approximately 21 kDa. It can activate the p21-activated kinase family either directly or indirectly, influencing the activity of various downstream effectors and playing a role in regulating the cytoskeleton, cell movement, and cell cycle. RhoJ's expression and activity are controlled by multiple upstream factors at different levels, including expression, subcellular localization, and activation. High RhoJ expression is generally associated with a poor prognosis for cancer patients and is mainly due to an increased number of tumor blood vessels and abnormal expression in malignant cells. RhoJ promotes tumor progression through several pathways, particularly in tumor angiogenesis and drug resistance. Clinical data also indicates that high RhoJ expression is closely linked to the pathological features of tumor malignancy. There are various cancer treatment methods that target RhoJ signaling, such as direct binding to inhibit the RhoJ effector pocket, inhibiting RhoJ expression, blocking RhoJ upstream and downstream signals, and indirectly inhibiting RhoJ's effect. RhoJ is an emerging cancer biomarker and a significant target for future cancer clinical research and drug development.
Collapse
Affiliation(s)
- Jinze Shen
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Xinming Su
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Shana Wang
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Zehua Wang
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China
| | - Chenming Zhong
- Medical Genetics Center, School of Medicine, Ningbo University, Ningbo, Zhejiang, China
| | - Yi Huang
- Department of Neurosurgery, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China.
| | - Shiwei Duan
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, Zhejiang, China.
| |
Collapse
|
9
|
Bou Malhab LJ, Schmidt S, Fagotto-Kaufmann C, Pion E, Gadea G, Roux P, Fagotto F, Debant A, Xirodimas DP. An Anti-Invasive Role for Mdmx through the RhoA GTPase under the Control of the NEDD8 Pathway. Cells 2024; 13:1625. [PMID: 39404389 PMCID: PMC11475522 DOI: 10.3390/cells13191625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Mdmx (Mdm4) is established as an oncogene mainly through repression of the p53 tumour suppressor. On the other hand, anti-oncogenic functions for Mdmx have also been proposed, but the underlying regulatory pathways remain unknown. Investigations into the effect of inhibitors for the NEDD8 pathway in p53 activation, human cell morphology, and in cell motility during gastrulation in Xenopus embryos revealed an anti-invasive function of Mdmx. Through stabilisation and activation of the RhoA GTPase, Mdmx is required for the anti-invasive effects of NEDDylation inhibitors. Mechanistically, through its Zn finger domain, Mdmx preferentially interacts with the inactive GDP-form of RhoA. This protects RhoA from degradation and allows for RhoA targeting to the plasma membrane for its subsequent activation. The effect is transient, as prolonged NEDDylation inhibition targets Mdmx for degradation, which subsequently leads to RhoA destabilisation. Surprisingly, Mdmx degradation requires non-NEDDylated (inactive) Culin4A and the Mdm2 E3-ligase. This study reveals that Mdmx can control cell invasion through RhoA stabilisation/activation, which is potentially linked to the reported anti-oncogenic functions of Mdmx. As inhibitors of the NEDD8 pathway are in clinical trials, the status of Mdmx may be a critical determinant for the anti-tumour effects of these inhibitors.
Collapse
Affiliation(s)
- Lara J. Bou Malhab
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates
| | - Susanne Schmidt
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Christine Fagotto-Kaufmann
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Emmanuelle Pion
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Gilles Gadea
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Pierre Roux
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Francois Fagotto
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Anne Debant
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| | - Dimitris P. Xirodimas
- CRBM, Cell Biology Research Centre of Montpellier, Université de Montpellier, CNRS, 34293 Montpellier, France; (S.S.); (C.F.-K.); (E.P.); (G.G.); (P.R.); (F.F.)
| |
Collapse
|
10
|
Reffai A, Hori M, Adusumilli R, Bermudez A, Bouzoubaa A, Pitteri S, Bennani Mechita M, Mallick P. A Proteomic Analysis of Nasopharyngeal Carcinoma in a Moroccan Subpopulation. Cancers (Basel) 2024; 16:3282. [PMID: 39409902 PMCID: PMC11476039 DOI: 10.3390/cancers16193282] [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: 07/22/2024] [Revised: 09/14/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
BACKGROUND Nasopharyngeal carcinoma (NPC) is a distinct cancer of the head and neck that is highly prevalent in Southeast Asia and North Africa. Though an extensive analysis of environmental and genetic contributors has been performed, very little is known about the proteome of this disease. A proteomic analysis of formalin-fixed paraffin-embedded (FFPE) tissues can provide valuable information on protein expression and molecular patterns for both increasing our understanding of the disease and for biomarker discovery. To date, very few NPC proteomic studies have been performed, and none focused on patients from Morocco and North Africa. METHODS Label-free Liquid Chromatography-Tandem Mass Spectrometry (LC-MS/MS) was used to perform a proteomic analysis of FFPE tissue samples from a cohort of 41 NPC tumor samples of Morocco and North Africa origins. The LC-MS/MS data from this cohort were analyzed alongside 21 healthy controls using MaxQuant 2.4.2.0. A differential expression analysis was performed using the MSstats package in R. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional annotations were carried out using the DAVID bioinformatic tool. RESULTS 3341 proteins were identified across our NPC cases, revealing three main clusters and five DEPs with prognostic significance. The sex disparity of NPC was investigated from a proteomic perspective in which 59 DEPs were found between males and females, with significantly enriched terms associated with the immune response and gene expression. Furthermore, 26 DEPs were observed between patients with early and advanced stages of NPC with a significant cluster related to the immune response, implicating up-regulated DEPs such as IGHA, IGKC, and VAT1. Across both datasets, 6532 proteins were quantified between NPC patients and healthy controls. Among them, 1507 differentially expressed proteins (DEPs) were observed. GO and KEGG pathway analyses showed enriched terms of DEPs related to increased cellular activity, cell proliferation, and survival. PI3K and MAPK proteins as well as RAC1 BCL2 and PPIA were found to be overexpressed between cancer tissues and healthy controls. EBV infection was also one of the enriched pathways implicating its latent genes like LMP1 and LMP2 that activate several proteins and signaling pathways including NF-Kappa B, MAPK, and JAK-STAT pathways. CONCLUSION Our findings unveil the proteomic landscape of NPC for the first time in the Moroccan population. These studies additionally may provide a foundation for identifying potential biomarkers. Further research is still needed to help develop tools for the early diagnosis and treatment of NPC in Moroccan and North African populations.
Collapse
Affiliation(s)
- Ayman Reffai
- Intelligent Automation and BioMed Genomics Laboratory, Biology Department, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University-Tetouan, Tangier 90000, Morocco
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michelle Hori
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Ravali Adusumilli
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Abel Bermudez
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | | | - Sharon Pitteri
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mohcine Bennani Mechita
- Intelligent Automation and BioMed Genomics Laboratory, Biology Department, Faculty of Sciences and Techniques of Tangier, Abdelmalek Essaadi University-Tetouan, Tangier 90000, Morocco
| | - Parag Mallick
- Canary Center for Cancer Early Detection, School of Medicine, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
11
|
Sajib MS, Zahra FT, Lamprou M, Akwii RG, Park JH, Osorio M, Tullar P, Doci CL, Zhang C, Huveneers S, Van Buul JD, Wang MH, Markiewski MM, Srivastava SK, Zheng Y, Gutkind JS, Hu J, Bickel U, Maeda DY, Zebala JA, Lionakis MS, Trasti S, Mikelis CM. Tumor-induced endothelial RhoA activation mediates tumor cell transendothelial migration and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.22.614304. [PMID: 39372784 PMCID: PMC11451620 DOI: 10.1101/2024.09.22.614304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The endothelial barrier plays an active role in transendothelial tumor cell migration during metastasis, however, the endothelial regulatory elements of this step remain obscure. Here we show that endothelial RhoA activation is a determining factor during this process. Breast tumor cell-induced endothelial RhoA activation is the combined outcome of paracrine IL-8-dependent and cell-to-cell contact β 1 integrin-mediated mechanisms, with elements of this pathway correlating with clinical data. Endothelial-specific RhoA blockade or in vivo deficiency inhibited the transendothelial migration and metastatic potential of human breast tumor and three murine syngeneic tumor cell lines, similar to the pharmacological blockade of the downstream RhoA pathway. These findings highlight endothelial RhoA as a potent, universal target in the tumor microenvironment for anti-metastatic treatment of solid tumors.
Collapse
|
12
|
Zhao C, Wu Z, Yao Z, Zhang F, Zhao R, Cao X, Ling S, Jiang X. The tumorigenic effect of the high expression of ABRACL in glioma and its potential as a therapeutic target. Heliyon 2024; 10:e36597. [PMID: 39286126 PMCID: PMC11402703 DOI: 10.1016/j.heliyon.2024.e36597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Gliomas are the most common malignant intracranial tumors, with no effective treatments. Better understanding and identification of novel targets are urgently warranted. Actin-binding Rho activating C-terminal like (ABRACL) has been reported as an oncogene in several cancer types. However, the potential roles of ABRACL in the tumorigenesis of malignant glioma remain unknown. We discovered that ABRACL is highly expressed in different sub-types of gliomas in both CGGA and TCGA databases, which was further validated in glioblastoma cell lines and normal human astrocyte lines. RT-qPCR, Western blotting and immunohistochemistry demonstrated that ABRACL expression in glioma tissues was upregulated along with the increasing WHO grades. Further survival analysis of glioma patients also revealed that the overall survival of patients in the ABRACL high expression level group were significantly shorter than those in the low expression level group. Knockdown of ABRACL inhibited the proliferation, cell migration, invasion and cytodynamics behaviors in glioma cell lines via activating STAT3 signaling, which also induced apoptosis and cell cycle arrest. Conversely, overexpressing ABRACL promoted cell renewing and migration, enabled more flexible cell deformation, supporting ABRACL being a bona fide oncogene. Intracranial orthotopic xenograft experiment further confirmed that ABRACL downregulation significantly suppressed glioma growth. These results have demonstrated that the tumorigenic effect of ABRACL is partly mediated by STAT3, whose expression also correlates with clinical prognosis. ABRACL facilitates glioma malignancy phenotype through regulating the cytoskeleton by activating STAT3 pathway, suggesting that it may represent a potential therapeutic target for glioblastoma.
Collapse
Affiliation(s)
- Chenhui Zhao
- Department of Neurosurgery, Shandong Provincial Third Hospital, Shandong University, Jinan, China
| | - Zeyu Wu
- Translational Research Institute for Neurological Disorders, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wannan Medical College, Wuhu, China
| | - Zhipeng Yao
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Fan Zhang
- School of Chemistry and Chemical Engineering, Southeast University, Nanjing, China
| | - Rui Zhao
- Translational Research Institute for Neurological Disorders, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wannan Medical College, Wuhu, China
| | - Xiaoxiang Cao
- Translational Research Institute for Neurological Disorders, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wannan Medical College, Wuhu, China
| | - Shizhang Ling
- Translational Research Institute for Neurological Disorders, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wannan Medical College, Wuhu, China
| | - Xiaochun Jiang
- Department of Neurosurgery, Shandong Provincial Third Hospital, Shandong University, Jinan, China
| |
Collapse
|
13
|
Liu C, Chen S, Zhang Y, Zhou X, Wang H, Wang Q, Lan X. Mechanisms of Rho GTPases in regulating tumor proliferation, migration and invasion. Cytokine Growth Factor Rev 2024:S1359-6101(24)00075-3. [PMID: 39317522 DOI: 10.1016/j.cytogfr.2024.09.002] [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: 08/28/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
The occurrence of most cancers is due to the clonal proliferation of tumor cells, immune evasion, and the ability to spread to other body parts. Rho GTPases, a family of small GTPases, are key regulators of cytoskeleton reorganization and cell polarity. Additionally, Rho GTPases are key proteins that induce the proliferation and metastasis of tumor cells. This review focuses on the complex regulatory mechanisms of Rho GTPases, exploring their critical role in promoting tumor cell proliferation and dissemination. Regarding tumor cell proliferation, attention is given to the role of Rho GTPases in regulating the cell cycle and mitosis. In terms of tumor cell dissemination, the focus is on the role of Rho GTPases in regulating cell migration and invasion. Overall, this review elucidates the mechanisms of Rho GTPases members in the development of tumor cells, aiming to provide theoretical references for the treatment of mammalian tumor diseases and related applications.
Collapse
Affiliation(s)
- Cheng Liu
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| | - Shutao Chen
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| | - Yu Zhang
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| | - Xinyi Zhou
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| | - Haiwei Wang
- Chongqing Academy Of Animal Sciences, Chongqing 402460, China.
| | - Qigui Wang
- Chongqing Academy Of Animal Sciences, Chongqing 402460, China.
| | - Xi Lan
- College Of Animal, Science And Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
14
|
Li Y, Gu A, Yang L, Wang Q. RAC1 serves as a prognostic factor and correlated with immune infiltration in liver hepatocellular carcinoma. J Cancer Res Clin Oncol 2024; 150:418. [PMID: 39264423 PMCID: PMC11393158 DOI: 10.1007/s00432-024-05933-w] [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: 06/20/2024] [Accepted: 08/28/2024] [Indexed: 09/13/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (LIHC) has severe consequences due to late diagnosis and the lack of effective therapies. Currently, potential biomarkers for the diagnosis and prognosis of LIHC have not been systematically evaluated. Previous studies have reported that RAC1 is associated with the B cell receptor signaling pathway in various tumor microenvironments, but its relationship with LIHC remains unclear. We investigated the relationship between RAC1 and the prognosis and immune infiltration microenvironment of LIHC, exploring its potential as a prognostic biomarker for this type of cancer. METHODS In this study, we analyzed data from The Cancer Genome Atlas (TCGA) using the Wilcoxon signed-rank test and logistic regression to assess the association between RAC1 expression and clinical characteristics in LIHC patients. Additionally, Kaplan-Meier and Cox regression methods were employed to confirm the impact of RAC1 expression levels on overall survival. Immunohistochemistry was used to validate RAC1 protein expression in LIHC. We constructed RAC1 knockdown LIHC cells and studied the effects of RAC1 protein on cell proliferation and migration at both cellular and animal levels. RESULTS RAC1 expression levels were significantly elevated in LIHC tissues compared to normal tissues. High RAC1 expression was strongly associated with advanced pathological stages and was identified as an independent factor negatively affecting overall survival. At both cellular and animal levels, RAC1 knockdown significantly inhibited the proliferation and migration of LIHC cells. Furthermore, RAC1 expression was positively correlated with the infiltration of Th2 cells and macrophages in the tumor microenvironment, suggesting that RAC1 may contribute to the deterioration of the tumor immunosuppressive microenvironment and potentially lead to reduced patient survival. CONCLUSION These findings indicate that RAC1 expression promotes LIHC proliferation and migration and influences the landscape of immune cell infiltration in the tumor microenvironment. Based on these results, RAC1 is proposed as a potential prognostic biomarker for LIHC, associated with both cancer progression and tumor immune cell infiltration.
Collapse
Affiliation(s)
- Yuan Li
- Department of Chemotherapy, The Second Hospital of Nanjing,Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Aidong Gu
- Department of Hepatobiliary Surgery, The Second Hospital of Nanjing,Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Lili Yang
- Department of Chemotherapy, The Second Hospital of Nanjing,Affiliated to Nanjing University of Chinese Medicine, Nanjing, China
| | - Qingbo Wang
- Department of Chemotherapy, The Second Hospital of Nanjing,Affiliated to Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
15
|
Tabassum R, Dilshad E. A comparative anticancer analysis of iron oxide nanoparticles of Hippophae rhamnoides and Cichorium intybus found in the Karakoram Range of Gilgit Baltistan against liver cancer targeting the RhoA gene. Drug Dev Ind Pharm 2024:1-10. [PMID: 39226126 DOI: 10.1080/03639045.2024.2400209] [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: 05/02/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/05/2024]
Abstract
OBJECTIVE The current research work focused on the evaluation of of H. rhamnoides and C. intybus Fe2O3 NPs against liver cancer cell line (HepG2) by performing antiproliferative assay targeting the RhoA gene and apoptotic pathway genes and proteins. METHODS Fe2O3 NPs were synthesized using extracts of H. rhamnoides and C. intybus and characterized by UV-Vis spectroscopy, FTIR, SEM/EDS and XRD. MTT assay was used to study cytotoxicity against the HepG2 cells. Real-time qPCR and ELISA were used for the gene and protein analysis. RESULTS An absorbance peak at 300 nm for H. rhamnoides and 289 nm for C. intybus nanoparticles were observed by UV-Vis analysis. The FTIR bands of H. rhamnoide Fe2O3 NPs suggested the presence of aldehydes, alcohols and polyols whereas bands of C. intybus Fe2O3 NPs suggested the presence of carboxyl groups, hydroxyl groups, alkynes and amines. The size of Fe2O3 NPs was found to be 27 ± 5nm for H. rhamnoides and 84 ± 4nm for C. intybus. The IC50 value of 41.69 µM for H. rhmnoides and 71.04 µM for C. intybus Fe2O3 NPs compared to plant extract (78.10 and 96.03 µM for H. rhamnoides and C. intybus, respectively) were found against HepG2 cells. The gene expression and protein levels of RhoA were decreased whereas those of bax, caspase 3, caspase 8 and caspase 9 were found increased. CONCLUSION Nanoparticles and extract of H. rhamnoides were found more effective as compared to C. intybus, which was evident by the results of cytotoxicity and analysis of studied genes and proteins.
Collapse
Affiliation(s)
- Rukhsana Tabassum
- Department of Bioinformatics and Biosciences, Faculty of Health and Life Sciences, Capital University of Science and Technology (CUST), Islamabad, Pakistan
| | - Erum Dilshad
- Department of Bioinformatics and Biosciences, Faculty of Health and Life Sciences, Capital University of Science and Technology (CUST), Islamabad, Pakistan
| |
Collapse
|
16
|
Pipili A, Babteen NA, Kuwair L, Jannet MB, Quist J, Ong KKV, Pitaluga R, Grigoriadis AG, Tutt A, Wells CM. PAK6 acts downstream of IQGAP3 to promote contractility in triple negative breast cancer cells. Cell Signal 2024; 121:111233. [PMID: 38763182 DOI: 10.1016/j.cellsig.2024.111233] [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: 03/21/2024] [Revised: 05/14/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Breast cancer is a heterogeneous disease that remains the most common malignancy among women worldwide. During genomic analysis of breast tumours, mRNA levels of IQGAP3 were found to be upregulated in triple negative tumours. IQGAP3 was subsequently found to be expressed across a panel of triple negative breast cancer (TNBC) cell lines. Depleting expression levels of IQGAP3 delivered elongated cells, disrupted cell migration, and inhibited the ability of cells to form specialised invasive adhesion structures, termed invadopodia. The morphological changes induced by IQGAP3 depletion were found to be dependent on RhoA. Indeed, reduced expression of IQGAP3 disrupted RhoA activity and actomyosin contractility. Interestingly, IQGAP3 was also found to interact with p-21 activated kinase 6 (PAK6); a protein already associated with the regulation of cell morphology. Moreover, PAK6 depletion phenocopied IQGAP3 depletion in these cells. Whereas PAK6 overexpression rescued the IQGAP3 depletion phenotype. Our work points to an important PAK6-IQGAP3-RhoA pathway that drives the cellular contractility of breast cancer cells promoting both cell migration and adhesive invasion of these cells. As this phenotype is relevant to the process of metastasis and re-seeding of metastasis, the pharmacological targeting of PAK6 could lead to clinical benefit in TNBC patients.
Collapse
Affiliation(s)
- Aikaterini Pipili
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Nouf A Babteen
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK; Department of Biological Science, Collage of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Lujain Kuwair
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Mahfuja Bulu Jannet
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Jelmar Quist
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Karine K V Ong
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Ryan Pitaluga
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Anita G Grigoriadis
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK
| | - Andrew Tutt
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK; Tony Robins Breast Cancer Now Research Centre, The Institute of Cancer Research, London, UK
| | - Claire M Wells
- School of Cancer and Pharmaceutical Sciences, Kings College London, London, UK.
| |
Collapse
|
17
|
Bashir I, Dilshad E. A comparative study of Mentha longifolia var. asiatica and Zygophyllum arabicum ZnO nanoparticles against breast cancer targeting Rab22A gene. PLoS One 2024; 19:e0308982. [PMID: 39213285 PMCID: PMC11364221 DOI: 10.1371/journal.pone.0308982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Breast cancer is the most frequently diagnosed cancer worldwide, and the incidence rate has increased enormously over the last three decades. Rab proteins are members of the Rab GTPase superfamily. The aberrant function of these proteins leads to the development of tumors. Mentha longifolia var. asiatica and Zygophyllum arabicum have been known for their therapeutic potential for ages. The present study aimed to synthesize ZnO nanoparticles encapsulated with the extracts of M. longifolia var. asiatica and Z. arabicum and evaluating their therapeutic potential against breast cancer, targeting the Rab22A gene and its protein. UV-Vis spectrophotometer showed characteristic absorbance peaks at 295 nm and 345 nm for Z. arabicum and M. longifolia var. asiatica ZnONPs, respectively. The FTIR bands of Z. arabicum nanoparticles suggested the presence of aldehydes, alcohols, and polyols whereas bands of M. longifolia var. asiatica ZnONPs suggested the presence of carboxyl groups, hydroxyl groups, alkynes, and amines. SEM revealed the size of Z. arabicum ZnO NPs to be 25 ± 4 nm with a spherical shape as compared to nanoparticles of M. longifolia var. asiatica having a size of 35 ± 6 nm with a hexagonal shape. EDX determined the elemental composition of both particles. The cytotoxicity of both plant extracts and respective NPs was determined against the MCF-7 breast cancer cell line, which was found to be significant with an IC50 value of 51.68 μM for Z. arabicum and 88.02 μM for M. longifolia var. asiatica ZnO compared to plant extracts (64.01 μM and 107.9 μM for Z. arabicum and M. longifolia var. asiatica). The gene expression and protein levels of Rab22A were decreased in nanoparticle-treated cells as compared to the control group. The apoptotic role of synthesized nanoparticles against the MCF-7 cell line was also determined by the expression of apoptotic pathway genes and proteins (bax, caspase 3, caspase 8 and caspase 9). All samples showed significant apoptotic activity by activating intrinsic and extrinsic pathway genes. The activity of Z. arabicum was more eminent as compared to M. longifolia var. asiatica which was evident by the greater expression of studied genes and proteins as determined by Real-time qPCR and ELISA. This is the first-ever report describing the comparative analysis of the efficacy of Z. arabicum and M. longifolia var. asiatica ZnONPs against breast cancer.
Collapse
Affiliation(s)
- Iqra Bashir
- Department of Bioinformatics and Biosciences, Faculty of Health and Life Sciences, Capital University of Science and Technology (CUST), Islamabad, Pakistan
| | - Erum Dilshad
- Department of Bioinformatics and Biosciences, Faculty of Health and Life Sciences, Capital University of Science and Technology (CUST), Islamabad, Pakistan
| |
Collapse
|
18
|
Ramos R, Vinyals A, Campos-Martin R, Cabré E, Bech JJ, Vaquero J, Gonzalez-Sanchez E, Bertran E, Ferreres JR, Lorenzo D, De La Torre CG, Fabregat I, Caminal JM, Fabra À. New Insights into the Exosome-Induced Migration of Uveal Melanoma Cells and the Pre-Metastatic Niche Formation in the Liver. Cancers (Basel) 2024; 16:2977. [PMID: 39272836 PMCID: PMC11394004 DOI: 10.3390/cancers16172977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/14/2024] [Accepted: 08/25/2024] [Indexed: 09/15/2024] Open
Abstract
UM is an aggressive intraocular tumor characterized by high plasticity and a propensity to metastasize in the liver. However, the underlying mechanisms governing liver tropism remain poorly understood. Given the emerging significance of exosomes, we sought to investigate the contribution of UM-derived exosomes to specific steps of the metastatic process. Firstly, we isolated exosomes from UM cells sharing a common genetic background and different metastatic properties. A comparison of protein cargo reveals an overrepresentation of proteins related to cytoskeleton remodeling and actin filament-based movement in exosomes derived from the parental cells that may favor the detachment of cells from the primary site. Secondly, we assessed the role of macrophages in reprogramming the HHSCs by exosomes. The activation of HHSCs triggered a pro-inflammatory and pro-fibrotic environment through cytokine production, upregulation of extracellular matrix molecules, and the activation of signaling pathways. Finally, we found that activated HHSCs promote increased adhesion and migration of UM cells. Our findings shed light on the pivotal role of exosomes in pre-metastatic niche construction in the liver.
Collapse
Affiliation(s)
- Raquel Ramos
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
| | - Antònia Vinyals
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
| | - Rafael Campos-Martin
- Division of Neurogenetics and Molecular Psychiatry, Department of Psychiatry and Psychotherapy, University of Cologne, 50937 Cologne, Germany
| | - Eduard Cabré
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
| | - Joan Josep Bech
- Clinical Proteomics Unit, IDIBELL, 08908 Barcelona, Spain
- Proteomic Unit, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, 08916 Badalona, Spain
| | - Javier Vaquero
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
- HepatoBiliary Tumors Lab, Centro de Investigación del Cancer and Instituto de Biologia Molecular y Celular del Cancer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain
| | - Ester Gonzalez-Sanchez
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
- HepatoBiliary Tumors Lab, Centro de Investigación del Cancer and Instituto de Biologia Molecular y Celular del Cancer, CSIC-Universidad de Salamanca, 37007 Salamanca, Spain
- Department of Physiological Sciences, Faculty of Pharmacy, University of Salamanca, 37008 Salamanca, Spain
| | - Esther Bertran
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
| | - Josep Ramon Ferreres
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
- Dermatology Service, IDIBELL, Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Daniel Lorenzo
- Ocular Translational Eye Research Unit, Ophthalmology Department, Spanish Ocular Oncology National Referral Center (CSUR), Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Carolina G De La Torre
- Clinical Proteomics Unit, IDIBELL, 08908 Barcelona, Spain
- Proteomic Unit, Josep Carreras Leukaemia Research Institute (IJC), Ctra de Can Ruti, 08916 Badalona, Spain
| | - Isabel Fabregat
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
| | - Jose Maria Caminal
- Ocular Translational Eye Research Unit, Ophthalmology Department, Spanish Ocular Oncology National Referral Center (CSUR), Hospital Universitari de Bellvitge, 08907 Barcelona, Spain
| | - Àngels Fabra
- TGF-β and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08907 Barcelona, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBEREHD), ISCIII, 28029 Madrid, Spain
| |
Collapse
|
19
|
Lin Y, Ramelot TA, Senyuz S, Gursoy A, Jang H, Nussinov R, Keskin O, Zheng Y. Tumor-derived RHOA mutants interact with effectors in the GDP-bound state. Nat Commun 2024; 15:7176. [PMID: 39169042 PMCID: PMC11339415 DOI: 10.1038/s41467-024-51445-z] [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: 07/26/2023] [Accepted: 08/07/2024] [Indexed: 08/23/2024] Open
Abstract
RHOA mutations are found at diverse residues in various cancer types, implying mutation- and cell-specific mechanisms of tumorigenesis. Here, we focus on the underlying mechanisms of two gain-of-function RHOA mutations, A161P and A161V, identified in adult T-cell leukemia/lymphoma. We find that RHOAA161P and RHOAA161V are both fast-cycling mutants with increased guanine nucleotide dissociation/association rates compared with RHOAWT and show reduced GTP-hydrolysis activity. Crystal structures reveal an altered nucleotide association in RHOAA161P and an open nucleotide pocket in RHOAA161V. Both mutations perturb the dynamic properties of RHOA switch regions and shift the conformational landscape important for RHOA activity, as shown by 31P NMR and molecular dynamics simulations. Interestingly, RHOAA161P and RHOAA161V can interact with effectors in the GDP-bound state. 1H-15N HSQC NMR spectra support the existence of an active population in RHOAA161V-GDP. The distinct interaction mechanisms resulting from the mutations likely favor an RHOAWT-like "ON" conformation, endowing GDP-bound state effector binding activity.
Collapse
Affiliation(s)
- Yuan Lin
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| | - Theresa A Ramelot
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY, USA
| | - Simge Senyuz
- Computational Sciences and Engineering, Koc University, Rumelifeneri Yolu, Istanbul, Turkey
| | - Attila Gursoy
- Department of Computer Engineering, Koc Univeristy, Rumelifeneri Yolu, Istanbul, Turkey
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, MD, USA
| | - Ozlem Keskin
- Department of Chemical and Biological Engineering, Koc Univeristy, Rumelifeneri Yolu, Istanbul, Turkey
| | - Yi Zheng
- Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
20
|
Bannoura SF, Khan HY, Uddin MH, Mohammad RM, Pasche BC, Azmi AS. Targeting guanine nucleotide exchange factors for novel cancer drug discovery. Expert Opin Drug Discov 2024; 19:949-959. [PMID: 38884380 PMCID: PMC11380440 DOI: 10.1080/17460441.2024.2368242] [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: 02/28/2024] [Accepted: 06/10/2024] [Indexed: 06/18/2024]
Abstract
INTRODUCTION Guanine nucleotide exchange factors (GEFs) regulate the activation of small GTPases (G proteins) of the Ras superfamily proteins controlling cellular functions. Ras superfamily proteins act as 'molecular switches' that are turned 'ON' by guanine exchange. There are five major groups of Ras family GTPases: Ras, Ran, Rho, Rab and Arf, with a variety of different GEFs regulating their GTP loading. GEFs have been implicated in various diseases including cancer. This makes GEFs attractive targets to modulate signaling networks controlled by small GTPases. AREAS COVERED In this review, the roles and mechanisms of GEFs in malignancy are outlined. The mechanism of guanine exchange activity by GEFs on a small GTPase is illustrated. Then, some examples of GEFs that are significant in cancer are presented with a discussion on recent progress in therapeutic targeting efforts using a variety of approaches. EXPERT OPINION Recently, GEFs have emerged as potential therapeutic targets for novel cancer drug development. Targeting small GTPases is challenging; thus, targeting their activation by GEFs is a promising strategy. Most GEF-targeted drugs are still in preclinical development. A deeper biological understanding of the underlying mechanisms of GEF activity and utilizing advanced technology are necessary to enhance drug discovery for GEFs in cancer.
Collapse
Affiliation(s)
- Sahar F Bannoura
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Husain Yar Khan
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Md Hafiz Uddin
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Ramzi M Mohammad
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Boris C Pasche
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - Asfar S Azmi
- Department of Oncology, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
21
|
DeBenedictis JN, Murrell C, Hauser D, van Herwijnen M, Elen B, de Kok TM, van Breda SG. Effects of Different Combinations of Phytochemical-Rich Fruits and Vegetables on Chronic Disease Risk Markers and Gene Expression Changes: Insights from the MiBLEND Study, a Randomized Trial. Antioxidants (Basel) 2024; 13:915. [PMID: 39199161 PMCID: PMC11351619 DOI: 10.3390/antiox13080915] [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: 05/29/2024] [Revised: 07/13/2024] [Accepted: 07/19/2024] [Indexed: 09/01/2024] Open
Abstract
Adequate fruit and vegetable (F and V) intake, as recommended by the World Health Organization (over 400 g/day), is linked to reduced chronic disease risk. However, human intervention trials, especially with whole F and V and in complex combinations, are lacking. The MiBlend Study explored the effects of various phytochemical-rich F and V combinations on chronic disease risk markers, phytochemical absorption, and gene expression in blood. This randomized cross-over study involved participants consuming two of seven different F and V blends for 2 weeks (450 g/day), following a 2-week low F and V intake period (50 g/day). Each blend represented major phytochemical classes (flavonoids, anthocyanins, carotenoids, and glucosinolates) or combinations thereof. Markers of chronic disease risk, including DNA damage, oxidative stress, and retinal microvasculature, were measured. Increasing F and V intake significantly improved plasma antioxidant capacity, DNA damage protection, and retinal arteriolar dilation. Flavonoid-rich, carotenoid-rich, and complex blends notably reduced DNA damage susceptibility. Anthocyanin-rich and carotenoid-rich interventions were most effective in boosting antioxidant capacity, while blends high in flavonoids, especially combined with anthocyanins, significantly improved retinal microvasculature. Gene expression analysis revealed changes in DNA repair, signal transduction, and transcription processes, indicating mechanisms for these health benefits. The study suggests specific F and V blends can provide targeted health improvements, emphasizing the importance of both overall F and V intake and the specific phytochemical composition for personalized preventive strategies.
Collapse
Affiliation(s)
- Julia N. DeBenedictis
- Toxicogenomics Department, GROW School of Oncology & Reproduction, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Courtney Murrell
- Toxicogenomics Department, GROW School of Oncology & Reproduction, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Duncan Hauser
- Toxicogenomics Department, GROW School of Oncology & Reproduction, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Marcel van Herwijnen
- Toxicogenomics Department, GROW School of Oncology & Reproduction, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Bart Elen
- Flemish Institute for Technological Research (VITO), 2400 Mol, Belgium
| | - Theo M. de Kok
- Toxicogenomics Department, GROW School of Oncology & Reproduction, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Simone G. van Breda
- Toxicogenomics Department, GROW School of Oncology & Reproduction, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
22
|
Arici MK, Tuncbag N. Unveiling hidden connections in omics data via pyPARAGON: an integrative hybrid approach for disease network construction. Brief Bioinform 2024; 25:bbae399. [PMID: 39163205 PMCID: PMC11334722 DOI: 10.1093/bib/bbae399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 06/26/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024] Open
Abstract
Network inference or reconstruction algorithms play an integral role in successfully analyzing and identifying causal relationships between omics hits for detecting dysregulated and altered signaling components in various contexts, encompassing disease states and drug perturbations. However, accurate representation of signaling networks and identification of context-specific interactions within sparse omics datasets in complex interactomes pose significant challenges in integrative approaches. To address these challenges, we present pyPARAGON (PAgeRAnk-flux on Graphlet-guided network for multi-Omic data integratioN), a novel tool that combines network propagation with graphlets. pyPARAGON enhances accuracy and minimizes the inclusion of nonspecific interactions in signaling networks by utilizing network rather than relying on pairwise connections among proteins. Through comprehensive evaluations on benchmark signaling pathways, we demonstrate that pyPARAGON outperforms state-of-the-art approaches in node propagation and edge inference. Furthermore, pyPARAGON exhibits promising performance in discovering cancer driver networks. Notably, we demonstrate its utility in network-based stratification of patient tumors by integrating phosphoproteomic data from 105 breast cancer tumors with the interactome and demonstrating tumor-specific signaling pathways. Overall, pyPARAGON is a novel tool for analyzing and integrating multi-omic data in the context of signaling networks. pyPARAGON is available at https://github.com/netlab-ku/pyPARAGON.
Collapse
Affiliation(s)
- Muslum Kaan Arici
- Graduate School of Informatics, Middle East Technical University, Ankara 06800, Turkey
| | - Nurcan Tuncbag
- Chemical and Biological Engineering, College of Engineering, Koc University, Istanbul 34450, Turkey
- School of Medicine, Koc University, Istanbul 34450, Turkey
- Koc University Research Center for Translational Medicine (KUTTAM), Koc University, Istanbul 34450, Turkey
| |
Collapse
|
23
|
Guruvaiah P, Gupta R. IκBα kinase inhibitor BAY 11-7082 promotes anti-tumor effect in RAS-driven cancers. J Transl Med 2024; 22:642. [PMID: 38982514 PMCID: PMC11233160 DOI: 10.1186/s12967-024-05384-4] [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: 03/20/2024] [Accepted: 06/08/2024] [Indexed: 07/11/2024] Open
Abstract
BACKGROUND Oncogenic mutations in the RAS gene are associated with uncontrolled cell growth, a hallmark feature contributing to tumorigenesis. While diverse therapeutic strategies have been diligently applied to treat RAS-mutant cancers, successful targeting of the RAS gene remains a persistent challenge in the field of cancer therapy. In our study, we discover a promising avenue for addressing this challenge. METHODS In this study, we tested the viability of several cell lines carrying oncogenic NRAS, KRAS, and HRAS mutations upon treatment with IkappaBalpha (IκBα) inhibitor BAY 11-7082. We performed both cell culture-based viability assay and in vivo subcutaneous xenograft-based assay to confirm the growth inhibitory effect of BAY 11-7082. We also performed large RNA sequencing analysis to identify differentially regulated genes and pathways in the context of oncogenic NRAS, KRAS, and HRAS mutations upon treatment with BAY 11-7082. RESULTS We demonstrate that oncogenic NRAS, KRAS, and HRAS activate the expression of IκBα kinase. BAY 11-7082, an inhibitor of IκBα kinase, attenuates the growth of NRAS, KRAS, and HRAS mutant cancer cells in cell culture and in mouse model. Mechanistically, BAY 11-7082 inhibitor treatment leads to suppression of the PI3K-AKT signaling pathway and activation of apoptosis in all RAS mutant cell lines. Additionally, we find that BAY 11-7082 treatment results in the downregulation of different biological pathways depending upon the type of RAS protein that may also contribute to tumor growth inhibition. CONCLUSION Our study identifies BAY 11-7082 to be an efficacious inhibitor for treating RAS oncogene (HRAS, KRAS, and NRAS) mutant cancer cells. This finding provides new therapeutic opportunity for effective treatment of RAS-mutant cancers.
Collapse
Affiliation(s)
- Praveen Guruvaiah
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Romi Gupta
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
- O'Neal Comprehensive Cancer Center, The University of Alabama at Birmingham, Birmingham, AL, 35233, USA.
| |
Collapse
|
24
|
Xu N, Jiang J, Jiang F, Dong G, Meng L, Wang M, Chen J, Li C, Shi Y, He S, Li R. CircCDC42-encoded CDC42-165aa regulates macrophage pyroptosis in Klebsiella pneumoniae infection through Pyrin inflammasome activation. Nat Commun 2024; 15:5730. [PMID: 38977695 PMCID: PMC11231140 DOI: 10.1038/s41467-024-50154-x] [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: 08/29/2023] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
The circular RNA (circRNA) family is a group of endogenous non-coding RNAs (ncRNAs) that have critical functions in multiple physiological and pathological processes, including inflammation, cancer, and cardiovascular diseases. However, their roles in regulating innate immune responses remain unclear. Here, we define Cell division cycle 42 (CDC42)-165aa, a protein encoded by circRNA circCDC42, which is overexpressed in Klebsiella pneumoniae (KP)-infected alveolar macrophages. High levels of CDC42-165aa induces the hyperactivation of Pyrin inflammasomes and aggravates alveolar macrophage pyroptosis, while the inhibition of CDC42-165aa reduces lung injury in mice after KP infection by inhibiting Pyrin inflammasome-mediated pyroptosis. Overall, these results demonstrate that CDC42-165aa stimulates Pyrin inflammasome by inhibiting CDC42 GTPase activation and provides a potential clinical target for pathogenic bacterial infection in clinical practice.
Collapse
Affiliation(s)
- Nana Xu
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
- Laboratory of Morphology, Xuzhou Medical University, Xuzhou, China
| | - Jiebang Jiang
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Fei Jiang
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
- Department of Laboratory Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Guokai Dong
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
- Laboratory of Morphology, Xuzhou Medical University, Xuzhou, China
| | - Li Meng
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
- Laboratory of Morphology, Xuzhou Medical University, Xuzhou, China
| | - Meng Wang
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
- Laboratory of Morphology, Xuzhou Medical University, Xuzhou, China
| | - Jing Chen
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China
| | - Cong Li
- Xuzhou Key Laboratory of Emergency Medicine, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Yongping Shi
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China.
| | - Sisi He
- Department of Oncology, The Second Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Rongpeng Li
- Jiangsu Province Engineering Research Center of Cardiovascular Drugs Targeting Endothelial Cells, School of Life Sciences, Jiangsu Normal University, Xuzhou, China.
| |
Collapse
|
25
|
Brindani N, Vuong LM, La Serra MA, Salvador N, Menichetti A, Acquistapace IM, Ortega JA, Veronesi M, Bertozzi SM, Summa M, Girotto S, Bertorelli R, Armirotti A, Ganesan AK, De Vivo M. Discovery of CDC42 Inhibitors with a Favorable Pharmacokinetic Profile and Anticancer In Vivo Efficacy. J Med Chem 2024; 67:10401-10424. [PMID: 38866385 PMCID: PMC11215724 DOI: 10.1021/acs.jmedchem.4c00855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
We previously reported trisubstituted pyrimidine lead compounds, namely, ARN22089 and ARN25062, which block the interaction between CDC42 with its specific downstream effector, a PAK protein. This interaction is crucial for the progression of multiple tumor types. Such inhibitors showed anticancer efficacy in vivo. Here, we describe a second class of CDC42 inhibitors with favorable drug-like properties. Out of the 25 compounds here reported, compound 15 (ARN25499) stands out as the best lead compound with an improved pharmacokinetic profile, increased bioavailability, and efficacy in an in vivo PDX tumor mouse model. Our results indicate that these CDC42 inhibitors represent a promising chemical class toward the discovery of anticancer drugs, with ARN25499 as an additional lead candidate for preclinical development.
Collapse
Affiliation(s)
- Nicoletta Brindani
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Linh M. Vuong
- Department
of Dermatology, University of California, Irvine, California 92697, United States
| | - Maria Antonietta La Serra
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Noel Salvador
- Department
of Dermatology, University of California, Irvine, California 92697, United States
| | - Andrea Menichetti
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Isabella Maria Acquistapace
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Jose Antonio Ortega
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Marina Veronesi
- Structural
Biophysics Facility, Istituto Italiano di
Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Sine Mandrup Bertozzi
- Analytical
Chemistry Facility, Istituto Italiano di
Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Maria Summa
- Translational
Pharmacology Facility, Istituto Italiano
di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Stefania Girotto
- Structural
Biophysics Facility, Istituto Italiano di
Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Rosalia Bertorelli
- Translational
Pharmacology Facility, Istituto Italiano
di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Andrea Armirotti
- Analytical
Chemistry Facility, Istituto Italiano di
Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Anand K. Ganesan
- Department
of Dermatology, University of California, Irvine, California 92697, United States
| | - Marco De Vivo
- Molecular
Modeling and Drug Discovery Lab, Istituto
Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| |
Collapse
|
26
|
Liu W, Wang X, Yu H, Yan G, Shen S, Gao M, Zhang X. Integrated Platform for Large-Scale Quantitative Profiling of Phosphotyrosine Signaling Complexes Based on Cofractionation/Mass Spectrometry and Complex-Centric Algorithm. Anal Chem 2024; 96:9849-9858. [PMID: 38836774 DOI: 10.1021/acs.analchem.4c00285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
The scarcity and dynamic nature of phosphotyrosine (pTyr)-modified proteins pose a challenge for researching protein complexes with pTyr modification, which are assembled through multiple protein-protein interactions. We developed an integrated complex-centric platform for large-scale quantitative profiling of pTyr signaling complexes based on cofractionation/mass spectrometry (CoFrac-MS) and a complex-centric algorithm. We initially constructed a trifunctional probe based on pTyr superbinder (SH2-S) for specifically binding and isolation of intact pTyr protein complexes. Then, the CoFrac-MS strategy was employed for the identification of pTyr protein complexes by integrating ion exchange chromatography in conjunction with data independent acquisition mass spectrometry. Furthermore, we developed a novel complex-centric algorithm for quantifying protein complexes based on the protein complex elution curve. Utilizing this algorithm, we effectively quantified 216 putative protein complexes. We further screened 21 regulated pTyr protein complexes related to the epidermal growth factor signal. Our study engenders a comprehensive framework for the intricate examination of pTyr protein complexes and presents, for the foremost occasion, a quantitative landscape delineating the composition of pTyr protein complexes in HeLa cells.
Collapse
Affiliation(s)
- Wei Liu
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Xuantang Wang
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Hailong Yu
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Guoquan Yan
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Mingxia Gao
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Xiangmin Zhang
- Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
27
|
Zhang Y, Zhang H, Zhao S, Qi Z, He Y, Zhang X, Wu W, Yan K, Hu L, Sun S, Tang X, Zhou Q, Chen F, Gu A, Wang L, Zhang Z, Yu B, Wang D, Han Y, Xie L, Ji Y. S-Nitrosylation of Septin2 Exacerbates Aortic Aneurysm and Dissection by Coupling the TIAM1-RAC1 Axis in Macrophages. Circulation 2024; 149:1903-1920. [PMID: 38357802 DOI: 10.1161/circulationaha.123.066404] [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: 07/21/2023] [Accepted: 01/26/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND S-Nitrosylation (SNO), a prototypic redox-based posttranslational modification, is involved in cardiovascular disease. Aortic aneurysm and dissection are high-risk cardiovascular diseases without an effective cure. The aim of this study was to determine the role of SNO of Septin2 in macrophages in aortic aneurysm and dissection. METHODS Biotin-switch assay combined with liquid chromatography-tandem mass spectrometry was performed to identify the S-nitrosylated proteins in aortic tissue from both patients undergoing surgery for aortic dissection and Apoe-/- mice infused with angiotensin II. Angiotensin II-induced aortic aneurysm model and β-aminopropionitrile-induced aortic aneurysm and dissection model were used to determine the role of SNO of Septin2 (SNO-Septin2) in aortic aneurysm and dissection development. RNA-sequencing analysis was performed to recapitulate possible changes in the transcriptome profile of SNO-Septin2 in macrophages in aortic aneurysm and dissection. Liquid chromatography-tandem mass spectrometry and coimmunoprecipitation were used to uncover the TIAM1-RAC1 (Ras-related C3 botulinum toxin substrate 1) axis as the downstream target of SNO-Septin2. Both R-Ketorolac and NSC23766 treatments were used to inhibit the TIAM1-RAC1 axis. RESULTS Septin2 was identified S-nitrosylated at cysteine 111 (Cys111) in both aortic tissue from patients undergoing surgery for aortic dissection and Apoe-/- mice infused with Angiotensin II. SNO-Septin2 was demonstrated driving the development of aortic aneurysm and dissection. By RNA-sequencing, SNO-Septin2 in macrophages was demonstrated to exacerbate vascular inflammation and extracellular matrix degradation in aortic aneurysm. Next, TIAM1 (T lymphoma invasion and metastasis-inducing protein 1) was identified as a SNO-Septin2 target protein. Mechanistically, compared with unmodified Septin2, SNO-Septin2 reduced its interaction with TIAM1 and activated the TIAM1-RAC1 axis and consequent nuclear factor-κB signaling pathway, resulting in stronger inflammation and extracellular matrix degradation mediated by macrophages. Consistently, both R-Ketorolac and NSC23766 treatments protected against aortic aneurysm and dissection by inhibiting the TIAM1-RAC1 axis. CONCLUSIONS SNO-Septin2 drives aortic aneurysm and dissection through coupling the TIAM1-RAC1 axis in macrophages and activating the nuclear factor-κB signaling pathway-dependent inflammation and extracellular matrix degradation. Pharmacological blockade of RAC1 by R-Ketorolac or NSC23766 may therefore represent a potential treatment against aortic aneurysm and dissection.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Hao Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Shuang Zhao
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Zhenhua Qi
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Yiwei He
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Xuhong Zhang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Wencheng Wu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Ke Yan
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Lulu Hu
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Shixiu Sun
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
| | - Xinlong Tang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Institute of Cardiothoracic Vascular Disease, Nanjing University, China (X.T., Q.Z., D.W.)
| | - Qing Zhou
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Institute of Cardiothoracic Vascular Disease, Nanjing University, China (X.T., Q.Z., D.W.)
| | - Feng Chen
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
- Department of Forensic Medicine (F.C.), Nanjing Medical University, China
| | - Aihua Gu
- School of Public Health (A.G.), Nanjing Medical University, China
| | - Liansheng Wang
- Departments of Cardiology, First Affiliated Hospital of Nanjing Medical University, China (L.W.)
| | - Zhiren Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Heilongjiang, PR China (Z.Z., Y.J.)
| | - Bo Yu
- Department of Cardiology, Second Affiliated Hospital of Harbin Medical University, Key Laboratory of Myocardial Ischemia, Ministry of Education, Harbin Medical University, Heilongjiang, China (B.Y.)
| | - Dongjin Wang
- Department of Thoracic and Cardiovascular Surgery, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Institute of Cardiothoracic Vascular Disease, Nanjing University, China (X.T., Q.Z., D.W.)
| | - Yi Han
- Department of Geriatrics, the First Affiliated Hospital of Nanjing Medical University, China (Y.H.)
| | - Liping Xie
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
- Gusu School, Nanjing Medical University, Suzhou, China (L.X., Y.J.)
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine; Key Laboratory of Targeted Intervention of Cardiovascular Disease; Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Jiangsu, China (Y.Z., H.Z., S.Z., Z.Q., Y.H., X.Z., W.W., K.Y., L.H., S.S., F.C., L.X., Y.J.)
- Gusu School, Nanjing Medical University, Suzhou, China (L.X., Y.J.)
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Harbin Medical University, Heilongjiang, PR China (Z.Z., Y.J.)
| |
Collapse
|
28
|
Liang B, Wu Q, Wang Y, Shi Y, Sun F, Huang Q, Li G, Liu Y, Zhang S, Xu X, Yao G, Peng J, Zhai X, Wu J, Tan Y, Wu Z, Zhou R, Li S, Wu J, Yang M, Liao W, Shi M. Cdc42-driven endosomal cholesterol transport promotes collateral resistance in HER2-positive gastric cancer. Cancer Lett 2024; 587:216702. [PMID: 38336288 DOI: 10.1016/j.canlet.2024.216702] [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: 09/08/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Resistance to trastuzumab and the poor efficacy of subsequent chemotherapy have become major challenges for HER2-positive gastric cancer (GC). As resistance evolves, tumor cells may acquire a new drug susceptibility profile, profoundly impacting the subsequent treatment selection and patient survival. However, the interplay between trastuzumab and other types of drugs in HER2-positive GC remains elusive. In our study, we utilized resistant cell lines and tissue specimens to map the drug susceptibility profile of trastuzumab-resistant GC, discovering that resistance to trastuzumab induces collateral resistance to commonly used chemotherapeutic agents. Additionally, patients with collateral resistance distinguished by a 13-gene scoring model in HER2-positive GC cohorts are predicted to have a poor prognosis and may be sensitive to cholesterol-lowering drugs. Mechanistically, endosomal cholesterol transport is further confirmed to enrich cholesterol in the plasma membrane, contributing to collateral resistance through the Hedgehog-ABCB1 axis. As a driver for cholesterol, Cdc42 is activated by the formation of the NPC1-TβRI-Cdc42 complex to facilitate endosomal cholesterol transport. We demonstrated that inhibiting Cdc42 activation with ZCL278 reduces cholesterol levels in the plasma membrane and reverses collateral resistance between trastuzumab and chemotherapy in vitro and in vivo. Collectively, our findings verify the phenomena and mechanism of collateral resistance between trastuzumab and chemotherapy, and propose a potential therapeutic target and strategy in the second-line treatment for trastuzumab-resistant HER2-positive GC.
Collapse
Affiliation(s)
- Bishan Liang
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Qijing Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Yawen Wang
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Yulu Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Fei Sun
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Qiong Huang
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Guanjun Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Yajing Liu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical Oncology, Breast Tumor Center, Phase I Clinical Trial Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 510060, Guangzhou, China
| | - Shuyi Zhang
- Department of Oncology, Huizhou Municipal Central Hospital, 516008, Huizhou, Guangdong, China
| | - Xin Xu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Guangyu Yao
- Department of General Surgery, Breast Center, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Jianjun Peng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, 510060, Guangzhou, Guangdong, China
| | - Xiaohui Zhai
- Department of Medical Oncology, The Sixth Affiliated Hospital of Sun Yat-sen University, 510655, Guangzhou, Guangdong, China
| | - Jing Wu
- Department of Oncology, The People's Hospital of Foshan, 528010, Foshan, Guangdong, China
| | - Yujing Tan
- Department of Radiation Oncology, Zhujiang Hospital, Southern Medical University, 510280, Guangzhou, Guangdong, China
| | - Zhenzhen Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Rui Zhou
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Shaowei Li
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Jianhua Wu
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Ming Yang
- Shandong Provincial Key Laboratory of Radiation Oncology, Cancer Research Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, 271016, Jinan, Shandong, China
| | - Wangjun Liao
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China
| | - Min Shi
- Department of Oncology, Nanfang Hospital, Southern Medical University, 510515, Guangzhou, Guangdong, China.
| |
Collapse
|
29
|
Karcini A, Mercier NR, Lazar IM. Proteomic Assessment of SKBR3/HER2+ Breast Cancer Cellular Response to Lapatinib and Investigational Ipatasertib Kinase Inhibitors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.02.587656. [PMID: 38617302 PMCID: PMC11014527 DOI: 10.1101/2024.04.02.587656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Modern cancer treatment approaches aim at achieving cancer remission by using targeted and personalized therapies, as well as harnessing the power of the immune system to recognize and eliminate the cancer cells. To overcome a relatively short-lived response due to the development of resistance to the administered drugs, combination therapies have been pursued, as well. To expand the outlook of combination therapies, the objective of this study was to use high-throughput data generation technologies such as mass spectrometry and proteomics to investigate the response of HER2+ breast cancer cells to a mixture of two kinase inhibitors that has not been adopted yet as a standard treatment regime. The broader landscape of biological processes that are affected by inhibiting two major pathways that sustain the growth and survival of cancer cells, i.e., EGFR and PI3K/AKT, was investigated by treating SKBR3/HER2+ breast cancer cells with Lapatinib or a mixture of Lapatinib/Ipatasertib small molecule drugs. Changes in protein expression and/or activity in response to the drug treatments were assessed by using two complementary quantitative proteomic approaches based on peak area and peptide spectrum match measurements. Over 900 proteins matched by three unique peptide sequences (FDR<0.05) were affected by the exposure of cells to the drugs. The work corroborated the anti-proliferative activity of Lapatinib and Ipatasertib, and, in addition to cell cycle and growth arrest processes enabled the identification of several multi-functional proteins with roles in cancer-supportive hallmark processes. Among these, immune response, adhesion and migration emerged as particularly relevant to the ability to effectively suppress the proliferation and dissemination of cancer cells. The supplementation of Lapatinib with Ipatasertib further affected the expression or activity of additional transcription factors and proteins involved in gene expression, trafficking, DNA repair, and development of multidrug resistance. Furthermore, over fifty of the affected proteins represented approved or investigational targets in the DrugBank database, which through their protein-protein interaction networks can inform the selection of effective therapeutic partners. Altogether, our findings exposed a broad plethora of yet untapped opportunities that can be further explored for enhancing the anti-cancer effects of each drug as well as of many other multi-drug therapies that target the EGFR/ERBB2 and PI3K/AKT pathways. The data are available via ProteomeXchange with identifier PXD051094.
Collapse
Affiliation(s)
- Arba Karcini
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060; Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA 24061
| | - Nicole R. Mercier
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060; Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA 24061
| | - Iulia M. Lazar
- Department of Biological Sciences, Virginia Tech, Blacksburg, VA 24060; Department of Biological Sciences, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA 24061
- Fralin Life Sciences Institute, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA 24061
- Division of Systems Biology, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA 24061
- Carilion School of Medicine, Virginia Tech, 1981 Kraft Drive, Blacksburg, VA 24061
| |
Collapse
|
30
|
Mikhael S, Kurdi A, Khoueiry-Zgheib N, Tahtouh R, Nasr R, Hilal G. Evaluating synergistic effects of metformin and simvastatin on ovarian cancer cells. PLoS One 2024; 19:e0298127. [PMID: 38489280 PMCID: PMC10942021 DOI: 10.1371/journal.pone.0298127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 01/19/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Ovarian Cancer (OC) stands as the most lethal gynecological malignancy, presenting an urgent clinical challenge in the quest to improve response rates. One approach to address this challenge is through drug repurposing, exemplified by the investigation of metabolic-modulating drugs such as Metformin (MTF) and Simvastatin (SIM). This study aims to explore the molecular mechanisms contributing to the potential synergistic anti-cancer effects between MTF and SIM on ovarian cancer cells. METHODS We assessed the effects of the combination on the proliferation and viability of two cell lines OVCAR-3 and SKOV-3. IC50 concentrations of MTF and SIM were determined using a proliferation assay, followed by subtoxic concentrations to explore the potential synergistic effects on the viability of both cell lines. Transcriptomic analysis was conducted on OVCAR-3 treated cells, and the findings were validated by assessing the expression levels of differentially expressed genes (DEGs) through real-time PCR in both cell lines SK-OV-3 and OVCAR-3. RESULTS Cytotoxicity analysis guided the selection of treatment concentrations as such MTF 10 mM and SIM 5 μM. The combined treatment of MTF and SIM demonstrated a synergistic inhibition of proliferation and viability in both cell lines. In OVCAR-3, exclusive identification of 507 DEGs was seen in the combination arm. Upregulation of FOXO3, RhoA, and TNFα, along with downregulation of PIK3R1, SKP2, and ATP6V1D levels, was observed in OVCAR-3 treated cells. Real-time PCR validation confirmed the consistency of expression levels for the mentioned DEGs. CONCLUSION Our data strongly supports the presence of synergy between MTF and SIM in OC cells. The combination's effect is associated with the dysregulation of genes in the key regulators AMPK and mTOR alongside other interconnected pathways.
Collapse
Affiliation(s)
- Sara Mikhael
- Laboratory of Cancer and Metabolism, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Abdullah Kurdi
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nathalie Khoueiry-Zgheib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Roula Tahtouh
- Laboratory of Cancer and Metabolism, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| | - Rihab Nasr
- Department of Anatomy, Cell Biology, and Physiological Sciences, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - George Hilal
- Laboratory of Cancer and Metabolism, Faculty of Medicine, Saint-Joseph University, Beirut, Lebanon
| |
Collapse
|
31
|
Ravichandran Y, Hänisch J, Murray K, Roca V, Dingli F, Loew D, Sabatet V, Boëda B, Stradal TE, Etienne-Manneville S. The distinct localization of CDC42 isoforms is responsible for their specific functions during migration. J Cell Biol 2024; 223:e202004092. [PMID: 38386112 PMCID: PMC10883850 DOI: 10.1083/jcb.202004092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/26/2023] [Accepted: 01/04/2024] [Indexed: 02/23/2024] Open
Abstract
The small G-protein CDC42 is an evolutionary conserved polarity protein and a key regulator of polarized cell functions, including directed cell migration. In vertebrates, alternative splicing gives rise to two CDC42 proteins: the ubiquitously expressed isoform (CDC42u) and the brain isoform (CDC42b), which only differ in their carboxy-terminal sequence, including the CAAX motif essential for their association with membranes. We show that these divergent sequences do not directly affect the range of CDC42's potential binding partners but indirectly influence CDC42-driven signaling by controlling the subcellular localization of the two isoforms. In astrocytes and neural precursors, which naturally express both variants, CDC42u associates with the leading-edge plasma membrane of migrating cells, where it recruits the Par6-PKCζ complex to fulfill its polarity function. In contrast, CDC42b mainly localizes to intracellular membrane compartments, where it regulates N-WASP-mediated endocytosis. Both CDC42 isoforms contribute their specific functions to promote the chemotaxis of neural precursors, demonstrating that their expression pattern is decisive for tissue-specific cell behavior.
Collapse
Affiliation(s)
- Yamini Ravichandran
- UMR3691 CNRS, Equipe Labellisée Ligue 2023, Université de Paris, Cell Polarity, Migration and Cancer Unit, Institut Pasteur, Paris, France
- Collège Doctoral, Sorbonne Université, Paris, France
| | - Jan Hänisch
- UMR3691 CNRS, Equipe Labellisée Ligue 2023, Université de Paris, Cell Polarity, Migration and Cancer Unit, Institut Pasteur, Paris, France
| | - Kerren Murray
- UMR3691 CNRS, Equipe Labellisée Ligue 2023, Université de Paris, Cell Polarity, Migration and Cancer Unit, Institut Pasteur, Paris, France
| | - Vanessa Roca
- UMR3691 CNRS, Equipe Labellisée Ligue 2023, Université de Paris, Cell Polarity, Migration and Cancer Unit, Institut Pasteur, Paris, France
| | - Florent Dingli
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | - Damarys Loew
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | - Valentin Sabatet
- PSL Research University, Centre de Recherche, Laboratoire de Spectrométrie de Masse Protéomique, Institut Curie, Paris, France
| | - Batiste Boëda
- UMR3691 CNRS, Equipe Labellisée Ligue 2023, Université de Paris, Cell Polarity, Migration and Cancer Unit, Institut Pasteur, Paris, France
| | - Theresia E. Stradal
- Helmholtz Centre for Infection Research, Inhoffenstrasse 7, Braunschweig, Germany
| | - Sandrine Etienne-Manneville
- UMR3691 CNRS, Equipe Labellisée Ligue 2023, Université de Paris, Cell Polarity, Migration and Cancer Unit, Institut Pasteur, Paris, France
| |
Collapse
|
32
|
Akompong SK, Li Y, Gong W, Ye L, Liu J. Recently reported cell migration inhibitors: Opportunities and challenges for antimetastatic agents. Drug Discov Today 2024; 29:103906. [PMID: 38309689 DOI: 10.1016/j.drudis.2024.103906] [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: 09/25/2023] [Revised: 01/17/2024] [Accepted: 01/25/2024] [Indexed: 02/05/2024]
Abstract
Antimetastatic agents are highly desirable for cancer treatment because of the severe medical challenges and high mortality resulting from tumor metastasis. Having demonstrated antimetastatic effects in numerous in vitro and in vivo studies, migration inhibitors present significant opportunities for developing a new class of anticancer drugs. To provide a useful overview on the latest research in migration inhibitors, this article first discusses their therapeutic significance, targetable proteins, and developmental avenues. Subsequently it reviews over 20 representative migration inhibitors reported in recent journals in terms of their inhibitory mechanism, potency, and potential clinical utility. The relevance of the target proteins to cellular migratory function is focused on as it is crucial for assessing the overall efficacy of the inhibitors.
Collapse
Affiliation(s)
- Samuel K Akompong
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Yang Li
- Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Wenxue Gong
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China
| | - Long Ye
- School of Chemistry and Chemical Engineering, Wuhan University of Science and Technology, Wuhan 430081, China.
| | - Jinping Liu
- Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
33
|
Woo S, Strasser L. Atypical RhoUV GTPases in development and disease. Biochem Soc Trans 2024; 52:89-97. [PMID: 38314621 PMCID: PMC10903452 DOI: 10.1042/bst20230212] [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: 10/24/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/06/2024]
Abstract
RhoU and RhoV are members of the Rho family of small GTPases that comprise their own subfamily. RhoUV GTPases are classified as atypical due to the kinetics of their GTP/GDP binding cycles. They also possess unique N- and C-termini that regulate their subcellular localization and activity. RhoU and RhoV have been linked to cytoskeletal regulation, cell adhesion, and cell migration. They each exhibit distinct expression patterns during embryonic development and diseases such as cancer metastasis, suggesting they have specialized functions. In this review, we will discuss the known functions of RhoU and RhoV, with a focus on their roles in early development, organogenesis, and disease.
Collapse
Affiliation(s)
- Stephanie Woo
- Department of Molecular Cell Biology, University of California, Merced, CA, U.S.A
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, U.S.A
| | - Leesa Strasser
- Department of Molecular Cell Biology, University of California, Merced, CA, U.S.A
- Quantitative and Systems Biology Graduate Program, University of California, Merced, CA, U.S.A
| |
Collapse
|
34
|
Patyal P, Zhang X, Verma A, Azhar G, Wei JY. Inhibitors of Rho/MRTF/SRF Transcription Pathway Regulate Mitochondrial Function. Cells 2024; 13:392. [PMID: 38474356 PMCID: PMC10931493 DOI: 10.3390/cells13050392] [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: 12/30/2023] [Revised: 02/19/2024] [Accepted: 02/20/2024] [Indexed: 03/14/2024] Open
Abstract
RhoA-regulated gene transcription by serum response factor (SRF) and its transcriptional cofactor myocardin-related transcription factors (MRTFs) signaling pathway has emerged as a promising therapeutic target for pharmacological intervention in multiple diseases. Altered mitochondrial metabolism is one of the major hallmarks of cancer, therefore, this upregulation is a vulnerability that can be targeted with Rho/MRTF/SRF inhibitors. Recent advances identified a novel series of oxadiazole-thioether compounds that disrupt the SRF transcription, however, the direct molecular target of these compounds is unclear. Herein, we demonstrate the Rho/MRTF/SRF inhibition mechanism of CCG-203971 and CCG-232601 in normal cell lines of human lung fibroblasts and mouse myoblasts. Further studies investigated the role of these molecules in targeting mitochondrial function. We have shown that these molecules hyperacetylate histone H4K12 and H4K16 and regulate the genes involved in mitochondrial function and dynamics. These small molecule inhibitors regulate mitochondrial function as a compensatory mechanism by repressing oxidative phosphorylation and increasing glycolysis. Our data suggest that these CCG molecules are effective in inhibiting all the complexes of mitochondrial electron transport chains and further inducing oxidative stress. Therefore, our present findings highlight the therapeutic potential of CCG-203971 and CCG-232601, which may prove to be a promising approach to target aberrant bioenergetics.
Collapse
Affiliation(s)
| | | | | | | | - Jeanne Y. Wei
- Donald W. Reynolds Department of Geriatrics and Institute on Aging, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (P.P.); (X.Z.); (A.V.); (G.A.)
| |
Collapse
|
35
|
Kopsida M, Liu N, Kotti A, Wang J, Jensen L, Jothimani G, Hildesjo C, Haapaniemi S, Zhong W, Pathak S, Sun XF. RhoB expression associated with chemotherapy response and prognosis in colorectal cancer. Cancer Cell Int 2024; 24:75. [PMID: 38355625 PMCID: PMC10867990 DOI: 10.1186/s12935-024-03236-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 01/20/2024] [Indexed: 02/16/2024] Open
Abstract
PURPOSE To examine the role of RhoB expression in relation to chemotherapy response, clinical outcomes and associated signaling pathways in colorectal cancer patients. MATERIALS AND METHODS The study included 5 colon cancer cell lines, zebrafish embryos and 260 colorectal cancer patients treated with 5-fluorouracil (5-FU) and oxaliplatin (OXL). The methods consisted of CRISPR/Cas9, reactive oxygen species (ROS), caspase-3 activity, autophagy flux, in-silico RNA sequencing and immunohistochemistry. Gene expression analysis and pathway analysis were conducted using RNA-seq data. RESULTS All cancer lines tested, including SW480, SW480-KO13 (RhoB knockout), SW480-KO55 (RhoB knockout), HCT116 and HCT116-OE (RhoB overexpressed), exhibited cytotoxicity to 5-FU and OXL. RhoB knockout cell lines demonstrated significantly reduced migration compared to the control cell lines. Furthermore, RhoB played a role in caspase-3-dependent apoptosis, regulation of ROS production and autophagic flux. The mRNA sequencing data indicated lower expression levels of oncogenes in RhoB knockout cell lines. The zebrafish model bearing SW480-KO showed a light trend toward tumor regression. RhoB expression by immunohistochemistry in patients was increased from normal mucosa to tumor samples. In patients who received chemotherapy, high RhoB expression was related to worse survival compared to low RhoB expression. Furthermore, the molecular docking analysis revealed that OXL had a higher binding affinity for RhoB than 5-FU, with a binding affinity of -7.8 kcal/mol and HADDOCK predicted molecular interactions between RhoB and caspase 3 protein. Gene-set enrichment analysis supported these findings, showing that enrichment of DNA damage response pathway and p53 signaling in RhoB overexpression treatment group, while the RhoB knockout treatment group exhibited enrichment in the negative regulation pathway of cell migration. CONCLUSION RhoB was negatively associated with chemotherapy response and survival in colorectal cancers. Therefore, RhoB inhibition may enhance chemotherapeutic responses and patient survival.
Collapse
Affiliation(s)
- Maria Kopsida
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Na Liu
- Department of Gastroenterology, Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Angeliki Kotti
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Jing Wang
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lasse Jensen
- Department of Medical and Health Sciences, Linköping University, Linköping, Sweden
| | - Ganesan Jothimani
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India
| | - Camilla Hildesjo
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Staffan Haapaniemi
- Department of Surgery and Department of Biomedical and Clinical Sciences, Linköping University, Norrköping, Sweden
| | - Wen Zhong
- Science for Life Laboratory, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Surajit Pathak
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Tamil Nadu, India.
| | - Xiao-Feng Sun
- Department of Oncology and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.
| |
Collapse
|
36
|
Sanchez C, Ramirez A, Hodgson L. Unravelling molecular dynamics in living cells: Fluorescent protein biosensors for cell biology. J Microsc 2024. [PMID: 38357769 PMCID: PMC11324865 DOI: 10.1111/jmi.13270] [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: 10/16/2023] [Revised: 01/11/2024] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Genetically encoded, fluorescent protein (FP)-based Förster resonance energy transfer (FRET) biosensors are microscopy imaging tools tailored for the precise monitoring and detection of molecular dynamics within subcellular microenvironments. They are characterised by their ability to provide an outstanding combination of spatial and temporal resolutions in live-cell microscopy. In this review, we begin by tracing back on the historical development of genetically encoded FP labelling for detection in live cells, which lead us to the development of early biosensors and finally to the engineering of single-chain FRET-based biosensors that have become the state-of-the-art today. Ultimately, this review delves into the fundamental principles of FRET and the design strategies underpinning FRET-based biosensors, discusses their diverse applications and addresses the distinct challenges associated with their implementation. We place particular emphasis on single-chain FRET biosensors for the Rho family of guanosine triphosphate hydrolases (GTPases), pointing to their historical role in driving our understanding of the molecular dynamics of this important class of signalling proteins and revealing the intricate relationships and regulatory mechanisms that comprise Rho GTPase biology in living cells.
Collapse
Affiliation(s)
- Colline Sanchez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Andrea Ramirez
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Louis Hodgson
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, New York, USA
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
37
|
Haspel N, Jang H, Nussinov R. Allosteric Activation of RhoA Complexed with p115-RhoGEF Deciphered by Conformational Dynamics. J Chem Inf Model 2024; 64:862-873. [PMID: 38215280 DOI: 10.1021/acs.jcim.3c01412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2024]
Abstract
The Ras homologue family member A (RhoA) is a member of the Rho family, a subgroup of the Ras superfamily. RhoA interacts with the 115 kDa guanine nucleotide exchange factor (p115-RhoGEF), which assists in activation and binding with downstream effectors. Here, we use molecular dynamics (MD) simulations and essential dynamics analysis of the inactive RhoA-GDP and active RhoA-GTP, when bound to p115-RhoGEF to decipher the mechanism of RhoA activation at the structural level. We observe that inactive RhoA-GDP maintains its position near the catalytic site on the Dbl homology (DH) domain of p115-RhoGEF through the interaction of its Switch I region with the DH domain. We further show that the active RhoA-GTP is engaged in more interactions with the p115-RhoGEF membrane-bound Pleckstrin homology (PH) domain as compared to RhoA-GDP. We hypothesize that the role of the interactions between the active RhoA-GTP and the PH domain is to help release it from the DH domain upon activation. Our results support this premise, and our simulations uncover the beginning of this process and provide structural details. They also point to allosteric communication pathways that take part in RhoA activation to promote and strengthen the interaction between the active RhoA-GTP and the PH domain. Allosteric regulation also occurs among other members of the Rho superfamily. Collectively, we suggest that in the activation process, the role of the RhoA-GTP interaction with the PH domain is to release RhoA-GTP from the DH domain after activation, making it available to downstream effectors.
Collapse
Affiliation(s)
- Nurit Haspel
- Department of Computer Science, University of Massachusetts Boston, Boston, Massachusetts 02125, United States
| | - Hyunbum Jang
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
| | - Ruth Nussinov
- Computational Structural Biology Section, Frederick National Laboratory for Cancer Research in the Cancer Innovation Laboratory, National Cancer Institute, Frederick, Maryland 21702, United States
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
38
|
Kant R, Mishra N, Kandhari K, Saba L, Michel C, Reisdorph R, Tewari-Singh N, Pantcheva MB, Petrash JM, Agarwal C, Agarwal R. Dexamethasone targets actin cytoskeleton signaling and inflammatory mediators to reverse sulfur mustard-induced toxicity in rabbit corneas. Toxicol Appl Pharmacol 2024; 483:116834. [PMID: 38266871 PMCID: PMC10923037 DOI: 10.1016/j.taap.2024.116834] [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: 11/27/2023] [Revised: 01/17/2024] [Accepted: 01/21/2024] [Indexed: 01/26/2024]
Abstract
PURPOSE Sulfur mustard (SM), a bi-functional alkylating agent, was used during World War I and the Iran-Iraq war. SM toxicity is ten times higher in eyes than in other tissues. Cornea is exceptionally susceptible to SM-injuries due to its anterior positioning and mucous-aqueous interphase. Ocular SM exposure induces blepharitis, photosensitivity, dry eye, epithelial defects, limbal ischemia and stem cell deficiency, and mustard gas keratopathy leading to temporary or permanent vision impairments. We demonstrated that dexamethasone (Dex) is a potent therapeutic intervention against SM-induced corneal injuries; however, its mechanism of action is not well known. Investigations employing proteomic profiling (LC-MS/MS) to understand molecular mechanisms behind SM-induced corneal injury and Dex efficacy were performed in the rabbit cornea exposed to SM and then received Dex treatment. PEAKS studio was used to extract, search, and summarize peptide identity. Ingenuity Pathway Analysis was used for pathway identification. Validation was performed using immunofluorescence. One-Way ANOVA (FDR < 0.05; p < 0.005) and Student's t-test (p < 0.05) were utilized for analyzing proteomics and IF data, respectively. Proteomic analysis revealed that SM-exposure upregulated tissue repair pathways, particularly actin cytoskeleton signaling and inflammation. Prominently dysregulated proteins included lipocalin2, coronin1A, actin-related protein2, actin-related protein2/3 complex subunit2, actin-related protein2/3 complex subunit4, cell division cycle42, ezrin, bradykinin/kininogen1, moesin, and profilin. Upregulated actin cytoskeleton signaling increases F-actin formation, dysregulating cell shape and motility. Dex reversed SM-induced increases in the aforementioned proteins levels to near control expression profiles. Dex aids corneal wound healing and improves corneal integrity via actin cytoskeletal signaling and anti-inflammatory effects following SM-induced injuries.
Collapse
Affiliation(s)
- Rama Kant
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Neha Mishra
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Kushal Kandhari
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Laura Saba
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Cole Michel
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Richard Reisdorph
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Neera Tewari-Singh
- Department of Pharmacology and Toxicology, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Mina B Pantcheva
- Department of Ophthalmology, School of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - J Mark Petrash
- Department of Ophthalmology, School of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Chapla Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA
| | - Rajesh Agarwal
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado-Anschutz Medical Campus, Aurora, CO, USA.
| |
Collapse
|
39
|
Xu Y, Liu X, Cao J, Wu Y, Jiang Q, Luo B. Rho GTPase-activating protein 1 promotes hepatocellular carcinoma progression via modulation by CircPIP5K1A/MiR-101-3p. Hepatol Res 2024; 54:174-188. [PMID: 37792600 DOI: 10.1111/hepr.13972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 09/04/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
AIM There has been an increased focus on regulating cell function with Rho family GTPases, including proliferation, migration/invasion, polarity, and adhesion. Due to the challenges involved in targeting Rho family GTPases directly, it may be more effective to target their regulators, such as Rho GTPase-activating protein 1 (ARHGAP1). This present research was performed to define the clinical significance of ARHGAP1 expression, as well as its regulatory mechanisms in hepatocellular carcinoma. METHODS ARHGAP1 and miR-101-3p expression of liver cancer patients, and their relevance with clinicopathological characteristics and prognosis were analyzed by the Cancer Genome Atlas sequencing data, and verified using samples of hepatocellular carcinoma patients. The interactions between miR-101-3p and ARHGAP1 or circPIP5K1A were validated by bioinformatic analyses, as well as confirmed by quantitative reverse transcription polymerase chain reaction, western blotting, and dual-luciferase reporter analysis. Plate clonality assays, cell adhesion and migration experiments, and proliferation experiments were used for assessing the participation of the circPIP5K1A/miR-101-3p/ARHGAP1 pathway in cell proliferation and motility. RESULTS Elevated ARHGAP1 and reduced miR-101-3p expression are related to poorer survival. MiR-101-3p targets ARHGAP1 to suppress hepatocellular carcinoma cell colony formation and invasion, whereas miR-101-3p inhibitor reverses liver cancer proliferation and metastasis suppression caused by ARHGAP1 knockdown. In addition, circPIP5K1A, which is mainly distributed in the cytosol, showed carcinogenic effects by sponging miR-101-3p, thus regulating ARHGAP1 expression. CONCLUSIONS ARHGAP1 serves as an oncogenic gene in liver cancer, and the expression thereof is regulated by circPIP5K1A through sponging miR-101-3p.
Collapse
Affiliation(s)
- Yanni Xu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Xiaodi Liu
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Jincheng Cao
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Ye Wu
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Qiongchao Jiang
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Baoming Luo
- Department of Ultrasound, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
40
|
Klein DC, Lardo SM, Hainer SJ. The ncBAF Complex Regulates Transcription in AML Through H3K27ac Sensing by BRD9. CANCER RESEARCH COMMUNICATIONS 2024; 4:237-252. [PMID: 38126767 PMCID: PMC10831031 DOI: 10.1158/2767-9764.crc-23-0382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 11/02/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
The non-canonical BAF complex (ncBAF) subunit BRD9 is essential for acute myeloid leukemia (AML) cell viability but has an unclear role in leukemogenesis. Because BRD9 is required for ncBAF complex assembly through its DUF3512 domain, precise bromodomain inhibition is necessary to parse the role of BRD9 as a transcriptional regulator from that of a scaffolding protein. To understand the role of BRD9 bromodomain function in regulating AML, we selected a panel of five AML cell lines with distinct driver mutations, disease classifications, and genomic aberrations and subjected these cells to short-term BRD9 bromodomain inhibition. We examined the bromodomain-dependent growth of these cell lines, identifying a dependency in AML cell lines but not HEK293T cells. To define a mechanism through which BRD9 maintains AML cell survival, we examined nascent transcription, chromatin accessibility, and ncBAF complex binding genome-wide after bromodomain inhibition. We identified extensive regulation of transcription by BRD9 bromodomain activity, including repression of myeloid maturation factors and tumor suppressor genes, while standard AML chemotherapy targets were repressed by inhibition of the BRD9 bromodomain. BRD9 bromodomain activity maintained accessible chromatin at both gene promoters and gene-distal putative enhancer regions, in a manner that qualitatively correlated with enrichment of BRD9 binding. Furthermore, we identified reduced chromatin accessibility at GATA, ETS, and AP-1 motifs and increased chromatin accessibility at SNAIL-, HIC-, and TP53-recognized motifs after BRD9 inhibition. These data suggest a role for BRD9 in regulating AML cell differentiation through modulation of accessibility at hematopoietic transcription factor binding sites. SIGNIFICANCE The bromodomain-containing protein BRD9 is essential for AML cell viability, but it is unclear whether this requirement is due to the protein's role as an epigenetic reader. We inhibited this activity and identified altered gene-distal chromatin regulation and transcription consistent with a more mature myeloid cell state.
Collapse
Affiliation(s)
- David C. Klein
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Santana M. Lardo
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Sarah J. Hainer
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
41
|
Morel A, Douat C, Blangy A, Vives V. Bone resorption by osteoclasts involves fine tuning of RHOA activity by its microtubule-associated exchange factor GEF-H1. Front Physiol 2024; 15:1342024. [PMID: 38312316 PMCID: PMC10834693 DOI: 10.3389/fphys.2024.1342024] [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: 11/21/2023] [Accepted: 01/08/2024] [Indexed: 02/06/2024] Open
Abstract
Bone health is controlled by the balance between bone formation by osteoblasts and degradation by osteoclasts. A disequilibrium in favor of bone resorption leads to osteolytic diseases characterized by decreased bone density. Osteoclastic resorption is dependent on the assembly of an adhesion structure: the actin ring, also called podosome belt or sealing zone, which is composed of a unique patterning of podosomes stabilized by microtubules. A better understanding of the molecular mechanisms regulating the crosstalk between actin cytoskeleton and microtubules network is key to find new treatments to inhibit bone resorption. Evidence points to the importance of the fine tuning of the activity of the small GTPase RHOA for the formation and maintenance of the actin ring, but the underlying mechanism is not known. We report here that actin ring disorganization upon microtubule depolymerization is mediated by the activation of the RHOA-ROCK signaling pathway. We next show the involvement of GEF-H1, one of RHOA guanine exchange factor highly expressed in osteoclasts, which has the particularity of being negatively regulated by sequestration on microtubules. Using a CRISPR/Cas9-mediated GEF-H1 knock-down osteoclast model, we demonstrate that RHOA activation upon microtubule depolymerization is mediated by GEF-H1 release. Interestingly, although lower levels of GEF-H1 did not impact sealing zone formation in the presence of an intact microtubule network, sealing zone was smaller leading to impaired resorption. Altogether, these results suggest that a fine tuning of GEF-H1 through its association with microtubules, and consequently of RHOA activity, is essential for osteoclast sealing zone stability and resorption function.
Collapse
Affiliation(s)
- Anne Morel
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Christophe Douat
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Anne Blangy
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| | - Virginie Vives
- CRBM (Montpellier cell Biology Research Center), Univ Montpellier, CNRS (National Center for Scientific Research), Montpellier, France
| |
Collapse
|
42
|
Huang H, Wang S, Guan Y, Ren J, Liu X. Molecular basis and current insights of atypical Rho small GTPase in cancer. Mol Biol Rep 2024; 51:141. [PMID: 38236467 DOI: 10.1007/s11033-023-09140-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Accepted: 12/11/2023] [Indexed: 01/19/2024]
Abstract
Atypical Rho GTPases are a subtype of the Rho GTPase family that are involved in diverse cellular processes. The typical Rho GTPases, led by RhoA, Rac1 and Cdc42, have been well studied, while relative studies on atypical Rho GTPases are relatively still limited and have great exploration potential. With the increase in studies, current evidence suggests that atypical Rho GTPases regulate multiple biological processes and play important roles in the occurrence and development of human cancers. Therefore, this review mainly discusses the molecular basis of atypical Rho GTPases and their roles in cancer. We summarize the sequence characteristics, subcellular localization and biological functions of each atypical Rho GTPase. Moreover, we review the recent advances and potential mechanisms of atypical Rho GTPases in the development of multiple cancers. A comprehensive understanding and extensive exploration of the biological functions of atypical Rho GTPases and their molecular mechanisms in tumors will provide important insights into the pathophysiology of tumors and the development of cancer therapeutic strategies.
Collapse
Affiliation(s)
- Hua Huang
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Sijia Wang
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Yifei Guan
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China
| | - Jing Ren
- Department of Plastic and Reconstructive Surgery, The First Medical Center, Chinese PLA (People's Liberation Army) General Hospital, Beijing, 100853, China.
| | - Xinhui Liu
- Center of Excellence for Environmental Safety and Biological Effects, Faculty of Environment and Life, Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing University of Technology, Beijing, 100124, China.
- Faculty of Environment and Life, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
43
|
Cowell E, Jaber H, Kris LP, Fitzgerald MG, Sanders VM, Norbury AJ, Eyre NS, Carr JM. Vav proteins do not influence dengue virus replication but are associated with induction of phospho-ERK, IL-6, and viperin mRNA following DENV infection in vitro. Microbiol Spectr 2024; 12:e0239123. [PMID: 38054722 PMCID: PMC10782993 DOI: 10.1128/spectrum.02391-23] [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: 06/07/2023] [Accepted: 11/03/2023] [Indexed: 12/07/2023] Open
Abstract
IMPORTANCE Dengue disease is characterized by an inflammatory-mediated immunopathology, with elevated levels of circulating factors including TNF-α and IL-6. If the damaging inflammatory pathways could be blocked without loss of antiviral responses or exacerbating viral replication, then this would be of potential therapeutic benefit. The study here has investigated the Vav guanine exchange factors as a potential alternative signaling pathway that may drive dengue virus (DENV)-induced inflammatory responses, with a focus on Vav1 and 2. While Vav proteins were positively associated with mRNA for inflammatory cytokines, blocking Vav signaling didn't affect DENV replication but prevented DENV-induction of p-ERK and enhanced IL-6 (inflammatory) and viperin (antiviral) mRNA. These initial data suggest that Vav proteins could be a target that does not compromise control of viral replication and should be investigated further for broader impact on host inflammatory responses, in settings such as antibody-dependent enhancement of infection and in different cell types.
Collapse
Affiliation(s)
- Evangeline Cowell
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Hawraa Jaber
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Luke P. Kris
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Madeleine G. Fitzgerald
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Valeria M. Sanders
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Aidan J. Norbury
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Nicholas S. Eyre
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| | - Jillian M. Carr
- College of Medicine and Public Health and Flinders Health and Medical Research Institute, Flinders University, Bedford Park, Adelaide, South Australia, Australia
| |
Collapse
|
44
|
Thapa R, Afzal O, Afzal M, Gupta G, Bhat AA, Hassan Almalki W, Kazmi I, Alzarea SI, Saleem S, Arora P, Singh SK, Dua K. From LncRNA to metastasis: The MALAT1-EMT axis in cancer progression. Pathol Res Pract 2024; 253:154959. [PMID: 38029713 DOI: 10.1016/j.prp.2023.154959] [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: 10/12/2023] [Revised: 11/11/2023] [Accepted: 11/16/2023] [Indexed: 12/01/2023]
Abstract
Cancer is a complex disease that causes abnormal genetic changes and unchecked cellular growth. It also causes a disruption in the normal regulatory processes that leads to the creation of malignant tissue. The complex interplay of genetic, environmental, and epigenetic variables influences its etiology. Long non-coding RNAs (LncRNAs) have emerged as pivotal contributors within the intricate landscape of cancer biology, orchestrating an array of multifaceted cellular processes that substantiate the processes of carcinogenesis and metastasis. Metastasis is a crucial driver of cancer mortality. Among these, MALAT1 (Metastasis-Associated Lung Adenocarcinoma Transcript 1) has drawn a lot of interest for its function in encouraging metastasis via controlling the Epithelial-Mesenchymal Transition (EMT) procedure. MALAT1 exerts a pivotal influence on the process of EMT, thereby promoting metastasis to distant organs. The mechanistic underpinning of this phenomenon involves the orchestration of an intricate regulatory network encompassing transcription factors, signalling cascades, and genes intricately associated with the EMT process by MALAT1. Its crucial function in transforming tumor cells into an aggressive phenotype is highlighted by its capacity to influence the expression of essential EMT effectors such as N-cadherin, E-cadherin, and Snail. An understanding of the MALAT1-EMT axis provides potential therapeutic approaches for cancer intervention. Targeting MALAT1 or its downstream EMT effectors may reduce the spread of metastatic disease and improve the effectiveness of already available therapies. Understanding the MALAT1-EMT axis holds significant clinical implications. Therefore, directing attention towards MALAT1 or its downstream mediators could present innovative therapeutic strategies for mitigating metastasis and improving patient prognosis. This study highlights the importance of MALAT1 in cancer biology and its potential for cutting back on metastatic disease with novel treatment strategies.
Collapse
Affiliation(s)
- Riya Thapa
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Obaid Afzal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al Kharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, India; School of Pharmacy, Graphic Era Hill University, Dehradun 248007, India.
| | - Asif Ahmad Bhat
- School of Pharmacy, Suresh Gyan Vihar University, Mahal Road, Jagatpura, Jaipur, India
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sami I Alzarea
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka, Al-Jouf, Saudi Arabia
| | - Shakir Saleem
- Department of Public Health. College of Health Sciences, Saudi Electronic University, Riyadh, Saudi Arabia
| | - Poonam Arora
- SGT College of Pharmacy, SGT University, Gurugram, Haryana, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India; Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW 2007, Australia; Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW 2007, Australia
| |
Collapse
|
45
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [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: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
46
|
Kozlova A, Sarygina E, Deinichenko K, Radko S, Ptitsyn K, Khmeleva S, Kurbatov L, Spirin P, Prassolov V, Ilgisonis E, Lisitsa A, Ponomarenko E. Comparison of Alternative Splicing Landscapes Revealed by Long-Read Sequencing in Hepatocyte-Derived HepG2 and Huh7 Cultured Cells and Human Liver Tissue. BIOLOGY 2023; 12:1494. [PMID: 38132320 PMCID: PMC10740679 DOI: 10.3390/biology12121494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 11/17/2023] [Accepted: 11/25/2023] [Indexed: 12/23/2023]
Abstract
The long-read RNA sequencing developed by Oxford Nanopore Technologies provides a direct quantification of transcript isoforms, thereby making it possible to present alternative splicing (AS) profiles as arrays of single splice variants with different abundances. Additionally, AS profiles can be presented as arrays of genes characterized by the degree of alternative splicing (the DAS-the number of detected splice variants per gene). Here, we successfully utilized the DAS to reveal biological pathways influenced by the alterations in AS in human liver tissue and the hepatocyte-derived malignant cell lines HepG2 and Huh7, thus employing the mathematical algorithm of gene set enrichment analysis. Furthermore, analysis of the AS profiles as abundances of single splice variants by using the graded tissue specificity index τ provided the selection of the groups of genes expressing particular splice variants specifically in liver tissue, HepG2 cells, and Huh7 cells. The majority of these splice variants were translated into proteins products and appeal to be in focus regarding further insights into the mechanisms underlying cell malignization. The used metrics are intrinsically suitable for transcriptome-wide AS profiling using long-read sequencing.
Collapse
Affiliation(s)
- Anna Kozlova
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Elizaveta Sarygina
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Kseniia Deinichenko
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Sergey Radko
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Konstantin Ptitsyn
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Svetlana Khmeleva
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Leonid Kurbatov
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Pavel Spirin
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (P.S.); (V.P.)
| | - Vladimir Prassolov
- Department of Cancer Cell Biology, Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Vavilova 32, 119991 Moscow, Russia; (P.S.); (V.P.)
| | - Ekaterina Ilgisonis
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Andrey Lisitsa
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| | - Elena Ponomarenko
- Institute of Biomedical Chemistry, Pogodinskaya Street 10, 119121 Moscow, Russia (S.R.)
| |
Collapse
|
47
|
Ma S, Wang Y, Li W, Qiu S, Zhang X, Niu R, Zhao F, Zheng Y. Integrated analysis identities Rho GTPases related molecular map in patients with gastric carcinoma. Sci Rep 2023; 13:21443. [PMID: 38052924 PMCID: PMC10698149 DOI: 10.1038/s41598-023-48294-z] [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] [Accepted: 11/24/2023] [Indexed: 12/07/2023] Open
Abstract
The intricate involvement of Rho GTPases in a multitude of human malignancies and their diverse array of biological functions has garnered substantial attention within the scientific community. However, their expression pattern and potential role in gastric cancer (GC) remain unclear. In this study, we successfully identified two distinct subtypes associated with Rho GTPase-related gene (RGG) through consensus clustering analysis, which exhibited significant disparities in overall survival and the tumor microenvironment. Subsequently, an extensively validated risk model termed RGGscore was meticulously constructed to prognosticate the outcomes of GC patients. This model was further assessed and validated using an external cohort. Notably, the high RGGscore group was indicative of a poorer prognosis. Univariate and multivariate Cox regression analyses unveiled the RGGscore as an autonomous prognostic indicator for GC patients. Subsequent external validation, utilizing two cohorts of patients who underwent immunotherapy, demonstrated a significant correlation between a low RGGscore and improved response to immunotherapy. Additionally, the expression levels of three genes associated with RGGscore were examined using qRT-PCR. Taken together, a pioneering RGGscore model has been successfully established, showcasing its potential efficacy in offering valuable therapeutic guidance for GC.
Collapse
Affiliation(s)
- Shaowei Ma
- Department of Gastrointestinal Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ying Wang
- Department of Cardiology, Xingtai Third Hospital, Xingtai, 054000, China
| | - Weibo Li
- Department of Gastrointestinal Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Shaofan Qiu
- Department of Gastrointestinal Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Xiangyu Zhang
- Department of Gastrointestinal Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China
| | - Ren Niu
- Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Fangchao Zhao
- Department of Thoracic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| | - Yu Zheng
- Department of Gastrointestinal Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, China.
| |
Collapse
|
48
|
Soh JEC, Shimizu A, Sato A, Ogita H. Novel cardiovascular protective effects of RhoA signaling and its therapeutic implications. Biochem Pharmacol 2023; 218:115899. [PMID: 37907138 DOI: 10.1016/j.bcp.2023.115899] [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: 09/19/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/02/2023]
Abstract
Ras homolog gene family member A (RhoA) belongs to the Rho GTPase superfamily, which was first studied in cancers as one of the essential regulators controlling cellular function. RhoA has long attracted attention as a key molecule involved in cell signaling and gene transcription, through which it affects cellular processes. A series of studies have demonstrated that RhoA plays crucial roles under both physiological states and pathological conditions in cardiovascular diseases. RhoA has been identified as an important regulator in cardiac remodeling by regulating actin stress fiber dynamics and cytoskeleton formation. However, its underlying mechanisms remain poorly understood, preventing definitive conclusions being drawn about its protective role in the cardiovascular system. In this review, we outline the characteristics of RhoA and its related signaling molecules, and present an overview of RhoA classical function and the corresponding cellular responses of RhoA under physiological and pathological conditions. Overall, we provide an update on the novel signaling under RhoA in the cardiovascular system and its potential clinical and therapeutic targets in cardiovascular medicine.
Collapse
Affiliation(s)
- Joanne Ern Chi Soh
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akio Shimizu
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Akira Sato
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Hisakazu Ogita
- Division of Molecular Medical Biochemistry, Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan.
| |
Collapse
|
49
|
Pereira M, Glogova A, Haagsma J, Stewart J, Shepherd TG, Petrik J. Mutant p53 murine oviductal epithelial cells induce progression of high-grade serous carcinoma and are most sensitive to simvastatin therapy in vitro and in vivo. J Ovarian Res 2023; 16:218. [PMID: 37986175 PMCID: PMC10662458 DOI: 10.1186/s13048-023-01307-x] [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: 02/14/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023] Open
Abstract
High-grade serous carcinoma (HGSC) is the most common and aggressive subtype of epithelial ovarian cancer, characterized by gain-of-function TP53 mutations originating in the fallopian tube epithelium. Therapeutic intervention occurs at advanced metastatic disease, due to challenges in early-stage diagnosis, with common disease recurrence and therapy resistance despite initial therapy success. The mevalonate pathway is exploited by many cancers and is potently inhibited by statin drugs. Statins have shown anti-cancer activity in many, but not all cancers. Here, we investigated the role of p53 status in relation to mevalonate pathway signaling in murine oviductal epithelial (OVE) cells and identified OVE cell sensitivity to statin inhibition. We found that p53R175H mutant and Trp53 knockout OVE cells have increased mevalonate pathway signaling compared to p53 wild-type OVE cells. Through orthotopic implantation to replicate the fallopian tube origin of HGSC, p53R175H mutant cells upregulated the mevalonate pathway to drive progression to advanced-stage ovarian cancer, and simvastatin treatment abrogated this effect. Additionally, simvastatin was more efficacious at inhibiting cell metabolic activity in OVE cells than atorvastatin, rosuvastatin and pravastatin. In vitro, simvastatin demonstrated potent effects on cell proliferation, apoptosis, invasion and migration in OVE cells regardless of p53 status. In vivo, simvastatin induced ovarian cancer disease regression through decreased primary ovarian tumor weight and increased apoptosis. Simvastatin also significantly increased cytoplasmic localization of HMG-CoA reductase in ovarian tumors. Downstream of the mevalonate pathway, simvastatin had no effect on YAP or small GTPase activity. This study suggests that simvastatin can induce anti-tumor effects and could be an important inhibitor of ovarian cancer progression.
Collapse
Affiliation(s)
- Madison Pereira
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Alice Glogova
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jacob Haagsma
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Julia Stewart
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Trevor G Shepherd
- The Mary & John Knight Translational Ovarian Cancer Research Unit, London Regional Cancer Program, London, ON, Canada
- Department of Anatomy & Cell Biology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Obstetrics & Gynaecology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
50
|
Magalhaes YT, Forti FL. ROCK inhibition reduces the sensitivity of mutant p53 glioblastoma to genotoxic stress through a Rac1-driven ROS production. Int J Biochem Cell Biol 2023; 164:106474. [PMID: 37778694 DOI: 10.1016/j.biocel.2023.106474] [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: 05/12/2023] [Revised: 09/16/2023] [Accepted: 09/21/2023] [Indexed: 10/03/2023]
Abstract
Resistance to radio and chemotherapy in Glioblastoma (GBM) is correlated with its malignancy, invasiveness, and aggressiveness. The Rho GTPase pathway plays important roles in these processes, but its involvement in the GBM response to genotoxic treatments remains unsolved. Inhibition of this signaling pathway has emerged as a promising approach for the treatment of CNS injuries and diseases, proving to be a strong candidate for therapeutic approaches. To this end, Rho-associated kinases (ROCK), classic downstream effectors of small Rho GTPases, were targeted for pharmacological inhibition using Y-27632 in GBM cells, expressing the wild-type or mutated p53 gene, and exposed to genotoxic stress by gamma ionizing radiation (IR) or cisplatin (PT). The use of the ROCK inhibitor (ROCKi) had opposite effects in these cells: in cells expressing wild-type p53, ROCKi reduced survival and DNA repair capacity (reduction of γH2AX foci and accumulation of strand breaks) after stress promoted by IR or PT; in cells expressing the mutant p53 protein, both treatments promoted longer survival and more efficient DNA repair, responses further enhanced by ROCKi. The target DNA repair mechanisms of ROCK inhibition were, respectively, an attenuation of NHEJ and NER pathways in wild-type p53 cells, and a stimulation of HR and NER pathways in mutant p53 cells. These effects were accompanied by the formation of reactive oxygen species (ROS) induced by genotoxic stress only in mutant p53 cells but potentiated by ROCKi and reversed by p53 knockdown. N-acetyl-L-cysteine (NAC) treatment or Rac1 knockdown completely eliminated ROCKi's p53-dependent actions, since ROCK inhibition specifically elevated Rac-GTP levels only in mutant p53 cells. Combining IR or PT and ROCKi treatments broadens our understanding of the sensitivity and resistance of, respectively, GBM expressing wild-type or mutant p53 to genotoxic agents. Our proposal may be a determining factor in improving the efficiency and assertiveness of CNS antitumor therapies based on ROCK inhibitors. SIGNIFICANCE: The use of ROCK inhibitors in association with radio or chemotherapy modulates GBM resistance and sensitivity depending on the p53 activity, suggesting the potential value of this protein as therapeutic target for tumor pre-sensitization strategies.
Collapse
Affiliation(s)
- Yuli Thamires Magalhaes
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Fabio Luis Forti
- Laboratory of Signaling in Biomolecular Systems, Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil.
| |
Collapse
|