1
|
Linke JA, Munn LL, Jain RK. Compressive stresses in cancer: characterization and implications for tumour progression and treatment. Nat Rev Cancer 2024; 24:768-791. [PMID: 39390249 DOI: 10.1038/s41568-024-00745-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/12/2024]
Abstract
Beyond their many well-established biological aberrations, solid tumours create an abnormal physical microenvironment that fuels cancer progression and confers treatment resistance. Mechanical forces impact tumours across a range of biological sizes and timescales, from rapid events at the molecular level involved in their sensing and transmission, to slower and larger-scale events, including clonal selection, epigenetic changes, cell invasion, metastasis and immune response. Owing to challenges with studying these dynamic stimuli in biological systems, the mechanistic understanding of the effects and pathways triggered by abnormally elevated mechanical forces remains elusive, despite clear correlations with cancer pathophysiology, aggressiveness and therapeutic resistance. In this Review, we examine the emerging and diverse roles of physical forces in solid tumours and provide a comprehensive framework for understanding solid stress mechanobiology. We first review the physiological importance of mechanical forces, especially compressive stresses, and discuss their defining characteristics, biological context and relative magnitudes. We then explain how abnormal compressive stresses emerge in tumours and describe the experimental challenges in investigating these mechanically induced processes. Finally, we discuss the clinical translation of mechanotherapeutics that alleviate solid stresses and their potential to synergize with chemotherapy, radiotherapy and immunotherapies.
Collapse
Affiliation(s)
- Julia A Linke
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
2
|
Zhang N, Wu J, Gao S, Peng H, Li H, Gibson C, Wu S, Zhu J, Zheng Q. pH-Controlled Chemoselective Rapid Azo-Coupling Reaction (CRACR) Enables Global Profiling of Serotonylation Proteome in Cancer Cells. J Proteome Res 2024; 23:4457-4466. [PMID: 39208062 DOI: 10.1021/acs.jproteome.4c00409] [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] [Indexed: 09/04/2024]
Abstract
Serotonylation has been identified as a novel protein posttranslational modification for decades, where an isopeptide bond is formed between the glutamine residue and serotonin through transamination. Transglutaminase 2 (also known as TGM2 or TGase2) was proven to act as the main "writer" enzyme for this PTM, and a number of key regulatory proteins (including small GTPases, fibronectin, fibrinogen, serotonin transporter, and histone H3) have been characterized as the substrates of serotonylation. However, due to the lack of pan-specific antibodies for serotonylated glutamine, the precise enrichment and proteomic profiling of serotonylation still remain challenging. In our previous research, we developed an aryldiazonium probe to specifically label protein serotonylation in a bioorthogonal manner, which depended on a pH-controlled chemoselective rapid azo-coupling reaction. Here, we report the application of a photoactive aryldiazonium-biotin probe for the global profiling of serotonylation proteome in cancer cells. Thus, over 1,000 serotonylated proteins were identified from HCT 116 cells, many of which are highly related to carcinogenesis. Moreover, a number of modification sites of these serotonylated proteins were determined, attributed to the successful application of our chemical proteomic approach. Overall, these findings provided new insights into the significant association between cellular protein serotonylation and cancer development, further suggesting that to target TGM2-mediated monoaminylation may serve as a promising strategy for cancer therapeutics.
Collapse
Affiliation(s)
- Nan Zhang
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Jinghua Wu
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Shuaixin Gao
- Human Nutrition Program, Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Haidong Peng
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Huapeng Li
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, Ohio 43210, United States
| | - Connor Gibson
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
| | - Sophia Wu
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Columbus Academy, Gahanna, Ohio 43230, United States
| | - Jiangjiang Zhu
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Human Nutrition Program, Department of Human Sciences, College of Education and Human Ecology, The Ohio State University, Columbus, Ohio 43210, United States
| | - Qingfei Zheng
- Department of Radiation Oncology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Center for Cancer Metabolism, James Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio 43210, United States
- Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Biological Chemistry and Pharmacology, College of Medicine, The Ohio State University, Columbus, Ohio 43210, United States
- Department of Medicinal Chemistry and Molecular Pharmacology, College of Pharmacy, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
3
|
Cao Z, Wei X, Xing S, Zhang J, Wang S, Yue L, Zhang J, Jiang N, Zhai X. Discovery of novel pyrazolo[1,5-a]pyrimidine derivatives as selective ROCK2 inhibitors with anti-breast cancer migration and invasion activities. Bioorg Chem 2024; 151:107675. [PMID: 39126868 DOI: 10.1016/j.bioorg.2024.107675] [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: 06/14/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024]
Abstract
Rho-associated coiled-coil kinase (ROCK) is involved in multiple cellular activities regulating the actin cytoskeleton, such as cell morphology, adhesion, and migration. The inhibition of ROCK is a feasible strategy to suppress breast cancer metastasis. Herein, based on Belumosudil, a series of pyrazolo[1,5-a]pyrimidine derivatives as selective ROCK2 inhibitors were designed and synthesized. Through systematic investigation of SARs, the piperazine analog 7u was identified with optimum ROCK2 inhibitory activity (IC50 = 36.8 nM) and excellent selectivity over the isoform protein ROCK1 (>250-fold). Intriguingly, upon treatment with 7u, the arrangement of the MDA-MB-231 cytoskeleton was affected accompanied by the alteration of morphology. Furthermore, cell scratch and transwell assays indicated that 7u inhibited MDA-MB-231 cell migration and invasion in a dose-dependent manner. Ultimately, the binding model of 7u with ROCK2 well accounted for the superior activities of 7u as a promising ROCK2 inhibitor with the potential application in breast cancer metastasis treatment.
Collapse
Affiliation(s)
- Zhi Cao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiujian Wei
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuming Xing
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiahao Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Shuqi Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Lingfeng Yue
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiahe Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Nan Jiang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Xin Zhai
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
4
|
Cui YF, Lu QH, Huang X, Lin WN, Huang T, Yang Q. [Effects of inhibition of Rho/ROCK pathway on proliferation and migration of airway smooth muscle cells and related mechanisms]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:974-981. [PMID: 39267514 PMCID: PMC11404462 DOI: 10.7499/j.issn.1008-8830.2405119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 09/17/2024]
Abstract
OBJECTIVES To investigate the effects and molecular mechanisms of inhibition of the Ras homolog gene (Rho)/Rho-associated coiled-coil forming protein kinase (ROCK) pathway on the proliferation and migration of airway smooth muscle cells involving myocardin (MYOCD). METHODS Human airway smooth muscle cells were infected with the adenoviral vector Ad-ZsGreen-shRNA-hROCK1 in vitro. The cells were randomly divided into four groups: ROCK1 gene silencing control (shNC) group, shNC + arachidonic acid (AA, Rho/ROCK pathway activator) group, ROCK1 gene silencing (shROCK1) group, and shROCK1 + AA group (n=3 each). Quantitative real-time polymerase chain reaction and Western blot were used to detect the expression levels of ROCK1 and MYOCD mRNA and protein. ELISA was employed to measure the levels of globular actin and filamentous actin, while immunofluorescent staining and scratch assays were utilized to assess cell proliferation and migration. RESULTS Compared to the shNC + AA group, the shROCK1 + AA group exhibited decreased levels of ROCK1 and MYOCD mRNA and protein expression, reduced expression levels of globular actin and filamentous actin, and diminished cell proliferation and migration capabilities (P<0.05). CONCLUSIONS Inhibition of the Rho/ROCK pathway suppresses the proliferation and migration of airway smooth muscle cells, which may be associated with the downregulation of MYOCD.
Collapse
Affiliation(s)
- Yun-Fei Cui
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong 518036, China
| | - Qing-Hua Lu
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong 518036, China
| | - Xiao Huang
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong 518036, China
| | - Wei-Nan Lin
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong 518036, China
| | - Ting Huang
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong 518036, China
| | - Qin Yang
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, Guangdong 518036, China
| |
Collapse
|
5
|
Jia W, Czabanka M, Broggini T. Cell blebbing novel therapeutic possibilities to counter metastasis. Clin Exp Metastasis 2024:10.1007/s10585-024-10308-z. [PMID: 39222238 DOI: 10.1007/s10585-024-10308-z] [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: 05/23/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
Cells constantly reshape there plasma membrane and cytoskeleton during physiological and pathological processes (Hagmann et al. in J Cell Biochem 73:488-499, 1999). Cell blebbing, the formation of bulges or protrusions on the cell membrane, is related to mechanical stress, changes in intracellular pressure, chemical signals, or genetic anomalies. These membrane bulges interfere with the force balance of actin filaments, microtubules, and intermediate filaments, the basic components of the cytoskeleton (Charras in J Microsc 231:466-478, 2008). In the past, these blebs with circular structures were considered apoptotic markers (Blaser et al. in Dev Cell 11:613-627, 2006). Cell blebbing activates phagocytes and promotes the rapid removal of intrinsic compartments. However, recent studies have revealed that blebbing is associated with dynamic cell reorganization and alters the movement of cells in-vivo and in-vitro (Charras and Paluch in Nat Rev Mol Cell Biol 9:730-736, 2008). During tumor progression, blebbing promotes invasion of cancer cells into blood, and lymphatic vessels, facilitating tumor progression and metastasis (Weems et al. in Nature 615:517-525, 2023). Blebbing is a dominant feature of tumor cells generally absent in normal cells. Restricting tumor blebbing reduces anoikis resistance (survival in suspension) (Weems et al. in Nature 615:517-525, 2023). Hence, therapeutic intervention with targeting blebbing could be highly selective for proliferating pro-metastatic tumor cells, providing a novel therapeutic pathway for tumor metastasis with minimal side effects. Here, we review the association between cell blebbing and tumor cells, to uncover new research directions and strategies for metastatic cancer therapy. Finaly, we aim to identify the druggable targets of metastatic cancer in relation to cell blebbing.
Collapse
Affiliation(s)
- Weiyi Jia
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
| | - Marcus Czabanka
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany
| | - Thomas Broggini
- Department of Neurosurgery, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt, Germany.
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, Germany.
| |
Collapse
|
6
|
Xu QX, Guo L, Li Y, Wang ZW, Hu P, Yang GM, Pan Y. In silico screening-based discovery of benzamide derivatives as inhibitors of Rho-associated kinase-1 (ROCK1). J Biomol Struct Dyn 2024; 42:7467-7484. [PMID: 37668086 DOI: 10.1080/07391102.2023.2253918] [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: 02/09/2023] [Accepted: 07/17/2023] [Indexed: 09/06/2023]
Abstract
As a pivotal node in modulating various cell behaviors, Rho-associated kinase-1 (ROCK1) has attracted significant attention as a promising therapeutic target in a variety of diseases. Benzamide has been widely reported as a ROCK1 inhibitors in recent years. To better understand its pharmacological properties and to explore its potential inhibitors, a series of ROCK1 inhibitors derived from N-methyl-4-(4-pyrazolidinyl) benzamides (MPBs) were investigated by using three-dimensional quantitative structure-activity relationship (3D-QSAR) models, pharmacophore models, molecular docking, and molecular dynamics (MD) simulation. The comparative Molecular Field Analysis (CoMFA) model (q2 = 0.616, R2 = 0.972, ONC = 4, and r2pred = 0.983) and the best Comparative Molecular Similarity Indices Analysis (CoMSIA) model (q2= 0.740, R2 = 0.982, ONC = 6, and r2pred = 0.824) exhibited reliable predictability with satisfactory validation parameters. In the subsequent virtual screening, VS03 and VS05 were identified to have superior predicted activities and higher docking scores, meanwhile they demonstrated to be reasonably stable in the binding pocket through MD simulations. These results provide a significant theoretical direction for the rational design and development of novel ROCK1 inhibitors.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Qi-Xuan Xu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liang Guo
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuan Li
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhao-Wei Wang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Po Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Guang-Ming Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yang Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
7
|
Sun H, Gao Y, Ma X, Deng Y, Bi L, Li L. Mechanism and application of feedback loops formed by mechanotransduction and histone modifications. Genes Dis 2024; 11:101061. [PMID: 39071110 PMCID: PMC11282412 DOI: 10.1016/j.gendis.2023.06.030] [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: 10/03/2022] [Revised: 03/24/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2024] Open
Abstract
Mechanical stimulation is the key physical factor in cell environment. Mechanotransduction acts as a fundamental regulator of cell behavior, regulating cell proliferation, differentiation, apoptosis, and exhibiting specific signature alterations during the pathological process. As research continues, the role of epigenetic science in mechanotransduction is attracting attention. However, the molecular mechanism of the synergistic effect between mechanotransduction and epigenetics in physiological and pathological processes has not been clarified. We focus on how histone modifications, as important components of epigenetics, are coordinated with multiple signaling pathways to control cell fate and disease progression. Specifically, we propose that histone modifications can form regulatory feedback loops with signaling pathways, that is, histone modifications can not only serve as downstream regulators of signaling pathways for target gene transcription but also provide feedback to regulate signaling pathways. Mechanotransduction and epigenetic changes could be potential markers and therapeutic targets in clinical practice.
Collapse
Affiliation(s)
- Han Sun
- Department of Hematology and Oncology, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Yafang Gao
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Xinyu Ma
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Yizhou Deng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| | - Lintao Bi
- Department of Hematology and Oncology, China-Japan Union Hospital, Jilin University, Changchun, Jilin 130021, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, China
| |
Collapse
|
8
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [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: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
9
|
Shultz KD, Al Anbari YF, Wright NT. I told you to stop: obscurin's role in epithelial cell migration. Biochem Soc Trans 2024; 52:1947-1956. [PMID: 39051125 DOI: 10.1042/bst20240564] [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: 05/13/2024] [Revised: 07/02/2024] [Accepted: 07/11/2024] [Indexed: 07/27/2024]
Abstract
The giant cytoskeletal protein obscurin contains multiple cell signaling domains that influence cell migration. Here, we follow each of these pathways, examine how these pathways modulate epithelial cell migration, and discuss the cross-talk between these pathways. Specifically, obscurin uses its PH domain to inhibit phosphoinositide-3-kinase (PI3K)-dependent migration and its RhoGEF domain to activate RhoA and slow cell migration. While obscurin's effect on the PI3K pathway agrees with the literature, obscurin's effect on the RhoA pathway runs counter to most other RhoA effectors, whose activation tends to lead to enhanced motility. Obscurin also phosphorylates cadherins, and this may also influence cell motility. When taken together, obscurin's ability to modulate three independent cell migration pathways is likely why obscurin knockout cells experience enhanced epithelial to mesenchymal transition, and why obscurin is a frequently mutated gene in several types of cancer.
Collapse
Affiliation(s)
- Kamrin D Shultz
- Department of Chemistry and Biochemistry, James Madison University, 901 Carrier Dr., Harrisonburg, VA 22807, U.S.A
| | - Yasmin F Al Anbari
- Department of Chemistry and Biochemistry, James Madison University, 901 Carrier Dr., Harrisonburg, VA 22807, U.S.A
| | - Nathan T Wright
- Department of Chemistry and Biochemistry, James Madison University, 901 Carrier Dr., Harrisonburg, VA 22807, U.S.A
| |
Collapse
|
10
|
Xu J, Dong K, Bai X, Zhang M, Du Q, Chen L, Yang J. GluOC promotes proliferation and metastasis of TNBC through the ROCK1 signaling pathway. Cancer Cell Int 2024; 24:263. [PMID: 39054484 PMCID: PMC11270849 DOI: 10.1186/s12935-024-03445-8] [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: 02/14/2024] [Accepted: 07/09/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND Triple negative breast cancer (TNBC) is a type of breast cancer that is negative for oestrogen receptor, progesterone receptor and human epidermal growth factor receptor 2, is highly malignant and aggressive, lacks of corresponding targeted therapy, and has a relatively poor prognosis. Therefore, understanding the mechanism of TNBC development and formulating effective treatment strategies for inducing cell death are still urgent tasks in the treatment of TNBC. Research has shown that uncarboxylated osteocalcin can promote the proliferation of prostate cancer, lung adenocarcinoma and TNBC cells, but the mechanism by which GluOC affects TNBC growth and metastasis needs further study. METHODS MDA-MB-231 breast cancer cells were used for in vitro cell analysis. Key target molecules or pathways were identified by RNA sequencing, and migration ability was detected by scratch assays, Transwell assays, cell adhesion assays and western blot analysis. Fluorescence staining, colony detection, qRT‒PCR and flow cytometry were used to detect apoptosis, oxidative stress, the cell cycle and the stemness of cancer cells, and a xenotransplantation model in BALB/C nude mice was used for in vivo analysis. RESULTS This study demonstrated that GluOC facilitates the migration of MDA-MB-231 breast cancer cells through the ROCK1/MYPT1/MLC2 signalling pathway and promotes the proliferation of TNBC cells via the ROCK1/JAK2/PIK3CA/AKT signalling pathway. Experiments in nude mice demonstrated that GluOC promoted tumour cell proliferation and metastasis in tumour-bearing mice, which further clarified the molecular mechanism of TNBC growth and invasion. CONCLUSION Our findings highlight the importance of GluOC in driving TNBC progression and its association with poor patient outcomes. This study clarifies the functional effects of GluOC on TNBC growth, providing insight into the molecular basis of TNBC and potentially providing new ideas for developing targeted therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Jiaojiao Xu
- Medical School, University of Chinese Academy of Sciences, Beijing, 101400, China
| | - Keting Dong
- Medical School, University of Chinese Academy of Sciences, Beijing, 101400, China
| | - Xue Bai
- Medical School, University of Chinese Academy of Sciences, Beijing, 101400, China
| | - Miao Zhang
- Medical School, University of Chinese Academy of Sciences, Beijing, 101400, China
| | - Qian Du
- Medical School, University of Chinese Academy of Sciences, Beijing, 101400, China
| | - Lei Chen
- Medical School, University of Chinese Academy of Sciences, Beijing, 101400, China
| | - Jianhong Yang
- Medical School, University of Chinese Academy of Sciences, Beijing, 101400, China.
| |
Collapse
|
11
|
Khan F, Pitstick L, Lara J, Ventrella R. Rho-Associated Protein Kinase Activity Is Required for Tissue Homeostasis in the Xenopus laevis Ciliated Epithelium. J Dev Biol 2024; 12:17. [PMID: 38921484 PMCID: PMC11204898 DOI: 10.3390/jdb12020017] [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: 05/08/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 06/27/2024] Open
Abstract
Lung epithelial development relies on the proper balance of cell proliferation and differentiation to maintain homeostasis. When this balance is disturbed, it can lead to diseases like cancer, where cells undergo hyperproliferation and then can undergo migration and metastasis. Lung cancer is one of the deadliest cancers, and even though there are a variety of therapeutic approaches, there are cases where treatment remains elusive. The rho-associated protein kinase (ROCK) has been thought to be an ideal molecular target due to its role in activating oncogenic signaling pathways. However, in a variety of cases, inhibition of ROCK has been shown to have the opposite outcome. Here, we show that ROCK inhibition with y-27632 causes abnormal epithelial tissue development in Xenopus laevis embryonic skin, which is an ideal model for studying lung cancer development. We found that treatment with y-27632 caused an increase in proliferation and the formation of ciliated epithelial outgrowths along the tail edge. Our results suggest that, in certain cases, ROCK inhibition can disturb tissue homeostasis. We anticipate that these findings could provide insight into possible mechanisms to overcome instances when ROCK inhibition results in heightened proliferation. Also, these findings are significant because y-27632 is a common pharmacological inhibitor used to study ROCK signaling, so it is important to know that in certain in vivo developmental models and conditions, this treatment can enhance proliferation rather than lead to cell cycle suppression.
Collapse
Affiliation(s)
- Fayhaa Khan
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (F.K.); (J.L.)
| | - Lenore Pitstick
- Department of Biochemistry and Molecular Genetics, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA;
| | - Jessica Lara
- Biomedical Sciences Program, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA; (F.K.); (J.L.)
| | - Rosa Ventrella
- Precision Medicine Program, College of Graduate Studies, Midwestern University, Downers Grove, IL 60515, USA
| |
Collapse
|
12
|
Cho H, Kim K. Multi-functional nanomedicines for combinational cancer immunotherapy that transform cold tumors to hot tumors. Expert Opin Drug Deliv 2024; 21:627-638. [PMID: 38682272 DOI: 10.1080/17425247.2024.2348656] [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: 01/15/2024] [Accepted: 04/24/2024] [Indexed: 05/01/2024]
Abstract
INTRODUCTION Currently, cancer immunotherapy is widely used as a groundbreaking method that can completely cure advanced cancers. However, this new immunotherapy has the challenge of low patient response, which is often due to many patients' tumors having an immunosuppressive environment, known as cold tumors. AREAS COVERED This review aims to introduce various nanomedicine-derived combinational cancer immunotherapy that can transform cold tumor into hot tumors. Initially, we discuss new technologies for combinational immunotherapy based on multifunctional nanomedicines that can deliver combinational immunogenic cell death (ICD) inducers, immune checkpoint blockades (ICBs) and immune modulators (IMs) to targeted tumor tissues at the same time. Ultimately, we highlight how multifunctional nanomedicines for combinational cancer immunotherapy can be used to transform cold tumor into hot tumors against advanced cancers. EXPERT OPINION Nanomedicine-derived combinational cancer immunotherapy for delivering multiple ICD inducers, ICBs, and IMs at the same time is recognized as a new potential technology that can activate tumor immunity and simultaneously increase the therapeutic efficacy of immune cells that can transform effectively the cold tumors into hot tumors. Finally, nanomedicine-derived combinational cancer immunotherapy can solve the serious problems of low therapeutic efficacy that occurs when treating single drug or simple combinational drugs in cancer immunotherapy.
Collapse
Affiliation(s)
- Hanhee Cho
- Graduate School of Pharmaceutical Sciences, College of Pharmacy, Ewha Woman's University, Seoul, Republic of Korea
| | | |
Collapse
|
13
|
Liu X, Cai YD, Chiu JC. Regulation of protein O-GlcNAcylation by circadian, metabolic, and cellular signals. J Biol Chem 2024; 300:105616. [PMID: 38159854 PMCID: PMC10810748 DOI: 10.1016/j.jbc.2023.105616] [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: 07/24/2023] [Revised: 12/12/2023] [Accepted: 12/13/2023] [Indexed: 01/03/2024] Open
Abstract
O-linked β-N-acetylglucosamine (O-GlcNAcylation) is a dynamic post-translational modification that regulates thousands of proteins and almost all cellular processes. Aberrant O-GlcNAcylation has been associated with numerous diseases, including cancer, neurodegenerative diseases, cardiovascular diseases, and type 2 diabetes. O-GlcNAcylation is highly nutrient-sensitive since it is dependent on UDP-GlcNAc, the end product of the hexosamine biosynthetic pathway (HBP). We previously observed daily rhythmicity of protein O-GlcNAcylation in a Drosophila model that is sensitive to the timing of food consumption. We showed that the circadian clock is pivotal in regulating daily O-GlcNAcylation rhythms given its control of the feeding-fasting cycle and hence nutrient availability. Interestingly, we reported that the circadian clock also modulates daily O-GlcNAcylation rhythm by regulating molecular mechanisms beyond the regulation of food consumption time. A large body of work now indicates that O-GlcNAcylation is likely a generalized cellular status effector as it responds to various cellular signals and conditions, such as ER stress, apoptosis, and infection. In this review, we summarize the metabolic regulation of protein O-GlcNAcylation through nutrient availability, HBP enzymes, and O-GlcNAc processing enzymes. We discuss the emerging roles of circadian clocks in regulating daily O-GlcNAcylation rhythm. Finally, we provide an overview of other cellular signals or conditions that impact O-GlcNAcylation. Many of these cellular pathways are themselves regulated by the clock and/or metabolism. Our review highlights the importance of maintaining optimal O-GlcNAc rhythm by restricting eating activity to the active period under physiological conditions and provides insights into potential therapeutic targets of O-GlcNAc homeostasis under pathological conditions.
Collapse
Affiliation(s)
- Xianhui Liu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA
| | - Yao D Cai
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA
| | - Joanna C Chiu
- Department of Entomology and Nematology, College of Agricultural and Environmental Sciences, University of California, Davis, California, USA.
| |
Collapse
|
14
|
Kang JH, Kawano T, Murata M, Toita R. Vascular calcification and cellular signaling pathways as potential therapeutic targets. Life Sci 2024; 336:122309. [PMID: 38042282 DOI: 10.1016/j.lfs.2023.122309] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/21/2023] [Accepted: 11/24/2023] [Indexed: 12/04/2023]
Abstract
Increased vascular calcification (VC) is observed in patients with cardiovascular diseases such as atherosclerosis, diabetes, and chronic kidney disease. VC is divided into three types according to its location: intimal, medial, and valvular. Various cellular signaling pathways are associated with VC, including the Wnt, mitogen-activated protein kinase, phosphatidylinositol-3 kinase/Akt, cyclic nucleotide-dependent protein kinase, protein kinase C, calcium/calmodulin-dependent kinase II, adenosine monophosphate-activated protein kinase/mammalian target of rapamycin, Ras homologous GTPase, apoptosis, Notch, and cytokine signaling pathways. In this review, we discuss the literature concerning the key cellular signaling pathways associated with VC and their role as potential therapeutic targets. Inhibitors to these pathways represent good candidates for use as potential therapeutic agents for the prevention and treatment of VC.
Collapse
Affiliation(s)
- Jeong-Hun Kang
- National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan.
| | - Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Riki Toita
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology (AIST), 1-8-31 Midorigaoka, Ikeda, Osaka, 563-8577, Japan; AIST-Osaka University Advanced Photonics and Biosensing Open Innovation Laboratory, AIST, 2-1 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
15
|
Mukhopadhyay S, Huang HY, Lin Z, Ranieri M, Li S, Sahu S, Liu Y, Ban Y, Guidry K, Hu H, Lopez A, Sherman F, Tan YJ, Lee YT, Armstrong AP, Dolgalev I, Sahu P, Zhang T, Lu W, Gray NS, Christensen JG, Tang TT, Velcheti V, Khodadadi-Jamayran A, Wong KK, Neel BG. Genome-Wide CRISPR Screens Identify Multiple Synthetic Lethal Targets That Enhance KRASG12C Inhibitor Efficacy. Cancer Res 2023; 83:4095-4111. [PMID: 37729426 PMCID: PMC10841254 DOI: 10.1158/0008-5472.can-23-2729] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/22/2023]
Abstract
Non-small lung cancers (NSCLC) frequently (∼30%) harbor KRAS driver mutations, half of which are KRASG12C. KRAS-mutant NSCLC with comutated STK11 and/or KEAP1 is particularly refractory to conventional, targeted, and immune therapy. Development of KRASG12C inhibitors (G12Ci) provided a major therapeutic advance, but resistance still limits their efficacy. To identify genes whose deletion augments efficacy of the G12Cis adagrasib (MRTX-849) or adagrasib plus TNO155 (SHP2i), we performed genome-wide CRISPR/Cas9 screens on KRAS/STK11-mutant NSCLC lines. Recurrent, potentially targetable, synthetic lethal (SL) genes were identified, including serine-threonine kinases, tRNA-modifying and proteoglycan synthesis enzymes, and YAP/TAZ/TEAD pathway components. Several SL genes were confirmed by siRNA/shRNA experiments, and the YAP/TAZ/TEAD pathway was extensively validated in vitro and in mice. Mechanistic studies showed that G12Ci treatment induced gene expression of RHO paralogs and activators, increased RHOA activation, and evoked ROCK-dependent nuclear translocation of YAP. Mice and patients with acquired G12Ci- or G12Ci/SHP2i-resistant tumors showed strong overlap with SL pathways, arguing for the relevance of the screen results. These findings provide a landscape of potential targets for future combination strategies, some of which can be tested rapidly in the clinic. SIGNIFICANCE Identification of synthetic lethal genes with KRASG12C using genome-wide CRISPR/Cas9 screening and credentialing of the ability of TEAD inhibition to enhance KRASG12C efficacy provides a roadmap for combination strategies. See related commentary by Johnson and Haigis, p. 4005.
Collapse
Affiliation(s)
- Suman Mukhopadhyay
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Hsin-Yi Huang
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Ziyan Lin
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, United States
| | - Michela Ranieri
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Shuai Li
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Soumyadip Sahu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yingzhuo Liu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yi Ban
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Kayla Guidry
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Hai Hu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Alfonso Lopez
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Fiona Sherman
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yi Jer Tan
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Yeuan Ting Lee
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Amanda P. Armstrong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Igor Dolgalev
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Priyanka Sahu
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Tinghu Zhang
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | - Wenchao Lu
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | - Nathanael S. Gray
- Department of Chemical and Systems Biology, ChEM-H, Stanford Cancer Institute, School of Medicine, Stanford University, California, United States
| | | | - Tracy T. Tang
- Vivace Therapeutics, Inc., San Mateo, California, United States
| | - Vamsidhar Velcheti
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Alireza Khodadadi-Jamayran
- Applied Bioinformatics Laboratories, Office of Science and Research, New York University Grossman School of Medicine, New York, United States
| | - Kwok-Kin Wong
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| | - Benjamin G. Neel
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, NYU Langone Health, New York, United States
| |
Collapse
|
16
|
Shin YJ, Jo EH, Oh Y, Kim DS, Hyun S, Yu A, Hong HK, Cho YB. Improved Drug-Response Prediction Model of APC Mutant Colon Cancer Patient-Derived Organoids for Precision Medicine. Cancers (Basel) 2023; 15:5531. [PMID: 38067236 PMCID: PMC10705195 DOI: 10.3390/cancers15235531] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 10/08/2024] Open
Abstract
Colorectal cancer is the third most common cancer in the world, with an annual incidence of 2 million cases. The success of first-line chemotherapy plays a crucial role in determining the disease outcome. Therefore, there is an increasing demand for precision medicine to predict drug responses and optimize chemotherapy in order to increase patient survival and reduce the related side effects. Patient-derived organoids have become a popular in vitro screening model for drug-response prediction for precision medicine. However, there is no established correlation between oxaliplatin and drug-response prediction. Here, we suggest that organoid culture conditions can increase resistance to oxaliplatin during drug screening, and we developed a modified medium condition to address this issue. Notably, while previous studies have shown that survivin is a mechanism for drug resistance, our study observed consistent survivin expression irrespective of the culture conditions and oxaliplatin treatment. However, clusterin induced apoptosis inhibition and cell survival, demonstrating a significant correlation with drug resistance. This study's findings are expected to contribute to increasing the accuracy of drug-response prediction in patient-derived APC mutant colorectal cancer organoids, thereby providing reliable precision medicine and improving patient survival rates.
Collapse
Affiliation(s)
- Yong Jae Shin
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea; (Y.J.S.); (E.H.J.); (Y.O.); (D.S.K.); (H.K.H.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Eun Hae Jo
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea; (Y.J.S.); (E.H.J.); (Y.O.); (D.S.K.); (H.K.H.)
| | - Yunjeong Oh
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea; (Y.J.S.); (E.H.J.); (Y.O.); (D.S.K.); (H.K.H.)
| | - Da Som Kim
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea; (Y.J.S.); (E.H.J.); (Y.O.); (D.S.K.); (H.K.H.)
| | - Seungyoon Hyun
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea;
| | - Ahran Yu
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
| | - Hye Kyung Hong
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea; (Y.J.S.); (E.H.J.); (Y.O.); (D.S.K.); (H.K.H.)
| | - Yong Beom Cho
- Innovative Institute for Precision Medicine, Samsung Medical Center, Seoul 06351, Republic of Korea; (Y.J.S.); (E.H.J.); (Y.O.); (D.S.K.); (H.K.H.)
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Republic of Korea;
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University, Seoul 06351, Republic of Korea;
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon-si 16419, Republic of Korea
| |
Collapse
|
17
|
Wu J, Liang Y, Fu P, Feng A, Lu Q, Unwalla HJ, Marciano DP, Black SM, Wang T. Sphingosine-1-Phosphate Receptor 3 Induces Endothelial Barrier Loss via ADAM10-Mediated Vascular Endothelial-Cadherin Cleavage. Int J Mol Sci 2023; 24:16083. [PMID: 38003272 PMCID: PMC10671260 DOI: 10.3390/ijms242216083] [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: 09/27/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/26/2023] Open
Abstract
Mechanical ventilation (MV) is a life-supporting strategy employed in the Intensive Care Unit (ICU). However, MV-associated mechanical stress exacerbates existing lung inflammation in ICU patients, resulting in limited improvement in mortality and a condition known as Ventilator-Induced Lung Injury (VILI). Sphingosine-1-phosphate (S1P) is a circulating bioactive lipid that maintains endothelial integrity primarily through S1P receptor 1 (S1PR1). During VILI, mechanical stress upregulates endothelial S1PR3 levels. Unlike S1PR1, S1PR3 mediates endothelial barrier disruption through Rho-dependent pathways. However, the specific impact of elevated S1PR3 on lung endothelial function, apart from Rho activation, remains poorly understood. In this study, we investigated the effects of S1PR3 in endothelial pathobiology during VILI using an S1PR3 overexpression adenovirus. S1PR3 overexpression caused cytoskeleton rearrangement, formation of paracellular gaps, and a modified endothelial response towards S1P. It resulted in a shift from S1PR1-dependent barrier enhancement to S1PR3-dependent barrier disruption. Moreover, S1PR3 overexpression induced an ADAM10-dependent cleavage of Vascular Endothelial (VE)-cadherin, which hindered endothelial barrier recovery. S1PR3-induced cleavage of VE-cadherin was at least partially regulated by S1PR3-mediated NFκB activation. Additionally, we employed an S1PR3 inhibitor TY-52156 in a murine model of VILI. TY-52156 effectively attenuated VILI-induced increases in bronchoalveolar lavage cell counts and protein concentration, suppressed the release of pro-inflammatory cytokines, and inhibited lung inflammation as assessed via a histological evaluation. These findings confirm that mechanical stress associated with VILI increases S1PR3 levels, thereby altering the pulmonary endothelial response towards S1P and impairing barrier recovery. Inhibiting S1PR3 is validated as an effective therapeutic strategy for VILI.
Collapse
Affiliation(s)
- Jialin Wu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Ying Liang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Panfeng Fu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Anlin Feng
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Qing Lu
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
| | - Hoshang J. Unwalla
- Department of Immunology and Nanomedicine, Florida International University, Miami, FL 33199, USA
| | - David P. Marciano
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Cellular Biology and Pharmacology, Florida International University, Miami, FL 33199, USA
| | - Stephen M. Black
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular Biology and Pharmacology, Florida International University, Miami, FL 33199, USA
| | - Ting Wang
- Center for Translational Science, Florida International University, Port Saint Lucie, FL 34987, USA
- Department of Environmental Health Sciences, Florida International University, Miami, FL 33199, USA
- Department of Cellular Biology and Pharmacology, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
18
|
Xu X, Zhang G, Li Z, Li D, Chen R, Huang C, Li Y, Li B, Yu H, Chu XM. MINOCA biomarkers: Non-atherosclerotic aspects. Clin Chim Acta 2023; 551:117613. [PMID: 37871762 DOI: 10.1016/j.cca.2023.117613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Myocardial infarction in the absence of obstructive coronary artery disease (MINOCA) is an important subtype of myocardial infarction. Although comprising less than 50% stenosis in the main epicardial coronary arteries, it constitutes a severe health risk. A variety of approaches have been recommended, but definitive diagnosis remains elusive. In addition, the lack of a comprehensive understanding of underlying pathophysiology makes clinical management difficult and unpredictable. This review highlights ongoing efforts to identify relevant biomarkers in MINOCA to improve diagnosis, individualize treatment and better predict outcomes.
Collapse
Affiliation(s)
- Xiaojian Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China.
| | - Guoliang Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China.
| | - Zhaoqing Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China.
| | - Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China.
| | - Ruolan Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China.
| | - Chao Huang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China.
| | - Yonghong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China.
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao 266000, China; Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China.
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China.
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao 266100, China; The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao 266071, China.
| |
Collapse
|
19
|
Hua L, Anjum F, Shafie A, Ashour AA, Almalki AA, Alqarni AA, Banjer HJ, Almaghrabi SA, He S, Xu N. Identifying promising GSK3β inhibitors for cancer management: a computational pipeline combining virtual screening and molecular dynamics simulations. Front Chem 2023; 11:1200490. [PMID: 37284581 PMCID: PMC10239944 DOI: 10.3389/fchem.2023.1200490] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 05/05/2023] [Indexed: 06/08/2023] Open
Abstract
Glycogen synthase kinase-3 (GSK3β), a serine/threonine protein kinase, has been discovered as a novel target for anticancer drugs. Although GSK3β is involved in multiple pathways linked to the etiology of various cancers, no specific GSK3β inhibitor has been authorized for cancer therapy. Most of its inhibitors have toxicity effects therefore, there is a need to develop safe and more potent inhibitors. In this study, a library of 4,222 anti-cancer compounds underwent rigorous computational screening to identify potential candidates for targeting the binding pocket of GSK3β. The screening process involved various stages, including docking-based virtual screening, physicochemical and ADMET analysis, and molecular dynamics simulations. Ultimately, two hit compounds, BMS-754807 and GSK429286A, were identified as having high binding affinities to GSK3β. BMS-754807 and GSK429286A exhibited binding affinities of -11.9, and -9.8 kcal/mol, respectively, which were greater than that of the positive control (-7.6 kcal/mol). Further, molecular dynamics simulations for 100 ns were employed to optimize the interaction between the compounds and GSK3β, and the simulations demonstrated that the interaction was stable and consistent throughout the study. These hits were also anticipated to have good drug-like properties. Finally, this study suggests that BMS-754807 and GSK429286A may undergo experimental validation to evaluate their potential as cancer treatments in clinical settings.
Collapse
Affiliation(s)
- Libo Hua
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Amal Adnan Ashour
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, Faculty of Dentistry, Taif University, Taif, Saudi Arabia
| | - Abdulraheem Ali Almalki
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Ali Abdullah Alqarni
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, Faculty of Dentistry, Taif University, Taif, Saudi Arabia
| | - Hamsa Jameel Banjer
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Sarah Abdullah Almaghrabi
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Center for Innovations in Personalized Medicine (CIPM), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Shan He
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan, China
- Institute for Nano Scale and Technology, College of Science and Engineering, Flinders University, Bedford Park, SA, Australia
- College of Engineering, Information Technology and Environment, Charles Darwin University, Darwin, NT, Australia
| | - Nenggui Xu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acupuncture Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
20
|
Tennstedt P, Oh-Hohenhorst SJ. Advances, Limitations and Future Challenges in the Management of Immunotherapy for Hematological Diseases and Solid Tumors. Int J Mol Sci 2023; 24:ijms24108812. [PMID: 37240158 DOI: 10.3390/ijms24108812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Our immune system is able to attack cancer cells by recognizing cellular mistakes and destroying them [...].
Collapse
Affiliation(s)
- Pierre Tennstedt
- Martini-Klinik Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| | - Su Jung Oh-Hohenhorst
- Martini-Klinik Prostate Cancer Center, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany
| |
Collapse
|
21
|
Shakiba D, Genin GM, Zustiak SP. Mechanobiology of cancer cell responsiveness to chemotherapy and immunotherapy: Mechanistic insights and biomaterial platforms. Adv Drug Deliv Rev 2023; 196:114771. [PMID: 36889646 PMCID: PMC10133187 DOI: 10.1016/j.addr.2023.114771] [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: 08/30/2022] [Revised: 12/17/2022] [Accepted: 03/03/2023] [Indexed: 03/08/2023]
Abstract
Mechanical forces are central to how cancer treatments such as chemotherapeutics and immunotherapies interact with cells and tissues. At the simplest level, electrostatic forces underlie the binding events that are critical to therapeutic function. However, a growing body of literature points to mechanical factors that also affect whether a drug or an immune cell can reach a target, and to interactions between a cell and its environment affecting therapeutic efficacy. These factors affect cell processes ranging from cytoskeletal and extracellular matrix remodeling to transduction of signals by the nucleus to metastasis of cells. This review presents and critiques the state of the art of our understanding of how mechanobiology impacts drug and immunotherapy resistance and responsiveness, and of the in vitro systems that have been of value in the discovery of these effects.
Collapse
Affiliation(s)
- Delaram Shakiba
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, USA
| | - Guy M Genin
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Mechanical Engineering and Materials Science, Washington University, St. Louis, MO, USA.
| | - Silviya P Zustiak
- NSF Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, USA; Department of Biomedical Engineering, School of Science and Engineering, Saint Louis University, St. Louis, MO, USA.
| |
Collapse
|
22
|
Guo Y, Kanamarlapudi V. Molecular Analysis of SARS-CoV-2 Spike Protein-Induced Endothelial Cell Permeability and vWF Secretion. Int J Mol Sci 2023; 24:5664. [PMID: 36982738 PMCID: PMC10053386 DOI: 10.3390/ijms24065664] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
Coronavirus disease COVID-19, which is caused by severe acute respiratory syndrome coronavirus SARS-CoV-2, has become a worldwide pandemic in recent years. In addition to being a respiratory disease, COVID-19 is a 'vascular disease' since it causes a leaky vascular barrier and increases blood clotting by elevating von Willebrand factor (vWF) levels in the blood. In this study, we analyzed in vitro how the SARS-CoV-2 spike protein S1 induces endothelial cell (EC) permeability and its vWF secretion, and the underlying molecular mechanism for it. We showed that the SARS-CoV-2 spike protein S1 receptor-binding domain (RBD) is sufficient to induce endothelial permeability and vWF-secretion through the angiotensin-converting enzyme (ACE)2 in an ADP-ribosylation factor (ARF)6 activation-dependent manner. However, the mutants, including those in South African and South Californian variants of SARS-CoV-2, in the spike protein did not affect its induced EC permeability and vWF secretion. In addition, we have identified a signaling cascade downstream of ACE2 for the SARS-CoV-2 spike protein-induced EC permeability and its vWF secretion by using pharmacological inhibitors. The knowledge gained from this study could be useful in developing novel drugs or repurposing existing drugs for treating infections of SARS-CoV-2, particularly those strains that respond poorly to the existing vaccines.
Collapse
|
23
|
Barcelo J, Samain R, Sanz-Moreno V. Preclinical to clinical utility of ROCK inhibitors in cancer. Trends Cancer 2023; 9:250-263. [PMID: 36599733 DOI: 10.1016/j.trecan.2022.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/25/2022] [Accepted: 12/02/2022] [Indexed: 01/03/2023]
Abstract
ROCK belongs to the AGC family of Ser/Thr protein kinases that are involved in many cellular processes. ROCK-driven actomyosin contractility regulates cytoskeletal dynamics underpinning cell migration, proliferation, and survival in many cancer types. ROCK1/2 play key protumorigenic roles in several subtypes and stages of cancer development. Therefore, successfully targeting ROCK and its downstream effectors presents an interesting avenue for cancer treatment. Because local use of ROCK inhibitors will reduce the side effects of systemic administration, we propose different therapeutic strategies and latest-generation ROCK inhibitors for use in the clinic.
Collapse
Affiliation(s)
- Jaume Barcelo
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Remi Samain
- Barts Cancer Institute, Queen Mary University of London, London, UK
| | | |
Collapse
|
24
|
Zeng C, Long M, Lu Y. Monensin synergizes with chemotherapy in uveal melanoma through suppressing RhoA. Immunopharmacol Immunotoxicol 2023; 45:35-42. [PMID: 36043455 DOI: 10.1080/08923973.2022.2112219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE Uveal melanoma (UM) is the common primary cancer of the eye and new treatments are needed. Substantial evidence has shown that an antibiotic monensin is an attractive candidate for the development of anti-cancer drug. In this study, we investigated the potential of repositioning monensin for the treatment of UM in the pre-clinical setting. MATERIALS AND METHODS Cellular activity assays were performed using multiple cell lines representing UM models with different cellular origins and genetic profiling and normal cells as control. Combination studies were performed using Chou-Talalay method. Mechanism studies were performed using immunoblotting and ELISA. RESULTS Monensin was effective against all tested UM cell lines and less effective against normal fibroblast cells. Monensin induced G0/G1 arrest and thus decreased S phase, leading to UM cell growth inhibition. It also inhibited migration and induced apoptosis in UM cells. In addition, the combination of monensin and dacarbazine was synergistic in targeting UM cells. Our mechanistic studies showed that monensin specifically decreased activity of RhoA without affecting other small GTPases, such as Ras and Rac1. Consistently, monensin decreased phosphorylation of downstream effectors of RhoA signaling, including ROCK, MYPT1 and MLC. Rescue studies using RhoA activator calpeptin showed that calpeptin significantly abolished the inhibitory effects of monensin on RhoA activity, proliferation, migration and survival, confirming that RhoA is the target of monensin in UM cells. CONCLUSIONS Our study demonstrates that monensin is a potent inhibitor of UM and synergizes with chemotherapy, via suppressing RhoA activity and RhoA-mediated signaling. Our findings suggest that monensin may be a potential lead compound for further development into a drug for UM treatment.
Collapse
Affiliation(s)
- Chaoxia Zeng
- Hainan Eye Hospital and Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Haikou, PR China
| | - Mingxia Long
- Department of Nursing, Wuhan Third Hospital-Tongren Hospital of Wuhan University, Wuhan, PR China
| | - Ying Lu
- Department of Integrated Traditional Chinese and Western Medicine, Wuhan Third Hospital -Tongren Hospital of Wuhan University, Wuhan, PR China
| |
Collapse
|
25
|
Hyeon DY, Nam D, Han Y, Kim DK, Kim G, Kim D, Bae J, Back S, Mun DG, Madar IH, Lee H, Kim SJ, Kim H, Hyun S, Kim CR, Choi SA, Kim YR, Jeong J, Jeon S, Choo YW, Lee KB, Kwon W, Choi S, Goo T, Park T, Suh YA, Kim H, Ku JL, Kim MS, Paek E, Park D, Jung K, Baek SH, Jang JY, Hwang D, Lee SW. Proteogenomic landscape of human pancreatic ductal adenocarcinoma in an Asian population reveals tumor cell-enriched and immune-rich subtypes. NATURE CANCER 2023; 4:290-307. [PMID: 36550235 DOI: 10.1038/s43018-022-00479-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/04/2022] [Indexed: 12/24/2022]
Abstract
We report a proteogenomic analysis of pancreatic ductal adenocarcinoma (PDAC). Mutation-phosphorylation correlations identified signaling pathways associated with somatic mutations in significantly mutated genes. Messenger RNA-protein abundance correlations revealed potential prognostic biomarkers correlated with patient survival. Integrated clustering of mRNA, protein and phosphorylation data identified six PDAC subtypes. Cellular pathways represented by mRNA and protein signatures, defining the subtypes and compositions of cell types in the subtypes, characterized them as classical progenitor (TS1), squamous (TS2-4), immunogenic progenitor (IS1) and exocrine-like (IS2) subtypes. Compared with the mRNA data, protein and phosphorylation data further classified the squamous subtypes into activated stroma-enriched (TS2), invasive (TS3) and invasive-proliferative (TS4) squamous subtypes. Orthotopic mouse PDAC models revealed a higher number of pro-tumorigenic immune cells in TS4, inhibiting T cell proliferation. Our proteogenomic analysis provides significantly mutated genes/biomarkers, cellular pathways and cell types as potential therapeutic targets to improve stratification of patients with PDAC.
Collapse
Affiliation(s)
- Do Young Hyeon
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Dowoon Nam
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Youngmin Han
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Duk Ki Kim
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea.,Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Seoul, Republic of Korea
| | - Gibeom Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.,Creative Research Initiatives Center for Epigenetic Code and Diseases, Seoul National University, Seoul, Republic of Korea
| | - Daeun Kim
- Department of Biological Sciences, College of Natural Sciences and Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Jingi Bae
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Seunghoon Back
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Dong-Gi Mun
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Inamul Hasan Madar
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Hangyeore Lee
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Su-Jin Kim
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Hokeun Kim
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea
| | - Sangyeop Hyun
- Department of Biological Sciences, College of Natural Sciences and Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea
| | - Chang Rok Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.,Creative Research Initiatives Center for Epigenetic Code and Diseases, Seoul National University, Seoul, Republic of Korea
| | - Seon Ah Choi
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.,Creative Research Initiatives Center for Epigenetic Code and Diseases, Seoul National University, Seoul, Republic of Korea
| | - Yong Ryoul Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.,Creative Research Initiatives Center for Epigenetic Code and Diseases, Seoul National University, Seoul, Republic of Korea
| | - Juhee Jeong
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Suwan Jeon
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Yeon Woong Choo
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Kyung Bun Lee
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | - Wooil Kwon
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seunghyuk Choi
- Department of Computer Science, Hanyang University, Seoul, Republic of Korea
| | - Taewan Goo
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
| | - Taesung Park
- Department of Statistics, Seoul National University, Seoul, Republic of Korea
| | - Young-Ah Suh
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Hongbeom Kim
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Ja-Lok Ku
- Korean Cell Line Bank, Laboratory of Cell Biology, Cancer Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Eunok Paek
- Department of Computer Science, Hanyang University, Seoul, Republic of Korea
| | - Daechan Park
- Department of Biological Sciences, College of Natural Sciences and Department of Molecular Science and Technology, Ajou University, Suwon, Republic of Korea.
| | - Keehoon Jung
- Department of Anatomy and Cell Biology and Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul, Republic of Korea. .,Institute of Allergy and Clinical Immunology, Seoul National University Medical Research Center, Seoul, Republic of Korea.
| | - Sung Hee Baek
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea. .,Creative Research Initiatives Center for Epigenetic Code and Diseases, Seoul National University, Seoul, Republic of Korea.
| | - Jin-Young Jang
- Department of Surgery and Cancer Research Institute, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Daehee Hwang
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea.
| | - Sang-Won Lee
- Department of Chemistry and Center for Proteogenome Research, Korea University, Seoul, Republic of Korea.
| |
Collapse
|
26
|
George S, Martin JAJ, Graziani V, Sanz-Moreno V. Amoeboid migration in health and disease: Immune responses versus cancer dissemination. Front Cell Dev Biol 2023; 10:1091801. [PMID: 36699013 PMCID: PMC9869768 DOI: 10.3389/fcell.2022.1091801] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 12/15/2022] [Indexed: 01/07/2023] Open
Abstract
Cell migration is crucial for efficient immune responses and is aberrantly used by cancer cells during metastatic dissemination. Amoeboid migrating cells use myosin II-powered blebs to propel themselves, and change morphology and direction. Immune cells use amoeboid strategies to respond rapidly to infection or tissue damage, which require quick passage through several barriers, including blood, lymph and interstitial tissues, with complex and varied environments. Amoeboid migration is also used by metastatic cancer cells to aid their migration, dissemination and survival, whereby key mechanisms are hijacked from professionally motile immune cells. We explore important parallels observed between amoeboid immune and cancer cells. We also consider key distinctions that separate the lifespan, state and fate of these cell types as they migrate and/or fulfil their function. Finally, we reflect on unexplored areas of research that would enhance our understanding of how tumour cells use immune cell strategies during metastasis, and how to target these processes.
Collapse
|
27
|
Piccolo S, Panciera T, Contessotto P, Cordenonsi M. YAP/TAZ as master regulators in cancer: modulation, function and therapeutic approaches. NATURE CANCER 2023; 4:9-26. [PMID: 36564601 PMCID: PMC7614914 DOI: 10.1038/s43018-022-00473-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 10/31/2022] [Indexed: 12/24/2022]
Abstract
Our understanding of the function of the transcriptional regulators YAP and TAZ (YAP/TAZ) in cancer is advancing. In this Review, we provide an update on recent progress in YAP/TAZ biology, their regulation by Hippo signaling and mechanotransduction and highlight open questions. YAP/TAZ signaling is an addiction shared by multiple tumor types and their microenvironments, providing many malignant attributes. As such, it represents an important vulnerability that may offer a broad window of therapeutic efficacy, and here we give an overview of the current treatment strategies and pioneering clinical trials.
Collapse
Affiliation(s)
- Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- IFOM-ETS, the AIRC Institute of Molecular Oncology, Milan, Italy.
| | - Tito Panciera
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | | |
Collapse
|
28
|
Tang Y, Ye C, Zeng J, Zhu P, Cheng S, Zeng W, Yang B, Liu Y, Yu Y. Identification of a basement membrane-based risk scoring system for prognosis prediction and individualized therapy in clear cell renal cell carcinoma. Front Genet 2023; 14:1038924. [PMID: 36816030 PMCID: PMC9935575 DOI: 10.3389/fgene.2023.1038924] [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: 09/07/2022] [Accepted: 01/20/2023] [Indexed: 02/05/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) belongs to one of the 10 most frequently diagnosed cancers worldwide and has a poor prognosis at the advanced stage. Although multiple therapeutic agents have been proven to be curative in ccRCC, their clinical application was limited due to the lack of reliable biomarkers. Considering the important role of basement membrane (BM) in tumor metastasis and TME regulation, we investigated the expression of BM-related genes in ccRCC and identified prognostic BM genes through differentially expression analysis and univariate cox regression analysis. Then, BM-related ccRCC subtypes were recognized through consensus non-negative matrix factorization based on the prognostic BM genes and evaluated with regard to clinical and TME features. Next, utilizing the differentially expressed genes between the BM-related subtypes, a risk scoring system BMRS was established after serial analysis of univariate cox regression analysis, lasso regression analysis, and multivariate cox regression analysis. Time-dependent ROC curve revealed the satisfactory prognosis predictive capacity of BMRS with internal, and external validation. Multivariate analysis proved the independent predictive ability of BMRS and a BMRS-based nomogram was constructed for clinical application. Some featured mutants were discovered through genomic analysis of the BMRS risk groups. Meanwhile, the BMRS groups were found to have distinct immune scores, immune cell infiltration levels, and immune-related functions. Moreover, with the help of data from The Cancer Immunome Atlas (TCIA) and Genomics of Drug Sensitivity in Cancer (GDSC), the potential of BMRS in predicting therapeutic response was evaluated and some possible therapeutic compounds were proposed through ConnectivityMap (CMap). For the practicability of BMRS, we validated the expression of BMRS-related genes in clinical samples. After all, we identified BM-related ccRCC subtypes with distinct clinical and TME features and constructed a risk scoring system for the prediction of prognosis, therapeutic responses, and potential therapeutic agents of ccRCC. As ccRCC systemic therapy continues to evolve, the risk scoring system BMRS we reported may assist in individualized medication administration.
Collapse
Affiliation(s)
- Yanlin Tang
- Shantou University Medical College, Shantou, China
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chujin Ye
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jiayi Zeng
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ping Zhu
- Department of Immunology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Shouyu Cheng
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Weinan Zeng
- Shantou University Medical College, Shantou, China
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Bowen Yang
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yanjun Liu
- Department of Immunology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
- *Correspondence: Yuming Yu, ; Yanjun Liu,
| | - Yuming Yu
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- *Correspondence: Yuming Yu, ; Yanjun Liu,
| |
Collapse
|
29
|
Carigga Gutierrez NM, Pujol-Solé N, Arifi Q, Coll JL, le Clainche T, Broekgaarden M. Increasing cancer permeability by photodynamic priming: from microenvironment to mechanotransduction signaling. Cancer Metastasis Rev 2022; 41:899-934. [PMID: 36155874 DOI: 10.1007/s10555-022-10064-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 09/06/2022] [Indexed: 01/25/2023]
Abstract
The dense cancer microenvironment is a significant barrier that limits the penetration of anticancer agents, thereby restraining the efficacy of molecular and nanoscale cancer therapeutics. Developing new strategies to enhance the permeability of cancer tissues is of major interest to overcome treatment resistance. Nonetheless, early strategies based on small molecule inhibitors or matrix-degrading enzymes have led to disappointing clinical outcomes by causing increased chemotherapy toxicity and promoting disease progression. In recent years, photodynamic therapy (PDT) has emerged as a novel approach to increase the permeability of cancer tissues. By producing excessive amounts of reactive oxygen species selectively in the cancer microenvironment, PDT increases the accumulation, penetration depth, and efficacy of chemotherapeutics. Importantly, the increased cancer permeability has not been associated to increased metastasis formation. In this review, we provide novel insights into the mechanisms by which this effect, called photodynamic priming, can increase cancer permeability without promoting cell migration and dissemination. This review demonstrates that PDT oxidizes and degrades extracellular matrix proteins, reduces the capacity of cancer cells to adhere to the altered matrix, and interferes with mechanotransduction pathways that promote cancer cell migration and differentiation. Significant knowledge gaps are identified regarding the involvement of critical signaling pathways, and to which extent these events are influenced by the complicated PDT dosimetry. Addressing these knowledge gaps will be vital to further develop PDT as an adjuvant approach to improve cancer permeability, demonstrate the safety and efficacy of this priming approach, and render more cancer patients eligible to receive life-extending treatments.
Collapse
Affiliation(s)
| | - Núria Pujol-Solé
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Qendresa Arifi
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Jean-Luc Coll
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France
| | - Tristan le Clainche
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France.
| | - Mans Broekgaarden
- Université Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, 38000, Grenoble, France.
| |
Collapse
|
30
|
Gu Z, Yan Y, Yao H, Lin K, Li X. Targeting the LPA1 signalling pathway for fibrosis therapy: a patent review (2010-present). Expert Opin Ther Pat 2022; 32:1097-1122. [PMID: 36175357 DOI: 10.1080/13543776.2022.2130753] [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] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Fibrosis is a disease that damages organs and even causes death. Because of the complicated pathogenesis, the development of drugs for fibrosis is challenging. In the lysophosphatidic acid receptor type 1 (LPA1) signalling pathway, LPA1 and its downstream Rho-associated coiled-coil forming protein kinase (ROCK) are related to the process of fibrosis. Targeting LPA1 signalling pathway is a potential strategy for the treatment of fibrosis. AREA COVERED This review describes the process of fibrosis mediated by the LPA1 signalling pathway and then summarizes LPA1 antagonist patents reported since 2010 and ROCK inhibitor patents since 2017 according to their scaffolds based on the Cortellis Drug Discovery Intelligence database. Information on LPA1 antagonists entering clinical trials is integrated. EXPERT OPINION Over the past decade, a large number of antagonists targeting the LPA1 signalling pathway have been patented for fibrosis therapy. A limited number of compounds have entered clinical trials. Different companies and research groups have used different scaffolds when designing compounds for fibrosis therapy. Therefore, LPA1 and ROCK are competitive targets for the development of new therapies for fibrosis to provide a potential treatment method for fibrosis in the future.
Collapse
Affiliation(s)
- Zhihao Gu
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Yong Yan
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Hequan Yao
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Kejiang Lin
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Xuanyi Li
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
31
|
Schaefer A, Der CJ. RHOA takes the RHOad less traveled to cancer. Trends Cancer 2022; 8:655-669. [PMID: 35568648 DOI: 10.1016/j.trecan.2022.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 04/08/2022] [Accepted: 04/14/2022] [Indexed: 10/18/2022]
Abstract
RAS and RHO GTPases function as signaling nodes that regulate diverse cellular processes. Whereas RAS mutations were identified in human cancers nearly four decades ago, only recently have mutations in two RHO GTPases, RAC1 and RHOA, been identified in cancer. RAS mutations are found in a diverse spectrum of human cancer types. By contrast, RAC1 and RHOA mutations are associated with distinct and restricted cancer types. Despite a conservation of RAS and RAC1 residues that comprise mutational hotspots, RHOA mutations comprise highly divergent hotspots. Whereas RAS and RAC1 act as oncogenes, RHOA may act as both an oncogene and a tumor suppressor. Thus, while RAS and RHO each take different mutational paths, they arrive at the same biological destination as cancer drivers.
Collapse
Affiliation(s)
- Antje Schaefer
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Department of Pharmacology, Chapel Hill, NC 27599, USA
| | - Channing J Der
- University of North Carolina at Chapel Hill, Lineberger Comprehensive Cancer Center, Department of Pharmacology, Chapel Hill, NC 27599, USA.
| |
Collapse
|