1
|
Kary AD, Noelle H, Magin CM. Tissue-Informed Biomaterial Innovations Advance Pulmonary Regenerative Engineering. ACS Macro Lett 2025; 14:434-447. [PMID: 40102038 DOI: 10.1021/acsmacrolett.5c00075] [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: 03/20/2025]
Abstract
Irreversible progressive pulmonary diseases drastically reduce the patient quality of life, while transplantation remains the only definitive cure. Research into lung regeneration pathways holds significant potential to expand and promote the discovery of new treatment options. Polymeric biomaterials designed to replicate key tissue characteristics (i.e., biochemical composition and mechanical cues) show promise for creating environments in which to study chronic lung diseases and initiate lung tissue regeneration. In this Viewpoint, we explore how naturally derived materials can be employed alone or combined with engineered polymer systems to create advanced tissue culture platforms. Pulmonary tissue models have historically leveraged natural materials, including basement membrane extracts and a decellularized extracellular matrix, as platforms for lung regeneration studies. Here, we provide an overview of the progression of pulmonary regenerative engineering, exploring how innovations in the growing field of tissue-informed biomaterials have the potential to advance lung regeneration research by bridging the gap between biological relevance and mechanical precision.
Collapse
Affiliation(s)
- Anton D Kary
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Haley Noelle
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
2
|
Rejeb M, Lahmar A, Ghedira MB, Selmi A, Kosksi T, Debbabi N, Ghedira LC. Fish and bovine collagen promote higher migration and adhesion of dermal cells pre-treated with wound-healing herbal extracts. Tissue Cell 2025; 93:102762. [PMID: 39919404 DOI: 10.1016/j.tice.2025.102762] [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: 11/08/2024] [Revised: 01/01/2025] [Accepted: 01/25/2025] [Indexed: 02/09/2025]
Abstract
PURPOSE Dermal cells fabricate and interact with the extracellular matrix to preserve structural integrity and further healthy function during wound healing. Collagen is a critical component of the matrix, challenging collagen's stability during wound injury. Natural sources especially plant extracts can promote wound healing and interact with collagen to increase its action. In this context, we studied the effect of extracted fish and bovine collagen in controlling cell proliferation, migration, and adhesion in dermal cells pretreated with plant extract. METHODS An acid-solubilization procedure was used to extract collagen fish (CF) and bovine (CB). Three different hydro-ethanolic extracts were prepared Pistacia lentiscus leaves (PL), Calendula officinalis leaves (FL), and flowers (FS). Migration potency was determined using scratch assay. The covered surface area was estimated after 16 hours and 24 hours after cell seeding. The chemotaxis was determined by the Boyden chamber, and the film was coated with CF or CB (10 µg/mL). or poly-L-lysine (50 µg/mL). FINDINGS We show that CF and CB increase adherence and migration of 3T3-L1 cells, which are pretreated with PL, FL, and FS. In addition, we highlighted a significantly higher cell adhesion on the CF matrix compared to CB. However, in the case of cells pre-treated with PL, the attachment to CF and CB increased significantly compared to untreated cells. The exposition of Hacat cells to plant extracts regulates the secretion of MMP2 and MMP and the production of reactive oxygen species. CONCLUSION CF and CB promote higher migration and adhesion of dermal cells pre-treated with wound-healing herbal extracts. In future studies, composite dressings based on collagen, P. lentiscus, and C. officinalis extracts can potentially be developed for tissue regeneration.
Collapse
Affiliation(s)
- Marwa Rejeb
- Research Laboratory, Bioactive Natural Products and Biotechnology LR24ES14, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne Street, Monastir 5000, Tunisia.
| | - Aida Lahmar
- Research Laboratory, Bioactive Natural Products and Biotechnology LR24ES14, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne Street, Monastir 5000, Tunisia.
| | - Mohamed Bayrem Ghedira
- Research Laboratory, Bioactive Natural Products and Biotechnology LR24ES14, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne Street, Monastir 5000, Tunisia
| | - Arem Selmi
- Research Laboratory, Bioactive Natural Products and Biotechnology LR24ES14, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne Street, Monastir 5000, Tunisia
| | - Tahsine Kosksi
- Research Laboratory, Bioactive Natural Products and Biotechnology LR24ES14, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne Street, Monastir 5000, Tunisia
| | - Nawres Debbabi
- Research Laboratory, Bioactive Natural Products and Biotechnology LR24ES14, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne Street, Monastir 5000, Tunisia
| | - Leila Chekir Ghedira
- Research Laboratory, Bioactive Natural Products and Biotechnology LR24ES14, Faculty of Dental Medicine of Monastir, University of Monastir, Avicenne Street, Monastir 5000, Tunisia
| |
Collapse
|
3
|
AYTAÇ S, ÖZBEY G. Photodiagnosis and photodynamic recognition of cervical cancer with SEM and AFM images. PLoS One 2025; 20:e0316544. [PMID: 39913376 PMCID: PMC11801595 DOI: 10.1371/journal.pone.0316544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 12/12/2024] [Indexed: 02/09/2025] Open
Abstract
So far, the number of patients who die from cancer is quite high. Continuation of early detection research is important to reduce the number of deaths due to cancer. At the time of the literature review, images of the same patients taken from Scanning Electron Microscope (SEM) and Atomic Force Microscope (AFM) for early diagnosis of cervix cancer have not been addressed to date. This article, Photodiagnosis and Photodynamics with SEM and AFM images are valuable in recognizing cervical cancer and starting treatment early. Simultaneous examination of the, Photodiagnosis and Photodynamics with SEM and AFM cervix images of patients will provide us with a far more powerful solution than a one-way solution. Daubechies (db2, db3, db4, and db5), Coiflet (coif5, coif4, coif3, and coif2), Symlet (sym5, sym4, sym3, and sym2), and Biorthogonal (bior1.3, bior2.8, bior1.5, and bior3.3) 16 discrete wavelet transformation families (DWTF) have been applied to AFM and SEM images. One approximate and three detail coefficients have been obtained for each one AFM and SEM cervix images. Homogeneity, contrast, angular second moment, entropy, mean, standard deviation, correlation, cluster prominence, dissimilarity, and cluster shade values have been calculated for each of these one approximate and three detail coefficients. The classification rate found by the averages of the results obtained from the DWTF_JSD, DWTF_HD and DWTF_TD algorithms for AFM and SEM cervix images are 98.29% and 97.10%, respectively. According to these results, it has been determined that SEM images have lower classification rate than AFM images. It has been also observed that the surface roughness of the mAFM images was larger than nAFM and bAFM images. But, it was observed that the volume of particles of the mAFM images has been smaller than nAFM and bAFM images.
Collapse
Affiliation(s)
- Sevcan AYTAÇ
- Department of Electronic Technology, Firat University, Elazığ, Turkey
| | - Gürkan ÖZBEY
- Department of Obstetrics and Gynecology, Private Anadolu Hosital, Elazığ, Turkey
| |
Collapse
|
4
|
Anandi L, Garcia J, Ros M, Janská L, Liu J, Carmona-Fontaine C. Direct visualization of emergent metastatic features within an ex vivo model of the tumor microenvironment. Life Sci Alliance 2025; 8:e202403053. [PMID: 39419548 PMCID: PMC11487089 DOI: 10.26508/lsa.202403053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Ischemic conditions such as hypoxia and nutrient starvation, together with interactions with stromal cells, are critical drivers of metastasis. These conditions arise deep within tumor tissues, and thus, observing nascent metastases is exceedingly challenging. We thus developed the 3MIC-an ex vivo model of the tumor microenvironment-to study the emergence of metastatic features in tumor cells in a 3-dimensional (3D) context. Here, tumor cells spontaneously create ischemic-like conditions, allowing us to study how tumor spheroids migrate, invade, and interact with stromal cells under different metabolic conditions. Consistent with previous data, we show that ischemia increases cell migration and invasion, but the 3MIC allowed us to directly observe and perturb cells while they acquire these pro-metastatic features. Interestingly, our results indicate that medium acidification is one of the strongest pro-metastatic cues and also illustrate using the 3MIC to test anti-metastatic drugs on cells experiencing different metabolic conditions. Overall, the 3MIC can help dissecting the complexity of the tumor microenvironment for the direct observation and perturbation of tumor cells during the early metastatic process.
Collapse
Affiliation(s)
- Libi Anandi
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Jeremy Garcia
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Manon Ros
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Libuše Janská
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Josephine Liu
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Carlos Carmona-Fontaine
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| |
Collapse
|
5
|
Wu KY, Dave A, Daigle P, Tran SD. Advanced Biomaterials for Lacrimal Tissue Engineering: A Review. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5425. [PMID: 39597252 PMCID: PMC11595815 DOI: 10.3390/ma17225425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024]
Abstract
The lacrimal gland (LG) is vital for ocular health, producing tears that lubricate and protect the eye. Dysfunction of the LG leads to aqueous-deficient dry eye disease (DED), significantly impacting quality of life. Current treatments mainly address symptoms rather than the underlying LG dysfunction, highlighting the need for regenerative therapies. Tissue engineering offers a promising solution, with biomaterials playing crucial roles in scaffolding and supporting cell growth for LG regeneration. This review focuses on recent advances in biomaterials used for tissue engineering of the lacrimal gland. We discuss both natural and synthetic biomaterials that mimic the extracellular matrix and provide structural support for cell proliferation and differentiation. Natural biomaterials, such as Matrigel, decellularized extracellular matrices, chitosan, silk fibroin hydrogels, and human amniotic membrane are evaluated for their biocompatibility and ability to support lacrimal gland cells. Synthetic biomaterials, like polyethersulfone, polyesters, and biodegradable polymers (PLLA and PLGA), are assessed for their mechanical properties and potential to create scaffolds that replicate the complex architecture of the LG. We also explore the integration of growth factors and stem cells with these biomaterials to enhance tissue regeneration. Challenges such as achieving proper vascularization, innervation, and long-term functionality of engineered tissues are discussed. Advances in 3D bioprinting and scaffold fabrication techniques are highlighted as promising avenues to overcome current limitations.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Archan Dave
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Patrick Daigle
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
6
|
Wu Y, Wang H, Qu C, Deng X, Li N, Yue S, Xu W, Chen Y, Zhou M. Pig-derived ECM-SIS provides a novel matrix gel for tumor modeling. Biomed Phys Eng Express 2024; 10:065002. [PMID: 39178888 DOI: 10.1088/2057-1976/ad72fa] [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: 03/21/2024] [Accepted: 08/23/2024] [Indexed: 08/26/2024]
Abstract
The absence of effective extracellular matrix to mimic the natural tumor microenvironment remains a significant obstacle in cancer research. Matrigel, abundant in various biological matrix components, is limited in its application due to its high cost. This has prompted researchers to explore alternative matrix substitutes. Here, we have investigated the effects of the extracellular matrix derived from pig small intestinal submucosa (ECM-SIS) in xenograft tumor modeling. Our results showed that the pig-derived ECM-SIS effectively promotes the establishment of xenograft tumor models, with a tumor formation rate comparable to that of Matrigel. Furthermore, we showed that the pig-derived ECM-SIS exhibited lower immune rejection and fewer infiltrating macrophages than Matrigel. Gene sequencing analysis demonstrated only a 0.5% difference in genes between pig-derived ECM-SIS and Matrigel during the process of tumor tissue formation. These differentially expressed genes primarily participate in cellular processes, biological regulation, and metabolic processes. These findings emphasize the potential of pig-derived ECM-SIS as a cost-effective option for tumor modeling in cancer research.
Collapse
Affiliation(s)
- Yanhua Wu
- BGI-Shenzhen, BGI.Research,-Shenzhen, 518110, Guangdong Province, People's Republic of China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518110, Guangdong Province, People's Republic of China
- Liver-biotechnology (Shenzhen) Co., ltd, Shenzhen, 518110, Guangdong Province, People's Republic of China
| | - Hao Wang
- BGI-Shenzhen, BGI.Research,-Shenzhen, 518110, Guangdong Province, People's Republic of China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518110, Guangdong Province, People's Republic of China
- Liver-biotechnology (Shenzhen) Co., ltd, Shenzhen, 518110, Guangdong Province, People's Republic of China
| | - Changbo Qu
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, Guangdong Province, People's Republic of China
| | - Xuesong Deng
- Department of Hepatobiliary Surgery, Shenzhen Second People's Hospital, First Affiliated Hospital of Shenzhen University, Shenzhen, 518035, Guangdong Province, People's Republic of China
| | - Na Li
- BGI-Shenzhen, BGI.Research,-Shenzhen, 518110, Guangdong Province, People's Republic of China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518110, Guangdong Province, People's Republic of China
- Liver-biotechnology (Shenzhen) Co., ltd, Shenzhen, 518110, Guangdong Province, People's Republic of China
| | - Sile Yue
- BGI-Shenzhen, BGI.Research,-Shenzhen, 518110, Guangdong Province, People's Republic of China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518110, Guangdong Province, People's Republic of China
- Liver-biotechnology (Shenzhen) Co., ltd, Shenzhen, 518110, Guangdong Province, People's Republic of China
| | - Wenjing Xu
- BGI-Shenzhen, BGI.Research,-Shenzhen, 518110, Guangdong Province, People's Republic of China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518110, Guangdong Province, People's Republic of China
- Liver-biotechnology (Shenzhen) Co., ltd, Shenzhen, 518110, Guangdong Province, People's Republic of China
| | - Yinghua Chen
- Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, Guangdong Province, People's Republic of China
| | - Ming Zhou
- BGI-Shenzhen, BGI.Research,-Shenzhen, 518110, Guangdong Province, People's Republic of China
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518110, Guangdong Province, People's Republic of China
- Liver-biotechnology (Shenzhen) Co., ltd, Shenzhen, 518110, Guangdong Province, People's Republic of China
| |
Collapse
|
7
|
Chen C, Boché A, Wang Z, Lopez E, Peng J, Carreiras F, Schanne-Klein MC, Chen Y, Lambert A, Aimé C. The Balance Between Shear Flow and Extracellular Matrix in Ovarian Cancer-on-Chip. Adv Healthc Mater 2024; 13:e2400938. [PMID: 38829702 DOI: 10.1002/adhm.202400938] [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: 03/12/2024] [Revised: 04/30/2024] [Indexed: 06/05/2024]
Abstract
Ovarian cancer is the most lethal gynecologic cancer in developed countries. In the tumor microenvironment, the extracellular matrix (ECM) and flow shear stress are key players in directing ovarian cancer cells invasion. Artificial ECM models based only on ECM proteins are used to build an ovarian tumor-on-chip to decipher the crosstalk between ECM and shear stress on the migratory behavior and cellular heterogeneity of ovarian tumor cells. This work shows that in the shear stress regime of the peritoneal cavity, the ECM plays a major role in driving individual or collective ovarian tumor cells migration. In the presence of basement membrane proteins, migration is more collective than on type I collagen regardless of shear stress. With increasing shear stress, individual cell migration is enhanced; while, no significant impact on collective migration is measured. This highlights the central position that ECM and flow shear stress should hold in in vitro ovarian cancer models to deepen understanding of cellular responses and improve development of ovarian cancer therapeutic platforms. In this frame, adding flow provides significant improvement in biological relevance over the authors' previous work. Further steps for enhanced clinical relevance require not only multiple cell lines but also patient-derived cells and sera.
Collapse
Affiliation(s)
- Changchong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Alphonse Boché
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Zixu Wang
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Elliot Lopez
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Juan Peng
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Franck Carreiras
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Marie-Claire Schanne-Klein
- Laboratoire d'Optique et Biosciences (LOB), École polytechnique, CNRS, Inserm, Institut Polytechnique de Paris, Palaiseau, F-91128, France
| | - Yong Chen
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| | - Ambroise Lambert
- Equipe de Recherche sur les Relations Matrice Extracellulaire-Cellules, ERRMECe (EA1391), Groupe Matrice Extracellulaire et physiopathologie (MECuP), Institut des Matériaux, I-MAT (FD4122), CY Cergy Paris Université, Cergy, 95000, France
| | - Carole Aimé
- PASTEUR, Département de chimie, École normale supérieure, PSL University, Sorbonne Université, CNRS, Paris, 75005, France
| |
Collapse
|
8
|
Blázquez-Carmona P, Ruiz-Mateos R, Barrasa-Fano J, Shapeti A, Martín-Alfonso JE, Domínguez J, Van Oosterwyck H, Reina-Romo E, Sanz-Herrera JA. Quantitative atlas of collagen hydrogels reveals mesenchymal cancer cell traction adaptation to the matrix nanoarchitecture. Acta Biomater 2024; 185:281-295. [PMID: 38992411 DOI: 10.1016/j.actbio.2024.07.002] [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: 02/26/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
Collagen-based hydrogels are commonly used in mechanobiology to mimic the extracellular matrix. A quantitative analysis of the influence of collagen concentration and properties on the structure and mechanics of the hydrogels is essential for tailored design adjustments for specific in vitro conditions. We combined focused ion beam scanning electron microscopy and rheology to provide a detailed quantitative atlas of the mechanical and nanoscale three-dimensional structural alterations that occur when manipulating different hydrogel's physicochemistry. Moreover, we study the effects of such alterations on the phenotype of breast cancer cells and their mechanical interactions with the extracellular matrix. Regardless of the microenvironment's pore size, porosity or mechanical properties, cancer cells are able to reach a stable mesenchymal-like morphology. Additionally, employing 3D traction force microscopy, a positive correlation between cellular tractions and ECM mechanics is observed up to a critical threshold, beyond which tractions plateau. This suggests that cancer cells in a stable mesenchymal state calibrate their mechanical interactions with the ECM to keep their migration and invasiveness capacities unaltered. STATEMENT OF SIGNIFICANCE: The paper presents a thorough study on the mechanical microenvironment in breast cancer cells during their interaction with collagen based hydrogels of different compositions. The hydrogels' microstructure were obtained using state-of-the-art 3D microscopy, namely focused ion beam-scanning electron microscope (FIB-SEM). FIB-SEM was originally applied in this work to reconstruct complex fibered collagen microstructures within the nanometer range, to obtain key microarchitectural parameters. The mechanical microenvironment of cells was recovered using Traction Force Microscopy (TFM). The obtained results suggest that cells calibrate tractions such that they depend on mechanical, microstructural and physicochemical characteristics of the hydrogels, hence revealing a steric hindrance. We hypothesize that cancer cells studied in this paper tune their mechanical state to keep their migration and invasiveness capacities unaltered.
Collapse
Affiliation(s)
- Pablo Blázquez-Carmona
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Raquel Ruiz-Mateos
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Jorge Barrasa-Fano
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - Apeksha Shapeti
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - José Enrique Martín-Alfonso
- Escuela Técnica Superior de Ingeniería, Universidad de Huelva. Avda. de las Fuerzas Armadas s/n, 21007 Huelva, Spain
| | - Jaime Domínguez
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - Hans Van Oosterwyck
- Department of Mechanical Engineering, Biomechanics Section, KU Leuven, Celestijnenlaan 300. B-3001 Heverlee, Belgium
| | - Esther Reina-Romo
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain
| | - José Antonio Sanz-Herrera
- Escuela Técnica Superior de Ingeniería, Universidad de Sevilla. Avenida Camino de los Descubrimientos s/n, 41092 Sevilla, Spain; Instituto de Biomedicina de Sevilla (IBIS). C. Antonio Maura Montaner, 41013 Sevilla, Spain.
| |
Collapse
|
9
|
Garreta E, Moya-Rull D, Marco A, Amato G, Ullate-Agote A, Tarantino C, Gallo M, Esporrín-Ubieto D, Centeno A, Vilas-Zornoza A, Mestre R, Kalil M, Gorroñogoitia I, Zaldua AM, Sanchez S, Izquierdo Reyes L, Fernández-Santos ME, Prosper F, Montserrat N. Natural Hydrogels Support Kidney Organoid Generation and Promote In Vitro Angiogenesis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400306. [PMID: 38762768 DOI: 10.1002/adma.202400306] [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: 01/07/2024] [Revised: 05/14/2024] [Indexed: 05/20/2024]
Abstract
To date, strategies aiming to modulate cell to extracellular matrix (ECM) interactions during organoid derivation remain largely unexplored. Here renal decellularized ECM (dECM) hydrogels are fabricated from porcine and human renal cortex as biomaterials to enrich cell-to-ECM crosstalk during the onset of kidney organoid differentiation from human pluripotent stem cells (hPSCs). Renal dECM-derived hydrogels are used in combination with hPSC-derived renal progenitor cells to define new approaches for 2D and 3D kidney organoid differentiation, demonstrating that in the presence of these biomaterials the resulting kidney organoids exhibit renal differentiation features and the formation of an endogenous vascular component. Based on these observations, a new method to produce kidney organoids with vascular-like structures is achieved through the assembly of hPSC-derived endothelial-like organoids with kidney organoids in 3D. Major readouts of kidney differentiation and renal cell morphology are assessed exploiting these culture platforms as new models of nephrogenesis. Overall, this work shows that exploiting cell-to-ECM interactions during the onset of kidney differentiation from hPSCs facilitates and optimizes current approaches for kidney organoid derivation thereby increasing the utility of these unique cell culture platforms for personalized medicine.
Collapse
Affiliation(s)
- Elena Garreta
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, 08028, Spain
- University of Barcelona, Barcelona, 08028, Spain
| | - Daniel Moya-Rull
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, 08028, Spain
| | - Andrés Marco
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, 08028, Spain
| | - Gaia Amato
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, 08028, Spain
| | - Asier Ullate-Agote
- Regenerative Medicine Program, Centre for Applied Medical Research (CIMA), Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, 31008, Spain
| | - Carolina Tarantino
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, 08028, Spain
| | - Maria Gallo
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, 08028, Spain
| | - David Esporrín-Ubieto
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, Barcelona, 08028, Spain
| | - Alberto Centeno
- Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario A Coruña (CHUAC), Sergas, Universidade da Coruña (UDC), As Xubias, A Coruña, 15006, Spain
| | - Amaia Vilas-Zornoza
- Regenerative Medicine Program, Centre for Applied Medical Research (CIMA), Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Pamplona, 31008, Spain
| | - Rafael Mestre
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, Barcelona, 08028, Spain
| | - María Kalil
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, 08028, Spain
| | | | - Ane Miren Zaldua
- Leartiker S. Coop, Xemein Etorbidea 12A, Markina-Xemein, 48270, Spain
| | - Samuel Sanchez
- Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 10-12, Barcelona, 08028, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Passeig de Lluís Companys 23, Barcelona, 08010, Spain
| | | | - María Eugenia Fernández-Santos
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Madrid, 28009, Spain
- ATMPs Production Unit, Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, 28009, Spain
| | - Felipe Prosper
- Hematology Service and Cell Therapy Unit and Program of Hematology-Oncology CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN) and Instituto de Investigación Sanitaria de Navarra (IdISNA), Pamplona, 31008, Spain
- Centro de Investigación Biomedica en Red de Oncología (CIBERONC) and RICORS TERAV, Madrid, 28029, Spain
| | - Nuria Montserrat
- Pluripotency for Organ Regeneration, Institute for Bioengineering of Catalonia (IBEC), Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri i Reixac, 15-21, Barcelona, 08028, Spain
- Catalan Institute for Research and Advanced Studies (ICREA), Passeig de Lluís Companys 23, Barcelona, 08010, Spain
| |
Collapse
|
10
|
Asim S, Hayhurst E, Callaghan R, Rizwan M. Ultra-low content physio-chemically crosslinked gelatin hydrogel improves encapsulated 3D cell culture. Int J Biol Macromol 2024; 264:130657. [PMID: 38458282 PMCID: PMC11003839 DOI: 10.1016/j.ijbiomac.2024.130657] [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: 08/09/2023] [Revised: 02/24/2024] [Accepted: 03/04/2024] [Indexed: 03/10/2024]
Abstract
Gelatin-based hydrogels are extensively used for 3D cell culture, bioprinting, and tissue engineering due to their cell-adhesive nature and tunable physio-chemical properties. Gelatin hydrogels for 3D cell culture are often developed using high-gelatin content (frequently 10-15 % w/v) to ensure fast gelation and improved stability. While highly stable, such matrices restrict the growth of encapsulated cells due to creating a dense, restrictive environment around the encapsulated cells. Hydrogels with lower polymer content are known to improve 3D cell growth, yet fabrication of ultra-low concentration gelatin hydrogels is challenging while ensuring fast gelation and stability. Here, we demonstrate that physical gelation and photo-crosslinking in gelatin results in a fast-gelling hydrogel at a remarkably low gelatin concentration of 1 % w/v (GelPhy/Photo). The GelPhy/Photo hydrogel was highly stable, allowed uniform 3D distribution of cells, and significantly improved the spreading of encapsulated 3T3 fibroblast cells. Moreover, human cholangiocarcinoma (HuCCT-1) cells encapsulated in 1 % GelPhy/Photo matrix grew and self-assembled into epithelial cysts with lumen, which could not be achieved in a traditional high-concentration gelatin hydrogel. These findings pave the way to significantly improve existing gelatin hydrogels for 3D cell culture applications.
Collapse
Affiliation(s)
- Saad Asim
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Emma Hayhurst
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Rachel Callaghan
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI 49931, USA; Health Research Institute (HRI), Michigan Technological University, USA.
| |
Collapse
|
11
|
Tahara S, Sharma S, de Faria FCC, Sarchet P, Tomasello L, Rentsch S, Karna R, Calore F, Pollock RE. Comparison of three-dimensional cell culture techniques of dedifferentiated liposarcoma and their integration with future research. Front Cell Dev Biol 2024; 12:1362696. [PMID: 38500686 PMCID: PMC10945377 DOI: 10.3389/fcell.2024.1362696] [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: 12/28/2023] [Accepted: 02/13/2024] [Indexed: 03/20/2024] Open
Abstract
Background: Dedifferentiated liposarcoma is a formidable sarcoma subtype due to its high local recurrence rate and resistance to medical treatment. While 2D cell cultures are still commonly used, 3D cell culture systems have emerged as a promising alternative, particularly scaffold-based techniques that enable the creation of 3D models with more accurate cell-stroma interactions. Objective: To investigate how 3D structures with or without the scaffold existence would affect liposarcoma cell lines growth morphologically and biologically. Methods: Lipo246 and Lipo863 cell lines were cultured in 3D using four different methods; Matrigel® ECM scaffold method, Collagen ECM scaffold method, ULA plate method and Hanging drop method, in addition to conventional 2D cell culture methods. All samples were processed for histopathological analysis (HE, IHC and DNAscope™), Western blot, and qPCR; moreover, 3D collagen-based models were treated with different doses of SAR405838, a well-known inhibitor of MDM2, and cell viability was assessed in comparison to 2D model drug response. Results: Regarding morphology, cell lines behaved differently comparing the scaffold-based and scaffold-free methods. Lipo863 formed spheroids in Matrigel® but not in collagen, while Lipo246 did not form spheroids in either collagen or Matrigel®. On the other hand, both cell lines formed spheroids using scaffold-free methods. All samples retained liposarcoma characteristic, such as high level of MDM2 protein expression and MDM2 DNA amplification after being cultivated in 3D. 3D collagen samples showed higher cell viability after SAR40538 treatment than 2D models, while cells sensitive to the drug died by apoptosis or necrosis. Conclusion: Our results prompt us to extend our investigation by applying our 3D models to further oncological relevant applications, which may help address unresolved questions about dedifferentiated liposarcoma biology.
Collapse
Affiliation(s)
- Sayumi Tahara
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Soumya Sharma
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Fernanda Costas Casal de Faria
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Patricia Sarchet
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Luisa Tomasello
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Sydney Rentsch
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Roma Karna
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Federica Calore
- Department of Cancer Biology and Genetics, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Raphael E. Pollock
- Department of Surgery, Division of Surgical Oncology, The James Comprehensive Cancer Center, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| |
Collapse
|
12
|
Park S, Laskow TC, Chen J, Guha P, Dawn B, Kim D. Microphysiological systems for human aging research. Aging Cell 2024; 23:e14070. [PMID: 38180277 PMCID: PMC10928588 DOI: 10.1111/acel.14070] [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: 06/30/2023] [Revised: 11/28/2023] [Accepted: 11/30/2023] [Indexed: 01/06/2024] Open
Abstract
Recent advances in microphysiological systems (MPS), also known as organs-on-a-chip (OoC), enable the recapitulation of more complex organ and tissue functions on a smaller scale in vitro. MPS therefore provide the potential to better understand human diseases and physiology. To date, numerous MPS platforms have been developed for various tissues and organs, including the heart, liver, kidney, blood vessels, muscle, and adipose tissue. However, only a few studies have explored using MPS platforms to unravel the effects of aging on human physiology and the pathogenesis of age-related diseases. Age is one of the risk factors for many diseases, and enormous interest has been devoted to aging research. As such, a human MPS aging model could provide a more predictive tool to understand the molecular and cellular mechanisms underlying human aging and age-related diseases. These models can also be used to evaluate preclinical drugs for age-related diseases and translate them into clinical settings. Here, we provide a review on the application of MPS in aging research. First, we offer an overview of the molecular, cellular, and physiological changes with age in several tissues or organs. Next, we discuss previous aging models and the current state of MPS for studying human aging and age-related conditions. Lastly, we address the limitations of current MPS and present future directions on the potential of MPS platforms for human aging research.
Collapse
Affiliation(s)
- Seungman Park
- Department of Mechanical EngineeringUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Thomas C. Laskow
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jingchun Chen
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Prasun Guha
- Nevada Institute of Personalized MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
- School of Life SciencesUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Buddhadeb Dawn
- Department of Internal Medicine, Kirk Kerkorian School of MedicineUniversity of Nevada, Las VegasLas VegasNevadaUSA
| | - Deok‐Ho Kim
- Department of MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical EngineeringJohns Hopkins UniversityBaltimoreMarylandUSA
- Center for Microphysiological SystemsJohns Hopkins UniversityBaltimoreMarylandUSA
| |
Collapse
|
13
|
Mohammadimatin P, Parvin P, Jafargholi A, Jahanbakhshi A, Ahmadinouri F, Tabibkhooei A, Heidari O, Salarinejad S. Signal enhancement in spark-assisted laser-induced breakdown spectroscopy for discrimination of glioblastoma and oligodendroglioma lesions. BIOMEDICAL OPTICS EXPRESS 2023; 14:5795-5816. [PMID: 38021132 PMCID: PMC10659799 DOI: 10.1364/boe.497234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/16/2023] [Accepted: 09/18/2023] [Indexed: 12/01/2023]
Abstract
Here, the discrimination of two types of lethal brain cancers, i.e., glioblastoma multiforme (GBM) and oligodendroglioma (OG) are investigated under the laser-induced breakdown spectroscopy (LIBS) and the electrical spark-assisted laser-induced breakdown spectroscopy (SA-LIBS) in order to discriminate the human brain glioma lesions against the infiltrated tissues. It is shown there are notable differences between the plasma emissions over the brain gliomas against those of infiltrated tissues. In fact, a notable enhancement appears in the characteristic emissions in favor of SA-LIBS against those of conventional LIB spectra. Moreover, the plasma properties such as temperature, electron density, and degree of ionization are probed through the data processing of the plasma emissions. The corresponding parameters, taken from SA-LIBS data, attest to be lucidly larger than those of LIBS up to one order of magnitude. In addition, the ionic species such as Mg II characteristic line at 279 nm and caII emission at 393 nm are notably enhanced in favor of SA-LIBS. In general, the experimental evidence verifies that SA-LIBS is beneficial in the discrimination and grading of GBM/OG neoplasia against healthy (infiltrate) tissues in the early stages.
Collapse
Affiliation(s)
- Parisa Mohammadimatin
- Department of Physics and Energy
Engineering, Amirkabir University of Technology, P.O. Box 15875-4413, Tehran, Iran
| | - Parviz Parvin
- Department of Physics and Energy
Engineering, Amirkabir University of Technology, P.O. Box 15875-4413, Tehran, Iran
| | - Amir Jafargholi
- Department of Electronic and Electrical
Engineering, University College London
(UCL), United
Kingdom
| | - Amin Jahanbakhshi
- Stem Cell and Regenerative Medicine
Research Center, Iran University of Medical
Sciences, P.O. Box, 1997667665, Tehran, Iran
| | - Fatemeh Ahmadinouri
- Department of Physics and Energy
Engineering, Amirkabir University of Technology, P.O. Box 15875-4413, Tehran, Iran
| | - Alireza Tabibkhooei
- Skull Base Research Center, Department of
Neurosurgery, Iran University of Medical
Sciences, P.O. Box, 1997667665, Tehran, Iran
| | - Omid Heidari
- Department of Physics and Energy
Engineering, Amirkabir University of Technology, P.O. Box 15875-4413, Tehran, Iran
| | - Sareh Salarinejad
- Shohada-e-Tajrish Hospital, Department of
Pathology, Faculty of Medicine, Shahid Beheshti
University of Medical Sciences, P.O. box 1985717443,
Tehran, Iran
| |
Collapse
|
14
|
Lipreri MV, Di Pompo G, Boanini E, Graziani G, Sassoni E, Baldini N, Avnet S. Bone on-a-chip: a 3D dendritic network in a screening platform for osteocyte-targeted drugs. Biofabrication 2023; 15:045019. [PMID: 37552982 DOI: 10.1088/1758-5090/acee23] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 08/08/2023] [Indexed: 08/10/2023]
Abstract
Age-related musculoskeletal disorders, including osteoporosis, are frequent and associated with long lasting morbidity, in turn significantly impacting on healthcare system sustainability. There is therefore a compelling need to develop reliable preclinical models of disease and drug screening to validate novel drugs possibly on a personalized basis, without the need ofin vivoassay. In the context of bone tissue, although the osteocyte (Oc) network is a well-recognized therapeutic target, currentin vitropreclinical models are unable to mimic its physiologically relevant and highly complex structure. To this purpose, several features are needed, including an osteomimetic extracellular matrix, dynamic perfusion, and mechanical cues (e.g. shear stress) combined with a three-dimensional (3D) culture of Oc. Here we describe, for the first time, a high throughput microfluidic platform based on 96-miniaturized chips for large-scale preclinical evaluation to predict drug efficacy. We bioengineered a commercial microfluidic device that allows real-time visualization and equipped with multi-chips by the development and injection of a highly stiff bone-like 3D matrix, made of a blend of collagen-enriched natural hydrogels loaded with hydroxyapatite nanocrystals. The microchannel, filled with the ostemimetic matrix and Oc, is subjected to passive perfusion and shear stress. We used scanning electron microscopy for preliminary material characterization. Confocal microscopy and fluorescent microbeads were used after material injection into the microchannels to detect volume changes and the distribution of cell-sized objects within the hydrogel. The formation of a 3D dendritic network of Oc was monitored by measuring cell viability, evaluating phenotyping markers (connexin43, integrin alpha V/CD51, sclerostin), quantification of dendrites, and responsiveness to an anabolic drug. The platform is expected to accelerate the development of new drug aimed at modulating the survival and function of osteocytes.
Collapse
Affiliation(s)
| | - Gemma Di Pompo
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Elisa Boanini
- Department of Chemistry 'Giacomo Ciamician', University of Bologna, Bologna, Italy
| | - Gabriela Graziani
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Enrico Sassoni
- Department of Civil, Chemical, Environmental and Materials Engineering, University of Bologna, Bologna, Italy
| | - Nicola Baldini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Biomedical Science, Technologies, and Nanobiotecnologiy Lab, IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Sofia Avnet
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| |
Collapse
|
15
|
Bakar AJBA, Azam NSM, Sevakumaran V, Ismail WIBW, Razali MH, Razak SIBA, Amin KAM. Effectiveness of collagen and gatifloxacin in improving the healing and antibacterial activities of gellan gum hydrogel films as dressing materials. Int J Biol Macromol 2023; 245:125494. [PMID: 37348586 DOI: 10.1016/j.ijbiomac.2023.125494] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 06/04/2023] [Accepted: 06/18/2023] [Indexed: 06/24/2023]
Abstract
The demand for advanced wound care products is rapidly increasing nowadays. In this study, gellan gum/collagen (GG/C) hydrogel films containing gatifloxacin (GAT) were developed to investigate their properties as wound dressing materials. ATR-FTIR, swelling, water content, water vapor transmission rate (WVTR), and thermal properties were investigated. The mechanical properties of the materials were tested in dry and wet conditions to understand the performance of the materials after exposure to wound exudate. Drug release by Franz diffusion was measured with all samples showing 100 % cumulative drug release after 40 min. Strong antibacterial activities against Staphylococcus aureus and Staphylococcus epidermis were observed for Gram-positive bacteria, while Escherichia coli and Pseudomonas aeruginosa were observed for Gram-negative bacteria. The in-vivo cytotoxicity of GG/C-GAT was assessed by wound contraction in rats, which was 95 % for GG/C-GAT01. Hematoxylin and eosin and Masson's trichrome staining revealed the appearance of fresh full epidermis and granulation tissue, indicating that all wounds had passed through the proliferation phase. The results demonstrate the promising properties of the materials to be used as dressing materials.
Collapse
Affiliation(s)
| | - Nurul Shahirah Mohd Azam
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030, Terengganu, Malaysia
| | - Vigneswari Sevakumaran
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030, Terengganu, Malaysia
| | - Wan Iryani Bt Wan Ismail
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030, Terengganu, Malaysia
| | - Mohd Hasmizam Razali
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030, Terengganu, Malaysia
| | - Saiful Izwan Bin Abdul Razak
- BioInspired Device and Tissue Engineering Research Group, Faculty of Engineering, School of Biomedical Engineering and Health Sciences, Universiti Teknologi Malaysia, Skudai 81300, Johor, Malaysia
| | - Khairul Anuar Mat Amin
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, 21030, Terengganu, Malaysia.
| |
Collapse
|
16
|
Ellis K, Wood R. The Comparative Invasiveness of Endometriotic Cell Lines to Breast and Endometrial Cancer Cell Lines. Biomolecules 2023; 13:1003. [PMID: 37371583 DOI: 10.3390/biom13061003] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 06/08/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
Endometriosis is an invasive condition that affects 10% of women (and people assigned as female at birth) worldwide. The purpose of this study was to characterize the relative invasiveness of three available endometriotic cell lines (EEC12Z, iEc-ESCs, tHESCs) to cancer cell lines (MDA-MB-231, SW1353 and EM-E6/E7/TERT) and assess whether the relative invasiveness was consistent across different invasion assays. All cell lines were subjected to transwell, spheroid drop, and spheroid-gel invasion assays, and stained for vimentin, cytokeratin, E-Cadherin and N-Cadherin to assess changes in expression. In all assays, endometriotic cell lines showed comparable invasiveness to the cancer cell lines used in this study, with no significant differences in invasiveness identified. EEC12Z cells that had invaded within the assay periods showed declines in E-Cadherin expression compared to cells that had not invaded within the assay period, without significant changes in N-Cadherin expression, which may support the hypothesis that an epithelial-to-mesenchymal transition is an influence on the invasiveness shown by this peritoneal endometriosis cell line.
Collapse
Affiliation(s)
- Katherine Ellis
- Department of Chemical and Process Engineering, University of Canterbury, Christchurch 8041, New Zealand
- Endometriosis New Zealand, Christchurch 8041, New Zealand
| | - Rachael Wood
- Department of Chemical and Process Engineering, University of Canterbury, Christchurch 8041, New Zealand
- The Biomolecular Interaction Centre, University of Canterbury, Christchurch 8041, New Zealand
| |
Collapse
|
17
|
Rajendran AK, Sankar D, Amirthalingam S, Kim HD, Rangasamy J, Hwang NS. Trends in mechanobiology guided tissue engineering and tools to study cell-substrate interactions: a brief review. Biomater Res 2023; 27:55. [PMID: 37264479 DOI: 10.1186/s40824-023-00393-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
Sensing the mechanical properties of the substrates or the matrix by the cells and the tissues, the subsequent downstream responses at the cellular, nuclear and epigenetic levels and the outcomes are beginning to get unraveled more recently. There have been various instances where researchers have established the underlying connection between the cellular mechanosignalling pathways and cellular physiology, cellular differentiation, and also tissue pathology. It has been now accepted that mechanosignalling, alone or in combination with classical pathways, could play a significant role in fate determination, development, and organization of cells and tissues. Furthermore, as mechanobiology is gaining traction, so do the various techniques to ponder and gain insights into the still unraveled pathways. This review would briefly discuss some of the interesting works wherein it has been shown that specific alteration of the mechanical properties of the substrates would lead to fate determination of stem cells into various differentiated cells such as osteoblasts, adipocytes, tenocytes, cardiomyocytes, and neurons, and how these properties are being utilized for the development of organoids. This review would also cover various techniques that have been developed and employed to explore the effects of mechanosignalling, including imaging of mechanosensing proteins, atomic force microscopy (AFM), quartz crystal microbalance with dissipation measurements (QCMD), traction force microscopy (TFM), microdevice arrays, Spatio-temporal image analysis, optical tweezer force measurements, mechanoscanning ion conductance microscopy (mSICM), acoustofluidic interferometric device (AID) and so forth. This review would provide insights to the researchers who work on exploiting various mechanical properties of substrates to control the cellular and tissue functions for tissue engineering and regenerative applications, and also will shed light on the advancements of various techniques that could be utilized to unravel the unknown in the field of cellular mechanobiology.
Collapse
Affiliation(s)
- Arun Kumar Rajendran
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
| | - Deepthi Sankar
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Sivashanmugam Amirthalingam
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hwan D Kim
- Department of Polymer Science and Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
- Department of Biomedical Engineering, Korea National University of Transportation, Chungju, 27469, Republic of Korea
| | - Jayakumar Rangasamy
- Polymeric Biomaterials Lab, School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| | - Nathaniel S Hwang
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul, 08826, Republic of Korea.
- Institute of Engineering Research, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul, 08826, Republic of Korea.
- Bio-MAX/N-Bio Institute, Institute of Bio-Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
18
|
Raskov H, Gaggar S, Tajik A, Orhan A, Gögenur I. The Matrix Reloaded-The Role of the Extracellular Matrix in Cancer. Cancers (Basel) 2023; 15:2057. [PMID: 37046716 PMCID: PMC10093330 DOI: 10.3390/cancers15072057] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023] Open
Abstract
As the core component of all organs, the extracellular matrix (ECM) is an interlocking macromolecular meshwork of proteins, glycoproteins, and proteoglycans that provides mechanical support to cells and tissues. In cancer, the ECM can be remodelled in response to environmental cues, and it controls a plethora of cellular functions, including metabolism, cell polarity, migration, and proliferation, to sustain and support oncogenesis. The biophysical and biochemical properties of the ECM, such as its structural arrangement and being a reservoir for bioactive molecules, control several intra- and intercellular signalling pathways and induce cytoskeletal changes that alter cell shapes, behaviour, and viability. Desmoplasia is a major component of solid tumours. The abnormal deposition and composition of the tumour matrix lead to biochemical and biomechanical alterations that determine disease development and resistance to treatment. This review summarises the complex roles of ECM in cancer and highlights the possible therapeutic targets and how to potentially remodel the dysregulated ECM in the future. Furthering our understanding of the ECM in cancer is important as the modification of the ECM will probably become an important tool in the characterisation of individual tumours and personalised treatment options.
Collapse
Affiliation(s)
- Hans Raskov
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Shruti Gaggar
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Asma Tajik
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
| | - Adile Orhan
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Oncology, Zealand University Hospital, 4000 Roskilde, Denmark
| | - Ismail Gögenur
- Center for Surgical Science, Zealand University Hospital, Lykkebækvej 1, 4600 Køge, Denmark
- Department of Clinical Medicine, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
19
|
Abedin MJ, Michelhaugh SK, Mittal S, Berdichevsky Y. 3D models of glioblastoma interaction with cortical cells. Front Bioeng Biotechnol 2023; 11:1150772. [PMID: 36970613 PMCID: PMC10033518 DOI: 10.3389/fbioe.2023.1150772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Introduction: Glioblastoma (GBM) invasiveness and ability to infiltrate deep into the brain tissue is a major reason for the poor patient prognosis for this type of brain cancer. Behavior of glioblastoma cells, including their motility, and expression of invasion-promoting genes such as matrix metalloprotease-2 (MMP2), are strongly influenced by normal cells found in the brain parenchyma. Cells such as neurons may also be influenced by the tumor, as many glioblastoma patients develop epilepsy. In vitro models of glioblastoma invasiveness are used to supplement animal models in a search for better treatments, and need to combine capability for high-throughput experiments with capturing bidirectional interactions between GBM and brain cells.Methods: In this work, two 3D in vitro models of GBM-cortical interactions were investigated. A matrix-free model was created by co-culturing GBM and cortical spheroids, and a matrix-based model was created by embedding cortical cells and a GBM spheroid in Matrigel.Results: Rapid GBM invasion occurred in the matrix-based model, and was enhanced by the presence of cortical cells. Little invasion occurred in the matrix-free model. In both types of models, presence of GBM cells resulted in a significant increase in paroxysmal neuronal activity.Discussion: Matrix-based model may be better suited for studying GBM invasion in an environment that includes cortical cells, while matrix-free model may be useful in investigation of tumor-associated epilepsy.
Collapse
Affiliation(s)
- Md Joynal Abedin
- Department of Bioengineering, Lehigh University, Bethlehem, PA, United States
| | | | - Sandeep Mittal
- Fralin Biomedical Research Institute, Virginia Tech, Roanoke, VA, United States
| | - Yevgeny Berdichevsky
- Department of Bioengineering, Lehigh University, Bethlehem, PA, United States
- Department of Electrical and Computer Engineering, Bethlehem, PA, United States
- *Correspondence: Yevgeny Berdichevsky,
| |
Collapse
|
20
|
Merino-Casallo F, Gomez-Benito MJ, Hervas-Raluy S, Garcia-Aznar JM. Unravelling cell migration: defining movement from the cell surface. Cell Adh Migr 2022; 16:25-64. [PMID: 35499121 PMCID: PMC9067518 DOI: 10.1080/19336918.2022.2055520] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Accepted: 03/10/2022] [Indexed: 12/13/2022] Open
Abstract
Cell motility is essential for life and development. Unfortunately, cell migration is also linked to several pathological processes, such as cancer metastasis. Cells' ability to migrate relies on many actors. Cells change their migratory strategy based on their phenotype and the properties of the surrounding microenvironment. Cell migration is, therefore, an extremely complex phenomenon. Researchers have investigated cell motility for more than a century. Recent discoveries have uncovered some of the mysteries associated with the mechanisms involved in cell migration, such as intracellular signaling and cell mechanics. These findings involve different players, including transmembrane receptors, adhesive complexes, cytoskeletal components , the nucleus, and the extracellular matrix. This review aims to give a global overview of our current understanding of cell migration.
Collapse
Affiliation(s)
- Francisco Merino-Casallo
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Maria Jose Gomez-Benito
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Silvia Hervas-Raluy
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| | - Jose Manuel Garcia-Aznar
- Multiscale in Mechanical and Biological Engineering (M2BE), Aragon Institute of Engineering Research (I3A), Zaragoza, Spain
- Department of Mechanical Engineering, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
21
|
Nascentes Melo LM, Lesner NP, Sabatier M, Ubellacker JM, Tasdogan A. Emerging metabolomic tools to study cancer metastasis. Trends Cancer 2022; 8:988-1001. [PMID: 35909026 DOI: 10.1016/j.trecan.2022.07.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/24/2022] [Accepted: 07/06/2022] [Indexed: 12/24/2022]
Abstract
Metastasis is responsible for 90% of deaths in patients with cancer. Understanding the role of metabolism during metastasis has been limited by the development of robust and sensitive technologies that capture metabolic processes in metastasizing cancer cells. We discuss the current technologies available to study (i) metabolism in primary and metastatic cancer cells and (ii) metabolic interactions between cancer cells and the tumor microenvironment (TME) at different stages of the metastatic cascade. We identify advantages and disadvantages of each method and discuss how these tools and technologies will further improve our understanding of metastasis. Studies investigating the complex metabolic rewiring of different cells using state-of-the-art metabolomic technologies have the potential to reveal novel biological processes and therapeutic interventions for human cancers.
Collapse
Affiliation(s)
| | - Nicholas P Lesner
- Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Marie Sabatier
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Jessalyn M Ubellacker
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
| | - Alpaslan Tasdogan
- Department of Dermatology, University Hospital Essen and German Cancer Consortium, Partner Site, Essen, Germany.
| |
Collapse
|
22
|
Krysko DV, Demuynck R, Efimova I, Naessens F, Krysko O, Catanzaro E. In Vitro Veritas: From 2D Cultures to Organ-on-a-Chip Models to Study Immunogenic Cell Death in the Tumor Microenvironment. Cells 2022; 11:3705. [PMID: 36429133 PMCID: PMC9688238 DOI: 10.3390/cells11223705] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 11/16/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
Abstract
Immunogenic cell death (ICD) is a functionally unique form of cell death that promotes a T-cell-dependent anti-tumor immune response specific to antigens originating from dying cancer cells. Many anticancer agents and strategies induce ICD, but despite their robust effects in vitro and in vivo on mice, translation into the clinic remains challenging. A major hindrance in antitumor research is the poor predictive ability of classic 2D in vitro models, which do not consider tumor biological complexity, such as the contribution of the tumor microenvironment (TME), which plays a crucial role in immunosuppression and cancer evasion. In this review, we describe different tumor models, from 2D cultures to organ-on-a-chip technology, as well as spheroids and perfusion bioreactors, all of which mimic the different degrees of the TME complexity. Next, we discuss how 3D cell cultures can be applied to study ICD and how to increase the translational potential of the ICD inducers. Finally, novel research directions are provided regarding ICD in the 3D cellular context which may lead to novel immunotherapies for cancer.
Collapse
Affiliation(s)
- Dmitri V. Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Robin Demuynck
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Iuliia Efimova
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Faye Naessens
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| | - Olga Krysko
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod 603022, Russia
| | - Elena Catanzaro
- Cell Death Investigation and Therapy Laboratory, Department of Human Structure and Repair, Ghent University, 9000 Ghent, Belgium
- Cancer Research Institute Ghent, 9000 Ghent, Belgium
| |
Collapse
|
23
|
Divya G, Madhura R, Khetan V, Rishi P, Narayanan J. Understanding the mechano and chemo response of retinoblastoma tumor cells. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
24
|
Vasudevan J, Jiang K, Fernandez J, Lim CT. Extracellular matrix mechanobiology in cancer cell migration. Acta Biomater 2022; 163:351-364. [PMID: 36243367 DOI: 10.1016/j.actbio.2022.10.016] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 09/11/2022] [Accepted: 10/06/2022] [Indexed: 11/01/2022]
Abstract
The extracellular matrix (ECM) is pivotal in modulating tumor progression. Besides chemically stimulating tumor cells, it also offers physical support that orchestrates the sequence of events in the metastatic cascade upon dynamically modulating cell mechanosensation. Understanding this translation between matrix biophysical cues and intracellular signaling has led to rapid growth in the interdisciplinary field of cancer mechanobiology in the last decade. Substantial efforts have been made to develop novel in vitro tumor mimicking platforms to visualize and quantify the mechanical forces within the tissue that dictate tumor cell invasion and metastatic growth. This review highlights recent findings on tumor matrix biophysical cues such as fibrillar arrangement, crosslinking density, confinement, rigidity, topography, and non-linear mechanics and their implications on tumor cell behavior. We also emphasize how perturbations in these cues alter cellular mechanisms of mechanotransduction, consequently enhancing malignancy. Finally, we elucidate engineering techniques to individually emulate the mechanical properties of tumors that could help serve as toolkits for developing and testing ECM-targeted therapeutics on novel bioengineered tumor platforms. STATEMENT OF SIGNIFICANCE: Disrupted ECM mechanics is a driving force for transitioning incipient cells to life-threatening malignant variants. Understanding these ECM changes can be crucial as they may aid in developing several efficacious drugs that not only focus on inducing cytotoxic effects but also target specific matrix mechanical cues that support and enhance tumor invasiveness. Designing and implementing an optimal tumor mimic can allow us to predictively map biophysical cue-modulated cell behaviors and facilitate the design of improved lab-grown tumor models with accurately controlled structural features. This review focuses on the abnormal changes within the ECM during tumorigenesis and its implications on tumor cell-matrix mechanoreciprocity. Additionally, it accentuates engineering approaches to produce ECM features of varying levels of complexity which is critical for improving the efficiency of current engineered tumor tissue models.
Collapse
|
25
|
Lekka M. Applicability of atomic force microscopy to determine cancer-related changes in cells. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2022; 380:20210346. [PMID: 35909354 DOI: 10.1098/rsta.2021.0346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 02/24/2022] [Indexed: 06/15/2023]
Abstract
The determination of mechanical properties of living cells as an indicator of cancer progression has become possible with the development of local measurement techniques such as atomic force microscopy (AFM). Its most important advantage is a nanoscopic character, implying that very local alterations can be quantified. The results gathered from AFM measurements of various cancers show that, for most cancers, individual cells are characterized by the lower apparent Young's modulus, denoting higher cell deformability. The measured value depends on various factors, like the properties of substrates used for cell growth, force loading rate or indentation depth. Despite this, the results proved the AFM capability to recognize mechanically altered cells. This can significantly impact the development of methodological approaches toward the precise identification of pathological cells. This article is part of the theme issue 'Nanocracks in nature and industry'.
Collapse
Affiliation(s)
- Małgorzata Lekka
- Department of Biophysical Microstructures, Institute of Nuclear Physics, Polish Academy of Sciences, PL-31342 Krakow, Poland
| |
Collapse
|
26
|
Senent Y, Tavira B, Pio R, Ajona D. The complement system as a regulator of tumor-promoting activities mediated by myeloid-derived suppressor cells. Cancer Lett 2022; 549:215900. [PMID: 36087681 DOI: 10.1016/j.canlet.2022.215900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/25/2022] [Accepted: 08/26/2022] [Indexed: 11/16/2022]
Abstract
Tumor progression relies on the interaction between tumor cells and their surrounding tumor microenvironment (TME), which also influences therapeutic responses. The complement system, an essential part of innate immunity, has been traditionally considered an effector arm against tumors. However, established tumors co-opt complement-mediated immune responses in the TME to support chronic inflammation, activate cancer-related signaling pathways and hamper antitumor immune responses. In this context, myeloid-derived suppressor cells (MDSCs), a heterogeneous population of myeloid progenitors with immunosuppressive functions, are recognized as major mediators of tumor-associated complement activities. This review focuses on the impact of complement activation within the TME, with a special emphasis on MDSC functions and the involvement of the C5a/C5aR1 axis. We also discuss the translation of these findings into therapeutic advances based on complement inhibition.
Collapse
Affiliation(s)
- Yaiza Senent
- Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain; Cancer Center University of Navarra (CCUN), Pamplona, Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain
| | - Beatriz Tavira
- Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain; Cancer Center University of Navarra (CCUN), Pamplona, Spain; University of Navarra, School of Medicine, Department of Pathology, Anatomy and Physiology, Pamplona, Spain
| | - Ruben Pio
- Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain; Cancer Center University of Navarra (CCUN), Pamplona, Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain; Navarra Institute for Health Research (IdISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| | - Daniel Ajona
- Cima-University of Navarra, Program in Solid Tumors, Pamplona, Spain; Cancer Center University of Navarra (CCUN), Pamplona, Spain; University of Navarra, School of Sciences, Department of Biochemistry and Genetics, Pamplona, Spain; Navarra Institute for Health Research (IdISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
27
|
Rodríguez‐Pena A, Armendariz E, Oyarbide A, Morales X, Ortiz‐Espinosa S, Ruiz‐Fernández de Córdoba B, Cochonneau D, Cornago I, Heymann D, Argemi J, D'Avola D, Sangro B, Lecanda F, Pio R, Cortés‐Domínguez I, Ortiz‐de‐Solórzano C. Design and validation of a tunable inertial microfluidic system for the efficient enrichment of circulating tumor cells in blood. Bioeng Transl Med 2022; 7:e10331. [PMID: 36176621 PMCID: PMC9472016 DOI: 10.1002/btm2.10331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 03/11/2022] [Accepted: 04/04/2022] [Indexed: 11/08/2022] Open
Abstract
The analysis of circulating tumor cells (CTCs) in blood is a powerful noninvasive alternative to conventional tumor biopsy. Inertial-based separation is a promising high-throughput, marker-free sorting strategy for the enrichment and isolation of CTCs. Here, we present and validate a double spiral microfluidic device that efficiently isolates CTCs with a fine-tunable cut-off value of 9 μm and a separation range of 2 μm. We designed the device based on computer simulations that introduce a novel, customized inertial force term, and provide practical fabrication guidelines. We validated the device using calibration beads, which allowed us to refine the simulations and redesign the device. Then we validated the redesigned device using blood samples and a murine model of metastatic breast cancer. Finally, as a proof of principle, we tested the device using peripheral blood from a patient with hepatocellular carcinoma, isolating more than 17 CTCs/ml, with purity/removal values of 96.03% and 99.99% of white blood cell and red blood cells, respectively. These results confirm highly efficient CTC isolation with a stringent cut-off value and better separation results than the state of the art.
Collapse
Affiliation(s)
- Alejandro Rodríguez‐Pena
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | | | - Alvaro Oyarbide
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | - Xabier Morales
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | - Sergio Ortiz‐Espinosa
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
- Department of Biochemistry and Genetics, School of SciencesUniversity of NavarraPamplonaSpain
| | - Borja Ruiz‐Fernández de Córdoba
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | - Denis Cochonneau
- Institut de Cancérologie de l'Ouest, “Tumor Heterogeneity and Precision Medicine” Lab., Blvd Jacques MonodSaint‐HerblainFrance
| | - Iñaki Cornago
- Automotive and Mechatronics R&D Foundation (Naitec)PamplonaSpain
| | - Dominique Heymann
- Institut de Cancérologie de l'Ouest, “Tumor Heterogeneity and Precision Medicine” Lab., Blvd Jacques MonodSaint‐HerblainFrance
- Nantes Université, CNRS, US2B, UMR 6286NantesFrance
| | - Josepmaría Argemi
- Liver Unit, Clinica Universitaria de NavarraPamplona (Navarra)Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepáticas y Digestivas (CIBEREHD)Spain
| | - Delia D'Avola
- Liver Unit, Clinica Universitaria de NavarraPamplona (Navarra)Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepáticas y Digestivas (CIBEREHD)Spain
| | - Bruno Sangro
- Liver Unit, Clinica Universitaria de NavarraPamplona (Navarra)Spain
- Centro de Investigación Biomédica en Red de Enfermedades hepáticas y Digestivas (CIBEREHD)Spain
| | - Fernando Lecanda
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
- Department of Pathology, Anatomy and PhysiologyUniversity of NavarraPamplonaSpain
| | - Ruben Pio
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
- Department of Biochemistry and Genetics, School of SciencesUniversity of NavarraPamplonaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
| | - Iván Cortés‐Domínguez
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
| | - Carlos Ortiz‐de‐Solórzano
- Program in Solid TumorsCenter for Applied Medical Research (CIMA)PamplonaSpain
- Oncology DivisionNavarra's Health Research Institute (IDISNA)PamplonaSpain
- Centro de Investigación Biomédica en Red Cáncer (CIBERONC)MadridSpain
| |
Collapse
|
28
|
Sala F, Ficorella C, Osellame R, Käs JA, Martínez Vázquez R. Microfluidic Lab-on-a-Chip for Studies of Cell Migration under Spatial Confinement. BIOSENSORS 2022; 12:bios12080604. [PMID: 36004998 PMCID: PMC9405557 DOI: 10.3390/bios12080604] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022]
Abstract
Understanding cell migration is a key step in unraveling many physiological phenomena and predicting several pathologies, such as cancer metastasis. In particular, confinement has been proven to be a key factor in the cellular migration strategy choice. As our insight in the field improves, new tools are needed in order to empower biologists’ analysis capabilities. In this framework, microfluidic devices have been used to engineer the mechanical and spatial stimuli and to investigate cellular migration response in a more controlled way. In this work, we will review the existing technologies employed in the realization of microfluidic cellular migration assays, namely the soft lithography of PDMS and hydrogels and femtosecond laser micromachining. We will give an overview of the state of the art of these devices, focusing on the different geometrical configurations that have been exploited to study specific aspects of cellular migration. Our scope is to highlight the advantages and possibilities given by each approach and to envisage the future developments in in vitro migration studies under spatial confinement in microfluidic devices.
Collapse
Affiliation(s)
- Federico Sala
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Carlotta Ficorella
- Peter Debye Institute for Soft Matter Physics, University of Leipzig, 04109 Leipzig, Germany
| | - Roberto Osellame
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
| | - Josef A. Käs
- Peter Debye Institute for Soft Matter Physics, University of Leipzig, 04109 Leipzig, Germany
| | - Rebeca Martínez Vázquez
- Institute for Photonics and Nanotechnologies, CNR, Piazza Leonardo da Vinci 32, 20133 Milano, Italy
- Correspondence:
| |
Collapse
|
29
|
Song Q, Zhang Y, Zhou M, Xu Y, Zhang Q, Wu L, Liu S, Zhang M, Zhang L, Wu Z, Peng W, Liu X, Zhao C. The Culture Dish Surface Influences the Phenotype and Dissociation Strategy in Distinct Mouse Macrophage Populations. Front Immunol 2022; 13:920232. [PMID: 35874686 PMCID: PMC9299442 DOI: 10.3389/fimmu.2022.920232] [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: 04/14/2022] [Accepted: 06/10/2022] [Indexed: 12/04/2022] Open
Abstract
The nature of the culture dish surface and the technique used to detach adherent cells could very likely influence the cell viability and cell membrane protein integrity of harvested macrophages. Several previous studies assessed the detachment efficacies of enzymatic and non-enzymatic methods for harvesting the single cell suspensions of macrophages, but a comprehensive study assessing different dissociation methods and culture conditions for detaching functionally different macrophage populations has not yet been reported. In this study, via the well-established GM-CSF and M-CSF differentiated bone marrow derived macrophage models (GM-BMDMs and M-BMDMs), we compared four commonly used enzymatic (trypsin and accutase) and non-enzymatic (PBS and EDTA) dissociation methods along with necessary mechanical detaching steps (scraping and pipetting) to evaluate the viable cell recovery and cell surface marker integrality of GM-BMDMs and M-BMDMs cultured on standard cell culture dish (TC dish), or on culture dish (noTC dish) that was not conditioned to enhance adherence. The data showed that accutase yielded a better recovery of viable cells comparing with PBS and EDTA, especially for tightly adherent GM-BMDMs on TC dishes, with a relatively higher level of detected cell membrane marker F4/80 than trypsin. An additional gradient centrifugation-based dead cell removal approach could increase the proportion of viable cells for TC cultured GM-BMDMs after accutase dissociation. Furthermore, transcriptome analysis was performed to evaluate the putative influence of culture dishes. At steady state, BMDMs cultured on noTC dishes exhibited more proinflammatory gene expression signatures (e.g. IL6, CXCL2 and ILlβ) and functions (e.g. TNF and IL17 signaling pathways). Similar inflammatory responses were observed upon LPS challenge regardless of culture conditions and differentiation factors. However, in LPS treated samples, the difference of gene expression patterns, signaling pathways and molecular functions between TC and noTC cultured BMDMs were largely dependent on the types of growth factors (M-CSF and GM-CSF). This observation might provide valuable information for in vitro macrophage studies.
Collapse
Affiliation(s)
- Qiaoling Song
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yazhuo Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Mingming Zhou
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Yuting Xu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Qianyue Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Lihong Wu
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Shan Liu
- Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Minghui Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China
| | - Lei Zhang
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zhihua Wu
- College of Food Science and Engineering, Ocean University of China, Qingdao, China
| | - Weixun Peng
- Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Xutao Liu
- Samueli School of Engineering, University of California Los Angeles, Los Angeles, CA, United States
| | - Chenyang Zhao
- School of Medicine and Pharmacy, Ocean University of China, Qingdao, China.,Innovation Platform of Marine Drug Screening and Evaluation, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
30
|
Guo T, He C, Venado A, Zhou Y. Extracellular Matrix Stiffness in Lung Health and Disease. Compr Physiol 2022; 12:3523-3558. [PMID: 35766837 PMCID: PMC10088466 DOI: 10.1002/cphy.c210032] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The extracellular matrix (ECM) provides structural support and imparts a wide variety of environmental cues to cells. In the past decade, a growing body of work revealed that the mechanical properties of the ECM, commonly known as matrix stiffness, regulate the fundamental cellular processes of the lung. There is growing appreciation that mechanical interplays between cells and associated ECM are essential to maintain lung homeostasis. Dysregulation of ECM-derived mechanical signaling via altered mechanosensing and mechanotransduction pathways is associated with many common lung diseases. Matrix stiffening is a hallmark of lung fibrosis. The stiffened ECM is not merely a sequelae of lung fibrosis but can actively drive the progression of fibrotic lung disease. In this article, we provide a comprehensive view on the role of matrix stiffness in lung health and disease. We begin by summarizing the effects of matrix stiffness on the function and behavior of various lung cell types and on regulation of biomolecule activity and key physiological processes, including host immune response and cellular metabolism. We discuss the potential mechanisms by which cells probe matrix stiffness and convert mechanical signals to regulate gene expression. We highlight the factors that govern matrix stiffness and outline the role of matrix stiffness in lung development and the pathogenesis of pulmonary fibrosis, pulmonary hypertension, asthma, chronic obstructive pulmonary disease (COPD), and lung cancer. We envision targeting of deleterious matrix mechanical cues for treatment of fibrotic lung disease. Advances in technologies for matrix stiffness measurements and design of stiffness-tunable matrix substrates are also explored. © 2022 American Physiological Society. Compr Physiol 12:3523-3558, 2022.
Collapse
Affiliation(s)
- Ting Guo
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA.,Department of Respiratory Medicine, the Second Xiangya Hospital, Central-South University, Changsha, Hunan, China
| | - Chao He
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| | - Aida Venado
- Pulmonary, Critical Care, Allergy and Sleep Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | - Yong Zhou
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Alabama, USA
| |
Collapse
|
31
|
Luthold C, Hallal T, Labbé DP, Bordeleau F. The Extracellular Matrix Stiffening: A Trigger of Prostate Cancer Progression and Castration Resistance? Cancers (Basel) 2022; 14:cancers14122887. [PMID: 35740556 PMCID: PMC9221142 DOI: 10.3390/cancers14122887] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 02/06/2023] Open
Abstract
Despite advancements made in diagnosis and treatment, prostate cancer remains the second most diagnosed cancer among men worldwide in 2020, and the first in North America and Europe. Patients with localized disease usually respond well to first-line treatments, however, up to 30% develop castration-resistant prostate cancer (CRPC), which is often metastatic, making this stage of the disease incurable and ultimately fatal. Over the last years, interest has grown into the extracellular matrix (ECM) stiffening as an important mediator of diseases, including cancers. While this process is increasingly well-characterized in breast cancer, a similar in-depth look at ECM stiffening remains lacking for prostate cancer. In this review, we scrutinize the current state of literature regarding ECM stiffening in prostate cancer and its potential association with disease progression and castration resistance.
Collapse
Affiliation(s)
- Carole Luthold
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
| | - Tarek Hallal
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
| | - David P. Labbé
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montréal, QC H4A 3J1, Canada;
- Division of Urology, Department of Surgery, McGill University, Montréal, QC H4A 3J1, Canada
- Correspondence: (D.P.L.); (F.B.)
| | - François Bordeleau
- Centre de Recherche sur le Cancer, Université Laval, Québec, QC G1R 3S3, Canada;
- Division of Oncology, Centre de Recherche du CHU de Québec-Université Laval, Hôtel-Dieu de Québec, Québec, QC G1R 3S3, Canada
- Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Correspondence: (D.P.L.); (F.B.)
| |
Collapse
|
32
|
Mahdieh Z, Cherne MD, Fredrikson JP, Sidar B, Sanchez HS, Chang CB, Bimczok D, Wilking JN. Granular Matrigel: restructuring a trusted extracellular matrix material for improved permeability. Biomed Mater 2022; 17:045020. [PMID: 35609584 DOI: 10.1088/1748-605x/ac7306] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/24/2022] [Indexed: 11/11/2022]
Abstract
Matrigel is a polymeric extracellular matrix material produced by mouse cancer cells. Over the past four decades, Matrigel has been shown to support a wide variety of two- and three-dimensional cell and tissue culture applications including organoids. Despite widespread use, transport of molecules, cells, and colloidal particles through Matrigel can be limited. These limitations restrict cell growth, viability, and function and limit Matrigel applications. A strategy to improve transport through a hydrogel without modifying the chemistry or composition of the gel is to physically restructure the material into microscopic microgels and then pack them together to form a porous material. These 'granular' hydrogels have been created using a variety of synthetic hydrogels, but granular hydrogels composed of Matrigel have not yet been reported. Here we present a drop-based microfluidics approach for structuring Matrigel into a three-dimensional, mesoporous material composed of packed Matrigel microgels, which we call granular Matrigel. We show that restructuring Matrigel in this manner enhances the transport of colloidal particles and human dendritic cells (DCs) through the gel while providing sufficient mechanical support for culture of human gastric organoids (HGOs) and co-culture of human DCs with HGOs.
Collapse
Affiliation(s)
- Zahra Mahdieh
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Michelle D Cherne
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States of America
| | - Jacob P Fredrikson
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Barkan Sidar
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Humberto S Sanchez
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Connie B Chang
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| | - Diane Bimczok
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT, United States of America
| | - James N Wilking
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, United States of America
- Center for Biofilm Engineering, Montana State University, Bozeman, MT, United States of America
| |
Collapse
|
33
|
Mehta P, Rahman Z, Ten Dijke P, Boukany PE. Microfluidics meets 3D cancer cell migration. Trends Cancer 2022; 8:683-697. [PMID: 35568647 DOI: 10.1016/j.trecan.2022.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 03/23/2022] [Accepted: 03/30/2022] [Indexed: 01/12/2023]
Abstract
An early step of metastasis requires a complex and coordinated migration of invasive tumor cells into the surrounding tumor microenvironment (TME), which contains extracellular matrix (ECM). It is being appreciated that 3D matrix-based microfluidic models have an advantage over conventional in vitro and animal models to study tumor progression events. Recent microfluidic models have enabled recapitulation of key mechanobiological features present within the TME to investigate collective cancer cell migration and invasion. Microfluidics also allows for functional interrogation and therapeutic manipulation of specific steps to study the dynamic aspects of tumor progression. In this review, we focus on recent developments in cancer cell migration and how microfluidic strategies have evolved to address the physiological complexities of the TME to visualize migration modes adapted by various tumor cells.
Collapse
Affiliation(s)
- Pranav Mehta
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands; Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Zaid Rahman
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology and Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands.
| | - Pouyan E Boukany
- Department of Chemical Engineering, Delft University of Technology, Van der Maasweg 9, 2629 HZ, Delft, The Netherlands.
| |
Collapse
|
34
|
Choi JR, Kozalak G, di Bari I, Babar Q, Niknam Z, Rasmi Y, Yong KW. In Vitro Human Cancer Models for Biomedical Applications. Cancers (Basel) 2022; 14:2284. [PMID: 35565413 PMCID: PMC9099454 DOI: 10.3390/cancers14092284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/16/2022] [Accepted: 04/30/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide, and its incidence is steadily increasing. Although years of research have been conducted on cancer treatment, clinical treatment options for cancers are still limited. Animal cancer models have been widely used for studies of cancer therapeutics, but these models have been associated with many concerns, including inaccuracy in the representation of human cancers, high cost and ethical issues. Therefore, in vitro human cancer models are being developed quickly to fulfill the increasing demand for more relevant models in order to get a better knowledge of human cancers and to find novel treatments. This review summarizes the development of in vitro human cancer models for biomedical applications. We first review the latest development in the field by detailing various types of in vitro human cancer models, including transwell-based models, tumor spheroids, microfluidic tumor-microvascular systems and scaffold-based models. The advantages and limitations of each model, as well as their biomedical applications, are summarized, including therapeutic development, assessment of tumor cell migration, metastasis and invasion and discovery of key cancer markers. Finally, the existing challenges and future perspectives are briefly discussed.
Collapse
Affiliation(s)
- Jane Ru Choi
- Life Sciences Centre, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 1Z3, Canada;
| | - Gül Kozalak
- Faculty of Engineering and Natural Sciences (FENS), Sabanci University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano-Diagnostics (EFSUN), Sabanci University, Istanbul 34956, Turkey
| | - Ighli di Bari
- Dialysis and Transplantation Unit, Department of Emergency and Organ Transplantation-Nephrology, University of Bari, 70124 Bari, Italy;
| | - Quratulain Babar
- Department of Biochemistry, Government College University, Faisalabad 38000, Pakistan;
| | - Zahra Niknam
- Proteomics Research Center, Shahid Behesti University of Medical Sciences, Tehran 1983969411, Iran;
| | - Yousef Rasmi
- Department of Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 5714783734, Iran
- Cellular and Molecular Research Center, Urmia University of Medical Sciences, Urmia 5714783734, Iran
| | - Kar Wey Yong
- Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB T6G 2R7, Canada
| |
Collapse
|
35
|
Physical Forces in Glioblastoma Migration: A Systematic Review. Int J Mol Sci 2022; 23:ijms23074055. [PMID: 35409420 PMCID: PMC9000211 DOI: 10.3390/ijms23074055] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 02/01/2023] Open
Abstract
The invasive capabilities of glioblastoma (GBM) define the cancer’s aggressiveness, treatment resistance, and overall mortality. The tumor microenvironment influences the molecular behavior of cells, both epigenetically and genetically. Current forces being studied include properties of the extracellular matrix (ECM), such as stiffness and “sensing” capabilities. There is currently limited data on the physical forces in GBM—both relating to how they influence their environment and how their environment influences them. This review outlines the advances that have been made in the field. It is our hope that further investigation of the physical forces involved in GBM will highlight new therapeutic options and increase patient survival. A search of the PubMed database was conducted through to 23 March 2022 with the following search terms: (glioblastoma) AND (physical forces OR pressure OR shear forces OR compression OR tension OR torsion) AND (migration OR invasion). Our review yielded 11 external/applied/mechanical forces and 2 tumor microenvironment (TME) forces that affect the ability of GBM to locally migrate and invade. Both external forces and forces within the tumor microenvironment have been implicated in GBM migration, invasion, and treatment resistance. We endorse further research in this area to target the physical forces affecting the migration and invasion of GBM.
Collapse
|
36
|
Narkar AR, Tong Z, Soman P, Henderson JH. Smart biomaterial platforms: Controlling and being controlled by cells. Biomaterials 2022; 283:121450. [PMID: 35247636 PMCID: PMC8977253 DOI: 10.1016/j.biomaterials.2022.121450] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/23/2022] [Accepted: 02/26/2022] [Indexed: 02/07/2023]
Abstract
Across diverse research and application areas, dynamic functionality-such as programmable changes in biochemical property, in mechanical property, or in microscopic or macroscopic architecture-is an increasingly common biomaterials design criterion, joining long-studied criteria such as cytocompatibility and biocompatibility, drug release kinetics, and controlled degradability or long-term stability in vivo. Despite tremendous effort, achieving dynamic functionality while simultaneously maintaining other desired design criteria remains a significant challenge. Reversible dynamic functionality, rather than one-time or one-way dynamic functionality, is of particular interest but has proven especially challenging. Such reversible functionality could enable studies that address the current gap between the dynamic nature of in vivo biological and biomechanical processes, such as cell traction, cell-extracellular matrix (ECM) interactions, and cell-mediated ECM remodeling, and the static nature of the substrates and ECM constructs used to study the processes. This review assesses dynamic materials that have traditionally been used to control cell activity and static biomaterial constructs, experimental and computational techniques, with features that may inform continued advances in reversible dynamic materials. Taken together, this review presents a perspective on combining the reversibility of smart materials and the in-depth dynamic cell behavior probed by static polymers to design smart bi-directional ECM platforms that can reversibly and repeatedly communicate with cells.
Collapse
Affiliation(s)
- Ameya R Narkar
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, United States; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, United States.
| | - Zhuoqi Tong
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, United States; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, United States.
| | - Pranav Soman
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, United States; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, United States.
| | - James H Henderson
- BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, United States; Department of Biomedical and Chemical Engineering, Syracuse University, Syracuse, NY, 13244, United States.
| |
Collapse
|
37
|
Wieleba I, Wojas-Krawczyk K, Krawczyk P, Milanowski J. Clinical Application Perspectives of Lung Cancers 3D Tumor Microenvironment Models for In Vitro Cultures. Int J Mol Sci 2022; 23:ijms23042261. [PMID: 35216378 PMCID: PMC8876687 DOI: 10.3390/ijms23042261] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/01/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Despite the enormous progress and development of modern therapies, lung cancer remains one of the most common causes of death among men and women. The key element in the development of new anti-cancer drugs is proper planning of the preclinical research phase. The most adequate basic research exemplary for cancer study are 3D tumor microenvironment in vitro models, which allow us to avoid the use of animal models and ensure replicable culture condition. However, the question tormenting the scientist is how to choose the best tool for tumor microenvironment research, especially for extremely heterogenous lung cancer cases. In the presented review we are focused to explain the key factors of lung cancer biology, its microenvironment, and clinical gaps related to different therapies. The review summarized the most important strategies for in vitro culture models mimicking the tumor–tumor microenvironmental interaction, as well as all advantages and disadvantages were depicted. This knowledge could facilitate the right decision to designate proper pre-clinical in vitro study, based on available analytical tools and technical capabilities, to obtain more reliable and personalized results for faster introduction them into the future clinical trials.
Collapse
|
38
|
An Innovative Customized Biomimetic Hydrogel for Drug Screening Application Potential: Biocompatibility and Cell Invasion Ability. Int J Mol Sci 2022; 23:ijms23031488. [PMID: 35163411 PMCID: PMC8835991 DOI: 10.3390/ijms23031488] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 01/02/2023] Open
Abstract
The ability of Pluronic F127 (PF127) conjugated with tetrapeptide Gly-Arg-Gly-Asp (GRGD) as a sequence of Arg-Gly-Asp (RGD) peptide to form the investigated potential hydrogel (hereafter referred to as 3DG bioformer (3BE)) to produce spheroid, biocompatibility, and cell invasion ability, was assessed in this study. The fibroblast cell line (NIH 3T3), osteoblast cell line (MG-63), and human breast cancer cell line (MCF-7) were cultured in the 3BE hydrogel and commercial product (Matrigel) for comparison. The morphology of spheroid formation was evaluated via optical microscopy. The cell viability was observed through cell counting Kit-8 assay, and cell invasion was investigated via Boyden chamber assay. Analytical results indicated that 3BE exhibited lower spheroid formation than Matrigel. However, the 3BE appeared biocompatible to NIH 3T3, MG-63, and MCF-7 cells. Moreover, cell invasion ability and cell survival rate after invasion through the 3BE was displayed to be comparable to Matrigel. Thus, these findings demonstrate that the 3BE hydrogel has a great potential as an alternative to a three-dimensional cell culture for drug screening applications.
Collapse
|
39
|
Ortiz-Espinosa S, Morales X, Senent Y, Alignani D, Tavira B, Macaya I, Ruiz B, Moreno H, Remírez A, Sainz C, Rodriguez-Pena A, Oyarbide A, Ariz M, Andueza MP, Valencia K, Teijeira A, Hoehlig K, Vater A, Rolfe B, Woodruff TM, Lopez-Picazo JM, Vicent S, Kochan G, Escors D, Gil-Bazo I, Perez-Gracia JL, Montuenga LM, Lambris JD, Ortiz de Solorzano C, Lecanda F, Ajona D, Pio R. Complement C5a induces the formation of neutrophil extracellular traps by myeloid-derived suppressor cells to promote metastasis. Cancer Lett 2021; 529:70-84. [PMID: 34971753 DOI: 10.1016/j.canlet.2021.12.027] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 12/22/2021] [Accepted: 12/24/2021] [Indexed: 02/06/2023]
Abstract
Myeloid-derived suppressor cells (MDSCs) play a major role in cancer progression. In this study, we investigated the mechanisms by which complement C5a increases the capacity of polymorphonuclear MDSCs (PMN-MDSCs) to promote tumor growth and metastatic spread. Stimulation of PMN-MDSCs with C5a favored the invasion of cancer cells via a process dependent on the formation of neutrophil extracellular traps (NETs). NETosis was dependent on the production of high mobility group box 1 (HMGB1) by cancer cells. Moreover, C5a induced the surface expression of the HMGB1 receptors TLR4 and RAGE in PMN-MDSCs. In a mouse lung metastasis model, inhibition of C5a, C5a receptor-1 (C5aR1) or NETosis reduced the number of circulating-tumor cells (CTCs) and the metastatic burden. In support of the translational relevance of these findings, C5a was able to stimulate migration and NETosis in PMN-MDSCs obtained from lung cancer patients. Furthermore, myeloperoxidase (MPO)-DNA complexes, as markers of NETosis, were elevated in lung cancer patients and significantly correlated with C5a levels. In conclusion, C5a induces the formation of NETs from PMN-MDSCs in the presence of cancer cells, which may facilitate cancer cell dissemination and metastasis.
Collapse
Affiliation(s)
- Sergio Ortiz-Espinosa
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain
| | - Xabier Morales
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Imaging Platform, CIMA, Pamplona, Spain
| | - Yaiza Senent
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain
| | - Diego Alignani
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Cytometry Unit, Cima-University of Navarra, Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Beatriz Tavira
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain
| | - Irati Macaya
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain
| | - Borja Ruiz
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain
| | - Haritz Moreno
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain
| | - Ana Remírez
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Cristina Sainz
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Alejandro Rodriguez-Pena
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Imaging Platform, CIMA, Pamplona, Spain
| | - Alvaro Oyarbide
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Imaging Platform, CIMA, Pamplona, Spain
| | - Mikel Ariz
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Imaging Platform, CIMA, Pamplona, Spain
| | - Maria P Andueza
- Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Karmele Valencia
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Alvaro Teijeira
- Program in Immunology and Immunotherapy, Cima-University of Navarra, Pamplona, Spain
| | | | | | - Barbara Rolfe
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Queensland, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Queensland, Australia
| | - Jose Maria Lopez-Picazo
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Silvestre Vicent
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - Grazyna Kochan
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Immunomodulation Group, Navarrabiomed-Biomedical Research Center, Pamplona, Spain
| | - David Escors
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Immunomodulation Group, Navarrabiomed-Biomedical Research Center, Pamplona, Spain
| | - Ignacio Gil-Bazo
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Jose Luis Perez-Gracia
- Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; Department of Oncology, Clínica Universidad de Navarra, Pamplona, Spain
| | - Luis M Montuenga
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain; Department of Pathology, Anatomy and Physiology, School of Medicine, University of Navarra, Pamplona, Spain
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carlos Ortiz de Solorzano
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Imaging Platform, CIMA, Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Fernando Lecanda
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| | - Daniel Ajona
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain.
| | - Ruben Pio
- Program in Solid Tumors, Cima-University of Navarra, Pamplona, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain; Navarra's Health Research Institute (IDISNA), Pamplona, Spain; Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Madrid, Spain
| |
Collapse
|
40
|
Herring B, Jang S, Whitt J, Goliwas K, Aburjania Z, Dudeja V, Ren B, Berry J, Bibb J, Frost A, Chen H, Rose JB, Jaskula-Sztul R. Ex Vivo Modeling of Human Neuroendocrine Tumors in Tissue Surrogates. Front Endocrinol (Lausanne) 2021; 12:710009. [PMID: 35002949 PMCID: PMC8734644 DOI: 10.3389/fendo.2021.710009] [Citation(s) in RCA: 2] [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: 05/14/2021] [Accepted: 11/10/2021] [Indexed: 12/24/2022] Open
Abstract
Few models exist for studying neuroendocrine tumors (NETs), and there are mounting concerns that the currently available array of cell lines is not representative of NET biology. The lack of stable patient-derived NET xenograft models further limits the scientific community's ability to make conclusions about NETs and their response to therapy in patients. To address these limitations, we propose the use of an ex vivo 3D flow-perfusion bioreactor system for culturing and studying patient-derived NET surrogates. Herein, we demonstrate the utility of the bioreactor system for culturing NET surrogates and provide methods for evaluating the efficacy of therapeutic agents on human NET cell line xenograft constructs and patient-derived NET surrogates. We also demonstrate that patient-derived NET tissues can be propagated using the bioreactor system and investigate the near-infrared (NIR) dye IR-783 for its use in monitoring their status within the bioreactor. The results indicate that the bioreactor system and similar 3D culture models may be valuable tools for culturing patient-derived NETs and monitoring their response to therapy ex vivo.
Collapse
Affiliation(s)
- Brendon Herring
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Samuel Jang
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jason Whitt
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kayla Goliwas
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zviadi Aburjania
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vikas Dudeja
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bin Ren
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joel Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - James Bibb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andra Frost
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John Bart Rose
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Renata Jaskula-Sztul
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
41
|
Boghdady CM, Kalashnikov N, Mok S, McCaffrey L, Moraes C. Revisiting tissue tensegrity: Biomaterial-based approaches to measure forces across length scales. APL Bioeng 2021; 5:041501. [PMID: 34632250 PMCID: PMC8487350 DOI: 10.1063/5.0046093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 09/08/2021] [Indexed: 12/18/2022] Open
Abstract
Cell-generated forces play a foundational role in tissue dynamics and homeostasis and are critically important in several biological processes, including cell migration, wound healing, morphogenesis, and cancer metastasis. Quantifying such forces in vivo is technically challenging and requires novel strategies that capture mechanical information across molecular, cellular, and tissue length scales, while allowing these studies to be performed in physiologically realistic biological models. Advanced biomaterials can be designed to non-destructively measure these stresses in vitro, and here, we review mechanical characterizations and force-sensing biomaterial-based technologies to provide insight into the mechanical nature of tissue processes. We specifically and uniquely focus on the use of these techniques to identify characteristics of cell and tissue "tensegrity:" the hierarchical and modular interplay between tension and compression that provide biological tissues with remarkable mechanical properties and behaviors. Based on these observed patterns, we highlight and discuss the emerging role of tensegrity at multiple length scales in tissue dynamics from homeostasis, to morphogenesis, to pathological dysfunction.
Collapse
Affiliation(s)
| | - Nikita Kalashnikov
- Department of Chemical Engineering, McGill University, Montréal, Québec H3A 0C5, Canada
| | - Stephanie Mok
- Department of Chemical Engineering, McGill University, Montréal, Québec H3A 0C5, Canada
| | | | | |
Collapse
|
42
|
Banerjee A, Khan MP, Barui A, Datta P, Chowdhury AR, Bhowmik K. Finite element analysis of the influence of cyclic strain on cells anchored to substrates with varying properties. Med Biol Eng Comput 2021; 60:171-187. [PMID: 34782982 DOI: 10.1007/s11517-021-02453-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/20/2021] [Indexed: 12/27/2022]
Abstract
The response of cytoskeleton to mechanical cues plays a pivotal role in understanding several aspects of cellular growth, migration, and cell-cell and cell-matrix interactions under normal and diseased conditions. Finite element analysis (FEA) has become a powerful computational technique to study the response of cytoskeleton in the maintenance of overall cellular mechanics. With the revelation of role of external mechanical microenvironment on cell mechanics, FEA models have also been developed to simulate the effect of substrate stiffness on the mechanical properties of cancer cells. However, the models developed so far model cellular response under static mode, whereas in physiological condition, cells always experience dynamic loading conditions. To develop a more accurate model of cell-extracellular matrix (ECM) interactions, this paper models the cytoskeleton and other parts of the cell by beam and solid elements respectively, assuming spherical morphology of the cell. The stiffness and roughness of extracellular matrix were varied. Furthermore, static and dynamic sinusoidal loads were applied through a flat plate indenter on the cell along with providing sinusoidal strain at the substrate. It is observed that due to axial loading, cell reaches a plastic region, and when the sinusoidal loading is added to the axial load, the cell experiences permanent deformation. Degradation of the cytoskeleton elements and a physiologically more relevant spherical cap shape of the cell were also considered during the analysis. This study suggests that asperity topology of the substrate and indirect cyclic load can play a significant role in the shape alterations and motion of a cell.
Collapse
Affiliation(s)
- Abhinaba Banerjee
- Department of Mechanical Engineering, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Mohammed Parvez Khan
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Ananya Barui
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| | - Pallab Datta
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Kolkata, 700054, West Bengal, India
| | - Amit Roy Chowdhury
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India. .,Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India.
| | - Krishnendu Bhowmik
- Department of Aerospace Engineering and Applied Mechanics, Indian Institute of Engineering Science and Technology, Shibpur, Howrah, 711103, India
| |
Collapse
|
43
|
Torab P, Yan Y, Ahmed M, Yamashita H, Warrick JI, Raman JD, DeGraff DJ, Wong PK. Intratumoral Heterogeneity Promotes Collective Cancer Invasion through NOTCH1 Variation. Cells 2021; 10:3084. [PMID: 34831307 PMCID: PMC8619970 DOI: 10.3390/cells10113084] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 10/29/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
Abstract
Cellular and molecular heterogeneity within tumors has long been associated with the progression of cancer to an aggressive phenotype and a poor prognosis. However, how such intratumoral heterogeneity contributes to the invasiveness of cancer is largely unknown. Here, using a tumor bioengineering approach, we investigate the interaction between molecular subtypes within bladder microtumors and the corresponding effects on their invasiveness. Our results reveal heterogeneous microtumors formed by multiple molecular subtypes possess enhanced invasiveness compared to individual cells, even when both cells are not invasive individually. To examine the molecular mechanism of intratumoral heterogeneity mediated invasiveness, live single cell biosensing, RNA interference, and CRISPR-Cas9 gene editing approaches were applied to investigate and control the composition of the microtumors. An agent-based computational model was also developed to evaluate the influence of NOTCH1 variation on DLL4 expression within a microtumor. The data indicate that intratumoral variation in NOTCH1 expression can lead to upregulation of DLL4 expression within the microtumor and enhancement of microtumor invasiveness. Overall, our results reveal a novel mechanism of heterogeneity mediated invasiveness through intratumoral variation of gene expression.
Collapse
Affiliation(s)
- Peter Torab
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Yue Yan
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; (Y.Y.); (M.A.)
| | - Mona Ahmed
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; (Y.Y.); (M.A.)
| | - Hironobu Yamashita
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (H.Y.); (J.I.W.); (D.J.D.)
| | - Joshua I. Warrick
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (H.Y.); (J.I.W.); (D.J.D.)
- Penn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USA;
| | - Jay D. Raman
- Penn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USA;
| | - David J. DeGraff
- Department of Pathology and Laboratory Medicine, The Pennsylvania State University, Hershey, PA 17033, USA; (H.Y.); (J.I.W.); (D.J.D.)
- Penn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USA;
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Pak Kin Wong
- Department of Mechanical Engineering, The Pennsylvania State University, University Park, PA 16802, USA;
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA; (Y.Y.); (M.A.)
- Penn State Health Milton S., Hershey Medical Center, Department of Surgery, Hershey, PA 17033, USA;
| |
Collapse
|
44
|
Amer M, Shi L, Wolfenson H. The 'Yin and Yang' of Cancer Cell Growth and Mechanosensing. Cancers (Basel) 2021; 13:4754. [PMID: 34638240 PMCID: PMC8507527 DOI: 10.3390/cancers13194754] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 09/07/2021] [Accepted: 09/16/2021] [Indexed: 01/06/2023] Open
Abstract
In cancer, two unique and seemingly contradictory behaviors are evident: on the one hand, tumors are typically stiffer than the tissues in which they grow, and this high stiffness promotes their malignant progression; on the other hand, cancer cells are anchorage-independent-namely, they can survive and grow in soft environments that do not support cell attachment. How can these two features be consolidated? Recent findings on the mechanisms by which cells test the mechanical properties of their environment provide insight into the role of aberrant mechanosensing in cancer progression. In this review article, we focus on the role of high stiffness on cancer progression, with particular emphasis on tumor growth; we discuss the mechanisms of mechanosensing and mechanotransduction, and their dysregulation in cancerous cells; and we propose that a 'yin and yang' type phenomenon exists in the mechanobiology of cancer, whereby a switch in the type of interaction with the extracellular matrix dictates the outcome of the cancer cells.
Collapse
Affiliation(s)
- Malak Amer
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Lidan Shi
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| | - Haguy Wolfenson
- Department of Genetics and Developmental Biology, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 31096, Israel
| |
Collapse
|
45
|
Vernerey FJ, Lalitha Sridhar S, Muralidharan A, Bryant SJ. Mechanics of 3D Cell-Hydrogel Interactions: Experiments, Models, and Mechanisms. Chem Rev 2021; 121:11085-11148. [PMID: 34473466 DOI: 10.1021/acs.chemrev.1c00046] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hydrogels are highly water-swollen molecular networks that are ideal platforms to create tissue mimetics owing to their vast and tunable properties. As such, hydrogels are promising cell-delivery vehicles for applications in tissue engineering and have also emerged as an important base for ex vivo models to study healthy and pathophysiological events in a carefully controlled three-dimensional environment. Cells are readily encapsulated in hydrogels resulting in a plethora of biochemical and mechanical communication mechanisms, which recapitulates the natural cell and extracellular matrix interaction in tissues. These interactions are complex, with multiple events that are invariably coupled and spanning multiple length and time scales. To study and identify the underlying mechanisms involved, an integrated experimental and computational approach is ideally needed. This review discusses the state of our knowledge on cell-hydrogel interactions, with a focus on mechanics and transport, and in this context, highlights recent advancements in experiments, mathematical and computational modeling. The review begins with a background on the thermodynamics and physics fundamentals that govern hydrogel mechanics and transport. The review focuses on two main classes of hydrogels, described as semiflexible polymer networks that represent physically cross-linked fibrous hydrogels and flexible polymer networks representing the chemically cross-linked synthetic and natural hydrogels. In this review, we highlight five main cell-hydrogel interactions that involve key cellular functions related to communication, mechanosensing, migration, growth, and tissue deposition and elaboration. For each of these cellular functions, recent experiments and the most up to date modeling strategies are discussed and then followed by a summary of how to tune hydrogel properties to achieve a desired functional cellular outcome. We conclude with a summary linking these advancements and make the case for the need to integrate experiments and modeling to advance our fundamental understanding of cell-matrix interactions that will ultimately help identify new therapeutic approaches and enable successful tissue engineering.
Collapse
Affiliation(s)
- Franck J Vernerey
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States.,Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Shankar Lalitha Sridhar
- Department of Mechanical Engineering, University of Colorado at Boulder, 1111 Engineering Drive, Boulder, Colorado 80309-0428, United States
| | - Archish Muralidharan
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States
| | - Stephanie J Bryant
- Materials Science and Engineering Program, University of Colorado at Boulder, 4001 Discovery Drive, Boulder, Colorado 80309-613, United States.,Department of Chemical and Biological Engineering, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States.,BioFrontiers Institute, University of Colorado at Boulder, 3415 Colorado Avenue, Boulder, Colorado 80309-0596, United States
| |
Collapse
|
46
|
Parisi C, Qin K, Fernandes FM. Colonization versus encapsulation in cell-laden materials design: porosity and process biocompatibility determine cellularization pathways. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2021; 379:20200344. [PMID: 34334019 DOI: 10.1098/rsta.2020.0344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/28/2021] [Indexed: 06/13/2023]
Abstract
Seeding materials with living cells has been-and still is-one of the most promising approaches to reproduce the complexity and the functionality of living matter. The strategies to associate living cells with materials are limited to cell encapsulation and colonization, however, the requirements for these two approaches have been seldom discussed systematically. Here we propose a simple two-dimensional map based on materials' pore size and the cytocompatibility of their fabrication process to draw, for the first time, a guide to building cellularized materials. We believe this approach may serve as a straightforward guideline to design new, more relevant materials, able to seize the complexity and the function of biological materials. This article is part of the theme issue 'Bio-derived and bioinspired sustainable advanced materials for emerging technologies (part 1)'.
Collapse
Affiliation(s)
- Cleo Parisi
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| | - Kankan Qin
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| | - Francisco M Fernandes
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
47
|
Kim J, Jang J, Cho DW. Recapitulating the Cancer Microenvironment Using Bioprinting Technology for Precision Medicine. MICROMACHINES 2021; 12:1122. [PMID: 34577765 PMCID: PMC8472267 DOI: 10.3390/mi12091122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/20/2022]
Abstract
The complex and heterogenous nature of cancer contributes to the development of cancer cell drug resistance. The construction of the cancer microenvironment, including the cell-cell interactions and extracellular matrix (ECM), plays a significant role in the development of drug resistance. Traditional animal models used in drug discovery studies have been associated with feasibility issues that limit the recapitulation of human functions; thus, in vitro models have been developed to reconstruct the human cancer system. However, conventional two-dimensional and three-dimensional (3D) in vitro cancer models are limited in their ability to emulate complex cancer microenvironments. Advances in technologies, including bioprinting and cancer microenvironment reconstruction, have demonstrated the potential to overcome some of the limitations of conventional models. This study reviews some representative bioprinted in vitro models used in cancer research, particularly fabrication strategies for modeling and consideration of essential factors needed for the reconstruction of the cancer microenvironment. In addition, we highlight recent studies that applied such models, including application in precision medicine using advanced bioprinting technologies to fabricate biomimetic cancer models. Furthermore, we discuss current challenges in 3D bioprinting and suggest possible strategies to construct in vitro models that better mimic the pathophysiology of the cancer microenvironment for application in clinical settings.
Collapse
Affiliation(s)
- Jisoo Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
| | - Jinah Jang
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Department of Creative IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| | - Dong-Woo Cho
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea;
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
48
|
Lev-Ari S, Starr AN, Vexler A, Kalich-Philosoph L, Yoo HS, Kwon KR, Yadgar M, Bondar E, Bar-Shai A, Volovitz I, Schwarz Y. Rh2-enriched Korean ginseng (Ginseng Rh2+) inhibits tumor growth and development of metastasis of non-small cell lung cancer. Food Funct 2021; 12:8068-8077. [PMID: 34286798 DOI: 10.1039/d1fo00643f] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BACKGROUND AND OBJECTIVE While there are multiple studies on the anti-tumoral effects of Panax ginseng as active ingredients (one or more ginsenosides derived from the extract) or as a whole plant extract, there is a lack of studies to assess the effects Panax ginseng's of active ingredients combined with the whole plant extract. Our aim was to study the effect of whole ginseng, enriched in the anti-tumoral Rh2 component and other ginsenosides (Ginseng Rh2+), on the metastatic capacity of non-small cell lung cancer (NSCLC). METHODS We evaluated the effects of Ginseng Rh2+ on survival, migration and motility, induction of apoptosis, and expression of its apoptosis-related proteins in non-small cell lung cancer (NSCLC) cells in vitro and on primary tumor growth and metastatic capacity in a syngeneic mouse lung cancer model in vivo. The effects of Ginseng Rh2+ on NSCLC cells were studied in vitro using: a colorimetric tetrazolium salt (XTT) assay, annexin V-FITC/PI, western blotting, wound healing motility assay, Transwell migration and cell adhesion assays. In vivo, mice were inoculated with Lewis mouse lung carcinoma cells subcutaneously to evaluate local tumor growth, or intravenously to evaluate the effects of Ginseng Rh2+ on development of experimental metastases. Mice were treated by intraperitoneal administration of Ginseng Rh2+ (0.005-0.5 g kg-1) on days 6, 10, and 14 after tumor injection. RESULTS We found that Ginseng Rh2+ increased the apoptosis of NSCLC cells in vitro, demonstrating dose dependent down-regulation of the Bcl-2 anti-apoptotic gene and concurrent up-regulation of the Bax pro-apoptotic gene. Ginseng Rh2+ inhibited the tumor cells' capacity to attach to the ECM-related matrix and reduced cell migration. In vivo, Ginseng Rh2+ inhibited local tumor growth and reduced the development of experimental lung metastases. CONCLUSION Our study suggests that Ginseng Rh2+ may potentially be used as a therapeutic agent for treatment of NSCLC.
Collapse
Affiliation(s)
- Shahar Lev-Ari
- Laboratory of Herbal Medicine and Cancer Research, Institute of Oncology, Tel-Aviv Sourasky Medical Center, affiliated to Tel-Aviv University, Israel.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Barbato MG, Pereira RC, Mollica H, Palange A, Ferreira M, Decuzzi P. A permeable on-chip microvasculature for assessing the transport of macromolecules and polymeric nanoconstructs. J Colloid Interface Sci 2021; 594:409-423. [PMID: 33774397 DOI: 10.1016/j.jcis.2021.03.053] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/23/2021] [Accepted: 03/09/2021] [Indexed: 01/19/2023]
Abstract
HYPOTHESIS The selective permeation of molecules and nanomedicines across the diseased vasculature dictates the success of a therapeutic intervention. Yet, in vitro assays cannot recapitulate relevant differences between the physiological and pathological microvasculature. Here, a double-channel microfluidic device was engineered to comprise vascular and extravascular compartments connected through a micropillar membrane with tunable permeability. EXPERIMENTS The vascular compartment was coated by endothelial cells to achieve permeability values ranging from ~0.1 μm/sec, following a cyclic adenosine monophosphate (cAMP) pre-treatment (25 μg/mL), up to ~2 μm/sec, upon exposure to Mannitol, Lexiscan or in the absence of cells. Fluorescent microscopy was used to monitor the vascular behavior of 250 kDa Dextran molecules, 200 nm polystyrene nanoparticles (PB), and 1,000 × 400 nm discoidal polymeric nanoconstructs (DPN), under different permeability and flow conditions. FINDINGS In the proposed on-chip microvasculature, it was confirmed that permeation enhancers could favor the perivascular accumulation of ~200 nm, in a dose and time dependent fashion, while have no effect on larger particles. Moreover, the microfluidic device was used to interrogate the role of particle deformability in vascular dynamics. In the presence of a continuous endothelium, soft DPN attached to the vasculature more avidly at sub-physiological flows (100 μm/sec) than rigid DPN, whose deposition was larger at higher flow rates (1 mm/sec). The proposed double-channel microfluidic device can be efficiently used to systematically analyze the vascular behavior of drug delivery systems to enhance their tissue specific accumulation.
Collapse
Affiliation(s)
- Maria Grazia Barbato
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; Department of Informatics, Bioengineering, Robotics and System Engineering (DIBRIS), University of Genoa, Via Dodecaneso 25, 16146 Genoa, Italy
| | - Rui C Pereira
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Hilaria Mollica
- I.R.C.C.S. Istituto Giannina Gaslini, Via Gerolamo Gaslini 3, 16147 Genoa, Italy
| | - AnnaLisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy.
| |
Collapse
|
50
|
Transcending toward Advanced 3D-Cell Culture Modalities: A Review about an Emerging Paradigm in Translational Oncology. Cells 2021; 10:cells10071657. [PMID: 34359827 PMCID: PMC8304089 DOI: 10.3390/cells10071657] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is a disorder characterized by an uncontrollable overgrowth and a fast-moving spread of cells from a localized tissue to multiple organs of the body, reaching a metastatic state. Throughout years, complexity of cancer progression and invasion, high prevalence and incidence, as well as the high rise in treatment failure cases leading to a poor patient prognosis accounted for continuous experimental investigations on animals and cellular models, mainly with 2D- and 3D-cell culture. Nowadays, these research models are considered a main asset to reflect the physiological events in many cancer types in terms of cellular characteristics and features, replication and metastatic mechanisms, metabolic pathways, biomarkers expression, and chemotherapeutic agent resistance. In practice, based on research perspective and hypothesis, scientists aim to choose the best model to approach their understanding and to prove their hypothesis. Recently, 3D-cell models are seen to be highly incorporated as a crucial tool for reflecting the true cancer cell microenvironment in pharmacokinetic and pharmacodynamics studies, in addition to the intensity of anticancer drug response in pharmacogenomics trials. Hence, in this review, we shed light on the unique characteristics of 3D cells favoring its promising usage through a comparative approach with other research models, specifically 2D-cell culture. Plus, we will discuss the importance of 3D models as a direct reflector of the intrinsic cancer cell environment with the newest multiple methods and types available for 3D-cells implementation.
Collapse
|