1
|
Bezold MG, Dollinger BR, DeJulius CR, Keech MC, Hanna AR, Kittel AR, Yu F, Gupta MK, D'Arcy R, Brunger JM, Duvall CL. Shear-thinning hydrogel for allograft cell transplantation and externally controlled transgene expression. Biomaterials 2025; 314:122812. [PMID: 39288619 DOI: 10.1016/j.biomaterials.2024.122812] [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: 06/20/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
This work establishes the design of a fully synthetic, shear-thinning hydrogel platform that is injectable and can isolate engineered, allogeneic cell therapies from the host. We utilized RAFT to generate a library of linear random copolymers of N,N-dimethylacrylamide (DMA) and 2-vinyl-4,4-dimethyl azlactone (VDMA) with variable mol% VDMA and degree of polymerization. Poly(DMA-co-VDMA) copolymers were subsequently modified with either adamantane (Ad) or β-cyclodextrin (Cd) through amine-reactive VDMA to prepare hydrogel precursor macromers containing complementary guest-host pairing pendant groups that, when mixed, form shear-thinning hydrogels. Rheometric evaluation of the hydrogel library enabled identification of lead macromer structures comprising 15 mol% pendants (Ad or Cd) and a degree of polymerization of 1000; mixing of these Ad and Cd functionalized precursors yielded hydrogels possessing storage modulus above 1000 Pa, tan(δ) values below 1 and high yield strain, which are target characteristics of robust but injectable shear-thinning gels. This modular system proved amenable to nanoparticle integration with surface-modified nanoparticles displaying Ad. The addition of the Ad-functionalized nanoparticles simultaneously improved mechanical properties of the hydrogels and enabled extended hydrogel retention of a model small molecule in vivo. In studies benchmarking against alginate, a material traditionally used for cell encapsulation, the lead hydrogel showed significantly less fibrous encapsulation in a subcutaneous implant site. Finally, this platform was utilized to encapsulate and extend in vivo longevity of inducible transgene-engineered mesenchymal stem cells in an allogeneic transplant model. The hydrogels remained intact and blocked infiltration by host cells, consequently extending the longevity of grafted cell function relative to a benchmark, shear-thinning hyaluronic acid-based gel. In sum, the new synthetic, shear-thinning hydrogel system presented here shows potential for further development as an injectable platform for delivery and in situ drug modulation of allograft and engineered cell therapies.
Collapse
Affiliation(s)
- Mariah G Bezold
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Bryan R Dollinger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Megan C Keech
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Andrew R Hanna
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Anna R Kittel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Mukesh K Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Richard D'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
2
|
Tang M, Feng X, Ma L, Yu Y, Zhu H, Fu Y, Sun K, Wu X, Wang J, Li X, Zhang Y. Utilizing superheated steam to prepare traditional Chinese twice-cooked pork bellies, exploring its effects on the texture and flavor of fat layers. Meat Sci 2024; 217:109616. [PMID: 39089087 DOI: 10.1016/j.meatsci.2024.109616] [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: 05/02/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/03/2024]
Abstract
Fat greatly impacts the overall texture and flavor of pork belly. Twice-cooked pork bellies (TPB), typically boiled and sliced before "back to pot" being stir-fried, is a classic Sichuan cuisine among stir-fried dishes. In this study, the effects of substituting conventional pan-frying (PCV) with superheated steam (SHS) technology on the sensory, texture, microstructure and flavor of the fat layers were investigated. SHS was used as an alternative to boiling (120 °C for 15, 20, 25, and 30 min), and "back to pot" stir-frying was also by SHS. TPB precooked for 25 min (P25) with SHS performed better quality characteristics than PCV, with less collagen fiber disruption and lipid droplet area, resulting in a lower hardness and higher shear force. Besides, the low-oxygen environment of SHS retarded the lipid peroxidation, showing a significantly lower MDA content than PCV. Differently, PCV exhibited more grassy and fatty flavors, while P25 exhibited a unique aroma of fruity and creamy due to its higher UFA/SFA ratios in the pre-cooking stage. Overall, the sensory scores of P25 were comparable to those of PCV (with no significant difference), revealing that SHS is expected to be applied to the industrial production of stir-fried dishes.
Collapse
Affiliation(s)
- Mi Tang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Xin Feng
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Liang Ma
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China; Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Chongqing 400715, China
| | - Yong Yu
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Hankun Zhu
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Yu Fu
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Kangting Sun
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Xiaoqian Wu
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Academy of Animal Science, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Jinyong Wang
- Chongqing Academy of Animal Science, Chongqing 402460, China; National Center of Technology Innovation for Pigs, Chongqing 402460, China
| | - Xiang Li
- Culinary Institute of Sichuan Tourism University, Chengdu 610100, China
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China; Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Chongqing 400715, China.
| |
Collapse
|
3
|
Goder Orbach D, Roitman I, Coster Kimhi G, Zilberman M. Formulation-Property Effects in Novel Injectable and Resilient Natural Polymer-Based Hydrogels for Soft Tissue Regeneration. Polymers (Basel) 2024; 16:2879. [PMID: 39458707 PMCID: PMC11511563 DOI: 10.3390/polym16202879] [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: 09/08/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
The development of injectable hydrogels for soft tissue regeneration has gained significant attention due to their minimally invasive application and ability to conform precisely to the shape of irregular tissue cavities. This study presents a novel injectable porous scaffold based on natural polymers that undergoes in situ crosslinking, forming a highly resilient hydrogel with tailorable mechanical and physical properties to meet the specific demands of soft tissue repair. By adjusting the formulation, we achieved a range of stiffness values that closely mimic the mechanical characteristics of native tissues while maintaining very high resilience (>90%). The effects of gelatin, alginate, and crosslinker concentrations, as well as porosity, on the hydrogel's properties were elucidated. The main results indicated a compression modulus range of 2.7-89 kPa, which fits all soft tissues, and gelation times ranging from 5 to 30 s, which enable the scaffold to be successfully used in various operations. An increase in gelatin and crosslinker concentrations results in a higher modulus and lower gelation time, i.e., a stiffer hydrogel that is created in a shorter time. In vitro cell viability tests on human fibroblasts were performed and indicated high biocompatibility. Our findings demonstrate that these injectable hydrogel scaffolds offer a promising solution for enhancing soft tissue repair and regeneration, providing a customizable and resilient framework that is expected to support tissue integration and healing with minimal surgical intervention.
Collapse
Affiliation(s)
| | | | | | - Meital Zilberman
- Department of Biomedical Engineering, Tel-Aviv University, Tel Aviv 6997801, Israel; (D.G.O.); (I.R.); (G.C.K.)
| |
Collapse
|
4
|
Pitton M, Urzì C, Farè S, Contessi Negrini N. Visible light photo-crosslinking of biomimetic gelatin-hyaluronic acid hydrogels for adipose tissue engineering. J Mech Behav Biomed Mater 2024; 158:106675. [PMID: 39068848 DOI: 10.1016/j.jmbbm.2024.106675] [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: 08/26/2023] [Revised: 04/23/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Tissue engineering (TE) of adipose tissue (AT) is a promising strategy that can provide 3D constructs to be used for in vitro modelling, overcoming the limitations of 2D cell cultures by closely replicating the complex breast tissue extracellular matrix (ECM), cell-cell, and cell-ECM interactions. However, the challenge in developing 3D constructs of AT resides in designing artificial matrices that can mimic the structural properties of native AT and support adipocytes biological functions. Herein, we developed photocrosslinkable hydrogels by employing gelatin methacrylate (GelMA) and hyaluronic acid methacrylate (HAMA) to mimic the collagenous and glycosaminoglycan components of AT microenvironment, respectively. The physico-mechanical properties of the hydrogels were tuned to target AT biomimetic properties by varying the hydrogel formulation (with or without hyaluronic acid), and the amount of photoinitiator (ruthenium/sodium persulfate) used to crosslink the hydrogels via visible light. The physical and mechanical properties of the developed hydrogels were tuned by varying the material formulation and the photoinitiator concentration. Preadipocytes were encapsulated inside the hydrogels and differentiated into mature adipocytes. Findings enlightened that HAMA addition in hybrid hydrogels boosted an increased lipid accumulation. The engineered biomimetic adipocyte-based constructs resulted promising as scaffolds or 3D in vitro models of AT.
Collapse
Affiliation(s)
- Matteo Pitton
- Department of Chemistry, Materials and Chemical Engineering, Politecnico di Milano, Italy
| | - Christian Urzì
- Department of Chemistry, Materials and Chemical Engineering, Politecnico di Milano, Italy
| | - Silvia Farè
- Department of Chemistry, Materials and Chemical Engineering, Politecnico di Milano, Italy; National Interuniversity Consortium of Materials Science and Technology, Florence, Italy.
| | | |
Collapse
|
5
|
Fujikawa T, Yamada Y. In vivo video microscopy of the rupturing process of thin blood vessels to clarify the mechanism of bruising caused by blunt impact: an animal study. Biomed Eng Online 2024; 23:94. [PMID: 39261896 PMCID: PMC11389484 DOI: 10.1186/s12938-024-01284-2] [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: 06/17/2024] [Accepted: 08/27/2024] [Indexed: 09/13/2024] Open
Abstract
BACKGROUND The thresholds of mechanical inputs for bruising caused by blunt impact are important in the fields of machine safety and forensics. However, reliable data on these thresholds remain inadequate owing to a lack of in vivo experiments, which are crucial for investigating the occurrence of bruising. Since experiments involving live human participants are limited owing to ethical concerns, finite-element method (FEM) simulations of the bruising mechanism should be used to compensate for the lack of experimental data by estimating the thresholds under various conditions, which requires clarifying the mechanism of formation of actual bruises. Therefore, this study aimed to visualize the mechanism underlying the formation of bruises caused by blunt impact to enable FEM simulations to estimate the thresholds of mechanical inputs for bruising. METHODS In vivo microscopy of a transparent glass catfish subjected to blunt contact with an indenter was performed. The fish were anesthetized by immersing them in buffered MS-222 (75-100 mg/L) and then fixed on a subject tray. The indenter, made of transparent acrylic and having a rectangular contact area with dimensions of 1.0 mm × 1.5 mm, was loaded onto the lateral side of the caudal region of the fish. Blood vessels and surrounding tissues were examined through the transparent indenter using a microscope equipped with a video camera. The contact force was measured using a force-sensing table. RESULTS One of the processes of rupturing thin blood vessels, which are an essential component of the bruising mechanism, was observed and recorded as a movie. The soft tissue surrounding the thin blood vessel extended in a plane perpendicular to the compressive contact force. Subsequently, the thin blood vessel was pulled into a straight configuration. Next, it was stretched in the axial direction and finally ruptured. CONCLUSION The results obtained indicate that the extension of the surrounding tissue in the direction perpendicular to the contact force as well as the extension of the thin blood vessels are important factors in the bruising mechanism, which must be reproduced by FEM simulation to estimate the thresholds.
Collapse
Affiliation(s)
- Tatsuo Fujikawa
- Department of Mechanical Engineering, College of Engineering, Nihon University, 1 Nakagawara, Tokusada, Tamuramachi, Koriyama, 963-8642, Japan.
| | - Yoji Yamada
- Department of Mechanical System Engineering, Nagoya University, Furo-Cho, Chikusa-Ku, Nagoya, 464-8603, Japan
| |
Collapse
|
6
|
Zheng Y, Wang D, Beeghly G, Fischbach C, Shattuck MD, O'Hern CS. Computational modeling of the physical features that influence breast cancer invasion into adipose tissue. APL Bioeng 2024; 8:036104. [PMID: 38966325 PMCID: PMC11223776 DOI: 10.1063/5.0209019] [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: 03/18/2024] [Accepted: 06/21/2024] [Indexed: 07/06/2024] Open
Abstract
Breast cancer invasion into adipose tissue strongly influences disease progression and metastasis. The degree of cancer cell invasion into adipose tissue depends on both biochemical signaling and the mechanical properties of cancer cells, adipocytes, and other key components of adipose tissue. We model breast cancer invasion into adipose tissue using discrete element method simulations of active, cohesive spherical particles (cancer cells) invading into confluent packings of deformable polyhedra (adipocytes). We quantify the degree of invasion by calculating the interfacial area At between cancer cells and adipocytes. We determine the long-time value of At vs the activity and strength of the cohesion between cancer cells, as well as the mechanical properties of the adipocytes and extracellular matrix in which adipocytes are embedded. We show that the degree of invasion collapses onto a master curve as a function of the dimensionless energy scale Ec , which grows linearly with the cancer cell velocity persistence time and fluctuations, is inversely proportional to the system pressure, and is offset by the cancer cell cohesive energy. WhenE c > 1 , cancer cells will invade the adipose tissue, whereas forE c < 1 , cancer cells and adipocytes remain de-mixed. We also show that At decreases when the adipocytes are constrained by the ECM by an amount that depends on the spatial heterogeneity of the adipose tissue.
Collapse
Affiliation(s)
| | - Dong Wang
- Department of Mechanical Engineering and Materials Science, Yale University, New Haven, Connecticut 06520, USA
| | - Garrett Beeghly
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Claudia Fischbach
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York 14853, USA
| | - Mark D. Shattuck
- Benjamin Levich Institute and Physics Department, City College of New York, New York, New York 10031, USA
| | | |
Collapse
|
7
|
O'Shea TC, Croland KJ, Salem A, Urbanski R, Schultz KM. A Rheological Study on the Effect of Tethering Pro- and Anti-Inflammatory Cytokines into Hydrogels on Human Mesenchymal Stem Cell Migration, Degradation, and Morphology. Biomacromolecules 2024; 25:5121-5137. [PMID: 38961715 DOI: 10.1021/acs.biomac.4c00508] [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] [Indexed: 07/05/2024]
Abstract
Polymer-peptide hydrogels are being designed as implantable materials that deliver human mesenchymal stem cells (hMSCs) to treat wounds. Most wounds can progress through the healing process without intervention. During the normal healing process, cytokines are released from the wound to create a concentration gradient, which causes directed cell migration from the native niche to the wound site. Our work takes inspiration from this process and uniformly tethers cytokines into the scaffold to measure changes in cell-mediated degradation and motility. This is the first step in designing cytokine concentration gradients into the material to direct cell migration. We measure changes in rheological properties, encapsulated cell-mediated pericellular degradation and migration in a hydrogel scaffold with covalently tethered cytokines, either tumor necrosis factor-α (TNF-α) or transforming growth factor-β (TGF-β). TNF-α is expressed in early stages of wound healing causing an inflammatory response. TGF-β is released in later stages of wound healing causing an anti-inflammatory response in the surrounding tissue. Both cytokines cause directed cell migration. We measure no statistically significant difference in modulus or the critical relaxation exponent when tethering either cytokine in the polymeric network without encapsulated hMSCs. This indicates that the scaffold structure and rheology is unchanged by the addition of tethered cytokines. Increases in hMSC motility, morphology and cell-mediated degradation are measured using a combination of multiple particle tracking microrheology (MPT) and live-cell imaging in hydrogels with tethered cytokines. We measure that tethering TNF-α into the hydrogel increases cellular remodeling on earlier days postencapsulation and tethering TGF-β into the scaffold increases cellular remodeling on later days. We measure tethering either TGF-β or TNF-α enhances cell stretching and, subsequently, migration. This work provides rheological characterization that can be used to design new materials that present chemical cues in the pericellular region to direct cell migration.
Collapse
Affiliation(s)
- Thomas C O'Shea
- Purdue University, Davidson School of Chemical Engineering, 480 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| | - Kiera J Croland
- University of Colorado at Boulder, Department of Chemical and Biological Engineering, 3415 Colorado Ave, Boulder, Colorado 80303, United States
| | - Ahmad Salem
- Lehigh University, Department of Chemical and Biomolecular Engineering, 124 East Morton Street, Bethlehem, Pennsylvania 18015, United States
| | - Rylie Urbanski
- Lehigh University, Department of Chemical and Biomolecular Engineering, 124 East Morton Street, Bethlehem, Pennsylvania 18015, United States
| | - Kelly M Schultz
- Purdue University, Davidson School of Chemical Engineering, 480 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|
8
|
Shukla P, Bera AK, Yeleswarapu S, Pati F. High Throughput Bioprinting Using Decellularized Adipose Tissue-Based Hydrogels for 3D Breast Cancer Modeling. Macromol Biosci 2024; 24:e2400035. [PMID: 38685795 DOI: 10.1002/mabi.202400035] [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: 01/29/2024] [Revised: 03/29/2024] [Indexed: 05/02/2024]
Abstract
3D bioprinting allows rapid automated fabrication and can be applied for high throughput generation of biomimetic constructs for in vitro drug screening. Decellularized extracellular matrix (dECM) hydrogel is a popular biomaterial choice for tissue engineering and studying carcinogenesis as a tumor microenvironmental mimetic. This study proposes a method for high throughput bioprinting with decellularized adipose tissue (DAT) based hydrogels for 3D breast cancer modeling. A comparative analysis of decellularization protocol using detergent-based and detergent-free decellularization methods for caprine-origin adipose tissue is performed, and the efficacy of dECM hydrogel for 3D cancer modeling is assessed. Histological, biochemical, morphological, and biological characterization and analysis showcase the cytocompatibility of DAT hydrogel. The rheological property of DAT hydrogel and printing process optimization is assessed to select a bioprinting window to attain 3D breast cancer models. The bioprinted tissues are characterized for cellular viability and tumor cell-matrix interactions. Additionally, an approach for breast cancer modeling is shown by performing rapid high throughput bioprinting in a 96-well plate format, and in vitro drug screening using 5-fluorouracil is performed on 3D bioprinted microtumors. The results of this study suggest that high throughput bioprinting of cancer models can potentially have downstream clinical applications like multi-drug screening platforms and personalized disease models.
Collapse
Affiliation(s)
- Priyanshu Shukla
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana, 502284, India
| | - Ashis Kumar Bera
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana, 502284, India
| | - Sriya Yeleswarapu
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana, 502284, India
| | - Falguni Pati
- Department of Biomedical Engineering, Indian Institute of Technology Hyderabad, Kandi, Sangareddy, Telangana, 502284, India
| |
Collapse
|
9
|
Cesanelli L, Minderis P, Degens H, Satkunskiene D. Passive mechanical properties of adipose tissue and skeletal muscle from C57BL/6J mice. J Mech Behav Biomed Mater 2024; 155:106576. [PMID: 38744119 DOI: 10.1016/j.jmbbm.2024.106576] [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: 12/06/2023] [Revised: 02/01/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024]
Abstract
Skeletal muscle and adipose tissue are characterized by unique structural features finely tuned to meet specific functional demands. In this study, we investigated the passive mechanical properties of soleus (SOL), extensor digitorum longus (EDL) and diaphragm (DIA) muscles, as well as subcutaneous (SAT), visceral (VAT) and brown (BAT) adipose tissues from 13 C57BL/6J mice. Thereto, alongside stress-relaxation assessments we subjected isolated muscles and adipose tissues (ATs) to force-extension tests up to 10% and 30% of their optimal length, respectively. Peak passive stress was highest in the DIA, followed by the SOL and lowest in the EDL (p < 0.05). SOL displayed also the highest Young's modulus and hysteresis among muscles (p < 0.05). BAT demonstrated highest peak passive stress and Young's modulus followed by VAT (p < 0.05), while SAT showed the highest hysteresis (p < 0.05). When comparing data across all six biological specimens at fixed passive force intervals (i.e., 20-40 and 50-70 mN), skeletal muscles exhibited significantly higher peak stresses and strains than ATs (p < 0.05). Young's modulus was higher in skeletal muscles than in ATs (p < 0.05). Muscle specimens exhibited slower force relaxation in the first phase compared to ATs (p < 0.05), while there was no significant difference in behavior between muscles and AT in the second phase of relaxation. The study revealed distinctive mechanical behaviors specific to different tissues, and even between different muscles and ATs. These variations in mechanical properties are likely such to optimize the specific functions performed by each biological tissue.
Collapse
Affiliation(s)
- L Cesanelli
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania; Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania.
| | - P Minderis
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania
| | - H Degens
- Institute of Sport Science and Innovations, Lithuanian Sports University, Kaunas, Lithuania; Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - D Satkunskiene
- Department of Health Promotion and Rehabilitation, Lithuanian Sports University, Kaunas, Lithuania
| |
Collapse
|
10
|
Goder Orbach D, Zilberman M. Formulation Effects on the Mechano-Physical Properties of In Situ-Forming Resilient Hydrogels for Breast Tissue Regeneration. J Funct Biomater 2024; 15:176. [PMID: 39057298 PMCID: PMC11277960 DOI: 10.3390/jfb15070176] [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: 05/17/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
The need for a long-term solution for filling the defects created during partial mastectomies due to breast cancer diagnosis has not been met to date. All available defect-filling methods are non-permanent and necessitate repeat procedures. Here, we report on novel injectable porous hydrogel structures based on the natural polymers gelatin and alginate, which are designed to serve for breast reconstruction and regeneration following partial mastectomy. The effects of the formulation parameters on the mechanical and physical properties were thoroughly studied. The modulus in compression and tension were in the range of native breast tissue. Both increased with the increase in the crosslinker concentration and the polymer-air ratio. Resilience was very high, above 93% for most studied formulations, allowing the scaffold to be continuously deformed without changing its shape. The combination of high resilience and low elastic modulus is favored for adipose tissue regeneration. The physical properties of gelation time and water uptake are controllable and are affected mainly by the alginate and N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide hydrochloride (EDC) concentrations and less by the polymer-air ratio. In vitro cell viability tests were performed on mouse preadipocytes and indicated high biocompatibility. The minimally invasive nature of this approach, along with the excellent properties of the scaffold, will enable the filling of complex voids while simultaneously decreasing surgical costs and greatly improving patient well-being.
Collapse
Affiliation(s)
| | - Meital Zilberman
- Department of Biomedical Engineering, Faculty of Engineering, Tel-Aviv University, Tel-Aviv 69978, Israel;
| |
Collapse
|
11
|
Wyle Y, Lu N, Hepfer J, Sayal R, Martinez T, Wang A. The Role of Biophysical Factors in Organ Development: Insights from Current Organoid Models. Bioengineering (Basel) 2024; 11:619. [PMID: 38927855 PMCID: PMC11200479 DOI: 10.3390/bioengineering11060619] [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: 04/17/2024] [Revised: 05/26/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Biophysical factors play a fundamental role in human embryonic development. Traditional in vitro models of organogenesis focused on the biochemical environment and did not consider the effects of mechanical forces on developing tissue. While most human tissue has a Young's modulus in the low kilopascal range, the standard cell culture substrate, plasma-treated polystyrene, has a Young's modulus of 3 gigapascals, making it 10,000-100,000 times stiffer than native tissues. Modern in vitro approaches attempt to recapitulate the biophysical niche of native organs and have yielded more clinically relevant models of human tissues. Since Clevers' conception of intestinal organoids in 2009, the field has expanded rapidly, generating stem-cell derived structures, which are transcriptionally similar to fetal tissues, for nearly every organ system in the human body. For this reason, we conjecture that organoids will make their first clinical impact in fetal regenerative medicine as the structures generated ex vivo will better match native fetal tissues. Moreover, autologously sourced transplanted tissues would be able to grow with the developing embryo in a dynamic, fetal environment. As organoid technologies evolve, the resultant tissues will approach the structure and function of adult human organs and may help bridge the gap between preclinical drug candidates and clinically approved therapeutics. In this review, we discuss roles of tissue stiffness, viscoelasticity, and shear forces in organ formation and disease development, suggesting that these physical parameters should be further integrated into organoid models to improve their physiological relevance and therapeutic applicability. It also points to the mechanotransductive Hippo-YAP/TAZ signaling pathway as a key player in the interplay between extracellular matrix stiffness, cellular mechanics, and biochemical pathways. We conclude by highlighting how frontiers in physics can be applied to biology, for example, how quantum entanglement may be applied to better predict spontaneous DNA mutations. In the future, contemporary physical theories may be leveraged to better understand seemingly stochastic events during organogenesis.
Collapse
Affiliation(s)
- Yofiel Wyle
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
| | - Nathan Lu
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Jason Hepfer
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Rahul Sayal
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Taylor Martinez
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
| | - Aijun Wang
- Department of Surgery, School of Medicine, University of California-Davis, Sacramento, CA 95817, USA; (Y.W.); (N.L.); (J.H.); (R.S.); (T.M.)
- Institute for Pediatric Regenerative Medicine, Shriners Children’s, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California-Davis, Davis, CA 95616, USA
- Center for Surgical Bioengineering, Department of Surgery, School of Medicine, University of California, Davis, 4625 2nd Ave., Research II, Suite 3005, Sacramento, CA 95817, USA
| |
Collapse
|
12
|
Jones A, Netto J, Foote T, Ruliffson B, Whittington C. Combined effects of matrix stiffness and obesity-associated signaling directs progressive phenotype in PANC-1 pancreatic cancer cells in vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.11.598541. [PMID: 38915620 PMCID: PMC11195209 DOI: 10.1101/2024.06.11.598541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
Obesity is a leading risk factor of pancreatic ductal adenocarcinoma (PDAC) that contributes to poor disease prognosis and outcomes. Retrospective studies have identified this link, but interactions surrounding obesity and PDAC are still unclear. Research has shifted to contributions of fibrosis (desmoplasia) on malignancy, which involves increased deposition of collagens and other extracellular matrix (ECM) molecules and increased ECM crosslinking, all of which contribute to increased tissue stiffening. However, fibrotic stiffening is underrepresented as a model feature in current PDAC models. Fibrosis is shared between PDAC and obesity, and can be leveraged for in vitro model design, as current animal obesity models of PDAC are limited in their ability to isolate individual components of fibrosis to study cell behavior. In the current study, methacrylated type I collagen (PhotoCol®) was photo-crosslinked to pathological stiffness levels to recapitulate fibrotic ECM stiffening. PANC-1 cells were encapsulated within PhotoCol®, and the tumor-tissue constructs were prepared to represent normal (healthy) (~600 Pa) and pathological (~2000 Pa) tissues. Separately, human mesenchymal stem cells were differentiated into adipocytes representing lean (2D differentiation) and obese fat tissue (3D collagen matrix differentiation), and conditioned media was applied to PANC-1 tumor-tissue constructs. Conditioned media from obese adipocytes showed increased vimentin expression, a hallmark of invasiveness and progression, that was not seen after exposure to media from lean adipocytes or control media. Characterization of the obese adipocyte secretome suggested that some PANC-1 differences may arise from increased interleukin-8 and -10 compared to lean adipocytes. Additionally, high matrix stiffness associated induced an amoeboid morphology in PANC-1 cells that was not present at low stiffness. Amoeboid morphology is an accessory to epithelial-to-mesenchymal transition and is used to navigate complex ECM environments. This plasticity has greater implications for treatment efficacy of metastatic cancers. Overall, this work 1) highlights the importance of investigating PDAC-obesity interactions to study the effects on disease progression and persistence, 2) establishes PhotoCol® as a matrix material that can be leveraged to study amoeboid morphology and invasion in PDAC, and 3) emphasizes the importance of integrating both biophysical and biochemical interactions associated within both pathologies for in vitro PDAC models.
Collapse
Affiliation(s)
- A.E Jones
- Worcester Polytechnic Institute, Department of Biomedical Engineering
| | - J.F. Netto
- Worcester Polytechnic Institute, Department of Biomedical Engineering
| | - T.L. Foote
- Worcester Polytechnic Institute, Department of Biomedical Engineering
| | - B.N.K. Ruliffson
- Worcester Polytechnic Institute, Department of Biomedical Engineering
| | - C.F. Whittington
- Worcester Polytechnic Institute, Department of Biomedical Engineering
| |
Collapse
|
13
|
Blade SP, Falkowski DJ, Bachand SN, Pagano SJ, Chin L. Mechanobiology of Adipocytes. BIOLOGY 2024; 13:434. [PMID: 38927314 PMCID: PMC11200640 DOI: 10.3390/biology13060434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/08/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024]
Abstract
The growing obesity epidemic necessitates increased research on adipocyte and adipose tissue function and disease mechanisms that progress obesity. Historically, adipocytes were viewed simply as storage for excess energy. However, recent studies have demonstrated that adipocytes play a critical role in whole-body homeostasis, are involved in cell communication, experience forces in vivo, and respond to mechanical stimuli. Changes to the adipocyte mechanical microenvironment can affect function and, in some cases, contribute to disease. The aim of this review is to summarize the current literature on the mechanobiology of adipocytes. We reviewed over 100 papers on how mechanical stress is sensed by the adipocyte, the effects on cell behavior, and the use of cell culture scaffolds, particularly those with tunable stiffness, to study adipocyte behavior, adipose cell and tissue mechanical properties, and computational models. From our review, we conclude that adipocytes are responsive to mechanical stimuli, cell function and adipogenesis can be dictated by the mechanical environment, the measurement of mechanical properties is highly dependent on testing methods, and current modeling practices use many different approaches to recapitulate the complex behavior of adipocytes and adipose tissue. This review is intended to aid future studies by summarizing the current literature on adipocyte mechanobiology.
Collapse
Affiliation(s)
- Sean P. Blade
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Dylan J. Falkowski
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Sarah N. Bachand
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| | - Steven J. Pagano
- Department of Mechanical Engineering, Widener University, Chester, PA 19013, USA;
| | - LiKang Chin
- Department of Biomedical Engineering, Widener University, Chester, PA 19013, USA; (S.P.B.); (D.J.F.); (S.N.B.)
| |
Collapse
|
14
|
Elsafty O, Berkey CA, Dauskardt RH. Insights and mechanics-driven modeling of human cutaneous impact injuries. J Mech Behav Biomed Mater 2024; 153:106456. [PMID: 38442507 DOI: 10.1016/j.jmbbm.2024.106456] [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: 06/02/2023] [Accepted: 02/03/2024] [Indexed: 03/07/2024]
Abstract
Cutaneous damage mechanisms related to dynamic fragment impacts are dependent on the impact angle, impact energy, and fragment characteristics including shape, volume, contact friction, and orientation. Understanding the cutaneous injury mechanism and its relationship to the fragment parameters is lacking compromising damage classification, treatment, and protection. Here we develop a high-fidelity dynamic mechanics-driven model for partial-thickness skin injuries and demonstrate the influence of fragment parameters on the injury mechanism and damage sequence. The model quantitatively predicts the wound shape, area, and depth into the skin layers for selected impact angles, kinetic energy density, and the fragment projectile type including shape and material. The detailed sequence of impact damage including epidermal tearing that occurs ahead of the fragments initial contact location, subsequent stripping of the epidermal/dermal junction, and crushing of the underlying dermis are revealed. We demonstrate that the fragment contact friction with skin plays a key role in redistributing impact energy affecting the extent of epidermal tearing and dermal crushing. Furthermore, projectile edges markedly affect injury severity dependent on the orientation of the edge during initial impact. The model provides a quantitative framework for understanding the detailed mechanisms of cutaneous damage and a basis for the design of protective equipment.
Collapse
Affiliation(s)
- Omar Elsafty
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Christopher A Berkey
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Reinhold H Dauskardt
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
15
|
Micek HM, Yang N, Dutta M, Rosenstock L, Ma Y, Hielsberg C, McCord M, Notbohm J, McGregor S, Kreeger PK. The role of Piezo1 mechanotransduction in high-grade serous ovarian cancer: Insights from an in vitro model of collective detachment. SCIENCE ADVANCES 2024; 10:eadl4463. [PMID: 38669327 PMCID: PMC11051664 DOI: 10.1126/sciadv.adl4463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 03/22/2024] [Indexed: 04/28/2024]
Abstract
Slowing peritoneal spread in high-grade serous ovarian cancer (HGSOC) would improve patient prognosis and quality of life. HGSOC spreads when single cells and spheroids detach, float through the peritoneal fluid and take over new sites, with spheroids thought to be more aggressive than single cells. Using our in vitro model of spheroid collective detachment, we determine that increased substrate stiffness led to the detachment of more spheroids. We identified a mechanism where Piezo1 activity increased MMP-1/MMP-10, decreased collagen I and fibronectin, and increased spheroid detachment. Piezo1 expression was confirmed in omental masses from patients with stage III/IV HGSOC. Using OV90 and CRISPR-modified PIEZO1-/- OV90 in a mouse xenograft model, we determined that while both genotypes efficiently took over the omentum, loss of Piezo1 significantly decreased ascitic volume, tumor spheroids in the ascites, and the number of macroscopic tumors in the mesentery. These results support that slowing collective detachment may benefit patients and identify Piezo1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Hannah M. Micek
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ning Yang
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Mayuri Dutta
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Lauren Rosenstock
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Yicheng Ma
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Caitlin Hielsberg
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Molly McCord
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Biophysics Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jacob Notbohm
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Mechanical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Biophysics Program, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Stephanie McGregor
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Pamela K. Kreeger
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| |
Collapse
|
16
|
Major G, Ahn M, Cho WW, Santos M, Wise J, Phillips E, Wise SG, Jang J, Rnjak-Kovacina J, Woodfield T, Lim KS. Programming temporal stiffness cues within extracellular matrix hydrogels for modelling cancer niches. Mater Today Bio 2024; 25:101004. [PMID: 38420142 PMCID: PMC10900776 DOI: 10.1016/j.mtbio.2024.101004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Extracellular matrix (ECM) stiffening is a common occurrence during the progression of many diseases, such as breast cancer. To accurately mimic the pathophysiological context of disease within 3D in vitro models, there is high demand for smart biomaterials which replicate the dynamic and temporal mechanical cues of diseased states. This study describes a preclinical disease model, using breast cancer as an example, which replicates the dynamic plasticity of the tumour microenvironment by incorporating temporal (3-week progression) biomechanical cues within a tissue-specific hydrogel microenvironment. The composite hydrogel formulation, integrating adipose-derived decellularised ECM (AdECM) and silk fibroin, was initially crosslinked using a visible light-mediated system, and then progressively stiffened through spontaneous secondary structure interactions inherent between the polymer chains (∼10-15 kPa increase, with a final stiffness of 25 kPa). When encapsulated and cultured in vitro, MCF-7 breast cancer cells initially formed numerous, large spheroids (>1000 μm2 in area), however, with progressive temporal stiffening, cells demonstrated growth arrest and underwent phenotypic changes resulting in intratumoral heterogeneity. Unlike widely-investigated static mechanical models, this stiffening hydrogel allowed for progressive phenotypic changes to be observed, and fostered the development of mature organoid-like spheroids, which mimicked both the organisation and acinar-structures of mature breast epithelium. The spheroids contained a central population of cells which expressed aggressive cellular programs, evidenced by increased fibronectin expression and reduction of E-cadherin. The phenotypic heterogeneity observed using this model is more reflective of physiological tumours, demonstrating the importance of establishing temporal cues within preclinical models in future work. Overall, the developed model demonstrated a novel strategy to uncouple ECM biomechanical properties from the cellular complexities of the disease microenvironment and offers the potential for wide applicability in other 3D in vitro disease models through addition of tissue-specific dECM materials.
Collapse
Affiliation(s)
- Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Minjun Ahn
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Won-Woo Cho
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Miguel Santos
- Applied Materials Group, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Jessika Wise
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Steven G Wise
- Applied Materials Group, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Jinah Jang
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
- Tyree Institute of Health Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Khoon S Lim
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
- Light-Activated Materials Group, School of Medical Sciences, University of Sydney, Australia
| |
Collapse
|
17
|
Stricher M, Vigneron P, Delbecq F, Sarde CO, Egles C. The microalga Volvox carteri as a cell supportive building block for tissue engineering. Mater Today Bio 2024; 25:101013. [PMID: 38464496 PMCID: PMC10923841 DOI: 10.1016/j.mtbio.2024.101013] [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: 12/22/2023] [Revised: 02/18/2024] [Accepted: 02/26/2024] [Indexed: 03/12/2024] Open
Abstract
Background V. carteri f. nagariensis constitutes, in its most simplified form, a cellularized spheroid built around and stabilised by a form of primitive extracellular matrix (ECM). Methods We developed a modular approach to soft tissue engineering, by compact stacking V. carteri-based building blocks. This approach is made possible by the structure and cell adhesive properties of these building blocks, which results from the composition of their algal ECM. Results A primary biocompatibility assessment demonstrated the cytocompatibility of the algal suspension, its histogenesis-promoting properties, and that it did not induce an inflammatory response in vitro. These results allowed us to consider the use of this algal suspension for soft tissue augmentation, and to initiate an in vivo biocompatibility study. V. carteri exhibited cellular fate-directing properties, causing (i) fibroblasts to take on an alkaline phosphatase+ stem-cell-like phenotype and (ii) both human adipose-derived stem cells and mouse embryonic stem cells to differentiate into preadipocytes to adipocytes. The ability of V. carteri to support histogenesis and adipogenesis was also observed in vivo by subcutaneous tissue augmentation of athymic mice, highlighting the potential of V. carteri to support or influence tissue regeneration. Conclusions We present for the first time V. carteri as an innovative and inspiring biomaterial for tissue engineering and soft tissue regeneration. Its strategies in terms of shape, structure and composition can be central in the design of a new generation of bio-inspired heterogeneous biomaterials recapitulating more appropriately the complexity of body tissues when guiding their regeneration.
Collapse
Affiliation(s)
- Mathilde Stricher
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
| | - Pascale Vigneron
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
| | - Frederic Delbecq
- Université de Technologie de Compiègne, ESCOM, TIMR (Integrated Transformations of Renewable Matter), Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
| | - Claude-Olivier Sarde
- Université de Technologie de Compiègne, ESCOM, TIMR (Integrated Transformations of Renewable Matter), Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
| | - Christophe Egles
- Université de Technologie de Compiègne, CNRS, Biomechanics and Bioengineering, Centre de Recherche Royallieu, CEDEX CS 60 319, 60 203, Compiègne, France
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, Normandie Univ, PBS UMR 6270, 55 Rue Saint-Germain, 27 000, Évreux, France
| |
Collapse
|
18
|
Hadagali P, Fischer SL, Callaghan JP, Cronin DS. Quantifying the Importance of Active Muscle Repositioning a Finite Element Neck Model in Flexion Using Kinematic, Kinetic, and Tissue-Level Responses. Ann Biomed Eng 2024; 52:510-525. [PMID: 37923814 DOI: 10.1007/s10439-023-03396-7] [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: 08/18/2023] [Accepted: 10/24/2023] [Indexed: 11/06/2023]
Abstract
PURPOSE Non-neutral neck positions are important initial conditions in impact scenarios, associated with a higher incidence of injury. Repositioning in finite element (FE) neck models is often achieved by applying external boundary conditions (BCs) to the head while constraining the first thoracic vertebra (T1). However, in vivo, neck muscles contract to achieve a desired head and neck position generating initial loads and deformations in the tissues. In the present study, a new muscle-based repositioning method was compared to traditional applied BCs using a contemporary FE neck model for forward head flexion of 30°. METHODS For the BC method, an external moment (2.6 Nm) was applied to the head with T1 fixed, while for the muscle-based method, the flexors and extensors were co-contracted under gravity loading to achieve the target flexion. RESULTS The kinematic response from muscle contraction was within 10% of the in vivo experimental data, while the BC method differed by 18%. The intervertebral disc forces from muscle contraction were agreeable with the literature (167 N compression, 12 N shear), while the BC methodology underpredicted the disc forces owing to the lack of spine compression. Correspondingly, the strains in the annulus fibrosus increased by an average of 60% across all levels due to muscle contraction compared to BC method. CONCLUSION The muscle repositioning method enhanced the kinetic response and subsequently led to differences in tissue-level responses compared to the conventional BC method. The improved kinematics and kinetics quantify the importance of repositioning FE neck models using active muscles to achieve non-neutral neck positions.
Collapse
Affiliation(s)
- Prasannaah Hadagali
- Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Steven L Fischer
- Kinesiology and Health Sciences, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Jack P Callaghan
- Kinesiology and Health Sciences, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada
| | - Duane S Cronin
- Mechanical and Mechatronics Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON, N2L 3G1, Canada.
| |
Collapse
|
19
|
Thai NLB, Beaman HT, Perlman M, Obeng EE, Du C, Monroe MBB. Chitosan Poly(vinyl alcohol) Methacrylate Hydrogels for Tissue Engineering Scaffolds. ACS APPLIED BIO MATERIALS 2024. [PMID: 38380883 DOI: 10.1021/acsabm.3c01209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
A major challenge in tissue engineering scaffolds is controlling scaffold degradation rates during healing while maintaining mechanical properties to support tissue formation. Hydrogels are three-dimensional matrices that are widely applied as tissue scaffolds based on their unique properties that can mimic the extracellular matrix. In this study, we develop a hybrid natural/synthetic hydrogel platform to tune the properties for tissue engineering scaffold applications. We modified chitosan and poly(vinyl alcohol) (PVA) with photo-cross-linkable methacrylate functional groups and then synthesized a library of chitosan PVA methacrylate hydrogels (ChiPVAMA) with two different photoinitiators, Irgacure 2959 (I2959) and lithium phenyl-2,4,6-trimethylbenzoylphosphinate (LAP). ChiPVAMA hydrogels showed tunability in degradation rates and mechanical properties based on both the polymer content and photoinitiator type. This tunability could enable their application in a range of tissue scaffold applications. In a 2D scratch wound healing assay, all hydrogel samples induced faster wound closure compared to a gauze clinical wound dressing control. NIH/3T3 cells encapsulated in hydrogels showed a high viability (∼92%) over 14 days, demonstrating the capacity of this system as a supportive cell scaffold. In addition, hydrogels containing a higher chitosan content demonstrated a high antibacterial capacity. Overall, ChiPVAMA hydrogels provide a potential tissue engineering scaffold that is tunable, degradable, and suitable for cell growth.
Collapse
Affiliation(s)
- Nghia Le Ba Thai
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Henry T Beaman
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Megan Perlman
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Ernest E Obeng
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Changling Du
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| | - Mary Beth B Monroe
- Department of Biomedical and Chemical Engineering, Syracuse Biomaterials Institute, and BioInspired Syracuse: Institute for Material and Living Systems, Syracuse University, Syracuse, New York 13244, United States
| |
Collapse
|
20
|
Major G, Simcock J, Kumar A, Kleffmann T, Woodfield TBF, Lim KS. Comprehensive Matrisome Profiling of Human Adipose Tissue for Soft Tissue Reconstruction. Adv Biol (Weinh) 2024; 8:e2300448. [PMID: 37953659 DOI: 10.1002/adbi.202300448] [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: 08/25/2023] [Revised: 10/14/2023] [Indexed: 11/14/2023]
Abstract
For effective translation of research from tissue engineering and regenerative medicine domains, the cell-instructive extracellular matrix (ECM) of specific tissues must be accurately realized. As adipose tissue is gaining traction as a biomaterial for soft tissue reconstruction, with highly variable clinical outcomes obtained, a quantitative investigation of the adipose tissue matrisome is overdue. In this study, the human adipose tissue matrisome is profiled using quantitative sequential windowed acquisition of all theoretical fragment ion spectra - mass spectrometry (SWATH-MS) proteomics across a cohort of 13 fat-grafting patients, to provide characterization of ECM proteins within the tissue, and to understand human population variation. There are considerable differences in the expression of matrisome proteins across the patient cohort, with age and lipoaspirate collection technique contributing to the greatest variation across the core matrisome. A high abundance of basement membrane proteins (collagen IV and heparan sulfate proteoglycan) is detected, as well as fibrillar collagens I and II, reflecting the hierarchical structure of the tissue. This study provides a comprehensive proteomic evaluation of the adipose tissue matrisome and contributes to an enhanced understanding of the influence of the matrisome in adipose-related pathologies by providing a healthy reference cohort and details an experimental pipeline that can be further exploited for future biomaterial development.
Collapse
Affiliation(s)
- Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, 8011, New Zealand
| | - Jeremy Simcock
- Department of Surgery, University of Otago, Christchurch, 8011, New Zealand
| | - Abhishek Kumar
- Centre for Protein Research, Research Infrastructure Centre, University of Otago, Dunedin, 9054, New Zealand
| | - Torsten Kleffmann
- Centre for Protein Research, Research Infrastructure Centre, University of Otago, Dunedin, 9054, New Zealand
| | - Tim B F Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, 8011, New Zealand
| | - Khoon S Lim
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, 8011, New Zealand
- Light-Activated Biomaterials Group, School of Medical Science, University of Sydney, Sydney, NSW, 2006, Australia
| |
Collapse
|
21
|
Carobbio S, Pellegrinelli V, Vidal-Puig A. Adipose Tissue Dysfunction Determines Lipotoxicity and Triggers the Metabolic Syndrome: Current Challenges and Clinical Perspectives. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:231-272. [PMID: 39287854 DOI: 10.1007/978-3-031-63657-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
The adipose tissue organ is organised as distinct anatomical depots located all along the body axis, and it is constituted of three different types of adipocytes: white, beige and brown, which are integrated with vascular, immune, neural, and extracellular stroma cells. These distinct adipocytes serve different specialised functions. The main function of white adipocytes is to ensure healthy storage of excess nutrients/energy and its rapid mobilisation to supply the demand of energy imposed by physiological cues in other organs, whereas brown and beige adipocytes are designed for heat production through uncoupling lipid oxidation from energy production. The concerted action of the three types of adipocytes/tissues ensures an optimal metabolic status. However, when one or several of these adipose depots become dysfunctional because of sustained lipid/nutrient overload, then insulin resistance and associated metabolic complications ensue. These metabolic alterations close a vicious cycle that negatively affects the adipose tissue functionality and compromises global metabolic homeostasis. Optimising white adipose tissue expandability and ensuring its functional metabolic flexibility and/or promoting brown/beige mediated thermogenic activity are complementary strategies that counteract obesity and its associated lipotoxic metabolic effects. However, the development of these therapeutic approaches requires a deep understanding of adipose tissue in all broad aspects. In this chapter, we will discuss the characteristics of the different adipose tissue depots with respect to origins and precursors recruitment, plasticity, cellular composition, and expandability capacity potential as well as molecular and metabolic characteristic signatures in both physiological and pathophysiological conditions. Current antilipotoxic strategies for future clinical application are also discussed in this chapter.
Collapse
Affiliation(s)
- Stefania Carobbio
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| | - Vanessa Pellegrinelli
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Antonio Vidal-Puig
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
- Centro de Investigación Principe Felipe, Valencia, Spain.
| |
Collapse
|
22
|
Wu H, He Z, Yang L, Li H. Exploring the formation of a transparent fat portion in bacon after heating based on physicochemical characteristics and microstructure. Food Chem X 2023; 20:100964. [PMID: 38144753 PMCID: PMC10740067 DOI: 10.1016/j.fochx.2023.100964] [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: 03/01/2023] [Revised: 10/07/2023] [Accepted: 10/23/2023] [Indexed: 12/26/2023] Open
Abstract
Bacons, which possess a transparent fat tissue after heating, have high commercial value in China owing to their good sensory quality. This study was performed to explore the formation of transparent fat tissue by comparing the physicochemical characteristics and microstructures of transparent and non-transparent fat tissues. The physicochemical characteristics and microstructure of fat tissue were found to be significantly affected by drying, which increased the saturated fatty acid content and oxidation level, and decreased the moisture content and water activity (p < 0.05). Shrivelled adipocytes were observed in fat tissue after drying. Transparent and non-transparent fat tissues differed significantly in terms of moisture, fat content, texture, and fatty acid composition (p < 0.05). Multivariate statistical analysis indicated that low moisture content might be the major factor in the formation of transparent tissue, while the destruction of adipocytes also contributed to such formation.
Collapse
Affiliation(s)
- Han Wu
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Zhifei He
- College of Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
| | - Li Yang
- College of Food Science, Southwest University, Chongqing 400715, China
| | - Hongjun Li
- College of Food Science, Southwest University, Chongqing 400715, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, China
| |
Collapse
|
23
|
Footner E, Firipis K, Liu E, Baker C, Foley P, Kapsa RMI, Pirogova E, O'Connell C, Quigley A. Layer-by-Layer Analysis of In Vitro Skin Models. ACS Biomater Sci Eng 2023; 9:5933-5952. [PMID: 37791888 DOI: 10.1021/acsbiomaterials.3c00283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
In vitro human skin models are evolving into versatile platforms for the study of skin biology and disorders. These models have many potential applications in the fields of drug testing and safety assessment, as well as cosmetic and new treatment development. The development of in vitro skin models that accurately mimic native human skin can reduce reliance on animal models and also allow for more precise, clinically relevant testing. Recent advances in biofabrication techniques and biomaterials have led to the creation of increasingly complex, multilayered skin models that incorporate important functional components of skin, such as the skin barrier, mechanical properties, pigmentation, vasculature, hair follicles, glands, and subcutaneous layer. This improved ability to recapitulate the functional aspects of native skin enhances the ability to model the behavior and response of native human skin, as the complex interplay of cell-to-cell and cell-to-material interactions are incorporated. In this review, we summarize the recent developments in in vitro skin models, with a focus on their applications, limitations, and future directions.
Collapse
Affiliation(s)
- Elizabeth Footner
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Kate Firipis
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Emily Liu
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Chris Baker
- Department of Dermatology, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Skin Health Institute, Carlton, VIC 3053, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Peter Foley
- Department of Dermatology, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Skin Health Institute, Carlton, VIC 3053, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Robert M I Kapsa
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Clinical Neurosciences and Neurological Research, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Elena Pirogova
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Cathal O'Connell
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Anita Quigley
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Clinical Neurosciences and Neurological Research, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
24
|
Kho ASK, Béguin S, O'Cearbhaill ED, Ní Annaidh A. Mechanical characterisation of commercial artificial skin models. J Mech Behav Biomed Mater 2023; 147:106090. [PMID: 37717289 DOI: 10.1016/j.jmbbm.2023.106090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/19/2023] [Accepted: 08/23/2023] [Indexed: 09/19/2023]
Abstract
Understanding of the mechanical properties of skin is crucial in evaluating the performance of skin-interfacing medical devices. Artificial skin models (ASMs) have rapidly gained attention as they are able to overcome the challenges in ethically sourcing consistent and representative ex vivo animal or human tissue models. Although some ASMs have become commercialised, a thorough understanding of the mechanical properties of the skin models is crucial to ensure that they are suitable for the purpose of the study. In the present study, skin and fat layers of ASMs (Simulab®, LifeLike®, SynDaver® and Parafilm®) were mechanically characterised through hardness, needle insertion, tensile and compression testing. Different boundary constraint conditions (minimally and highly constrained) were investigated for needle insertion testing, while anisotropic properties of the skin models were investigated through different specimen orientations during tensile testing. Analysis of variance (ANOVA) tests were performed to compare the mechanical properties between the skin models. Properties of the skin models were compared against literature to determine the suitability of the skin models based on the material property of interest. All skin models offer relatively consistent mechanical performance, providing a solid basis for benchtop evaluation of skin-interfacing medical device performance. Through prioritising models with mechanical properties that are consistent with human skin data, and with limited variance, researchers can use the data presented here as a toolbox to select the most appropriate ASM for their particular application.
Collapse
Affiliation(s)
- Antony S K Kho
- UCD Centre for Biomedical Engineering, University College Dublin, Belfield Dublin 4, Ireland; I-Form Advanced Manufacturing Research Centre, School of Mechanical & Materials Engineering, University College Dublin, Belfield Dublin 4, Ireland; BD Research Centre Ireland Ltd, Carysfort Avenue, Blackrock, Ireland
| | - Steve Béguin
- BD Research Centre Ireland Ltd, Carysfort Avenue, Blackrock, Ireland
| | - Eoin D O'Cearbhaill
- UCD Centre for Biomedical Engineering, University College Dublin, Belfield Dublin 4, Ireland; I-Form Advanced Manufacturing Research Centre, School of Mechanical & Materials Engineering, University College Dublin, Belfield Dublin 4, Ireland; UCD Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield Dublin 4, Ireland; The Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Aisling Ní Annaidh
- UCD Centre for Biomedical Engineering, University College Dublin, Belfield Dublin 4, Ireland; I-Form Advanced Manufacturing Research Centre, School of Mechanical & Materials Engineering, University College Dublin, Belfield Dublin 4, Ireland; UCD Charles Institute of Dermatology, School of Medicine, University College Dublin, Belfield Dublin 4, Ireland.
| |
Collapse
|
25
|
Escudero M, Vaysse L, Eke G, Peyrou M, Villarroya F, Bonnel S, Jeanson Y, Boyer L, Vieu C, Chaput B, Yao X, Deschaseaux F, Parny M, Raymond‐Letron I, Dani C, Carrière A, Malaquin L, Casteilla L. Scalable Generation of Pre-Vascularized and Functional Human Beige Adipose Organoids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301499. [PMID: 37731092 PMCID: PMC10625054 DOI: 10.1002/advs.202301499] [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: 03/07/2023] [Revised: 08/07/2023] [Indexed: 09/22/2023]
Abstract
Obesity and type 2 diabetes are becoming a global sociobiomedical burden. Beige adipocytes are emerging as key inducible actors and putative relevant therapeutic targets for improving metabolic health. However, in vitro models of human beige adipose tissue are currently lacking and hinder research into this cell type and biotherapy development. Unlike traditional bottom-up engineering approaches that aim to generate building blocks, here a scalable system is proposed to generate pre-vascularized and functional human beige adipose tissue organoids using the human stromal vascular fraction of white adipose tissue as a source of adipose and endothelial progenitors. This engineered method uses a defined biomechanical and chemical environment using tumor growth factor β (TGFβ) pathway inhibition and specific gelatin methacryloyl (GelMA) embedding parameters to promote the self-organization of spheroids in GelMA hydrogel, facilitating beige adipogenesis and vascularization. The resulting vascularized organoids display key features of native beige adipose tissue including inducible Uncoupling Protein-1 (UCP1) expression, increased uncoupled mitochondrial respiration, and batokines secretion. The controlled assembly of spheroids allows to translate organoid morphogenesis to a macroscopic scale, generating vascularized centimeter-scale beige adipose micro-tissues. This approach represents a significant advancement in developing in vitro human beige adipose tissue models and facilitates broad applications ranging from basic research to biotherapies.
Collapse
Affiliation(s)
- Mélanie Escudero
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
- LAAS‐CNRSUniversité de Toulouse, CNRS, INSAToulouse31400France
| | - Laurence Vaysse
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | - Gozde Eke
- LAAS‐CNRSUniversité de Toulouse, CNRS, INSAToulouse31400France
| | - Marion Peyrou
- CIBER “Fisiopatologia de la Obesidad y Nutrición”, Department of Biochemistry and Molecular BiomedicineUniversity of BarcelonaMadrid28029Spain
| | - Francesc Villarroya
- CIBER “Fisiopatologia de la Obesidad y Nutrición”, Department of Biochemistry and Molecular BiomedicineUniversity of BarcelonaMadrid28029Spain
| | - Sophie Bonnel
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | - Yannick Jeanson
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | - Louisa Boyer
- LAAS‐CNRSUniversité de Toulouse, CNRS, INSAToulouse31400France
| | - Christophe Vieu
- LAAS‐CNRSUniversité de Toulouse, CNRS, INSAToulouse31400France
| | - Benoit Chaput
- Service de Chirurgie plastique, reconstructrice et esthétiqueCentre Hospitalier Universitaire RangueilToulouse31400France
| | - Xi Yao
- Faculté de MédecineUniversité Côte d'AzurINSERM, CNRS, iBVNice06103France
| | - Frédéric Deschaseaux
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | - Mélissa Parny
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
- LabHPEC, Histology and Pathology DepartmentUniversité de Toulouse, ENVTToulouse31076France
| | - Isabelle Raymond‐Letron
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
- LabHPEC, Histology and Pathology DepartmentUniversité de Toulouse, ENVTToulouse31076France
| | - Christian Dani
- Faculté de MédecineUniversité Côte d'AzurINSERM, CNRS, iBVNice06103France
| | - Audrey Carrière
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| | | | - Louis Casteilla
- RESTORE Research CenterUniversité de Toulouse, INSERM 1301, CNRS 5070, EFS, ENVTToulouse31100France
| |
Collapse
|
26
|
Hodge JG, Robinson JL, Mellott AJ. Mesenchymal Stem Cell Extracellular Vesicles from Tissue-Mimetic System Enhance Epidermal Regeneration via Formation of Migratory Cell Sheets. Tissue Eng Regen Med 2023; 20:993-1013. [PMID: 37515738 PMCID: PMC10519905 DOI: 10.1007/s13770-023-00565-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 07/31/2023] Open
Abstract
BACKGROUND The secretome of adipose-derived mesenchymal stem cells (ASCs) offers a unique approach to understanding and treating wounds, including the critical process of epidermal regeneration orchestrated by keratinocytes. However, 2D culture techniques drastically alter the secretory dynamics of ASCs, which has led to ambiguity in understanding which secreted compounds (e.g., growth factors, exosomes, reactive oxygen species) may be driving epithelialization. METHODS A novel tissue-mimetic 3D hydrogel system was utilized to enhance the retainment of a more regenerative ASC phenotype and highlight the functional secretome differences between 2D and 3D. Subsequently, the ASC-secretome was stratified by molecular weight and the presence/absence of extracellular vesicles (EVs). The ASC-secretome fractions were then evaluated to assess for the capacity to augment specific keratinocyte activities. RESULTS Culture of ASCs within the tissue-mimetic system enhanced protein secretion ~ 50%, exclusively coming from the > 100 kDa fraction. The ASC-secretome ability to modulate epithelialization functions, including migration, proliferation, differentiation, and morphology, resided within the "> 100 kDa" fraction, with the 3D ASC-secretome providing the greatest improvement. 3D ASC EV secretion was enhanced two-fold and exhibited dose-dependent effects on epidermal regeneration. Notably, ASC-EVs induced morphological changes in keratinocytes reminiscent of native regeneration, including formation of stratified cell sheets. However, only 3D-EVs promoted collective cell sheet migration and an epithelial-to-mesenchymal-like transition in keratinocytes, whereas 2D-EVs contained an anti-migratory stimulus. CONCLUSION This study demonstrates how critical the culture environment is on influencing ASC-secretome regenerative capabilities. Additionally, the critical role of EVs in modulating epidermal regeneration is revealed and their translatability for future clinical therapies is discussed.
Collapse
Affiliation(s)
- Jacob G Hodge
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA
- Department of Plastic Surgery, University of Kansas Medical Center, 3901 Rainbow Blvd., Mail Stop: 3051, Kansas City, KS, USA
| | - Jennifer L Robinson
- Bioengineering Graduate Program, University of Kansas, Lawrence, KS, USA
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, KS, USA
- Department of Orthopaedics and Sports Medicine, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Adam J Mellott
- Department of Plastic Surgery, University of Kansas Medical Center, 3901 Rainbow Blvd., Mail Stop: 3051, Kansas City, KS, USA.
- Ronawk Inc., Olathe, KS, USA.
| |
Collapse
|
27
|
Ribezzi D, Gueye M, Florczak S, Dusi F, de Vos D, Manente F, Hierholzer A, Fussenegger M, Caiazzo M, Blunk T, Malda J, Levato R. Shaping Synthetic Multicellular and Complex Multimaterial Tissues via Embedded Extrusion-Volumetric Printing of Microgels. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2301673. [PMID: 37269532 DOI: 10.1002/adma.202301673] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/24/2023] [Indexed: 06/05/2023]
Abstract
In living tissues, cells express their functions following complex signals from their surrounding microenvironment. Capturing both hierarchical architectures at the micro- and macroscale, and anisotropic cell patterning remains a major challenge in bioprinting, and a bottleneck toward creating physiologically-relevant models. Addressing this limitation, a novel technique is introduced, termed Embedded Extrusion-Volumetric Printing (EmVP), converging extrusion-bioprinting and layer-less, ultra-fast volumetric bioprinting, allowing spatially pattern multiple inks/cell types. Light-responsive microgels are developed for the first time as bioresins (µResins) for light-based volumetric bioprinting, providing a microporous environment permissive for cell homing and self-organization. Tuning the mechanical and optical properties of gelatin-based microparticles enables their use as support bath for suspended extrusion printing, in which features containing high cell densities can be easily introduced. µResins can be sculpted within seconds with tomographic light projections into centimeter-scale, granular hydrogel-based, convoluted constructs. Interstitial microvoids enhanced differentiation of multiple stem/progenitor cells (vascular, mesenchymal, neural), otherwise not possible with conventional bulk hydrogels. As proof-of-concept, EmVP is applied to create complex synthetic biology-inspired intercellular communication models, where adipocyte differentiation is regulated by optogenetic-engineered pancreatic cells. Overall, EmVP offers new avenues for producing regenerative grafts with biological functionality, and for developing engineered living systems and (metabolic) disease models.
Collapse
Affiliation(s)
- Davide Ribezzi
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
| | - Marième Gueye
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
| | - Sammy Florczak
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
| | - Franziska Dusi
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Dieuwke de Vos
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, The Netherlands
| | - Francesca Manente
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, The Netherlands
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, Naples, 80131, Italy
| | - Andreas Hierholzer
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Martin Fussenegger
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel, CH-4058, Switzerland
- Faculty of Science, University of Basel, Mattenstrasse 26, Basel, CH-4058, Switzerland
| | - Massimiliano Caiazzo
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Universiteitsweg 99, Utrecht, 3584 CG, The Netherlands
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II", Via Pansini 5, Naples, 80131, Italy
| | - Torsten Blunk
- Department of Trauma, Hand, Plastic and Reconstructive Surgery, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Jos Malda
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, The Netherlands
| | - Riccardo Levato
- Department of Orthopaedics, University Medical Center Utrecht, Utrecht University, Utrecht, 3584 CX, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3584 CT, The Netherlands
| |
Collapse
|
28
|
Ivanovska IL, Tobin MP, Bai T, Dooling LJ, Discher DE. Small lipid droplets are rigid enough to indent a nucleus, dilute the lamina, and cause rupture. J Cell Biol 2023; 222:e202208123. [PMID: 37212777 PMCID: PMC10202833 DOI: 10.1083/jcb.202208123] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 03/24/2023] [Accepted: 05/04/2023] [Indexed: 05/23/2023] Open
Abstract
The nucleus in many cell types is a stiff organelle, but fat-filled lipid droplets (FDs) in cytoplasm are seen to indent and displace the nucleus. FDs are phase-separated liquids with a poorly understood interfacial tension γ that determines how FDs interact with other organelles. Here, micron-sized FDs remain spherical as they indent peri-nuclear actomyosin and the nucleus, while causing local dilution of Lamin-B1 independent of Lamin-A,C and sometimes triggering nuclear rupture. Focal accumulation of the cytosolic DNA sensor cGAS at the rupture site is accompanied by sustained mislocalization of DNA repair factors to cytoplasm, increased DNA damage, and delayed cell cycle. Macrophages show FDs and engulfed rigid beads cause similar indentation dilution. Spherical shapes of small FDs indicate a high γ, which we measure for FDs mechanically isolated from fresh adipose tissue as ∼40 mN/m. This value is far higher than that of protein condensates, but typical of oils in water and sufficiently rigid to perturb cell structures including nuclei.
Collapse
Affiliation(s)
- Irena L. Ivanovska
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael P. Tobin
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Tianyi Bai
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Lawrence J. Dooling
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| | - Dennis E. Discher
- Molecular and Cell Biophysics Lab, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
29
|
Wu L, Zhang Z. Impact of the Paraglottic Space on Voice Production in an MRI-Based Vocal Fold Model. J Voice 2023; 37:633.e15-633.e23. [PMID: 33752927 PMCID: PMC8449798 DOI: 10.1016/j.jvoice.2021.02.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE While the vocal fold is in direct contact anteriorly with the thyroid cartilage, posteriorly the vocal fold connects to the thyroid cartilage through a soft tissue layer in the paraglottic space. Currently the paraglottic space is often neglected in computational models of phonation, in which a fixed boundary condition is often imposed on the lateral surface of the vocal fold. The goal of this study was to investigate the effect of the paraglottic space on voice production in an MRI-based vocal fold model, and how this effect may be counteracted by vocal fold stiffening due to laryngeal muscle activation. METHODS Parametric simulation study using an MRI-based computational vocal fold model. RESULTS The results showed that the presence of the paraglottic space increased the mean and amplitude of the glottal area waveform, decreased the phonation frequency and closed quotient. For the particular vocal fold geometry used in this study, the presence of the paraglottic space also reduced the occurrence of irregular vocal fold vibration. These effects of the paraglottic space became smaller with increasing paraglottic space stiffness and to a lesser degree with vocal fold stiffening. CONCLUSIONS The results suggest that the paraglottic space may be neglected in qualitative evaluations of normal phonation, but needs to be included in simulations of pathological phonation or vocal fold posturing.
Collapse
Affiliation(s)
- Liang Wu
- Department of Head and Neck Surgery, University of California, Los Angeles, California
| | - Zhaoyan Zhang
- Department of Head and Neck Surgery, University of California, Los Angeles, California.
| |
Collapse
|
30
|
Tuttle T, McClintock D, Roccabianca S. Effects of swelling and anatomical location on the viscoelastic behavior of the porcine urinary bladder wall. J Mech Behav Biomed Mater 2023; 143:105926. [PMID: 37269604 DOI: 10.1016/j.jmbbm.2023.105926] [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: 12/20/2022] [Revised: 05/18/2023] [Accepted: 05/21/2023] [Indexed: 06/05/2023]
Abstract
The ability of the urinary bladder to perform its physiological function depends largely on its mechanical characteristics. Understanding the mechanics of this tissue is crucial to the development of accurate models of not just this specific organ, but of the pelvic floor overall. In this study, we tested porcine bladder to identify variations in the tissue's viscoelastic characteristics associated with anatomical locations and swelling. We investigated this relationship using a series of stress-relaxation experiments as well as a modified Maxwell-Wiechert model to aid in the interpretation of the experimental data. Our results highlight that tissue located near the neck of the bladder presents significantly different viscoelastic characteristics than the body of the organ. This supports what was previously observed and is a valuable contribution to the understanding of the location-specific properties of the bladder. We also tested the effect of swelling, revealing that the bladder's viscoelastic behavior is mostly independent of solution osmolarity in hypoosmotic solutions, but the use of a hyperosmotic solution can significantly affect its behavior. This is significant, since several urinary tract pathologies can lead to chronic inflammation and disrupt the urothelial barrier causing increased urothelial permeability, thus subjecting the bladder wall to non-physiologic osmotic challenge.
Collapse
Affiliation(s)
- Tyler Tuttle
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, 48823, USA
| | - Dillon McClintock
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, 48823, USA
| | - Sara Roccabianca
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, 48823, USA.
| |
Collapse
|
31
|
Hatt A, Lloyd R, Bolsterlee B, Bilston LE. Strain-dependent shear properties of human adipose tissue in vivo. J Mech Behav Biomed Mater 2023; 143:105924. [PMID: 37276651 DOI: 10.1016/j.jmbbm.2023.105924] [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: 04/04/2023] [Revised: 05/14/2023] [Accepted: 05/20/2023] [Indexed: 06/07/2023]
Abstract
INTRODUCTION Human adipose tissue (fat) deforms substantially under normal physiological loading and during impact. Thus, accurate data on strain-dependent stiffness of fat is essential for the creation of accurate biomechanical models. Previous studies on ex vivo samples reported human fat to be nonlinear and viscoelastic. When static compression is combined with magnetic resonance (MR) elastography (an imaging technique used to measure viscoelasticity in vivo), the large deformation properties of tissues can be determined. Here, we use magnetic resonance elastography to quantify fat shear modulus in vivo under increasing compressive strain and compare it to the underlying passive gluteal muscle. METHODS The right buttocks of ten female participants were incrementally compressed at four levels while MR elastography (50 Hz) and mDixon images were acquired. Maps of tissue shear modulus (G*) were reconstructed from the MR elastography phase images. Tissue strain was estimated from registration of deformed and undeformed mDixon images. Linear mixed models were fit to the natural logarithm of the compressive strain and shear modulus data for each tissue. RESULTS Shear modulus increased in an exponential relationship with compressive strain in fat: Gfat*=748.5*Cyy-1.18Pa, and to a lesser extent in muscle: Gmuscle*=956.4*Cyy-0.36Pa. The baseline (undeformed) stiffness of fat was significantly lower than that of muscle (mean G*fat = 752 Pa, mean G*muscle = 1000 Pa, paired samples t-test, t = -4.24, p = 0.001). However, fat exhibited a significantly higher degree of strain dependence (characterised by the exponent of the curve, t = -6.47, p = 0.0001). CONCLUSION Static compression of human adipose tissue results in an increase in apparent viscoelastic shear modulus (stiffness), in an exponentially increasing relationship. The relationships defined here can be used in the development of physiologically realistic computational models for impact, injury and biomechanical modelling.
Collapse
Affiliation(s)
- Alice Hatt
- Neuroscience Research Australia, PO Box 1165, Randwick, NSW, 2031, Australia
| | - Robert Lloyd
- Neuroscience Research Australia, PO Box 1165, Randwick, NSW, 2031, Australia; University of New South Wales, Faculty of Medicine & Health, 18 High St, Kensington, NSW, 2052, Australia
| | - Bart Bolsterlee
- Neuroscience Research Australia, PO Box 1165, Randwick, NSW, 2031, Australia; University of New South Wales, Graduate School of Biomedical Engineering, Library Rd, Kensington, NSW, 2033, Australia
| | - Lynne E Bilston
- Neuroscience Research Australia, PO Box 1165, Randwick, NSW, 2031, Australia; University of New South Wales, Faculty of Medicine & Health, 18 High St, Kensington, NSW, 2052, Australia.
| |
Collapse
|
32
|
Kuziel G, Moore BN, Arendt LM. Obesity and Fibrosis: Setting the Stage for Breast Cancer. Cancers (Basel) 2023; 15:cancers15112929. [PMID: 37296891 DOI: 10.3390/cancers15112929] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
Obesity is a rising health concern and is linked to a worsened breast cancer prognosis. Tumor desmoplasia, which is characterized by elevated numbers of cancer-associated fibroblasts and the deposition of fibrillar collagens within the stroma, may contribute to the aggressive clinical behavior of breast cancer in obesity. A major component of the breast is adipose tissue, and fibrotic changes in adipose tissue due to obesity may contribute to breast cancer development and the biology of the resulting tumors. Adipose tissue fibrosis is a consequence of obesity that has multiple sources. Adipocytes and adipose-derived stromal cells secrete extracellular matrix composed of collagen family members and matricellular proteins that are altered by obesity. Adipose tissue also becomes a site of chronic, macrophage-driven inflammation. Macrophages exist as a diverse population within obese adipose tissue and mediate the development of fibrosis through the secretion of growth factors and matricellular proteins and interactions with other stromal cells. While weight loss is recommended to resolve obesity, the long-term effects of weight loss on adipose tissue fibrosis and inflammation within breast tissue are less clear. Increased fibrosis within breast tissue may increase the risk for tumor development as well as promote characteristics associated with tumor aggressiveness.
Collapse
Affiliation(s)
- Genevra Kuziel
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
| | - Brittney N Moore
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| | - Lisa M Arendt
- Cancer Biology Graduate Program, University of Wisconsin-Madison, 1111 Highland Ave, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, 2015 Linden Drive, Madison, WI 53706, USA
| |
Collapse
|
33
|
Karanfil AS, Louis F, Matsusaki M. Biofabrication of vascularized adipose tissues and their biomedical applications. MATERIALS HORIZONS 2023; 10:1539-1558. [PMID: 36789675 DOI: 10.1039/d2mh01391f] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Recent advances in adipose tissue engineering and cell biology have led to the development of innovative therapeutic strategies in regenerative medicine for adipose tissue reconstruction. To date, the many in vitro and in vivo models developed for vascularized adipose tissue engineering cover a wide range of research areas, including studies with cells of various origins and types, polymeric scaffolds of natural and synthetic derivation, models presented using decellularized tissues, and scaffold-free approaches. In this review, studies on adipose tissue types with different functions, characteristics and body locations have been summarized with 3D in vitro fabrication approaches. The reason for the particular focus on vascularized adipose tissue models is that current liposuction and fat transplantation methods are unsuitable for adipose tissue reconstruction as the lack of blood vessels results in inadequate nutrient and oxygen delivery, leading to necrosis in situ. In the first part of this paper, current studies and applications of white and brown adipose tissues are presented according to the polymeric materials used, focusing on the studies which could show vasculature in vitro and after in vivo implantation, and then the research on adipose tissue fabrication and applications are explained.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
34
|
Frigo CA, Bellomo S, Bigatti M, Pellegrini R, Denza G, Di Stanislao E. A finite element analysis of load distribution during donning and orthostatic posture in the ITOP hybrid subischial socket. Prosthet Orthot Int 2023; 47:204-209. [PMID: 36701634 DOI: 10.1097/pxr.0000000000000209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 11/21/2022] [Indexed: 01/28/2023]
Abstract
INTRODUCTION Pressure and shear stresses applied to the stump of a transfemoral amputee wearing a newly designed prosthetic socket have been analyzed by a finite element modeling approach. METHODS The new socket was developed by the Istituto Tecnico Ortopedico Preneste, and it was named the "hybrid subischial socket." This work aimed at understanding the loads' distribution on the stump surface in 2 operative conditions: at the end of the wearing phase and during the orthostatic posture. The model of the stump was composed of 4 different materials: the femoral bone, the muscle tissue, the fat, and the skin layers. Except for the bone (rigid), the biological tissues were modeled as Neo-Hookean, and their mechanical properties were taken from the literature. The socket was composed of a containment frame, made of carbon fiber composite material, a shell made of flexible silicone, and a liner made of hyperelastic silicone. RESULTS The results of our simulation show that the main support areas are located in a proper position, in agreement with the ideal principles of this prosthetic design, and the maximum pressures are well below the pain threshold reported in the literature for the same contact areas. CONCLUSIONS We can conclude that although the upper rim of the socket is well below the ischiatic area, the new socket design allows for a safe and comfortable support of the body weight. This is in agreement with the evidence of a good functionality and acceptance of this prosthetics gathered in the many real applications.
Collapse
Affiliation(s)
- Carlo A Frigo
- Department of Electronics, Information and Bioengineering, Politenico di Milano, Milan, Italy
- Gait Analysis Lab, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
| | - Silvia Bellomo
- Department of Electronics, Information and Bioengineering, Politenico di Milano, Milan, Italy
| | - Matteo Bigatti
- Department of Electronics, Information and Bioengineering, Politenico di Milano, Milan, Italy
| | - Roberto Pellegrini
- ITOP SpA Officine Ortopediche, Prosthetics and Orthotics Clinic, Palestrina, Italy
| | - Gabriele Denza
- ITOP SpA Officine Ortopediche, Prosthetics and Orthotics Clinic, Palestrina, Italy
| | - Eugenio Di Stanislao
- ITOP SpA Officine Ortopediche, Prosthetics and Orthotics Clinic, Palestrina, Italy
| |
Collapse
|
35
|
Mariniello MD, Ghilli M, Favati B, Gerges I, Colizzi L, Tamplenizza M, Tocchio A, Martello F, Ghilardi M, Cossu MC, Danti S, Roncella M. Cell-free biomimetic polyurethane-based scaffold for breast reconstruction following non-malignant lesion resection. A first-in-human study. Breast Cancer 2023:10.1007/s12282-023-01446-5. [PMID: 36977972 DOI: 10.1007/s12282-023-01446-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Accepted: 02/27/2023] [Indexed: 03/30/2023]
Abstract
BACKGROUND Based on the volume of tissue removed, conservative surgery (BCS) cannot always guarantee satisfactory cosmetic results, unless resorting to more complex oncoplastic approaches. Investigating an alternative to optimize aesthetic outcomes minimizing surgical complexity, was the purpose of this study. We assessed an innovative surgical procedure based on the use of a biomimetic polyurethane-based scaffold intended for regenerating soft-tissue resembling fat, in patients undergoing BCS for non-malignant breast lesions. Safety and performance of the scaffold, and safety and feasibility of the entire implant procedure were evaluated. METHODS A volunteer sample of 15 female patients underwent lumpectomy with immediate device positioning, performing seven study visits with six-month follow-up. We evaluated incidence of adverse events (AEs), changes in breast appearance (using photographs and anthropomorphic measurements), interference with ultrasound and MRI (assessed by two independent investigators), investigator's satisfaction (through a VAS scale), patient's pain (through a VAS scale) and quality of life (QoL) (using the BREAST-Q© questionnaire). Data reported are the results of the interim analysis on the first 5 patients. RESULTS No AEs were device related nor serious. Breast appearance was unaltered and the device did not interference with imaging. High investigator's satisfaction, minimal post-operative pain and positive impact on QoL were also detected. CONCLUSIONS Albeit on a limited number of patients, data showed positive outcomes both in terms of safety and performance, paving the way to an innovative breast reconstructive approach with a potential remarkable impact on clinical application of tissue engineering. TRIAL REGISTRATION ClinicalTrials.gov (NCT04131972, October 18, 2019).
Collapse
Affiliation(s)
| | - Matteo Ghilli
- Breast Cancer Center, University Hospital of Pisa, Via Roma 57, 56126, Pisa, Italy
| | - Benedetta Favati
- Breast Radiology, Breast Cancer Center, University Hospital of Pisa, Via Roma 57, 56126, Pisa, Italy
| | | | - Livio Colizzi
- Breast Cancer Center, University Hospital of Pisa, Via Roma 57, 56126, Pisa, Italy
| | | | | | | | - Maria Ghilardi
- Breast Cancer Center, University Hospital of Pisa, Via Roma 57, 56126, Pisa, Italy
| | - Maria Cristina Cossu
- Breast Radiology, Breast Cancer Center, University Hospital of Pisa, Via Roma 57, 56126, Pisa, Italy
| | - Serena Danti
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56122, Pisa, Italy
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave., Cambridge, MA, 02139, USA
| | - Manuela Roncella
- Breast Cancer Center, University Hospital of Pisa, Via Roma 57, 56126, Pisa, Italy
| |
Collapse
|
36
|
Genç H, Cianciosi A, Lohse R, Stahlhut P, Groll J, Alexiou C, Cicha I, Jüngst T. Adjusting Degree of Modification and Composition of gelAGE-Based Hydrogels Improves Long-Term Survival and Function of Primary Human Fibroblasts and Endothelial Cells in 3D Cultures. Biomacromolecules 2023; 24:1497-1510. [PMID: 36786807 DOI: 10.1021/acs.biomac.2c01536] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
This study aimed to develop a suitable hydrogel-based 3D platform to support long-term culture of primary endothelial cells (ECs) and fibroblasts. Two hydrogel systems based on allyl-modified gelatin (gelAGE), G1MM and G2LH, were cross-linked via thiol-ene click reaction with a four-arm thiolated polyethylene glycol (PEG-4-SH). Compared to G1MM, the G2LH hydrogel was characterized by the lower polymer content and cross-linking density with a softer matrix and homogeneous and open porosity. Cell viability in both hydrogels was comparable, although the G2LH-based platform supported better F-actin organization, cell-cell interactions, and collagen and fibronectin production. In co-cultures, early morphogenesis leading to tubular-like structures was observed within 2 weeks. Migration of fibroblasts out of spheroids embedded in the G2LH hydrogels started after 5 days of incubation. Taken together, the results demonstrated that the G2LH hydrogel fulfilled the demands of both ECs and fibroblasts to enable long-term culture and matrix remodeling.
Collapse
Affiliation(s)
- Hatice Genç
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Endowed Professorship for Nanomedicine, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Alessandro Cianciosi
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Würzburg 97070, Germany
| | - Raphael Lohse
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Endowed Professorship for Nanomedicine, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Philipp Stahlhut
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Würzburg 97070, Germany
| | - Jürgen Groll
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Würzburg 97070, Germany
| | - Christoph Alexiou
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Endowed Professorship for Nanomedicine, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Iwona Cicha
- Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Endowed Professorship for Nanomedicine, Department of Otorhinolaryngology, Head and Neck Surgery, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen 91054, Germany
| | - Tomasz Jüngst
- Department of Functional Materials in Medicine and Dentistry at the Institute of Functional Materials and Biofabrication (IFB), University of Würzburg and KeyLab Polymers for Medicine of the Bavarian Polymer Institute (BPI), Würzburg 97070, Germany
| |
Collapse
|
37
|
Hodge JG, Decker HE, Robinson JL, Mellott AJ. Tissue-mimetic culture enhances mesenchymal stem cell secretome capacity to improve regenerative activity of keratinocytes and fibroblasts in vitro. Wound Repair Regen 2023; 31:367-383. [PMID: 36866522 DOI: 10.1111/wrr.13076] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/18/2023] [Accepted: 02/21/2023] [Indexed: 03/04/2023]
Abstract
Mesenchymal stem/stromal cells (MSCs) are a heterogenous population of multipotent and highly secretory cells currently being investigated in the field of wound healing for their ability to augment tissue responses. The adaptive response of MSC populations to the rigid substrate of current 2D culture systems has been considered to result in a deterioration of regenerative 'stem-like' properties. In this study, we characterise how the improved culture of adipose-derived mesenchymal stem cells (ASCs) within a tissue-mimetic 3D hydrogel system, that is mechanically similar to native adipose tissue, enhances their regenerative capabilities. Notably, the hydrogel system contains a porous microarchitecture that permits mass transport, enabling efficient collection of secreted cellular compounds. By utilising this 3D system, ASCs retained a significantly higher expression of ASC 'stem-like' markers while demonstrating a significant reduction in senescent populations, relative to 2D. Additionally, culture of ASCs within the 3D system resulted in enhanced secretory activity with significant increases in the secretion of proteinaceous factors, antioxidants and extracellular vesicles (EVs) within the conditioned media (CM) fraction. Lastly, treatment of wound healing cells, keratinocytes (KCs) and fibroblasts (FBs), with ASC-CM from the 2D and 3D systems resulted in augmented functional regenerative activity, with ASC-CM from the 3D system significantly increasing KC and FB metabolic, proliferative and migratory activity. This study demonstrates the potential beneficial role of MSC culture within a tissue-mimetic 3D hydrogel system that more closely mimics native tissue mechanics, and subsequently how the improved phenotype augments secretory activity and potential wound healing capabilities of the MSC secretome.
Collapse
Affiliation(s)
- Jacob G Hodge
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | | | - Jennifer L Robinson
- Bioengineering Graduate Program, University of Kansas, Lawrence, Kansas, USA
- Department of Chemical and Petroleum Engineering, University of Kansas, Lawrence, Kansas, USA
| | - Adam J Mellott
- Department of Plastic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
- Ronawk, LLC, Olathe, Kansas, USA
| |
Collapse
|
38
|
Arnold N, Scott J, Bush TR. A review of the characterizations of soft tissues used in human body modeling: Scope, limitations, and the path forward. J Tissue Viability 2023; 32:286-304. [PMID: 36878737 DOI: 10.1016/j.jtv.2023.02.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 02/06/2023] [Accepted: 02/10/2023] [Indexed: 02/27/2023]
Abstract
Soft tissue material properties are vital to human body models that evaluate interactions between the human body and its environment. Such models evaluate internal stress/strain responses in soft tissues to investigate issues like pressure injuries. Numerous constitutive models and parameters have been used to represent mechanical behavior of soft tissues in biomechanical models under quasi-static loading. However, researchers reported that generic material properties cannot accurately represent specific target populations due to large inter-individual variability. Two challenges that exist are experimental mechanical characterization and constitutive modeling of biological soft tissues and personalization of constitutive parameters using non-invasive, non-destructive bedside testing methods. It is imperative to understand the scope and appropriate applications for reported material properties. Thus, the goal of this paper was to compile studies from which soft tissue material properties were obtained and categorize them by source of tissue samples, methods used to quantify deformation, and material models used to describe tissues. The collected studies displayed wide ranges of material properties, and factors that affected the properties included whether tissue samples were in vivo or ex vivo, from humans or animals, the body region tested, body position during in vivo studies, deformation measurements, and material models used to describe tissues. Because of the factors that affected reported material properties, it is clear that much progress has been made in understanding soft tissue responses to loading, yet there is a need to broaden the scope of reported soft tissue material properties and better match reported properties to appropriate human body models.
Collapse
Affiliation(s)
- Nicole Arnold
- Department of Mechanical Engineering, Michigan State University, 428 S Shaw Lane, Rm. 2555 Engineering Building, East Lansing, MI, 48824-1226, USA
| | - Justin Scott
- Department of Mechanical Engineering, Michigan State University, 428 S Shaw Lane, Rm. 2555 Engineering Building, East Lansing, MI, 48824-1226, USA
| | - Tamara Reid Bush
- Department of Mechanical Engineering, Michigan State University, 428 S Shaw Lane, Rm. 2555 Engineering Building, East Lansing, MI, 48824-1226, USA.
| |
Collapse
|
39
|
Wang CJ, Noble PB, Elliot JG, James AL, Wang KCW. From Beneath the Skin to the Airway Wall: Understanding the Pathological Role of Adipose Tissue in Comorbid Asthma-Obesity. Compr Physiol 2023; 13:4321-4353. [PMID: 36715283 DOI: 10.1002/cphy.c220011] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article provides a contemporary report on the role of adipose tissue in respiratory dysfunction. Adipose tissue is distributed throughout the body, accumulating beneath the skin (subcutaneous), around organs (visceral), and importantly in the context of respiratory disease, has recently been shown to accumulate within the airway wall: "airway-associated adipose tissue." Excessive adipose tissue deposition compromises respiratory function and increases the severity of diseases such as asthma. The mechanisms of respiratory impairment are inflammatory, structural, and mechanical in nature, vary depending on the anatomical site of deposition and adipose tissue subtype, and likely contribute to different phenotypes of comorbid asthma-obesity. An understanding of adipose tissue-driven pathophysiology provides an opportunity for diagnostic advancement and patient-specific treatment. As an exemplar, the potential impact of airway-associated adipose tissue is highlighted, and how this may change the management of a patient with asthma who is also obese. © 2023 American Physiological Society. Compr Physiol 13:4321-4353, 2023.
Collapse
Affiliation(s)
- Carolyn J Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Peter B Noble
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - John G Elliot
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia
| | - Alan L James
- Department of Pulmonary Physiology and Sleep Medicine, West Australian Sleep Disorders Research Institute, Sir Charles Gairdner Hospital, Nedlands, Western Australia, Australia.,Medical School, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Kimberley C W Wang
- School of Human Sciences, The University of Western Australia, Crawley, Western Australia, Australia.,Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|
40
|
Sun K, Li X, Scherer PE. Extracellular Matrix (ECM) and Fibrosis in Adipose Tissue: Overview and Perspectives. Compr Physiol 2023; 13:4387-4407. [PMID: 36715281 PMCID: PMC9957663 DOI: 10.1002/cphy.c220020] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Fibrosis in adipose tissue is a major driver of obesity-related metabolic dysregulation. It is characterized by an overaccumulation of extracellular matrix (ECM) during unhealthy expansion of adipose tissue in response to over nutrition. In obese adipose-depots, hypoxia stimulates multiple pro-fibrotic signaling pathways in different cell populations, thereby inducing the overproduction of the ECM components, including collagens, noncollagenous proteins, and additional enzymatic components of ECM synthesis. As a consequence, local fibrosis develops. The result of fibrosis-induced mechanical stress not only triggers cell necrosis and inflammation locally in adipose tissue but also leads to system-wide lipotoxicity and insulin resistance. A better understanding of the mechanisms underlying the obesity-induced fibrosis will help design therapeutic approaches to reduce or reverse the pathological changes associated with obese adipose tissue. Here, we aim to summarize the major advances in the field, which include newly identified fibrotic factors, cell populations that contribute to the fibrosis in adipose tissue, as well as novel mechanisms underlying the development of fibrosis. We further discuss the potential therapeutic strategies to target fibrosis in adipose tissue for the treatment of obesity-linked metabolic diseases and cancer. © 2023 American Physiological Society. Compr Physiol 13:4387-4407, 2023.
Collapse
Affiliation(s)
- Kai Sun
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Xin Li
- Center for Metabolic and Degenerative Diseases, Institute of Molecular Medicine, University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Philipp E. Scherer
- Department of Internal Medicine, Touchstone Diabetes Center, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
- Department of Cell Biology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
41
|
Neumann EE, Doherty S, Bena J, Erdemir A. Role of multi-layer tissue composition of musculoskeletal extremities for prediction of in vivo surface indentation response and layer deformations. PLoS One 2023; 18:e0284721. [PMID: 37083580 PMCID: PMC10121013 DOI: 10.1371/journal.pone.0284721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 04/05/2023] [Indexed: 04/22/2023] Open
Abstract
Emergent mechanics of musculoskeletal extremities (surface indentation stiffness and tissue deformation characteristics) depend on the underlying composition and mechanics of each soft tissue layer (i.e. skin, fat, and muscle). Limited experimental studies have been performed to explore the layer specific relationships that contribute to the surface indentation response. The goal of this study was to examine through statistical modeling how the soft tissue architecture contributed to the aggregate mechanical surface response across 8 different sites of the upper and lower extremities. A publicly available dataset was used to examine the relationship of soft tissue thickness (fat and muscle) to bulk tissue surface compliance. Models required only initial tissue layer thicknesses, making them usable in the future with only a static ultrasound image. Two physics inspired models (series of linear springs), which allowed reduced statistical representations (combined locations and location specific), were explored to determine the best predictability of surface compliance and later individual layer deformations. When considering the predictability of the experimental surface compliance, the physics inspired combined locations model showed an improvement over the location specific model (percent difference of 25.4 +/- 27.9% and 29.7 +/- 31.8% for the combined locations and location specific models, respectively). While the statistical models presented in this study show that tissue compliance relies on the individual layer thicknesses, it is clear that there are other variables that need to be accounted for to improve the model. In addition, the individual layer deformations of fat and muscle tissues can be predicted reasonably well with the physics inspired models, however additional parameters may improve the robustness of the model outcomes, specifically in regard to capturing subject specificity.
Collapse
Affiliation(s)
- Erica E Neumann
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
- Computational Biomodeling (CoBi) Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
| | - Sean Doherty
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
- Computational Biomodeling (CoBi) Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
| | - James Bena
- Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, United States of America
| | - Ahmet Erdemir
- Department of Biomedical Engineering, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
- Computational Biomodeling (CoBi) Core, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States of America
| |
Collapse
|
42
|
de Guzman RC, Meer AS, Mathews AA, Israel AR, Moses MT, Sams CM, Deegan DB. Reduced fibrous capsule elastic fibers from biologic ECM-enveloped CIEDs in minipigs, supported with a novel compression mechanics model. Biomed Mater Eng 2022:BME221488. [PMID: 36617774 DOI: 10.3233/bme-221488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Fibrous capsules (Fb) in response to cardiovascular implantable electronic devices (CIEDs), including a pacemaker (P) system, can produce patient discomfort and difficulties in revision surgery due partially to their increased compressive strength, previously linked to elevated tissue fibers. OBJECTIVE A preliminary study to quantify structural proteins, determine if biologic extracellular matrix-enveloped CIEDs (PECM) caused differential Fb properties, and to implement a realistic mechanical model. METHODS Retrieved Fb (-P and -PECM) from minipigs were subjected to biomechanical (shear oscillation and uniaxial compression) and histological (collagen I and elastin) analyses. RESULTS Fb-PECM showed significant decreases compared to Fb-P in: low strain-loss modulus (390 vs. 541 Pa) across angular frequencies, high strain-compressive elastic modulus (1043 vs. 2042 kPa), and elastic fiber content (1.92 vs. 3.15 μg/mg tissue). Decreases in elastin were particularly noted closer to the implant's surface (Fb-PECM = 71% vs. Fb-P = 143% relative to dermal elastin at mid-tangential sections) and verified with a solid mechanics hyperelasticity with direction-dependent fiber viscoelasticity compression simulation (r2 ≥ 98.9%). CONCLUSIONS The biologic envelope composed of decellularized porcine small intestine submucosa ECM for CIEDs promoted fibrous tissues with less elastic fibers. Novel compression modeling analyses directly correlated this singular reduction to more desirable subcutaneous tissue mechanics.
Collapse
Affiliation(s)
- Roche C de Guzman
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY, USA
| | - Allison S Meer
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY, USA.,Department of Biology, Hofstra University, Hempstead, NY, USA
| | - Aidan A Mathews
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY, USA.,Department of Biology, Hofstra University, Hempstead, NY, USA
| | - Atara R Israel
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY, USA
| | - Michael T Moses
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY, USA
| | - Clarence M Sams
- Bioengineering Program, Department of Engineering, Hofstra University, Hempstead, NY, USA
| | | |
Collapse
|
43
|
Kollech HG, Chao MR, Stark AC, German RZ, Paniello RC, Christensen MB, Barkmeier-Kraemer JM, Vande Geest JP. Extracellular matrix deformations of the porcine recurrent laryngeal nerve in response to hydrostatic pressure. Acta Biomater 2022; 153:364-373. [PMID: 36152909 PMCID: PMC10627241 DOI: 10.1016/j.actbio.2022.09.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 08/22/2022] [Accepted: 09/15/2022] [Indexed: 11/25/2022]
Abstract
Damage to the recurrent laryngeal nerve (RLN) caused by supraphysiological compression or tension imposed by adjacent tissue structures, such as the aorta, may contribute to onset of idiopathic unilateral vocal fold paralysis (iUVP) resulting in difficulty speaking, breathing, and swallowing. We previously demonstrated in adolescent pigs that the right RLN epineurium exhibits uniform composition of adipose tissue, with larger quantities along its length within the neck region in contrast to the left RLN that shows greater collagen composition in the thoracic region and greater quantities of adipose tissue in the neck region. In contrast, the epineurium in piglets was primarily composed of collagen tissue that remained uniform along the length of the left and right RLNs. Tensile testing of the left and right RLN in piglets and pigs showed associated differences in strain by RLN side and segment by age. The goal of this study was to investigate how external hydrostatic compression of the RLN affects the nerve's connective tissue and microstructure. RLN segments were harvested from the distal (cervical/neck) regions and proximal (subclavian for the right RLN, thoracic for the left RLN) regions from eight adolescent pigs and nine piglets. RLN segments were isolated and assessed under fluid compression to test hypotheses regarding epineurium composition and response to applied forces. Second harmonic generation (SHG) imaging of epineurial collagen was conducted at 0, 40, and 80 mmHg of compression. The cartesian strain tensor, principal strain (Eps1), and principal direction of the RLN collagen fibers were determined at each pressure step. Significantly larger values of the 1st principal strain occurred in the proximal segments of the pig left RLN when compared to the same segment in piglets (p = 0.001, pig = 0.0287 [IQR = 0.0161 - 0.0428], piglet = 0.0061 [IQR = 0.0033 - 0.0156]). Additionally, the median transverse strain Eyy) for the second pressure increment was larger in the right proximal segment of pigs compared to piglets (p < 0.001, pig = 0.0122 [IQR = 0.0033 - 0.0171], piglet = 0.0013 [IQR = 0.00001 - 0.0028]). Eyy values were significantly larger in the right proximal RLN versus the left proximal RLNs in pigs but not in piglets (p < 0.001). In contrast to piglets, histological analysis of pig RLN demonstrated increased axial alignment of epineurial and endoneurial collagen in response to compressive pressure. These findings support the hypothesis that the biomechanical response of the RLN to compressive pressure changed from being similar to being different between the right and left RLNs during development in the porcine model. Further investigation of these findings associated with age-related onset of idiopathic UVP may illuminate underlying etiologic mechanisms. STATEMENT OF SIGNIFICANCE: Damage to the recurrent laryngeal nerve (RLN) caused by compression imposed by the aorta may contribute to the onset of left-sided idiopathic unilateral vocal fold paralysis resulting in difficulty speaking, breathing, and swallowing. The goal of this study was to investigate how compression affects the connective tissue and microstructure of the RLN. We quantified the pressure induced deformation of the RLN using multiphoton imaging as a function of both location (proximal versus distal) and age (piglets, adolescent pigs). Our results demonstrate that the biomechanical response of the RLN to compression changes in the right versus left RLN throughout development, providing further evidence that the the left RLN is exposed to increasing dynamic loads with age.
Collapse
Affiliation(s)
- Hirut G Kollech
- Computational Modeling and Simulation Program, University of Pittsburgh, Pittsburgh, PA, USA
| | - Melissa R Chao
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| | - Amanda C Stark
- National Center for Voice and Speech, University of Utah, Salt Lake City, UT, USA
| | - Rebecca Z German
- Department of Anatomy and Neurobiology, Northeast Ohio Medical University (NEOMED), Rootstown, OH, USA
| | - Randal C Paniello
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Julie M Barkmeier-Kraemer
- Department of Surgery, Division of Otolaryngology-Head and Neck Surgery, University of Utah, UT, USA
| | - Jonathan P Vande Geest
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; Department of Mechanical Engineering and Material Science, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
44
|
Albrecht FB, Schmidt FF, Volz AC, Kluger PJ. Bioprinting of 3D Adipose Tissue Models Using a GelMA-Bioink with Human Mature Adipocytes or Human Adipose-Derived Stem Cells. Gels 2022; 8:gels8100611. [PMID: 36286112 PMCID: PMC9601941 DOI: 10.3390/gels8100611] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 12/31/2022] Open
Abstract
Adipose tissue is related to the development and manifestation of multiple diseases, demonstrating the importance of suitable in vitro models for research purposes. In this study, adipose tissue lobuli were explanted, cultured, and used as an adipose tissue control to evaluate in vitro generated adipose tissue models. During culture, lobule exhibited a stable weight, lactate dehydrogenase, and glycerol release over 15 days. For building up in vitro adipose tissue models, we adapted the biomaterial gelatin methacryloyl (GelMA) composition and handling to homogeneously mix and bioprint human primary mature adipocytes (MA) and adipose-derived stem cells (ASCs), respectively. Accelerated cooling of the bioink turned out to be essential for the homogeneous distribution of lipid-filled MAs in the hydrogel. Last, we compared manual and bioprinted GelMA hydrogels with MA or ASCs and the explanted lobules to evaluate the impact of the printing process and rate the models concerning the physiological reference. The viability analyses demonstrated no significant difference between the groups due to additive manufacturing. The staining of intracellular lipids and perilipin A suggest that GelMA is well suited for ASCs and MA. Therefore, we successfully constructed physiological in vitro models by bioprinting MA-containing GelMA bioinks.
Collapse
Affiliation(s)
- Franziska B. Albrecht
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
- Faculty of Natural Science, University of Hohenheim, 70599 Stuttgart, Germany
| | - Freia F. Schmidt
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Ann-Cathrin Volz
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Petra J. Kluger
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany
- Correspondence: ; Tel.: +49-7121-271-2061
| |
Collapse
|
45
|
Mechanical Behavior of Subcutaneous and Visceral Abdominal Adipose Tissue in Patients with Obesity. Processes (Basel) 2022. [DOI: 10.3390/pr10091798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The mechanical characterization of adipose tissues is important for various medical purposes, including plastic surgery and biomechanical applications, such as computational human body models for the simulation of surgical procedures or injury prediction, for example, in the evaluation of vehicle crashworthiness. In this context, the measurement of human subcutaneous adipose tissue (SAT) and visceral adipose tissue (VAT) mechanical properties in relation to subject characteristics may be really relevant. The aim of this work was to properly characterize the mechanical response of adipose tissues in patients with obesity. Then, the data were exploited to develop a reliable finite element model of the adipose tissues characterized by a constitutive material model that accounted for nonlinear elasticity and time dependence. Mechanical tests have been performed on both SAT and VAT specimens, which have been harvested from patients with severe obesity during standard laparoscopic sleeve gastrectomy intervention. The experimental campaign included indentation tests, which permitted us to obtain the initial/final indentation stiffnesses for each specimen. Statistical results revealed a higher statistical stiffness in SAT than in VAT, with an initial/final indentation stiffness of 1.65 (SD ± 0.29) N/30.30 (SD ± 20) N compared to 1.29 (SD ± 0.30) N/21.00 (SD ± 16) N. Moreover, the results showed that gender, BMI, and age did not significantly affect the stiffness. The experimental results were used in the identification of the constitutive parameters to be inserted in the constitutive material model. Such constitutive characterization of VAT and SAT mechanics can be the starting point for the future development of more accurate computational models of the human adipose tissue and, in general, of the human body for the optimization of numerous medical and biomechanical procedures and applications.
Collapse
|
46
|
Xue W, Yu SY, Kuss MA, Kong Y, Shi W, Chung S, Kim SY, Duan B. 3D bioprinted white adipose model for in vitro study of cancer-associated cachexia induced adipose tissue remodeling. Biofabrication 2022; 14. [PMID: 35504266 DOI: 10.1088/1758-5090/ac6c4b] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 05/03/2022] [Indexed: 11/11/2022]
Abstract
Cancer-associated cachexia (CAC) is a complex metabolic and behavioral syndrome with multiple manifestations that involve systemic inflammation, weight loss, and adipose lipolysis. It impacts the quality of life of patients and is the direct cause of death in 20-30% of cancer patients. The severity of fat loss and adipose tissue remodeling negatively correlate with patients' survival outcomes. To address the mechanism of fat loss and design potential approaches to prevent the process, it will be essential to understand CAC pathophysiology through white adipose tissue models. In the present study, an engineered human white adipose tissue (eWAT) model based on three-dimensional (3D) bioprinting was developed and treated with pancreatic cancer cell-conditioned medium (CM) to mimic the status of CAC in vitro. We found that the CM treatment significantly increased the lipolysis and accumulation of the extracellular matrix (ECM). The 3D eWATs were further vascularized to study the influence of vascularization on lipolysis and CAC progression, which was largely unknown. Results demonstrated that CM treatment improved the angiogenesis of vascularized eWATs (veWATs), and veWATs demonstrated decreased glycerol release but increased Ucp1 expression, compared to eWATs. Many unique inflammatory cytokines (IL-8, CXCL-1, GM-CSF, etc) from the CM were detected and supposed to contribute to eWAT lipolysis, Ucp1 up-regulation, and ECM development. In response to CM treatment, eWATs also secreted inflammatory adipokines related to the metastatic ability of cancer, muscle atrophy, and vascularization (NGAL, CD54, IGFBP-2, etc). Our work demonstrated that the eWAT is a robust model for studying cachectic fat loss and the accompanying remodeling of adipose tissue. It is therefore a useful tool for future research exploring CAC physiologies and developing potential therapies.
Collapse
Affiliation(s)
- Wen Xue
- University of Nebraska Medical Center, DRCII, Omaha, 68198-7400, UNITED STATES
| | - Seok-Yeong Yu
- Regenerative Medicine, University of Nebraska Medical Center, DRCII R6035, Omaha, Nebraska, 68198-7400, UNITED STATES
| | - Mitchell A Kuss
- Regenerative Medicine, University of Nebraska Medical Center, DRCII, Omaha, Nebraska, 68106, UNITED STATES
| | - Yunfan Kong
- University of Nebraska Medical Center, DRCII, Omaha, 68198-7400, UNITED STATES
| | - Wen Shi
- University of Nebraska Medical Center, DRCII, Omaha, Nebraska, 68106, UNITED STATES
| | - Soonkyu Chung
- University of Massachusetts Amherst, UMA, Amherst, Massachusetts, 01003, UNITED STATES
| | - So-Youn Kim
- Regenerative Medicine, University of Nebraska Medical Center, DRCII R6035, Omaha, Nebraska, 68198-7400, UNITED STATES
| | - Bin Duan
- Regenerative Medicine, University of Nebraska Medical Center, DRCII R6035, Omaha, Nebraska, 68198-7400, UNITED STATES
| |
Collapse
|
47
|
Applications of Stem Cell Therapy and Adipose-Derived Stem Cells for Skin Repair. CURRENT DERMATOLOGY REPORTS 2022. [DOI: 10.1007/s13671-022-00357-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
48
|
Unamuno X, Gómez-Ambrosi J, Becerril S, Álvarez-Cienfuegos FJ, Ramírez B, Rodríguez A, Ezquerro S, Valentí V, Moncada R, Mentxaka A, Llorente M, Silva C, Elizalde MDLR, Catalán V, Frühbeck G. Changes in mechanical properties of adipose tissue after bariatric surgery driven by extracellular matrix remodelling and neovascularization are associated with metabolic improvements. Acta Biomater 2022; 141:264-279. [PMID: 35007786 DOI: 10.1016/j.actbio.2022.01.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/08/2023]
Abstract
Biomechanical properties of adipose tissue (AT) are closely involved in the development of obesity-associated comorbidities. Bariatric surgery (BS) constitutes the most effective option for a sustained weight loss in addition to improving obesity-associated metabolic diseases including type 2 diabetes (T2D). We aimed to determine the impact of weight loss achieved by BS and caloric restriction (CR) on the biomechanical properties of AT. BS but not CR changed the biomechanical properties of epididymal white AT (EWAT) from a diet-induced obesity rat model, which were associated with metabolic improvements. We found decreased gene expression levels of collagens and Lox together with increased elastin and Mmps mRNA levels in EWAT after BS, which were also associated with the biomechanical properties. Moreover, an increased blood vessel density was observed in EWAT after surgery, confirmed by an upregulation of Acta2 and Antxr1 gene expression levels, which was also correlated with the biomechanical properties. Visceral AT from patients with obesity showed increased stiffness after tensile tests compared to the EWAT from the animal model. This study uncovers new insights into EWAT adaptation after BS with decreased collagen crosslink and synthesis as well as an increased degradation together with enhanced blood vessel density providing, simultaneously, higher stiffness and more ductility. STATEMENT OF SIGNIFICANCE: Biomechanical properties of the adipose tissue (AT) are closely involved in the development of obesity-associated comorbidities. In this study, we show for the first time that biomechanical properties of AT determined by E, UTS and strain at UTS are decreased in obesity, being increased after bariatric surgery by the promotion of ECM remodelling and neovascularization. Moreover, these changes in biomechanical properties are associated with improvements in metabolic homeostasis. Consistently, a better characterization of the plasticity and biomechanical properties of the AT after bariatric surgery opens up a new field for the development of innovative strategies for the reduction of fibrosis and inflammation in AT as well as to better understand obesity and its associated comorbidities.
Collapse
|
49
|
Routzong MR, Martin LC, Rostaminia G, Abramowitch S. Urethral support in female urinary continence part 2: a computational, biomechanical analysis of Valsalva. Int Urogynecol J 2022; 33:551-561. [PMID: 33787951 DOI: 10.1007/s00192-021-04694-1] [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: 11/11/2020] [Accepted: 01/10/2021] [Indexed: 10/21/2022]
Abstract
INTRODUCTION AND HYPOTHESIS In Part 1, we observed urethral mechanics during Valsalva that oppose current continence theories. In this study, we utilize a finite element model to elucidate the role of supportive tissues on the urethra during Valsalva. By determining the sensitivity of urethral motion and deformations to variations in tissue stiffnesses, we formulate new hypotheses regarding mechanisms of urethral passive closure. METHODS Anatomy was segmented from a nulliparous, continent woman at rest. The model was tuned such that urethral motion during Valsalva matched that observed in that patient. Urethra and surrounding tissue material properties were varied using Latin hypercube sampling to perform a sensitivity analysis. As in Part 1, urethral length, proximal and distal swinging, and shape parameters were measured at peak Valsalva for 50 simulations, and partial rank correlation coefficients were calculated between all model inputs and outputs. Cumulative influence factors determined which tissue properties were meaningfully influential (≥ 0.5). RESULTS The material properties of the urethra, perineal membrane, bladder, and paraurethral connective tissues meaningfully influenced urethral motion, deformation, and shape. Reduction of the urethral stiffness and/or the perineal membrane soft constraint resulted in simulated urethral motions and shapes associated with stress urinary incontinence in Part 1. CONCLUSIONS The data from Parts 1 and 2 suggest that connective tissues guide the controlled swinging motion and deformation of the urethra needed for passive closure during Valsalva. The swinging and kinking quantified in Part 1 and simulated in Part 2 are inconsistent with current continence theories.
Collapse
Affiliation(s)
- Megan R Routzong
- Department of Bioengineering, University of Pittsburgh, 300 O'Hara Street, 406 Benedum Hall, Pittsburgh, PA, 15260, USA
| | - Liam C Martin
- Department of Bioengineering, University of Pittsburgh, 300 O'Hara Street, 406 Benedum Hall, Pittsburgh, PA, 15260, USA
| | - Ghazaleh Rostaminia
- Female Pelvic Medicine and Reconstructive Surgery (FPMRS), Division of Urogynecology, University of Chicago Pritzker School of Medicine, NorthShore University HealthSystem, Skokie, IL, USA
| | - Steven Abramowitch
- Department of Bioengineering, University of Pittsburgh, 300 O'Hara Street, 406 Benedum Hall, Pittsburgh, PA, 15260, USA.
| |
Collapse
|
50
|
Yee C, Dickson KA, Muntasir MN, Ma Y, Marsh DJ. Three-Dimensional Modelling of Ovarian Cancer: From Cell Lines to Organoids for Discovery and Personalized Medicine. Front Bioeng Biotechnol 2022; 10:836984. [PMID: 35223797 PMCID: PMC8866972 DOI: 10.3389/fbioe.2022.836984] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 01/19/2022] [Indexed: 12/11/2022] Open
Abstract
Ovarian cancer has the highest mortality of all of the gynecological malignancies. There are several distinct histotypes of this malignancy characterized by specific molecular events and clinical behavior. These histotypes have differing responses to platinum-based drugs that have been the mainstay of therapy for ovarian cancer for decades. For histotypes that initially respond to a chemotherapeutic regime of carboplatin and paclitaxel such as high-grade serous ovarian cancer, the development of chemoresistance is common and underpins incurable disease. Recent discoveries have led to the clinical use of PARP (poly ADP ribose polymerase) inhibitors for ovarian cancers defective in homologous recombination repair, as well as the anti-angiogenic bevacizumab. While predictive molecular testing involving identification of a genomic scar and/or the presence of germline or somatic BRCA1 or BRCA2 mutation are in clinical use to inform the likely success of a PARP inhibitor, no similar tests are available to identify women likely to respond to bevacizumab. Functional tests to predict patient response to any drug are, in fact, essentially absent from clinical care. New drugs are needed to treat ovarian cancer. In this review, we discuss applications to address the currently unmet need of developing physiologically relevant in vitro and ex vivo models of ovarian cancer for fundamental discovery science, and personalized medicine approaches. Traditional two-dimensional (2D) in vitro cell culture of ovarian cancer lacks critical cell-to-cell interactions afforded by culture in three-dimensions. Additionally, modelling interactions with the tumor microenvironment, including the surface of organs in the peritoneal cavity that support metastatic growth of ovarian cancer, will improve the power of these models. Being able to reliably grow primary tumoroid cultures of ovarian cancer will improve the ability to recapitulate tumor heterogeneity. Three-dimensional (3D) modelling systems, from cell lines to organoid or tumoroid cultures, represent enhanced starting points from which improved translational outcomes for women with ovarian cancer will emerge.
Collapse
Affiliation(s)
- Christine Yee
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Kristie-Ann Dickson
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Mohammed N. Muntasir
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Yue Ma
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
| | - Deborah J. Marsh
- Translational Oncology Group, School of Life Sciences, Faculty of Science, University of Technology Sydney, Ultimo, NSW, Australia
- Northern Clinical School, Faculty of Medicine and Health, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|