1
|
Park G, Grey JA, Mourkioti F, Han WM. 3D Mechanical Confinement Directs Muscle Stem Cell Fate and Function. Adv Biol (Weinh) 2025; 9:e2400717. [PMID: 40040295 PMCID: PMC12001014 DOI: 10.1002/adbi.202400717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/07/2025] [Indexed: 03/06/2025]
Abstract
Muscle stem cells (MuSCs) play a crucial role in skeletal muscle regeneration, residing in a niche that undergoes dimensional and mechanical changes throughout the regeneration process. This study investigates how 3D confinement and stiffness encountered by MuSCs during the later stages of regeneration regulate their function, including stemness, activation, proliferation, and differentiation. An asymmetric 3D hydrogel bilayer platform is engineered with tunable physical constraints to mimic the regenerating MuSC niche. These results demonstrate that increased 3D confinement maintains Pax7 expression, reduces MuSC activation and proliferation, inhibits differentiation, and is associated with smaller nuclear size and decreased H4K16ac levels, suggesting that mechanical confinement modulates both nuclear architecture and epigenetic regulation. MuSCs in unconfined 2D environments exhibit larger nuclei and higher H4K16ac expression compared to those in more confined 3D conditions, leading to progressive activation, expansion, and myogenic commitment. This study highlights the importance of 3D mechanical cues in MuSC fate regulation, with 3D confinement acting as a mechanical brake on myogenic commitment, offering novel insights into the mechano-epigenetic mechanisms that govern MuSC behavior during muscle regeneration.
Collapse
Affiliation(s)
- GaYoung Park
- Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Josh A. Grey
- Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Department of CellDevelopmentand Regenerative BiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Black Family Stem Cell InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Institute for Regenerative MedicineIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| | - Foteini Mourkioti
- Department of Orthopaedic SurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Department of Cell and Developmental BiologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Institute for Regenerative MedicineMusculoskeletal ProgramPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Woojin M. Han
- Department of OrthopaedicsIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Department of CellDevelopmentand Regenerative BiologyIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Black Family Stem Cell InstituteIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
- Institute for Regenerative MedicineIcahn School of Medicine at Mount SinaiNew YorkNY10029USA
| |
Collapse
|
2
|
Novak CM, Wheat JS, Ghadiali SN, Ballinger MN. Mechanomemory of pulmonary fibroblasts demonstrates reversibility of transcriptomics and contraction phenotypes. Biomaterials 2025; 314:122830. [PMID: 39276408 DOI: 10.1016/j.biomaterials.2024.122830] [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: 03/27/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
Fibroblasts are cells responsible for producing extracellular matrix (ECM) components, which provides physical support for organs. Although these mesenchymal cells are responsive to mechanical cues in their environment, the permanence of these mechanophenotypes is not well defined. We investigated the mechanomemory of lung fibroblasts and determined how switching culture conditions modulate cell responses and function. Primary murine lung fibroblasts were isolated and cultured on 2D tissue culture plates or within 3D collagen hydrogels and were then passaged within the same or opposite culture condition to assess changes in gene expression, protein production, fibroblast subpopulation, contractile behavior, and traction forces. Compared to fibroblasts isolated on 2D tissue culture plates, fibroblasts within 3D hydrogels exhibited a decreased activation phenotype including reduced contraction profiles, diminished cell traction forces and decreased αSMA gene expression. Cells initially isolated via 2D culture and then cultured in 3D hydrogels exhibited a reversal in activation phenotype as measured by gene expression and contraction profiles. Bulk RNAseq identified groups of genes that exhibit reversible and non-reversable expression patterns. Overall, these findings indicate that lung fibroblasts have a mechanical memory that is altered by culture condition and can be reversible through precondition of cells within a softer 3D microenvironment.
Collapse
Affiliation(s)
- Caymen M Novak
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, 473 West 12th Ave, Columbus, OH, 43210, USA; Department of Mechanical Engineering, Bioengineering Program, University of Michigan Dearborn, 4901 Evergreen Rd, Dearborn, MI, 48128, USA.
| | - Jana S Wheat
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, 473 West 12th Ave, Columbus, OH, 43210, USA
| | - Samir N Ghadiali
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, 473 West 12th Ave, Columbus, OH, 43210, USA; Department of Biomedical Engineering, The Ohio State University, 140 West 19th Avenue Columbus, OH, 43210, USA
| | - Megan N Ballinger
- Dorothy M. Davis Heart and Lung Research Institute, Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Wexner Medical Center, The Ohio State University, 473 West 12th Ave, Columbus, OH, 43210, USA
| |
Collapse
|
3
|
Rudzinska-Radecka M, Turos-Korgul L, Mukherjee D, Podszywalow-Bartnicka P, Piwocka K, Guzowski J. High-throughput formulation of reproducible 3D cancer microenvironments for drug testing in myeloid leukemia. Biofabrication 2024; 17:015035. [PMID: 39622161 DOI: 10.1088/1758-5090/ad998d] [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: 06/28/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Leukemic microenvironment has been recognized as a factor that strongly supports the mechanisms of resistance. Therefore, targeting the microenvironment is currently one of the major directions in drug development and preclinical studies in leukemia. Despite the variety of available leukemia 3D culture models, the reproducible generation of miniaturized leukemic microenvironments, suitable for high-throughput drug testing, has remained a challenge. Here, we use droplet microfluidics to generate tens of thousands of highly monodisperse leukemic-bone marrow microenvironments within minutes. We employ gelatin methacryloyl (GelMA) as a model extracellular matrix (ECM) and tune the concentration of the biopolymer, check the impact of other components of the ECM (hyaluronic acid), cell concentration and the ratio of leukemic cells to bone marrow cells within the microbeads to establish the optimal conditions for microtissue formation. We administer model kinase inhibitor, imatinib, at various concentrations to the encapsulated leukemic microtissues, and, via comparing mono- and co-culture conditions (cancer alone vs cancer-stroma), we find that the stroma-leukemia crosstalk systematically protects the encapsulated cells against the drug-induced cytotoxicity. With that we demonstrate that our system mimics the physiological stroma-dependent protection. We discuss applicability of our model to (i) studying the role of direct- or close-contact interactions between the leukemia and bone marrow cells embedded in microscale 3D ECM on the stroma-mediated protection, and (ii) high-throughput screening of anti-cancer therapeutics in personalized leukemia therapies.
Collapse
Affiliation(s)
- M Rudzinska-Radecka
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
- Equal contribution.v
| | - L Turos-Korgul
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., Warsaw 02-093, Poland
- Equal contribution.v
| | - D Mukherjee
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - P Podszywalow-Bartnicka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., Warsaw 02-093, Poland
| | - K Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., Warsaw 02-093, Poland
| | - J Guzowski
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
4
|
Elhashani S, Glenn M, Raymant M, Schmid MC, Mielgo A. Expression of versican isoforms V0/V1 by pancreatic cancer associated fibroblasts increases fibroblast proliferation. Pancreatology 2024; 24:719-731. [PMID: 38719756 DOI: 10.1016/j.pan.2024.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 04/17/2024] [Accepted: 04/23/2024] [Indexed: 07/15/2024]
Abstract
BACKGROUND Versican is a large extracellular matrix (ECM) proteoglycan with four isoforms V0-3. Elevated V0/V1 levels in breast cancer and glioma regulate cell migration and proliferation, but the role of versican in pancreatic ductal adenocarcinoma (PDAC) remains unclear. METHODS In this study, we evaluated the expression levels of versican isoforms, as well as their cellular source and interacting partners, in vivo, in human and mouse primary and metastatic PDAC tumours and in vitro, in pancreatic tumour cells and fibroblasts using immunostaining, confocal microscopy and qPCR techniques. We also investigated the effect of versican expression on fibroblast proliferation and migration using genetic and pharmacological approaches. RESULTS We found that versican V0/V1 is highly expressed by cancer-associated fibroblasts (CAFs) in mouse and human primary and metastatic PDAC tumours. Our data also show that exposing fibroblasts to tumour-conditioned media upregulates V0 and V1 expressions, while Verbascoside (a CD44 inhibitor) downregulates V0/V1 expression. Importantly, V0/V1 knockdown significantly inhibits fibroblast proliferation. Mechanistically, we found that inhibiting hyaluronan synthesis does not affect versican co-localisation with CD44 in fibroblasts. CONCLUSION CAFs express high levels of versican V0/V1 in primary and liver metastatic PDAC tumours and versican V0/V1 supports fibroblast proliferation.
Collapse
Affiliation(s)
- Sufyan Elhashani
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Mark Glenn
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Meirion Raymant
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Michael C Schmid
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK
| | - Ainhoa Mielgo
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Ashton Street, Liverpool, L69 3GE, UK.
| |
Collapse
|
5
|
Ferretti L, Moccia V, Centelleghe C, Venerando A, Dettin M, Sieni E, Zamuner A, Caicci F, Castagnaro M, Zappulli V, Mazzariol S. Bottlenose dolphin (Tursiops truncatus) immortalized fibroblasts on novel 3D in vitro collagen-free scaffolds. PLoS One 2024; 19:e0304992. [PMID: 38861523 PMCID: PMC11166351 DOI: 10.1371/journal.pone.0304992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 05/22/2024] [Indexed: 06/13/2024] Open
Abstract
Dolphins, as apex predators, can be considered relevant sentinels of the health of marine ecosystems. The creation of 3D cell models to assess in vitro cell-to-cell and cell-to-matrix interactions in environmental-mimicking conditions, is of considerable interest. However, to date the establishment of cetacean 3D culture systems has not yet been accomplished. Thus, in this study, different 3D systems of bottlenose dolphin (Tursiops truncatus) skin fibroblasts have been analyzed. Particularly, novel scaffolds based on hyaluronic acid and ionic-complementary self-assembling peptides such as RGD-EAbuK and EAbuK-IKVAV have been compared to Matrigel. Histological and fluorescent staining, electron microscopy (TEM) analyses and viability assays have been performed and RT-PCR has been used to detect extracellular matrix (ECM) components produced by cells. Results showed that Matrigel induced cells to form aggregates with lower viability and no ECM production compared to the novel scaffolds. Moreover, scaffolds allowed dispersed cells to produce a collagenous ECM containing collagen1a1, laminin B1 and elastin. The HA-EAbuK-IKVAV scaffold resulted in the most suitable 3D model in terms of cell quantity and viability. The development of this innovative approach is the first step towards the possibility to create 3D in vitro models for this protected species.
Collapse
Affiliation(s)
- Lucrezia Ferretti
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Valentina Moccia
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Cinzia Centelleghe
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Andrea Venerando
- Department of Agrifood, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - Monica Dettin
- Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Elisabetta Sieni
- Department of Theoretical and Applied Sciences, Insubria University, Varese, Italy
| | - Annj Zamuner
- Department of Industrial Engineering, University of Padova, Padova, Italy
- Department of Civil, Environmental, and Architectural Engineering, University of Padova, Padova, Italy
| | | | - Massimo Castagnaro
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Valentina Zappulli
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| | - Sandro Mazzariol
- Department of Comparative Biomedicine and Food Science, University of Padua, Legnaro, Italy
| |
Collapse
|
6
|
Cassel SE, Huntington BM, Chen W, Lei P, Andreadis ST, Kloxin AM. Dynamic reporters for probing real-time activation of human fibroblasts from single cells to populations. APL Bioeng 2024; 8:026127. [PMID: 38938687 PMCID: PMC11209894 DOI: 10.1063/5.0166152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 06/06/2024] [Indexed: 06/29/2024] Open
Abstract
Activation of fibroblasts is pivotal for wound healing; however, persistent activation leads to maladaptive processes and is a hallmark of fibrosis, where disease mechanisms are only partially understood. Human in vitro model systems complement in vivo animal models for both hypothesis testing and drug evaluation to improve the identification of therapeutics relevant to human disease. Despite advances, a challenge remains in understanding the dynamics of human fibroblast responses to complex microenvironment stimuli, motivating the need for more advanced tools to investigate fibrotic mechanisms. This work established approaches for assessing the temporal dynamics of these responses using genetically encoded fluorescent reporters of alpha smooth muscle actin expression, an indicator of fibroblast activation. Specifically, we created a toolset of human lung fibroblast reporter cell lines from different origins (male, female; healthy, idiopathic pulmonary fibrosis) and used three different versions of the reporter with the fluorescent protein modified to exhibit different temporal stabilities, providing temporal resolution of protein expression processes over a range of timescales. Using this toolset, we demonstrated that reporters provide insight into population shifts in response to both mechanical and biochemical cues that are not detectable by traditional end point assessments with differential responses based on cell origin. Furthermore, individual cells can also be tracked over time, with opportunities for comparison to complementary end point measurements. The establishment of this reporter toolset enables dynamic cell investigations that can be translated into more complex synthetic culture environments for elucidating disease mechanisms and evaluating therapeutics for lung fibrosis and other complex biological processes more broadly.
Collapse
Affiliation(s)
- Samantha E. Cassel
- Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA
| | - Breanna M. Huntington
- Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA
| | - Wilfred Chen
- Chemical and Biomolecular Engineering, University of Delaware, Newark, Delaware 19716, USA
| | - Pedro Lei
- Chemical and Biological Engineering, University at Buffalo, Buffalo, New York 14260-4200, USA
| | - Stelios T. Andreadis
- Chemical and Biological Engineering, University at Buffalo, Buffalo, New York 14260-4200, USA
| | | |
Collapse
|
7
|
Chou Y, Alfarafisa NM, Ikezawa M, Khairani AF. Progress in the Development of Stem Cell-Derived Cell-Free Therapies for Skin Aging. Clin Cosmet Investig Dermatol 2023; 16:3383-3406. [PMID: 38021432 PMCID: PMC10676866 DOI: 10.2147/ccid.s434439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023]
Abstract
Introduction The skin is a vital organ as the body's largest barrier, but its function declines with aging. Therefore, research into effective regeneration treatments must continue to advance. Stem cell transplantation, a cell-based therapy, has become a popular skin-aging treatment, although it comes with drawbacks like host immune reactions. Stem cell-derived cell-free therapies have emerged as an alternative, backed by promising preclinical findings. Stem cell secretomes and extracellular vesicles (EVs) are the key components in cell-free therapy from stem cells. However, comprehensive reviews on the mechanisms of such treatments for skin aging are still limited. Purpose This review discusses stem cell-derived cell-free therapy's potential mechanisms of action related to skin aging prevention by identifying specific molecular targets suitable for the interventions. Methods A search identified 27 relevant in vitro studies on stem cell-derived cell-free therapy interventions in skin aging model cells without restricting publication years using PubMed, Scopus, and Google Scholar. Results Stem cell-derived cell-free therapy can prevent skin aging through various mechanisms, such as (1) involvement of multiple regenerative pathways [NFkb, AP-1, MAPK, P-AKT, NRF2, SIRT-1]; (2) oxidative stress regulation [by reducing oxidants (HO-1, NQO1) and enhancing antioxidants (SOD1, CAT, GP, FRAP)]; (3) preventing ECM degradation [by increasing elastin, collagen, HA, TIMP, and reducing MMP]; (4) regulating cell activity [by reducing cell senescence (SA-β-gal), apoptosis, and cell cycle arrest (P53, P12, P16); and enhancing autophagy, cell migration, and cell proliferation (Ki67)] (5) Regulating the inflammatory pathway [by reducing IL-6, IL-1, TNF-⍺, and increasing TGF-β]. Several clinical trials have also revealed improvements in wrinkles, elasticity, hydration, pores, and pigmentation. Conclusion Stem cell-derived cell-free therapy is a potential novel treatment for skin aging by cell rejuvenation through various molecular mechanisms.
Collapse
Affiliation(s)
- Yoan Chou
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Nayla Majeda Alfarafisa
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| | - Maiko Ikezawa
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Gunma University, Gunma, Japan
| | - Astrid Feinisa Khairani
- Graduate School of Master Program in Anti Aging and Aesthetic Medicine, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, West Java, Indonesia
| |
Collapse
|
8
|
Çapkın Yurtsever M, Güldağ G. TiO 2, CeO 2, and TiO 2-CeO 2 nanoparticles incorporated 2.5D chitosan hydrogels: Gelation behavior and cytocompatibility. J Mech Behav Biomed Mater 2023; 146:106088. [PMID: 37619284 DOI: 10.1016/j.jmbbm.2023.106088] [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: 06/12/2023] [Revised: 08/17/2023] [Accepted: 08/20/2023] [Indexed: 08/26/2023]
Abstract
In this study, gelation behavior and cytocompatibility of 2.5D chitosan hydrogels were investigated in the presence of TiO2, CeO2 and TiO2-CeO2 composite nanoparticles. Chemical co-precipitation method was used for nanoparticle synthesis and they were heat treated at 600 °C and 700 °C. Gelation of the chitosan solutions was carried out at 37 °C in the presence of glycerol phosphate and genipin as crosslinkers. The gelation time of chitosan was decreased by all of the nanoparticles whereas its elastic modulus was increased by nanoparticles addition. Chitosan solutions containing CeO2 or TiO2-CeO2 nanoparticles showed faster gel formation compared to chitosan solutions containing only TiO2 nanoparticles. CeO2@700 °C nanoparticles decreased the gelation time by 46% and increased elastic modulus by 14%. Average pore diameter of the hydrogel decreased from 127 ± 62 μm to 77 ± 33 μm, water uptake decreased 21% and thermal stability increased in the presence of CeO2@700 °C nanoparticles compared to chitosan hydrogel. Cell viability results indicated that chitosan hydrogels with or without nanoparticles created 2.5D environment supporting cellular proliferation approximately 1.5 times more than TCPS due to their high porous surfaces. Immunofluorescence images were also supported cell viability results. Therefore, CeO2 or TiO2-CeO2 composite nanoparticles incorporated 2.5D chitosan hydrogels may be alternative tissue engineering materials with their fast gelation, ease of use, low cost, light transparency, and cytocompatibility.
Collapse
Affiliation(s)
- Merve Çapkın Yurtsever
- Faculty of Engineering, Department of Bioengineering, Adana Alparslan Türkeş Science and Technology University, Adana, Türkiye.
| | - Gözde Güldağ
- Faculty of Engineering, Department of Bioengineering, Adana Alparslan Türkeş Science and Technology University, Adana, Türkiye
| |
Collapse
|
9
|
Louback RDA, Martins-Cardoso K, Tinoco LW, Collino F, de Barros APDN, Fortuna-Costa A, Monteiro RQ, Rossi MID, Lindoso RS. Aspirin Affects MDA-MB-231 Vesicle Production and Their Capacity to Induce Fibroblasts towards a Pro-Invasive State. Int J Mol Sci 2023; 24:12020. [PMID: 37569393 PMCID: PMC10419278 DOI: 10.3390/ijms241512020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/19/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Long-term administration of aspirin (ASA, acetylsalicylic acid) in oncogenic patients has been related to a reduction in cancer risk incidence, but its precise mechanism of action is unclear. The activation of cancer-associated fibroblasts (CAFs) is a key element in tumor progression and can be triggered by cancer-derived extracellular vesicles (EVs). Targeting the communication between cancer cells and the surrounding tumor microenvironment (TME) may control cancer progression. Our aim was to investigate the effect of ASA on breast cancer cells, focusing on EV secretion and their effect on the biological properties of CAFs. As a result, ASA was shown to reduce the amount and alter the size distribution of EVs produced by MDA-MB-231 tumor cells. Fibroblasts stimulated with EVs derived from MDA-MB-231 treated with ASA (EV-ASA) showed a lower expression of alpha-smooth muscle actin (α-SMA), matrix metalloproteinase-2 (MMP2) but not fibroblast activation protein (FAP) in respect to the ones stimulated with EVs from untreated breast cancer cells (EV-CTR). Furthermore, invasion assays using a three-dimensional (3D) fibroblast spheroid model showed reduced MDA-MB-231 invasion towards fibroblast spheroids pretreated with EV-ASA as compared to spheroids prepared with EV-CTR-stimulated fibroblasts. This suggests that ASA partially inhibits the ability of tumor EVs to stimulate CAFs to promote cancer invasion. In conclusion, ASA can interfere with tumor communication by reducing EV secretion by breast tumor cells as well as by interfering with their capacity to stimulate fibroblasts to become CAFs.
Collapse
Affiliation(s)
- Rafaela de Assiz Louback
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941590, Brazil; (R.d.A.L.); (A.P.D.N.d.B.); (A.F.-C.)
- Clementino Fraga Filho University Hospiyal, Federal University of Rio de Janeiro, Rio de Janeiro 21941913, Brazil
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941590, Brazil; (K.M.-C.); (R.Q.M.)
| | - Karina Martins-Cardoso
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941590, Brazil; (K.M.-C.); (R.Q.M.)
| | - Luzineide W. Tinoco
- Laboratory for Analysis and Development of Enzyme Inhibitors, Natural Products Research Institute, Federal University of Rio de Janeiro, Rio de Janeiro 21941590, Brazil;
| | - Federica Collino
- Department of Clinical Sciences and Community Health, University of Milano, 20122 Milan, Italy;
- Laboratory of Translational Research in Paediatric Nephro-Urology, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Ana Paula D. N. de Barros
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941590, Brazil; (R.d.A.L.); (A.P.D.N.d.B.); (A.F.-C.)
- Clementino Fraga Filho University Hospiyal, Federal University of Rio de Janeiro, Rio de Janeiro 21941913, Brazil
| | - Anneliese Fortuna-Costa
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941590, Brazil; (R.d.A.L.); (A.P.D.N.d.B.); (A.F.-C.)
- Clementino Fraga Filho University Hospiyal, Federal University of Rio de Janeiro, Rio de Janeiro 21941913, Brazil
| | - Robson Q. Monteiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro 21941590, Brazil; (K.M.-C.); (R.Q.M.)
| | - Maria Isabel Doria Rossi
- Institute of Biomedical Sciences, Federal University of Rio de Janeiro, Rio de Janeiro 21941590, Brazil; (R.d.A.L.); (A.P.D.N.d.B.); (A.F.-C.)
- Clementino Fraga Filho University Hospiyal, Federal University of Rio de Janeiro, Rio de Janeiro 21941913, Brazil
| | - Rafael Soares Lindoso
- Carlos Chagas Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro 21941902, Brazil
- National Center of Science and Technology for Regenerative Medicine/REGENERA, Federal University of Rio de Janeiro, Rio de Janeiro 21941902, Brazil
| |
Collapse
|
10
|
McKenzie TJ, Cawood C, Davis C, Ayres N. Synthesis of patterned polyHIPE-hydrogel composite materials using thiol-ene chemistry. J Colloid Interface Sci 2023; 645:502-512. [PMID: 37159992 DOI: 10.1016/j.jcis.2023.04.132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 04/17/2023] [Accepted: 04/24/2023] [Indexed: 05/11/2023]
Abstract
Elastomeric materials combining multiple properties within a single composite are highly desired in applications including biomaterials interfaces, actuators, and soft robotics. High spatial resolution is required to impart different properties across the composite for the intended application, but many techniques used to prepare these composites rely on multistep and complex methods. There is a need for the development of simple and efficient platforms to design layered composite materials. Here, we report the synthesis of horizontally- and vertically-patterned composites consisting of PDMS-based polymerized high internal phase emulsion (polyHIPE) porous elastomers and PDMS/PEG hydrogels. Composites with defined interfaces that were mechanically robust were prepared, and rheological analysis of the polyHIPE and hydrogel layers showed storage moduli values of ∼ 35 kPa and 45 kPa respectively. The compressive Young's Modulus and maximum strain of the polyHIPEs were dependent on the thiol to ene ratio in the formulation and obtained values ranging from 6 to 25 kPa and 50-65% respectively. The mechanical properties, total porosity of the polyHIPE, and swelling ratio of the hydrogel were unaffected by the patterning technique compared to non-patterned controls. PolyHIPE-hydrogel composite materials having up to 7-different horizontally pattered layers could be prepared that could expand and contract up hydration and drying.
Collapse
Affiliation(s)
- Tucker J McKenzie
- Department of Chemistry, The University of Cincinnati, P.O. Box 210172, Cincinnati, OH 45221, United States
| | - Christian Cawood
- Department of Chemistry, The University of Cincinnati, P.O. Box 210172, Cincinnati, OH 45221, United States
| | - Chelsea Davis
- Department of Chemistry, The University of Cincinnati, P.O. Box 210172, Cincinnati, OH 45221, United States
| | - Neil Ayres
- Department of Chemistry, The University of Cincinnati, P.O. Box 210172, Cincinnati, OH 45221, United States.
| |
Collapse
|
11
|
Woodley JP, Lambert DW, Asencio IO. Reduced Fibroblast Activation on Electrospun Polycaprolactone Scaffolds. Bioengineering (Basel) 2023; 10:bioengineering10030348. [PMID: 36978739 PMCID: PMC10045272 DOI: 10.3390/bioengineering10030348] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/14/2023] Open
Abstract
In vivo, quiescent fibroblasts reside in three-dimensional connective tissues and are activated in response to tissue injury before proliferating rapidly and becoming migratory and contractile myofibroblasts. When deregulated, chronic activation drives fibrotic disease. Fibroblasts cultured on stiff 2D surfaces display a partially activated phenotype, whilst many 3D environments limit fibroblast activation. Cell mechanotransduction, spreading, polarity, and integrin expression are controlled by material mechanical properties and micro-architecture. Between 3D culture systems, these features are highly variable, and the challenge of controlling individual properties without altering others has led to an inconsistent picture of fibroblast behaviour. Electrospinning offers greater control of mechanical properties and microarchitecture making it a valuable model to study fibroblast activation behaviour in vitro. Here, we present a comprehensive characterisation of the activation traits of human oral fibroblasts grown on a microfibrous scaffold composed of electrospun polycaprolactone. After over 7 days in the culture, we observed a reduction in proliferation rates compared to cells cultured in 2D, with low KI67 expression and no evidence of cellular senescence. A-SMA mRNA levels fell, and the expression of ECM protein-coding genes also decreased. Electrospun fibrous scaffolds, therefore, represent a tuneable platform to investigate the mechanisms of fibroblast activation and their roles in fibrotic disease.
Collapse
|
12
|
Li S, Sun Y, Chen Y, Lu J, Jiang G, Yu K, Wu Y, Mao Y, Jin H, Luo J, Dong S, Hu B, Ding Y, Liu A, Shen Y, Feng G, Yan S, He Y, Yan R. Sandwich Biomimetic Scaffold Based Tendon Stem/Progenitor Cell Alignment in a 3D Microenvironment for Functional Tendon Regeneration. ACS APPLIED MATERIALS & INTERFACES 2023; 15:4652-4667. [PMID: 36698266 DOI: 10.1021/acsami.2c16584] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Tendon injuries are some of the most commonly diagnosed musculoskeletal diseases. Tendon regeneration is sensitive to the topology of the substitute as it affects the cellular microenvironment and homeostasis. To bionic in vivo three-dimensional (3D) aligned microenvironment, an ordered 3D sandwich model was used to investigate the cell response in the tendon. First, high-resolution 3D printing provided parallel-grooved topographical cues on the hydrogel surface. Then the cells were seeded on its surface to acquire a 2D model. Afterward, an additional hydrogel coating layer was applied to the cells to create the 3D model. The interaction between cells and order structures in three-dimensions is yet to be explored. The study found that the tendon stem/progenitor cells (TSPCs) still maintain their ordering growth in the 3D model as in the 2D model. The study also found that the 3D-aligned TSPCs exhibited enhanced tenogenic differentiation through the PI3K-AKT signaling pathway and presented a less inflammatory phenotype than those in the 2D model. The in vivo implantation of such a 3D-aligned TSPC composite promoted tendon regeneration and mitigated heterotopic ossification in an Achilles defect model. These findings demonstrated that 3D-aligned TSPCs within a biomimetic topology environment are promising for functional tendon regeneration.
Collapse
Affiliation(s)
- Sihao Li
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Yuan Sun
- State Key Laboratory of Fluid Power and Mechatronic Systems, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, 450002, China
| | - Yazhou Chen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Jinwei Lu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Guangyao Jiang
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Kang Yu
- State Key Laboratory of Fluid Power and Mechatronic Systems, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, 450002, China
| | - Yifan Wu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Yufei Mao
- Medical College of Tianjin University, Tianjin University, Tianjin, 300072, China
| | - Hao Jin
- Key Lab. of Advanced Micro/Nano Electronic Devices & Smart Systems of Zhejiang, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Jikui Luo
- Key Lab. of Advanced Micro/Nano Electronic Devices & Smart Systems of Zhejiang, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Shurong Dong
- Key Lab. of Advanced Micro/Nano Electronic Devices & Smart Systems of Zhejiang, College of Information Science and Electronic Engineering, Zhejiang University, Hangzhou, 310027, China
- ZJU-Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, 311200, China
| | - Bin Hu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Yi Ding
- Basic Medical College, Naval Medical University, Shanghai, 200433, China
| | - An Liu
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Yu Shen
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Gang Feng
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Shigui Yan
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, College of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, 450002, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Ruijian Yan
- Department of Orthopedic Surgery, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310000, China
- Key Laboratory of Motor System Disease Research and Precision Therapy of Zhejiang Province, Hangzhou, 310000, China
- Clinical Research Center of Motor System Disease of Zhejiang Province, Hangzhou, 310000, China
| |
Collapse
|
13
|
Davis-Hall D, Thomas E, Peña B, Magin CM. 3D-bioprinted, phototunable hydrogel models for studying adventitial fibroblast activation in pulmonary arterial hypertension. Biofabrication 2022; 15:10.1088/1758-5090/aca8cf. [PMID: 36533728 PMCID: PMC9933849 DOI: 10.1088/1758-5090/aca8cf] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 12/05/2022] [Indexed: 12/10/2022]
Abstract
Pulmonary arterial hypertension (PAH) is a progressive disease of the lung vasculature, characterized by elevated pulmonary blood pressure, remodeling of the pulmonary arteries, and ultimately right ventricular failure. Therapeutic interventions for PAH are limited in part by the lack ofin vitroscreening platforms that accurately reproduce dynamic arterial wall mechanical properties. Here we present a 3D-bioprinted model of the pulmonary arterial adventitia comprised of a phototunable poly(ethylene glycol) alpha methacrylate (PEG-αMA)-based hydrogel and primary human pulmonary artery adventitia fibroblasts (HPAAFs). This unique biomaterial emulates PAH pathogenesisin vitrothrough a two-step polymerization reaction. First, PEG-αMA macromer was crosslinked off-stoichiometry by 3D bioprinting an acidic bioink solution into a basic gelatin support bath initiating a base-catalyzed thiol-ene reaction with synthetic and biodegradable crosslinkers. Then, matrix stiffening was induced by photoinitiated homopolymerization of unreacted αMA end groups. A design of experiments approach produced a hydrogel platform that exhibited an initial elastic modulus (E) within the range of healthy pulmonary arterial tissue (E= 4.7 ± 0.09 kPa) that was stiffened to the pathologic range of hypertensive tissue (E= 12.8 ± 0.47 kPa) and supported cellular proliferation over time. A higher percentage of HPAAFs cultured in stiffened hydrogels expressed the fibrotic marker alpha-smooth muscle actin than cells in soft hydrogels (88 ± 2% versus 65 ± 4%). Likewise, a greater percentage of HPAAFs were positive for the proliferation marker 5-ethynyl-2'-deoxyuridine (EdU) in stiffened models (66 ± 6%) compared to soft (39 ± 6%). These results demonstrate that 3D-bioprinted, phototunable models of pulmonary artery adventitia are a tool that enable investigation of fibrotic pathogenesisin vitro.
Collapse
Affiliation(s)
- Duncan Davis-Hall
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, United States of America
| | - Emily Thomas
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States of America
| | - Brisa Peña
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, United States of America
- CU-Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, United States of America
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States of America
| |
Collapse
|
14
|
Wang Y, Singer R, Liu X, Inman SJ, Cao Q, Zhou Q, Noble A, Li L, Arizpe Tafoya AV, Babi M, Ask K, Kolb MR, Ramsay S, Geng F, Zhang B, Shargall Y, Moran-Mirabal JM, Dabaghi M, Hirota JA. The CaT stretcher: An open-source system for delivering uniaxial strain to cells and tissues (CaT). Front Bioeng Biotechnol 2022; 10:959335. [PMID: 36329705 PMCID: PMC9622803 DOI: 10.3389/fbioe.2022.959335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 09/29/2022] [Indexed: 01/23/2025] Open
Abstract
Integration of mechanical cues in conventional 2D or 3D cell culture platforms is an important consideration for in vivo and ex vivo models of lung health and disease. Available commercial and published custom-made devices are frequently limited in breadth of applications, scalability, and customization. Herein we present a technical report on an open-source, cell and tissue (CaT) stretcher, with modularity for different in vitro and ex vivo systems, that includes the following features: 1) Programmability for modeling different breathing patterns, 2) scalability to support low to high-throughput experimentation, and 3) modularity for submerged cell culture, organ-on-chips, hydrogels, and live tissues. The strategy for connecting the experimental cell or tissue samples to the stretching device were designed to ensure that traditional biomedical outcome measurements including, but not limited to microscopy, soluble mediator measurement, and gene and protein expression remained possible. Lastly, to increase the uptake of the device within the community, the system was built with economically feasible and available components. To accommodate diverse in vitro and ex vivo model systems we developed a variety of chips made of compliant polydimethylsiloxane (PDMS) and optimized coating strategies to increase cell adherence and viability during stretch. The CaT stretcher was validated for studying mechanotransduction pathways in lung cells and tissues, with an increase in alpha smooth muscle actin protein following stretch for 24 h observed in independent submerged monolayer, 3D hydrogel, and live lung tissue experiments. We anticipate that the open-source CaT stretcher design will increase accessibility to studies of the dynamic lung microenvironment through direct implementation by other research groups or custom iterations on our designs.
Collapse
Affiliation(s)
- Yushi Wang
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Ryan Singer
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
| | - Xinyue Liu
- Department of Materials Science and Engineering, University of Toronto, Toronto, ON, Canada
| | - Seth J. Inman
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Quynh Cao
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Quan Zhou
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Alex Noble
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Laura Li
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Aidee Verónica Arizpe Tafoya
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
| | - Mouhanad Babi
- Centre for Advanced Light Microscopy, McMaster University, Hamilton, ON, Canada
| | - Kjetil Ask
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Michael G. DeGroote Centre for Learning and Discovery, Hamilton, ON, Canada
| | - Martin R. Kolb
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
| | - Scott Ramsay
- Department of Materials Science and Engineering, University of Toronto, Toronto, ON, Canada
| | - Fei Geng
- W Booth School of Engineering Practice and Technology, McMaster University, Hamilton, ON, Canada
| | - Boyang Zhang
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
- Department of Chemical Engineering, McMaster University, Hamilton, ON, Canada
| | - Yaron Shargall
- Division of Thoracic Surgery, Department of Surgery, McMaster University, Hamilton, ON, Canada
| | - Jose Manuel Moran-Mirabal
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, ON, Canada
- Centre for Advanced Light Microscopy, McMaster University, Hamilton, ON, Canada
| | - Mohammadhossein Dabaghi
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
| | - Jeremy A. Hirota
- Department of Medicine, Firestone Institute for Respiratory Health—Division of Respirology, McMaster University, Hamilton, ON, Canada
- School of Biomedical Engineering, McMaster University, Hamilton, ON, Canada
- McMaster Immunology Research Centre, McMaster University, Michael G. DeGroote Centre for Learning and Discovery, Hamilton, ON, Canada
- Department of Medicine, Division of Respiratory Medicine, University of British Columbia, Vancouver, BC, Canada
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
15
|
Bomb K, Pradhan L, Zhang Q, Jarai BM, Bhattacharjee A, Burris DL, Kloxin AM, Fromen CA. Destructive fibrotic teamwork: how both microenvironment stiffness and profibrotic interleukin 13 impair alveolar macrophage phenotype and function. Biomater Sci 2022; 10:5689-5706. [PMID: 36018297 PMCID: PMC9632634 DOI: 10.1039/d2bm00828a] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The pulmonary fibrotic microenvironment is characterized by increased stiffness of lung tissue and enhanced secretion of profibrotic soluble cues contributing to a feedback loop that leads to dysregulated wound healing and lung failure. Pinpointing the individual and tandem effects of profibrotic stimuli in impairing immune cell response remains difficult and is needed for improved therapeutic strategies. We utilized a statistical design of experiment (DOE) to investigate how microenvironment stiffness and interleukin 13 (IL13), a profibrotic soluble factor linked with disease severity, contribute to the impaired macrophage response commonly observed in pulmonary fibrosis. We used engineered bioinspired hydrogels of different stiffness, ranging from healthy to fibrotic lung tissue, and cultured murine alveolar macrophages (MH-S cells) with or without IL13 to quantify cell response and analyze independent and synergistic effects. We found that, while both stiffness and IL13 independently influence macrophage morphology, phenotype, phagocytosis and efferocytosis, these factors work synergistically to exacerbate impaired macrophage phenotype and efferocytosis. These unique findings provide insights into how macrophages in fibrotic conditions are not as effective in clearing debris, contributing to fibrosis initiation/progression, and more broadly inform how underlying drivers of fibrosis modulate immune cell response to facilitate therapeutic strategies.
Collapse
Affiliation(s)
- Kartik Bomb
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Lina Pradhan
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Qi Zhang
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | - Bader M Jarai
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| | | | - David L Burris
- Mechanical Engineering, University of Delaware, Newark, DE, USA
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
- Material Science and Engineering, University of Delaware, Newark, DE, USA
| | - Catherine A Fromen
- Chemical and Biomolecular Engineering, University of Delaware, Newark, DE, USA.
| |
Collapse
|
16
|
Freeman S, Calabro S, Williams R, Jin S, Ye K. Bioink Formulation and Machine Learning-Empowered Bioprinting Optimization. Front Bioeng Biotechnol 2022; 10:913579. [PMID: 35782492 PMCID: PMC9240914 DOI: 10.3389/fbioe.2022.913579] [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: 04/05/2022] [Accepted: 05/18/2022] [Indexed: 11/23/2022] Open
Abstract
Bioprinting enables the fabrication of complex, heterogeneous tissues through robotically-controlled placement of cells and biomaterials. It has been rapidly developing into a powerful and versatile tool for tissue engineering. Recent advances in bioprinting modalities and biofabrication strategies as well as new materials and chemistries have led to improved mimicry and development of physiologically relevant tissue architectures constituted with multiple cell types and heterogeneous spatial material properties. Machine learning (ML) has been applied to accelerate these processes. It is a new paradigm for bioprinting. In this review, we explore current trends in bioink formulation and how ML has been used to accelerate optimization and enable real-time error detection as well as to reduce the iterative steps necessary for bioink formulation. We examined how rheometric properties, including shear storage, loss moduli, viscosity, shear-thinning property of biomaterials affect the printability of a bioink. Furthermore, we scrutinized the interplays between yield shear stress and the printability of a bioink. Moreover, we systematically surveyed the application of ML in precision in situ surgical site bioprinting, closed-loop AI printing, and post-printing optimization.
Collapse
Affiliation(s)
- Sebastian Freeman
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
| | - Stefano Calabro
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
| | - Roma Williams
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, United States
| | - Sha Jin
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
- Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, NY, United States
| | - Kaiming Ye
- Department of Biomedical Engineering, Binghamton University, Binghamton, NY, United States
- Center of Biomanufacturing for Regenerative Medicine, Binghamton University, State University of New York (SUNY), Binghamton, NY, United States
| |
Collapse
|
17
|
Holl M, Rasch ML, Becker L, Keller AL, Schultze-Rhonhof L, Ruoff F, Templin M, Keller S, Neis F, Keßler F, Andress J, Bachmann C, Krämer B, Schenke-Layland K, Brucker SY, Marzi J, Weiss M. Cell Type-Specific Anti-Adhesion Properties of Peritoneal Cell Treatment with Plasma-Activated Media (PAM). Biomedicines 2022; 10:biomedicines10040927. [PMID: 35453677 PMCID: PMC9032174 DOI: 10.3390/biomedicines10040927] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/16/2022] Open
Abstract
Postoperative abdominal adhesions are responsible for serious clinical disorders. Administration of plasma-activated media (PAM) to cell type-specific modulated proliferation and protein biosynthesis is a promising therapeutic strategy to prevent pathological cell responses in the context of wound healing disorders. We analyzed PAM as a therapeutic option based on cell type-specific anti-adhesive responses. Primary human peritoneal fibroblasts and mesothelial cells were isolated, characterized and exposed to different PAM dosages. Cell type-specific PAM effects on different cell components were identified by contact- and marker-independent Raman imaging, followed by thorough validation by specific molecular biological methods. The investigation revealed cell type-specific molecular responses after PAM treatment, including significant cell growth retardation in peritoneal fibroblasts due to transient DNA damage, cell cycle arrest and apoptosis. We identified a therapeutic dose window wherein specifically pro-adhesive peritoneal fibroblasts were targeted, whereas peritoneal mesothelial cells retained their anti-adhesive potential of epithelial wound closure. Finally, we demonstrate that PAM treatment of peritoneal fibroblasts reduced the expression and secretion of pro-adhesive cytokines and extracellular matrix proteins. Altogether, we provide insights into biochemical PAM mechanisms which lead to cell type-specific pro-therapeutic cell responses. This may open the door for the prevention of pro-adhesive clinical disorders.
Collapse
Affiliation(s)
- Myriam Holl
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
| | - Marie-Lena Rasch
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
| | - Lucas Becker
- Institute of Biomedical Engineering, Eberhard Karls University Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University, 72076 Tübingen, Germany
| | - Anna-Lena Keller
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
| | - Laura Schultze-Rhonhof
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
| | - Felix Ruoff
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
| | - Markus Templin
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
| | - Silke Keller
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
| | - Felix Neis
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
| | - Franziska Keßler
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
| | - Jürgen Andress
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
| | - Cornelia Bachmann
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
| | - Bernhard Krämer
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
| | - Katja Schenke-Layland
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
- Institute of Biomedical Engineering, Eberhard Karls University Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University, 72076 Tübingen, Germany
- Department of Medicine/Cardiology, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| | - Sara Y. Brucker
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
| | - Julia Marzi
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
- Institute of Biomedical Engineering, Eberhard Karls University Tübingen, 72076 Tübingen, Germany;
- Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, Eberhard Karls University, 72076 Tübingen, Germany
| | - Martin Weiss
- Department of Women’s Health Tübingen, Eberhard Karls University Tübingen, 72076 Tübingen, Germany; (M.H.); (M.-L.R.); (L.S.-R.); (F.N.); (F.K.); (J.A.); (C.B.); (B.K.); (S.Y.B.)
- NMI Natural and Medical Sciences Institute, University Tübingen, 72770 Reutlingen, Germany; (A.-L.K.); (F.R.); (M.T.); (S.K.); (K.S.-L.); (J.M.)
- Correspondence:
| |
Collapse
|
18
|
Krieghoff J, Gronbach M, Schulz-Siegmund M, Hacker MC. Biodegradable macromers for implant bulk and surface engineering. Biol Chem 2021; 402:1357-1374. [PMID: 34433237 DOI: 10.1515/hsz-2021-0161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 08/09/2021] [Indexed: 11/15/2022]
Abstract
Macromers, polymeric molecules with at least two functional groups for cross-polymerization, are interesting materials to tailor mechanical, biochemical and degradative bulk and surface properties of implants for tissue regeneration. In this review we focus on macromers with at least one biodegradable building block. Manifold design options, such as choice of polymeric block(s), optional core molecule and reactive groups, as well as cross-co-polymerization with suitable anchor or linker molecules, allow the adaptation of macromer-based biomaterials towards specific application requirements in both hard and soft tissue regeneration. Implants can be manufactured from macromers using additive manufacturing as well as molding and templating approaches. This review summarizes and discusses the overall concept of biodegradable macromers and recent approaches for macromer processing into implants as well as techniques for surface modification directed towards bone regeneration. These aspects are reviewed including a focus on the authors' contributions to the field through research within the collaborative research project Transregio 67.
Collapse
Affiliation(s)
- Jan Krieghoff
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Mathis Gronbach
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Michaela Schulz-Siegmund
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany
| | - Michael C Hacker
- Medical Faculty, Pharmaceutical Technology, Leipzig University, Eilenburger Str. 15A, D-04317 Leipzig, Germany.,Collaborative Research Center (SFB-TRR67) "Functional Biomaterials for Controlling Healing Processes in Bone and Skin - From Material Science to Clinical Application", Leipzig and Dresden, Germany.,Institute of Pharmaceutics and Biopharmaceutics, Heinrich Heine University, Universitätsstrasse 1, D-40225 Düsseldorf, Germany
| |
Collapse
|
19
|
Selo MA, Sake JA, Kim KJ, Ehrhardt C. In vitro and ex vivo models in inhalation biopharmaceutical research - advances, challenges and future perspectives. Adv Drug Deliv Rev 2021; 177:113862. [PMID: 34256080 DOI: 10.1016/j.addr.2021.113862] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/11/2022]
Abstract
Oral inhalation results in pulmonary drug targeting and thereby reduces systemic side effects, making it the preferred means of drug delivery for the treatment of respiratory disorders such as asthma, chronic obstructive pulmonary disease or cystic fibrosis. In addition, the high alveolar surface area, relatively low enzymatic activity and rich blood supply of the distal airspaces offer a promising pathway to the systemic circulation. This is particularly advantageous when a rapid onset of pharmacological action is desired or when the drug is suffering from stability issues or poor biopharmaceutical performance following oral administration. Several cell and tissue-based in vitro and ex vivo models have been developed over the years, with the intention to realistically mimic pulmonary biological barriers. It is the aim of this review to critically discuss the available models regarding their advantages and limitations and to elaborate further which biopharmaceutical questions can and cannot be answered using the existing models.
Collapse
|
20
|
Abstract
Over the past decade, 3D culture models of human and animal cells have found their way into tissue differentiation, drug development, personalized medicine and tumour behaviour studies. Embryoid bodies (EBs) are in vitro 3D cultures established from murine pluripotential stem cells, whereas tumoroids are patient-derived in vitro 3D cultures. This thesis aims to describe a new implication of an embryoid body model and to characterize the patient-specific microenvironment of the parental tumour in relation to tumoroid growth rate. In this thesis, we described a high-throughput monitoring method, where EBs are used as a dynamic angiogenesis model. In this model, digital image analysis (DIA) is implemented on immunohistochemistry (IHC) stained sections of the cultures over time. Furthermore, we have investigated the correlation between the genetic profile and inflammatory microenvironment of parental tumours on the in vitro growth rate of tumoroids. The EBs were cultured in spinner flasks. The samples were collected at days 4, 6, 9, 14, 18 and 21, dehydrated and embedded in paraffin. The histological sections were IHC stained for the endothelial marker CD31 and digitally scanned. The virtual whole-image slides were digitally analysed by Visiopharm® software. Histological evaluation showed vascular-like structures over time. The quantitative DIA was plausible to monitor significant increase in the total area of the EBs and an increase in endothelial differentiation. The tumoroids were established from 32 colorectal adenocarcinomas. The in vitro growth rate of the tumoroids was followed by automated microscopy over an 11-day period. The parental tumours were analysed by next-generation sequencing for KRAS, TP53, PIK3CA, SMAD4, MAP2K1, BRAF, FGFR3 and FBXW7 status. The tumoroids established from KRAS-mutated parental tumours showed a significantly higher growth rate compared to their wild-type counterparts. The density of CD3+ T lymphocytes and CD68+ macrophages was calculated in the centre of the tumours and at the invasive margin of the tumours. The high density of CD3+ cells and the low density of CD68+ cells showed a significant correlation with a higher growth rate of the tumoroids. In conclusion, a novel approach for histological monitoring of endothelial differentiation is presented in the stem cell-derived EBs. Furthermore, the KRAS status and density of CD3+ T cells and macrophages in the parental tumour influence the growth rate of the tumoroids. Our results indicate that these parameters should be included when tumoroids are to be implemented in personalized medicine.
Collapse
Affiliation(s)
- Nabi Mousavi
- Department of Pathology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
21
|
Woodley JP, Lambert DW, Asencio IO. Understanding Fibroblast Behavior in 3D Biomaterials. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:569-578. [PMID: 34102862 DOI: 10.1089/ten.teb.2021.0010] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Traditional monolayer culture fails to fully recapitulate the in vivo environment of connective tissue cells such as the fibroblast. When cultured on stiff two-dimensional (2D) plastic, fibroblasts become highly proliferative forming broad lamellipodia and stress fibers. Conversely, in different three-dimensional (3D) culture systems, fibroblasts have displayed a diverse array of features; from an "activated" phenotype like that observed in 2D cultures and by myofibroblasts, to a quiescent state that likely better represents in vivo fibroblasts at rest. Today, a plethora of microfabrication techniques have made 3D culture commonplace, for both tissue engineering purposes and in the study of basic biological interactions. However, establishing the in vivo mimetic credentials of different biomimetic materials is not always straightforward, particularly in the context of fibroblast responses. Fibroblast behavior is governed by the complex interplay of biological features such as integrin binding sites, material mechanical properties that influence cellular mechanotransduction, and microarchitectural features like pore and fiber size, as well as chemical cues. Furthermore, fibroblasts are a heterogeneous group of cells with specific phenotypic traits dependent on their tissue of origin. These features have made understanding the influence of biomaterials on fibroblast behavior a challenging task. In this study, we present a review of the strategies used to investigate fibroblast behavior with a focus on the material properties that influence fibroblast activation, a process that becomes pathological in fibrotic diseases and certain cancers.
Collapse
Affiliation(s)
- Joe P Woodley
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Daniel W Lambert
- Integrated Bioscience Group, The School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Ilida Ortega Asencio
- Bioengineering and Health Technologies Group, The School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
22
|
Tang LJW, Zaseela A, Toh CCM, Adine C, Aydar AO, Iyer NG, Fong ELS. Engineering stromal heterogeneity in cancer. Adv Drug Deliv Rev 2021; 175:113817. [PMID: 34087326 DOI: 10.1016/j.addr.2021.05.027] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/19/2021] [Accepted: 05/29/2021] [Indexed: 02/09/2023]
Abstract
Based on our exponentially increasing knowledge of stromal heterogeneity from advances in single-cell technologies, the notion that stromal cell types exist as a spectrum of unique subpopulations that have specific functions and spatial distributions in the tumor microenvironment has significant impact on tumor modeling for drug development and personalized drug testing. In this Review, we discuss the importance of incorporating stromal heterogeneity and tumor architecture, and propose an overall approach to guide the reconstruction of stromal heterogeneity in vitro for tumor modeling. These next-generation tumor models may support the development of more precise drugs targeting specific stromal cell subpopulations, as well as enable improved recapitulation of patient tumors in vitro for personalized drug testing.
Collapse
Affiliation(s)
- Leon Jia Wei Tang
- Department of Biological Sciences, National University of Singapore, Singapore
| | - Ayshath Zaseela
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | | | - Christabella Adine
- Department of Biomedical Engineering, National University of Singapore, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore
| | - Abdullah Omer Aydar
- Department of Biomedical Engineering, National University of Singapore, Singapore
| | - N Gopalakrishna Iyer
- National Cancer Centre Singapore, Singapore; Duke-NUS Medical School, Singapore.
| | - Eliza Li Shan Fong
- Department of Biomedical Engineering, National University of Singapore, Singapore; The N.1 Institute for Health, National University of Singapore, Singapore.
| |
Collapse
|
23
|
Xu Y, Koya R, Ask K, Zhao R. Engineered microenvironment for the study of myofibroblast mechanobiology. Wound Repair Regen 2021; 29:588-596. [PMID: 34118169 PMCID: PMC8254796 DOI: 10.1111/wrr.12955] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 06/10/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022]
Abstract
Myofibroblasts are mechanosensitive cells and a variety of their behaviours including differentiation, migration, force production and biosynthesis are regulated by the surrounding microenvironment. Engineered cell culture models have been developed to examine the effect of microenvironmental factors such as the substrate stiffness, the topography and strain of the extracellular matrix (ECM) and the shear stress on myofibroblast biology. These engineered models provide well-mimicked, pathophysiologically relevant experimental conditions that are superior to those enabled by the conventional two-dimensional (2D) culture models. In this perspective, we will review the recent advances in the development of engineered cell culture models for myofibroblasts and outline the findings on the myofibroblast mechanobiology under various microenvironmental conditions. These studies have demonstrated the power and utility of the engineered models for the study of microenvironment-regulated cellular behaviours. The findings derived using these models contribute to a greater understanding of how myofibroblast behaviour is regulated in tissue repair and pathological scar formation.
Collapse
Affiliation(s)
- Ying Xu
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260, USA
| | - Richard Koya
- Department of Obstetrics and Gynecology, University of Chicago Comprehensive Cancer Center, Biological Sciences Division, University of Chicago School of Medicine, Chicago, IL 60637, USA
| | - Kjetil Ask
- Department of Medicine, Div. Respirology, McMaster University, Hamilton, ON, Canada L8N 4A6
- The Research Institute of St. Joe’s Hamilton, Firestone Institute for Respiratory Health, Hamilton, ON, Canada L8N 4A6
| | - Ruogang Zhao
- Department of Biomedical Engineering, State University of New York at Buffalo, Buffalo, NY 14260, USA
| |
Collapse
|
24
|
Sawant M, Hinz B, Schönborn K, Zeinert I, Eckes B, Krieg T, Schuster R. A story of fibers and stress: Matrix-embedded signals for fibroblast activation in the skin. Wound Repair Regen 2021; 29:515-530. [PMID: 34081361 DOI: 10.1111/wrr.12950] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/13/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022]
Abstract
Our skin is continuously exposed to mechanical challenge, including shear, stretch, and compression. The extracellular matrix of the dermis is perfectly suited to resist these challenges and maintain integrity of normal skin even upon large strains. Fibroblasts are the key cells that interpret mechanical and chemical cues in their environment to turnover matrix and maintain homeostasis in the skin of healthy adults. Upon tissue injury, fibroblasts and an exclusive selection of other cells become activated into myofibroblasts with the task to restore skin integrity by forming structurally imperfect but mechanically stable scar tissue. Failure of myofibroblasts to terminate their actions after successful repair or upon chronic inflammation results in dysregulated myofibroblast activities which can lead to hypertrophic scarring and/or skin fibrosis. After providing an overview on the major fibrillar matrix components in normal skin, we will interrogate the various origins of fibroblasts and myofibroblasts in the skin. We then examine the role of the matrix as signaling hub and how fibroblasts respond to mechanical matrix cues to restore order in the confusing environment of a healing wound.
Collapse
Affiliation(s)
- Mugdha Sawant
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Katrin Schönborn
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Isabel Zeinert
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Beate Eckes
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Thomas Krieg
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Ronen Schuster
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Canada.,PhenomicAI, MaRS Centre, 661 University Avenue, Toronto, Canada
| |
Collapse
|
25
|
Shirazi J, Donzanti MJ, Nelson KM, Zurakowski R, Fromen CA, Gleghorn JP. Significant Unresolved Questions and Opportunities for Bioengineering in Understanding and Treating COVID-19 Disease Progression. Cell Mol Bioeng 2020; 13:259-284. [PMID: 32837585 PMCID: PMC7384395 DOI: 10.1007/s12195-020-00637-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/14/2020] [Indexed: 12/19/2022] Open
Abstract
COVID-19 is a disease that manifests itself in a multitude of ways across a wide range of tissues. Many factors are involved, and though impressive strides have been made in studying this novel disease in a very short time, there is still a great deal that is unknown about how the virus functions. Clinical data has been crucial for providing information on COVID-19 progression and determining risk factors. However, the mechanisms leading to the multi-tissue pathology are yet to be fully established. Although insights from SARS-CoV-1 and MERS-CoV have been valuable, it is clear that SARS-CoV-2 is different and merits its own extensive studies. In this review, we highlight unresolved questions surrounding this virus including the temporal immune dynamics, infection of non-pulmonary tissue, early life exposure, and the role of circadian rhythms. Risk factors such as sex and exposure to pollutants are also explored followed by a discussion of ways in which bioengineering approaches can be employed to help understand COVID-19. The use of sophisticated in vitro models can be employed to interrogate intercellular interactions and also to tease apart effects of the virus itself from the resulting immune response. Additionally, spatiotemporal information can be gleaned from these models to learn more about the dynamics of the virus and COVID-19 progression. Application of advanced tissue and organ system models into COVID-19 research can result in more nuanced insight into the mechanisms underlying this condition and elucidate strategies to combat its effects.
Collapse
Affiliation(s)
- Jasmine Shirazi
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Michael J. Donzanti
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Katherine M. Nelson
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Ryan Zurakowski
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| | - Catherine A. Fromen
- Department of Chemical and Biomolecular Engineering, University of Delaware, Newark, DE 19716 USA
| | - Jason P. Gleghorn
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Lab, Newark, DE 19716 USA
| |
Collapse
|