1
|
Duff D, Gargan S, Long A. Non-muscle myosin heavy chain IIA regulates cell morphology, stress fibre structure, and cell migration in FLO-1 oesophageal adenocarcinoma cells. Hum Cell 2025; 38:80. [PMID: 40164920 PMCID: PMC11958448 DOI: 10.1007/s13577-025-01196-w] [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: 08/07/2024] [Accepted: 02/27/2025] [Indexed: 04/02/2025]
Abstract
The incidence of oesophageal adenocarcinoma (OAC) is increasing at a rapid rate in Western countries. Early oesophageal cancer is often asymptomatic and metastatic disease is common at presentation leading to poor prognosis and survival rates. Cell migration is tightly controlled in the healthy cell but can become dysregulated in diseases such as OAC where increased cell motility and migration can contribute to metastasis. We investigated the role of an actin-based molecular motor, non-muscle myosin heavy chain IIA (NMHCIIA) in the migratory capacity of oesophageal adenocarcinoma cells. Immunofluorescence microscopy and ratiometric imaging demonstrated that NMHCIIA co-localises with F-actin at the leading edge and retracting rear of migrating FLO-1 OAC cells. siRNA-mediated depletion of NMHCIIA from FLO-1 cells altered cell morphology, gave rise to an increased number of stress fibre like structures and reduced FLO-1 cell migration. These findings suggest that NMHCIIA influences FLO-1 cell migration by regulating F-actin dynamics and the actin cytoskeleton, providing insight into the mechanisms of migration employed by OAC cells and identifying NMHCIIA as a potential therapeutic target for this disease.
Collapse
Affiliation(s)
- Deirdre Duff
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Siobhan Gargan
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland
| | - Aideen Long
- Trinity Translational Medicine Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
2
|
Stoessel MB, Stowell RD, Lowery RL, Le LHD, Vu AN, Whitelaw BS, Majewska AK. The effects of P2Y12 loss on microglial gene expression, dynamics, and injury response in the cerebellum and cerebral cortex. Brain Behav Immun 2025; 128:99-120. [PMID: 40174868 DOI: 10.1016/j.bbi.2025.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/26/2025] [Accepted: 03/29/2025] [Indexed: 04/04/2025] Open
Affiliation(s)
- Mark B Stoessel
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA; Neuroscience Graduate Program, University of Rochester, Rochester, NY 14642, USA.
| | - Rianne D Stowell
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA.
| | - Rebecca L Lowery
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA
| | - Linh H D Le
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA; Neuroscience Graduate Program, University of Rochester, Rochester, NY 14642, USA.
| | - Andy N Vu
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA
| | - Brendan S Whitelaw
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA; Neuroscience Graduate Program, University of Rochester, Rochester, NY 14642, USA; Medical Scientist Training Program, University of Rochester 14642, USA
| | - Ania K Majewska
- Department of Neuroscience, Delmonte Institute for Neuroscience, University of Rochester, Rochester NY 14642, USA; Center for Visual Science, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
3
|
Diniz-Filho JF, Silva ACM, Teixeira AL, Sousa BLN, Santos-Oliveira R, Silva GEB, dos Santos CC, Alencar LMR. Penile Cancer: Innovations in Ultrastructural and Vibrational Markers. ACS OMEGA 2025; 10:3449-3461. [PMID: 39926554 PMCID: PMC11800048 DOI: 10.1021/acsomega.4c07293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 01/13/2025] [Accepted: 01/17/2025] [Indexed: 02/11/2025]
Abstract
Penile cancer (PCa) is a disease that manifests predominantly as squamous cell carcinomas (SCCs), which, although rare, represents a significant public health problem, especially in regions with less socioeconomic development. One of the biggest challenges in managing this disease is the difficulty in differentiating tumor subtypes, making accurate diagnosis and treatment challenging. In this context, new characterization techniques are needed to investigate these tumors more completely. Atomic force microscopy (AFM) and Raman spectroscopy (RS) are valuable in this context, providing quantitative and qualitative ultrastructural data and vibrational signatures of the analyzed samples. In this study, AFM and RS techniques were employed to investigate subtypes of penile cancer, including the highly aggressive basaloid subtype, which is closely associated with human papillomavirus (HPV), and the sarcomatoid subtype, comparing them with nontumorous tissues. The AFM results revealed nanoscale changes in the ultrastructural properties of tumor samples, such as increased roughness in tumor tissues, with emphasis on the basaloid type associated with the HPV virus, and reduction in the surface area and volume of tumor tissues at the nanoscale, suggesting deeper tissue infiltration and greater deformability of tumor samples at the nanoscale. RS results detected significant spectral differences between normal and cancerous tissues and between tumor subtypes, particularly in vibrational modes related to proteins and lipids. Principal component analysis (PCA) confirmed a strong discriminative power between control and PCa groups. The data presented here offers new insights into the characteristics of penile tumors that, when integrated with clinical analyses, could improve the understanding of penile cancer behavior, contributing to more accurate diagnostic methods and targeted treatments.
Collapse
Affiliation(s)
- Joel Félix
Silva Diniz-Filho
- Biophysics
and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Ana Caroline Muniz Silva
- Biophysics
and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Antônio
Augusto Lima Teixeira
- Immunofluorescence
and Electron Microscopy Laboratory (LIME/HUUFMA), Department of Medicine, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Bruna Larissa Nolêto Sousa
- Immunofluorescence
and Electron Microscopy Laboratory (LIME/HUUFMA), Department of Medicine, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Ralph Santos-Oliveira
- Brazilian
Nuclear Energy Commission, Institute of
Nuclear Engineering, Laboratory of Nanoradiopharmacy and Synthesis
of New Radiopharmaceuticals, Rio de Janeiro 21941906, Brazil
- State
University of Rio de Janeiro, Laboratory of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro 21941906, Brazil
| | - Gyl Eanes Barros Silva
- Immunofluorescence
and Electron Microscopy Laboratory (LIME/HUUFMA), Department of Medicine, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Clenilton Costa dos Santos
- Biophysics
and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Biophysics
and Nanosystems Laboratory, Department of Physics, Federal University of Maranhão, São Luís, Maranhão 65080-805, Brazil
| |
Collapse
|
4
|
Rolfs LA, Falat EJ, Gutzman JH. myh9b is a critical non-muscle myosin II encoding gene that interacts with myh9a and myh10 during zebrafish development in both compensatory and redundant pathways. G3 (BETHESDA, MD.) 2025; 15:jkae260. [PMID: 39503257 PMCID: PMC11708221 DOI: 10.1093/g3journal/jkae260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024]
Abstract
Non-muscle myosin (NMII) motor proteins have diverse developmental functions due to their roles in cell shape changes, cell migration, and cell adhesion. Zebrafish are an ideal vertebrate model system to study the NMII encoding myh genes and proteins due to high sequence homology, established gene editing tools, and rapid ex utero development. In humans, mutations in the NMII encoding MYH genes can lead to abnormal developmental processes and disease. This study utilized zebrafish myh9a, myh9b, and myh10 null mutants to examine potential genetic interactions and roles for each gene in development. It was determined that the myh9b gene is the most critical NMII encoding gene, as myh9b mutants develop pericardial edema and have a partially penetrant lethal phenotype, which was not observed in the other myh mutants. This study also established that genetic interactions occur between the zebrafish myh9a, myh9b, and myh10 genes where myh9b is required for the expression of both myh9a and myh10, and myh10 is required for the expression of myh9b. Additionally, protein analyses suggested that enhanced NMII protein stability in some mutant backgrounds may play a role in compensation. Finally, double mutant studies revealed different and more severe phenotypes at earlier time points than single mutants, suggesting roles for tissue specific genetic redundancy, and in some genotypes, haploinsufficiency. These mutants are the first in vivo models allowing for the study of complete loss of the NMIIA and NMIIB proteins, establishing them as valuable tools to elucidate the role of NMII encoding myh genes in development and disease.
Collapse
Affiliation(s)
- Laura A Rolfs
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Elizabeth J Falat
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| | - Jennifer H Gutzman
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA
| |
Collapse
|
5
|
Kyriazi D, Voth L, Bader A, Ewert W, Gerlach J, Elfrink K, Franz P, Tsap MI, Schirmer B, Damiano-Guercio J, Hartmann FK, Plenge M, Salari A, Schöttelndreier D, Strienke K, Bresch N, Salinas C, Gutzeit HO, Schaumann N, Hussein K, Bähre H, Brüsch I, Claus P, Neumann D, Taft MH, Shcherbata HR, Ngezahayo A, Bähler M, Amiri M, Knölker HJ, Preller M, Tsiavaliaris G. An allosteric inhibitor of RhoGAP class-IX myosins suppresses the metastatic features of cancer cells. Nat Commun 2024; 15:9947. [PMID: 39550360 PMCID: PMC11569205 DOI: 10.1038/s41467-024-54181-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 11/01/2024] [Indexed: 11/18/2024] Open
Abstract
Aberrant Ras homologous (Rho) GTPase signalling is a major driver of cancer metastasis, and GTPase-activating proteins (GAPs), the negative regulators of RhoGTPases, are considered promising targets for suppressing metastasis, yet drug discovery efforts have remained elusive. Here, we report the identification and characterization of adhibin, a synthetic allosteric inhibitor of RhoGAP class-IX myosins that abrogates ATPase and motor function, suppressing RhoGTPase-mediated modes of cancer cell metastasis. In human and murine adenocarcinoma and melanoma cell models, including three-dimensional spheroid cultures, we reveal anti-migratory and anti-adhesive properties of adhibin that originate from local disturbances in RhoA/ROCK-regulated signalling, affecting actin-dynamics and actomyosin-based cell-contractility. Adhibin blocks membrane protrusion formation, disturbs remodelling of cell-matrix adhesions, affects contractile ring formation, and disrupts epithelial junction stability; processes severely impairing single/collective cell migration and cytokinesis. Combined with the non-toxic, non-pathological signatures of adhibin validated in organoids, mouse and Drosophila models, this mechanism of action provides the basis for developing anti-metastatic cancer therapies.
Collapse
Affiliation(s)
- Despoina Kyriazi
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Lea Voth
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Almke Bader
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Wiebke Ewert
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
- Institute for Functional Gene Analytics (IFGA), Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | | | - Kerstin Elfrink
- Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany
| | - Peter Franz
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Mariana I Tsap
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany
| | - Bastian Schirmer
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | | | - Falk K Hartmann
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Masina Plenge
- Department of Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz Universität Hannover, Hannover, Germany
| | - Azam Salari
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | - Katharina Strienke
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Nadine Bresch
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | - Claudio Salinas
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | | | - Nora Schaumann
- Institute for Pathology, Hannover Medical School, Hannover, Germany
| | - Kais Hussein
- Institute of Pathology, KRH Klinikum Nordstadt, Hannover, Germany
| | - Heike Bähre
- Research Core Unit Mass Spectrometry-Metabolomics, Hannover Medical School, Hanover, Germany
| | - Inga Brüsch
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Peter Claus
- SMATHERIA gGmbH-Non-Profit Biomedical Research Institute, Hannover, Germany
| | - Detlef Neumann
- Institute of Pharmacology, Hannover Medical School, Hannover, Germany
| | - Manuel H Taft
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
| | | | - Anaclet Ngezahayo
- Department of Cell Physiology and Biophysics, Institute of Cell Biology and Biophysics, Leibniz Universität Hannover, Hannover, Germany
| | - Martin Bähler
- Institute of Integrative Cell Biology and Physiology, University of Münster, Münster, Germany
| | - Mahdi Amiri
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany
| | | | - Matthias Preller
- Institute for Biophysical Chemistry, Hannover Medical School, Hannover, Germany
- Institute for Functional Gene Analytics (IFGA), Bonn-Rhein-Sieg University of Applied Sciences, Rheinbach, Germany
| | | |
Collapse
|
6
|
Montero-Calle A, Garranzo-Asensio M, Poves C, Sanz R, Dziakova J, Peláez-García A, de Los Ríos V, Martinez-Useros J, Fernández-Aceñero MJ, Barderas R. In-Depth Proteomic Analysis of Paraffin-Embedded Tissue Samples from Colorectal Cancer Patients Revealed TXNDC17 and SLC8A1 as Key Proteins Associated with the Disease. J Proteome Res 2024; 23:4802-4820. [PMID: 39441737 DOI: 10.1021/acs.jproteome.3c00749] [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] [Indexed: 10/25/2024]
Abstract
A deeper understanding of colorectal cancer (CRC) biology would help to identify specific early diagnostic markers. Here, we conducted quantitative proteomics on FFPE healthy, adenoma, and adenocarcinoma tissue samples from six stage I sporadic CRC patients to identify dysregulated proteins during early CRC development. Two independent quantitative 10-plex TMT experiments were separately performed. After protein extraction, trypsin digestion, and labeling, proteins were identified and quantified by using a Q Exactive mass spectrometer. A total of 2681 proteins were identified and quantified after data analysis and bioinformatics with MaxQuant and the R program. Among them, 284 and 280 proteins showed significant upregulation and downregulation (expression ratio ≥1.5 or ≤0.67, p-value ≤0.05), respectively, in adenoma and/or adenocarcinoma compared to healthy tissue. Ten dysregulated proteins were selected to study their role in CRC by WB, IHC, TMA, and ELISA using tissue and plasma samples from CRC patients, individuals with premalignant colorectal lesions (adenomas), and healthy individuals. In vitro loss-of-function cell-based assays and in vivo experiments using three CRC cell lines with different metastatic properties assessed the important roles of SLC8A1 and TXNDC17 in CRC and liver metastasis. Additionally, SLC8A1 and TXNDC17 protein levels in plasma possessed the diagnostic ability of early CRC stages.
Collapse
Affiliation(s)
- Ana Montero-Calle
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- Proteomics Core UCCTs, Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
| | - María Garranzo-Asensio
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
| | - Carmen Poves
- Gastroenterology Unit, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Rodrigo Sanz
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Jana Dziakova
- Surgical Digestive Department, Hospital Universitario Clínico San Carlos, E-28040 Madrid, Spain
| | - Alberto Peláez-García
- Molecular Pathology and Therapeutic Targets Group, Hospital La Paz Institute for Health Research (IdiPAZ), E-28046 Madrid, Spain
| | | | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute-Fundación Jiménez Díaz, Fundación Jiménez Díaz University Hospital/Universidad Autónoma de Madrid (IIS-FJD/UAM), E-28040 Madrid, Spain
- Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos University, Alcorcón, E-28922 Madrid, Spain
| | | | - Rodrigo Barderas
- Chronic Disease Programme (UFIEC), Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- Proteomics Core UCCTs, Instituto de Salud Carlos III, Majadahonda, E-28220 Madrid, Spain
- CIBER of Frailty and Healthy Aging (CIBERFES), E-28029 Madrid, Spain
| |
Collapse
|
7
|
Stoessel MB, Stowell RD, Lowery RL, Le L, Vu AN, Whitelaw BS, Majewska AK. The effects of P2Y12 loss on microglial gene expression, dynamics, and injury response in the cerebellum and cerebral cortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614526. [PMID: 39386439 PMCID: PMC11463386 DOI: 10.1101/2024.09.25.614526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Despite the emerging consensus that microglia are critical to physiological and pathological brain function, it is unclear how microglial roles and their underlying mechanisms differ between brain regions. Microglia throughout the brain express common markers, such as the purinergic receptor P2Y12, that delineate them from peripheral macrophages. P2Y12 is a critical sensor of injury but also contributes to the sensing of neuronal activity and remodeling of synapses, with microglial loss of P2Y12 resulting in behavioral deficits. P2Y12 has largely been studied in cortical microglia, despite the fact that a growing body of evidence suggests that microglia exhibit a high degree of regional specialization. Cerebellar microglia, in particular, exhibit transcriptional, epigenetic, and functional profiles that set them apart from their better studied cortical and hippocampal counterparts. Here, we demonstrate that P2Y12 deficiency does not alter the morphology, distribution, or dynamics of microglia in the cerebellum. In fact, loss of P2Y12 does little to disturb the distinct transcriptomic profiles of cortical and cerebellar microglia. However, unlike in cortex, P2Y12 is not required for a full microglial response to focal injury, suggesting that cerebellar and cortical microglia use different cues to respond to injury. Finally, we show that P2Y12 deficiency impairs cerebellar learning in a delay eyeblink conditioning task, a common test of cerebellar plasticity and circuit function. Our findings suggest not only region-specific roles of microglial P2Y12 signaling in the focal injury response, but also indicate a conserved role for P2Y12 in microglial modulation of plasticity across regions.
Collapse
|
8
|
Lv HB, Wu QY, Zhang YJ, Quan SW, Ma N, Dai YQ, Sun Y. Study on the expression and prognostic relationship of MYL6B in liver cancer based on bioinformatics. World J Clin Oncol 2024; 15:1188-1197. [PMID: 39351463 PMCID: PMC11438851 DOI: 10.5306/wjco.v15.i9.1188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/21/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
BACKGROUND Primary liver cancer is a prevalent and deadly cancer type. Despite treatment advances, prognosis remains poor, with high recurrence rates. Early detection is crucial but challenging due to the disease's insidious nature. Myosin proteins play significant roles in cancer development, influencing cell migration, invasion, and tumor suppression. MYL6B, a myosin light chain, is involved in various cellular processes and has been associated with poor prognosis in colorectal adenocarcinoma and potential as a biomarker in breast cancer. AIM To investigate the expression of MYL6B in liver hepatocellular carcinoma (LIHC) and its impact on prognosis and potential mechanisms of action using bioinformatics methods. METHODS The expression of MYL6B in pan-cancer and normal tissues was analyzed using the gene expression profiling interactive analysis 2 and tumor immune estimation resource databases. The expression level of MYL6B in LIHC tissues and its relationship with prognosis were analyzed, immunohistochemical analysis of MYL6B and its effect on immune cell infiltration, and the protein network were further studied. RESULTS MYL6B was highly expressed in diffuse large b-cell lymphoma, LIHC, pancreatic adenocarcinoma, skin cutaneous melanoma, thymoma, uterine corpus endometrial carcinoma, uterine carcinosarcoma, and lowly expressed in kidney chromophobe, acute myeloid leukemia, testicular germ cell tumors. The expression level of MYL6B was significantly different between cancer and normal tissues. It had a significant impact on both overall survival and disease-free survival. MYL6B is highly expressed in hepatocellular carcinoma and its expression level increases with cancer progression. High MYL6B expression is associated with poor prognosis in terms of overall survival and recurrence-free survival. The immunohistochemical level of MYL6B is high in hepatocellular carcinoma tissues, and MYL6B has a high level of immune infiltration inflammation. In protein network analysis, MYL6B is correlated with MYL2, MYL6, MYL9, MYLK4, MYLK2, MYL12A, MYL12B, MYH11, MYH9 and MYH10. CONCLUSION The expression level of MYL6B in LIHC was significantly higher than in normal liver tissues, and it was correlated with the degree of differentiation survival rate, and immune infiltration. MYL6B is a potential target for LIHC treatment.
Collapse
Affiliation(s)
- Hai-Bing Lv
- Department of General Surgery, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Qing-Yun Wu
- Department of General Surgery, Xianning Central Hospital, Xianning 437000, Hubei Province, China
| | - Yu-Jiao Zhang
- Department of Medical oncology, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Sheng-Wei Quan
- Department of General Surgery, Beidahuang Group General Hospital, Harbin 150000, Heilongjiang Province, China
| | - Ning Ma
- Department of General Surgery, Daqing Oilfield General Hospital, Daqing 163000, Heilongjiang Province, China
| | - Yu-Qing Dai
- College of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui Province, China
| | - Yan Sun
- Department of General Surgery, Second Affiliated Hospital of Harbin Medical University, Harbin 150000, Heilongjiang Province, China
| |
Collapse
|
9
|
Leineweber WD, Rowell MZ, Ranamukhaarachchi SK, Walker A, Li Y, Villazon J, Mestre-Farrera A, Hu Z, Yang J, Shi L, Fraley SI. Divergent iron regulatory states contribute to heterogeneity in breast cancer aggressiveness. iScience 2024; 27:110661. [PMID: 39262774 PMCID: PMC11387597 DOI: 10.1016/j.isci.2024.110661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/19/2024] [Accepted: 07/31/2024] [Indexed: 09/13/2024] Open
Abstract
Contact with dense collagen I (Col1) can induce collective invasion of triple negative breast cancer (TNBC) cells and transcriptional signatures linked to poor patient prognosis. However, this response is heterogeneous and not well understood. Using phenotype-guided sequencing analysis of invasive vs. noninvasive subpopulations, we show that these two phenotypes represent opposite sides of the iron response protein 1 (IRP1)-mediated response to cytoplasmic labile iron pool (cLIP) levels. Invasive cells upregulate iron uptake and utilization machinery characteristic of a low cLIP response, which includes contractility regulating genes that drive migration. Non-invasive cells upregulate iron sequestration machinery characteristic of a high cLIP response, which is accompanied by upregulation of actin sequestration genes. These divergent IRP1 responses result from Col1-induced transient expression of heme oxygenase I (HO-1), which cleaves heme and releases iron. These findings lend insight into the emerging theory that heme and iron fluxes regulate TNBC aggressiveness.
Collapse
Affiliation(s)
- William D. Leineweber
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maya Z. Rowell
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | | | - Alyssa Walker
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Yajuan Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Jorge Villazon
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Aida Mestre-Farrera
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Zhimin Hu
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Jing Yang
- Department of Pharmacology, Moores Cancer Center, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
- Department of Pediatrics, University of California, San Diego, School of Medicine, La Jolla, CA 92093, USA
| | - Lingyan Shi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephanie I. Fraley
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
10
|
Katsuta H, Sokabe M, Hirata H. From stress fiber to focal adhesion: a role of actin crosslinkers in force transmission. Front Cell Dev Biol 2024; 12:1444827. [PMID: 39193363 PMCID: PMC11347286 DOI: 10.3389/fcell.2024.1444827] [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: 06/06/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
The contractile apparatus, stress fiber (SF), is connected to the cell adhesion machinery, focal adhesion (FA), at the termini of SF. The SF-FA complex is essential for various mechanical activities of cells, including cell adhesion to the extracellular matrix (ECM), ECM rigidity sensing, and cell migration. This mini-review highlights the importance of SF mechanics in these cellular activities. Actin-crosslinking proteins solidify SFs by attenuating myosin-driven flows of actin and myosin filaments within the SF. In the solidified SFs, viscous slippage between actin filaments in SFs and between the filaments and the surrounding cytosol is reduced, leading to efficient transmission of myosin-generated contractile force along the SFs. Hence, SF solidification via actin crosslinking ensures exertion of a large force to FAs, enabling FA maturation, ECM rigidity sensing and cell migration. We further discuss intracellular mechanisms for tuning crosslinker-modulated SF mechanics and the potential relationship between the aberrance of SF mechanics and pathology including cancer.
Collapse
Affiliation(s)
- Hiroki Katsuta
- Department of Cardiovascular Physiology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Masahiro Sokabe
- Human Information Systems Laboratories, Kanazawa Institute of Technology, Hakusan, Japan
| | - Hiroaki Hirata
- Department of Applied Bioscience, Kanazawa Institute of Technology, Hakusan, Japan
| |
Collapse
|
11
|
Xie X, Sauer F, Grosser S, Lippoldt J, Warmt E, Das A, Bi D, Fuhs T, Käs JA. Effect of non-linear strain stiffening in eDAH and unjamming. SOFT MATTER 2024; 20:1996-2007. [PMID: 38323652 PMCID: PMC10900305 DOI: 10.1039/d3sm00630a] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024]
Abstract
In cell clusters, the prominent factors at play encompass contractility-based enhanced tissue surface tension and cell unjamming transition. The former effect pertains to the boundary effect, while the latter constitutes a bulk effect. Both effects share outcomes of inducing significant elongation in cells. This elongation is so substantial that it surpasses the limits of linear elasticity, thereby giving rise to additional effects. To investigate these effects, we employ atomic force microscopy (AFM) to analyze how the mechanical properties of individual cells change under such considerable elongation. Our selection of cell lines includes MCF-10A, chosen for its pronounced demonstration of the extended differential adhesion hypothesis (eDAH), and MDA-MB-436, selected due to its manifestation of cell unjamming behavior. In the AFM analyses, we observe a common trend in both cases: as elongation increases, both cell lines exhibit strain stiffening. Notably, this effect is more prominent in MCF-10A compared to MDA-MB-436. Subsequently, we employ AFM on a dynamic range of 1-200 Hz to probe the mechanical characteristics of cell spheroids, focusing on both surface and bulk mechanics. Our findings align with the results from single cell investigations. Specifically, MCF-10A cells, characterized by strong contractile tissue tension, exhibit the greatest stiffness on their surface. Conversely, MDA-MB-436 cells, which experience significant elongation, showcase their highest stiffness within the bulk region. Consequently, the concept of single cell strain stiffening emerges as a crucial element in understanding the mechanics of multicellular spheroids (MCSs), even in the case of MDA-MB-436 cells, which are comparatively softer in nature.
Collapse
Affiliation(s)
- Xiaofan Xie
- Soft Matter Physics Division, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Germany.
| | - Frank Sauer
- Soft Matter Physics Division, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Germany.
| | - Steffen Grosser
- Soft Matter Physics Division, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Germany.
| | - Jürgen Lippoldt
- Soft Matter Physics Division, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Germany.
| | - Enrico Warmt
- Soft Matter Physics Division, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Germany.
| | - Amit Das
- Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - Dapeng Bi
- Department of Physics, Northeastern University, Boston, MA 02115, USA
| | - Thomas Fuhs
- Soft Matter Physics Division, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Germany.
| | - Josef A Käs
- Soft Matter Physics Division, Peter Debye Institute for Soft Matter Physics, University of Leipzig, Germany.
| |
Collapse
|
12
|
Leineweber WD, Rowell MZ, Ranamukhaarachchi S, Walker A, Li Y, Villazon J, Farrera AM, Hu Z, Yang J, Shi L, Fraley SI. Divergent iron-regulatory states contribute to heterogeneity in breast cancer aggressiveness. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.23.546216. [PMID: 37425829 PMCID: PMC10327122 DOI: 10.1101/2023.06.23.546216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/11/2023]
Abstract
Primary tumors with similar mutational profiles can progress to vastly different outcomes where transcriptional state, rather than mutational profile, predicts prognosis. A key challenge is to understand how distinct tumor cell states are induced and maintained. In triple negative breast cancer cells, invasive behaviors and aggressive transcriptional signatures linked to poor patient prognosis can emerge in response to contact with collagen type I. Herein, collagen-induced migration heterogeneity within a TNBC cell line was leveraged to identify transcriptional programs associated with invasive versus non-invasive phenotypes and implicate molecular switches. Phenotype-guided sequencing revealed that invasive cells upregulate iron uptake and utilization machinery, anapleurotic TCA cycle genes, actin polymerization promoters, and a distinct signature of Rho GTPase activity and contractility regulating genes. The non-invasive cell state is characterized by actin and iron sequestration modules along with glycolysis gene expression. These unique tumor cell states are evident in patient tumors and predict divergent outcomes for TNBC patients. Glucose tracing confirmed that non-invasive cells are more glycolytic than invasive cells, and functional studies in cell lines and PDO models demonstrated a causal relationship between phenotype and metabolic state. Mechanistically, the OXPHOS dependent invasive state resulted from transient HO-1 upregulation triggered by contact with dense collagen that reduced heme levels and mitochondrial chelatable iron levels. This induced expression of low cytoplasmic iron response genes regulated by ACO1/IRP1. Knockdown or inhibition of HO-1, ACO1/IRP1, MRCK, or OXPHOS abrogated invasion. These findings support an emerging theory that heme and iron flux serve as important regulators of TNBC aggressiveness.
Collapse
|
13
|
Solomatina ES, Nishkomaeva EN, Kovaleva AV, Tvorogova AV, Potashnikova DM, Saidova AA. Parameters of Cell Death and Proliferation of Prostate Cancer Cells with Altered Expression of Myosin 1C Isoforms. DOKL BIOCHEM BIOPHYS 2024; 514:16-22. [PMID: 38189886 PMCID: PMC11021239 DOI: 10.1134/s1607672923700588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 10/17/2023] [Accepted: 10/17/2023] [Indexed: 01/09/2024]
Abstract
Myosin 1C is a monomeric myosin motor with a truncated tail domain. Such motors are referred as slow "tension sensors." Three isoforms of myosin 1C differ in short N-termed amino acid sequences, the functional differences between isoforms have not been elucidated. Myosin 1C isoform A was described as a diagnostic marker for prostate cancer, but its role in tumor transformation remains unknown. Based on data on the functions of myosin 1C, we hypothesized the potential role of myosin 1C isoforms in maintaining the tumor phenotype of prostate cancer cells. In our work, we showed that a decrease in the expression level of myosin 1C isoform C leads to an increase in the proliferative activity of prostate tumor cells.
Collapse
Affiliation(s)
- E S Solomatina
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - E N Nishkomaeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - A V Kovaleva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, Moscow State University, Moscow, Russia
| | - A V Tvorogova
- Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russia
| | - D M Potashnikova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - A A Saidova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia.
- Faculty of Biology, Moscow State University, Moscow, Russia.
| |
Collapse
|
14
|
Wollscheid HP, Ulrich HD. Chromatin meets the cytoskeleton: the importance of nuclear actin dynamics and associated motors for genome stability. DNA Repair (Amst) 2023; 131:103571. [PMID: 37738698 DOI: 10.1016/j.dnarep.2023.103571] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 09/11/2023] [Accepted: 09/13/2023] [Indexed: 09/24/2023]
Abstract
The actin cytoskeleton is of fundamental importance for numerous cellular processes, including intracellular transport, cell plasticity, and cell migration. However, functions of filamentous actin (F-actin) in the nucleus remain understudied due to the comparatively low abundance of nuclear actin and the resulting experimental limitations to its visualization. Owing to recent technological advances such as super-resolution microscopy and the development of nuclear-specific actin probes, essential roles of the actin cytoskeleton in the context of genome maintenance are now emerging. In addition to the contributions of monomeric actin as a component of multiple important nuclear protein complexes, nuclear actin has been found to undergo polymerization in response to DNA damage and DNA replication stress. Consequently, nuclear F-actin plays important roles in the regulation of intra-nuclear mobility of repair and replication foci as well as the maintenance of nuclear shape, two important aspects of efficient stress tolerance. Beyond actin itself, there is accumulating evidence for the participation of multiple actin-binding proteins (ABPs) in the surveillance of genome integrity, including nucleation factors and motor proteins of the myosin family. Here we summarize recent findings highlighting the importance of actin cytoskeletal factors within the nucleus in key genome maintenance pathways.
Collapse
Affiliation(s)
- Hans-Peter Wollscheid
- Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, Mainz D - 55128, Germany.
| | - Helle D Ulrich
- Institute of Molecular Biology gGmbH (IMB), Ackermannweg 4, Mainz D - 55128, Germany.
| |
Collapse
|
15
|
Yang A, Zeng K, Huang H, Liu D, Song X, Qian Y, Yu X, Liu D, Zha X, Zhang H, Chai X, Tu P, Hu Z. Usenamine A induces apoptosis and autophagic cell death of human hepatoma cells via interference with the Myosin-9/actin-dependent cytoskeleton remodeling. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 116:154895. [PMID: 37229890 DOI: 10.1016/j.phymed.2023.154895] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 05/09/2023] [Accepted: 05/19/2023] [Indexed: 05/27/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major cause of cancer-associated mortality worldwide. Myosin-9's role in HCC and the anti-HCC effect of the drugs targeting Myosin-9 remain poorly understood so far. Candidate antitumor agents obtained from natural products have attracted worldwide attention. Usenamine A is a novel product, which was first extracted in our laboratory from the lichen Usnea longissima. According to published reports, usenamine A exhibits good antitumor activity, while the mechanisms underlying its antitumor effects remain to be elucidated. PURPOSE The present study investigated the anti-hepatoma effect of usenamine A and the underlying molecular mechanisms, along with evaluating the therapeutic potential of targeting Myosin-9 in HCC. METHODS The CCK-8, Hoechst staining, and FACS assays were conducted in the present study to investigate how usenamine A affected the growth and apoptosis of human hepatoma cells. Moreover, TEM, acridine orange staining, and immunofluorescence assay were performed to explore the induction of autophagy by usenamine A in human hepatoma cells. The usenamine A-mediated regulation of protein expression in human hepatoma cells was analyzed using immunoblotting. MS analysis, SPR assay, CETSA, and molecular modeling were performed to identify the direct target of usenamine A. Immunofluorescence assay and co-immunoprecipitation assay were conducted to determine whether usenamine A affected the interaction between Myosin-9 and the actin present in human hepatoma cells. In addition, the anti-hepatoma effect of usenamine A was investigated in vivo using a xenograft tumor model and the IHC analysis. RESULTS The present study initially revealed that usenamine A could suppress the proliferation of HepG2 and SK-HEP-1 cells (hepatoma cell lines). Furthermore, usenamine A induced cell apoptosis via the activation of caspase-3. In addition, usenamine A enhanced autophagy. Moreover, usenamine A administration could dramatically suppress the carcinogenic ability of HepG2 cells, as evidenced by the nude mouse xenograft tumor model. Importantly, it was initially revealed that Myosin-9 was a direct target of usenamine A. Usenamine A could block cytoskeleton remodeling through the disruption of the interaction between Myosin-9 and actin. Myosin-9 participated in suppressing proliferation while inducing apoptosis and autophagy in response to treatment with usenamine A. In addition, Myosin-9 was revealed as a potential oncogene in HCC. CONCLUSIONS Usenamine A was initially revealed to suppress human hepatoma cells growth by interfering with the Myosin-9/actin-dependent cytoskeleton remodeling through the direct targeting of Myosin-9. Myosin-9 is, therefore, a promising candidate target for HCC treatment, while usenamine A may be utilized as a possible anti-HCC therapeutic, particularly in the treatment of HCC with aberrant Myosin-9.
Collapse
Affiliation(s)
- Ailin Yang
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Kewu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Huiming Huang
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dongxiao Liu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xiaomin Song
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yi Qian
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xuelong Yu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Dan Liu
- Proteomics Laboratory, Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing 100191, China
| | - Xiaojun Zha
- Department of Biochemistry & Molecular Biology, School of Basic Medicine, Anhui Medical University, Hefei 230032, China
| | - Hongbing Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Physiology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100005, China
| | - Xingyun Chai
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Pengfei Tu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| | - Zhongdong Hu
- Modern Research Center for Traditional Chinese Medicine, Beijing Research Institute of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China.
| |
Collapse
|
16
|
Groen L, Kloots I, Englert D, Seto K, Estafanos L, Smith P, Verhaegh GW, Mehra N, Schalken JA. Transcriptome Profiling of Circulating Tumor Cells to Predict Clinical Outcomes in Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2023; 24:ijms24109002. [PMID: 37240349 DOI: 10.3390/ijms24109002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/04/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
The clinical utility of circulating tumor cells (CTC) as a non-invasive multipurpose biomarker is broadly recognized. The earliest methods for enriching CTCs from whole blood rely on antibody-based positive selection. The prognostic utility of CTC enumeration using positive selection with the FDA-approved CellSearchTM system has been demonstrated in numerous studies. The capture of cells with specific protein phenotypes does not fully represent cancer heterogeneity and therefore does not realize the prognostic potential of CTC liquid biopsies. To avoid this selection bias, CTC enrichment based on size and deformability may provide better fidelity, i.e., facilitate the characterization of CTCs with any phenotype. In this study, the recently FDA-approved Parsortix® technology was used to enrich CTCs from prostate cancer (PCa) patients for transcriptome analysis using HyCEADTM technology. A tailored PCa gene panel allowed us to stratify metastatic castration-resistant prostate cancer (mCRPC) patients with clinical outcomes. In addition, our findings suggest that targeted CTC transcriptome profiling may be predictive of therapy response.
Collapse
Affiliation(s)
- Levi Groen
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Iris Kloots
- Department of Medical Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | | | - Kelly Seto
- ANGLE Biosciences Inc., Toronto, ON M9W 1B3, Canada
| | | | - Paul Smith
- ANGLE Biosciences Inc., Toronto, ON M9W 1B3, Canada
| | - Gerald W Verhaegh
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Niven Mehra
- Department of Medical Oncology, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| | - Jack A Schalken
- Department of Urology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
17
|
Jusue-Torres I, Tiv R, Ricarte-Filho JC, Mallisetty A, Contreras-Vargas L, Godoy-Calderon MJ, Khaddour K, Kennedy K, Valyi-Nagy K, David O, Menchaca M, Kottorou A, Koutras A, Dimitrakopoulos F, Abdelhady KM, Massad M, Rubinstein I, Feldman L, Stewart J, Shimamura T, Danilova L, Hulbert A. Myo1e overexpression in lung adenocarcinoma is associated with increased risk of mortality. Sci Rep 2023; 13:4107. [PMID: 36914720 PMCID: PMC10011530 DOI: 10.1038/s41598-023-30765-y] [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: 12/11/2022] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
This study aims to perform a comprehensive genomic analysis to assess the influence of overexpression of MYO1E in non-small cell lung carcinoma (NSCLC) and whether there are differences in survival and mortality risk in NSCLC patients depending on both DNA methylation and RNA expression of MYO1E. The DNA methylation probe cg13887966 was inversely correlated with MYO1E RNA expression in both LUAD and LUSC subpopulations showing that lower MYO1E RNA expression was associated with higher MYO1E DNA methylation. Late stages of lung cancer showed significantly lower MYO1E DNA methylation and significantly higher MYO1E RNA expression for LUAD but not for LUSC. Low DNA methylation as well as high RNA expression of MYO1E are associated with a shorter median survival time and an increased risk of mortality for LUAD, but not for LUSC. This study suggests that changes in MYO1E methylation and expression in LUAD patients may have an essential role in lung cancer's pathogenesis. It shows the utility of MYO1E DNA methylation and RNA expression in predicting survival for LUAD patients. Also, given the low normal expression of MYO1E in blood cells MYO1E DNA methylation has the potential to be used as circulating tumor marker in liquid biopsies.
Collapse
Affiliation(s)
| | - Richies Tiv
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | - Apurva Mallisetty
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Leglys Contreras-Vargas
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | | | - Karam Khaddour
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Kathleen Kennedy
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Klara Valyi-Nagy
- Department of Pathology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Odile David
- Department of Pathology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Martha Menchaca
- Department of Radiology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Anastasia Kottorou
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | - Angelos Koutras
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | - Foteinos Dimitrakopoulos
- Molecular Oncology Laboratory, Division of Oncology, Medical School, University of Patras, Patras, Greece
| | | | - Malek Massad
- Division of Cardiothoracic Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Israel Rubinstein
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA
- Division of Pulmonary, Critical Care, Sleep, and Allergy Medicine, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Lawrence Feldman
- Division of Hematology Oncology, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA
| | - John Stewart
- Department of Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
- Section of Surgical Oncology, Department of Surgery, Louisiana State University, New Orleans, LA, USA
| | - Takeshi Shimamura
- Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
- Division of Cardiothoracic Surgery, University of Illinois at Chicago College of Medicine, Chicago, IL, USA
| | - Ludmila Danilova
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Laboratory of Systems Biology and Computational Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Alicia Hulbert
- Cancer Center, University of Illinois at Chicago, Chicago, IL, USA.
- Medical and Research Services, Jesse Brown VA Medical Center, Chicago, IL, USA.
- Department of Surgery, University of Illinois College of Medicine, 909 South Wolcott Ave, COMRB Suite 5140, Chicago, IL, 60612, USA.
| |
Collapse
|
18
|
MYL5 as a Novel Prognostic Marker is Associated with Immune Infiltrating in Breast Cancer: A Preliminary Study. Breast J 2023; 2023:9508632. [PMID: 36846347 PMCID: PMC9957649 DOI: 10.1155/2023/9508632] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 01/20/2023] [Accepted: 02/01/2023] [Indexed: 02/19/2023]
Abstract
Background Myosin light chain plays a vital regulatory function in a large-scale cellular physiological procedure, however, the role of myosin light chain 5 (MYL5) in breast cancer has not been reported. In this study, we aimed to elucidate the effects of MYL5 on clinical prognosis and immune cell infiltration, and further explore the potential mechanism in breast cancer patients. Methods In this study, we first explored the expression pattern and prognostic value of MYL5 in breast cancer across multiple databases, including Oncomine, TCGA, GTEx, GEPIA2, PrognoScan, and Kaplan-Meier Plotter. The correlations of MYL5 expression with immune cell infiltration and associational gene markers in breast cancer were analyzed by using the TIMER, TIMER2.0, and TISIDB databases. The enrichment and prognosis analysis of MYL5-related genes were implemented by using LinkOmics datasets. Results We found that there was a low expression of MYL5 in breast cancer than in corresponding normal tissue by analyzing the data from Oncomine and TCGA datasets. Furthermore, research showed the prognosis of the MYL5 high-expression group was better than the low-expression group in breast cancer patients. Furthermore, MYL5 expression is markedly related to the tumor-infiltrating immune cells (TIICs), including cancer-associated fibroblast, B cell, CD8+ T cell, CD4+ T cell, macrophage, neutrophil, and dendritic cell, and related to immune molecules as well as the associated gene markers of TIICs. Conclusion MYL5 can serve as a prognostic signature in breast cancer and is associated with immune infiltration. This study first offers a relatively comprehensive understanding of the oncogenic roles of MYL5 for breast cancer.
Collapse
|
19
|
Diaz-Valencia JD, Estrada-Abreo LA, Rodríguez-Cruz L, Salgado-Aguayo AR, Patiño-López G. Class I Myosins, molecular motors involved in cell migration and cancer. Cell Adh Migr 2022; 16:1-12. [PMID: 34974807 PMCID: PMC8741282 DOI: 10.1080/19336918.2021.2020705] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/13/2023] Open
Abstract
Class I Myosins are a subfamily of motor proteins with ATPase activity and a characteristic structure conserved in all myosins: A N-Terminal Motor Domain, a central Neck and a C terminal Tail domain. Humans have eight genes for these myosins. Class I Myosins have different functions: regulate membrane tension, participate in endocytosis, exocytosis, intracellular trafficking and cell migration. Cell migration is influenced by many cellular components including motor proteins, like myosins. Recently has been reported that changes in myosin expression have an impact on the migration of cancer cells, the formation of infiltrates and metastasis. We propose that class I myosins might be potential markers for future diagnostic, prognostic or even as therapeutic targets in leukemia and other cancers.Abbreviations: Myo1g: Myosin 1g; ALL: Acute Lymphoblastic Leukemia, TH1: Tail Homology 1; TH2: Tail Homology 2; TH3: Tail Homology 3.
Collapse
Affiliation(s)
- Juan D. Diaz-Valencia
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| | - Laura A. Estrada-Abreo
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Leonor Rodríguez-Cruz
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Alfonso R. Salgado-Aguayo
- Rheumatic Diseases Laboratory, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Genaro Patiño-López
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| |
Collapse
|
20
|
Nguyen LTS, Jacob MAC, Parajón E, Robinson DN. Cancer as a biophysical disease: Targeting the mechanical-adaptability program. Biophys J 2022; 121:3573-3585. [PMID: 35505610 PMCID: PMC9617128 DOI: 10.1016/j.bpj.2022.04.039] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/30/2022] [Accepted: 04/28/2022] [Indexed: 11/02/2022] Open
Abstract
With the number of cancer cases projected to significantly increase over time, researchers are currently exploring "nontraditional" research fields in the pursuit of novel therapeutics. One emerging area that is steadily gathering interest revolves around cellular mechanical machinery. When looking broadly at the physical properties of cancer, it has been debated whether a cancer could be defined as either stiffer or softer across cancer types. With numerous articles supporting both sides, the evidence instead suggests that cancer is not particularly regimented. Instead, cancer is highly adaptable, allowing it to endure the constantly changing microenvironments cancer cells encounter, such as tumor compression and the shear forces in the vascular system and body. What allows cancer cells to achieve this adaptability are the particular proteins that make up the mechanical network, leading to a particular mechanical program of the cancer cell. Coincidentally, some of these proteins, such as myosin II, α-actinins, filamins, and actin, have either altered expression in cancer and/or some type of direct involvement in cancer progression. For this reason, targeting the mechanical system as a therapeutic strategy may lead to more efficacious treatments in the future. However, targeting the mechanical program is far from trivial. As involved as the mechanical program is in cancer development and metastasis, it also helps drive many other key cellular processes, such as cell division, cell adhesion, metabolism, and motility. Therefore, anti-cancer treatments targeting the mechanical program must take great care to avoid potential side effects. Here, we introduce the potential of targeting the mechanical program while also providing its challenges and shortcomings as a strategy for cancer treatment.
Collapse
Affiliation(s)
- Ly T S Nguyen
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Mark Allan C Jacob
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Eleana Parajón
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland
| | - Douglas N Robinson
- Department of Cell Biology, School of Medicine, Johns Hopkins University, Baltimore, Maryland.
| |
Collapse
|
21
|
Matsushita K, Kobayashi S, Akita H, Konno M, Asai A, Noda T, Iwagami Y, Asaoka T, Gotoh K, Mori M, Doki Y, Eguchi H, Ishii H. Clinicopathological significance of MYL9 expression in pancreatic ductal adenocarcinoma. Cancer Rep (Hoboken) 2022; 5:e1582. [PMID: 34821071 PMCID: PMC9575502 DOI: 10.1002/cnr2.1582] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/31/2021] [Accepted: 10/12/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Pancreatic ductal adenocarcinoma is one of the most aggressive malignancies, and often involves invasion and distant metastasis from the early tumor stages. Myosin II reportedly plays a key role in regulating tumor progression and metastasis. AIMS We examined whether myosin regulatory light polypeptide 9 (MYL9) regulates cancer cell proliferation. METHODS AND RESULTS To investigate the expression pattern and clinical significance of MYL9 in pancreatic ductal adenocarcinoma, we performed immunohistochemical analysis of samples collected from 101 patients with pancreatic ductal adenocarcinoma. The expression of MYL9 was investigated to evaluate its functional role and contribution to proliferation and apoptosis in pancreatic ductal adenocarcinoma cells in vitro. The results showed that MYL9 was predominantly expressed in the cytoplasm and membrane of pancreatic ductal adenocarcinoma cells. Multivariate analysis indicated that MYL9 acted as an independent prognostic factor for overall survival and distant metastasis-free survival. MYL9 expression was strongly associated with malignancy in in vitro analyses, including proliferation and anti-apoptotic activities. CONCLUSIONS Our findings suggest that MYL9 is an independent prognostic factor of pancreatic ductal adenocarcinoma. MYL9 is a crucial biomarker and potential therapeutic target for pancreatic ductal adenocarcinoma.
Collapse
Affiliation(s)
- Katsunori Matsushita
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Shogo Kobayashi
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Hirofumi Akita
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Masamitsu Konno
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Ayumu Asai
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Artificial Intelligence Research CenterThe Institute of Scientific and Industrial Research, Osaka UniversityIbarakiOsakaJapan
- Institute of Scientific and Industrial ResearchOsaka UniversityIbarakiOsakaJapan
| | - Takehiro Noda
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Yoshifumi Iwagami
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Tadafumi Asaoka
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Kunihito Gotoh
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Masaki Mori
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Department of Surgery and ScienceGraduate School of Medical Sciences, Kyushu UniversityFukuokaJapan
| | - Yuichiro Doki
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Hidetoshi Eguchi
- Department of Gastroenterological SurgeryGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| | - Hideshi Ishii
- Center of Medical Innovation and Translational ResearchGraduate School of Medicine, Osaka UniversitySuitaOsakaJapan
| |
Collapse
|
22
|
Jayathirtha M, Neagu AN, Whitham D, Alwine S, Darie CC. Investigation of the effects of downregulation of jumping translocation breakpoint (JTB) protein expression in MCF7 cells for potential use as a biomarker in breast cancer. Am J Cancer Res 2022; 12:4373-4398. [PMID: 36225631 PMCID: PMC9548009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 06/18/2022] [Indexed: 06/16/2023] Open
Abstract
MCF7 is a commonly used luminal type A non-invasive/poor-invasive human breast cancer cell line that does not usually migrate or invade compared with MDA-MB-231 highly metastatic cells, which emphasize an invasive and migratory behavior. Under special conditions, MCF7 cells might acquire invasive features. The aberration in expression and biological functions of the jumping translocation breackpoint (JTB) protein is associated with malignant transformation of cells, based on mitochondrial dysfunction, inhibition of tumor suppressive function of TGF-β, and involvement in cancer cell cycle. To investigate new putative functions of JTB by cellular proteomics, we analyzed the biological processes and pathways that are associated with the JTB protein downregulation. The results demonstrated that MCF7 cell line developed a more "aggressive" phenotype and behavior. Most of the proteins that were overexpressed in this experiment promoted the actin cytoskeleton reorganization that is involved in growth and metastatic dissemination of cancer cells. Some of these proteins are involved in the epithelial-mesenchymal transition (EMT) process (ACTBL2, TUBA4A, MYH14, CSPG5, PKM, UGDH, HSP90AA2, and MIF), in correlation with the energy metabolism reprogramming (PKM, UGDH), stress-response (HSP10, HSP70A1A, HSP90AA2), and immune and inflammatory response (MIF and ERp57-TAPBP). Almost all upregulated proteins in JTB downregulated condition promote viability, motility, proliferation, invasion, survival into a hostile microenvironment, metabolic reprogramming, and escaping of tumor cells from host immune control, leading to a more invasive phenotype for MCF7 cell line. Due to their downregulated condition, four proteins, such as CREBZF, KMT2B, SELENOS and CACNA1I are also involved in maintenance of the invasive phenotype of cancer cells, promoting cell proliferation, migration, invasion and tumorigenesis. Other downregulated proteins, such as MAZ, PLEKHG2, ENO1, TPI2, TOR2A, and CNNM1, may promote suppression of cancer cell growth, invasion, EMT, tumorigenic abilities, interacting with glucose and lipid metabolism, disrupting nuclear envelope stability, or suppressing apoptosis and developing anti-angiogenetic activities. Therefore, the main biological processes and pathways that may increase the tumorigenic potential of the MCF7 cells in JTB downregulated condition are related to the actin cytoskeleton organization, EMT, mitotic cell cycle, glycolysis and fatty acid metabolism, inflammatory response and macrophage activation, chemotaxis and migration, cellular response to stress condition (oxidative stress and hypoxia), transcription control, histone modification and ion transport.
Collapse
Affiliation(s)
- Madhuri Jayathirtha
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Anca-Narcisa Neagu
- Laboratory of Animal Histology, Faculty of Biology, “Alexandru Ioan Cuza” University of IasiCarol I bvd. No. 22, Iasi 700505, Romania
| | - Danielle Whitham
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Shelby Alwine
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| | - Costel C Darie
- Biochemistry & Proteomics Group, Department of Chemistry and Biomolecular Science, Clarkson UniversityPotsdam, NY 13699-5810, USA
| |
Collapse
|
23
|
Engevik MA, Engevik AC. Myosins and membrane trafficking in intestinal brush border assembly. Curr Opin Cell Biol 2022; 77:102117. [PMID: 35870341 DOI: 10.1016/j.ceb.2022.102117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 11/29/2022]
Abstract
Myosins are a class of motors that participate in a wide variety of cellular functions including organelle transport, cell adhesion, endocytosis and exocytosis, movement of RNA, and cell motility. Among the emerging roles for myosins is regulation of the assembly, morphology, and function of actin protrusions such as microvilli. The intestine harbors an elaborate apical membrane composed of highly organized microvilli. Microvilli assembly and function are intricately tied to several myosins including Myosin 1a, non-muscle Myosin 2c, Myosin 5b, Myosin 6, and Myosin 7b. Here, we review the research progress made in our understanding of myosin mediated apical assembly.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina
| | - Amy C Engevik
- Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina.
| |
Collapse
|
24
|
A functional role of S100A4/non-muscle myosin IIA axis for pro-tumorigenic vascular functions in glioblastoma. Cell Commun Signal 2022; 20:46. [PMID: 35392912 PMCID: PMC8991692 DOI: 10.1186/s12964-022-00848-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/16/2022] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Glioblastoma (GBM) is the most aggressive form of brain tumor and has vascular-rich features. The S100A4/non-muscle myosin IIA (NMIIA) axis contributes to aggressive phenotypes in a variety of human malignancies, but little is known about its involvement in GBM tumorigenesis. Herein, we examined the role of the S100A4/NMIIA axis during tumor progression and vasculogenesis in GBM. METHODS We performed immunohistochemistry for S100A4, NMIIA, and two hypoxic markers, hypoxia-inducible factor-1α (HIF-1α) and carbonic anhydrase 9 (CA9), in samples from 94 GBM cases. The functional impact of S100A4 knockdown and hypoxia were also assessed using a GBM cell line. RESULTS In clinical GBM samples, overexpression of S100A4 and NMIIA was observed in both non-pseudopalisading (Ps) and Ps (-associated) perinecrotic lesions, consistent with stabilization of HIF-1α and CA9. CD34(+) microvascular densities (MVDs) and the interaction of S100A4 and NMIIA were significantly higher in non-Ps perinecrotic lesions compared to those in Ps perinecrotic areas. In non-Ps perinecrotic lesions, S100A4(+)/HIF-1α(-) GBM cells were recruited to the surface of preexisting host vessels in the vascular-rich areas. Elevated vascular endothelial growth factor A (VEGFA) mRNA expression was found in S100A4(+)/HIF-1α(+) GBM cells adjacent to the vascular-rich areas. In addition, GBM patients with high S100A4 protein expression had significantly worse OS and PFS than did patients with low S100A4 expression. Knockdown of S100A4 in the GBM cell line KS-1 decreased migration capability, concomitant with decreased Slug expression; the opposite effects were elicited by blebbistatin-dependent inhibition of NMIIA. CONCLUSION S100A4(+)/HIF-1α(-) GBM cells are recruited to (and migrate along) preexisting vessels through inhibition of NMIIA activity. This is likely stimulated by extracellular VEGF that is released by S100A4(+)/HIF-1α(+) tumor cells in non-Ps perinecrotic lesions. In turn, these events engender tumor progression via acceleration of pro-tumorigenic vascular functions. Video abstract.
Collapse
|
25
|
Detection of Myosin 1g Overexpression in Pediatric Leukemia by Novel Monoclonal Antibodies. Int J Mol Sci 2022; 23:ijms23073912. [PMID: 35409272 PMCID: PMC8999415 DOI: 10.3390/ijms23073912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 02/23/2022] [Accepted: 03/01/2022] [Indexed: 01/27/2023] Open
Abstract
Myosin 1g (Myo1g) is a mechanoenzyme associated with actin filaments, expressed exclusively in hematopoietic cells, and involved in various cellular functions, including cell migration, adhesion, and membrane trafficking. Despite the importance of Myo1g in distinct functions, there is currently no monoclonal antibody (mAb) against Myo1g. mAbs are helpful tools for the detection of specific antigens in tumor cells and other tissues. The development of mAbs against targeted dysregulated molecules in cancer cells remains a crucial tool for aiding in the diagnosis and the treatment of patients. Using hybridoma technology, we generated a panel of hybridomas specific for Myo1g. ELISA, immunofluorescence, and Western blot assay results revealed the recognition of Myo1g by these novel monoclonal antibodies in normal and transformed T and B cells. Here, we report the development and application of new monoclonal antibodies against Myo1g for their potential use to detect its overexpression in acute lymphoblastic leukemia (ALL) patients.
Collapse
|
26
|
Deng S, Leong HC, Datta A, Gopal V, Kumar AP, Yap CT. PI3K/AKT Signaling Tips the Balance of Cytoskeletal Forces for Cancer Progression. Cancers (Basel) 2022; 14:1652. [PMID: 35406424 PMCID: PMC8997157 DOI: 10.3390/cancers14071652] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/13/2022] [Accepted: 03/21/2022] [Indexed: 02/01/2023] Open
Abstract
The PI3K/AKT signaling pathway plays essential roles in multiple cellular processes, which include cell growth, survival, metabolism, and motility. In response to internal and external stimuli, the PI3K/AKT signaling pathway co-opts other signaling pathways, cellular components, and cytoskeletal proteins to reshape individual cells. The cytoskeletal network comprises three main components, which are namely the microfilaments, microtubules, and intermediate filaments. Collectively, they are essential for many fundamental structures and cellular processes. In cancer, aberrant activation of the PI3K/AKT signaling cascade and alteration of cytoskeletal structures have been observed to be highly prevalent, and eventually contribute to many cancer hallmarks. Due to their critical roles in tumor progression, pharmacological agents targeting PI3K/AKT, along with cytoskeletal components, have been developed for better intervention strategies against cancer. In our review, we first discuss existing evidence in-depth and then build on recent advances to propose new directions for therapeutic intervention.
Collapse
Affiliation(s)
- Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
| | - Hin Chong Leong
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| | - Arpita Datta
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
| | - Vennila Gopal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
| | - Alan Prem Kumar
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore;
- Departments of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (S.D.); (V.G.)
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore;
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
27
|
Deng Y, Liu L, Feng W, Lin Z, Ning Y, Luo X. High Expression of MYL9 Indicates Poor Clinical Prognosis of Epithelial Ovarian Cancer. Recent Pat Anticancer Drug Discov 2021; 16:533-539. [PMID: 34551701 DOI: 10.2174/1574891x16666210706153740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 02/06/2021] [Accepted: 02/27/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND The prognosis of Epithelial Ovarian Cancer (EOC) is poor, but the prognostic biomarkers are neither sensitive nor specific. Therefore, it is very important to search novel prognostic biomarkers for EOC. OBJECTIVES The present study aimed to investigate Myosin Light Chain 9(MYL9) expression in Epithelial Ovarian Cancer (EOC) tissues (including paraffin-embedded and fresh tissue samples) and its relationship with clinicopathological characteristics, as well as its potential prognostic value in patients with EOC. METHODS Between March 2009 and December 2018, all of 184 paraffin-embedded cancer tissues from patients with EOC and 41 paratumor tissues, pathologically confirmed at the Memorial Hospital of Sun Yat-sen University and Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, were collected for the present study and were assessed for MYL9 protein expression patterns using Immunohistochemistry (IHC). Furthermore, from August 2013 to November 2019, 16 fresh EOC tissues and their paired paratumor tissues, pathologically confirmed at the Integrated Hospital of Traditional Chinese Medicine, Southern Medical University were analyzed using Reverse-Transcription Quantitative PCR (RT-qPCR) to detect MYL9 mRNA expression levels. RESULTS The results showed that MYL9 expression was higher in cancer tissues compared with that in paratumor tissues, and MYL9 overexpression was associated with shorter Recurrence Free Survival (RFS) and Overall Survival (OS) of EOC patients. Furthermore, multivariate Cox model analysis indicated that MYL9 overexpression was an independent poor survival prediction in patients with EOC. CONCLUSION MYL9 is upregulated in EOC and may serve as a useful patent of prognostic biomarker in EOC, and it may demonstrate an important value for the clinical treatment and supervision of patients with EOC.
Collapse
Affiliation(s)
- Yuao Deng
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Longyang Liu
- Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 13 Shiliugang ST, Guangzhou, 510315, China
| | - Weifeng Feng
- The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Zhongqiu Lin
- Department of Gynecology Oncology, The Memorial Hospital of Sun Yat-sen University, Guangzhou, 510120, China
| | - Yingxia Ning
- Department of Gynaecology and Obstetrics, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Xin Luo
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| |
Collapse
|
28
|
Estrada-Abreo LA, Rodríguez-Cruz L, Garfias-Gómez Y, Araujo-Cardenas JE, Antonio-Andrés G, Salgado-Aguayo AR, Orozco-Ruiz D, Torres-Nava JR, Díaz-Valencia JD, Huerta-Yépez S, Patiño-López G. High expression of Myosin 1g in pediatric acute lymphoblastic leukemia. Oncotarget 2021; 12:1937-1945. [PMID: 34548909 PMCID: PMC8448507 DOI: 10.18632/oncotarget.28055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 08/13/2021] [Indexed: 12/29/2022] Open
Abstract
Acute Lymphoblastic Leukemia (ALL) is the most frequent cancer in pediatric population. Although the treatment has improved and almost 85% of the children are cured about 20% suffer relapse, therefore finding molecules that participate in the pathogenesis of the disease for the identification of relapse and patients at risk is an urgent unmet need. Class I myosins are molecular motors involved in membrane tension, endocytosis, phagocytosis and cell migration and recently they have been shown important for development and aggressiveness of diverse cancer types, however Myo1g an hematopoietic specific myosin has not been studied in cancer so far. We evaluated the expression of Myo1g by qRT-PCR, Immunocytochemistry and Immunofluorescence in a cohort of 133 ALL patients and correlated the expression at diagnosis and after treatment with clinical features and treatment outcomes. We found high expression levels of Myo1g in Peripheral Blood Mononuclear Cells (PBMCs) from patients with ALL at diagnosis and those levels decreased after complete remission; furthermore, we found an increase in Myo1g expression on patients with 9:22 translocation and those who relapse. This study show that Myo1g is over expressed in ALL and that may participate in the pathogenesis of the disease specially in high-risk patients.
Collapse
Affiliation(s)
- Laura A Estrada-Abreo
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México.,Cell Biology and Flow Cytometry Laboratory, Department of Health Sciences, Universidad Autónoma Metropolitana, Iztapalapa, México
| | - Leonor Rodríguez-Cruz
- Cell Biology and Flow Cytometry Laboratory, Department of Health Sciences, Universidad Autónoma Metropolitana, Iztapalapa, México
| | - Yanelly Garfias-Gómez
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México
| | - Janeth E Araujo-Cardenas
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México
| | - Gabriela Antonio-Andrés
- Oncologic Diseases Research Unit, Hospital Infantil de México Federico Gómez, México City, México
| | - Alfonso R Salgado-Aguayo
- Laboratory of Research on Rheumatic Diseases, National Institute of Respiratory Diseases, Ismael Cosío Villegas, México City, México
| | | | | | - Juan D Díaz-Valencia
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México
| | - Sara Huerta-Yépez
- Oncologic Diseases Research Unit, Hospital Infantil de México Federico Gómez, México City, México
| | - Genaro Patiño-López
- Immunology and Proteomics Laboratory, Hospital Infantil de México Federico Gómez, México City, México
| |
Collapse
|
29
|
Ma J, Ma Y, Chen S, Guo S, Hu J, Yue T, Zhang J, Zhu J, Wang P, Chen G, Liu Y. SPARC enhances 5-FU chemosensitivity in gastric cancer by modulating epithelial-mesenchymal transition and apoptosis. Biochem Biophys Res Commun 2021; 558:134-140. [PMID: 33910127 DOI: 10.1016/j.bbrc.2021.04.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/02/2021] [Indexed: 12/27/2022]
Abstract
Previous studies have shown that secreted protein acidic and rich in cysteine (SPARC) proteins can inhibit the development of cancer cells in various ways, such as by inhibiting angiogenesis and inhibiting cell proliferation. In fact, SPARC proteins may have an effect on the chemoresistance of gastric cancer cells to 5-Fluorouracil (5-FU), which needs further research in the future. Therefore, the purpose of this study was to explore the relationship between SPARC proteins and the chemosensitivity of gastric cancer cells to 5-FU. In vitro, after SPARC protein levels were regulated by plasmid, siRNA and human recombinant SPARC protein transfection in MGC-803, SGC-7901 and BGC-823 cells, we detected epithelial-mesenchymal transition (EMT), apoptosis markers and cell viability after 5-FU treatment. In vivo, we implanted BGC-823 cells with stable SPARC overexpression into nude mice. Tumour size was measured to assess the effect of SPARC protein on tumour formation and 5-FU chemosensitivity. In SGC-7901 and BGC-823 cells, both endogenous and exogenous upregulation of SPARC protein levels decreased cell viability, destroyed cytoskeletal F-actin, inhibited cell migration, and downregulated a series of transcription factors to inhibit cell EMT; it also upregulated cell apoptosis-related proteins to promote cell apoptosis. However, we obtained opposite results in SPARC knockdown MGC-803 cells. In vivo, compared with the control group, the group engrafted with BGC-823 cells stably overexpressing SPARC had a significant smaller tumour size. After 5-FU treatment, the new tumour gradually decreased in size. Our results show that the SPARC protein could enhance 5-FU chemosensitivity in gastric cancer cell lines by inhibiting EMT and promoting cell apoptosis.
Collapse
Affiliation(s)
- Ju Ma
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Yongchen Ma
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Shanwen Chen
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Shihao Guo
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Jianwen Hu
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Taohua Yue
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Junling Zhang
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Jing Zhu
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Pengyuan Wang
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China
| | - Guowei Chen
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China.
| | - Yucun Liu
- Division of General Surgery, Peking University First Hospital, Peking University, Beijing, PR China.
| |
Collapse
|
30
|
Datta A, Deng S, Gopal V, Yap KCH, Halim CE, Lye ML, Ong MS, Tan TZ, Sethi G, Hooi SC, Kumar AP, Yap CT. Cytoskeletal Dynamics in Epithelial-Mesenchymal Transition: Insights into Therapeutic Targets for Cancer Metastasis. Cancers (Basel) 2021; 13:1882. [PMID: 33919917 PMCID: PMC8070945 DOI: 10.3390/cancers13081882] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 12/14/2022] Open
Abstract
In cancer cells, a vital cellular process during metastasis is the transformation of epithelial cells towards motile mesenchymal cells called the epithelial to mesenchymal transition (EMT). The cytoskeleton is an active network of three intracellular filaments: actin cytoskeleton, microtubules, and intermediate filaments. These filaments play a central role in the structural design and cell behavior and are necessary for EMT. During EMT, epithelial cells undergo a cellular transformation as manifested by cell elongation, migration, and invasion, coordinated by actin cytoskeleton reorganization. The actin cytoskeleton is an extremely dynamic structure, controlled by a balance of assembly and disassembly of actin filaments. Actin-binding proteins regulate the process of actin polymerization and depolymerization. Microtubule reorganization also plays an important role in cell migration and polarization. Intermediate filaments are rearranged, switching to a vimentin-rich network, and this protein is used as a marker for a mesenchymal cell. Hence, targeting EMT by regulating the activities of their key components may be a potential solution to metastasis. This review summarizes the research done on the physiological functions of the cytoskeleton, its role in the EMT process, and its effect on multidrug-resistant (MDR) cancer cells-highlight some future perspectives in cancer therapy by targeting cytoskeleton.
Collapse
Affiliation(s)
- Arpita Datta
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Shuo Deng
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Vennila Gopal
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Kenneth Chun-Hong Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
| | - Clarissa Esmeralda Halim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Mun Leng Lye
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Mei Shan Ong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117593, Singapore;
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Shing Chuan Hooi
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore;
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117593, Singapore;
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Celestial T. Yap
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore; (A.D.); (S.D.); (V.G.); (K.C.-H.Y.); (C.E.H.); (M.L.L.); (M.S.O.); (S.C.H.)
- Cancer Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117593, Singapore
- National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| |
Collapse
|
31
|
Gomig THB, Gontarski AM, Cavalli IJ, Souza RLRD, Lucena ACR, Batista M, Machado KC, Marchini FK, Marchi FA, Lima RS, Urban CDA, Marchi RD, Cavalli LR, Ribeiro EMDSF. Integrated analysis of label-free quantitative proteomics and bioinformatics reveal insights into signaling pathways in male breast cancer. Genet Mol Biol 2021; 44:e20190410. [PMID: 33656060 PMCID: PMC7926483 DOI: 10.1590/1678-4685-gmb-2019-0410] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 01/18/2021] [Indexed: 01/04/2023] Open
Abstract
Male breast cancer (MBC) is a rare malignancy that accounts for about 1.8% of all breast cancer cases. In contrast to the high number of the “omics” studies in breast cancer in women, only recently molecular approaches have been performed in MBC research. High-throughput proteomics based methodologies are promisor strategies to characterize the MBC proteomic signatures and their association with clinico-pathological parameters. In this study, the label-free quantification-mass spectrometry and bioinformatics approaches were applied to analyze the proteomic profiling of a MBC case using the primary breast tumor and the corresponding axillary metastatic lymph nodes and adjacent non-tumor breast tissues. The differentially expressed proteins were identified in the signaling pathways of granzyme B, sirtuins, eIF2, actin cytoskeleton, eNOS, acute phase response and calcium and were connected to the upstream regulators MYC, PI3K SMARCA4 and cancer-related chemical drugs. An additional proteomic comparative analysis was performed with a primary breast tumor of a female patient and revealed an interesting set of proteins, which were mainly involved in cancer biology. Together, our data provide a relevant data source for the MBC research that can help the therapeutic strategies for its management.
Collapse
Affiliation(s)
| | - Amanda Moletta Gontarski
- Universidade Federal do Paraná, Departamento de Genética, Programa de Pós-graduação em Genética, Curitiba, PR, Brazil
| | - Iglenir João Cavalli
- Universidade Federal do Paraná, Departamento de Genética, Programa de Pós-graduação em Genética, Curitiba, PR, Brazil
| | | | | | - Michel Batista
- Instituto Carlos Chagas, Laboratório de Genômica Funcional, Curitiba, PR, Brazil.,Fundação Oswaldo Cruz (Fiocruz), Plataforma de Espectrometria de Massas, Curitiba, PR, Brazil
| | | | - Fabricio Klerynton Marchini
- Instituto Carlos Chagas, Laboratório de Genômica Funcional, Curitiba, PR, Brazil.,Fundação Oswaldo Cruz (Fiocruz), Plataforma de Espectrometria de Massas, Curitiba, PR, Brazil
| | | | - Rubens Silveira Lima
- Hospital Nossa Senhora das Graças, Centro de Doenças da Mama, Curitiba, PR, Brazil
| | | | | | - Luciane Regina Cavalli
- Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil.,Georgetown University, Lombardi Comprehensive Cancer Center, Washington, USA
| | | |
Collapse
|
32
|
Naydenov NG, Lechuga S, Huang EH, Ivanov AI. Myosin Motors: Novel Regulators and Therapeutic Targets in Colorectal Cancer. Cancers (Basel) 2021; 13:741. [PMID: 33670106 PMCID: PMC7916823 DOI: 10.3390/cancers13040741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022] Open
Abstract
Colorectal cancer (CRC) remains the third most common cause of cancer and the second most common cause of cancer deaths worldwide. Clinicians are largely faced with advanced and metastatic disease for which few interventions are available. One poorly understood aspect of CRC involves altered organization of the actin cytoskeleton, especially at the metastatic stage of the disease. Myosin motors are crucial regulators of actin cytoskeletal architecture and remodeling. They act as mechanosensors of the tumor environments and control key cellular processes linked to oncogenesis, including cell division, extracellular matrix adhesion and tissue invasion. Different myosins play either oncogenic or tumor suppressor roles in breast, lung and prostate cancer; however, little is known about their functions in CRC. This review focuses on the functional roles of myosins in colon cancer development. We discuss the most studied class of myosins, class II (conventional) myosins, as well as several classes (I, V, VI, X and XVIII) of unconventional myosins that have been linked to CRC development. Altered expression and mutations of these motors in clinical tumor samples and their roles in CRC growth and metastasis are described. We also evaluate the potential of using small molecular modulators of myosin activity to develop novel anticancer therapies.
Collapse
Affiliation(s)
- Nayden G. Naydenov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| | - Susana Lechuga
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| | - Emina H. Huang
- Departments of Cancer Biology and Colorectal Surgery, Cleveland Clinic Foundation, Cleveland, OH 44195, USA;
| | - Andrei I. Ivanov
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; (N.G.N.); (S.L.)
| |
Collapse
|
33
|
Balestrieri K, Kew K, McDaniel M, Ramez M, Pittman HK, Murray G, Vohra NA, Verbanac KM. Proteomic identification of tumor- and metastasis-associated galectin-1 in claudin-low breast cancer. Biochim Biophys Acta Gen Subj 2021; 1865:129784. [PMID: 33166603 DOI: 10.1016/j.bbagen.2020.129784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 11/01/2020] [Accepted: 11/04/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Metastasis and mortality remain high among breast cancer patients with the claudin-low subtype because these tumors are aggressive, chemoresistant, and lack targeted therapies. Our objective was to utilize discovery-based proteomics to identify proteins associated with claudin-low primary and metastatic tumors to gain insight into pathways and mechanisms of tumor progression. METHODS We used nano-LC-MS/MS proteomics to analyze orthotopic and metastatic tumors from the syngeneic murine T11 tumor model, which displays gene expression profiles mirroring human claudin-low tumors. Galectin-1 identity, expression and spatial distribution were investigated by biochemical and immunochemical methods and MALDI/IMS. RNA seq data from mouse and human tumors in our study and publicly available microarray data were analyzed for differential galectin-1 expression across breast cancer subtypes. RESULTS Galectin-1, an N-acetyllactosamine-binding protein, exhibited the highest sequence coverage and high abundance rank order among nano-LC-MS/MS-identified proteins shared by T11 claudin-low tumors but not normal tissue. Label-free quantitation, Western immunoblot and ELISA confirmed galectin-1 identity and significant differential expression. MALDI/IMS spatial mapping and immunohistochemistry detected galectin-1 in T11 metastatic lung foci. Immunohistochemistry of human claudin-low tumors demonstrated intermediate-to-high intensity galectin-1 staining of tumor and stroma. Gene expression analysis of mouse and human tumors found the highest galectin-1 levels in the claudin-low breast cancer subtype. CONCLUSIONS Proteomics and genomics reveal high expression of galectin-1 protein and RNA in primary and metastatic claudin-low breast cancer. GENERAL SIGNIFICANCE This work endorses proteomic approaches in cancer research and supports further investigations of the function and significance of galectin-1 overexpression in claudin-low tumor progression.
Collapse
Affiliation(s)
- Kassondra Balestrieri
- Brody School of Medicine, East Carolina University, Department of Surgery, 600 Moye Boulevard, Greenville, NC 27834, United States of America
| | - Kimberly Kew
- Brody School of Medicine, East Carolina University, Department of Biochemistry and Molecular Biology, 600 Moye Boulevard, Greenville, NC 27834, United States of America
| | - Moses McDaniel
- Brody School of Medicine, East Carolina University, Department of Surgery, 600 Moye Boulevard, Greenville, NC 27834, United States of America
| | - Mohamed Ramez
- Brody School of Medicine, East Carolina University, Department of Surgery, 600 Moye Boulevard, Greenville, NC 27834, United States of America
| | - H Keith Pittman
- Brody School of Medicine, East Carolina University, Department of Surgery, 600 Moye Boulevard, Greenville, NC 27834, United States of America
| | - Gina Murray
- Brody School of Medicine, East Carolina University, Department of Pathology, 600 Moye Boulevard, Greenville, NC 27834, United States of America
| | - Nasreen A Vohra
- Brody School of Medicine, East Carolina University, Department of Surgery, 600 Moye Boulevard, Greenville, NC 27834, United States of America
| | - Kathryn M Verbanac
- Brody School of Medicine, East Carolina University, Department of Surgery, 600 Moye Boulevard, Greenville, NC 27834, United States of America.
| |
Collapse
|
34
|
Giwa A, Fatai A, Gamieldien J, Christoffels A, Bendou H. Identification of novel prognostic markers of survival time in high-risk neuroblastoma using gene expression profiles. Oncotarget 2020; 11:4293-4305. [PMID: 33245713 PMCID: PMC7679032 DOI: 10.18632/oncotarget.27808] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 10/27/2020] [Indexed: 12/11/2022] Open
Abstract
Neuroblastoma is the most common extracranial solid tumor in childhood. Patients in high-risk group often have poor outcomes with low survival rates despite several treatment options. This study aimed to identify a genetic signature from gene expression profiles that can serve as prognostic indicators of survival time in patients of high-risk neuroblastoma, and that could be potential therapeutic targets. RNA-seq count data was downloaded from UCSC Xena browser and samples grouped into Short Survival (SS) and Long Survival (LS) groups. Differential gene expression (DGE) analysis, enrichment analyses, regulatory network analysis and machine learning (ML) prediction of survival group were performed. Forty differentially expressed genes (DEGs) were identified including genes involved in molecular function activities essential for tumor proliferation. DEGs used as features for prediction of survival groups included EVX2, NHLH2, PRSS12, POU6F2, HOXD10, MAPK15, RTL1, LGR5, CYP17A1, OR10AB1P, MYH14, LRRTM3, GRIN3A, HS3ST5, CRYAB and NXPH3. An accuracy score of 82% was obtained by the ML classification models. SMIM28 was revealed to possibly have a role in tumor proliferation and aggressiveness. Our results indicate that these DEGs can serve as prognostic indicators of survival in high-risk neuroblastoma patients and will assist clinicians in making better therapeutic and patient management decisions.
Collapse
Affiliation(s)
- Abdulazeez Giwa
- SAMRC Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Azeez Fatai
- Department of Biochemistry, Lagos State University, Lagos, Nigeria
| | - Junaid Gamieldien
- SAMRC Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Alan Christoffels
- SAMRC Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Hocine Bendou
- SAMRC Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| |
Collapse
|
35
|
Park H, Kim D, Kim D, Park J, Koh Y, Yoon SS. Truncation of MYH8 tail in AML: a novel prognostic marker with increase cell migration and epithelial-mesenchymal transition utilizing RAF/MAPK pathway. Carcinogenesis 2020; 41:817-827. [PMID: 31430364 DOI: 10.1093/carcin/bgz146] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 07/29/2019] [Accepted: 08/18/2019] [Indexed: 12/20/2022] Open
Abstract
MYH8 is an actin-based motor protin involved in integrin-mediated cell adhesion and migration. Heretofore, the association of MYH8 mutation and cancer is unclear. In this study, we investigated the biologic significance of novel MYH8 tail truncation mutation, R1292X, in acute myeloid leukemia (AML) which was discovered by whole-exome sequencing and targeted re-sequencing of 209 AML patients. The patients harboring the mutation all relapsed within 3.8-20.9 months. To explore the functional consequence of the mutation in AML progress, we established knock-in cell lines using CRISPR-Cas9 genome editing. Using the established mutant model, we assessed traits of cancer progress. The mutant cells had improved motility, which was confirmed by immunofluorescence staining, wound healing, transwell migration and adhesion assay. The cell morphology and cell cycle were altered to be accessible to migration and epithelial-to-mesenchymal transition (EMT) transcription factors were also increased. The Raf and p44/42 MAPK pathway was a major regulator of these characteristics proved by a screening of signal transduction and inhibitor assay. Further, a public cancer genome database (cBioPortal) shows that MYH8 tail truncation mutations occurring near the R1292 position of the genome may have a significant function in cancer. In conclusion, truncation of MYH8 could be a novel prognostic marker related to poor prognosis by inducing cell migration and EMT features, and inhibition of the Raf/MAPK pathway would be a therapeutic strategy for AML patients with MYH8 tail truncation.
Collapse
Affiliation(s)
- Hyejoo Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Daeyoon Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Dongchan Kim
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Jihyun Park
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Youngil Koh
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital, 101, Daehak-ro, Jongno-gu, Seoul 03080, Republic of Korea
| |
Collapse
|
36
|
Abstract
Targeted cancer therapy aims to achieve specific elimination of cancerous but not normal cells. Recently, PIWI proteins, a subfamily of the PAZ-PIWI domain (PPD) protein family, have emerged as promising candidates for targeted cancer therapy. PPD proteins are essential for small noncoding RNA pathways. The Argonaute subfamily partners with microRNA and small interfering RNA, whereas the PIWI subfamily partners with PIWI-interacting RNA (piRNA). Both PIWI proteins and piRNA are mostly expressed in the germline and best known for their function in transposon silencing, with no detectable function in mammalian somatic tissues. However, PIWI proteins become aberrantly expressed in multiple types of somatic cancers, thus gaining interest in targeted therapy. Despite this, little is known about the regulatory mechanism of PIWI proteins in cancer. Here we report that one of the four PIWI proteins in humans, PIWIL1, is highly expressed in gastric cancer tissues and cell lines. Knocking out the PIWIL1 gene (PIWIL1-KO) drastically reduces gastric cancer cell proliferation, migration, metastasis, and tumorigenesis. RNA deep sequencing of gastric cancer cell line SNU-1 reveals that KO significantly changes the transcriptome, causing the up-regulation of most of its associated transcripts. Surprisingly, few bona fide piRNAs exist in gastric cancer cells. Furthermore, abolishing the piRNA-binding activity of PIWIL1 does not affect its oncogenic function. Thus, PIWIL1 function in gastric cancer cells is independent of piRNA. This piRNA-independent regulation involves interaction with the UPF1-mediated nonsense-mediated mRNA decay (NMD) mechanism. Altogether, our findings reveal a piRNA-independent function of PIWIL1 in promoting gastric cancer.
Collapse
|
37
|
Hiruta A, Oguri Y, Yokoi A, Matsumoto T, Oda Y, Tomohiro M, Hashimura M, Jiang Z, Tochimoto M, Nakagawa M, Saegusa M. S100A4/Nonmuscle Myosin IIA/p53 Axis Contributes to Aggressive Features in Ovarian High-Grade Serous Carcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:2304-2316. [PMID: 32805233 DOI: 10.1016/j.ajpath.2020.07.014] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Revised: 07/02/2020] [Accepted: 07/21/2020] [Indexed: 12/22/2022]
Abstract
S100A4 is a small calcium-binding protein that exerts its biological functions by interacting with nonmuscle myosin IIA (NMIIA) and p53. Although S100A4 promotes metastasis in several tumors, little is known about its involvement in the progression of ovarian high-grade serous carcinomas (HGSCs). Herein, we focused on functional roles of the S100A4/NMIIA/p53 axis in these tumors. In HGSC cell lines harboring mutant p53, knockdown (KD) of S100A4 reduced the expression of several epithelial-mesenchymal transition/cancer stem cell markers and the ALDH1high population, consistent with an inhibition of stemness features. S100A4-KD also increased apoptosis, decreased cell proliferation, and accelerated cell mobility. This was accompanied by increased Snail expression, which, in turn, was likely due to loss of p53 function. In contrast, specific inhibition of NMIIA by blebbistatin induced phenotypes that-with the exception of cell proliferation and mobility-were opposite to those observed in S100A4-KD cells. In clinical samples, cytoplasmic and/or nuclear interactions between S100A4, NMIIA, and mutant p53 were observed. In addition, high expression of S100A4, but not NMIIA or p53, was a significant and independent unfavorable prognostic factor in HGSC patients. These findings suggest that, via its interaction with NMIIA and p53, overexpressed S100A4 may induce epithelial-mesenchymal transition/cancer stem cell properties in HGSC and elicit several other tumor-associated phenotypes.
Collapse
Affiliation(s)
- Ai Hiruta
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yasuko Oguri
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Ako Yokoi
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Toshihide Matsumoto
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yusuke Oda
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Mikihisa Tomohiro
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Miki Hashimura
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Zesong Jiang
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Masataka Tochimoto
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Mayu Nakagawa
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Makoto Saegusa
- Department of Pathology, Kitasato University School of Medicine, Sagamihara, Japan.
| |
Collapse
|
38
|
The Expressions and Mechanisms of Sarcomeric Proteins in Cancers. DISEASE MARKERS 2020; 2020:8885286. [PMID: 32670437 PMCID: PMC7346232 DOI: 10.1155/2020/8885286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/07/2020] [Accepted: 06/13/2020] [Indexed: 02/07/2023]
Abstract
The sarcomeric proteins control the movement of cells in diverse species, whereas the deregulation can induce tumours in model organisms and occurs in human carcinomas. Sarcomeric proteins are recognized as oncogene and related to tumor cell metastasis. Recent insights into their expressions and functions have led to new cancer therapeutic opportunities. In this review, we appraise the evidence for the sarcomeric proteins as cancer genes and discuss cancer-relevant biological functions, potential mechanisms by which sarcomeric proteins activity is altered in cancer.
Collapse
|
39
|
Uray K, Major E, Lontay B. MicroRNA Regulatory Pathways in the Control of the Actin-Myosin Cytoskeleton. Cells 2020; 9:E1649. [PMID: 32660059 PMCID: PMC7408560 DOI: 10.3390/cells9071649] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022] Open
Abstract
MicroRNAs (miRNAs) are key modulators of post-transcriptional gene regulation in a plethora of processes, including actin-myosin cytoskeleton dynamics. Recent evidence points to the widespread effects of miRNAs on actin-myosin cytoskeleton dynamics, either directly on the expression of actin and myosin genes or indirectly on the diverse signaling cascades modulating cytoskeletal arrangement. Furthermore, studies from various human models indicate that miRNAs contribute to the development of various human disorders. The potentially huge impact of miRNA-based mechanisms on cytoskeletal elements is just starting to be recognized. In this review, we summarize recent knowledge about the importance of microRNA modulation of the actin-myosin cytoskeleton affecting physiological processes, including cardiovascular function, hematopoiesis, podocyte physiology, and osteogenesis.
Collapse
Affiliation(s)
- Karen Uray
- Correspondence: (K.U.); (B.L.); Tel.: +36-52-412345 (K.U. & B.L.)
| | | | - Beata Lontay
- Correspondence: (K.U.); (B.L.); Tel.: +36-52-412345 (K.U. & B.L.)
| |
Collapse
|
40
|
Tomić TT, Olausson J, Rehammar A, Deland L, Muth A, Ejeskär K, Nilsson S, Kristiansson E, Wassén ON, Abel F. MYO5B mutations in pheochromocytoma/paraganglioma promote cancer progression. PLoS Genet 2020; 16:e1008803. [PMID: 32511227 PMCID: PMC7329139 DOI: 10.1371/journal.pgen.1008803] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 07/01/2020] [Accepted: 04/27/2020] [Indexed: 12/20/2022] Open
Abstract
Identification of additional cancer-associated genes and secondary mutations driving the metastatic progression in pheochromocytoma and paraganglioma (PPGL) is important for subtyping, and may provide optimization of therapeutic regimens. We recently reported novel recurrent nonsynonymous mutations in the MYO5B gene in metastatic PPGL. Here, we explored the functional impact of these MYO5B mutations, and analyzed MYO5B expression in primary PPGL tumor cases in relation to mutation status. Immunohistochemistry and mRNA expression analysis in 30 PPGL tumors revealed an increased MYO5B expression in metastatic compared to non-metastatic cases. In addition, subcellular localization of MYO5B protein was altered from cytoplasmic to membranous in some metastatic tumors, and the strongest and most abnormal expression pattern was observed in a paraganglioma harboring a somatic MYO5B:p.G1611S mutation. In addition to five previously discovered MYO5B mutations, the present study of 30 PPGL (8 previous and 22 new samples) also revealed two, and hence recurrent, mutations in the gene paralog MYO5A. The three MYO5B missense mutations with the highest prediction scores (p.L587P, p.G1611S and p.R1641C) were selected and functionally validated using site directed mutagenesis and stable transfection into human neuroblastoma cells (SK-N-AS) and embryonic kidney cells (HEK293). In vitro analysis showed a significant increased proliferation rate in all three MYO5B mutated clones. The two somatically derived mutations, p.L587P and p.G1611S, were also found to increase the migration rate. Expression analysis of MYO5B mutants compared to wild type clones, demonstrated a significant enrichment of genes involved in migration, proliferation, cell adhesion, glucose metabolism, and cellular homeostasis. Our study validates the functional role of novel MYO5B mutations in proliferation and migration, and suggest the MYO5-pathway to be involved in the malignant progression in some PPGL tumors. Up to 25% of pheochromocytoma/paraganglioma (PPGL) cases develop metastatic disease with poor outcome and few treatment options. The disease mechanism is not fully understood, and to date there are no reliable markers to predict malignancy. We have recently discovered novel missense mutations in the non-conventional myosin 5 gene (MYO5B), an endosomal transport protein, which we now show enhances progression and migration in PPGLs. MYO5B mutations were preferentially found in patients with metastatic disease and SDH deficiency (germline SDHB-mutations). Abolished SDH activity result in a metabolic switch to aerobic glycolysis requiring increased glucose consumption. Since the MYO5B mutations were found to drive progression through downstream up-regulation of glucose metabolism genes, e.g. glucagon, we hypothesize that these mutations may fuel the pseudohypoxic state by altering glucose uptake in cancer cells. Our result is the first to link the myosin 5 genes to PPGL tumorigenesis. Further, it shows that the tumor progression route in PPGL is complex, with contribution from several genetic factors. An increasing number of studies show dysregulation and importance of the MYO5-proteins in cancer, but little is still known about the precise role and mechanism of mutations, hence more research in this area is needed.
Collapse
Affiliation(s)
- Tajana Tešan Tomić
- Department of Pathology and Genetics, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Josefin Olausson
- Department of Pathology and Genetics, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Anna Rehammar
- Department of Mathematical Sciences, Chalmers University of Technology and Biostatistics, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Lily Deland
- Department of Pathology and Genetics, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Andreas Muth
- Department of Surgery, Institute of Clinical Science, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Region Västra Götaland, Sahlgrenska University Hospital, Department of Surgery, Section of endocrine and sarcoma surgery, Gothenborg, Sweden
| | - Katarina Ejeskär
- School of Health and Education, University of Skövde, Skövde, Sweden
| | - Staffan Nilsson
- Department of Pathology and Genetics, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden.,Department of Mathematical Sciences, Chalmers University of Technology and Biostatistics, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Erik Kristiansson
- Department of Mathematical Sciences, Chalmers University of Technology and Biostatistics, School of Public Health and Community Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ola Nilsson Wassén
- Sahlgrenska Cancer Center, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Frida Abel
- Department of Pathology and Genetics, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
41
|
Cook AW, Gough RE, Toseland CP. Nuclear myosins - roles for molecular transporters and anchors. J Cell Sci 2020; 133:133/11/jcs242420. [PMID: 32499319 DOI: 10.1242/jcs.242420] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The myosin family of molecular motors are well-characterised cytoskeletal proteins. However, myosins are also present in the nucleus, where they have been shown to have roles in transcription, DNA repair and viral infections. Despite their involvement in these fundamental cellular processes, our understanding of these functions and their regulation remains limited. Recently, research on nuclear myosins has been gathering pace, and this Review will evaluate the current state of the field. Here, we will focus on the variation in structure of nuclear myosins, their nuclear import and their roles within transcription, DNA damage, chromatin organisation and viral infections. We will also consider both the biochemical and biophysical properties and restraints that are placed on these multifunctional motors, and how they link to their cytoplasmic counterparts. By highlighting these properties and processes, we show just how integral nuclear myosins are for cellular survival.
Collapse
Affiliation(s)
- Alexander W Cook
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Rosemarie E Gough
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | | |
Collapse
|
42
|
Ghosh P, Vidal C, Dey S, Zhang L. Mitochondria Targeting as an Effective Strategy for Cancer Therapy. Int J Mol Sci 2020; 21:E3363. [PMID: 32397535 PMCID: PMC7247703 DOI: 10.3390/ijms21093363] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are well known for their role in ATP production and biosynthesis of macromolecules. Importantly, increasing experimental evidence points to the roles of mitochondrial bioenergetics, dynamics, and signaling in tumorigenesis. Recent studies have shown that many types of cancer cells, including metastatic tumor cells, therapy-resistant tumor cells, and cancer stem cells, are reliant on mitochondrial respiration, and upregulate oxidative phosphorylation (OXPHOS) activity to fuel tumorigenesis. Mitochondrial metabolism is crucial for tumor proliferation, tumor survival, and metastasis. Mitochondrial OXPHOS dependency of cancer has been shown to underlie the development of resistance to chemotherapy and radiotherapy. Furthermore, recent studies have demonstrated that elevated heme synthesis and uptake leads to intensified mitochondrial respiration and ATP generation, thereby promoting tumorigenic functions in non-small cell lung cancer (NSCLC) cells. Also, lowering heme uptake/synthesis inhibits mitochondrial OXPHOS and effectively reduces oxygen consumption, thereby inhibiting cancer cell proliferation, migration, and tumor growth in NSCLC. Besides metabolic changes, mitochondrial dynamics such as fission and fusion are also altered in cancer cells. These alterations render mitochondria a vulnerable target for cancer therapy. This review summarizes recent advances in the understanding of mitochondrial alterations in cancer cells that contribute to tumorigenesis and the development of drug resistance. It highlights novel approaches involving mitochondria targeting in cancer therapy.
Collapse
Affiliation(s)
| | | | | | - Li Zhang
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, TX 75080, USA; (P.G.); (C.V.); (S.D.)
| |
Collapse
|
43
|
Navinés-Ferrer A, Martín M. Long-Tailed Unconventional Class I Myosins in Health and Disease. Int J Mol Sci 2020; 21:ijms21072555. [PMID: 32272642 PMCID: PMC7177449 DOI: 10.3390/ijms21072555] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 01/21/2023] Open
Abstract
Long-tailed unconventional class I myosin, Myosin 1E (MYO1E) and Myosin 1F (MYO1F) are motor proteins that use chemical energy from the hydrolysis of adenosine triphosphate (ATP) to produce mechanical work along the actin cytoskeleton. On the basis of their motor properties and structural features, myosins perform a variety of essential roles in physiological processes such as endocytosis, exocytosis, cell adhesion, and migration. The long tailed unconventional class I myosins are characterized by having a conserved motor head domain, which binds actin and hydrolyzes ATP, followed by a short neck with an isoleucine-glutamine (IQ) motif, which binds calmodulin and is sensitive to calcium, and a tail that contains a pleckstrin homology domain (PH), a tail homology 1 domain (TH1), wherein these domains allow membrane binding, a tail homology 2 domain (TH2), an ATP-insensitive actin-binding site domain, and a single Src homology 3 domain (SH3) susceptible to binding proline rich regions in other proteins. Therefore, these motor proteins are able to bind actin, plasma membrane, and other molecules (adaptor, kinases, membrane proteins) that contribute to their function, ranging from increasing membrane tension to molecular trafficking and cellular adhesion. MYO1E and MYO1F function in host self-defense, with a better defined role in innate immunity in cell migration and phagocytosis. Impairments of their function have been identified in patients suffering pathologies ranging from tumoral processes to kidney diseases. In this review, we summarize our current knowledge of specific features and functions of MYO1E and MYO1F in various tissues, as well as their involvement in disease.
Collapse
Affiliation(s)
- A. Navinés-Ferrer
- Biochemistry and Molecular Biology Unit, Biomedicine Department, Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Laboratory of Clinic and Experimental Respiratory Immunoallergy, IDIBAPS, 08036 Barcelona, Spain
| | - M. Martín
- Biochemistry and Molecular Biology Unit, Biomedicine Department, Faculty of Medicine, University of Barcelona, 08036 Barcelona, Spain
- Laboratory of Clinic and Experimental Respiratory Immunoallergy, IDIBAPS, 08036 Barcelona, Spain
- ARADyAL research network, Carlos III Health Institute, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-93-4024541; Fax: +34-93-4035882
| |
Collapse
|
44
|
Li X, Turanli B, Juszczak K, Kim W, Arif M, Sato Y, Ogawa S, Turkez H, Nielsen J, Boren J, Uhlen M, Zhang C, Mardinoglu A. Classification of clear cell renal cell carcinoma based on PKM alternative splicing. Heliyon 2020; 6:e03440. [PMID: 32095654 PMCID: PMC7033363 DOI: 10.1016/j.heliyon.2020.e03440] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 02/07/2020] [Accepted: 02/14/2020] [Indexed: 01/17/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) accounts for 70-80% of kidney cancer diagnoses and displays high molecular and histologic heterogeneity. Hence, it is necessary to reveal the underlying molecular mechanisms involved in progression of ccRCC to better stratify the patients and design effective treatment strategies. Here, we analyzed the survival outcome of ccRCC patients as a consequence of the differential expression of four transcript isoforms of the pyruvate kinase muscle type (PKM). We first extracted a classification biomarker consisting of eight gene pairs whose within-sample relative expression orderings (REOs) could be used to robustly classify the patients into two groups with distinct molecular characteristics and survival outcomes. Next, we validated our findings in a validation cohort and an independent Japanese ccRCC cohort. We finally performed drug repositioning analysis based on transcriptomic expression profiles of drug-perturbed cancer cell lines and proposed that paracetamol, nizatidine, dimethadione and conessine can be repurposed to treat the patients in one of the subtype of ccRCC whereas chenodeoxycholic acid, fenoterol and hexylcaine can be repurposed to treat the patients in the other subtype.
Collapse
Affiliation(s)
- Xiangyu Li
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Beste Turanli
- Department of Bioengineering, Istanbul Medeniyet University, Istanbul, Turkey
| | - Kajetan Juszczak
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Woonghee Kim
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Muhammad Arif
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Yusuke Sato
- Department of Pathology and Tumor Biology, Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Urology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Kyoto, Japan
- Department of Medicine, Centre for Hematology and Regenerative Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hasan Turkez
- Department of Molecular Biology and Genetics, Erzurum Technical University, Erzurum, 25240, Turkey
| | - Jens Nielsen
- Department of Biology and Biological Engineering, Chalmers University of Technology, Gothenburg, Sweden
| | - Jan Boren
- Department of Molecular and Clinical Medicine, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Mathias Uhlen
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
| | - Cheng Zhang
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, PR China
| | - Adil Mardinoglu
- Science for Life Laboratory, KTH - Royal Institute of Technology, Stockholm, Sweden
- Centre for Host–Microbiome Interactions, Dental Institute, King's College London, London, SE1 9RT, United Kingdom
| |
Collapse
|
45
|
Nguyen AV, Trompetto B, Tan XHM, Scott MB, Hu KHH, Deeds E, Butte MJ, Chiou PY, Rowat AC. Differential Contributions of Actin and Myosin to the Physical Phenotypes and Invasion of Pancreatic Cancer Cells. Cell Mol Bioeng 2020; 13:27-44. [PMID: 32030106 PMCID: PMC6981337 DOI: 10.1007/s12195-019-00603-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 10/04/2019] [Indexed: 02/07/2023] Open
Abstract
INTRODUCTION Metastasis is a fundamentally physical process in which cells deform through narrow gaps and generate forces to invade surrounding tissues. While it is commonly thought that increased cell deformability is an advantage for invading cells, we previously found that more invasive pancreatic ductal adenocarcinoma (PDAC) cells are stiffer than less invasive PDAC cells. Here we investigate potential mechanisms of the simultaneous increase in PDAC cell stiffness and invasion, focusing on the contributions of myosin II, Arp2/3, and formins. METHOD We measure cell invasion using a 3D scratch wound invasion assay and cell stiffness using atomic force microscopy (AFM). To determine the effects of actin- and myosin-mediated force generation on cell stiffness and invasion, we treat cells with pharmacologic inhibitors of myosin II (blebbistatin), Arp2/3 (CK-666), and formins (SMIFH2). RESULTS We find that the activity of myosin II, Arp2/3, and formins all contribute to the stiffness of PDAC cells. Interestingly, we find that the invasion of PDAC cell lines is differentially affected when the activity of myosin II, Arp2/3, or formins is inhibited, suggesting that despite having similar tissue origins, different PDAC cell lines may rely on different mechanisms for invasion. CONCLUSIONS These findings deepen our knowledge of the factors that regulate cancer cell mechanotype and invasion, and incite further studies to develop therapeutics that target multiple mechanisms of invasion for improved clinical benefit.
Collapse
Affiliation(s)
- Angelyn V. Nguyen
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
| | - Brittany Trompetto
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
| | | | - Michael B. Scott
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
- Present Address: Department of Radiology, Northwestern University Feinberg School of Medicine, Chicago, USA
- Department of Biomedical Engineering, Northwestern McCormick School of Engineering, Evanston, USA
| | | | - Eric Deeds
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, USA
| | - Manish J. Butte
- Department of Pediatrics, University of California, Los Angeles, USA
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, USA
| | - Pei Yu Chiou
- Department of Bioengineering, University of California, Los Angeles, USA
- Department of Mechanical and Aerospace Engineering, University of California, Los Angeles, USA
| | - Amy C. Rowat
- Department of Integrative Biology and Physiology, University of California, 610 Charles E Young Dr. East, Los Angeles, CA 90095 USA
- Department of Bioengineering, University of California, Los Angeles, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, USA
| |
Collapse
|
46
|
Prognostic and Predictive Molecular Biomarkers for Colorectal Cancer: Updates and Challenges. Cancers (Basel) 2020; 12:cancers12020319. [PMID: 32019056 PMCID: PMC7072488 DOI: 10.3390/cancers12020319] [Citation(s) in RCA: 150] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/20/2020] [Accepted: 01/22/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a leading cause of death among cancer patients. This heterogeneous disease is characterized by alterations in multiple molecular pathways throughout its development. Mutations in RAS, along with the mismatch repair gene deficiency, are currently routinely tested in clinics. Such biomarkers provide information for patient risk stratification and for the choice of the best treatment options. Nevertheless, reliable and powerful prognostic markers that can identify “high-risk” CRC patients, who might benefit from adjuvant chemotherapy, in early stages, are currently missing. To bridge this gap, genomic information has increasingly gained interest as a potential method for determining the risk of recurrence. However, due to several limitations of gene-based signatures, these have not yet been clinically implemented. In this review, we describe the different molecular markers in clinical use for CRC, highlight new markers that might become indispensable over the next years, discuss recently developed gene expression-based tests and highlight the challenges in biomarker research.
Collapse
|
47
|
Mechanics of actin filaments in cancer onset and progress. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2020; 355:205-243. [DOI: 10.1016/bs.ircmb.2020.05.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
48
|
Klingler-Hoffmann M, Mittal P, Hoffmann P. The Emerging Role of Cytoskeletal Proteins as Reliable Biomarkers. Proteomics 2019; 19:e1800483. [PMID: 31525818 DOI: 10.1002/pmic.201800483] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/12/2019] [Indexed: 12/26/2022]
Abstract
Cytoskeletal proteins are essential building blocks of cells. More than 100 cytoskeletal and cytoskeleton-associated proteins are known and for some, their function and regulation are understood in great detail. Apart from cell shape and support, they facilitate many processes such as intracellular signaling and transport, and cancer related processes such as proliferation, migration, and invasion. During the last decade, comparative proteomic studies have identified cytoskeletal proteins as in vitro markers for tumor progression and metastasis. Here, these results are summarized and a number of unrelated studies are highlighted, identifying the same cytoskeletal proteins as potential biomarkers. These findings might indicate that the abundance of these potential markers of tumor progression is associated with the biological outcome and are independent of the cancer origin. This correlates well with recently published results from the Cancer Genome Atlas, indicating that cancers show remarkable similarities in their analyzed molecular information, independent of their organ of origin. It is postulated that the quantification of cytoskeletal proteins in healthy tissues, tumors, in adjacent tissues, and in stroma, is a great source of molecular information, which might not only be used to classify tumors, but more importantly to predict patients' outcome or even best treatment choices.
Collapse
Affiliation(s)
- Manuela Klingler-Hoffmann
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, 5095, Australia
| | - Parul Mittal
- Adelaide Proteomics Centre, School of Biological Sciences, University of Adelaide, Adelaide, 5005, Australia
| | - Peter Hoffmann
- Future Industries Institute, University of South Australia, Mawson Lakes Campus, Mawson Lakes, 5095, Australia
| |
Collapse
|
49
|
Ko YS, Bae JA, Kim KY, Kim SJ, Sun EG, Lee KH, Kim N, Kang H, Seo YW, Kim H, Chung IJ, Kim KK. MYO1D binds with kinase domain of the EGFR family to anchor them to plasma membrane before their activation and contributes carcinogenesis. Oncogene 2019; 38:7416-7432. [DOI: 10.1038/s41388-019-0954-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 07/26/2019] [Accepted: 08/02/2019] [Indexed: 12/13/2022]
|
50
|
Zhan S, Wang T, Wang M, Li J, Ge W. In-Depth Proteomics Analysis to Identify Biomarkers of Papillary Thyroid Cancer Patients Older Than 45 Years with Different Degrees of Lymph Node Metastases. Proteomics Clin Appl 2019; 13:e1900030. [PMID: 31148369 DOI: 10.1002/prca.201900030] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 05/20/2019] [Indexed: 01/08/2023]
Abstract
PURPOSE Investigations of the molecular mechanisms underlying the metastatic phenotype of papillary thyroid cancer (PTC) and identification of novel candidate biomarkers to better predict PTC metastasis are urgently required. EXPERIMENTAL DESIGN Tandem mass tag-based quantitative proteomics approach is used to identify differentially expressed proteins (DEPs) in PTC tumorous tissues with different degrees of lymph node metastases (LNMs). Furthermore, DEPs and their clinical significance are analyzed in another independent Cancer Genome Atlas dataset. RESULTS The protein profiles among tumorous tissues with different degrees of LNMs are clearly distinguished, while the protein profiles in normal tissues are remarkably similar. DEPs in tumorous tissues are mostly enriched in the categories associated with pathological hallmarks of cancer, including extracellular matrix, metabolism, and cell growth. The expression patterns of six DEPs (LAMC2, LAMB3, ATP5A1, MYO1G, S100A4, and FAS) are confirmed by the Cancer Genome Atlas dataset. Additionally, the elevated expression of LAMC2 and MYO1G mRNA levels in tumorous tissues show a positive relationship with unfavorable variables, including larger tumor size, LNMs, high AJCC staging, BRAFV600E mutation, and poor prognosis. CONCLUSIONS AND CLINICAL RELEVANCE LAMC2 and MYO1G are identified as potential candidate biomarkers for the prediction of PTC metastasis and prognosis.
Collapse
Affiliation(s)
- Shaohua Zhan
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, National Key Laboratory of Medical Molecular Biology & Department of Immunology, No. 5 Dongdan Santiao, Dongcheng, Beijing, 100005, China.,National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Tianxiao Wang
- Key Laboratory of Carcinogenesis and Translational Research Department of Head and Neck Surgery, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Meng Wang
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Jinming Li
- National Center for Clinical Laboratories, Beijing Hospital, National Center of Gerontology, Beijing, 100730, China
| | - Wei Ge
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, National Key Laboratory of Medical Molecular Biology & Department of Immunology, No. 5 Dongdan Santiao, Dongcheng, Beijing, 100005, China.,Affiliated Hospital of Hebei University, No. 212, Yu Hua East Road, Nan Shi, Baoding, Hebei, 071000, China
| |
Collapse
|