1
|
López de Andrés J, Rodríguez-Santana C, de Lara-Peña L, Jiménez G, Escames G, Marchal JA. A bioengineered tumor matrix-based scaffold for the evaluation of melatonin efficacy on head and neck squamous cancer stem cells. Mater Today Bio 2024; 29:101246. [PMID: 39351489 PMCID: PMC11440243 DOI: 10.1016/j.mtbio.2024.101246] [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: 07/28/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) presents a significant challenge worldwide due to its aggressiveness and high recurrence rates post-treatment, often linked to cancer stem cells (CSCs). Melatonin shows promise as a potent tumor suppressor; however, the effects of melatonin on CSCs remain unclear, and the development of models that closely resemble tumor heterogeneity could help to better understand the effects of this molecule. This study developed a tumor scaffold based on patient fibroblast-derived decellularized extracellular matrix that mimics the HNSCC microenvironment. Our study investigates the antitumoral effects of melatonin within this context. We validated its strong antiproliferative effect on HNSCC CSCs and the reduction of tumor invasion and migration markers, even in a strongly chemoprotective environment, as it is required to increase the minimum doses necessary to impact tumor viability compared to the non-scaffolded tumorspheres culture. Moreover, melatonin exhibited no cytotoxic effects on healthy cells co-cultured in the tumor hydrogel. This scaffold-based platform allows an in vitro study closer to HNSCC tumor reality, including CSCs, stromal component, and a biomimetic matrix, providing a new valuable research tool in precision oncology.
Collapse
Affiliation(s)
- Julia López de Andrés
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada, Spain
- BioFab i3D Lab-Biofabrication and 3D (Bio)printing Singular Laboratory, University of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - César Rodríguez-Santana
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, University of Granada, Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Laura de Lara-Peña
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada, Spain
- BioFab i3D Lab-Biofabrication and 3D (Bio)printing Singular Laboratory, University of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| | - Gema Jiménez
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada, Spain
- BioFab i3D Lab-Biofabrication and 3D (Bio)printing Singular Laboratory, University of Granada, Granada, Spain
- Department of Health Sciences, University of Jaén, Jaen, Spain
| | - Germaine Escames
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada, Spain
- Instituto de Biotecnología, Centro de Investigación Biomédica, University of Granada, Granada, Spain
- Department of Physiology, Faculty of Medicine, University of Granada, Granada, Spain
- Centro de Investigación Biomédica en Red Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Investigación Biosanitaria (Ibs), Granada, San Cecilio University Hospital, Granada, Spain
| | - Juan Antonio Marchal
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada, University of Granada, Granada, Spain
- BioFab i3D Lab-Biofabrication and 3D (Bio)printing Singular Laboratory, University of Granada, Granada, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, Spain
| |
Collapse
|
2
|
Bhattacharya T, Kumari M, Kaur K, Kaity S, Arumugam S, Ravichandiran V, Roy S. Decellularized extracellular matrix-based bioengineered 3D breast cancer scaffolds for personalized therapy and drug screening. J Mater Chem B 2024; 12:8843-8867. [PMID: 39162395 DOI: 10.1039/d4tb00680a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Breast cancer (BC) is the second deadliest cancer after lung cancer. Similar to all cancers, it is also driven by a 3D microenvironment. The extracellular matrix (ECM) is an essential component of the 3D tumor micro-environment, wherein it functions as a scaffold for cells and provides metabolic support. BC is characterized by alterations in the ECM. Various studies have attempted to mimic BC-specific ECMs using artificial materials, such as Matrigel. Nevertheless, research has proven that naturally derived decellularized extracellular matrices (dECMs) are superior in providing the essential in vivo-like cues needed to mimic a cancer-like environment. Developing in vitro 3-D BC models is not straightforward and requires extensive analysis of the data established by researchers. For the benefit of researchers, in this review, we have tried to highlight all developmental studies that have been conducted by various scientists so far. The analysis of the conclusions drawn from these studies is also discussed. The advantages and drawbacks of the decellularization methods employed for generating BC scaffolds will be covered, and the review will shed light on how dECM scaffolds help develop a BC environment. The later stages of the article will also focus on immunogenicity issues arising from decellularization and the origin of the tissue. Finally, this review will also discuss the biofabrication of matrices, which is the core part of the bioengineering process.
Collapse
Affiliation(s)
- Teeshyo Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Mamta Kumari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Somasundaram Arumugam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| |
Collapse
|
3
|
Villares E, Gerecht S. Engineered Biomaterials and Model Systems to Study YAP/TAZ in Cancer. ACS Biomater Sci Eng 2024; 10:5550-5561. [PMID: 39190867 DOI: 10.1021/acsbiomaterials.4c01170] [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: 08/29/2024]
Abstract
The transcriptional coactivators yes-associated protein (YAP) and transcriptional coactivator with PDZ-binding motif (TAZ) are master regulators involved in a multitude of cancer types and a wide range of tumorigenic events, including cancer stem cell renewal, invasion, metastasis, tumor precursor emergence, and drug resistance. YAP/TAZ are known to be regulated by several external cues and stimuli, such as extracellular matrix stiffness, cell spreading, cell geometry, and shear stress. Therefore, there is a need in the field of cancer research to develop and design relevant in vitro models that can accurately reflect the complex biochemical and biophysical cues of the tumor microenvironment central to the YAP/TAZ signaling nexus. While much progress has been made, this remains a major roadblock to advancing research in this field. In this review, we highlight the current engineered biomaterials and in vitro model systems that can be used to advance our understanding of how YAP/TAZ shapes several aspects of cancer. We begin by discussing current 2D and 3D hydrogel systems that model the YAP/TAZ response to ECM stiffness. We then examine the current trends in organoid culture systems and the use of microfluidics to model the effects of cellular density and shear stress on YAP/TAZ. Finally, we analyze the ongoing pitfalls of the present models used and important future directions in engineering systems that will advance our current knowledge of YAP/TAZ in cancer.
Collapse
Affiliation(s)
- Emma Villares
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27705, United States
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27705, United States
| |
Collapse
|
4
|
Faleeva M, Ahmad S, Theofilatos K, Lynham S, Watson G, Whitehead M, Marhuenda E, Iskratsch T, Cox S, Shanahan CM. Sox9 Accelerates Vascular Aging by Regulating Extracellular Matrix Composition and Stiffness. Circ Res 2024; 134:307-324. [PMID: 38179698 PMCID: PMC10826924 DOI: 10.1161/circresaha.123.323365] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 12/21/2023] [Accepted: 12/30/2023] [Indexed: 01/06/2024]
Abstract
BACKGROUND Vascular calcification and increased extracellular matrix (ECM) stiffness are hallmarks of vascular aging. Sox9 (SRY-box transcription factor 9) has been implicated in vascular smooth muscle cell (VSMC) osteo/chondrogenic conversion; however, its relationship with aging and calcification has not been studied. METHODS Immunohistochemistry was performed on human aortic samples from young and aged patients. Young and senescent primary human VSMCs were induced to produce ECM, and Sox9 expression was manipulated using adenoviral overexpression and depletion. ECM properties were characterized using atomic force microscopy and proteomics, and VSMC phenotype on hydrogels and the ECM were examined using confocal microscopy. RESULTS In vivo, Sox9 was not spatially associated with vascular calcification but correlated with the senescence marker p16 (cyclin-dependent kinase inhibitor 2A). In vitro Sox9 showed mechanosensitive responses with increased expression and nuclear translocation in senescent cells and on stiff matrices. Sox9 was found to regulate ECM stiffness and organization by orchestrating changes in collagen (Col) expression and reducing VSMC contractility, leading to the formation of an ECM that mirrored that of senescent cells. These ECM changes promoted phenotypic modulation of VSMCs, whereby senescent cells plated on ECM synthesized from cells depleted of Sox9 returned to a proliferative state, while proliferating cells on a matrix produced by Sox9 expressing cells showed reduced proliferation and increased DNA damage, reiterating features of senescent cells. LH3 (procollagen-lysine, 2-oxoglutarate 5-dioxygenase 3) was identified as an Sox9 target and key regulator of ECM stiffness. LH3 is packaged into extracellular vesicles and Sox9 promotes extracellular vesicle secretion, leading to increased LH3 deposition within the ECM. CONCLUSIONS These findings highlight the crucial role of ECM structure and composition in regulating VSMC phenotype. We identify a positive feedback cycle, whereby cellular senescence and increased ECM stiffening promote Sox9 expression, which, in turn, drives further ECM modifications to further accelerate stiffening and senescence.
Collapse
Affiliation(s)
- Maria Faleeva
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Sadia Ahmad
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Konstantinos Theofilatos
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Steven Lynham
- Proteomics Facility, Centre of Excellence for Mass Spectrometry (S.L.) King’s College London, United Kingdom
| | - Gabriel Watson
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Meredith Whitehead
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| | - Emilie Marhuenda
- School of Engineering and Material Science, Queen Mary University of London, United Kingdom (E.M., T.I.)
| | - Thomas Iskratsch
- School of Engineering and Material Science, Queen Mary University of London, United Kingdom (E.M., T.I.)
| | - Susan Cox
- Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine (S.C.) King’s College London, United Kingdom
| | - Catherine M. Shanahan
- British Heart Foundation (BHF) Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences (M.F., S.A., K.T., G.W., M.W., C.M.S.) King’s College London, United Kingdom
| |
Collapse
|
5
|
Xu F, Dawson C, Hoare T. Multicellular Layered Nanofibrous Poly(Oligo Ethylene Glycol Methacrylate) (POEGMA)-Based Hydrogel Scaffolds via Reactive Cell Electrospinning. Adv Biol (Weinh) 2023; 7:e2300052. [PMID: 37271858 DOI: 10.1002/adbi.202300052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/06/2023] [Indexed: 06/06/2023]
Abstract
While hydrogels are demonstrated to be effective scaffolds for soft tissue engineering, existing fabrication techniques pose limitations in terms of being able to reproduce both the micro/nanofibrous structures of native extracellular matrix as well as the spatial arrangement of different cell types inherent of more complex tissues. Herein, a reactive cell electrospinning strategy is described using hydrazide and aldehyde-functionalized poly(oligoethylene glycol methacrylate) precursor polymers that can create nanofibrous hydrogel scaffolds with controllable local cell gradients using a sequential all-aqueous process that does not require additives or external energy. Cells can be encapsulated directly during the fabrication process in different layers within the scaffold, enabling localized segregation of different cell types within the structures without compromising their capacity to proliferate (≈4-fold increase in cell density over a 14 day incubation period). This sequential reactive electrospinning approach thus offers promise to generate coculture fibrous hydrogel networks in which both the nanoscale architecture and the cell distribution can be controlled, as it is essential to recreate more complex types of tissues.
Collapse
Affiliation(s)
- Fei Xu
- Department of Chemical Engineering, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Chloe Dawson
- Department of Chemical Engineering, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main St W, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
6
|
Kundu AN, Dougan CE, Mahmoud S, Kilic A, Panagiotou A, Richbourg N, Irakoze N, Peyton SR. Tenascin-C Activation of Lung Fibroblasts in a 3D Synthetic Lung Extracellular Matrix Mimic. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301493. [PMID: 37227134 PMCID: PMC10528529 DOI: 10.1002/adma.202301493] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/26/2023] [Indexed: 05/26/2023]
Abstract
The lung extracellular matrix (ECM) maintains the structural integrity of the tissue and regulates the phenotype and functions of resident fibroblasts. Lung-metastatic breast cancer alters these cell-ECM interactions, promoting fibroblast activation. There is a need for bio-instructive ECM models that match the ECM composition and biomechanics of the lung to study these cell-matrix interactions in vitro. Here, a synthetic, bioactive hydrogel is synthesized that mimics the native lung modulus and includes a representative distribution of the most abundant ECM peptide motifs responsible for integrin-binding and matrix metalloproteinase (MMP)-mediated degradation in the lung, which enables quiescent culture of human lung fibroblasts (HLFs). Stimulation with transforming growth factor β1 (TGF-β1), metastatic breast cancer conditioned media (CM), or tenascin-C-derived integrin-binding peptide activated hydrogel-encapsulated HLFs demonstrates multiple environmental methods to activate HLFs in a lung ECM-mimicking hydrogel. This lung hydrogel platform is a tunable, synthetic approach to studying the independent and combinatorial effects of ECM in regulating fibroblast quiescence and activation.
Collapse
Affiliation(s)
- Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Carey E. Dougan
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Samar Mahmoud
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Alara Kilic
- Department of Biochemistry and Molecular Biology, University of Massachusetts Amherst
| | - Alexi Panagiotou
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
| | - Nathan Richbourg
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Ninette Irakoze
- Department of Chemical Engineering, University of Massachusetts Amherst
| | - Shelly R. Peyton
- Department of Chemical Engineering, University of Massachusetts Amherst
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst
- Institute for Applied Life Sciences, University of Massachusetts Amherst, 240 Thatcher Way, Life Sciences Laboratory N531, Amherst, MA 01003
| |
Collapse
|
7
|
Blood Inflammatory-like and Lung Resident-like Eosinophils Affect Migration of Airway Smooth Muscle Cells and Their ECM-Related Proliferation in Asthma. Int J Mol Sci 2023; 24:ijms24043469. [PMID: 36834879 PMCID: PMC9958882 DOI: 10.3390/ijms24043469] [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: 12/30/2022] [Revised: 01/27/2023] [Accepted: 02/07/2023] [Indexed: 02/11/2023] Open
Abstract
Airway remodeling is a hallmark feature of asthma, and one of its key structural changes is increased airway smooth muscle (ASM) mass and disturbed extracellular matrix (ECM) homeostasis. Eosinophil functions in asthma are broadly defined; however, we lack knowledge about eosinophil subtypes' interaction with lung structural cells and their effect on the airway's local microenvironment. Therefore, we investigated the effect of blood inflammatory-like eosinophils (iEOS-like) and lung resident-like eosinophils (rEOS-like) on ASM cells via impact on their migration and ECM-related proliferation in asthma. A total of 17 non-severe steroid-free allergic asthma (AA), 15 severe eosinophilic asthma (SEA) patients, and 12 healthy control subjects (HS) were involved in this study. Peripheral blood eosinophils were enriched using Ficoll gradient centrifugation and magnetic separation, subtyped by using magnetic separation against CD62L. ASM cell proliferation was assessed by AlamarBlue assay, migration by wound healing assay, and gene expression by qRT-PCR analysis. We found that blood iEOS-like and rEOS-like cells from AA and SEA patients' upregulated genes expression of contractile apparatus proteins, COL1A1, FN, TGF-β1 in ASM cells (p < 0.05), and SEA eosinophil subtypes demonstrated the highest effect on sm-MHC, SM22, and COL1A1 gene expression. Moreover, AA and SEA patients' blood eosinophil subtypes promoted migration of ASM cells and their ECM-related proliferation, compared with HS (p < 0.05) with the higher effect of rEOS-like cells. In conclusion, blood eosinophil subtypes may contribute to airway remodeling by upregulating contractile apparatus and ECM component production in ASM cells, further promoting their migration and ECM-related proliferation, with a stronger effect of rEOS-like cells and in SEA.
Collapse
|
8
|
Jones CE, Sharick JT, Sizemore ST, Cukierman E, Strohecker AM, Leight JL. A miniaturized screening platform to identify novel regulators of extracellular matrix alignment. CANCER RESEARCH COMMUNICATIONS 2022; 2:1471-1486. [PMID: 36530465 PMCID: PMC9757767 DOI: 10.1158/2767-9764.crc-22-0157] [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: 04/13/2022] [Revised: 08/03/2022] [Accepted: 10/31/2022] [Indexed: 11/06/2022]
Abstract
Extracellular matrix alignment contributes to metastasis in a number of cancers and is a known prognostic stromal factor; however, the mechanisms controlling matrix organization remain unclear. Cancer-associated fibroblasts (CAF) play a critical role in this process, particularly via matrix production and modulation of key signaling pathways controlling cell adhesion and contractility. Stroma normalization, as opposed to elimination, is a highly sought strategy, and screening for drugs that effectively alter extracellular matrix (ECM) alignment is a practical way to identify novel CAF-normalizing targets that modulate ECM organization. To meet this need, we developed a novel high-throughput screening platform in which fibroblast-derived matrices were produced in 384-well plates, imaged with automated confocal microscopy, and analyzed using a customized MATLAB script. This platform is a technical advance because it miniaturizes the assay, eliminates costly and time-consuming experimental steps, and streamlines data acquisition and analysis to enable high-throughput screening applications. As a proof of concept, this platform was used to screen a kinase inhibitor library to identify modulators of matrix alignment. A number of novel potential regulators were identified, including several receptor tyrosine kinases (c-MET, tropomyosin receptor kinase 1 (NTRK1), HER2/ERBB2) and the serine/threonine kinases protein kinase A, C, and G (PKA, PKC, and PKG). The expression of these regulators was analyzed in publicly available patient datasets to examine the association between stromal gene expression and patient outcomes.
Collapse
Affiliation(s)
- Caitlin E. Jones
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
| | - Joe T. Sharick
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
| | - Steven T. Sizemore
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
- Department of Radiation Oncology, The Ohio State University, Columbus, Ohio
| | - Edna Cukierman
- Cancer Signaling and Epigenetics, The Marvin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple Health, Philadelphia, Pennsylvania
| | - Anne Marie Strohecker
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
- Department of Cancer Biology and Genetics, The Ohio State University, Columbus, Ohio
| | - Jennifer L. Leight
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- The James Comprehensive Cancer Center, Program in Cancer Biology, The Ohio State University, Columbus, Ohio
| |
Collapse
|
9
|
Sahan AZ, Baday M, Patel CB. Biomimetic Hydrogels in the Study of Cancer Mechanobiology: Overview, Biomedical Applications, and Future Perspectives. Gels 2022; 8:gels8080496. [PMID: 36005097 PMCID: PMC9407355 DOI: 10.3390/gels8080496] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/26/2022] [Accepted: 07/02/2022] [Indexed: 11/18/2022] Open
Abstract
Hydrogels are biocompatible polymers that are tunable to the system under study, allowing them to be widely used in medicine, bioprinting, tissue engineering, and biomechanics. Hydrogels are used to mimic the three-dimensional microenvironment of tissues, which is essential to understanding cell–cell interactions and intracellular signaling pathways (e.g., proliferation, apoptosis, growth, and survival). Emerging evidence suggests that the malignant properties of cancer cells depend on mechanical cues that arise from changes in their microenvironment. These mechanobiological cues include stiffness, shear stress, and pressure, and have an impact on cancer proliferation and invasion. The hydrogels can be tuned to simulate these mechanobiological tissue properties. Although interest in and research on the biomedical applications of hydrogels has increased in the past 25 years, there is still much to learn about the development of biomimetic hydrogels and their potential applications in biomedical and clinical settings. This review highlights the application of hydrogels in developing pre-clinical cancer models and their potential for translation to human disease with a focus on reviewing the utility of such models in studying glioblastoma progression.
Collapse
Affiliation(s)
- Ayse Z. Sahan
- Biomedical Sciences Graduate Program, Department of Pharmacology, School of Medicine, University California at San Diego, 9500 Gilman Drive, San Diego, CA 92093, USA
| | - Murat Baday
- Department of Neurology and Neurological Sciences, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Precision Health and Integrated Diagnostics Center, School of Medicine, Stanford University, Stanford, CA 94305, USA
- Correspondence: (M.B.); (C.B.P.)
| | - Chirag B. Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Neuroscience Graduate Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
- Cancer Biology Program, The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences (GSBS), Houston, TX 77030, USA
- Correspondence: (M.B.); (C.B.P.)
| |
Collapse
|
10
|
Shologu N, Gurdal M, Szegezdi E, FitzGerald U, Zeugolis DI. Macromolecular crowding in the development of a three-dimensional organotypic human breast cancer model. Biomaterials 2022; 287:121642. [PMID: 35724540 DOI: 10.1016/j.biomaterials.2022.121642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 05/31/2022] [Accepted: 06/14/2022] [Indexed: 11/02/2022]
Abstract
Although cell-derived matrices are at the forefront of scientific research and technological innovation for the development of in vitro tumour models, their two-dimensional structure and low extracellular matrix composition restrict their capacity to accurately predict toxicity of candidate molecules. Herein, we assessed the potential of macromolecular crowding (a biophysical phenomenon that significantly enhances and accelerates extracellular matrix deposition, resulting in three-dimensional tissue surrogates) in improving cell-derived matrices in vitro tumour models. Among the various decellularisation protocols assessed (NH4OH, DOC, SDS/EDTA, NP40), the NP40 appeared to be the most effective in removing cellular matter and the least destructive to the deposited matrix. Among the various cell types (mammary, skin, lung fibroblasts) used to produce the cell-derived matrices, the mammary fibroblast derived matrices produced under macromolecular crowding conditions and decellularised with NP40 resulted in significant increase in focal adhesion molecules, matrix metalloproteinases and proinflammatory cytokines, when seeded with MDA-MB-231 cells. Further, macromolecular crowding derived matrices significantly increased doxorubicin resistance and reduced the impact of intracellular reactive oxygen species mediated cell death. Collectively our data clearly illustrate the potential of macromolecular crowding in the development of cell-derived matrices-based in vitro tumour models that more accurately resemble the tumour microenvironment.
Collapse
Affiliation(s)
- Naledi Shologu
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Mehmet Gurdal
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| | - Eva Szegezdi
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Apoptosis Research Centre, Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Una FitzGerald
- Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Galway Neuroscience Centre, Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular & Biomedical Research and School of Mechanical & Materials Engineering, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
11
|
Doryab A, Schmid O. Bioactive Cell-Derived ECM Scaffold Forms a Unique Cellular Microenvironment for Lung Tissue Engineering. Biomedicines 2022; 10:biomedicines10081791. [PMID: 35892691 PMCID: PMC9394345 DOI: 10.3390/biomedicines10081791] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic lung diseases are one of the leading causes of death worldwide. Lung transplantation is currently the only causal therapeutic for lung diseases, which is restricted to end-stage disease and limited by low access to donor lungs. Lung tissue engineering (LTE) is a promising approach to regenerating a replacement for at least a part of the damaged lung tissue. Currently, lung regeneration is limited to a simplified local level (e.g., alveolar−capillary barrier) due to the sophisticated and complex structure and physiology of the lung. Here, we introduce an extracellular matrix (ECM)-integrated scaffold using a cellularization−decellularization−recellularization technique. This ECM-integrated scaffold was developed on our artificial co-polymeric BETA (biphasic elastic thin for air−liquid interface cell culture conditions) scaffold, which were initially populated with human lung fibroblasts (IMR90 cell line), as the main generator of ECM proteins. Due to the interconnected porous structure of the thin (<5 µm) BETA scaffold, the cells can grow on and infiltrate into the scaffold and deposit their own ECM. After a mild decellularization procedure, the ECM proteins remained on the scaffold, which now closely mimicked the cellular microenvironment of pulmonary cells more realistically than the plain artificial scaffolds. We assessed several decellularization methods and found that 20 mM NH4OH and 0.1% Triton X100 with subsequent DNase treatment completely removed the fibroblasts (from the first cellularization) and maintains collagen I and IV as the key ECM proteins on the scaffold. We also showed the repopulation of the primary fibroblast from human (without chronic lung disease (non-CLD) donors) and human bronchial epithelial (16HBE14o−) cells on the ECM-integrated BETA scaffold. With this technique, we developed a biomimetic scaffold that can mimic both the physico-mechanical properties and the native microenvironment of the lung ECM. The results indicate the potential of the presented bioactive scaffold for LTE application.
Collapse
|
12
|
Modification of the alginate hydrogel with fibroblast‐ and Schwann cell‐derived extracellular matrix potentiates differentiation of mesenchymal stem cells toward neuron‐like cells. J Appl Polym Sci 2022. [DOI: 10.1002/app.52501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
13
|
Poon S, Ailles LE. Modeling the Role of Cancer-Associated Fibroblasts in Tumor Cell Invasion. Cancers (Basel) 2022; 14:962. [PMID: 35205707 PMCID: PMC8870277 DOI: 10.3390/cancers14040962] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/06/2023] Open
Abstract
The major cause of cancer-related deaths can be attributed to the metastatic spread of tumor cells-a dynamic and complex multi-step process beginning with tumor cells acquiring an invasive phenotype to allow them to travel through the blood and lymphatic vessels to ultimately seed at a secondary site. Over the years, various in vitro models have been used to characterize specific steps in the cascade to collectively begin providing a clearer picture of the puzzle of metastasis. With the discovery of the TME's supporting role in activating tumor cell invasion and metastasis, these models have evolved in parallel to accommodate features of the TME and to observe its interactions with tumor cells. In particular, CAFs that reside in reactive tumor stroma have been shown to play a substantial pro-invasive role through their matrix-modifying functions; accordingly, this warranted further investigation with the development and use of invasion assays that could include these stromal cells. This review explores the growing toolbox of assays used to study tumor cell invasion, from the simple beginnings of a tumor cell and extracellular matrix set-up to the advent of models that aim to more closely recapitulate the interplay between tumor cells, CAFs and the extracellular matrix. These models will prove to be invaluable tools to help tease out the intricacies of tumor cell invasion.
Collapse
Affiliation(s)
- Stephanie Poon
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada;
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Laurie E. Ailles
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5G 1L7, Canada;
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON M5G 1L7, Canada
| |
Collapse
|
14
|
Pant T, Gaikwad G, Jain D, Dandekar P, Jain R. Establishment and characterization of lung co-culture spheroids for paclitaxel loaded Eudragit® RL 100 nanoparticle evaluation. Biotechnol Prog 2021; 37:e3203. [PMID: 34427389 DOI: 10.1002/btpr.3203] [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: 06/08/2021] [Revised: 08/04/2021] [Accepted: 08/23/2021] [Indexed: 11/09/2022]
Abstract
3D cell cultures are regarded as a better and more relevant approach for screening drugs and therapeutics, particularly due to their likeness with the in vivo conditions. Spheroids offer an intermediate platform between in vitro and in vivo models, for conducting tumor-based investigations. In this study, a simple setup was developed for consistent generation of lung co-culture spheroids, which were developed using the cancer cell lines A549, NCI H460, and fibroblast cells WI-38. The potential of these spheroids for evaluating the toxicity of Eudragit® RL 100 nanoparticles (ENP) was explored. Monodisperse ENP, having the size range of 140-200 nm was prepared using the nanoprecipitation method. These were loaded with the poorly water-soluble anticancer drug paclitaxel. The evaluation of toxicity and uptake of drug-loaded ENP revealed that 2D monolayers were more sensitive to treatment than 3D spheroids. Within spheroids, co-cultures were more resistant to the treatment than monocultures. Overall, our findings demonstrated that the lung co-culture spheroids were a suitable model for accelerating the efficacy and toxicity-related investigations of novel drug delivery systems.
Collapse
Affiliation(s)
- Tejal Pant
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Ganesh Gaikwad
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Dhiraj Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Ratnesh Jain
- Department of Chemical Engineering, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
15
|
Emmert M, Somorowsky F, Ebert J, Görick D, Heyn A, Rosenberger E, Wahl M, Heinrich D. Modulation of Mammalian Cell Behavior by Nanoporous Glass. Adv Biol (Weinh) 2021; 5:e2000570. [PMID: 33960740 DOI: 10.1002/adbi.202000570] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 03/31/2021] [Indexed: 11/08/2022]
Abstract
The introduction of novel bioactive materials to manipulate living cell behavior is a crucial topic for biomedical research and tissue engineering. Biomaterials or surface patterns that boost specific cell functions can enable innovative new products in cell culture and diagnostics. This study investigates the influence of the intrinsically nano-patterned surface of nanoporous glass membranes on the behavior of mammalian cells. Three different cell lines and primary human mesenchymal stem cells (hMSCs) proliferate readily on nanoporous glass membranes with mean pore sizes between 10 and 124 nm. In both proliferation and mRNA expression experiments, L929 fibroblasts show a distinct trend toward mean pore sizes >80 nm. For primary hMSCs, excellent proliferation is observed on all nanoporous surfaces. hMSCs on samples with 17 nm pore size display increased expression of COL10, COL2A1, and SOX9, especially during the first two weeks of culture. In the upside down culture, SK-MEL-28 cells on nanoporous glass resist the gravitational force and proliferate well in contrast to cells on flat references. The effect of paclitaxel treatment of MDA-MB-321 breast cancer cells is already visible after 48 h on nanoporous membranes and strongly pronounced in comparison to reference samples, underlining the material's potential for functional drug screening.
Collapse
Affiliation(s)
- Martin Emmert
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany.,Julius-Maximilians-Universität Würzburg, Chemical Technology of Material Synthesis, Röntgenring 11, 97070, Würzburg, Germany
| | - Ferdinand Somorowsky
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Jutta Ebert
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Dominik Görick
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Andreas Heyn
- Julius-Maximilians-Universität Würzburg, Chemical Technology of Material Synthesis, Röntgenring 11, 97070, Würzburg, Germany
| | - Eva Rosenberger
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany
| | - Moritz Wahl
- Julius-Maximilians-Universität Würzburg, Chemical Technology of Material Synthesis, Röntgenring 11, 97070, Würzburg, Germany
| | - Doris Heinrich
- Fraunhofer Institute for Silicate Research ISC, Neunerplatz 2, 97082, Würzburg, Germany.,Leiden University, LION Leiden Institute of Physics, Niels Bohrweg 2, Leiden, 2333 CA, The Netherlands
| |
Collapse
|
16
|
Fang C, Guo F, Zhao X, Zhang Z, Lu J, Pan H, Xu T, Li W, Yang M, Huang Y, Zhao Y, Zhao S. Biological mechanisms of growth performance and meat quality in porcine muscle tissue. Anim Biotechnol 2021; 33:1246-1254. [PMID: 33704018 DOI: 10.1080/10495398.2021.1886939] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Growth performance and meat quality are important traits for pig production. The aim of the present study was to investigate the molecular mechanisms underlying growth performance and meat quality, and to identify novel target molecules for predicting the growth performance and meat quality. The differentially expressed genes (DEGs) in Diannan small ears pigs (DSP) and Landrace pigs (LP) were assessed by RNA-sequencing analyzing technology. A total of 339 DEGs were obtained between DSP and LP. 146 DEGs were upregulated in LP compared with DSP and 193 DEGs were upregulated in DSP compared with LP. The DEGs were significantly enriched in 26 GO and 3 KEGG pathways. The protein-protein interaction (PPI) network with 201 nodes and 382 edges was constructed and 5 modules were extracted from the entire network. The identified upregulated expression of genes involved in glycolysis and myogenesis as well as extracellular matrix may be associated with fast body and muscle deposition rates in LP. Increased expression of genes involved in PPAR signaling pathway and fatty acid metabolism as well as oxidative phosphate processes could be related to the intramuscular fat deposition and meat quality in DSP. The present study may provide an improved understanding of the growth performance and meat quality.
Collapse
Affiliation(s)
- Chen Fang
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Fei Guo
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Xiaoqi Zhao
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China.,Institute of Herbivorous Livestock, Yunnan Academy of Animal Sciences, Kunming, China
| | - Zining Zhang
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Junlan Lu
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Hongbin Pan
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Taojie Xu
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Weizhen Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Minghua Yang
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Ying Huang
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| | - Yanguang Zhao
- Research Institute of Pig and Animal Nutrition, Yunnan Academy of Animal Sciences, Kunming, China
| | - Sumei Zhao
- Yunnan Key Laboratory of Animal Nutrition and Feed Science, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
17
|
Harikrishnan K, Joshi O, Madangirikar S, Balasubramanian N. Cell Derived Matrix Fibulin-1 Associates With Epidermal Growth Factor Receptor to Inhibit Its Activation, Localization and Function in Lung Cancer Calu-1 Cells. Front Cell Dev Biol 2020; 8:522. [PMID: 32719793 PMCID: PMC7348071 DOI: 10.3389/fcell.2020.00522] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 06/02/2020] [Indexed: 12/11/2022] Open
Abstract
Epidermal Growth Factor Receptor (EGFR) is a known promoter of tumor progression and is overexpressed in lung cancers. Growth factor receptors (including EGFR) are known to interact with extracellular matrix (ECM) proteins, which regulate their activation and function. Fibulin-1 (FBLN1) is a major component of the ECM in lung tissue, and its levels are known to be downregulated in non-small cell lung cancers (NSCLC). To test the possible role FBLN1 isoforms could have in regulating EGFR signaling and function in lung cancer, we performed siRNA mediated knockdown of FBLN1C and FBLN1D in NSCLC Calu-1 cells. Their loss significantly increased basal (with serum) and EGF (Epidermal Growth Factor) mediated EGFR activation without affecting net EGFR levels. Overexpression of FBLN1C and FBLN1D also inhibits EGFR activation confirming their regulatory crosstalk. Loss of FBLN1C and FBLN1D promotes EGFR-dependent cell migration, inhibited upon Erlotinib treatment. Mechanistically, both FBLN1 isoforms interact with EGFR, their association not dependent on its activation. Notably, cell-derived matrix (CDM) enriched FBLN1 binds EGFR. Calu-1 cells plated on CDM derived from FBLN1C and FBLN1D knockdown cells show a significant increase in EGF mediated EGFR activation. This promotes cell adhesion and spreading with active EGFR enriched at membrane ruffles. Both adhesion and spreading on CDMs is significantly reduced by Erlotinib treatment. Together, these findings show FBLN1C/1D, as part of the ECM, can bind and regulate EGFR activation and function in NSCLC Calu-1 cells. They further highlight the role tumor ECM composition could have in influencing EGFR dependent lung cancers.
Collapse
Affiliation(s)
| | - Omkar Joshi
- Indian Institute of Science Education and Research, Pune, India
| | | | | |
Collapse
|
18
|
Hassan G, Seno M. Blood and Cancer: Cancer Stem Cells as Origin of Hematopoietic Cells in Solid Tumor Microenvironments. Cells 2020; 9:cells9051293. [PMID: 32455995 PMCID: PMC7290570 DOI: 10.3390/cells9051293] [Citation(s) in RCA: 20] [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: 03/18/2020] [Revised: 05/19/2020] [Accepted: 05/20/2020] [Indexed: 12/27/2022] Open
Abstract
The concepts of hematopoiesis and the generation of blood and immune cells from hematopoietic stem cells are some steady concepts in the field of hematology. However, the knowledge of hematopoietic cells arising from solid tumor cancer stem cells is novel. In the solid tumor microenvironment, hematopoietic cells play pivotal roles in tumor growth and progression. Recent studies have reported that solid tumor cancer cells or cancer stem cells could differentiate into hematopoietic cells. Here, we discuss efforts and research that focused on the presence of hematopoietic cells in tumor microenvironments. We also discuss hematopoiesis from solid tumor cancer stem cells and clarify the notion of differentiation of solid tumor cancer stem cells into non-cancer hematopoietic stem cells.
Collapse
Affiliation(s)
- Ghmkin Hassan
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan;
- Department of Microbiology and Biochemistry, Faculty of Pharmacy, Damascus University, Damascus 10769, Syria
| | - Masaharu Seno
- Graduate School of Interdisciplinary Science and Engineering in Health Systems, Okayama University, Okayama 700-8530, Japan;
- Correspondence: ; Tel.: +81-86-251-8216
| |
Collapse
|
19
|
Ferreira LP, Gaspar VM, Mano JF. Decellularized Extracellular Matrix for Bioengineering Physiomimetic 3D in Vitro Tumor Models. Trends Biotechnol 2020; 38:1397-1414. [PMID: 32416940 DOI: 10.1016/j.tibtech.2020.04.006] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/10/2020] [Accepted: 04/14/2020] [Indexed: 02/06/2023]
Abstract
Recent advances in the extraction and purification of decellularized extracellular matrix (dECM) obtained from healthy or malignant tissues open new avenues for engineering physiomimetic 3D in vitro tumor models, which closely recapitulate key biomolecular hallmarks and the dynamic cancer cell-ECM interactions in the tumor microenvironment. We review current and upcoming methodologies for chemical modification of dECM-based biomaterials and advanced bioprocessing into organotypic 3D solid tumor models. A comprehensive review of disruptive advances and shortcomings of exploring dECM-based biomaterials for recapitulating the native tumor-supporting matrix is also provided. We hope to drive the discussion on how 3D dECM testing platforms can be leveraged for generating microphysiological tumor surrogates that generate more robust and predictive data on therapeutic bioperformance.
Collapse
Affiliation(s)
- Luís P Ferreira
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO, Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
20
|
Kuo PH, Teng YH, Cin AL, Han W, Huang PW, Wang LHC, Chou YT, Yang JL, Tseng YL, Kao M, Chang MDT. Heparan sulfate targeting strategy for enhancing liposomal drug accumulation and facilitating deep distribution in tumors. Drug Deliv 2020; 27:542-555. [PMID: 32241176 PMCID: PMC7170378 DOI: 10.1080/10717544.2020.1745326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Nanoparticles (NPs), such as liposomes, effectively evade the severe toxicity of unexpected accumulation and passively shuttle drugs into tumor tissues by enhanced permeability and retention. In the case of non-small cell lung cancer and pancreatic ductal adenocarcinoma, cancer-associated fibroblasts promote the aggregation of a gel-like extracellular matrix that forms a physical barrier in the desmoplastic stroma of the tumor. These stroma are composed of protein networks and glycosaminoglycans (GAGs) that greatly compromise tumor-penetrating performance, leading to insufficient extravasation and tissue penetration of NPs. Moreover, the presence of heparan sulfate (HS) and related proteoglycans on the cell surface and tumor extracellular matrix may serve as molecular targets for NP-mediated drug delivery. Here, a GAG-binding peptide (GBP) with high affinity for HS and high cell-penetrating activity was used to develop an HS-targeting delivery system. Specifically, liposomal doxorubicin (L-DOX) was modified by post-insertion with the GBP. We show that the in vitro uptake of L-DOX in A549 lung adenocarcinoma cells increased by GBP modification. Cellular uptake of GBP-modified L-DOX (L-DOX-GBP) was diminished in the presence of extracellular HS but not in the presence of other GAGs, indicating that the interaction with HS is critical for the cell surface binding of L-DOX-GBP. The cytotoxicity of doxorubicin positively correlated with the molecular composition of GBP. Moreover, GBP modification improved the in vivo distribution and anticancer efficiency of L-DOX, with enhanced desmoplastic targeting and extensive distribution. Taken together, GBP modification may greatly improve the tissue distribution and delivery efficiency of NPs against HS-abundant desmoplastic stroma-associated neoplasm.
Collapse
Affiliation(s)
- Ping-Hsueh Kuo
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Hsien Teng
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ann-Lun Cin
- Operations Center for Industry Collaboration, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen Han
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.,Graduate Program of Biotechnology in Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | | | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Ting Chou
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Jia-Ling Yang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | | | - Minhsiung Kao
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Margaret Dah-Tsyr Chang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan.,Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
21
|
Bairagi D, Biswas P, Basu K, Hazra S, Hermida-Merino D, Sinha DK, Hamley IW, Banerjee A. Self-Assembling Peptide-Based Hydrogel: Regulation of Mechanical Stiffness and Thermal Stability and 3D Cell Culture of Fibroblasts. ACS APPLIED BIO MATERIALS 2019; 2:5235-5244. [DOI: 10.1021/acsabm.9b00424] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Dipayan Bairagi
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Parijat Biswas
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Kingshuk Basu
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Soumyajit Hazra
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | | | - Deepak Kumar Sinha
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Ian W. Hamley
- Department of Chemistry, University of Reading, Whiteknights, Reading RG6, 6AD, United Kingdom
| | - Arindam Banerjee
- School of Biological Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, India
| |
Collapse
|
22
|
Saforo D, Omer L, Smolenkov A, Barve A, Casson L, Boyd N, Clark G, Siskind L, Beverly L. Primary lung cancer samples cultured under microenvironment-mimetic conditions enrich for mesenchymal stem-like cells that promote metastasis. Sci Rep 2019; 9:4177. [PMID: 30862796 PMCID: PMC6414636 DOI: 10.1038/s41598-019-40519-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 02/01/2019] [Indexed: 12/27/2022] Open
Abstract
The tumor microenvironment (TME) is composed of a heterogeneous biological ecosystem of cellular and non-cellular elements including transformed tumor cells, endothelial cells, immune cells, activated fibroblasts or myofibroblasts, stem and progenitor cells, as well as the cytokines and matrix that they produce. The constituents of the TME stroma are multiple and varied, however cancer associated fibroblasts (CAF) and their contribution to the TME are important in tumor progression. CAF are hypothesized to arise from multiple progenitor cell types, including mesenchymal stem cells. Currently, isolation of TME stroma from patients is complicated by issues such as limited availability of biopsy material and cell stress incurred during lengthy adaptation to atmospheric oxygen (20% O2) in cell culture, limiting pre-clinical studies of patient tumor stromal interactions. Here we describe a microenvironment mimetic in vitro cell culturing system that incorporates elements of the in vivo lung environment, including lung fibroblast derived extracellular matrix and physiological hypoxia (5% O2). Using this system, we easily isolated and rapidly expanded stromal progenitors from patient lung tumor resections without complex sorting methods or growth supplements. These progenitor populations retained expression of pluripotency markers, secreted factors associated with cancer progression, and enhanced tumor cell growth and metastasis. An understanding of the biology of these progenitor cell populations in a TME-like environment may advance our ability to target these cells and limit their effects on promoting cancer metastasis.
Collapse
Affiliation(s)
- Douglas Saforo
- University of Louisville, Department of Pharmacology & Toxicology, Louisville, KY, 40202, USA
| | - Linda Omer
- University of Louisville, Department of Biochemistry & Molecular Genetics, Louisville, KY, 40202, USA
- University of Louisville, Cardiovascular Innovation Institute, Louisville, KY, 40202, USA
| | - Andrei Smolenkov
- University of Louisville, James Graham Brown Cancer Center, Louisville, KY, 40202, USA
| | - Aditya Barve
- University of Louisville, Department of Pharmacology & Toxicology, Louisville, KY, 40202, USA
| | - Lavona Casson
- University of Louisville, James Graham Brown Cancer Center, Louisville, KY, 40202, USA
| | - Nolan Boyd
- University of Louisville, Cardiovascular Innovation Institute, Louisville, KY, 40202, USA
- University of Louisville, Department of Physiology, Louisville, KY, 40202, USA
| | - Geoffrey Clark
- University of Louisville, Department of Pharmacology & Toxicology, Louisville, KY, 40202, USA
- University of Louisville, James Graham Brown Cancer Center, Louisville, KY, 40202, USA
| | - Leah Siskind
- University of Louisville, Department of Pharmacology & Toxicology, Louisville, KY, 40202, USA.
- University of Louisville, James Graham Brown Cancer Center, Louisville, KY, 40202, USA.
| | - Levi Beverly
- University of Louisville, Department of Pharmacology & Toxicology, Louisville, KY, 40202, USA.
- University of Louisville, James Graham Brown Cancer Center, Louisville, KY, 40202, USA.
- University of Louisville, Department of Medicine, Louisville, KY, 40202, USA.
| |
Collapse
|
23
|
Covalent immobilization of fibroblast-derived matrix on metallic stent for expeditious re-endothelialization. J IND ENG CHEM 2019. [DOI: 10.1016/j.jiec.2018.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
24
|
Horodyska J, Wimmers K, Reyer H, Trakooljul N, Mullen AM, Lawlor PG, Hamill RM. RNA-seq of muscle from pigs divergent in feed efficiency and product quality identifies differences in immune response, growth, and macronutrient and connective tissue metabolism. BMC Genomics 2018; 19:791. [PMID: 30384851 PMCID: PMC6211475 DOI: 10.1186/s12864-018-5175-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 10/16/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Feed efficiency (FE) is an indicator of efficiency in converting energy and nutrients from feed into a tissue that is of major environmental and economic significance. The molecular mechanisms contributing to differences in FE are not fully elucidated, therefore the objective of this study was to profile the porcine Longissimus thoracis et lumborum (LTL) muscle transcriptome, examine the product quality from pigs divergent in FE and investigate the functional networks underpinning the potential relationship between product quality and FE. RESULTS RNA-Seq (n = 16) and product quality (n = 40) analysis were carried out in the LTL of pigs differing in FE status. A total of 272 annotated genes were differentially expressed with a P < 0.01. Functional annotation revealed a number of biological events related to immune response, growth, carbohydrate & lipid metabolism and connective tissue indicating that these might be the key mechanisms governing differences in FE. Five most significant bio-functions altered in FE groups were 'haematological system development & function', 'lymphoid tissue structure & development', 'tissue morphology', 'cellular movement' and 'immune cell trafficking'. Top significant canonical pathways represented among the differentially expressed genes included 'IL-8 signalling', 'leukocyte extravasation signalling, 'sphingosine-1-phosphate signalling', 'PKCθ signalling in T lymphocytes' and 'fMLP signalling in neutrophils'. A minor impairment in the quality of meat, in relation to texture and water-holding capacity, produced by high-FE pigs was observed. High-FE pigs also had reduced intramuscular fat content and improved nutritional profile in terms of fatty acid composition. CONCLUSIONS Ontology analysis revealed enhanced activity of adaptive immunity and phagocytes in high-FE pigs suggesting more efficient conserving of resources, which can be utilised for other important biological processes. Shifts in carbohydrate conversion into glucose in FE-divergent muscle may underpin the divergent evolution of pH profile in meat from the FE-groups. Moreover, altered amino acid metabolism and increased mobilisation & flux of calcium may influence growth in FE-divergent muscle. Furthermore, decreased degradation of fibroblasts in FE-divergent muscle could impact on collagen turnover and alter tenderness of meat, whilst enhanced lipid degradation in high-FE pigs may potentially underlie a more efficient fat metabolism in these animals.
Collapse
Affiliation(s)
- Justyna Horodyska
- Teagasc, Food Research Centre, Ashtown, Dublin, 15, Ireland.,Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | - Klaus Wimmers
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany.,Faculty of Agricultural and Environmental Sciences, University Rostock, Rostock, Germany
| | - Henry Reyer
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | - Nares Trakooljul
- Leibniz Institute for Farm Animal Biology (FBN), Institute for Genome Biology, Dummerstorf, Germany
| | | | - Peadar G Lawlor
- Teagasc, Pig Development Department, AGRIC, Moorepark, Fermoy, Co. Cork, Ireland
| | - Ruth M Hamill
- Teagasc, Food Research Centre, Ashtown, Dublin, 15, Ireland.
| |
Collapse
|
25
|
Senthebane DA, Jonker T, Rowe A, Thomford NE, Munro D, Dandara C, Wonkam A, Govender D, Calder B, Soares NC, Blackburn JM, Parker MI, Dzobo K. The Role of Tumor Microenvironment in Chemoresistance: 3D Extracellular Matrices as Accomplices. Int J Mol Sci 2018; 19:E2861. [PMID: 30241395 PMCID: PMC6213202 DOI: 10.3390/ijms19102861] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 09/17/2018] [Accepted: 09/18/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The functional interplay between tumor cells and their adjacent stroma has been suggested to play crucial roles in the initiation and progression of tumors and the effectiveness of chemotherapy. The extracellular matrix (ECM), a complex network of extracellular proteins, provides both physical and chemicals cues necessary for cell proliferation, survival, and migration. Understanding how ECM composition and biomechanical properties affect cancer progression and response to chemotherapeutic drugs is vital to the development of targeted treatments. METHODS 3D cell-derived-ECMs and esophageal cancer cell lines were used as a model to investigate the effect of ECM proteins on esophageal cancer cell lines response to chemotherapeutics. Immunohistochemical and qRT-PCR evaluation of ECM proteins and integrin gene expression was done on clinical esophageal squamous cell carcinoma biopsies. Esophageal cancer cell lines (WHCO1, WHCO5, WHCO6, KYSE180, KYSE 450 and KYSE 520) were cultured on decellularised ECMs (fibroblasts-derived ECM; cancer cell-derived ECM; combinatorial-ECM) and treated with 0.1% Dimethyl sulfoxide (DMSO), 4.2 µM cisplatin, 3.5 µM 5-fluorouracil and 2.5 µM epirubicin for 24 h. Cell proliferation, cell cycle progression, colony formation, apoptosis, migration and activation of signaling pathways were used as our study endpoints. RESULTS The expression of collagens, fibronectin and laminins was significantly increased in esophageal squamous cell carcinomas (ESCC) tumor samples compared to the corresponding normal tissue. Decellularised ECMs abrogated the effect of drugs on cancer cell cycling, proliferation and reduced drug induced apoptosis by 20⁻60% that of those plated on plastic. The mitogen-activated protein kinase-extracellular signal-regulated kinase (MEK-ERK) and phosphoinositide 3-kinase-protein kinase B (PI3K/Akt) signaling pathways were upregulated in the presence of the ECMs. Furthermore, our data show that concomitant addition of chemotherapeutic drugs and the use of collagen- and fibronectin-deficient ECMs through siRNA inhibition synergistically increased cancer cell sensitivity to drugs by 30⁻50%, and reduced colony formation and cancer cell migration. CONCLUSION Our study shows that ECM proteins play a key role in the response of cancer cells to chemotherapy and suggest that targeting ECM proteins can be an effective therapeutic strategy against chemoresistant tumors.
Collapse
Affiliation(s)
- Dimakatso Alice Senthebane
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Tina Jonker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Arielle Rowe
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Nicholas Ekow Thomford
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Daniella Munro
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Collet Dandara
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Ambroise Wonkam
- Pharmacogenetics Research Group, Division of Human Genetics, Department of Pathology and Institute of Infectious Diseases and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Dhirendra Govender
- Division of Anatomical Pathology, Faculty of Health Sciences, University of Cape Town, NHLS-Groote Schuur Hospital, Cape Town 7925, South Africa.
| | - Bridget Calder
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - Nelson C Soares
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - Jonathan M Blackburn
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town 7925, South Africa.
| | - M Iqbal Parker
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
| | - Kevin Dzobo
- Division of Medical Biochemistry and Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Anzio Road, Observatory, Cape Town 7925, South Africa.
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Cape Town Component, Wernher and Beit Building (South), UCT Campus, Anzio Road, Observatory, Cape Town 7925, South Africa.
| |
Collapse
|
26
|
Kotwal GJ, Martin MD, Chien S. Significant upregulation of U1 and U4 spliceosomal snRNAs by ATP nanoliposomes explains acceleration of wound healing, due to increased pre-mRNA processing to functional mRNA. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1289-1299. [PMID: 29627519 DOI: 10.1016/j.nano.2018.03.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 03/12/2018] [Accepted: 03/21/2018] [Indexed: 01/06/2023]
Abstract
Delayed wound healing is one of the hallmarks of diabetic complications and certain autoimmune inflammatory diseases. Extensive wound healing studies in rabbits have indicated that the delivery of ATP encapsulated in unilamellar nanoliposomes causes rapid cell proliferation and fast tracks the wound healing process. In the current study, we explored the possible molecular mechanism underlying this response by comparing gene expression in cultured rabbit kidney cells treated with either ATP nanoliposomes (containing 1 mg Mg-ATP/ml formulation) or control nanoliposomes (containing 1 mg/ml unmetabolisable gamma-thio-ATP/ml formulation). High-quality total RNA was isolated 24 h from the cells and subjected to RNA seq technology, which revealed significant overexpression of specific noncoding RNAs. The U1 spliceosomal RNA, U1 snRNA, was upregulated more than 250-fold following treatment with ATP nanoliposomes. This multifunctional U1 spliceosomal RNA may function in transcription by speeding up the critical splicing step, thereby facilitating faster processing of pre-mRNA to translatable mRNA.
Collapse
Affiliation(s)
| | | | - Sufan Chien
- Department of Surgery, University of Louisville School of Medicine, Louisville, KY, USA; Noveratech LLC, iHUB, Louisville, KY, USA
| |
Collapse
|
27
|
A New Chapter for Mesenchymal Stem Cells: Decellularized Extracellular Matrices. Stem Cell Rev Rep 2017; 13:587-597. [DOI: 10.1007/s12015-017-9757-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
28
|
The effects of porosity and stiffness of genipin cross-linked egg white simulating aged extracellular matrix on proliferation and aggregation of ovarian cancer cells. Colloids Surf A Physicochem Eng Asp 2017. [DOI: 10.1016/j.colsurfa.2017.02.031] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
|
29
|
Biophysical regulation of mouse embryonic stem cell fate and genomic integrity by feeder derived matrices. Biomaterials 2017; 119:9-22. [DOI: 10.1016/j.biomaterials.2016.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 12/07/2016] [Indexed: 01/01/2023]
|
30
|
Guo Z, Zhang T, Fang K, Liu P, Li M, Gu N. The effect of porosity and stiffness of glutaraldehyde cross-linked egg white scaffold simulating aged extracellular matrix on distribution and aggregation of ovarian cancer cells. Colloids Surf A Physicochem Eng Asp 2016. [DOI: 10.1016/j.colsurfa.2016.05.050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
|
31
|
Liu GX, Xi HQ, Sun XY, Geng ZJ, Yang SW, Lu YJ, Wei B, Chen L. Isoprenaline Induces Periostin Expression in Gastric Cancer. Yonsei Med J 2016; 57:557-64. [PMID: 26996552 PMCID: PMC4800342 DOI: 10.3349/ymj.2016.57.3.557] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 12/01/2015] [Accepted: 12/10/2015] [Indexed: 11/27/2022] Open
Abstract
PURPOSE Periostin mediates critical steps in gastric cancer and is involved in various signaling pathways. However, the roles of periostin in promoting gastric cancer metastasis are not clear. The aim of this study was to investigate the relevance between periostin expression and gastric cancer progression and the role of stress-related hormones in the regulation of cancer development and progression. MATERIALS AND METHODS Normal, cancerous and metastatic gastric tissues were collected from patients diagnosed with advanced gastric cancer. The in vivo expression of periostin was evaluated by in situ hybridization and immunofluorescent staining. Meanwhile, human gastric adenocarcinoma cell lines MKN-45 and BGC-803 were used to detect the in vitro expression of periostin by using quantitative real-time polymerase chain reaction (PCR) and western blotting. RESULTS Periostin is expressed in the stroma of the primary gastric tumors and metastases, but not in normal gastric tissue. In addition, we observed that periostin is located mainly in pericryptal fibroblasts, but not in the tumor cells, and strongly correlated to the expression of α-smooth muscle actin (SMA). Furthermore, the distribution patterns of periostin were broader as the clinical staging of tumors progressed. We also identified a role of stress-related signaling in promoting cancer development and progression, and found that isoprenaline upregulated expression levels of periostin in gastric cancer cells. CONCLUSION These findings suggest that the distribution pattern of periostin was broader as the clinical staging of the tumor progressed and found that isoprenaline upregulated expression levels of periostin in gastric cancer cells.
Collapse
Affiliation(s)
- Guo-Xiao Liu
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Hong-Qing Xi
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China
| | - Xiao-Yan Sun
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Trauma Center of Postgraduate Medical School, Chinese PLA General Hospital, Beijing, China
| | - Zhi-Jun Geng
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Trauma Center of Postgraduate Medical School, Chinese PLA General Hospital, Beijing, China
| | - Shao-Wei Yang
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Trauma Center of Postgraduate Medical School, Chinese PLA General Hospital, Beijing, China
| | - Yan-Jie Lu
- Department of Pathology, Chengde Medical College, Chengde, Hebei Province, China
| | - Bo Wei
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China.
| | - Lin Chen
- Department of General Surgery, Chinese PLA General Hospital, Beijing, China.
| |
Collapse
|