1
|
Lin Y, Xu T, Jiang Q, Chen J, Zhang H, Reinach PS, Yan D, Qu J, Chen S. Fluorometholone inhibits corneal epithelial proliferation, migration via targeting Rho GTPases: RhoA, Rac1, and Cdc42. Exp Eye Res 2025; 256:110397. [PMID: 40268157 DOI: 10.1016/j.exer.2025.110397] [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: 01/14/2025] [Revised: 04/07/2025] [Accepted: 04/21/2025] [Indexed: 04/25/2025]
Abstract
Abnormal corneal epithelial hyperplasia is a common complication following refractive surgery. 0.1 % fluorometholone (FML) eye drops are commonly used for treatment. However, their efficacy varies among patients, potentially attributed to differences in the patient's microenvironment. The underlying reason remains incompletely understood. This study aimed to elucidate the molecular mechanisms of FML's action on corneal epithelial cells (CECs). The effects of FML on the cell viability, proliferation, cell cycle, and migration of human corneal epithelial cells (HCECs) were evaluated using MTS assay, EdU staining, flow cytometry, and scratch assay, respectively. Mouse corneal sections were immunofluorescently stained to assess cell proliferation. A corneal wound model, monitored by slit-lamp photography, was utilized to evaluate the impact of FML on wound healing. Gene expression alterations were detected via RNA sequencing. RT-qPCR and Western blot were employed to validate gene and protein expression in HCECs and mouse corneal epithelia. Proteomic analysis was conducted on tear samples from patients. FML treatment significantly inhibited CEC proliferation, migration, and wound healing. At the molecular level, FML treatment led to a remarkable downregulation of RhoA, Rac1, and Cdc42. Correspondingly, reductions in the downstream Erk and NF-κB signaling pathways were observed in both HCECs and mouse corneal epithelia. Moreover, these pathways were similarly downregulated in tear samples from clinical patients. In conclusion, FML inhibits CEC proliferation and migration by modulating the Rho GTPase signaling network, especially through RhoA/Rac1/Cdc42, thereby suppressing the Erk/NF-κB pathway.
Collapse
Affiliation(s)
- Yong Lin
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Tianyi Xu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Qiuruo Jiang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jialu Chen
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hua Zhang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Peter Sol Reinach
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Dongsheng Yan
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jia Qu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| | - Shihao Chen
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
2
|
Sun R, Zhao Y, Liu Y, Zhang M, Qiu Z, Ma X, Wei L, Lu W, Liu Z, Jiang J. Extracellular matrix stiffness in endometrial cancer: driving progression and modulating treatment sensitivity via the ROCK1/YAP1 axis. Cell Death Dis 2025; 16:380. [PMID: 40368918 PMCID: PMC12078694 DOI: 10.1038/s41419-025-07697-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 04/16/2025] [Accepted: 04/25/2025] [Indexed: 05/16/2025]
Abstract
Endometrial cancer (EC) is among the most prevalent gynecological malignancies, with advanced or recurrent cases posing significant treatment challenges due to limited responses to conventional therapies. Growing evidence highlights the critical role of extracellular matrix (ECM) stiffness in driving tumor progression by shaping the tumor microenvironment. In this study, we demonstrate that ECM stiffness is significantly higher in EC tissues compared to normal endometrium, correlating with elevated expression of ROCK1, a mechanosensitive kinase. Using atomic force microscopy (AFM), we quantified ECM stiffness, while polyacrylamide gels with varying stiffness were employed to mimic ECM conditions in vitro. Bioinformatics analyses, immunofluorescence, Western blotting, and co-immunoprecipitation experiments revealed that ROCK1 modulates the phosphorylation of YAP1, promoting its nuclear localization and transcriptional activity, thereby driving aggressive tumor behaviors, including enhanced proliferation, migration, invasion, and reduced apoptosis. Pharmacological inhibition of ROCK1 with Y-27632 mitigated these effects, suppressing tumor growth, restoring apoptosis, and inducing cell cycle arrest. Treatment with Y-27632 improved sensitivity to chemotherapy and radiotherapy, and significantly enhanced macrophage-mediated phagocytosis, thereby boosting anti-tumor immune responses. In hormone-resistant EC cells, ROCK1 inhibition restored sensitivity to progesterone therapy. Notably, in vivo experiments in a xenograft mouse model confirmed the therapeutic potential of Y-27632, as combination therapy with progesterone showed superior tumor-suppressive effects compared to monotherapy. These findings underscore the dual role of ECM stiffness and ROCK1 in driving tumor progression and influencing treatment outcomes. By elucidating the relationship between ECM stiffness, ROCK1/YAP1 signaling, and treatment sensitivity, this study highlights the potential of targeting the ROCK1/YAP1 axis as a therapeutic strategy. ROCK1 serves as both a biomarker for prognosis and a target for improving personalized treatment approaches, offering new avenues to enhance clinical outcomes for EC patients.
Collapse
Affiliation(s)
- Rui Sun
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Ying Zhao
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Yao Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Mengyao Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
| | - Ziyi Qiu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaohong Ma
- Department of Obstetrics and Gynecology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Lina Wei
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Wei Lu
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Zhiming Liu
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.
| | - Jie Jiang
- Department of Obstetrics and Gynecology, Qilu Hospital of Shandong University, Jinan, China.
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
3
|
Chen R, Zhang R, Ke F, Guo X, Zeng F, Liu Q. Mechanisms of breast cancer metastasis: the role of extracellular matrix. Mol Cell Biochem 2025; 480:2771-2796. [PMID: 39652293 DOI: 10.1007/s11010-024-05175-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/23/2024] [Indexed: 05/03/2025]
Abstract
The components of the extracellular matrix (ECM) are dynamic, and they mediate mechanical signals that modulate cellular behaviors. Disruption of the ECM can induce the migration and invasion of cancer cells via specific signaling pathways and cytokines. Metastasis is a leading cause of high mortality in malignancies, and early intervention can improve survival rates. However, breast cancer is frequently diagnosed subsequent to metastasis, resulting in poor prognosis and distant metastasis poses substantial hurdles in therapy. In breast cancer, there is notable tissue remodeling of ECM proteins, with several identified as essential components for metastasis. Moreover, specific ECM molecules, receptors, enzymes, and various signaling pathways play crucial roles in breast cancer metastasis, drug treatment, and resistance. The in-depth consideration of these elements could provide potential therapeutic targets to enhance the survival rates and quality of life for breast cancer patients. This review explores the mechanisms by which alterations in the ECM contribute to breast cancer metastasis and discusses current clinical applications targeting ECM in breast cancer treatment, offering valuable perspectives for future ECM-based therapies.
Collapse
Affiliation(s)
- Rui Chen
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Ranqi Zhang
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China
| | - Famin Ke
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Xiurong Guo
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China
| | - Fancai Zeng
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| | - Qiuyu Liu
- School of Pharmacy, Southwest Medical University, Luzhou, 646000, China.
- Laboratory of Biochemistry and Molecular Biology, School of Basic Medical Science, Southwest Medical University, Luzhou, 646000, China.
| |
Collapse
|
4
|
Wen J, Sun Y, Ma L, Zu T, Wang N, Zhang T, Liang J, Zhang Y, Lu H, Wu Y, Zhang S. Y-27632 Suppresses the Growth and Migration of Oral Squamous Cell Carcinoma, but Upregulates Autophagy by Suppressing mTOR Effectors. J Oral Pathol Med 2025; 54:207-216. [PMID: 39921531 DOI: 10.1111/jop.13603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 12/04/2024] [Accepted: 01/01/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND The Rho-associated protein kinase (ROCK) inhibitor Y-27632 is a potential immunotherapeutic agent for cancer treatment. Y-27632 blocks the growth and migration of oral squamous cell carcinoma (OSCC) CAL-27 cells. However, detailed studies on the underlying mechanisms have not yet been reported. METHODS We investigated the effects of Y-27632 on the proliferation, migration, and invasion of OSCC cells (CAL-27, SCC-4, and SCC-9) using the Cell Counting Kit-8 assay, ethynyl-2'-deoxyuridine staining, cell scratch, and transwell assay in vitro. Next, ROCK1/2 was knocked down using siRNA to confirm that the effects of Y-27632 were mediated by the inhibition of ROCK activity. A xenograft mouse model was used to verify the effects of Y-27632 in vivo. The mechanisms underlying Y-27632-induced tumor suppression were detected using western blotting and qRT-PCR. RESULTS Our data demonstrated that Y-27632 potently inhibited OSCC cells (CAL-27, SCC-4, and SCC-9) by inhibiting ROCK activity. In vivo assays confirmed that Y-27632 suppressed OSCC growth by reducing cell proliferation. Biochemical assays demonstrated that Y-27632 inactivated the AKT pathway, and treatment with SC79, an AKT activator, rescued the cell growth and migration inhibition elicited by Y-27632. Further investigation revealed that Y-27632 enhanced autophagy by suppressing the AKT/mTOR pathway. CONCLUSION Our study demonstrated that Y-27632 significantly suppressed the growth and migration of OSCC cells and upregulated autophagy via the AKT/mTOR pathway, thus providing a potential therapeutic drug for patients with OSCC.
Collapse
Affiliation(s)
- Jie Wen
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yunhan Sun
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Li Ma
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Tingjian Zu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Na Wang
- Department of Stomatology, Affiliated Hospital of Heze Medical College, Heze, Shandong, China
| | - Tianqi Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jin Liang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yulei Zhang
- School of Stomatology, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Haoyang Lu
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yihua Wu
- Department of Periodontology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Shizhou Zhang
- Department of Oral and Maxillofacial Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| |
Collapse
|
5
|
Master K, El Khalki L, Bayachou M, Sossey‐Alaoui K. Role of WAVE3 as an actin binding protein in the pathology of triple negative breast cancer. Cytoskeleton (Hoboken) 2025; 82:130-144. [PMID: 39021344 PMCID: PMC11904861 DOI: 10.1002/cm.21898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Breast cancer, a prevalent global health concern, has sparked extensive research efforts, particularly focusing on triple negative breast cancer (TNBC), a subtype lacking estrogen receptor (ER), progesterone receptor, and epidermal growth factor receptor. TNBC's aggressive nature and resistance to hormone-based therapies heightens the risk of tumor progression and recurrence. Actin-binding proteins, specifically WAVE3 from the Wiskott-Aldrich syndrome protein (WASP) family, have emerged as major drivers in understanding TNBC biology. This review delves into the intricate molecular makeup of TNBC, shedding light on actin's fundamental role in cellular processes. Actin, a structural element in the cytoskeleton, regulates various cellular pathways essential for homeostasis. Its dynamic nature enables functions such as cell migration, motility, intracellular transport, cell division, and signal transduction. Actin-binding proteins, including WAVE3, play pivotal roles in these processes. WAVE3, a member of the WASP family, remains the focus of this review due to its potential involvement in TNBC progression. While actin-binding proteins are studied for their roles in healthy cellular cycles, their significance in TNBC remains underexplored. This review aims to discuss WAVE3's impact on TNBC, exploring its molecular makeup, functions, and significance in tumor progression. The intricate structure of WAVE3, featuring elements like the verprolin-cofilin-acidic domain and regulatory elements, plays a crucial role in regulating actin dynamics. Dysregulation of WAVE3 in TNBC has been linked to enhanced cell migration, invasion, extracellular matrix remodeling, epithelial-mesenchymal transition, tumor proliferation, and therapeutic resistance. Understanding the role of actin-binding proteins in cancer biology has potential clinical implications, making them potential prognostic biomarkers and promising therapeutic targets. The review emphasizes the need for further research into actin-binding proteins' clinical applications, diagnostic value, and therapeutic interventions. In conclusion, this comprehensive review explores the complex interplay between actin and actin-binding proteins, with special emphasis on WAVE3, in the context of TNBC. By unraveling the molecular intricacies, structural characteristics, and functional significance, the review paves the way for future research directions, clinical applications, and potential therapeutic strategies in the challenging landscape of TNBC.
Collapse
Affiliation(s)
- Kruyanshi Master
- Department of ChemistryCleveland State UniversityClevelandOhioUSA
| | - Lamyae El Khalki
- MetroHealth SystemClevelandOhioUSA
- Case Western Reserve UniversityClevelandOhioUSA
- Case Comprehensive Cancer CenterClevelandOhioUSA
| | - Mekki Bayachou
- Department of ChemistryCleveland State UniversityClevelandOhioUSA
| | - Khalid Sossey‐Alaoui
- MetroHealth SystemClevelandOhioUSA
- Case Western Reserve UniversityClevelandOhioUSA
- Case Comprehensive Cancer CenterClevelandOhioUSA
| |
Collapse
|
6
|
Deng K, Pei M, Li B, Yang N, Wang Z, Wan X, Zhong Z, Yang Z, Chen Y. Signal pathways involved in contrast-induced acute kidney injury. Front Physiol 2024; 15:1490725. [PMID: 39655278 PMCID: PMC11625813 DOI: 10.3389/fphys.2024.1490725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Contrast-induced acute kidney injury (CI-AKI) has emerged as a global public health concern, ranking as the third most prevalent cause of hospital-acquired acute kidney injury, which is related to adverse outcomes. However, its precise pathogenesis remains elusive. Consequently, researchers are dedicated to uncovering CI-AKI's pathophysiology and signaling pathways, including inflammation, oxidative stress, apoptosis, and ferroptosis, to improve prevention and treatment. This review thoroughly analyzes the signaling pathways and their interactions associated with CI-AKI, assesses the impact of various research models on pathway analysis, and explores more precise targeted treatment and prevention approaches. Aims to furnish a robust theoretical foundation for the molecular mechanisms underpinning clinical treatments.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yanling Chen
- Department of Pathophysiology, Zhuhai Campus of Zunyi Medical University, Zhuhai, Guangdong, China
| |
Collapse
|
7
|
Seifi Z, Khazaei M, Cheraghali D, Rezakhani L. Decellularized tissues as platforms for digestive system cancer models. Heliyon 2024; 10:e31589. [PMID: 38845895 PMCID: PMC11153114 DOI: 10.1016/j.heliyon.2024.e31589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 05/17/2024] [Accepted: 05/20/2024] [Indexed: 06/09/2024] Open
Abstract
The extracellular matrix (ECM) is a multifunctional network of macromolecules that regulate various cellular functions and physically support the tissues. Besides physiological conditions, the ECM also changes during pathological conditions such as cancer. As tumor cells proliferate, notable changes occur in the quantity and makeup of the surrounding ECM. Therefore, the role of this noncellular component of tissues in studies of tumor microenvironments should be considered. So far, many attempts have been made to create 2-dimensional (2D) or 3-dimensional (3D) models that can replicate the intricate connections within the tumor microenvironment. Decellularized tissues are proper scaffolds that imitate the complex nature of native ECM. This review aims to summarize 3D models of digestive system cancers based on decellularized ECMs. These ECM-based scaffolds will enable us to study the interactive communication between cells and their surrounding environment which brings new potential for a better understanding of the pathophysiology of cancer.
Collapse
Affiliation(s)
- Zahra Seifi
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mozafar Khazaei
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Danial Cheraghali
- Department of Mechanical Engineering, New Jersey Institute of Technology, NJ, USA
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
8
|
Xiong J, Xiao R, Zhao J, Zhao Q, Luo M, Li F, Zhang W, Wu M. Matrix stiffness affects tumor-associated macrophage functional polarization and its potential in tumor therapy. J Transl Med 2024; 22:85. [PMID: 38246995 PMCID: PMC10800063 DOI: 10.1186/s12967-023-04810-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/17/2023] [Indexed: 01/23/2024] Open
Abstract
The extracellular matrix (ECM) plays critical roles in cytoskeletal support, biomechanical transduction and biochemical signal transformation. Tumor-associated macrophage (TAM) function is regulated by matrix stiffness in solid tumors and is often associated with poor prognosis. ECM stiffness-induced mechanical cues can activate cell membrane mechanoreceptors and corresponding mechanotransducers in the cytoplasm, modulating the phenotype of TAMs. Currently, tuning TAM polarization through matrix stiffness-induced mechanical stimulation has received increasing attention, whereas its effect on TAM fate has rarely been summarized. A better understanding of the relationship between matrix stiffness and macrophage function will contribute to the development of new strategies for cancer therapy. In this review, we first introduced the overall relationship between macrophage polarization and matrix stiffness, analyzed the changes in mechanoreceptors and mechanotransducers mediated by matrix stiffness on macrophage function and tumor progression, and finally summarized the effects of targeting ECM stiffness on tumor prognosis to provide insight into this new field.
Collapse
Affiliation(s)
- Jiaqiang Xiong
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Rourou Xiao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Jiahui Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qiuyan Zhao
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Manwen Luo
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Feng Li
- Department of Medical Genetics, School of Basic Medical Sciences, Wuhan University, Wuhan, 430071, China.
- Hubei Provincial Key Laboratory of Allergy and Immunology, Wuhan, 430071, China.
| | - Wei Zhang
- Department of Obstetrics and Gynecology, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430032, China.
| |
Collapse
|
9
|
Wang L, Zhou Y, Cui H, Zhuang X, Cheng C, Weng Y, Liu H, Wang S, Pan X, Cui Y, Zhang W. IGH repertoire analysis at scale: deciphering the complexity of B cell infiltration and migration in esophageal squamous cell carcinoma. Cancer Gene Ther 2024; 31:131-147. [PMID: 37985722 DOI: 10.1038/s41417-023-00689-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 10/10/2023] [Accepted: 11/02/2023] [Indexed: 11/22/2023]
Abstract
Tumor-infiltrating B-lineage cells have become predictors of prognosis and immunotherapy responses in various cancers. However, limited knowledge about their infiltration and migration patterns has hindered the understanding of their anti-tumor functions. Here, we examined the immunoglobulin heavy chain (IGH) repertoires in 496 multi-regional tumor, 107 normal tissue, and 48 metastatic lymph node samples obtained from 107 patients with esophageal squamous cell carcinoma (ESCC). Our study revealed higher IgG-type B-lineage cells infiltration in tumors than in healthy tissue, which was associated with improved patient outcomes. Genes such as ACTN1, COL6A5, and pathways like focal adhesion, which shapes the physical structure of tumors, could affect B-lineage cell infiltration. Notably, the IGH sequence was used as an identity-tag to monitor B cell migration, and their infiltration schema within the tumor were depicted based on our multi-regional tumor specimens. This analysis revealed an escalation in B cell clones overlapped between metastatic lymph nodes and tumors. Therefore, the Lymph Node Activation Index was defined, which could predict the outcomes of patients with lymph node metastasis. This research introduces a novel framework for probing B cell infiltration and migration within the tumor microenvironment using large-scale transcriptome data, while simultaneously providing fresh perspectives on B cell immunology within ESCC.
Collapse
Affiliation(s)
- Longlong Wang
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China
| | - Yong Zhou
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China
| | - Heyang Cui
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China
| | - Xuehan Zhuang
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China
| | - Chen Cheng
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China
| | - Yongjia Weng
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China
| | - Huijuan Liu
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China
| | - Shubin Wang
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China
| | - Xinghua Pan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| | - Yongping Cui
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China.
- Key Laboratory of Cellular Physiology of the Ministry of Education, Department of Pathology, Shanxi Medical University, Taiyuan, Shanxi, 030001, China.
| | - Weimin Zhang
- Cancer Institute, Department of Oncology, Peking University Shenzhen Hospital, Shenzhen Peking University-the Hong Kong University of Science and Technology (PKU-HKUST) Medical Center, Shenzhen, Guangdong, 518035, China.
- Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, Guangdong, 518028, China.
- Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Molecular Oncology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
10
|
Budi HS, Farhood B. Targeting oral tumor microenvironment for effective therapy. Cancer Cell Int 2023; 23:101. [PMID: 37221555 DOI: 10.1186/s12935-023-02943-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Oral cancers are among the common head and neck malignancies. Different anticancer therapy modalities such as chemotherapy, immunotherapy, radiation therapy, and also targeted molecular therapy may be prescribed for targeting oral malignancies. Traditionally, it has been assumed that targeting malignant cells alone by anticancer modalities such as chemotherapy and radiotherapy suppresses tumor growth. In the last decade, a large number of experiments have confirmed the pivotal role of other cells and secreted molecules in the tumor microenvironment (TME) on tumor progression. Extracellular matrix and immunosuppressive cells such as tumor-associated macrophages, myeloid-derived suppressor cells (MDSCs), cancer-associated fibroblasts (CAFs), and regulatory T cells (Tregs) play key roles in the progression of tumors like oral cancers and resistance to therapy. On the other hand, infiltrated CD4 + and CD8 + T lymphocytes, and natural killer (NK) cells are key anti-tumor cells that suppress the proliferation of malignant cells. Modulation of extracellular matrix and immunosuppressive cells, and also stimulation of anticancer immunity have been suggested to treat oral malignancies more effectively. Furthermore, the administration of some adjuvants or combination therapy modalities may suppress oral malignancies more effectively. In this review, we discuss various interactions between oral cancer cells and TME. Furthermore, we also review the basic mechanisms within oral TME that may cause resistance to therapy. Potential targets and approaches for overcoming the resistance of oral cancers to various anticancer modalities will also be reviewed. The findings for targeting cells and potential therapeutic targets in clinical studies will also be reviewed.
Collapse
Affiliation(s)
- Hendrik Setia Budi
- Department of Oral Biology, Dental Pharmacology, Faculty of Dental Medicine, Universitas Airlangga, Surabaya, Indonesia.
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran.
| |
Collapse
|
11
|
Sagini MN, Klika KD, Owen RW, Berger MR. Khasianine Affects the Expression of Sugar-Sensitive Proteins in Pancreatic Cancer Cells, Which Are Altered in Data from the Rat Model and Patients. ACS Pharmacol Transl Sci 2023; 6:727-737. [PMID: 37200805 PMCID: PMC10186360 DOI: 10.1021/acsptsci.3c00013] [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: 01/19/2023] [Indexed: 05/20/2023]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy with no effective treatment, particularly in the advanced stage. This study explored the antiproliferative activity of khasianine against pancreatic cancer cell lines of human (Suit2-007) and rat (ASML) origin. Khasianine was purified from Solanum incanum fruits by silica gel column chromatography and analyzed by LC-MS and NMR spectroscopy. Its effect in pancreatic cancer cells was evaluated by cell proliferation assay, chip array and mass spectrometry. Proteins showing sensitivity to sugars, i.e. sugar-sensitive lactosyl-Sepharose binding proteins (LSBPs), were isolated from Suit2-007 cells by competitive affinity chromatography. The eluted fractions included galactose-, glucose-, rhamnose- and lactose-sensitive LSBPs. The resulting data were analyzed by Chipster, Ingenuity Pathway Analysis (IPA) and GraphPad Prism. Khasianine inhibited proliferation of Suit2-007 and ASML cells with IC50 values of 50 and 54 μg/mL, respectively. By comparative analysis, khasianine downregulated lactose-sensitive LSBPs the most (126%) and glucose-sensitive LSBPs the least (85%). Rhamnose-sensitive LSBPs overlapped significantly with lactose-sensitive LSBPs and were the most upregulated in data from patients (23%) and a pancreatic cancer rat model (11.5%). From IPA, the Ras homolog family member A (RhoA) emerged as one of the most activated signaling pathways involving rhamnose-sensitive LSBPs. Khasianine altered the mRNA expression of sugar-sensitive LSBPs, some of which were modulated in data from patients and the rat model. The antiproliferative effect of khasianine in pancreatic cancer cells and the downregulation of rhamnose-sensitive proteins underscore the potential of khasianine in treating pancreatic cancer.
Collapse
Affiliation(s)
- Micah N. Sagini
- Toxicology
and Chemotherapy Unit, German Cancer Research
Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| | - Karel D. Klika
- Molecular
Structure Analysis, German Cancer Research
Center (DKFZ), Im Neuenheimer
Feld 280, 69120 Heidelberg, Germany
| | - Robert W. Owen
- Biochemistry
and Biomarkers Unit, German Cancer Research
Center (DKFZ), Im Neuenheimer
Feld 580, 69120 Heidelberg, Germany
| | - Martin R. Berger
- Toxicology
and Chemotherapy Unit, German Cancer Research
Center (DKFZ), Im Neuenheimer Feld 580, 69120 Heidelberg, Germany
| |
Collapse
|
12
|
Zhao G, Ren Y, Yan J, Zhang T, Lu P, Lei J, Rao H, Kang X, Cao Z, Peng F, Peng C, Rao C, Li Y. Neoprzewaquinone A Inhibits Breast Cancer Cell Migration and Promotes Smooth Muscle Relaxation by Targeting PIM1 to Block ROCK2/STAT3 Pathway. Int J Mol Sci 2023; 24:ijms24065464. [PMID: 36982538 PMCID: PMC10051292 DOI: 10.3390/ijms24065464] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Salvia miltiorrhiza Bunge (Danshen) has been widely used to treat cancer and cardiovascular diseases in Chinese traditional medicine. Here, we found that Neoprzewaquinone A (NEO), an active component of S. miltiorrhiza, selectively inhibits PIM1. We showed that NEO potently inhibits PIM1 kinase at nanomolar concentrations and significantly suppresses the growth, migration, and Epithelial-Mesenchymal Transition (EMT) in the triple-negative breast cancer cell line, MDA-MB-231 in vitro. Molecular docking simulations revealed that NEO enters the PIM1 pocket, thereby triggering multiple interaction effects. Western blot analysis revealed that both NEO and SGI-1776 (a specific PIM1 inhibitor), inhibited ROCK2/STAT3 signaling in MDA-MB-231 cells, indicating that PIM1 kinase modulates cell migration and EMT via ROCK2 signaling. Recent studies indicated that ROCK2 plays a key role in smooth muscle contraction, and that ROCK2 inhibitors effectively control the symptoms of high intraocular pressure (IOP) in glaucoma patients. Here, we showed that NEO and SGI-1776 significantly reduce IOP in normal rabbits and relax pre-restrained thoracic aortic rings in rats. Taken together, our findings indicated that NEO inhibits TNBC cell migration and relaxes smooth muscles mainly by targeting PIM1 and inhibiting ROCK2/STAT3 signaling, and that PIM1 may be an effective target for IOP and other circulatory diseases.
Collapse
Affiliation(s)
- Guiying Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yali Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jie Yan
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tingrui Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Peng Lu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jieting Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Huanan Rao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Xin Kang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Zhixing Cao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Fu Peng
- West China School of Pharmacy, Sichuan University, Chengdu 611137, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chaolong Rao
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- R&D Center for Efficiency, Safety and Application in Chinese Materia Medica with Medical and Edible Values, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Correspondence: (C.R.); (Y.L.)
| | - Yuzhi Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
- Correspondence: (C.R.); (Y.L.)
| |
Collapse
|
13
|
Wang Y, He J, Zhang J, Zhang N, Zhou Y, Wu F. Cell migration induces apoptosis in osteosarcoma cell via inhibition of Wnt-β-catenin signaling pathway. Colloids Surf B Biointerfaces 2023; 223:113142. [PMID: 36669438 DOI: 10.1016/j.colsurfb.2023.113142] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/17/2022] [Accepted: 01/09/2023] [Indexed: 01/15/2023]
Abstract
The current design scheme on anti-cancer materials is mainly through tuning the mechanical properties of the materials to induce apoptosis in cancer cells, with the involvement of Rho/ROCK signaling pathway. We hypothesize that tuning the motility is another potential important approach to modifying the tumor microenvironment and inducing tumor apoptosis. To this aim, we have prepared RGD-modified substrates to regulate cell motility through modification of RGD with different concentrations, and systematically examined the effect of motility on the apoptosis of tumor cells, and the potential involvement of Wnt signaling pathway. Our studies indicated that RGD modification could be readily used to tune the motility of cancer cells. High RGD concentration significantly suppressed the migration of cancer cells, leading to significantly increased apoptosis rate, about three times of that of the unmodified samples. Western-blot analysis also showed that cell with low motility expressed more caspase-3 and PARP proteins. Further RNA sequence study strongly suggested that low motility inhibited the canonical Wnt signaling pathway, which in turn led to the activation of the mitochondria-associated caspase signaling pathway, and ultimately to the apoptosis of osteosarcoma cells. Activation of the Wnt-β-catenin pathway through HLY78 significantly suppressed the apoptosis of MG-63 cells, further suggesting the critical role of Wnt pathway in motility-regulated-apoptosis of tumor cells. Our findings shed insights to understand the underlying mechanisms that induced the tumor cell apoptosis, and might provide new strategy for designing the novel anti-tumor materials.
Collapse
Affiliation(s)
- Yao Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, PR China
| | - Jing He
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, PR China
| | - Junwei Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, PR China
| | - Nihui Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, PR China
| | - Yong Zhou
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610064, PR China.
| | - Fang Wu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu 610064, PR China.
| |
Collapse
|
14
|
Chen X, Wei X, Ma S, Xie H, Huang S, Yao M, Zhang L. Cysteine and glycine rich protein 2 exacerbates vascular fibrosis in pulmonary hypertension through the nuclear translocation of yes-associated protein and transcriptional coactivator with PDZ-binding motif. Toxicol Appl Pharmacol 2022; 457:116319. [PMID: 36414118 DOI: 10.1016/j.taap.2022.116319] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 11/21/2022]
Abstract
Pulmonary hypertension (PH) is a serious cardiovascular disease with a poor prognosis and high mortality. The pathogenesis of PH is complex, and the main pathological changes in PH are abnormal hypertrophy and vessel stiffness. Cysteine and glycine rich protein 2 (Csrp2), a member of the LIM-only family plays a key role in the response to vascular injury. However, its roles in vascular fibrosis and PH have not been clarified. Therefore, this study aimed to investigate whether Csrp2 can promote vascular fibrosis and to further explore the possible mechanisms. Csrp2 expression was increased in both the pulmonary vasculature of rats with PH and hypoxic pulmonary vascular smooth muscle cells (PASMCs). Hypoxia activated TGF-β1 and its downstream effector, SP1. Additionally, hypoxia activated the ROCK pathway and inhibited KLF4 expression. Silencing SP1 and overexpressing KLF4 reversed the hypoxia-induced increase in Csrp2 expression. Csrp2 knockdown decreased the expression of extracellular matrix (ECM) proteins and inhibited the nuclear translocation and expression of YAP/TAZ in hypoxic PASMCs. These results indicate that hypoxia induces Csrp2 expression through the TGF-β1/SP1 and ROCK/KLF4 pathways. Elevated Csrp2 promoted the nuclear translocation and expression of YAP/TAZ, leading to vascular fibrosis and the development of PH.
Collapse
Affiliation(s)
- Xinghe Chen
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China; Department of Pediatric Surgery, The First Affiliated Hospital of Fujian Medical University, Fujian Medical University, Fuzhou, China
| | - Xiaozhen Wei
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Saijie Ma
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Huating Xie
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Sirui Huang
- The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Mengge Yao
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Li Zhang
- Department of Cardiac Surgery, Fujian Medical University Union Hospital, Fuzhou, China; The Key Laboratory of Fujian Province Universities on Ion Channel and Signal Transduction in Cardiovascular Diseases, Department of Physiology and Pathophysiology, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
15
|
Xiong X, Chen C, Yang J, Ma L, Wang X, Zhang W, Yuan Y, Peng M, Li L, Luo P. Characterization of the basement membrane in kidney renal clear cell carcinoma to guide clinical therapy. Front Oncol 2022; 12:1024956. [DOI: 10.3389/fonc.2022.1024956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
BackgroundRenal cell carcinoma (RCC) is the most common kidney cancer in adults. According to the histological features, it could be divided into several subtypes, of which the most common one is kidney renal clear cell carcinoma (KIRC), which contributed to more than 90% of cases for RCC and usually ends with a dismal outcome. Previous studies suggested that basement membrane genes (BMGs) play a pivotal role in tumor development. However, the significance and prognostic value of BMGs in KIRC still wrap in the mist.MethodsKIRC data were downloaded from the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases. A prognostic risk score (PRS) model based on BMGs was established using univariate and least absolute shrinkage and selection operator (LASSO) and the Cox regression analysis was performed for prognostic prediction. The Kaplan-Meier analysis, univariate Cox regression, multivariate Cox regression, receiver operating characteristic (ROC) curves, nomogram, and calibration curves were utilized to evaluate and validate the PRS model. All KIRC cases were divided into the high-risk score (HRS) group and the low-risk score (LRS) group according to the median risk scores. In addition, single-sample gene set enrichment analysis (ssGSEA), immune analysis, tumor microenvironment (TME) analysis, principal component analysis (PCA), and half-maximal inhibitory concentration (IC50) were also applied. Expression levels of BMGs were confirmed by qRT-PCR in both human renal cancer cell lines and tissues.ResultsWe established the BMGs-based prognostic model according to the following steps. Within the TCGA cohort, patients’ prognosis of the HRS group was significantly worse than that of the LRS group, which was consistent with the analysis results of the GEO cohort. PCA patterns were significantly distinct for LRS and HRS groups and pathological features of the HRS group were more malignant compared with the LRS group. Correlation analysis of the PRS model and TME features, such as immune cell scores, stromal cell scores, and ESTIMATE values, revealed a higher immune infiltration in the HRS group compared with the LRS group. The chemotherapeutic response was also evaluated in KIRC treatment. It showed that the HRS group exhibited stronger chemoresistance to chemotherapeutics like FR-180204, GSK1904529A, KIN001-102, and YM201636. The therapeutic reactivity of the other 27 chemotherapeutic agents was summarized as well. Furthermore, the FREM2 level was measured in both human kidney tissues and associated cell lines, which suggested that lower FREM2 expression prompts a severer pathology and clinical ending.ConclusionsOur study showed that KIRC is associated with a unique BMG expression pattern. The risk scores related to the expression levels of 10 BMGs were assessed by survival status, TME, pathological features, and chemotherapeutic resistance. All results suggested that FREM2 could be a potential candidate for KIRC prognosis prediction. In this study, we established a valid model and presented new therapeutic targets for the KIRC prognosis prediction as well as the clinical treatment recommendation, and finally, facilitated precision tumor therapy for every single individual.
Collapse
|
16
|
Wang W, Li C, Zhuang C, Zhang H, Wang Q, Fan X, Qi M, Sun R, Yu J. Research on the Mechanism and Prevention of Hypertension Caused by Apatinib Through the RhoA/ROCK Signaling Pathway in a Mouse Model of Gastric Cancer. Front Cardiovasc Med 2022; 9:873829. [PMID: 35811723 PMCID: PMC9262125 DOI: 10.3389/fcvm.2022.873829] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Hypertension is one of the main adverse effects of antiangiogenic tumor drugs and thus limits their application. The mechanism of hypertension caused by tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factors is mainly related to inhibition of the nitric oxide (NO) pathway and activation of the endothelin pathway, as well as vascular rarefaction and increased salt sensitivity; consequently, prevention and treatment differ for this type of hypertension compared with primary hypertension. Apatinib is a highly selective TKI approved in China for the treatment of advanced or metastatic gastric cancer. The RhoA/ROCK pathway is involved in the pathogenesis of hypertension and mediates smooth muscle contraction, eNOS inhibition, endothelial dysfunction and vascular remodeling. In this study, in vivo experiments were performed to explore whether the RhoA/ROCK signaling pathway is part of a possible mechanism of apatinib in the treatment of gastric cancer-induced hypertension and the impairment of vascular remodeling and left ventricular function. Y27632, a selective small inhibitor of both ROCK1 and ROCK2, was combined with apatinib, and its efficacy was evaluated, wherein it can reduce hypertension induced by apatinib treatment in gastric cancer mice and weaken the activation of the RhoA/ROCK pathway by apatinib and a high-salt diet (HSD). Furthermore, Y-27632 improved aortic remodeling, fibrosis, endothelial dysfunction, superior mesenteric artery endothelial injury, left ventricular dysfunction and cardiac fibrosis in mice by weakening the activation of the RhoA/ROCK pathway. The expression of RhoA/ROCK pathway-related proteins and relative mRNA levels in mice after apatinib intervention were analyzed by various methods, and blood pressure and cardiac function indexes were compared. Endothelial and cardiac function and collagen levels in the aorta were also measured to assess vascular and cardiac fibrosis and to provide a basis for the prevention and treatment of this type of hypertension.
Collapse
|
17
|
Ma J, Huang L, Gao YB, Li MX, Chen LL, Yang L. M2 macrophage facilitated angiogenesis in cutaneous squamous cell carcinoma via circ_TNFRSF21/miR-3619-5p/ROCK axis. Kaohsiung J Med Sci 2022; 38:761-771. [PMID: 35593591 DOI: 10.1002/kjm2.12555] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 12/30/2021] [Accepted: 04/05/2022] [Indexed: 11/09/2022] Open
Abstract
In recent years, the role of circular RNA in cancer cells has been studied broadly; however, the functional significance of circular RNA in the regulation of the tumor microenvironment (TME) is not fully understood. In this study, we aimed to reveal the role of circ_TNFRSF21 in M2 macrophage-induced cutaneous squamous cell carcinoma (cSCC) angiogenesis. Quantitative polymerase chain reaction and Western blotting were performed to determine the levels of the indicated genes. Direct binding between circ_TNFRSF21 and miR-3619-5p, miR-3619-5p, and ROCK2 was verified by dual-luciferase activity. The migration and invasion of human umbilical vein endothelial cells were evaluated by wound healing and transwell assays. Tube formation was performed to detect in vitro angiogenesis. Circ_TNFRSF21 and ROCK2 were upregulated in cSCC tissue, while miR-3619-5p was downregulated. Circ_TNFRSF21 negatively regulated the expression of miR-3619-5p, while miR-3619-5p negatively regulated the expression of ROCK2. miR-3619-5p suppressed tube formation by inhibiting ROCK signaling. M2 macrophages facilitated tube formation via the circ_TNFRSF21/miR-3619-5p/ROCK2 axis. Our present study revealed that circ_TNFRSF21 was elevated in M2 macrophages and mediated M2 macrophage-induced tube formation in vitro.
Collapse
Affiliation(s)
- Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lei Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yan-Bin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Min-Xiong Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Liang-Long Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
18
|
Feng F, Liu M, Pan L, Wu J, Wang C, Yang L, Liu W, Xu W, Lei M. Biomechanical Regulatory Factors and Therapeutic Targets in Keloid Fibrosis. Front Pharmacol 2022; 13:906212. [PMID: 35614943 PMCID: PMC9124765 DOI: 10.3389/fphar.2022.906212] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/25/2022] [Indexed: 01/10/2023] Open
Abstract
Keloids are fibroproliferative skin disorder caused by abnormal healing of injured or irritated skin and are characterized by excessive extracellular matrix (ECM) synthesis and deposition, which results in excessive collagen disorders and calcinosis, increasing the remodeling and stiffness of keloid matrix. The pathogenesis of keloid is very complex, and may include changes in cell function, genetics, inflammation, and other factors. In this review, we aim to discuss the role of biomechanical factors in keloid formation. Mechanical stimulation can lead to excessive proliferation of wound fibroblasts, deposition of ECM, secretion of more pro-fibrosis factors, and continuous increase of keloid matrix stiffness. Matrix mechanics resulting from increased matrix stiffness further activates the fibrotic phenotype of keloid fibroblasts, thus forming a loop that continuously invades the surrounding normal tissue. In this process, mechanical force is one of the initial factors of keloid formation, and matrix mechanics leads to further keloid development. Next, we summarized the mechanotransduction pathways involved in the formation of keloids, such as TGF-β/Smad signaling pathway, integrin signaling pathway, YAP/TAZ signaling pathway, and calcium ion pathway. Finally, some potential biomechanics-based therapeutic concepts and strategies are described in detail. Taken together, these findings underscore the importance of biomechanical factors in the formation and progression of keloids and highlight their regulatory value. These findings may help facilitate the development of pharmacological interventions that can ultimately prevent and reduce keloid formation and progression.
Collapse
Affiliation(s)
- Fan Feng
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Mingying Liu
- School of Comprehensive Health Management, Xihua University, Chengdu, China
| | - Lianhong Pan
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Jiaqin Wu
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Chunli Wang
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Li Yang
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Wanqian Liu
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- *Correspondence: Wanqian Liu, ; Wei Xu, ; Mingxing Lei,
| | - Wei Xu
- Chongqing Clinical Research Center for Dermatology, Chongqing Key Laboratory of Integrative Dermatology Research, Department of Dermatology, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
- *Correspondence: Wanqian Liu, ; Wei Xu, ; Mingxing Lei,
| | - Mingxing Lei
- National Innovation and Attracting Talents “111” Base, Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
- *Correspondence: Wanqian Liu, ; Wei Xu, ; Mingxing Lei,
| |
Collapse
|
19
|
Targeting extracellular matrix stiffness and mechanotransducers to improve cancer therapy. J Hematol Oncol 2022; 15:34. [PMID: 35331296 PMCID: PMC8943941 DOI: 10.1186/s13045-022-01252-0] [Citation(s) in RCA: 244] [Impact Index Per Article: 81.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/09/2022] [Indexed: 02/06/2023] Open
Abstract
Cancer microenvironment is critical for tumorigenesis and cancer progression. The extracellular matrix (ECM) interacts with tumor and stromal cells to promote cancer cells proliferation, migration, invasion, angiogenesis and immune evasion. Both ECM itself and ECM stiffening-induced mechanical stimuli may activate cell membrane receptors and mechanosensors such as integrin, Piezo1 and TRPV4, thereby modulating the malignant phenotype of tumor and stromal cells. A better understanding of how ECM stiffness regulates tumor progression will contribute to the development of new therapeutics. The rapidly expanding evidence in this research area suggests that the regulators and effectors of ECM stiffness represent potential therapeutic targets for cancer. This review summarizes recent work on the regulation of ECM stiffness in cancer, the effects of ECM stiffness on tumor progression, cancer immunity and drug resistance. We also discuss the potential targets that may be druggable to intervene ECM stiffness and tumor progression. Based on these advances, future efforts can be made to develop more effective and safe drugs to interrupt ECM stiffness-induced oncogenic signaling, cancer progression and drug resistance.
Collapse
|
20
|
Kolesnikoff N, Chen CH, Samuel M. Interrelationships between the extracellular matrix and the immune microenvironment that govern epithelial tumour progression. Clin Sci (Lond) 2022; 136:361-377. [PMID: 35260891 PMCID: PMC8907655 DOI: 10.1042/cs20210679] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/18/2022] [Accepted: 02/24/2022] [Indexed: 12/19/2022]
Abstract
Solid tumours are composed of cancer cells characterised by genetic mutations that underpin the disease, but also contain a suite of genetically normal cells and the extracellular matrix (ECM). These two latter components are constituents of the tumour microenvironment (TME), and are key determinants of tumour biology and thereby the outcomes for patients. The tumour ECM has been the subject of intense research over the past two decades, revealing key biochemical and mechanobiological principles that underpin its role in tumour cell proliferation and survival. However, the ECM also strongly influences the genetically normal immune cells within the microenvironment, regulating not only their proliferation and survival, but also their differentiation and access to tumour cells. Here we review recent advances in our knowledge of how the ECM regulates the tumour immune microenvironment and vice versa, comparing normal skin wound healing to the pathological condition of tumour progression.
Collapse
Affiliation(s)
- Natasha Kolesnikoff
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Chun-Hsien Chen
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
| | - Michael Susithiran Samuel
- Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, South Australia, Australia
- Adelaide Medical School, Faculty of Health Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
21
|
Youn M, Smith SM, Lee AG, Chae HD, Spiteri E, Erdmann J, Galperin I, Jones LM, Donato M, Abidi P, Bittencourt H, Lacayo N, Dahl G, Aftandilian C, Davis KL, Matthews JA, Kornblau SM, Huang M, Sumarsono N, Redell MS, Fu CH, Chen IM, Alonzo TA, Eklund E, Gotlib J, Khatri P, Sweet-Cordero EA, Hijiya N, Sakamoto KM. Comparison of the Transcriptomic Signatures in Pediatric and Adult CML. Cancers (Basel) 2021; 13:cancers13246263. [PMID: 34944883 PMCID: PMC8699058 DOI: 10.3390/cancers13246263] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/20/2022] Open
Abstract
Simple Summary To investigate whether pediatric and adult chronic myeloid leukemia (CML) have unique molecular characteristics, we studied the transcriptomic signature of pediatric and adult CML cells using high-throughput RNA sequencing. We identified differentially expressed genes and pathways unique to pediatric CML cells compared to adult CML cells. The Rho pathway was significantly dysregulated in pediatric CML cells compared to adult CML cells, suggesting the potential importance in the pathogenesis of pediatric CML. Our study is the first to compare transcriptome profiles of CML across different age groups. A better understanding of the biology of CML across different ages may inform future treatment approaches. Abstract Children with chronic myeloid leukemia (CML) tend to present with higher white blood counts and larger spleens than adults with CML, suggesting that the biology of pediatric and adult CML may differ. To investigate whether pediatric and adult CML have unique molecular characteristics, we studied the transcriptomic signature of pediatric and adult CML CD34+ cells and healthy pediatric and adult CD34+ control cells. Using high-throughput RNA sequencing, we found 567 genes (207 up- and 360 downregulated) differentially expressed in pediatric CML CD34+ cells compared to pediatric healthy CD34+ cells. Directly comparing pediatric and adult CML CD34+ cells, 398 genes (258 up- and 140 downregulated), including many in the Rho pathway, were differentially expressed in pediatric CML CD34+ cells. Using RT-qPCR to verify differentially expressed genes, VAV2 and ARHGAP27 were significantly upregulated in adult CML CD34+ cells compared to pediatric CML CD34+ cells. NCF1, CYBB, and S100A8 were upregulated in adult CML CD34+ cells but not in pediatric CML CD34+ cells, compared to healthy controls. In contrast, DLC1 was significantly upregulated in pediatric CML CD34+ cells but not in adult CML CD34+ cells, compared to healthy controls. These results demonstrate unique molecular characteristics of pediatric CML, such as dysregulation of the Rho pathway, which may contribute to clinical differences between pediatric and adult patients.
Collapse
Affiliation(s)
- Minyoung Youn
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Stephanie M. Smith
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Alex Gia Lee
- Department of Pediatrics, University of California, San Francisco, CA 94143, USA; (A.G.L.); (E.A.S.-C.)
| | - Hee-Don Chae
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Elizabeth Spiteri
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA;
- Cytogenetics Laboratory, Stanford Health Care, Stanford, CA 94304, USA; (J.E.); (I.G.)
| | - Jason Erdmann
- Cytogenetics Laboratory, Stanford Health Care, Stanford, CA 94304, USA; (J.E.); (I.G.)
| | - Ilana Galperin
- Cytogenetics Laboratory, Stanford Health Care, Stanford, CA 94304, USA; (J.E.); (I.G.)
| | - Lara Murphy Jones
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; (M.D.); (P.K.)
| | - Michele Donato
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; (M.D.); (P.K.)
- Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, CA 94305, USA
| | - Parveen Abidi
- Division of Hematology, Department of Medicine, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.A.); (J.G.)
| | - Henrique Bittencourt
- Hematology-Oncology Division, Charles Bruneau Cancer Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC H3T 1C5, Canada;
| | - Norman Lacayo
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Gary Dahl
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Catherine Aftandilian
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Kara L. Davis
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Jairo A. Matthews
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (J.A.M.); (S.M.K.)
| | - Steven M. Kornblau
- Department of Leukemia, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA; (J.A.M.); (S.M.K.)
| | - Min Huang
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Nathan Sumarsono
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
| | - Michele S. Redell
- Division of Pediatric Hematology/Oncology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Cecilia H. Fu
- Division of Hematology/Oncology, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA;
| | - I-Ming Chen
- Department of Pathology, University of New Mexico Comprehensive Cancer Center, Albuquerque, NM 87102, USA;
| | - Todd A. Alonzo
- Department of Preventive Medicine, University of Southern California, Los Angeles, CA 90032, USA;
| | - Elizabeth Eklund
- Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Jason Gotlib
- Division of Hematology, Department of Medicine, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; (P.A.); (J.G.)
| | - Purvesh Khatri
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; (M.D.); (P.K.)
- Stanford Center for Biomedical Informatics Research, Stanford University, Stanford, CA 94305, USA
| | | | - Nobuko Hijiya
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY 10032, USA;
| | - Kathleen M. Sakamoto
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; (M.Y.); (S.M.S.); (H.-D.C.); (L.M.J.); (N.L.); (G.D.); (C.A.); (K.L.D.); (M.H.); (N.S.)
- Correspondence: ; Tel.: +1-650-725-7126; Fax: +1-650-723-6700
| |
Collapse
|
22
|
Kochetkova M, Samuel MS. Differentiation of the tumor microenvironment: are CAFs the Organizer? Trends Cell Biol 2021; 32:285-294. [PMID: 34895986 DOI: 10.1016/j.tcb.2021.11.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/18/2021] [Accepted: 11/19/2021] [Indexed: 12/13/2022]
Abstract
Cancers contain a suite of genetically stable cells within an extracellular matrix, collectively termed the tumor microenvironment (TME). The TME strongly influences disease outcome for patients. Gleaning clues from the literature, we propose that the TME should be viewed not as disparate populations of cells constituting a pathological lesion, but as a cohesive tissue constituting a novel pathological organ, arising from the coordinated differentiation of its constituent cell types - a process we have termed tumor-associated neodifferentiation (TAND). We also discuss why cancer-associated fibroblasts (CAFs) may assume the role of Organizer of this organ, directing the recruitment and differentiation of cells within the TME. Viewing the microenvironment in this way will reveal new cancer vulnerabilities that may be exploited for therapy.
Collapse
Affiliation(s)
- Marina Kochetkova
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia
| | - Michael Susithiran Samuel
- Centre for Cancer Biology, an alliance between SA Pathology and the University of South Australia, Adelaide, SA 5000, Australia; Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, SA 5000, Australia.
| |
Collapse
|
23
|
Huang K, Liu J, Chen Q, Feng D, Wu H, Aldanakh A, Jian Y, Xu Z, Wang S, Yang D. The effect of mechanical force in genitourinary malignancies. Expert Rev Anticancer Ther 2021; 22:53-64. [PMID: 34726963 DOI: 10.1080/14737140.2022.2000864] [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: 10/19/2022]
Abstract
INTRODUCTION Mechanical force is attributed to the formation of tumor blood vessels, influences cancer cell invasion and metastasis, and promotes reprogramming of the energy metabolism. Currently, therapy strategies for the tumor microenvironment are being developed progressively. The purpose of this article is to discuss the molecular mechanism, diagnosis, and treatment of mechanical force in urinary tract cancers and outline the medications used in the mechanical microenvironment. AREAS COVERED This review covers the complex mechanical elements in the microenvironment of urinary system malignancies, focusing on mechanical molecular mechanisms for diagnosis and treatment. EXPERT OPINION The classification of various mechanical forces, such as matrix stiffness, shear force, and other forces, is relatively straightforward. However, little is known about the molecular process of mechanical forces in urinary tract malignancies. Because mechanical therapy is still controversial, it is critical to understand the molecular basis of mechanical force before adding mechanical therapy solutions.
Collapse
Affiliation(s)
- Kai Huang
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Junqiang Liu
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Qiwei Chen
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China.,School of Information Science and Technology, Dalian Maritime University, Dalian City, China
| | - Dan Feng
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Haotian Wu
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Abdullah Aldanakh
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| | - Yuli Jian
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Zhongyang Xu
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Shujing Wang
- Department of Biochemistry, Institute of Glycobiology, Dalian Medical University, Dalian, China
| | - Deyong Yang
- Department of Urology, First Affifiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
24
|
Smoking-associated upregulation of CBX3 suppresses ARHGAP24 expression to activate Rac1 signaling and promote tumor progression in lung adenocarcinoma. Oncogene 2021; 41:538-549. [PMID: 34785774 PMCID: PMC8782721 DOI: 10.1038/s41388-021-02114-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/27/2021] [Accepted: 11/01/2021] [Indexed: 01/10/2023]
Abstract
Although tobacco smoking is a risk factor for lung adenocarcinoma (LUAD), the mechanisms by which tobacco smoking induces LUAD development remain elusive. Histone methylation levels in human bronchial epithelial cells have been reported to increase after exposure to cigarettes. In this study, we explored the mechanisms regulating histone methylation in LUAD in response to smoking. We found that the histone H3K9 methylation reader CBX3 was upregulated in current smokers with LUAD, and that CBX3 overexpression promoted LUAD progression. Functional enrichment analyses revealed that CBX3 regulated the activation of Rho GTPases in LUAD. We also found that by forming a complex with TRIM28, TRIM24, and RBBP4, CBX3 repressed the expression of ARHGAP24 and increased the amount of active Rac1 in LUAD cells. Collectively, these results suggest that smoking associated upregulation of CBX3 promotes LUAD progression by activating the ARHGAP24/Rac1 pathway. Hence, the CBX3/ARHGAP24/Rac1 axis may represent a promising therapeutic target in smoking-induced LUAD.
Collapse
|
25
|
He Y, Liu T, Dai S, Xu Z, Wang L, Luo F. Tumor-Associated Extracellular Matrix: How to Be a Potential Aide to Anti-tumor Immunotherapy? Front Cell Dev Biol 2021; 9:739161. [PMID: 34733848 PMCID: PMC8558531 DOI: 10.3389/fcell.2021.739161] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/28/2021] [Indexed: 02/05/2023] Open
Abstract
The development of cancer immunotherapy, particularly immune checkpoint blockade therapy, has made major breakthroughs in the therapy of cancers. However, less than one-third of the cancer patients obtain significant and long-lasting therapeutic effects by cancer immunotherapy. Over the past few decades, cancer-related inflammations have been gradually more familiar to us. It’s known that chronic inflammation in tumor microenvironment (TME) plays a predominant role in tumor immunosuppression. Tumor-associated extracellular matrix (ECM), as a core member of TME, has been a research hotspot recently. A growing number of studies indicate that tumor-associated ECM is one of the major obstacles to realizing more successful cases of cancer immunotherapy. In this review, we discussed the potential application of tumor-associated ECM in the cancer immunity and its aide potentialities to anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Yingying He
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Oncology Department, People's Hospital of Deyang City, Deyang, China
| | - Tao Liu
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China.,Department of Oncology, The First Affiliated Hospital of Chengdu Medical College, Chengdu Medical College, Chengdu, China
| | - Shuang Dai
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Zihan Xu
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Li Wang
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Feng Luo
- Department of Medical Oncology, Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Upregulation of miR-345-5p suppresses cell growth of lung adenocarcinoma by regulating ras homolog family member A (RhoA) and Rho/Rho associated protein kinase (Rho/ROCK) pathway. Chin Med J (Engl) 2021; 134:2619-2628. [PMID: 34748526 PMCID: PMC8577671 DOI: 10.1097/cm9.0000000000001804] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Microribose nucleic acids (miRNAs) are implicated in the progression of lung adenocarcinoma. MicroRNA-345-5p (miR-345-5p) is a recently identified anti-oncogene in some human cancers, but its functional role and possible molecular mechanism in lung adenocarcinoma remain unknown. This study aimed to identify the biological function and underlying mechanism of miR-345-5p in lung adenocarcinoma cells. METHODS In this study, lung adenocarcinoma tissues and adjacent tissues were collected in the First Affiliated Hospital of Anhui Medical University between April 2016 and February 2017. The expression of miR-345-5p and ras homolog family member A (RhoA) in lung adenocarcinoma tissues and human lung adenocarcinoma cell lines (A549, H1650, PC-9, and H441) was detected by reverse transcription quantitative polymerase chain reaction analysis. Functional assays including colony formation, flow cytometry analysis, wound healing, and transwell assays were performed to assess the proliferation, apoptosis, migration, and invasion of lung adenocarcinoma cells. In addition, RNA pulldown and luciferase reporter assays were conducted to evaluate the relationship between miR-345-5p and RhoA. Difference between the two groups was analyzed with Student's t test, while that among multiple groups was analyzed with one-way analysis of variance. RESULTS MiR-345-5p expression displayed lower level in lung adenocarcinoma tissues (0.241 ± 0.095 vs.1.000 ± 0.233, t = 19.247, P < 0.001) and cell lines (F = 56.992, P < 0.001) than control tissues and cells. Functional experiments demonstrated that upregulation of miR-345-5p inhibited the malignant phenotypes of lung adenocarcinoma cells via suppressing cell proliferation, migration, invasion, and facilitating cell apoptosis. Additionally, RhoA was verified to be the downstream target of miR-345-5p. Expression of RhoA was downregulated by overexpression of miR-345-5p in PC-9 (0.321 ± 0.047 vs. 1.000 ± 0.127, t = 8.536, P < 0.001) and H1650 (0.398 ± 0.054 vs. 1.000 ± 0.156, t = 4.429, P = 0.011) cells. Rescue assays revealed that overexpression of RhoA rescued the suppressive effects of miR-345-5p upregulation on proliferation, migration, and invasion of lung adenocarcinoma cells. Further, miR-345-5p was found to regulate the Rho/Rho-associated protein kinase (ROCK) signaling pathway by downregulation of RhoA in lung adenocarcinoma cells. CONCLUSIONS MiR-345-5p plays a tumor suppressor role in lung adenocarcinoma cells by downregulating RhoA to inactivate the Rho/ROCK pathway.
Collapse
|
27
|
Luo Y, Yin S, Lu J, Zhou S, Shao Y, Bao X, Wang T, Qiu Y, Yu H. Tumor microenvironment: a prospective target of natural alkaloids for cancer treatment. Cancer Cell Int 2021; 21:386. [PMID: 34284780 PMCID: PMC8290600 DOI: 10.1186/s12935-021-02085-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 07/08/2021] [Indexed: 12/17/2022] Open
Abstract
Malignant tumor has become one of the major diseases that seriously endangers human health. Numerous studies have demonstrated that tumor microenvironment (TME) is closely associated with patient prognosis. Tumor growth and progression are strongly dependent on its surrounding tumor microenvironment, because the optimal conditions originated from stromal elements are required for cancer cell proliferation, invasion, metastasis and drug resistance. The tumor microenvironment is an environment rich in immune/inflammatory cells and accompanied by a continuous, gradient of hypoxia and pH. Overcoming immunosuppressive environment and boosting anti-tumor immunity may be the key to the prevention and treatment of cancer. Most traditional Chinese medicine have been proved to have good anti-tumor activity, and they have the advantages of better therapeutic effect and few side effects in the treatment of malignant tumors. An increasing number of studies are giving evidence that alkaloids extracted from traditional Chinese medicine possess a significant anticancer efficiency via regulating a variety of tumor-related genes, pathways and other mechanisms. This paper reviews the anti-tumor effect of alkaloids targeting tumor microenvironment, and further reveals its anti-tumor mechanism through the effects of alkaloids on different components in tumor microenvironment.
Collapse
Affiliation(s)
- Yanming Luo
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shuangshuang Yin
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jia Lu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Shiyue Zhou
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yingying Shao
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xiaomei Bao
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Tao Wang
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin, 300070, China.
| | - Haiyang Yu
- Tianjin State Key Laboratory of Component-Based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
28
|
Implant Fibrosis and the Underappreciated Role of Myofibroblasts in the Foreign Body Reaction. Cells 2021; 10:cells10071794. [PMID: 34359963 PMCID: PMC8304203 DOI: 10.3390/cells10071794] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/08/2021] [Accepted: 07/13/2021] [Indexed: 02/06/2023] Open
Abstract
Body implants and implantable medical devices have dramatically improved and prolonged the life of countless patients. However, our body repair mechanisms have evolved to isolate, reject, or destroy any object that is recognized as foreign to the organism and inevitably mounts a foreign body reaction (FBR). Depending on its severity and chronicity, the FBR can impair implant performance or create severe clinical complications that will require surgical removal and/or replacement of the faulty device. The number of review articles discussing the FBR seems to be proportional to the number of different implant materials and clinical applications and one wonders, what else is there to tell? We will here take the position of a fibrosis researcher (which, coincidentally, we are) to elaborate similarities and differences between the FBR, normal wound healing, and chronic healing conditions that result in the development of peri-implant fibrosis. After giving credit to macrophages in the inflammatory phase of the FBR, we will mainly focus on the activation of fibroblastic cells into matrix-producing and highly contractile myofibroblasts. While fibrosis has been discussed to be a consequence of the disturbed and chronic inflammatory milieu in the FBR, direct activation of myofibroblasts at the implant surface is less commonly considered. Thus, we will provide a perspective how physical properties of the implant surface control myofibroblast actions and accumulation of stiff scar tissue. Because formation of scar tissue at the surface and around implant materials is a major reason for device failure and extraction surgeries, providing implant surfaces with myofibroblast-suppressing features is a first step to enhance implant acceptance and functional lifetime. Alternative therapeutic targets are elements of the myofibroblast mechanotransduction and contractile machinery and we will end with a brief overview on such targets that are considered for the treatment of other organ fibroses.
Collapse
|
29
|
Xuan B, Ghosh D, Dawson MR. Contributions of the distinct biophysical phenotype of polyploidal giant cancer cells to cancer progression. Semin Cancer Biol 2021; 81:64-72. [PMID: 33992783 DOI: 10.1016/j.semcancer.2021.05.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 05/03/2021] [Accepted: 05/10/2021] [Indexed: 01/27/2023]
Abstract
Polyploid giant cancer cells (PGCCs) are a commonly observed histological feature of human tumors and are particularly prominent in late stage and drug resistant cancers. The chromosomal duplication conferred by their aneuploidy gives rise to DNA damage resistance and complex tumor cell karyotypes, a driving factor in chemotherapy resistance and disease relapse. Furthermore, PGCCs also exhibit key cytoskeletal features that give rise to a distinct biophysical phenotype, including increased density of polymerized actin and vimentin intermediate filaments, nuclear and cytoskeletal stiffening, increased traction force, and migratory persistence. Despite recent research highlighting the role PGCCs play in cancer progression, this population of tumor cells remains poorly characterized in terms of their biophysical properties. In this review, we will discuss the various aspects of their biomolecular phenotype, such as increased stemness as well as a mixed EMT signature. These features have been extensively associated with tumorigenesis and recurrence, and aggressive cancers. Additionally, we will also examine the distinct PGCC cytoskeletal features of actin and filamentous vimentin. Specifically, how the differential organization of these networks serve to support their increased size and drive migratory persistence. These findings could shed light on potential therapeutic strategies that allow for specific elimination or mitigation of the invasive potential of these polyploid cancer cells. Lastly, we will examine how the biophysical and molecular phenotype of PGCCs combine to tip the scale in favor of promoting cancer progression, presenting an important target in the clinical treatment of cancer.
Collapse
Affiliation(s)
- Botai Xuan
- Brown University, Department of Molecular Pharmacology, Physiology, & Biotechnology, Providence, 02912, USA
| | - Deepraj Ghosh
- Brown University, Department of Molecular Pharmacology, Physiology, & Biotechnology, Providence, 02912, USA
| | - Michelle R Dawson
- Brown University, Department of Molecular Pharmacology, Physiology, & Biotechnology, Providence, 02912, USA; Brown University, Center for Biomedical Engineering, Providence, 02912, USA; Brown University, School of Engineering, Providence, 02912, USA.
| |
Collapse
|
30
|
ARHGEF10L Promotes Cervical Tumorigenesis via RhoA-Mediated Signaling. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:6683264. [PMID: 33833821 PMCID: PMC8012150 DOI: 10.1155/2021/6683264] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/10/2021] [Accepted: 03/17/2021] [Indexed: 11/17/2022]
Abstract
Background Rho guanine nucleotide exchange factor 10-like protein (ARHGEF10L) is a member of the guanine nucleotide exchange factor family, which regulates Rho GTPase activities, thus contributing to tumorigenesis. Our previous study demonstrated a strong association between the ARHGEF10L gene and the risk of cervical carcinoma. This study investigated the pathogenic role and mechanism of ARHGEF10L in cervical tumors. Methods The HeLa cell line, which was derived from cervical carcinoma, was transfected with ARHGEF10L-overexpressing plasmids or anti-ARHGEF10L siRNA. Cell counting kit-8 assays, wound-healing assays, and cell apoptosis assays were performed to investigate the effects of ARHGEF10L on cell activities. A Rho pull-down assay and RNA-sequencing analysis were performed to investigate the pathogenic pathway of ARHGEF10L involvement in cervical tumors. Results ARHGEF10L overexpression promoted cell proliferation and migration, reduced cell apoptosis, and induced epithelial-to-mesenchymal transition (EMT) via downregulation of E-cadherin and upregulation of N-cadherin and Slug in transfected HeLa cells. The overexpression of ARHGEF10L also upregulated GTP-RhoA, ROCK1, and phospho-ezrin/radixin/moesin (ERM) expression in HeLa cells. RNA-sequencing analysis detected altered transcription of 31 genes in HeLa cells with ARHGEF10L overexpression. Kyoto Encyclopedia of Genes and Genomes (KEGG) and gene ontology (GO) pathway analyses identified significant differences in cyclin-dependent protein serine/threonine kinase activity, cell responses to vitamin A, and Toll-like receptor signaling pathways. Both real-time PCR and Western blotting verified the increased expression of heat shock 70 kDa protein 6 (HSPA6) in ARHGEF10L-overexpressing HeLa cells. Since we reported that ARHGEF10L played a role through RhoA-ROCK1-ERM signaling, an important pathway in tumorigenesis, and stimulated EMT and HSPA6 expression in liver tumors and gastric tumor cells, we suggest that ARHGEF10L is a novel oncogene in many tumors.
Collapse
|
31
|
Xie L, Zeng Y. Therapeutic Potential of Exosomes in Pulmonary Fibrosis. Front Pharmacol 2020; 11:590972. [PMID: 33343360 PMCID: PMC7746877 DOI: 10.3389/fphar.2020.590972] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 11/12/2020] [Indexed: 12/13/2022] Open
Abstract
Pulmonary fibrosis is closely associated with the recruitment of fibroblasts from capillary vessels with damaged endothelial cells, the epithelial mesenchymal transition (EMT) of type II alveolar epithelial cells, and the transformation of fibroblasts to myofibroblasts. Recent studies suggest that EMT is a key factor in the pathogenesis of pulmonary fibrosis, as the disruption of EMT-related effector molecules can inhibit the occurrence and development of PF. With the numerous advancements made in molecular biology in recent years, researchers have discovered that exosomes and their cargos, such as miRNAs, lncRNAs, and proteins, can promote or inhibit the EMT, modulate the transformation of fibroblasts into myofibroblasts, contribute to the proliferation of fibroblasts and promote immunoregulatory and mitochondrial damage during pulmonary fibrosis. Exosomes are key factors regulating the differentiation of bone marrow mesenchymal stem cells (BMSCs) into myofibroblasts. Interestingly, exosomes derived from BMSCs under pathological and physiological conditions may promote or inhibit the EMT of type II alveolar epithelial cells and the transformation of fibroblasts into myofibroblasts to regulate pulmonary fibrosis. Thus, exosomes may become a new direction in the study of drugs for the treatment of pulmonary fibrosis.
Collapse
Affiliation(s)
- Linshen Xie
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Ye Zeng
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Boyle ST, Mittal P, Kaur G, Hoffmann P, Samuel MS, Klingler-Hoffmann M. Uncovering Tumor-Stroma Inter-relationships Using MALDI Mass Spectrometry Imaging. J Proteome Res 2020; 19:4093-4103. [PMID: 32870688 DOI: 10.1021/acs.jproteome.0c00511] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Tumorigenesis involves a complex interplay between genetically modified cancer cells and their adjacent normal tissue, the stroma. We used an established breast cancer mouse model to investigate this inter-relationship. Conditional activation of Rho-associated protein kinase (ROCK) in a model of mammary tumorigenesis enhances tumor growth and progression by educating the stroma and enhancing the production and remodeling of the extracellular matrix. We used peptide matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) to quantify the proteomic changes occurring within tumors and their stroma in their regular spatial context. Peptides were ranked according to their ability to discriminate between the two groups, using a receiver operating characteristic tool. Peptides were identified by liquid chromatography tandem mass spectrometry, and protein expression was validated by quantitative immunofluorescence using an independent set of tumor samples. We have identified and validated four key proteins upregulated in ROCK-activated mammary tumors relative to those expressing kinase-dead ROCK, namely, collagen I, α-SMA, Rab14, and tubulin-β4. Rab14 and tubulin-β4 are expressed within tumor cells, whereas collagen I is localized within the stroma. α-SMA is predominantly localized within the stroma but is also expressed at higher levels in the epithelia of ROCK-activated tumors. High expression of COL1A, the gene encoding the pro-α 1 chain of collagen, correlates with cancer progression in two human breast cancer genomic data sets, and high expression of COL1A and ACTA2 (the gene encoding α-SMA) are associated with a low survival probability (COLIA, p = 0.00013; ACTA2, p = 0.0076) in estrogen receptor-negative breast cancer patients. To investigate whether ROCK-activated tumor cells cause stromal cancer-associated fibroblasts (CAFs) to upregulate expression of collagen I and α-SMA, we treated CAFs with medium conditioned by primary mammary tumor cells in which ROCK had been activated. This led to abundant production of both proteins in CAFs, clearly highlighting the inter-relationship between tumor cells and CAFs and identifying CAFs as the potential source of high levels of collagen 1 and α-SMA and associated enhancement of tissue stiffness. Our research emphasizes the capacity of MALDI-MSI to quantitatively assess tumor-stroma inter-relationships and to identify potential prognostic factors for cancer progression in human patients, using sophisticated mouse cancer models.
Collapse
Affiliation(s)
- Sarah T Boyle
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide SA 5000, Australia
| | - Parul Mittal
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia
| | - Gurjeet Kaur
- Institute for Research in Molecular Medicine, Universiti Sains Malaysia, Minden 11800 Pulau Pinang, Malaysia
| | - Peter Hoffmann
- Future Industries Institute, University of South Australia, Mawson Lakes SA 5095, Australia
| | - Michael S Samuel
- Centre for Cancer Biology, SA Pathology and the University of South Australia, Adelaide SA 5000, Australia.,Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, Adelaide SA 5000, Australia
| | | |
Collapse
|
33
|
Li L, Xu X, Du Y, Zhang M, Feng Y, Qian X, Li S, Du T, Peng X, Chen F. ATPR induces acute promyelocytic leukemia cells differentiation and growth arrest by blockade of SHP2/Rho/ROCK1 pathway. Toxicol Appl Pharmacol 2020; 399:115053. [DOI: 10.1016/j.taap.2020.115053] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 05/04/2020] [Accepted: 05/12/2020] [Indexed: 01/04/2023]
|
34
|
Bin J, Nie S, Tang Z, Kang A, Fu Z, Hu Y, Liao Q, Xiong W, Zhou Y, Tang Y, Jiang J. Long noncoding RNA EPB41L4A-AS1 functions as an oncogene by regulating the Rho/ROCK pathway in colorectal cancer. J Cell Physiol 2020; 236:523-535. [PMID: 32557646 DOI: 10.1002/jcp.29880] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 12/20/2022]
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors worldwide. In terms of cancer-related death, colon cancer ranks second and third among men and women, respectively, and the incidence is increasing annually. Accumulating evidence have indicated that long noncoding RNA (lncRNA) plays an important role in tumorigenesis. In this study, we found that lncRNA EPB41L4A-AS1 was highly expressed in CRC tissues and was associated with poor prognosis and tumor metastasis in patients with CRC. In vitro studies showed that the knockdown of EPB41L4A-AS1 inhibited the proliferation, migration, invasion, and epithelial-mesenchymal transition of CRC cells. Mechanically, we found that EPB41L4A-AS1 may participate in the development of CRC by activating the Rho/Rho-associated protein kinase signaling pathway. Collectively, these results demonstrated that EPB41L4A-AS1 can promote the proliferation, invasion, and migration of CRC, and it may be a novel biomarker for the diagnosis and targeted treatment of CRC.
Collapse
Affiliation(s)
- Jie Bin
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Shaolin Nie
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Ziyuan Tang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Anding Kang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhongping Fu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yingbin Hu
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianjin Liao
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Central Laboratory, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Wei Xiong
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute, Central South University, Changsha, Hunan, China
| | - Yujuan Zhou
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yanyan Tang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Central Laboratory, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiarui Jiang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China.,Department of Colorectal Surgery, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
35
|
Palhares LCGF, Barbosa JS, Scortecci KC, Rocha HAO, Brito AS, Chavante SF. In vitro antitumor and anti-angiogenic activities of a shrimp chondroitin sulfate. Int J Biol Macromol 2020; 162:1153-1165. [PMID: 32553958 DOI: 10.1016/j.ijbiomac.2020.06.100] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 06/01/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
Abstract
Thrombin triggers cellular responses that are crucial for development and progression of cancer, such as proliferation, migration, oncogene expression and angiogenesis. Thus, biomolecules capable of inhibiting this protease have become targets in cancer research. The present work describes the in vitro antitumor properties of a chondroitin sulfate with anti-thrombin activity, isolated from the Litopenaeus vannamei shrimp (sCS). Although the compound was unable to induce cytotoxicity or cell death and/or cell cycle changes after 24 h incubation, it showed a long-term antiproliferative effect, reducing the tumor colony formation of melanoma cells by 75% at 100 μg/mL concentration and inhibiting the anchorage-independent colony formation. sCS reduced 66% of melanoma cell migration in the wound healing assay and 70% in the transwell assay. The compound also decreased melanin and TNF-α content of melanoma cells by 52% and 75% respectively. Anti-angiogenic experiments showed that sCS promoted 100% reduction of tubular structure formation at 100 μg/mL. These results are in accordance with the sCS-mediated in vitro expression of genes related to melanoma development (Cx-43, MAPK, RhoA, PAFR, NFKB1 and VEGFA). These findings bring a new insight to CS molecules in cancer biology that can contribute to ongoing studies for new approaches in designing anti-tumor therapy.
Collapse
Affiliation(s)
- Lais C G F Palhares
- Programa de Pós-graduação em Bioquímica e Biologia Molecular, Departamento de Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Jefferson S Barbosa
- Programa de Pós-graduação em Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil; Instituto Federal de Educação, Ciência e Tecnologia do Rio Grande do Norte, Campus São Gonçalo do Amarante, RN, Brazil
| | - Kátia C Scortecci
- Departamento de Biologia celular e genética, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Hugo A O Rocha
- Programa de Pós-graduação em Bioquímica e Biologia Molecular, Departamento de Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil
| | - Adriana S Brito
- Programa de Pós-graduação em Bioquímica e Biologia Molecular, Departamento de Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil; Faculdade de Ciências da Saúde do Trairi, Universidade Federal do Rio Grande do Norte, Santa Cruz, RN, Brazil.
| | - Suely F Chavante
- Programa de Pós-graduação em Bioquímica e Biologia Molecular, Departamento de Bioquímica, Universidade Federal do Rio Grande do Norte, Natal, RN, Brazil.
| |
Collapse
|
36
|
Cancer associated fibroblast: Mediators of tumorigenesis. Matrix Biol 2020; 91-92:19-34. [PMID: 32450219 DOI: 10.1016/j.matbio.2020.05.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 05/08/2020] [Accepted: 05/12/2020] [Indexed: 02/07/2023]
Abstract
It is well accepted that the tumor microenvironment plays a pivotal role in cancer onset, development, and progression. The majority of clinical interventions are designed to target either cancer or stroma cells. These emphases have been directed by one of two prevailing theories in the field, the Somatic Mutation Theory and the Tissue Organization Field Theory, which represent two seemingly opposing concepts. This review proposes that the two theories are mutually inclusive and should be concurrently considered for cancer treatments. Specifically, this review discusses the dynamic and reciprocal processes between stromal cells and extracellular matrices, using pancreatic cancer as an example, to demonstrate the inclusivity of the theories. Furthermore, this review highlights the functions of cancer associated fibroblasts, which represent the major stromal cell type, as important mediators of the known cancer hallmarks that the two theories attempt to explain.
Collapse
|
37
|
Li X, Wang J. Mechanical tumor microenvironment and transduction: cytoskeleton mediates cancer cell invasion and metastasis. Int J Biol Sci 2020; 16:2014-2028. [PMID: 32549750 PMCID: PMC7294938 DOI: 10.7150/ijbs.44943] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/15/2020] [Indexed: 12/13/2022] Open
Abstract
Metastasis is a complicated, multistep process that is responsible for over 90% of cancer-related death. Metastatic disease or the movement of cancer cells from one site to another requires dramatic remodeling of the cytoskeleton. The regulation of cancer cell migration is determined not only by biochemical factors in the microenvironment but also by the biomechanical contextual information provided by the extracellular matrix (ECM). The responses of the cytoskeleton to chemical signals are well characterized and understood. However, the mechanisms of response to mechanical signals in the form of externally applied force and forces generated by the ECM are still poorly understood. Furthermore, understanding the way cellular mechanosensors interact with the physical properties of the microenvironment and transmit the signals to activate the cytoskeletal movements may help identify an effective strategy for the treatment of cancer. Here, we will discuss the role of tumor microenvironment during cancer metastasis and how physical forces remodel the cytoskeleton through mechanosensing and transduction.
Collapse
Affiliation(s)
- Xingchen Li
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
| | - Jianliu Wang
- Department of Obstetrics and Gynecology, Peking University People's Hospital, Beijing, 100044, China
- Beijing Key Laboratory of Female Pelvic Floor Disorders Diseases, Beijing, 100044, China
| |
Collapse
|
38
|
Porazinski S, Parkin A, Pajic M. Rho-ROCK Signaling in Normal Physiology and as a Key Player in Shaping the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1223:99-127. [PMID: 32030687 DOI: 10.1007/978-3-030-35582-1_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The Rho-ROCK signaling network has a range of specialized functions of key biological importance, including control of essential developmental processes such as morphogenesis and physiological processes including homeostasis, immunity, and wound healing. Deregulation of Rho-ROCK signaling actively contributes to multiple pathological conditions, and plays a major role in cancer development and progression. This dynamic network is critical in modulating the intricate communication between tumor cells, surrounding diverse stromal cells and the matrix, shaping the ever-changing microenvironment of aggressive tumors. In this chapter, we overview the complex regulation of the Rho-ROCK signaling axis, its role in health and disease, and analyze progress made with key approaches targeting the Rho-ROCK pathway for therapeutic benefit. Finally, we conclude by outlining likely future trends and key questions in the field of Rho-ROCK research, in particular surrounding Rho-ROCK signaling within the tumor microenvironment.
Collapse
Affiliation(s)
- Sean Porazinski
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia.,Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia
| | - Ashleigh Parkin
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia
| | - Marina Pajic
- Personalised Cancer Therapeutics Lab, The Kinghorn Cancer Centre, Sydney, NSW, Australia. .,Faculty of Medicine, St Vincent's Clinical School, University of NSW, Sydney, NSW, Australia.
| |
Collapse
|