1
|
Gwon K, Dharmesh E, Nguyen KM, Schornack AMR, de Hoyos-Vega JM, Ceylan H, Stybayeva G, Peterson QP, Revzin A. Designing magnetic microcapsules for cultivation and differentiation of stem cell spheroids. MICROSYSTEMS & NANOENGINEERING 2024; 10:127. [PMID: 39261472 PMCID: PMC11390961 DOI: 10.1038/s41378-024-00747-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 05/03/2024] [Accepted: 06/05/2024] [Indexed: 09/13/2024]
Abstract
Human pluripotent stem cells (hPSCs) represent an excellent cell source for regenerative medicine and tissue engineering applications. However, there remains a need for robust and scalable differentiation of stem cells into functional adult tissues. In this paper, we sought to address this challenge by developing magnetic microcapsules carrying hPSC spheroids. A co-axial flow-focusing microfluidic device was employed to encapsulate stem cells in core-shell microcapsules that also contained iron oxide magnetic nanoparticles (MNPs). These microcapsules exhibited excellent response to an external magnetic field and could be held at a specific location. As a demonstration of utility, magnetic microcapsules were used for differentiating hPSC spheroids as suspension cultures in a stirred bioreactor. Compared to standard suspension cultures, magnetic microcapsules allowed for more efficient media change and produced improved differentiation outcomes. In the future, magnetic microcapsules may enable better and more scalable differentiation of hPSCs into adult cell types and may offer benefits for cell transplantation.
Collapse
Affiliation(s)
- Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
- Department of Biofibers and Biomaterials Science, Kyungpook National University, Daegu, Republic of Korea.
| | - Ether Dharmesh
- Department of Biomedical Engineering, Saint Louis University, St. Louis, MO, USA
| | - Kianna M Nguyen
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | | | - Jose M de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Hakan Ceylan
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Scottsdale, AZ, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Quinn P Peterson
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
2
|
Taroncher M, Gonzalez-Suarez AM, Gwon K, Romero S, Reyes-Figueroa AD, Rodríguez-Carrasco Y, Ruiz MJ, Stybayeva G, Revzin A, de Hoyos-Vega JM. Using Microfluidic Hepatic Spheroid Cultures to Assess Liver Toxicity of T-2 Mycotoxin. Cells 2024; 13:900. [PMID: 38891032 PMCID: PMC11172061 DOI: 10.3390/cells13110900] [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: 04/27/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/20/2024] Open
Abstract
The Fusarium fungi is found in cereals and feedstuffs and may produce mycotoxins, which are secondary metabolites, such as the T-2 toxin (T-2). In this work, we explored the hepatotoxicity of T-2 using microfluidic 3D hepatic cultures. The objectives were: (i) exploring the benefits of microfluidic 3D cultures compared to conventional 3D cultures available commercially (Aggrewell plates), (ii) establishing 3D co-cultures of hepatic cells (HepG2) and stellate cells (LX2) and assessing T-2 exposure in this model, (iii) characterizing the induction of metabolizing enzymes, and (iv) evaluating inflammatory markers upon T-2 exposure in microfluidic hepatic cultures. Our results demonstrated that, in comparison to commercial (large-volume) 3D cultures, spheroids formed faster and were more functional in microfluidic devices. The viability and hepatic function decreased with increasing T-2 concentrations in both monoculture and co-cultures. The RT-PCR analysis revealed that exposure to T-2 upregulates the expression of multiple Phase I and Phase II hepatic enzymes. In addition, several pro- and anti-inflammatory proteins were increased in co-cultures after exposure to T-2.
Collapse
Affiliation(s)
- Mercedes Taroncher
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55901, USA; (M.T.); (A.M.G.-S.); (G.S.)
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Valencia, Spain; (Y.R.-C.); (M.-J.R.)
| | - Alan M. Gonzalez-Suarez
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55901, USA; (M.T.); (A.M.G.-S.); (G.S.)
| | - Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55901, USA; (M.T.); (A.M.G.-S.); (G.S.)
| | - Samuel Romero
- Centro de Investigación en Matemáticas Unidad Monterrey, Apodaca 66628, NL, Mexico (A.D.R.-F.)
| | - Angel D. Reyes-Figueroa
- Centro de Investigación en Matemáticas Unidad Monterrey, Apodaca 66628, NL, Mexico (A.D.R.-F.)
- Consejo Nacional de Humanidades, Ciencias y Tecnologías, Ciudad de Mexico 03940, Mexico
| | - Yelko Rodríguez-Carrasco
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Valencia, Spain; (Y.R.-C.); (M.-J.R.)
| | - María-José Ruiz
- Research Group in Alternative Methods for Determining Toxics Effects and Risk Assessment of Contaminants and Mixtures (RiskTox), Laboratory of Food Chemistry and Toxicology, Faculty of Pharmacy, University of Valencia, Av. Vicent Andrés Estellés s/n, 46100 Valencia, Spain; (Y.R.-C.); (M.-J.R.)
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55901, USA; (M.T.); (A.M.G.-S.); (G.S.)
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55901, USA; (M.T.); (A.M.G.-S.); (G.S.)
| | - Jose M. de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN 55901, USA; (M.T.); (A.M.G.-S.); (G.S.)
| |
Collapse
|
3
|
Kemas AM, Zandi Shafagh R, Taebnia N, Michel M, Preiss L, Hofmann U, Lauschke VM. Compound Absorption in Polymer Devices Impairs the Translatability of Preclinical Safety Assessments. Adv Healthc Mater 2024; 13:e2303561. [PMID: 38053301 PMCID: PMC11469150 DOI: 10.1002/adhm.202303561] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Indexed: 12/07/2023]
Abstract
Organotypic and microphysiological systems (MPS) that can emulate the molecular phenotype and function of human tissues, such as liver, are increasingly used in preclinical drug development. However, despite their improved predictivity, drug development success rates have remained low with most compounds failing in clinical phases despite promising preclinical data. Here, it is tested whether absorption of small molecules to polymers commonly used for MPS fabrication can impact preclinical pharmacological and toxicological assessments and contribute to the high clinical failure rates. To this end, identical devices are fabricated from eight different MPS polymers and absorption of prototypic compounds with different physicochemical properties are analyzed. It is found that overall absorption is primarily driven by compound hydrophobicity and the number of rotatable bonds. However, absorption can differ by >1000-fold between polymers with polydimethyl siloxane (PDMS) being most absorptive, whereas polytetrafluoroethylene (PTFE) and thiol-ene epoxy (TEE) absorbed the least. Strikingly, organotypic primary human liver cultures successfully flagged hydrophobic hepatotoxins in lowly absorbing TEE devices at therapeutically relevant concentrations, whereas isogenic cultures in PDMS devices are resistant, resulting in false negative safety signals. Combined, these results can guide the selection of MPS materials and facilitate the development of preclinical assays with improved translatability.
Collapse
Affiliation(s)
- Aurino M. Kemas
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm17177Sweden
| | - Reza Zandi Shafagh
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm17177Sweden
- Dr. Margarete Fischer‐Bosch Institute of Clinical Pharmacology70376StuttgartGermany
- University of Tuebingen72074TuebingenGermany
- Division of Micro‐ and NanosystemsKTH Royal Institute of TechnologyStockholm10044Sweden
| | - Nayere Taebnia
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm17177Sweden
| | - Maurice Michel
- Department of Oncology and PathologyScience for Life LaboratoryKarolinska InstitutetStockholm17165Sweden
| | - Lena Preiss
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm17177Sweden
- Department of Drug Metabolism and Pharmacokinetics (DMPK)Merck KGaA64293DarmstadtGermany
| | - Ute Hofmann
- Dr. Margarete Fischer‐Bosch Institute of Clinical Pharmacology70376StuttgartGermany
| | - Volker M. Lauschke
- Department of Physiology and PharmacologyKarolinska InstitutetStockholm17177Sweden
- Dr. Margarete Fischer‐Bosch Institute of Clinical Pharmacology70376StuttgartGermany
- University of Tuebingen72074TuebingenGermany
| |
Collapse
|
4
|
Morimoto N, Murata A, Yamamoto Y, Narita F, Yamamoto M. Adhesive Sulfabetaine Polymer Hydrogels for the Sandwich Cell Culture. ACS OMEGA 2024; 9:11942-11949. [PMID: 38496950 PMCID: PMC10938316 DOI: 10.1021/acsomega.3c09708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/17/2024] [Accepted: 02/20/2024] [Indexed: 03/19/2024]
Abstract
Sandwich culture systems are techniques that cultivate cells by sandwiching them between the top and bottom substrates. Since the substrates can be separated, the system is expected to be applied to the construct layering of patterned cells and to the isolation of stacked cells. In this study, we prepared hydrogels composed of zwitterionic sulfabetaine polymers, poly[2-(2-(methacryloyloxyethyl)dimethylammonio)ethyl-1-sulfate] (PZBMA). The ZBMA homopolymers have been shown to form aggregates in aqueous solutions due to their intermolecular interactions. The water content of the PZBMA hydrogels in water was ∼70% regardless of N,N'-methylenebis(acrylamide), BIS, content as the cross-linker. The results indicated that the intermolecular interaction contributed more to the swelling behaviors than the chemical cross-linker. However, PZBMA hydrogels with 0.1 mol % BIS showed not only high elongation (∼850%) properties but also high adhesiveness and self-healing properties. When this PZBMA hydrogel was impregnated with collagen and subjected to sandwich culture using Madin-Darby canine kidney (MDCK) cells, a three-dimensional morphology of MDCK cell aggregates was constructed. Such a sulfabetaine hydrogel is expected to be developed for regenerative medicine.
Collapse
Affiliation(s)
- Nobuyuki Morimoto
- Department
of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02, Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
- Department
of Materials for Energy, Shimane University, 1060 Nishikawatsu, Matsue, Shimane 690-8504, Japan
| | - Atsuki Murata
- Department
of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02, Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | - Yuta Yamamoto
- Department
of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02, Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
| | - Fumio Narita
- Department
of Frontier Sciences for Advanced Environment, Graduate School of
Environmental Studies, Tohoku University, Sendai 980-8579, Japan
| | - Masaya Yamamoto
- Department
of Materials Processing, Graduate School of Engineering, Tohoku University, 6-6-02, Aramaki-aza Aoba, Aoba-ku, Sendai, Miyagi 980-8579, Japan
- Graduate
School of Biomedical Engineering,Tohoku
University, Sendai 980-8579, Japan
| |
Collapse
|
5
|
Badwaik HR, Sakure K, Giri TK. Hydrogels based on heparin and its conjugates. POLYSACCHARIDE HYDROGELS FOR DRUG DELIVERY AND REGENERATIVE MEDICINE 2024:69-87. [DOI: 10.1016/b978-0-323-95351-1.00013-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
6
|
Gwon K, Choi D, de Hoyos-Vega JM, Baskaran H, Gonzalez-Suarez AM, Lee S, Hong HJ, Nguyen KM, Dharmesh E, Sugahara G, Ishida Y, Saito T, Stybayeva G, Revzin A. Function of hepatocyte spheroids in bioactive microcapsules is enhanced by endogenous and exogenous hepatocyte growth factor. Bioact Mater 2023; 28:183-195. [PMID: 37266448 PMCID: PMC10230170 DOI: 10.1016/j.bioactmat.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 06/03/2023] Open
Abstract
The ability to maintain functional hepatocytes has important implications for bioartificial liver development, cell-based therapies, drug screening, and tissue engineering. Several approaches can be used to restore hepatocyte function in vitro, including coating a culture substrate with extracellular matrix (ECM), encapsulating cells within biomimetic gels (Collagen- or Matrigel-based), or co-cultivation with other cells. This paper describes the use of bioactive heparin-based core-shell microcapsules to form and cultivate hepatocyte spheroids. These microcapsules are comprised of an aqueous core that facilitates hepatocyte aggregation into spheroids and a heparin hydrogel shell that binds and releases growth factors. We demonstrate that bioactive microcapsules retain and release endogenous signals thus enhancing the function of encapsulated hepatocytes. We also demonstrate that hepatic function may be further enhanced by loading exogenous hepatocyte growth factor (HGF) into microcapsules and inhibiting transforming growth factor (TGF)-β1 signaling. Overall, bioactive microcapsules described here represent a promising new strategy for the encapsulation and maintenance of primary hepatocytes and will be beneficial for liver tissue engineering, regenerative medicine, and drug testing applications.
Collapse
Affiliation(s)
- Kihak Gwon
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Daheui Choi
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - José M. de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Harihara Baskaran
- Department of Chemical and Biomolecular Engineering, Case Western Reserve University, Cleveland, OH, USA
| | | | - Seonhwa Lee
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kianna M. Nguyen
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Ether Dharmesh
- Biomedical Engineering, Saint Louis University, St. Louis, MO, USA
| | - Go Sugahara
- University of Southern California, Keck School of Medicine, Department of Medicine, Division of Gastrointestinal and Liver Diseases, Los Angeles, CA, USA
- Research and Development Department, PhoenixBio, Co., Ltd, Kagamiyama, Higashi-Hiroshima, Hiroshima, Japan
| | - Yuji Ishida
- University of Southern California, Keck School of Medicine, Department of Medicine, Division of Gastrointestinal and Liver Diseases, Los Angeles, CA, USA
- Research and Development Department, PhoenixBio, Co., Ltd, Kagamiyama, Higashi-Hiroshima, Hiroshima, Japan
| | - Takeshi Saito
- University of Southern California, Keck School of Medicine, Department of Medicine, Division of Gastrointestinal and Liver Diseases, Los Angeles, CA, USA
- USC Research Center for Liver Diseases, Los Angeles, CA, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
7
|
Juraski AC, Sharma S, Sparanese S, da Silva VA, Wong J, Laksman Z, Flannigan R, Rohani L, Willerth SM. 3D bioprinting for organ and organoid models and disease modeling. Expert Opin Drug Discov 2023; 18:1043-1059. [PMID: 37431937 DOI: 10.1080/17460441.2023.2234280] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023]
Abstract
INTRODUCTION 3D printing, a versatile additive manufacturing technique, has diverse applications ranging from transportation, rapid prototyping, clean energy, and medical devices. AREAS COVERED The authors focus on how 3D printing technology can enhance the drug discovery process through automating tissue production that enables high-throughput screening of potential drug candidates. They also discuss how the 3D bioprinting process works and what considerations to address when using this technology to generate cell laden constructs for drug screening as well as the outputs from such assays necessary for determining the efficacy of potential drug candidates. They focus on how bioprinting how has been used to generate cardiac, neural, and testis tissue models, focusing on bio-printed 3D organoids. EXPERT OPINION The next generation of 3D bioprinted organ model holds great promises for the field of medicine. In terms of drug discovery, the incorporation of smart cell culture systems and biosensors into 3D bioprinted models could provide highly detailed and functional organ models for drug screening. By addressing current challenges of vascularization, electrophysiological control, and scalability, researchers can obtain more reliable and accurate data for drug development, reducing the risk of drug failures during clinical trials.
Collapse
Affiliation(s)
- Amanda C Juraski
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
- Department of Chemical Engineering, Polytechnic School, University of Sao Paulo, Sao Paulo, Brazil
| | - Sonali Sharma
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Sydney Sparanese
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Victor A da Silva
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
| | - Julie Wong
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Zachary Laksman
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Ryan Flannigan
- Department of Urologic Sciences, University of British Columbia, Vancouver BC, Canada
| | - Leili Rohani
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| | - Stephanie M Willerth
- Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria BC, Canada
- Faculty of Medicine, School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| |
Collapse
|
8
|
Vega JMDH, Hong HJ, Loutherback K, Stybayeva G, Revzin A. A Microfluidic Device for Long-Term Maintenance of Organotypic Liver Cultures. ADVANCED MATERIALS TECHNOLOGIES 2023; 8:2201121. [PMID: 36818276 PMCID: PMC9937715 DOI: 10.1002/admt.202201121] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Indexed: 06/03/2023]
Abstract
Liver cultures may be used for disease modeling, testing therapies and predicting drug-induced injury. The complexity of the liver cultures has evolved from hepatocyte monocultures to co-cultures with non-parenchymal cells and finally to precision-cut liver slices. The latter culture format retains liver's native biomolecular and cellular complexity and therefore holds considerable promise for in vitro testing. However, liver slices remain functional for ~72 h in vitro and display limited utility for some disease modeling and therapy testing applications that require longer culture times. This paper describes a microfluidic device for longer-term maintenance of functional organotypic liver cultures. Our microfluidic culture system was designed to enable direct injection of liver tissue into a culture chamber through a valve-enabled side port. Liver tissue was embedded in collagen and remained functional for up to 31 days, highlighted by continued production of albumin and urea. These organotypic cultures also expressed several enzymes involved in xenobiotic metabolism. Conversely, matched liver tissue embedded in collagen in a 96-well plate lost its phenotype and function within 3-5 days. The microfluidic organotypic liver cultures described here represent a significant advance in liver cultivation and may be used for future modeling of liver diseases or for individualized liver-directed therapies.
Collapse
Affiliation(s)
- José M. de Hoyos Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kevin Loutherback
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
9
|
Kaur I, Vasudevan A, Rawal P, Tripathi DM, Ramakrishna S, Kaur S, Sarin SK. Primary Hepatocyte Isolation and Cultures: Technical Aspects, Challenges and Advancements. Bioengineering (Basel) 2023; 10:131. [PMID: 36829625 PMCID: PMC9952008 DOI: 10.3390/bioengineering10020131] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/20/2023] Open
Abstract
Hepatocytes are differentiated cells that account for 80% of the hepatic volume and perform all major functions of the liver. In vivo, after an acute insult, adult hepatocytes retain their ability to proliferate and participate in liver regeneration. However, in vitro, prolonged culture and proliferation of viable and functional primary hepatocytes have remained the major and the most challenging goal of hepatocyte-based cell therapies and liver tissue engineering. The first functional cultures of rat primary hepatocytes between two layers of collagen gel, also termed as the "sandwich cultures", were reported in 1989. Since this study, several technical developments including choice of hydrogels, type of microenvironment, growth factors and culture conditions, mono or co-cultures of hepatocytes along with other supporting cell types have evolved for both rat and human primary hepatocytes in recent years. All these improvements have led to a substantial improvement in the number, life-span and hepatic functions of these cells in vitro for several downstream applications. In the current review, we highlight the details, limitations and prospects of different technical strategies being used in primary hepatocyte cultures. We discuss the use of newer biomaterials as scaffolds for efficient culture of primary hepatocytes. We also describe the derivation of mature hepatocytes from other cellular sources such as induced pluripotent stem cells, bone marrow stem cells and 3D liver organoids. Finally, we also explain the use of perfusion-based bioreactor systems and bioengineering strategies to support the long-term function of hepatocytes in 3D conditions.
Collapse
Affiliation(s)
- Impreet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Preety Rawal
- School of Biotechnology, Gautam Buddha University, Greater Noida 201312, India
| | - Dinesh M. Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Seeram Ramakrishna
- Department of Mechanical Engineering, National University of Singapore, Singapore 117581, Singapore
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| | - Shiv K. Sarin
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi 110070, India
| |
Collapse
|
10
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
11
|
de Hoyos-Vega JM, Hong HJ, Stybayeva G, Revzin A. Hepatocyte cultures: From collagen gel sandwiches to microfluidic devices with integrated biosensors. APL Bioeng 2021; 5:041504. [PMID: 34703968 PMCID: PMC8519630 DOI: 10.1063/5.0058798] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 09/21/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocytes are parenchymal cells of the liver responsible for drug detoxification, urea and bile production, serum protein synthesis, and glucose homeostasis. Hepatocytes are widely used for drug toxicity studies in bioartificial liver devices and for cell-based liver therapies. Because hepatocytes are highly differentiated cells residing in a complex microenvironment in vivo, they tend to lose hepatic phenotype and function in vitro. This paper first reviews traditional culture approaches used to rescue hepatic function in vitro and then discusses the benefits of emerging microfluidic-based culture approaches. We conclude by reviewing integration of hepatocyte cultures with bioanalytical or sensing approaches.
Collapse
Affiliation(s)
- Jose M. de Hoyos-Vega
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Hye Jin Hong
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Gulnaz Stybayeva
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| | - Alexander Revzin
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota 55902, USA
| |
Collapse
|
12
|
Stem Cells and Hydrogels for Liver Tissue Engineering: Synergistic Cure for Liver Regeneration. Stem Cell Rev Rep 2020; 16:1092-1104. [DOI: 10.1007/s12015-020-10060-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/12/2020] [Indexed: 02/06/2023]
|
13
|
Sengupta S, Johnson B, Seirup M, Ardalani H, Duffin B, Barrett-Wilt GA, Stewart R, Thomson JA. Co-culture with mouse embryonic fibroblasts improves maintenance of metabolic function of human small hepatocyte progenitor cells. Curr Res Toxicol 2020; 1:70-84. [PMID: 34345838 PMCID: PMC8320630 DOI: 10.1016/j.crtox.2020.08.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
Derivation and culture of small hepatocyte progenitor cells (SHPCs) capable of proliferating in vitro has been described in rodents and recently in humans. These cells are capable of engrafting in injured livers, however, they display de-differentiated morphology and reduced xenobiotic metabolism activity in culture over passages. Here we report that SHPCs derived from adult primary human hepatocytes (PHHs) and cultured on mouse embryonic fibroblasts (MEFs) not only display differentiated morphology and exhibit gene expression profiles similar to adult PHHs, but importantly, they retain their phenotype over several passages. Further, unlike previous reports, where extensive manipulations of culture conditions are required to convert SHPCs to metabolically functional hepatocytes, SHPCs in our co-culture system maintain expression of xenobiotic metabolism-associated genes. We show that SHPCs in co-culture are able to perform xenobiotic metabolism at rates equal to their parent PHHs as evidenced by the metabolism of acetaminophen to all of its major metabolites. In summary, we present an improved co-culture system that allows generation of SHPCs from adult PHHs that maintain their differentiated phenotype over multiple passages. Our findings would be useful for expansion of limited PHHs for use in studies of drug metabolism and toxicity testing.
Collapse
Affiliation(s)
- Srikumar Sengupta
- Morgridge Institute for Research, Madison, WI, United States of America
| | - Brian Johnson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, United States of America.,Institute for Quantitative Health Science and Engineering, Departments of Pharmacology & Toxicology and Biomedical Engineering, Michigan State University, East Lansing, MI, United States of America
| | - Morten Seirup
- Morgridge Institute for Research, Madison, WI, United States of America.,Dianomi Therapeutics, Madison, WI, United States of America
| | - Hamisha Ardalani
- Morgridge Institute for Research, Madison, WI, United States of America.,Beckman Coulter Life Sciences, San Jose, CA, United States of America
| | - Bret Duffin
- Morgridge Institute for Research, Madison, WI, United States of America
| | - Gregory A Barrett-Wilt
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI, United States of America
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI, United States of America
| | - James A Thomson
- Morgridge Institute for Research, Madison, WI, United States of America.,Department of Cell & Regenerative Biology, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States of America.,Department of Molecular, Cellular, & Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, United States of America
| |
Collapse
|
14
|
Menon N, Dang HX, Datla US, Moarefian M, Lawrence CB, Maher CA, Jones CN. Heparin-based hydrogel scaffolding alters the transcriptomic profile and increases the chemoresistance of MDA-MB-231 triple-negative breast cancer cells. Biomater Sci 2020; 8:2786-2796. [PMID: 32091043 PMCID: PMC7497406 DOI: 10.1039/c9bm01481k] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Accepted: 02/08/2020] [Indexed: 12/30/2022]
Abstract
The tumor microenvironment plays a critical role in the proliferation and chemoresistance of cancer cells. Growth factors (GFs) are known to interact with the extracellular matrix (ECM) via heparin binding sites, and these associations influence cell behavior. In the present study, we demonstrate the ability to define signals presented by the scaffold by pre-mixing growth factors, such as epidermal growth factor, into the heparin-based (HP-B) hydrogel prior to gelation. In the 3D biomimetic microenvironment, breast cancer cells formed spheroids within 24 hours of initial seeding. Despite higher number of proliferating cells in 2D cultures, 3D spheroids exhibited a higher degree of chemoresistance after 72 hours. Further, our RNA sequencing results highlighted the phenotypic changes influenced by solid-phase GF presentation. Wnt/β-catenin and TGF-β signaling were upregulated in the cells grown in the hydrogel, while apoptosis, IL2-STAT5 and PI3K-AKT-mTOR signaling were downregulated. With emerging technologies for precision medicine in cancer, this nature of fine-tuning the microenvironment is paramount for cultivation and downstream characterization of primary cancer cells and rare circulating tumor cells (CTCs), and effective screening of chemotherapeutic agents.
Collapse
Affiliation(s)
- Nidhi Menon
- Graduate Program in Translational Biology
, Medicine and Health
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
.
- Department of Biological Sciences
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| | - Ha X. Dang
- McDonnell Genome Institute
, Washington University in St. Louis
,
MO
63108
, USA
- Department of Medicine
, Washington University School of Medicine
,
St. Louis
, MO
63108
, USA
- Alvin J. Siteman Cancer Center
, Washington University in St. Louis
,
St. Louis
, MO
63108
, USA
| | - Udaya Sree Datla
- Graduate Program in Translational Biology
, Medicine and Health
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
.
- Department of Biological Sciences
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| | - Maryam Moarefian
- Department of Mechanical Engineering
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| | - Christopher B. Lawrence
- Department of Biological Sciences
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| | - Christopher A. Maher
- McDonnell Genome Institute
, Washington University in St. Louis
,
MO
63108
, USA
- Department of Medicine
, Washington University School of Medicine
,
St. Louis
, MO
63108
, USA
- Alvin J. Siteman Cancer Center
, Washington University in St. Louis
,
St. Louis
, MO
63108
, USA
- Department of Biomedical Engineering
, Washington University in St. Louis
,
MO
63108
, USA
| | - Caroline N. Jones
- Graduate Program in Translational Biology
, Medicine and Health
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
.
- Department of Biological Sciences
, Virginia Polytechnic Institute and State University
,
Blacksburg
, VA
24061
, USA
| |
Collapse
|
15
|
Li J, Wu C, Chu PK, Gelinsky M. 3D printing of hydrogels: Rational design strategies and emerging biomedical applications. MATERIALS SCIENCE AND ENGINEERING: R: REPORTS 2020; 140:100543. [DOI: 10.1016/j.mser.2020.100543] [Citation(s) in RCA: 370] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
16
|
Wei J, Lu J, Chen M, Xie S, Wang T, Li X. 3D spheroids generated on carbon nanotube-functionalized fibrous scaffolds for drug metabolism and toxicity screening. Biomater Sci 2019; 8:426-437. [PMID: 31746843 DOI: 10.1039/c9bm01310e] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The mechanical and electrical stimuli have a profound effect on the cellular behavior and function. In this study, a series of conductive nanofibrous scaffolds are developed by blend electrospinning of poly(styrene-co-maleic acid) (PSMA) and multiwalled-carbon nanotubes (CNTs), followed by grafting galactose as cell adhesion cues. When the mass ratios of CNTs to PSMA increase up to 5%, the alignment, Young's modulus and conductivity of fibrous scaffolds increase, whereas the average diameter, pore size and elongation at break decrease. Primary hepatocytes cultured on the scaffolds are self-assembled into 3D spheroids, which restores the hepatocyte polarity and sufficient expression of drug metabolism enzymes over an extended period of time. Among these conductive scaffolds, hepatocytes cultured on fibers containing 3% of CNTs (F3) show the highest clearance rates of model drugs, offering a better prediction of the in vivo data with a high correlation value. Moreover, the drug metabolism capability is maintained over 15 days and is more sensitive towards the inducers and inhibitors of metabolizing enzymes, demonstrating the applicability for drug-drug interaction studies. Thus, this culture system has been demonstrated as a reliable in vitro model for high-throughput screening of metabolism and toxicity in the early phases of drug development.
Collapse
Affiliation(s)
- Jiaojun Wei
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P.R. China.
| | | | | | | | | | | |
Collapse
|
17
|
Smithmyer ME, Cassel SE, Kloxin AM. Bridging 2D and 3D culture: probing impact of extracellular environment on fibroblast activation in layered hydrogels. AIChE J 2019; 65. [PMID: 32921797 DOI: 10.1002/aic.16837] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Many cell behaviors are significantly affected by cell culture geometry, though it remains unclear which geometry from two- to three-dimensional (2D to 3D) culture is appropriate for probing a specific cell function and mimicking native microenvironments. Toward addressing this, we established a 2.5D culture geometry, enabling initial cell spreading while reducing polarization to bridge between 2D and 3D geometries, and examined the responses of wound healing cells, human pulmonary fibroblasts, within it. To achieve this, we used engineered biomimetic hydrogels formed by photopolymerization, creating robust layered hydrogels with spread fibroblasts at the interface. We found that fibroblast responses were similar between 2D and 2.5D culture and different from 3D culture, with some underlying differences in mechanotransduction. These studies established the 2.5D cell culture geometry in conjunction with biomimetic synthetic matrices as a useful tool for investigations of fibroblast activation with relevance to the study of other cell functions and types.
Collapse
Affiliation(s)
- Megan E Smithmyer
- Chemical and Biomolecular Engineering, University of Delaware, Newark DE, 19716
| | - Samantha E Cassel
- Chemical and Biomolecular Engineering, University of Delaware, Newark DE, 19716
| | - April M Kloxin
- Chemical and Biomolecular Engineering, University of Delaware, Newark DE, 19716
- Materials Science and Engineering, University of Delaware, Newark DE. 19716
| |
Collapse
|
18
|
Leichner C, Jelkmann M, Bernkop-Schnürch A. Thiolated polymers: Bioinspired polymers utilizing one of the most important bridging structures in nature. Adv Drug Deliv Rev 2019; 151-152:191-221. [PMID: 31028759 DOI: 10.1016/j.addr.2019.04.007] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 04/16/2019] [Accepted: 04/16/2019] [Indexed: 12/13/2022]
Abstract
Thiolated polymers designated "thiomers" are obtained by covalent attachment of thiol functionalities on the polymeric backbone of polymers. In 1998 these polymers were first described as mucoadhesive and in situ gelling compounds forming disulfide bonds with cysteine-rich substructures of mucus glycoproteins and crosslinking through inter- and intrachain disulfide bond formation. In the following, it was shown that thiomers are able to form disulfides with keratins and membrane-associated proteins exhibiting also cysteine-rich substructures. Furthermore, permeation enhancing, enzyme inhibiting and efflux pump inhibiting properties were demonstrated. Because of these capabilities thiomers are promising tools for drug delivery guaranteeing a strongly prolonged residence time as well as sustained release on mucosal membranes. Apart from that, thiomers are used as drugs per se. In particular, for treatment of dry eye syndrome various thiolated polymers are in development and a first product has already reached the market. Within this review an overview about the thiomer-technology and its potential for different applications is provided discussing especially the outcome of studies in non-rodent animal models and that of numerous clinical trials. Moreover, an overview on product developments is given.
Collapse
|
19
|
Kiamehr M, Heiskanen L, Laufer T, Düsterloh A, Kahraman M, Käkelä R, Laaksonen R, Aalto-Setälä K. Dedifferentiation of Primary Hepatocytes is Accompanied with Reorganization of Lipid Metabolism Indicated by Altered Molecular Lipid and miRNA Profiles. Int J Mol Sci 2019; 20:ijms20122910. [PMID: 31207892 PMCID: PMC6627955 DOI: 10.3390/ijms20122910] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/10/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Aim: Primary human hepatocytes (PHHs) undergo dedifferentiation upon the two-dimensional (2D) culture, which particularly hinders their utility in long-term in vitro studies. Lipids, as a major class of biomolecules, play crucial roles in cellular energy storage, structure, and signaling. Here, for the first time, we mapped the alterations in the lipid profile of the dedifferentiating PHHs and studied the possible role of lipids in the loss of the phenotype of PHHs. Simultaneously, differentially expressed miRNAs associated with changes in the lipids and fatty acids (FAs) of the dedifferentiating PHHs were investigated. Methods: PHHs were cultured in monolayer and their phenotype was monitored morphologically, genetically, and biochemically for five days. The lipid and miRNA profile of the PHHs were analyzed by mass spectrometry and Agilent microarray, respectively. In addition, 24 key genes involved in the metabolism of lipids and FAs were investigated by qPCR. Results: The typical morphology of PHHs was lost from day 3 onward. Additionally, ALB and CYP genes were downregulated in the cultured PHHs. Lipidomics revealed a clear increase in the saturated fatty acids (SFA) and monounsaturated fatty acids (MUFA) containing lipids, but a decrease in the polyunsaturated fatty acids (PUFA) containing lipids during the dedifferentiation of PHHs. In line with this, FASN, SCD, ELOVL1, ELOVL3, and ELOVL7 were upregulated but ELOVL2 was downregulated in the dedifferentiated PHHs. Furthermore, differentially expressed miRNAs were identified, and the constantly upregulated miR-27a and miR-21, and downregulated miR-30 may have regulated the synthesis, accumulation and secretion of PHH lipids during the dedifferentiation. Conclusion: Our results showed major alterations in the molecular lipid species profiles, lipid-metabolizing enzyme expression as wells as miRNA profiles of the PHHs during their prolonged culture, which in concert could play important roles in the PHHs’ loss of phenotype. These findings promote the understanding from the dedifferentiation process and could help in developing optimal culture conditions, which better meet the needs of the PHHs and support their original phenotype.
Collapse
Affiliation(s)
- Mostafa Kiamehr
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland.
| | | | - Thomas Laufer
- Hummingbird Diagnostics GmbH, 69120 Heidelberg, Germany.
- Department of Human Genetics, Saarland University, 66421 Homburg, Germany.
| | | | - Mustafa Kahraman
- Hummingbird Diagnostics GmbH, 69120 Heidelberg, Germany.
- Clinical Bioinformatics, Saarland University, 66123 Saarbrücken, Germany.
| | - Reijo Käkelä
- Helsinki University Lipidomics Unit, Helsinki Institute for Life Science (HiLIFE) and Molecular and Integrative Biosciences Research Programme, University of Helsinki, FI-00014 Helsinki, Finland.
| | - Reijo Laaksonen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland.
- Zora Biosciences, 02150 Espoo, Finland.
| | - Katriina Aalto-Setälä
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland.
- Heart Hospital, Tampere University Hospital, 33520 Tampere, Finland.
| |
Collapse
|
20
|
He C, Ji H, Qian Y, Wang Q, Liu X, Zhao W, Zhao C. Heparin-based and heparin-inspired hydrogels: size-effect, gelation and biomedical applications. J Mater Chem B 2019; 7:1186-1208. [PMID: 32255159 DOI: 10.1039/c8tb02671h] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Heparin is the highest negatively charged biomolecule, which is a polysaccharide belonging to the glycosaminoglycan family, and its role as a regulator of various proteins, cells and tissues in the human body makes it an indispensable macromolecule. Heparin-based hydrogels are widely investigated in various applications including implantation, tissue engineering, biosensors, and drug-controlled release due to the 3D-constructs of hydrogels. However, heparin has supply and safety problems because it is usually derived from animal sources, and has the clinical limitations of bleeding and thrombocytopenia. Therefore, analogous heparin-mimicking polymers and hydrogels derived from non-animal and/or totally synthetic sources have been widely studied in recent years. In this review, the progress and potential biomedical applications of heparin-based and heparin-inspired hydrogels are highlighted. We classify the forms of these hydrogels by their size including macro-hydrogels, injectable hydrogels, and nano-hydrogels. Then, we summarize the various fabrication strategies for these hydrogels including chemical covalent bonding, physical conjugation, and the combination of chemical and physical interactions. Covalent bonding includes free radical polymerization of vinyl-containing components, amide bond formation reaction, Michael-type addition reaction, click-chemistry, divinyl sulfone crosslinking, and mussel-inspired coating. Hydrogels physically conjugated via host-guest interaction, electrostatic interaction, hydrogen bonding, and hydrophobic interaction are also discussed. Finally, we conclude with the challenges and future directions for the fabrication and the industrialization of heparin-based and heparin-inspired hydrogels. We believe that this review will attract more attention toward the design of heparin-based and heparin-inspired hydrogels, leading to future advancements in this emerging research field.
Collapse
Affiliation(s)
- Chao He
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, China.
| | | | | | | | | | | | | |
Collapse
|
21
|
Wang JZ, Xiong NY, Zhao LZ, Hu JT, Kong DC, Yuan JY. Review fantastic medical implications of 3D-printing in liver surgeries, liver regeneration, liver transplantation and drug hepatotoxicity testing: A review. Int J Surg 2018; 56:1-6. [PMID: 29886280 DOI: 10.1016/j.ijsu.2018.06.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 06/05/2018] [Indexed: 02/07/2023]
Abstract
The epidemiological trend in liver diseases becomes more serious worldwide. Several recent articles published by International Journal of Surgery in 2018 particularly emphasized the encouraging clinical benefits of hepatectomy, liver regeneration and liver transplantation, however, there are still many technical bottlenecks underlying these therapeutic approaches. Remarkably, a few preliminary studies have shown some clues to the role of three-dimensional (3D) printing in improving traditional therapy for liver diseases. Here, we concisely elucidated the curative applications of 3D-printing (no cells) and 3D Bio-printing (with hepatic cells), such as 3D-printed patient-specific liver models and devices for medical education, surgical simulation, hepatectomy and liver transplantation, 3D Bio-printed hepatic constructs for liver regeneration and artificial liver, 3D-printed liver tissues for evaluating drug's hepatotoxicity, and so on. Briefly, 3D-printed liver models and bioactive tissues may facilitate a lot of key steps to cure liver disorders, predictably bringing promising clinical benefits. This work further provides novel insights into facilitating treatment of hepatic carcinoma, promoting liver regeneration both in vivo and in vitro, expanding transplantable liver resources, maximizing therapeutic efficacy as well as minimizing surgical complications, medical hepatotoxicity, operational time, economic costs, etc.
Collapse
Affiliation(s)
- Jing-Zhang Wang
- Department of Medical Technology, College of Medicine, Affiliated Hospital, Hebei University of Engineering, Handan, 056002, PR China.
| | - Nan-Yan Xiong
- College of Medicine, Hebei University of Engineering, Handan, 056002, PR China
| | - Li-Zhen Zhao
- Department of Clinical Laboratory, Affiliated Hospital of Hebei University of Engineering, Handan, 056002, PR China
| | - Jin-Tian Hu
- Department of Clinical Laboratory, Affiliated Hospital of Hebei University of Engineering, Handan, 056002, PR China
| | - De-Cheng Kong
- College of Medicine, Hebei University of Engineering, Handan, 056002, PR China
| | - Jiang-Yong Yuan
- Department of Cardiology, Affiliated Hospital of Hebei University of Engineering, Handan, 056002, PR China.
| |
Collapse
|
22
|
Brown JH, Das P, DiVito MD, Ivancic D, Tan LP, Wertheim JA. Nanofibrous PLGA electrospun scaffolds modified with type I collagen influence hepatocyte function and support viability in vitro. Acta Biomater 2018; 73:217-227. [PMID: 29454157 PMCID: PMC5985221 DOI: 10.1016/j.actbio.2018.02.009] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 01/23/2018] [Accepted: 02/08/2018] [Indexed: 02/06/2023]
Abstract
A major challenge of maintaining primary hepatocytes in vitro is progressive loss of hepatocyte-specific functions, such as protein synthesis and cytochrome P450 (CYP450) catalytic activity. We developed a three-dimensional (3D) nanofibrous scaffold made from poly(l-lactide-co-glycolide) (PLGA) polymer using a newly optimized wet electrospinning technique that resulted in a highly porous structure that accommodated inclusion of primary human hepatocytes. Extracellular matrix (ECM) proteins (type I collagen or fibronectin) at varying concentrations were chemically linked to electrospun PLGA using amine coupling to develop an in vitro culture system containing the minimal essential ECM components of the liver micro-environment that preserve hepatocyte function in vitro. Cell-laden nanofiber scaffolds were tested in vitro to maintain hepatocyte function over a two-week period. Incorporation of type I collagen onto PLGA scaffolds (PLGA-Chigh: 100 µg/mL) led to 10-fold greater albumin secretion, 4-fold higher urea synthesis, and elevated transcription of hepatocyte-specific CYP450 genes (CYP3A4, 3.5-fold increase and CYP2C9, 3-fold increase) in primary human hepatocytes compared to the same cells grown within unmodified PLGA scaffolds over two weeks. These indices, measured using collagen-bonded scaffolds, were also higher than scaffolds coupled to fibronectin or an ECM control sandwich culture composed of type I collagen and Matrigel. Induction of CYP2C9 activity was also higher in these same type I collagen PLGA scaffolds compared to other ECM-modified or unmodified PLGA constructs and was equivalent to the ECM control at 7 days. Together, we demonstrate a minimalist ECM-based 3D synthetic scaffold that accommodates primary human hepatocyte inclusion into the matrix, maintains long-term in vitro survival and stimulates function, which can be attributed to coupling of type I collagen. STATEMENT OF SIGNIFICANCE Culturing primary hepatocytes within a three-dimensional (3D) structure that mimics the natural liver environment is a promising strategy for extending the function and viability of hepatocytes in vitro. In the present study we generate porous PLGA nanofibers, that are chemically modified with extracellular matrix proteins, to serve as 3D scaffolds for the in vitro culture of primary human hepatocytes. Our findings demonstrate that the use of ECM proteins, especially type I collagen, in a porous 3D environment helps to improve the synthetic function of primary hepatocytes over time. We believe the work presented within will provide insights to readers for drug toxicity and tissue engineering applications.
Collapse
Affiliation(s)
- Jessica H Brown
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Prativa Das
- Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Michael D DiVito
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - David Ivancic
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Lay Poh Tan
- Interdisciplinary Graduate School, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore; School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798 Singapore..
| | - Jason A Wertheim
- Comprehensive Transplant Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Department of Surgery, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States; Chemistry of Life Processes Institute, Northwestern University, Evanston, IL 60208, United States; Department of Surgery, Jesse Brown VA Medical Center, Chicago, IL 60612, United States; Simpson Querrey Institute for BioNanotechnology in Medicine, Northwestern University, Chicago, IL 60611, United States; Department of Biomedical Engineering, Northwestern University, Evanston, IL 60208, United States.
| |
Collapse
|
23
|
Ouyang L, Burdick JA, Sun W. Facile Biofabrication of Heterogeneous Multilayer Tubular Hydrogels by Fast Diffusion-Induced Gelation. ACS APPLIED MATERIALS & INTERFACES 2018; 10:12424-12430. [PMID: 29582989 DOI: 10.1021/acsami.7b19537] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Multilayer (ML) hydrogels are useful to achieve stepwise and heterogeneous control over the organization of biomedical materials and cells. There are numerous challenges in the development of fabrication approaches toward this, including the need for mild processing conditions that maintain the integrity of embedded compounds and the versatility in processing to introduce desired complexity. Here, we report a method to fabricate heterogeneous multilayered hydrogels based on diffusion-induced gelation. This technique uses the quick diffusion of ions and small molecules (i.e., photoinitiators) through gel-sol or gel-gel interfaces to produce hydrogel layers. Specifically, ionically (e.g., alginate-based) and covalently [e.g., gelatin methacryloyl (GelMA-based)] photocross-linked hydrogels are generated in converse directions from the same interface. The ML (e.g., seven layers) ionic hydrogels can be formed within seconds to minutes with thicknesses ranging from tens to hundreds of micrometers. The thicknesses of the covalent hydrogels are determined by the reaction time (or the molecule diffusion time). Multiwalled tubular structures (e.g., mimicking branched multiwalled vessels) are mainly investigated in this study based on a removable gel core, but this method can be generalized to other material patterns. The process is also demonstrated to support the encapsulation of viable cells and is compatible with a range of thermally reversible core materials (e.g., gelatin and Pluronic F127) and covalently cross-linked formulations (e.g., GelMA and methacrylated hyaluronic acid). This biofabrication process enhances our ability to fabricate a range of structures that are useful for biomedical applications.
Collapse
Affiliation(s)
- Liliang Ouyang
- Department of Mechanical Engineering , Tsinghua University , Beijing 100084 , China
| | - Jason A Burdick
- Department of Bioengineering , University of Pennsylvania , Philadelphia , Pennsylvania 19104 , United States
| | - Wei Sun
- Department of Mechanical Engineering , Tsinghua University , Beijing 100084 , China
- Department of Mechanical Engineering and Mechanics , Drexel University , Philadelphia , Pennsylvania 19104 , United States
| |
Collapse
|
24
|
Liaw CY, Ji S, Guvendiren M. Engineering 3D Hydrogels for Personalized In Vitro Human Tissue Models. Adv Healthc Mater 2018; 7. [PMID: 29345429 DOI: 10.1002/adhm.201701165] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Revised: 11/13/2017] [Indexed: 01/17/2023]
Abstract
There is a growing interest in engineering hydrogels for 3D tissue and disease models. The major motivation is to better mimic the physiological microenvironment of the disease and human condition. 3D tissue models derived from patients' own cells can potentially revolutionize the way treatment and diagnostic alternatives are developed. This requires development of tissue mimetic hydrogels with user defined and tunable properties. In this review article, a recent summary of 3D hydrogel platforms for in vitro tissue and disease modeling is given. Hydrogel design considerations and available hydrogel systems are summarized, followed by the types of currently available hydrogel models, such as bulk hydrogels, porous scaffolds, fibrous scaffolds, hydrogel microspheres, hydrogel sandwich systems, microwells, and 3D bioprinted constructs. Although hydrogels are utilized for a wide range of tissue models, this article focuses on liver and cancer models. This article also provides a detailed section on current challenges and future perspectives of hydrogel-based tissue models.
Collapse
Affiliation(s)
- Chya-Yan Liaw
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| | - Shen Ji
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| | - Murat Guvendiren
- Instructive Biomaterials and Additive Manufacturing Laboratory; Otto H. York Chemical; Biological and Pharmaceutical Engineering; Newark College of Engineering; New Jersey Institute of Technology; University Heights; 138 York Center Newark NJ 07102 USA
| |
Collapse
|
25
|
Siltanen C, Diakatou M, Lowen J, Haque A, Rahimian A, Stybayeva G, Revzin A. One step fabrication of hydrogel microcapsules with hollow core for assembly and cultivation of hepatocyte spheroids. Acta Biomater 2017; 50:428-436. [PMID: 28069506 DOI: 10.1016/j.actbio.2017.01.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/15/2016] [Accepted: 01/05/2017] [Indexed: 12/16/2022]
Abstract
3D hepatic microtissues can serve as valuable liver analogues for cell-based therapies and for hepatotoxicity screening during preclinical drug development. However, hepatocytes rapidly dedifferentiate in vitro, and typically require 3D culture systems or co-cultures for phenotype rescue. In this work we present a novel microencapsulation strategy, utilizing coaxial flow-focusing droplet microfluidics to fabricate microcapsules with liquid core and poly(ethylene glycol) (PEG) gel shell. When entrapped inside these capsules, primary hepatocytes rapidly formed cell-cell contacts and assembled into compact spheroids. High levels of hepatic function were maintained inside the capsules for over ten days. The microencapsulation approach described here is compatible with difficult-to-culture primary epithelial cells, allows for tuning gel mechanical properties and diffusivity, and may be used in the future for high density suspension cell cultures. STATEMENT OF SIGNIFICANCE Our paper combines an interesting new way for making capsules with cultivation of difficult-to-maintain primary epithelial cells (hepatocytes). The microcapsules described here will enable high density suspension culture of hepatocytes or other cells and may be used as building blocks for engineering tissues.
Collapse
|
26
|
Sgambato A, Cipolla L, Russo L. Bioresponsive Hydrogels: Chemical Strategies and Perspectives in Tissue Engineering. Gels 2016; 2:E28. [PMID: 30674158 PMCID: PMC6318637 DOI: 10.3390/gels2040028] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/28/2016] [Accepted: 10/08/2016] [Indexed: 12/28/2022] Open
Abstract
Disease, trauma, and aging account for a significant number of clinical disorders. Regenerative medicine is emerging as a very promising therapeutic option. The design and development of new cell-customised biomaterials able to mimic extracellular matrix (ECM) functionalities represents one of the major strategies to control the cell fate and stimulate tissue regeneration. Recently, hydrogels have received a considerable interest for their use in the modulation and control of cell fate during the regeneration processes. Several synthetic bioresponsive hydrogels are being developed in order to facilitate cell-matrix and cell-cell interactions. In this review, new strategies and future perspectives of such synthetic cell microenvironments will be highlighted.
Collapse
Affiliation(s)
- Antonella Sgambato
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy.
| | - Laura Cipolla
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy.
| | - Laura Russo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, 20126 Milano, Italy.
| |
Collapse
|
27
|
Haque A, Gheibi P, Gao Y, Foster E, Son KJ, You J, Stybayeva G, Patel D, Revzin A. Cell biology is different in small volumes: endogenous signals shape phenotype of primary hepatocytes cultured in microfluidic channels. Sci Rep 2016; 6:33980. [PMID: 27681582 PMCID: PMC5041105 DOI: 10.1038/srep33980] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 08/30/2016] [Indexed: 12/13/2022] Open
Abstract
The approaches for maintaining hepatocytes in vitro are aimed at recapitulating aspects of the native liver microenvironment through the use of co-cultures, surface coatings and 3D spheroids. This study highlights the effects of spatial confinement-a less studied component of the in vivo microenvironment. We demonstrate that hepatocytes cultured in low-volume microfluidic channels (microchambers) retain differentiated hepatic phenotype for 21 days whereas cells cultured in regular culture plates under identical conditions de-differentiate after 7 days. Careful consideration of nutrient delivery and oxygen tension suggested that these factors could not solely account for enhanced cell function in microchambers. Through a series of experiments involving microfluidic chambers of various heights and inhibition of key molecular pathways, we confirmed that phenotype of hepatocytes in small volumes was shaped by endogenous signals, both hepato-inductive growth factors (GFs) such as hepatocyte growth factor (HGF) and hepato-disruptive GFs such as transforming growth factor (TGF)-β1. Hepatocytes are not generally thought of as significant producers of GFs–this role is typically assigned to nonparenchymal cells of the liver. Our study demonstrates that, in an appropriate microenvironment, hepatocytes produce hepato-inductive and pro-fibrogenic signals at the levels sufficient to shape their phenotype and function.
Collapse
Affiliation(s)
- Amranul Haque
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Pantea Gheibi
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Yandong Gao
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Elena Foster
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Kyung Jin Son
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Jungmok You
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA.,Department of Plant and Environmental New Resources, Kyung Hee University, Youngin-si, Gyeonggi-do, South Korea
| | - Gulnaz Stybayeva
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Dipali Patel
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| | - Alexander Revzin
- Department of Biomedical Engineering, University of California Davis, CA 95616, USA
| |
Collapse
|
28
|
Ahadian S, Sadeghian RB, Salehi S, Ostrovidov S, Bae H, Ramalingam M, Khademhosseini A. Bioconjugated Hydrogels for Tissue Engineering and Regenerative Medicine. Bioconjug Chem 2015; 26:1984-2001. [PMID: 26280942 DOI: 10.1021/acs.bioconjchem.5b00360] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Samad Ahadian
- WPI-Advanced
Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
| | - Ramin Banan Sadeghian
- WPI-Advanced
Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
| | - Sahar Salehi
- WPI-Advanced
Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
| | - Serge Ostrovidov
- WPI-Advanced
Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
| | - Hojae Bae
- College
of Animal Bioscience and Technology, Department of Bioindustrial Technologies, Konkuk University, Hwayang-dong,
Kwangjin-gu, Seoul 143-701, Republic of Korea
| | - Murugan Ramalingam
- WPI-Advanced
Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
- Centre
for Stem Cell Research, Institute for Stem Cell Biology and Regenerative Medicine, Christian Medical College Campus, Vellore 632002, India
| | - Ali Khademhosseini
- WPI-Advanced
Institute for Materials Research, Tohoku University, Sendai 980-8577, Japan
- College
of Animal Bioscience and Technology, Department of Bioindustrial Technologies, Konkuk University, Hwayang-dong,
Kwangjin-gu, Seoul 143-701, Republic of Korea
- Department
of Medicine, Center for Biomedical Engineering, Brigham and Women’s Hospital, Harvard Medical School, Cambridge, Massachusetts 02139, United States
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02115, United States
| |
Collapse
|