1
|
Cheng VWT, Vaughn-Beaucaire P, Shaw GC, Kriegs M, Droop A, Psakis G, Mittelbronn M, Humphries M, Esteves F, Hayes J, Cockle JV, Knipp S, Rohwedder A, Ismail A, Rominiyi O, Collis SJ, Mavria G, Samarasekara J, Ladbury JE, Ketchen S, Morton R, Fagan S, Tams D, Myers K, McGarrity-Cottrell C, Dunning M, Boissinot M, Michalopoulos G, Prior S, Lam YW, Morrison EE, Short SC, Lawler SE, Brüning-Richardson A. ARHGAP12 and ARHGAP29 exert distinct regulatory effects on switching between two cell morphological states through GSK-3 activity. Cell Rep 2025; 44:115361. [PMID: 40053455 DOI: 10.1016/j.celrep.2025.115361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 12/26/2024] [Accepted: 02/06/2025] [Indexed: 03/09/2025] Open
Abstract
Cancer cells undergo morphological changes and phenotype switching to promote invasion into healthy tissues. Manipulating the transitional morphological states in cancer cells to prevent tumor dissemination may enhance survival and improve treatment response. We describe two members of the RhoGTPase activating protein (ARHGAP) family, ARHGAP12 and ARHGAP29, as regulators of transitional morphological states in glioma via Src kinase signaling events, leading to morphological changes that correspond to phenotype switching. Moreover, we establish a link between glycogen synthase kinase 3 (GSK-3) inhibition and β-catenin translocation in altering transcription of ARHGAP12 and ARHGAP29. Silencing ARHGAP12 causes loss of N-cadherin and adoption of mesenchymal morphology, a characteristic feature of aggressive cellular behavior. In patients with glioblastoma (GBM), we identify a link between ARHGAP12 and ARHGAP29 co-expression and recurrence after treatment. Consequently, we propose that further investigation of how ARHGAPs regulate transitional morphological events to drive cancer dissemination is warranted.
Collapse
Affiliation(s)
- Vinton W T Cheng
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK; Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Philippa Vaughn-Beaucaire
- School of Applied Sciences, Joseph Priestley Building, University of Huddersfield, Huddersfield HD1 3DH, UK
| | - Gary C Shaw
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK
| | - Malte Kriegs
- Department of Radiobiology & Radiation Oncology and UCCH Kinomics Core Facility, Hubertus Wald Tumorzentrum - University Cancer Center Hamburg (UCCH), University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246 Hamburg, Germany
| | - Alastair Droop
- Wellcome Trust Genome Campus, Wellcome Trust Institute, Hinxton CB10 1RQ, UK
| | - George Psakis
- School of Applied Sciences, Joseph Priestley Building, University of Huddersfield, Huddersfield HD1 3DH, UK
| | - Michel Mittelbronn
- Luxembourg Centre of Neuropathology, Luxembourg Institute of Health, L-3555 Dudelange, Luxembourg; National Center of Pathology (NCP), Laboratoire National de Santé (LNS), L-3555 Dudelange, Luxembourg; Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L-4362, 28 Esch-sur-Alzette, Luxembourg; Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg; Department of Cancer Research (DoCR), Luxembourg Institute of Health (LIH), L-1210 Luxembourg, Luxembourg
| | - Matt Humphries
- National Pathology Imaging Cooperative, Leeds Teaching Hospitals NHS Trust, St James's University Hospital, Leeds LS9 7TF, UK
| | - Filomena Esteves
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK
| | - Josie Hayes
- REVOLUTION Medicines, Redwood City, California 94036, USA
| | | | - Sabine Knipp
- School of Applied Sciences, Joseph Priestley Building, University of Huddersfield, Huddersfield HD1 3DH, UK; Zentrum fuer Medizinische Forschung, Johannes Kepler University, Linz, Austria
| | - Arndt Rohwedder
- Zentrum fuer Medizinische Forschung, Johannes Kepler University, Linz, Austria; School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Azzam Ismail
- Pathology Department, Leeds Teaching Hospitals NHS Trust, St James's University Hospital, Leeds LS9 7TF, UK
| | - Ola Rominiyi
- Department of Neuroscience, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2RX, UK
| | - Spencer J Collis
- Department of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Sheffield S10 2RX, UK
| | - Georgia Mavria
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK
| | | | - John E Ladbury
- School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Sophie Ketchen
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK; School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK
| | - Ruth Morton
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK
| | - Sarah Fagan
- School of Applied Sciences, Joseph Priestley Building, University of Huddersfield, Huddersfield HD1 3DH, UK
| | - Daniel Tams
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK
| | - Katie Myers
- Sheffield Bioinformatics Core, Faculty of Health, University of Sheffield, Sheffield, S10 2RX, UK
| | | | - Mark Dunning
- Sheffield Bioinformatics Core, Faculty of Health, University of Sheffield, Sheffield, S10 2RX, UK
| | - Marjorie Boissinot
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK
| | - George Michalopoulos
- School of Applied Sciences, Joseph Priestley Building, University of Huddersfield, Huddersfield HD1 3DH, UK
| | - Sally Prior
- School of Applied Sciences, Joseph Priestley Building, University of Huddersfield, Huddersfield HD1 3DH, UK
| | - Yun Wah Lam
- School of Applied Sciences, Joseph Priestley Building, University of Huddersfield, Huddersfield HD1 3DH, UK
| | - Ewan E Morrison
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK
| | - Susan C Short
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, University of Leeds, Leeds LS9 7TF, UK
| | - Sean E Lawler
- Department of Pathology & Laboratory Medicine, Legorreta Cancer Center, Brown University, Providence, Rhode Island 02903, USA
| | - Anke Brüning-Richardson
- School of Applied Sciences, Joseph Priestley Building, University of Huddersfield, Huddersfield HD1 3DH, UK.
| |
Collapse
|
2
|
Liao Z, Bao Q, Saijilahu, Chimedtseren C, Tumurbaatar K, Saijilafu. Research Progress on Biomaterials for Spinal Cord Repair. Int J Nanomedicine 2025; 20:1773-1787. [PMID: 39958319 PMCID: PMC11829652 DOI: 10.2147/ijn.s501121] [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: 10/24/2024] [Accepted: 01/22/2025] [Indexed: 02/18/2025] Open
Abstract
Spinal cord injury (SCI) is a very destructive disease of the central nervous system that often causes irreversible nerve damage. Unfortunately, the adult mammalian spinal cord displays little regenerative capacity after injury. In addition, the glial scars and inflammatory responses around the lesion site are another major obstacle for successful axon regeneration after SCI. However, biomaterials are highly biocompatible, and they could provide physical guidance to allow regenerating axon growth over the lesion site and restore functional neural circuits. In addition, combined or synergistic effects of spinal cord repair can be achieved by integrating different strategies, including the use of various biomaterials and microstructures, as well as combining bioactive molecules and living cells. Therefore, it is possible to use tissue engineering scaffolds to regulate the local microenvironment of the injured spinal cord, which may achieve better functional recovery in spinal cord injury repair. In this review, we summarize the latest progress in the treatment of SCI by biomaterials, and discussed its potential mechanism.
Collapse
Affiliation(s)
- Zhenglie Liao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Qianyi Bao
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| | - Saijilahu
- Tongliao Centers for Disease Control and Prevention, Tongliao, Inner Mongolia, People’s Republic of China
| | | | - Khaliunaa Tumurbaatar
- Institute of Traditional Medicine and Technology of Mongolia, Ulaanbaatar city, Mongolia
| | - Saijilafu
- Key Laboratory of Novel Targets and Drug Study for Neural Repair of Zhejiang Province, School of Medicine, Hangzhou City University, Hangzhou, People’s Republic of China
| |
Collapse
|
3
|
Škarková A, Pelantová M, Tolde O, Legátová A, Mateu R, Bušek P, Garcia‐Borja E, Šedo A, Etienne‐Manneville S, Rösel D, Brábek J. Microtubule-associated NAV3 regulates invasive phenotypes in glioblastoma cells. Brain Pathol 2025; 35:e13294. [PMID: 39097525 PMCID: PMC11669409 DOI: 10.1111/bpa.13294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 07/17/2024] [Indexed: 08/05/2024] Open
Abstract
Glioblastomas are aggressive brain tumors for which effective therapy is still lacking, resulting in dismal survival rates. These tumors display significant phenotypic plasticity, harboring diverse cell populations ranging from tumor core cells to dispersed, highly invasive cells. Neuron navigator 3 (NAV3), a microtubule-associated protein affecting microtubule growth and dynamics, is downregulated in various cancers, including glioblastoma, and has thus been considered a tumor suppressor. In this study, we challenge this designation and unveil distinct expression patterns of NAV3 across different invasion phenotypes. Using glioblastoma cell lines and patient-derived glioma stem-like cell cultures, we disclose an upregulation of NAV3 in invading glioblastoma cells, contrasting with its lower expression in cells residing in tumor spheroid cores. Furthermore, we establish an association between low and high NAV3 expression and the amoeboid and mesenchymal invasive phenotype, respectively, and demonstrate that overexpression of NAV3 directly stimulates glioblastoma invasive behavior in both 2D and 3D environments. Consistently, we observed increased NAV3 expression in cells migrating along blood vessels in mouse xenografts. Overall, our results shed light on the role of NAV3 in glioblastoma invasion, providing insights into this lethal aspect of glioblastoma behavior.
Collapse
Affiliation(s)
- Aneta Škarková
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of ScienceCharles UniversityVestecCzech Republic
| | - Markéta Pelantová
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of ScienceCharles UniversityVestecCzech Republic
| | - Ondřej Tolde
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of ScienceCharles UniversityVestecCzech Republic
| | - Anna Legátová
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of ScienceCharles UniversityVestecCzech Republic
| | - Rosana Mateu
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Petr Bušek
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Elena Garcia‐Borja
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | - Aleksi Šedo
- Laboratory of Cancer Cell Biology, Institute of Biochemistry and Experimental Oncology, First Faculty of MedicineCharles UniversityPragueCzech Republic
| | | | - Daniel Rösel
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of ScienceCharles UniversityVestecCzech Republic
| | - Jan Brábek
- Laboratory of Cancer Cell Invasion, Department of Cell Biology, BIOCEV, Faculty of ScienceCharles UniversityVestecCzech Republic
| |
Collapse
|
4
|
Jaworska K, Senior JJ, Brüning-Richardson A, Smith AM. The effect of elevating extracellular CaCl 2: Important considerations for tissue engineering applications. Tissue Cell 2024; 91:102615. [PMID: 39579735 DOI: 10.1016/j.tice.2024.102615] [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: 07/26/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
Polysaccharides such as sodium alginate, pectin and gellan gum are widely used biomaterials, for their ability to easily form hydrogels in the presence of divalent metal ions, such as calcium - a process often cited as a mild crosslinking mechanism. However, when using these materials as substrates for tissue engineering, there is a lack of extensive studies that investigate the impact of elevated calcium concentrations on cell health and behaviour. In this study, we performed an in-depth exploration to understand the potential effects of raising extracellular CaCl2 on cell viability, proliferation, morphology and migration. We used an established glioblastoma (GBM) cell line (U251), human dermal fibroblasts (HDF), and murine osteoblasts (MC3T3) to assess the consequences of using CaCl2 in tissue engineered models to help reevaluate biomaterial suitability and enhance standardisation practices in the field of tissue engineering. Our findings revealed that the addition of CaCl2 induced notable morphological changes in GBM cells when cultured in 3D hydrogels with excess CaCl2 added, leading to a transition from mesenchymal to amoeboid phenotypes, even at a concentration as low as 8 mM. Furthermore, cell viability was reduced in a concentration-dependent manner across all cell types, and migration was also affected. Despite the widespread use of high CaCl2 concentrations to facilitate scaffold gelation, our research unveils that there can be significant risks to cell viability, proliferation, morphology, and migration when such practices are not preceded by cell line-specific experimentation and thorough standardization procedures. This highlights the importance of careful consideration and optimisation of CaCl2 concentration when used as a crosslinking agent for hydrogels intended for use in tissue engineering applications that demand accurate recapitulation of cellular responses and physiological conditions.
Collapse
Affiliation(s)
- Kayley Jaworska
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, United Kingdom
| | - Jessica J Senior
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, United Kingdom
| | - Anke Brüning-Richardson
- Department of Physical and Life Sciences, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, United Kingdom
| | - Alan M Smith
- Department of Pharmacy, School of Applied Sciences, University of Huddersfield, Queensgate, Huddersfield HD1 3DH, United Kingdom.
| |
Collapse
|
5
|
Behrooz AB, Shojaei S. Mechanistic insights into mesenchymal-amoeboid transition as an intelligent cellular adaptation in cancer metastasis and resistance. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167332. [PMID: 38960056 DOI: 10.1016/j.bbadis.2024.167332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/26/2024] [Accepted: 06/26/2024] [Indexed: 07/05/2024]
Abstract
Malignant cell plasticity is an important hallmark of tumor biology and crucial for metastasis and resistance. Cell plasticity lets cancer cells adapt to and escape the therapeutic strategies, which is the leading cause of cancer patient mortality. Epithelial cells acquire mobility via epithelial-mesenchymal transition (EMT), whereas mesenchymal cells enhance their migratory ability and clonogenic potential by acquiring amoeboid characteristics through mesenchymal-amoeboid transition (MAT). Tumor formation, progression, and metastasis depend on the tumor microenvironment (TME), a complex ecosystem within and around a tumor. Through increased migration and metastasis of cancer cells, the TME also contributes to malignancy. This review underscores the distinction between invasion pattern morphological manifestations and the diverse structures found within the TME. Furthermore, the mechanisms by which amoeboid-associated characteristics promote resistance and metastasis and how these mechanisms may represent therapeutic opportunities are discussed.
Collapse
Affiliation(s)
- Amir Barzegar Behrooz
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada; Electrophysiology Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahla Shojaei
- Department of Human Anatomy and Cell Sciences, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada.
| |
Collapse
|
6
|
Thompson E, Prior S, Brüning-Richardson A. Traditional Plant-Derived Compounds Inhibit Cell Migration and Induce Novel Cytoskeletal Effects in Glioblastoma Cells. J Xenobiot 2024; 14:613-633. [PMID: 38804289 PMCID: PMC11130960 DOI: 10.3390/jox14020036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/02/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024] Open
Abstract
Glioblastomas (GBMs) are aggressive and invasive cancers of the brain, associated with high rates of tumour recurrence and poor patient outcomes despite initial treatment. Targeting cell migration is therefore of interest in highly invasive cancers such as GBMs, to prevent tumour dissemination and regrowth. One current aim of GBM research focuses on assessing the anti-migratory properties of novel or repurposed inhibitors, including plant-based drugs which display anti-cancer properties. We investigated the potential anti-migratory activity of plant-based products with known cytotoxic effects in cancers, using a range of two-dimensional (2D) and three-dimensional (3D) migration and invasion assays as well as immunofluorescence microscopy to determine the specific anti-migratory and phenotypic effects of three plant-derived compounds, Turmeric, Indigo and Magnolia bark, on established glioma cell lines. Migrastatic activity was observed in all three drugs, with Turmeric exerting the most inhibitory effect on GBM cell migration into scratches and from the spheroid edge at all the timepoints investigated (p < 0.001). We also observed novel cytoskeletal phenotypes affecting actin and the focal adhesion dynamics. As our in vitro results determined that Turmeric, Indigo and Magnolia are promising migrastatic drugs, we suggest additional experimentation at the whole organism level to further validate these novel findings.
Collapse
Affiliation(s)
| | - Sally Prior
- Correspondence: (S.P.); (A.B.-R.); Tel.: +44-01484-472518 (A.B.-R.)
| | | |
Collapse
|
7
|
Chillon TS, Demircan K, Hackler J, Heller RA, Kaghazian P, Moghaddam A, Schomburg L. Combined copper and zinc deficiency is associated with reduced SARS-CoV-2 immunization response to BNT162b2 vaccination. Heliyon 2023; 9:e20919. [PMID: 37886755 PMCID: PMC10597833 DOI: 10.1016/j.heliyon.2023.e20919] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 10/06/2023] [Accepted: 10/11/2023] [Indexed: 10/28/2023] Open
Abstract
The essential trace elements copper, selenium and zinc are of relevance for immunity and immune response to vaccination. In this longitudinal study, adult healthcare workers (n = 126) received two doses of an mRNA vaccine (BNT162b2), and longitudinal serum samples were prepared. Vaccine-induced antibodies and their neutralizing activity were analyzed, and the trace elements copper, zinc, and selenium along with the copper transporter ceruloplasmin were measured. Subjects with combined deficiency of copper and zinc, i.e. both in the lowest tertiles at baseline, displayed particularly low antibody titers at three (Double Q1: 13 AU/mL vs. not double Q1: 29 AU/mL) and six (Double Q1: 200 AU/mL vs. not double Q1: 425 AU/mL) weeks after vaccination (p < 0.05). The results indicate the potential importance of an adequate trace element status of copper and zinc for raising a strong vaccine-induced SARS-CoV-2 antibody response, and highlights the importance of considering combined micronutrient insufficiencies, as single deficiencies may synergize.
Collapse
Affiliation(s)
- Thilo Samson Chillon
- Max Rubner Center for Cardiovascular Metabolic Renal Research (CMR), Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straße 3-4, D-10115 Berlin, Germany
| | - Kamil Demircan
- Max Rubner Center for Cardiovascular Metabolic Renal Research (CMR), Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straße 3-4, D-10115 Berlin, Germany
| | - Julian Hackler
- Max Rubner Center for Cardiovascular Metabolic Renal Research (CMR), Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straße 3-4, D-10115 Berlin, Germany
| | - Raban A. Heller
- Max Rubner Center for Cardiovascular Metabolic Renal Research (CMR), Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straße 3-4, D-10115 Berlin, Germany
- Bundeswehr Hospital Berlin, Clinic of Traumatology and Orthopaedics, D-10115 Berlin, Germany
| | - Peyman Kaghazian
- Orthopedic and Trauma Surgery, Frohsinnstraße 12, D-63739 Aschaffenburg, Germany
| | - Arash Moghaddam
- Orthopedic and Trauma Surgery, Frohsinnstraße 12, D-63739 Aschaffenburg, Germany
| | - Lutz Schomburg
- Max Rubner Center for Cardiovascular Metabolic Renal Research (CMR), Institute for Experimental Endocrinology, Charité-Universitätsmedizin Berlin, Hessische Straße 3-4, D-10115 Berlin, Germany
| |
Collapse
|
8
|
Foda BM, Neubig RR. Role of Rho/MRTF in Aggressive Vemurafenib-Resistant Murine Melanomas and Immune Checkpoint Upregulation. Int J Mol Sci 2023; 24:13785. [PMID: 37762086 PMCID: PMC10531039 DOI: 10.3390/ijms241813785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Cutaneous melanoma is the deadliest skin cancer. Most have Ras-MAPK pathway (BRAFV600E or NRAS) mutations and highly effective targeted therapies exist; however, they and immune therapies are limited by resistance, in part driven by small GTPase (Rho and Rac) activation. To facilitate preclinical studies of combination therapies to provide durable responses, we describe the first mouse melanoma lines resistant to BRAF inhibitors. Treatment of mouse lines, YUMM1.7 and YUMMER, with vemurafenib (Vem), the BRAFV600E-selective inhibitor, resulted in high-level resistance (IC50 shifts 20-30-fold). Resistant cells showed enhanced activation of Rho and the downstream transcriptional coactivator, myocardin-related transcription factor (MRTF). Resistant cells exhibited increased stress fibers, nuclear translocation of MRTF-A, and an increased MRTF-A gene signature. Pharmacological inhibition of the Rho/MRTF pathway using CCG-257081 reduced viability of resistant lines and enhanced sensitivity to Vem. Remarkably, co-treatment of parental lines with Vem and CCG-257081 eliminated resistant colony development. Resistant cells grew more slowly in vitro, but they developed highly aggressive tumors with a shortened survival of tumor-bearing mice. Increased expression of immune checkpoint inhibitor proteins (ICIs) in resistant lines may contribute to aggressive in vivo behavior. Here, we introduce the first drug-resistant mouse melanoma models for assessing combinations of targeted and immune therapies.
Collapse
Affiliation(s)
- Bardees M. Foda
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48823, USA;
- Molecular Genetics and Enzymology Department, National Research Centre, Dokki 12622, Egypt
| | - Richard R. Neubig
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI 48823, USA;
- Nicholas V. Perricone, M.D. Division of Dermatology, Department of Medicine, Michigan State University, East Lansing, MI 48823, USA
| |
Collapse
|
9
|
Nunes JM, Kell DB, Pretorius E. Cardiovascular and haematological pathology in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS): A role for viruses. Blood Rev 2023; 60:101075. [PMID: 36963989 PMCID: PMC10027292 DOI: 10.1016/j.blre.2023.101075] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 03/25/2023]
Abstract
ME/CFS is a debilitating chronic condition that often develops after viral or bacterial infection. Insight from the study of Long COVID/Post Acute Sequelae of COVID-19 (PASC), the post-viral syndrome associated with SARS-CoV-2 infection, might prove to be useful for understanding pathophysiological mechanisms of ME/CFS. Disease presentation is similar between the two conditions, and a subset of Long COVID patients meet the diagnostic criteria for ME/CFS. Since Long COVID is characterized by significant vascular pathology - including endothelial dysfunction, coagulopathy, and vascular dysregulation - the question of whether or not the same biological abnormalities are of significance in ME/CFS arises. Cardiac abnormalities have for a while now been documented in ME/CFS cohorts, with recent studies demonstrating major deficits in cerebral blood flow, and hence vascular dysregulation. A growing body of research is demonstrating that ME/CFS is accompanied by platelet hyperactivation, anomalous clotting, a procoagulant phenotype, and endothelial dysfunction. Endothelial damage and dysregulated clotting can impair substance exchange between blood and tissues, and result in hypoperfusion, which may contribute to the manifestation of certain ME/CFS symptoms. Here we review the ME/CFS literature to summarize cardiovascular and haematological findings documented in patients with the condition, and, in this context, briefly discuss the potential role of previously-implicated pathogens. Overall, cardiac and haematological abnormalities are present within ME/CFS cohorts. While atherosclerotic heart disease is not significantly associated with ME/CFS, suboptimal cardiovascular function defined by reduced cardiac output, impaired cerebral blood flow, and vascular dysregulation are, and these abnormalities do not appear to be influenced by deconditioning. Rather, these cardiac abnormalities may result from dysfunction in the (autonomic) nervous system. Plenty of recently published studies are demonstrating significant platelet hyperactivity and endothelial dysfunction in ME/CFS, as well as anomalous clotting processes. It is of particular importance to determine to what extent these cardiovascular and haematological abnormalities contribute to symptom severity, and if these two systems can be targeted for therapeutic purposes. Viral reservoirs of herpesviruses exist in ME/CFS, and most likely contribute to cardiovascular and haematological dysfunction directly or indirectly. This review highlights the potential of studying cardiac functioning, the vasculature, and coagulation system in ME/CFS.
Collapse
Affiliation(s)
- Jean M Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa.
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK; The Novo Nordisk Foundation Centre for Biosustainability, Building 220, Chemitorvet 200, Technical University of Denmark, 2800 Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Stellenbosch, Private Bag X1, Matieland 7602, South Africa; Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool L69 7ZB, UK.
| |
Collapse
|
10
|
Malalasekera AP, Neththikumara N, Somasundaram P, Pathirana S, Ediriweera C, Ediriweera D, Goonewardena SAS, Perera ND, Abeygunasekara A, Jayasekara RW, Wettasinghe K, Lokuhetty MDS, Dissanayake VHW. Clinical Exome Gene Panel Analysis of a Cohort of Urothelial Bladder Cancer Patients from Sri Lanka. Asian Pac J Cancer Prev 2023; 24:1533-1542. [PMID: 37247272 PMCID: PMC10495915 DOI: 10.31557/apjcp.2023.24.5.1533] [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: 10/13/2022] [Accepted: 05/12/2023] [Indexed: 05/31/2023] Open
Abstract
BACKGROUND Bladder cancer has a high rate of recurrence and high mortality rates in those who progress to muscle invasive disease. Biomarkers and molecular sub classification of tumours beyond standard histopathology has been proposed to address therapeutic dilemmas. The Cancer Genome Atlas project and other studies have contributed to the enhanced knowledge base of the mutational landscape of urothelial bladder cancer. Once again, these are mostly from Caucasian and Chinese patients, with data from the rest of Asia and Sri Lanka being sparse. The objective of this study was to assess the genomic variations of a cohort of urothelial bladder cancer patients in Sri Lanka. METHODS The molecular genetic study was conducted on formalin fixed paraffin embedded tumour samples of 24 patients, prospectively enrolled from 2013 to 2017. The samples were sequenced and variant distribution performed based on a 70-gene panel. RESULTS Total number of filtered mutations in the 24 patients was 10453. Median mutations per patient were 450 (range 22-987). The predominant mutational change was C>T and G>A. The top 5 mutated genes in our cohort were SYNE1, SYNE2, KMT2C, LRP2, and ANK2. The genes were clustered into 3 groups dependent on the number of mutations per patient per gene. The genes of cluster 1 and 2 mapped to Chromatin modifying enzymes and Generic Transcription Pathway. The chromatin remodelling pathway accounted for the largest proportion (22%) of mutations. CONCLUSIONS Clinical exome sequencing utilising a gene panel yielded a high mutation rate in our patients. The predominant mutational change was C>T and G>A. Three clusters of genes were identified. SYNE1 was the gene with the most mutations. The mutations comprised predominantly of genes of the chromatin remodelling pathway.
Collapse
Affiliation(s)
- Ajith P Malalasekera
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| | - Nilaksha Neththikumara
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| | - Praveenan Somasundaram
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| | - Sajeewani Pathirana
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| | | | - Dileepa Ediriweera
- Health Data Science Unit, Faculty of Medicine, University of Kelaniya, Ragama, Sri Lanka.
| | | | - Neville D Perera
- Department of Urology, National Hospital of Sri Lanka, Colombo, Sri Lanka.
| | | | - Rohan W Jayasekara
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| | - Kalum Wettasinghe
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| | - M Dilani S Lokuhetty
- Department of Pathology, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| | - Vajira H W Dissanayake
- Department of Anatomy, Genetics and Biomedical Informatics, Faculty of Medicine, University of Colombo, Colombo, Sri Lanka.
| |
Collapse
|
11
|
Rosén E, Mangukiya HB, Elfineh L, Stockgard R, Krona C, Gerlee P, Nelander S. Inference of glioblastoma migration and proliferation rates using single time-point images. Commun Biol 2023; 6:402. [PMID: 37055469 PMCID: PMC10102065 DOI: 10.1038/s42003-023-04750-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 03/23/2023] [Indexed: 04/15/2023] Open
Abstract
Cancer cell migration is a driving mechanism of invasion in solid malignant tumors. Anti-migratory treatments provide an alternative approach for managing disease progression. However, we currently lack scalable screening methods for identifying novel anti-migratory drugs. To this end, we develop a method that can estimate cell motility from single end-point images in vitro by estimating differences in the spatial distribution of cells and inferring proliferation and diffusion parameters using agent-based modeling and approximate Bayesian computation. To test the power of our method, we use it to investigate drug responses in a collection of 41 patient-derived glioblastoma cell cultures, identifying migration-associated pathways and drugs with potent anti-migratory effects. We validate our method and result in both in silico and in vitro using time-lapse imaging. Our proposed method applies to standard drug screen experiments, with no change needed, and emerges as a scalable approach to screen for anti-migratory drugs.
Collapse
Affiliation(s)
- Emil Rosén
- Dept of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | | | - Ludmila Elfineh
- Dept of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Rebecka Stockgard
- Dept of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Cecilia Krona
- Dept of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden
| | - Philip Gerlee
- Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
- Mathematical Sciences, University of Gothenburg, Gothenburg, Sweden
| | - Sven Nelander
- Dept of Immunology, Genetics, and Pathology, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
12
|
Dual Targeting of EGFR and MTOR Pathways Inhibits Glioblastoma Growth by Modulating the Tumor Microenvironment. Cells 2023; 12:cells12040547. [PMID: 36831214 PMCID: PMC9954001 DOI: 10.3390/cells12040547] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/01/2023] [Accepted: 02/07/2023] [Indexed: 02/10/2023] Open
Abstract
Glioblastoma's (GBM) aggressive growth is driven by redundant activation of a myriad of signaling pathways and genomic alterations in tyrosine kinase receptors, such as epidermal growth factor receptor (EGFR), which is altered in over 50% of cases. Single agents targeting EGFR have not proven effective against GBM. In this study, we aimed to identify an effective anti-tumor regimen using pharmacogenomic testing of patient-derived GBM samples, in culture and in vivo. High-throughput pharmacological screens of ten EGFR-driven GBM samples identified the combination of erlotinib (EGFRi) and MLN0128 (a mammalian target of rapamycin inhibitor, or MTORi) as the most effective at inhibiting tumor cell viability. The anti-tumor activity of erlonitib+MLN0128 was synergistic and produced inhibition of the p-EGFR, mitogen-activated protein kinase (MAPK), and Phosphoinositide 3-kinase (PI3K) pathways in culture. Using an orthotopic murine model of GBM, we show that erlotinib+MLN0128 inhibited tumor growth in vivo and significantly prolonged the survival of tumor-bearing mice. Expression profiling of tumor tissues from treated mice revealed a unique gene signature induced by erlotinib+MLN0128, consisting of downregulation of immunosuppressive chemokines in the tumor microenvironment, including C-C motif chemokine ligand 2 (CCL2) and periostin. Lower periostin levels resulted in the inhibition of Iba1+ (tumor-promoting) macrophage infiltration of GBM xenografts. Taken together, our results demonstrate that pharmacological co-targeting of EGFR and MTOR using clinically available drugs represents an effective treatment paradigm for EGFR-driven GBMs, acting both by inhibiting tumor cell growth and modulating the immune tumor microenvironment.
Collapse
|
13
|
Faizuloev E, Gracheva A, Korchevaya E, Smirnova D, Samoilikov R, Pankratov A, Trunova G, Khokhlova V, Ammour Y, Petrusha O, Poromov A, Leneva I, Svitich O, Zverev V. Cold-adapted SARS-CoV-2 variants with different temperature sensitivity exhibit an attenuated phenotype and confer protective immunity. Vaccine 2023; 41:892-902. [PMID: 36528447 PMCID: PMC9744683 DOI: 10.1016/j.vaccine.2022.12.019] [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: 08/11/2022] [Revised: 11/28/2022] [Accepted: 12/09/2022] [Indexed: 12/14/2022]
Abstract
As novel SARS-CoV-2 Variants of Concern emerge, the efficacy of existing vaccines against COVID-19 is declining. A possible solution to this problem lies in the development of a live attenuated vaccine potentially able of providing cross-protective activity against a wide range of SARS-CoV-2 antigenic variants. Cold-adapted (ca) SARS-CoV-2 variants, Dubrovka-ca-B4 (D-B4) and Dubrovka-ca-D2 (D-D2), were obtained after long-term passaging of the Dubrovka (D) strain in Vero cells at reduced temperatures. Virulence, immunogenicity, and protective activity of SARS-CoV-2 variants were evaluated in experiments on intranasal infection of Syrian golden hamsters (Mesocricetus auratus). In animal model infecting with ca variants, the absence of body weight loss, the significantly lower viral titer and viral RNA concentration in animal tissues, the less pronounced inflammatory lesions in animal lungs as compared with the D strain indicated the reduced virulence of the virus variant. Single intranasal immunization with D-B4 and D-D2 variants induced the production of neutralizing antibodies in hamsters and protected them from infection with the D strain and the development of severe pneumonia. It was shown that for ca SARS-CoV-2 variants, the temperature-sensitive (ts) phenotype was not obligate for virulence reduction. Indeed, the D-B4 variant, which did not possess the ts phenotype but had lost the ability to infect human lung cells Calu-3, exhibited reduced virulence in hamsters. Consequently, the potential phenotypic markers of attenuation of ca SARS-CoV-2 variants are the ca phenotype, the ts phenotype, and the change in species specificity of the virus. This study demonstrates the great potential of SARS-CoV-2 cold adaptation as a strategy to develop a live attenuated COVID-19 vaccine.
Collapse
Affiliation(s)
- Evgeny Faizuloev
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia; Russian Medical Academy of Continuous Professional Education, Moscow, Russia.
| | | | | | - Daria Smirnova
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
| | - Roman Samoilikov
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
| | - Andrey Pankratov
- FSBI NMRRC of the Ministry of Health of the Russian Federation, P.A. Hertsen Moscow Oncology Research Institute, Moscow, Russia
| | - Galina Trunova
- FSBI NMRRC of the Ministry of Health of the Russian Federation, P.A. Hertsen Moscow Oncology Research Institute, Moscow, Russia
| | - Varvara Khokhlova
- FSBI NMRRC of the Ministry of Health of the Russian Federation, P.A. Hertsen Moscow Oncology Research Institute, Moscow, Russia
| | - Yulia Ammour
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
| | - Olga Petrusha
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
| | - Artem Poromov
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia,Peoples' Friendship University of Russia, Department of Biochemistry, Moscow, Russia
| | - Irina Leneva
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia
| | - Oxana Svitich
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia,I.M. Sechenov First Moscow State Medical University (Sechenov University), F.F. Erisman Institute of Public Health, Moscow, Russia
| | - Vitaly Zverev
- I.I. Mechnikov Research Institute of Vaccines and Sera, Moscow, Russia,I.M. Sechenov First Moscow State Medical University (Sechenov University), F.F. Erisman Institute of Public Health, Moscow, Russia
| |
Collapse
|
14
|
Allison SJ. Novel Anti-Cancer Agents and Cellular Targets and Their Mechanism(s) of Action. Biomedicines 2022; 10:biomedicines10081767. [PMID: 35892667 PMCID: PMC9332372 DOI: 10.3390/biomedicines10081767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- Simon J Allison
- School of Applied Sciences, University of Huddersfield, Huddersfield HD1 3DH, UK
| |
Collapse
|
15
|
Farhadi J, Mehrzad J, Mehrad-Majd H, Motavalizadehkakhky A. Clinical significance of TRIM29 expression in patients with gastric cancer. GASTROENTEROLOGY AND HEPATOLOGY FROM BED TO BENCH 2022; 15:131-138. [PMID: 35845310 PMCID: PMC9275739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 03/11/2022] [Indexed: 11/19/2022]
Abstract
Aim The present study aimed to evaluate the expression profile, prognostic value, and possible correlation of TRIM29 with β-catenin, Cyclin D, and Bcl2 in Iranian patients with GC. Background Tripartite Motif Containing 29 (TRIM29) has been reported to function as an oncogene or a tumor suppressor depending on the tumor type. This contextual function has created a controversial situation that needs to be fully delineated in various cancers. Although few studies have reported an elevated TRIM29 expression in gastric cancer (GC), its clinicopathological and prognostic values as well as possible molecular mechanisms are yet to be re-evaluated in different populations. Methods Real-time quantitative PCR was used to detect TRIM29, β-catenin, Cyclin D, and Bcl-2 expression in 40 GC and their adjacent normal tissues. Patients were further stratified into high and low expression subgroups based on their TRIM29 expression levels. The association of TRIM29 expression level with β-catenin, Cyclin D, BCL2, some clinicopathological features, and patients' overall survival (OS) was assessed using appropriate statistical analyses. Results The results showed a significantly higher TRIM29 expression level in GC tissues compared with their corresponding normal tissues (fold change=2.94, p=0.003). Patients with high TRIM29 expression levels exhibited poorer OS (HR=1.25, 95% CI: 1.06-1.47, p=0.007). High expression of TRIM29 was also associated with increased levels of β-catenin, Cyclin D, and Bcl-2 genes expression. Conclusion Overexpression of TRIM29 is associated with poor prognosis in patients with GC and is probably mediated through β-catenin/Cyclin D/Bcl2 pathway and can be considered as a potential independent prognostic marker.
Collapse
Affiliation(s)
- Javad Farhadi
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Jamshid Mehrzad
- Department of Biochemistry, Neyshabur Branch, Islamic Azad University, Neyshabur, Iran
| | - Hassan Mehrad-Majd
- Cancer Molecular Pathology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | |
Collapse
|