1
|
Yang J, Zhang T, Zhang L, Su X. A non-equilibrium dissipation system with tunable molecular fuel flux. NANOSCALE 2024; 16:4219-4228. [PMID: 38334944 DOI: 10.1039/d3nr06136a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2024]
Abstract
Cells convert macromolecule fuel into small molecule fuel through energy pathways, including glycolysis, the citric acid cycle, and oxidative phosphorylation. These processes drive vital dissipative networks or structures. Distinct from direct fuel (DF) utilization (directly acquire and utilize small molecule fuel), this macromolecule fuel mechanism is referred to as indirect fuel (IF) utilization, wherein the generation rate of small molecule fuel (fuel flux) can be effectively regulated. Here, we reported a bionic dissipation system with tunable fuel flux based on dynamic DNA nanotechnology. By regulating the rates of strand displacement and enzymatic reactions, we controlled the fuel flux and further tuned the strength of non-equilibrium transient states. Interestingly, we found that within a certain range, the fuel flux was positively correlated with the strength of the transient state. Once saturation was reached, it became negatively correlated. An appropriate fuel flux supports the maintenance of high-intensity non-equilibrium transients. Furthermore, we harnessed the dissipation system with tunable molecular fuel flux to regulate the dynamic assembly and disassembly of AuNPs. Different fuel fluxes resulted in varying assembly and disassembly rates and strengths for AuNPs, accomplishing a biomimetic process of regulating microtubule assembly through the control of fuel flux within living organisms. This work demonstrated a dissipation system with tunable molecular fuel flux, and we envision that this system holds significant potential for development in various fields such as biomimetics, synthetic biology, smart materials, biosensing, and artificial cells.
Collapse
Affiliation(s)
- Jiayu Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Tengfang Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Linghao Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Xin Su
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
2
|
Song D, Hu F, Huang C, Lan J, She X, Zhao C, Wu H, Liu A, Wu Q, Chen Y, Luo X, Feng Y, Yang X, Xu C, Hu J, Wang G. Tiam1 methylation by NSD2 promotes Rac1 signaling activation and colon cancer metastasis. Proc Natl Acad Sci U S A 2023; 120:e2305684120. [PMID: 38113258 PMCID: PMC10756287 DOI: 10.1073/pnas.2305684120] [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: 04/15/2023] [Accepted: 10/03/2023] [Indexed: 12/21/2023] Open
Abstract
Metastasis is a major cause of cancer therapy failure and mortality. However, targeting metastatic seeding and colonization remains a significant challenge. In this study, we identified NSD2, a histone methyltransferase responsible for dimethylating histone 3 at lysine 36, as being overexpressed in metastatic tumors. Our findings suggest that NSD2 overexpression enhances tumor metastasis both in vitro and in vivo. Further analysis revealed that NSD2 promotes tumor metastasis by activating Rac1 signaling. Mechanistically, NSD2 combines with and activates Tiam1 (T lymphoma invasion and metastasis 1) and promotes Rac1 signaling by methylating Tiam1 at K724. In vivo and in vitro studies revealed that Tiam1 K724 methylation could be a predictive factor for cancer prognosis and a potential target for metastasis inhibition. Furthermore, we have developed inhibitory peptide which was proved to inhibit tumor metastasis through blocking the interaction between NSD2 and Tiam1. Our results demonstrate that NSD2-methylated Tiam1 promotes Rac1 signaling and cancer metastasis. These results provide insights into the inhibition of tumor metastasis.
Collapse
Affiliation(s)
- Da Song
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Fuqing Hu
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Changsheng Huang
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Jingqin Lan
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Xiaowei She
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Chongchong Zhao
- Department of Protein Chemistry and Proteinomics Facility at Technology Center for Protein Sciences, Tsinghua University, Beijing100084, China
| | - Hong Wu
- Department of Integrative Cancer Center and Cancer Clinical Research Center, Sichuan Cancer Hospital and Institute Sichuan Cancer Center, School of Medicine University of Electronic Science and Technology, Chengdu610000, China
| | - Anyi Liu
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Qi Wu
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Yaqi Chen
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Xuelai Luo
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Yongdong Feng
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Xiangping Yang
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan430030, China
| | - Chuan Xu
- Department of Integrative Cancer Center and Cancer Clinical Research Center, Sichuan Cancer Hospital and Institute Sichuan Cancer Center, School of Medicine University of Electronic Science and Technology, Chengdu610000, China
| | - Junbo Hu
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| | - Guihua Wang
- Department of Gastrointestinal Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan430030, China
| |
Collapse
|
3
|
He S, Liu R, Luo Q, Song G. Tensile Overload Injures Human Alveolar Epithelial Cells through YAP/F-Actin/MAPK Signaling. Biomedicines 2023; 11:1833. [PMID: 37509472 PMCID: PMC10376431 DOI: 10.3390/biomedicines11071833] [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: 05/14/2023] [Revised: 06/18/2023] [Accepted: 06/23/2023] [Indexed: 07/30/2023] Open
Abstract
BACKGROUND Explosion shockwaves can generate overloaded mechanical forces and induce lung injuries. However, the mechanism of lung injuries caused by tensile overload is still unclear. METHODS Flow cytometry was used to detect the apoptosis of human alveolar epithelial cells (BEAS-2B) induced by tensile overload, and cell proliferation was detected using 5-ethynyl-2'-deoxyuridine (EdU). Immunofluorescence and Western blot analysis were used to identify the tensile overload on the actin cytoskeleton, proteins related to the mitogen-activated protein kinase (MAPK) signal pathway, and the Yes-associated protein (YAP). RESULTS Tensile overload reduced BEAS-2B cell proliferation and increased apoptosis. In terms of the mechanism, we found that tensile overload led to the depolymerization of the actin cytoskeleton, the activation of c-Jun N-terminal kinase (JNK) and extracellular-signal-regulated kinase 1/2 (ERK1/2), and the upregulation of YAP expression. Jasplakinolide (Jasp) treatment promoted the polymerization of the actin cytoskeleton and reduced the phosphorylation of tension-overload-activated JNK and ERK1/2 and the apoptosis of BEAS-2B cells. Moreover, the inhibition of the JNK and ERK1/2 signaling pathways, as well as the expression of YAP, also reduced apoptosis caused by tensile overload. CONCLUSION Our study establishes the role of the YAP/F-actin/MAPK axis in tensile-induced BEAS-2B cell injury and proposes new strategies for the treatment and repair of future lung injuries.
Collapse
Affiliation(s)
- Shan He
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Ruihan Liu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Qing Luo
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| | - Guanbin Song
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing 400030, China
| |
Collapse
|
4
|
Ahmed S, Alam W, Aschner M, Filosa R, Cheang WS, Jeandet P, Saso L, Khan H. Marine Cyanobacterial Peptides in Neuroblastoma: Search for Better Therapeutic Options. Cancers (Basel) 2023; 15:cancers15092515. [PMID: 37173981 PMCID: PMC10177606 DOI: 10.3390/cancers15092515] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/03/2023] [Accepted: 03/12/2023] [Indexed: 05/15/2023] Open
Abstract
Neuroblastoma is the most prevalent extracranial solid tumor in pediatric patients, originating from sympathetic nervous system cells. Metastasis can be observed in approximately 70% of individuals after diagnosis, and the prognosis is poor. The current care methods used, which include surgical removal as well as radio and chemotherapy, are largely unsuccessful, with high mortality and relapse rates. Therefore, attempts have been made to incorporate natural compounds as new alternative treatments. Marine cyanobacteria are a key source of physiologically active metabolites, which have recently received attention owing to their anticancer potential. This review addresses cyanobacterial peptides' anticancer efficacy against neuroblastoma. Numerous prospective studies have been carried out with marine peptides for pharmaceutical development including in research for anticancer potential. Marine peptides possess several advantages over proteins or antibodies, including small size, simple manufacturing, cell membrane crossing capabilities, minimal drug-drug interactions, minimal changes in blood-brain barrier (BBB) integrity, selective targeting, chemical and biological diversities, and effects on liver and kidney functions. We discussed the significance of cyanobacterial peptides in generating cytotoxic effects and their potential to prevent cancer cell proliferation via apoptosis, the activation of caspases, cell cycle arrest, sodium channel blocking, autophagy, and anti-metastasis behavior.
Collapse
Affiliation(s)
- Salman Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine Forchheimer, 209 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Rosanna Filosa
- Department of Science and Technology, University of Sannio, 82100 Benevento, Italy
| | - Wai San Cheang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Philippe Jeandet
- Faculty of Sciences, RIBP-USC INRAe 1488, University of Reims, 51100 Reims, France
| | - Luciano Saso
- Department of Physiology and Pharmacology "Vittorio Erspamer", Sapienza University, 00185 Rome, Italy
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| |
Collapse
|
5
|
Peng C, Guo S, Yang Z, Li X, Su Q, Mo W. A prognostic model for bladder cancer based on cytoskeleton-related genes. Medicine (Baltimore) 2023; 102:e33538. [PMID: 37115085 PMCID: PMC10146030 DOI: 10.1097/md.0000000000033538] [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: 02/07/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND A typical cancerous growth in the urinary tract, bladder cancer (BLCA) has a dismal survival rate and a poor chance of being cured. The cytoskeleton has been shown to be tightly related to tumor invasion and metastasis. Nevertheless, the expression of genes associated with the cytoskeleton and their prognostic significance in BLCA remain unknown. METHODS In our study, we performed differential expression analysis of cytoskeleton-related genes between BLCA versus normal bladder tissues. According to the outcomes of this analysis of differentially expressed genes, all BLCA cases doing nonnegative matrix decomposition clustering analysis be classified into different molecular subtypes and were subjected to Immune cell infiltration analysis. We then constructed a cytoskeleton-associated gene prediction model for BLCA, and performed risk score independent prognostic analysis and receiver operating characteristic curve analyses to evaluate and validate the prognostic value of the model. Furthermore, enrichment analysis, clinical correlation analysis of prognostic models, and immune cell correlation analysis were carried out. RESULTS We identified 546 differentially expressed genes that are linked to the cytoskeleton, including 314 up-regulated genes and 232 down-regulated genes. All BLCA cases doing nonnegative matrix decomposition clustering analysis could be classified into 2 molecular subtypes, and we observed differences (P < .05) in C1 and C2 immune scores about 9 cell types. Next, we obtained 129 significantly expressed cytoskeleton-related genes. A final optimized model was constructed consisting of 11 cytoskeleton-related genes. Survival curves and risk assessment predicted the prognostic risk in both groups of patients with BLCA. Survival curves and receiver operating characteristic curves were used to evaluate and validate the prognostic value of the model. Significant enrichment pathways for cytoskeleton-associated genes in bladder cancer samples were explored by Gene set enrichment analysis enrichment analysis. After we obtained the risk scores, a clinical correlation analysis was performed to examine which clinical traits were related to the risk scores. Finally, we demonstrated a correlation between different immune cells. CONCLUSION Cytoskeleton-related genes have an important predictive value for BLCA, and the prognostic model we constructed may enable personalized treatment of BLCA.
Collapse
Affiliation(s)
- Chunting Peng
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| | - Sufan Guo
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| | - Zheng Yang
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| | - Xiaohong Li
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| | - Qisheng Su
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| | - Wuning Mo
- Key Laboratory of Clinical Laboratory Medicine of Guangxi Department of Education, Department of Clinical Laboratory, the First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang, China
| |
Collapse
|
6
|
Guan G, Cannon RD, Coates DE, Mei L. Effect of the Rho-Kinase/ROCK Signaling Pathway on Cytoskeleton Components. Genes (Basel) 2023; 14:272. [PMID: 36833199 PMCID: PMC9957420 DOI: 10.3390/genes14020272] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/10/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The mechanical properties of cells are important in tissue homeostasis and enable cell growth, division, migration and the epithelial-mesenchymal transition. Mechanical properties are determined to a large extent by the cytoskeleton. The cytoskeleton is a complex and dynamic network composed of microfilaments, intermediate filaments and microtubules. These cellular structures confer both cell shape and mechanical properties. The architecture of the networks formed by the cytoskeleton is regulated by several pathways, a key one being the Rho-kinase/ROCK signaling pathway. This review describes the role of ROCK (Rho-associated coiled-coil forming kinase) and how it mediates effects on the key components of the cytoskeleton that are critical for cell behaviour.
Collapse
Affiliation(s)
- Guangzhao Guan
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Diagnostic and Surgical Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Richard D. Cannon
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| | - Dawn E. Coates
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
| | - Li Mei
- Sir John Walsh Research Institute, Faculty of Dentistry, University of Otago, Dunedin 9016, New Zealand
- Department of Oral Sciences, Faculty of Dentistry, University of Otago, 310 Great King Street, Dunedin 9016, New Zealand
| |
Collapse
|
7
|
Ahmed S, Alam W, Jeandet P, Aschner M, Alsharif KF, Saso L, Khan H. Therapeutic Potential of Marine Peptides in Prostate Cancer: Mechanistic Insights. Mar Drugs 2022; 20:md20080466. [PMID: 35892934 PMCID: PMC9330892 DOI: 10.3390/md20080466] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 02/07/2023] Open
Abstract
Prostate cancer (PCa) is the leading cause of cancer death in men, and its treatment is commonly associated with severe adverse effects. Thus, new treatment modalities are required. In this context, natural compounds have been widely explored for their anti-PCa properties. Aquatic organisms contain numerous potential medications. Anticancer peptides are less toxic to normal cells and provide an efficacious treatment approach via multiple mechanisms, including altered cell viability, apoptosis, cell migration/invasion, suppression of angiogenesis and microtubule balance disturbances. This review sheds light on marine peptides as efficacious and safe therapeutic agents for PCa.
Collapse
Affiliation(s)
- Salman Ahmed
- Department of Pharmacognosy, Faculty of Pharmacy and Pharmaceutical Sciences, University of Karachi, Karachi 75270, Pakistan;
| | - Waqas Alam
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan;
| | - Philippe Jeandet
- Research Unit “Induced Resistance and Plant Bioprotection”, Department of Biology and Biochemistry, Faculty of Sciences, University of Reims, EA 4707-USC INRAe 1488, SFR Condorcet FR CNRS 3417, P.O. Box 1039, CEDEX 02, 51687 Reims, France;
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA;
| | - Khalaf F. Alsharif
- Department of Clinical Laboratory, College of Applied Medical Science, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| | - Luciano Saso
- Department of Physiology and Pharmacology, “Vittorio Erspamer” Sapienza University, 00185 Rome, Italy;
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan;
- Correspondence:
| |
Collapse
|
8
|
Zhang Z, Xie H, Zuo W, Tang J, Zeng Z, Cai W, Lai L, Lu Y, Shen L, Dong X, Yin L, Tang D, Dai Y. Lysine 2-hydroxyisobutyrylation proteomics reveals protein modification alteration in the actin cytoskeleton pathway of oral squamous cell carcinoma. J Proteomics 2021; 249:104371. [PMID: 34500091 DOI: 10.1016/j.jprot.2021.104371] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/01/2021] [Accepted: 09/02/2021] [Indexed: 12/24/2022]
Abstract
As the most commonplace malignant carcinoma in the oral cavity, oral squamous cell carcinoma (OSCC) is highly invasive and prone to recurrence. The nosogenesis of OSCC are affected by epigenetics. Recently, a newly-found post-translational modification of lysine, 2-hydroxyisobutylation (Khib), has been proved to play a critical role in biological regulation. However, no research has evaluated the mechanism of Khib in oral cancer. Here, we performed liquid chromatography-mass spectrometry-based quantitative proteomics combined with bioinformatics analysis to reveal and evaluate Khib protein alterations in OSCC. Numerous proteins in OSCC undergo up-regulated modification of Khib. We quantified and identified 967 proteins with differential expression levels, and 617 2-hydroxyisobutylated proteins with 938 Khib sites. Among them, 125 proteins both differentially expressed and accompanied by obvious Khib modification were further identified and analyzed through KEGG-based and ingenuity pathway analysis (IPA). These proteins are enriched in the actin cytoskeleton regulatory pathway, and IPA predicted that they alter the state of actin aggregation and stability, hence impacting and regulating the actin cytoskeleton in OSCC. This is the first 2-hydroxyisobutylated modification proteomics performed for OSCC. Khib protein is significantly concentrated in the actin cytoskeleton regulatory pathway, indicating that this pathway may mediate the tumorigenesis or exacerbation of OSCC. SIGNIFICANCE: This is the first study that revealed the alterations of Khib protein in oral squamous cell carcinoma through LC-MS/MS-based modified proteomic. Our data showed that the protein in the actin cytoskeleton regulatory pathway was underwent significant Khib modification and abundance changes. We applied predictive function in IPA software to analyze and clarify that the aggregation of actin and the regulation of actin stability that mediated by the actin cytoskeleton regulatory pathway may be the potential mechanism of the occurrence and development of oral squamous cell carcinoma. Our research broadens the understanding of the pathogenesis of oral squamous cell carcinoma and provides new insights for future research.
Collapse
Affiliation(s)
- Zeyu Zhang
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, CN 518020, PR China; Department of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, PR China
| | - Hongliang Xie
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, CN 518020, PR China
| | - Wenxin Zuo
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, CN 518020, PR China
| | - Jianming Tang
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, CN 518020, PR China
| | - Zhipeng Zeng
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, CN 518020, PR China
| | - Wanxia Cai
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, CN 518020, PR China
| | - Liusheng Lai
- Guangxi Key Laboratory of Metabolic Diseases Research, Affiliated No. 924 Hospital, Southern Medical University, Guilin 541002, Guangxi, PR China
| | - Yongpin Lu
- Department of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, PR China
| | - Lingjun Shen
- Department of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, PR China
| | - Xiangnan Dong
- Department of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, PR China
| | - Lianghong Yin
- Department of Nephrology and Blood Purification, the First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, PR China.
| | - Donge Tang
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, CN 518020, PR China.
| | - Yong Dai
- The First Affiliated Hospital of Southern University of Science and Technology, the Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen, CN 518020, PR China; Guangxi Key Laboratory of Metabolic Diseases Research, Affiliated No. 924 Hospital, Southern Medical University, Guilin 541002, Guangxi, PR China.
| |
Collapse
|
9
|
Ciszewski WM, Wawro ME, Sacewicz-Hofman I, Sobierajska K. Cytoskeleton Reorganization in EndMT-The Role in Cancer and Fibrotic Diseases. Int J Mol Sci 2021; 22:ijms222111607. [PMID: 34769036 PMCID: PMC8583721 DOI: 10.3390/ijms222111607] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 10/21/2021] [Accepted: 10/26/2021] [Indexed: 12/11/2022] Open
Abstract
Chronic inflammation promotes endothelial plasticity, leading to the development of several diseases, including fibrosis and cancer in numerous organs. The basis of those processes is a phenomenon called the endothelial–mesenchymal transition (EndMT), which results in the delamination of tightly connected endothelial cells that acquire a mesenchymal phenotype. EndMT-derived cells, known as the myofibroblasts or cancer-associated fibroblasts (CAFs), are characterized by the loss of cell–cell junctions, loss of endothelial markers, and gain in mesenchymal ones. As a result, the endothelium ceases its primary ability to maintain patent and functional capillaries and induce new blood vessels. At the same time, it acquires the migration and invasion potential typical of mesenchymal cells. The observed modulation of cell shape, increasedcell movement, and invasion abilities are connected with cytoskeleton reorganization. This paper focuses on the review of current knowledge about the molecular pathways involved in the modulation of each cytoskeleton element (microfilaments, microtubule, and intermediate filaments) during EndMT and their role as the potential targets for cancer and fibrosis treatment.
Collapse
|
10
|
Yang C, Zhu S, Feng W, Chen X. Calponin 3 suppresses proliferation, migration and invasion of non-small cell lung cancer cells. Oncol Lett 2021; 22:634. [PMID: 34267826 PMCID: PMC8258620 DOI: 10.3892/ol.2021.12895] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Accepted: 05/28/2021] [Indexed: 02/07/2023] Open
Abstract
Calponin 3 (CNN3) is known to serve a role in certain types of cancer, such as gastric cancer and colorectal cancer. The present study investigated the clinical significance of CNN3 in non-small cell lung cancer (NSCLC) by evaluating its expression profile and relationship with disease prognosis using the Gene Expression Omnibus repository, Gene Expression Profiling Interactive Analysis 2 (GEPIA2) and Kaplan-Meier plotter analysis. CNN3 mRNA expression was measured using reverse transcription-quantitative PCR, while the protein expression level was measured using western blot analysis. Cell proliferation, cell cycle and apoptosis, and migration and invasion were analyzed using MTS assay, flow cytometry and Transwell assays, respectively. These results revealed that CNN3 mRNA expression was downregulated in NSCLC tissues compared with that in normal tissues. Additionally, CNN3 expression had a high diagnostic value based on the GSE2514 dataset and the data from The Cancer Genome Atlas and the Genotype Tissue Expression database, whereas it had a low diagnostic value based on the GSE10072 dataset. Furthermore, CNN3 expression was associated with survival in patients with lung adenocarcinoma (LUAD), whereas it was not associated with survival in patients with lung squamous cell carcinoma (LUSC) according to the Kaplan-Meier plotter results. According to the data from GEPIA2, and the GSE72094, GSE41271 and GSE31210 datasets, CNN3 expression was not associated with the prognosis of patients with LUAD and LUSC. The mRNA and protein expression levels of CNN3 were lower in two NSCLC cell lines (A549 and SK-MES-1) than in a human bronchial epithelial cell line (BEAS-2B). CNN3 overexpression suppressed cell proliferation, migration and invasion, induced G1-phase arrest, promoted apoptosis and suppressed PI3K/AKT signaling pathway activation in the NSCLC cell lines, whereas CNN3 overexpression had no effect on cell morphology. In conclusion, CNN3 suppressed the proliferation and metastasis of NSCLC cells by downregulating the PI3K/AKT signaling pathway, making it a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Chenglin Yang
- Traditional Chinese Medicine Department, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Shiping Zhu
- Traditional Chinese Medicine Department, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Weifeng Feng
- Traditional Chinese Medicine Department, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510632, P.R. China
| | - Xuexin Chen
- Department of Respiratory and Critical Care Medicine, The General Hospital of Yima Coal Industry Group Co. Ltd., Yima, Henan 472300, P.R. China
| |
Collapse
|
11
|
Vasilaki D, Bakopoulou A, Tsouknidas A, Johnstone E, Michalakis K. Biophysical interactions between components of the tumor microenvironment promote metastasis. Biophys Rev 2021; 13:339-357. [PMID: 34168685 PMCID: PMC8214652 DOI: 10.1007/s12551-021-00811-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Accepted: 05/03/2021] [Indexed: 02/07/2023] Open
Abstract
During metastasis, tumor cells need to adapt to their dynamic microenvironment and modify their mechanical properties in response to both chemical and mechanical stimulation. Physical interactions occur between cancer cells and the surrounding matrix including cell movements and cell shape alterations through the process of mechanotransduction. The latter describes the translation of external mechanical cues into intracellular biochemical signaling. Reorganization of both the cytoskeleton and the extracellular matrix (ECM) plays a critical role in these spreading steps. Migrating tumor cells show increased motility in order to cross the tumor microenvironment, migrate through ECM and reach the bloodstream to the metastatic site. There are specific factors affecting these processes, as well as the survival of circulating tumor cells (CTC) in the blood flow until they finally invade the secondary tissue to form metastasis. This review aims to study the mechanisms of metastasis from a biomechanical perspective and investigate cell migration, with a focus on the alterations in the cytoskeleton through this journey and the effect of biologic fluids on metastasis. Understanding of the biophysical mechanisms that promote tumor metastasis may contribute successful therapeutic approaches in the fight against cancer.
Collapse
Affiliation(s)
- Dimitra Vasilaki
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Athina Bakopoulou
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
| | - Alexandros Tsouknidas
- Laboratory for Biomaterials and Computational Mechanics, Department of Mechanical Engineering, University of Western Macedonia, Kozani, Greece
| | | | - Konstantinos Michalakis
- Department of Prosthodontics, School of Dentistry, Faculty of Health Sciences, Aristotle University of Thessaloniki, University Campus, 54124 Thessaloniki, Greece
- Division of Graduate Prosthodontics, Tufts University School of Dental Medicine, Boston, MA USA
- University of Oxford, Oxford, UK
| |
Collapse
|