1
|
Telang AC, Ference-Salo JT, McElliott MC, Chowdhury M, Beamish JA. Sustained alterations in proximal tubule gene expression in primary culture associate with HNF4A loss. Sci Rep 2024; 14:22927. [PMID: 39358473 PMCID: PMC11447228 DOI: 10.1038/s41598-024-73861-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 09/22/2024] [Indexed: 10/04/2024] Open
Abstract
Primary cultures of proximal tubule cells are widely used to model the behavior of kidney epithelial cells in vitro. However, de-differentiation of primary cells upon culture has been observed and appreciated for decades, yet the mechanisms driving this phenomenon remain poorly understood. This confounds the interpretation of experiments using primary kidney epithelial cells and prevents their use to engineer functional kidney tissue ex vivo. In this report, we measure the dynamics of cell-state transformations in early primary culture of mouse proximal tubules to identify key pathways and processes that correlate with and may drive de-differentiation. Our data show that the loss of proximal-tubule-specific genes is rapid, uniform, and sustained even after confluent, polarized epithelial monolayers develop. This de-differentiation occurs uniformly across many common culture condition variations. Changes in early culture were strongly associated with the loss of HNF4A. Exogenous re-expression of HNF4A can promote expression of a subset of proximal tubule genes in a de-differentiated proximal tubule cell line. Using genetically labeled proximal tubule cells, we show that selective pressures very early in culture influence which cells grow to confluence. Together, these data indicate that the loss of in vivo function in proximal tubule cultures occurs very early and suggest that the sustained loss of HNF4A is a key regulatory event mediating this change.
Collapse
Affiliation(s)
- Asha C Telang
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Jenna T Ference-Salo
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Madison C McElliott
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Mahboob Chowdhury
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA
| | - Jeffrey A Beamish
- Division of Nephrology, Department of Internal Medicine, University of Michigan, 1500 E. Medical Center Drive, SPC 5364, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
2
|
Ma C, Banan Sadeghian R, Negoro R, Fujimoto K, Araoka T, Ishiguro N, Takasato M, Yokokawa R. Efficient proximal tubule-on-chip model from hiPSC-derived kidney organoids for functional analysis of renal transporters. iScience 2024; 27:110760. [PMID: 39286490 PMCID: PMC11403423 DOI: 10.1016/j.isci.2024.110760] [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: 05/02/2024] [Revised: 06/24/2024] [Accepted: 08/14/2024] [Indexed: 09/19/2024] Open
Abstract
Renal transporters play critical roles in predicting potential drug-drug interactions. However, current in vitro models often fail to adequately express these transporters, particularly solute carrier proteins, including organic anion transporters (OAT1/3), and organic cation transporter 2 (OCT2). Here, we developed a hiPSC-derived kidney organoids-based proximal tubule-on-chip (OPTC) model that emulates in vivo renal physiology to assess transporter function. Compared to chips based on immortalized cells, OPTC derived from the two most commonly used differentiation protocols exhibited significant improvement in expression level and polarity of OAT1/3 and OCT2. Hence, the OPTC demonstrates enhanced functionality in efflux and uptake assessments, and nephrotoxicity. Furthermore, these functionalities are diminished upon adding inhibitors during substrate-inhibitor interactions, which were closer to in vivo observations. Overall, these results support that OPTC can reliably assess the role of renal transporters in drug transport and nephrotoxicity, paving the way for personalized models to assess renal transport and disease modeling.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | | | - Ryosuke Negoro
- Laboratory of Molecular Pharmacokinetics, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu 525-8577, Japan
| | - Kazuya Fujimoto
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| | - Toshikazu Araoka
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto 606-8507, Japan
| | - Naoki Ishiguro
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co. Ltd, Kobe, Japan
| | - Minoru Takasato
- RIKEN Center for Biosystems Dynamics Research (BDR), Kobe 650-0047, Japan
- Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto 615-8540, Japan
| |
Collapse
|
3
|
Wang H, Walles T, Wiese-Rischke C. Patient-Derived Lung Cancer "Sandwich Cultures" with a Preserved Tumor Microenvironment. Tissue Eng Part C Methods 2024; 30:27-37. [PMID: 38115596 PMCID: PMC10818046 DOI: 10.1089/ten.tec.2023.0199] [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: 08/01/2023] [Accepted: 11/02/2023] [Indexed: 12/21/2023] Open
Abstract
In the past, different spheroid-, organotypic-, and three-dimensional (3D) bioprinting lung cancer models were established for in vitro drug testing and personalized medicine. These tissue models cannot depict the tumor microenvironment (TME) and, therefore, research addressing tumor cell-TME interactions is limited. To overcome this hurdle, we applied patient-derived lung tumor samples to establish new in vitro models. To analyze the tissue model properties, we established two-dimensional (2D) and 3D coculture tissue models exposed to static and dynamic culture conditions that afforded tissue culture for up to 28 days. Our tissue models were characterized by hematoxylin eosin staining, M30 enzyme-linked immunosorbent assay, and immunofluorescence staining against specific lung cancer markers (TTF-1 and p40/p63), cancer-associated fibroblast (CAF) markers (α-SMA and MCT4), and fibronectin (FN). The 3D models were generated with higher success rate than the corresponding 2D model. The cell density of the static 3D model increased from 21 to 28 days, whereas the apoptosis decreased. The dynamic 3D model possessed an even higher cell density than the static 3D model. We identified lung cancer cells, CAFs, and FN. Therefore, a novel in vitro 3D lung cancer model was established, which simulated the TME for 28 days and possessed a structural complexity.
Collapse
Affiliation(s)
- Hailong Wang
- University Clinic for Cardiac and Thoracic Surgery, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Thorsten Walles
- University Clinic for Cardiac and Thoracic Surgery, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Cornelia Wiese-Rischke
- University Clinic for Cardiac and Thoracic Surgery, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
4
|
Khalil NN, Petersen AP, Song CJ, Chen Y, Takamoto K, Kellogg AC, Chen EZ, McMahon AP, McCain ML. User-friendly microfluidic system reveals native-like morphological and transcriptomic phenotypes induced by shear stress in proximal tubule epithelium. APL Bioeng 2023; 7:036106. [PMID: 37584027 PMCID: PMC10424157 DOI: 10.1063/5.0143614] [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: 01/24/2023] [Accepted: 07/24/2023] [Indexed: 08/17/2023] Open
Abstract
Drug-induced nephrotoxicity is a leading cause of drug attrition, partly due to the limited relevance of pre-clinical models of the proximal tubule. Culturing proximal tubule epithelial cells (PTECs) under fluid flow to mimic physiological shear stress has been shown to improve select phenotypes, but existing flow systems are expensive and difficult to implement by non-experts in microfluidics. Here, we designed and fabricated an accessible and modular flow system for culturing PTECs under physiological shear stress, which induced native-like cuboidal morphology, downregulated pathways associated with hypoxia, stress, and injury, and upregulated xenobiotic metabolism pathways. We also compared the expression profiles of shear-dependent genes in our in vitro PTEC tissues to that of ex vivo proximal tubules and observed stronger clustering between ex vivo proximal tubules and PTECs under physiological shear stress relative to PTECs under negligible shear stress. Together, these data illustrate the utility of our user-friendly flow system and highlight the role of shear stress in promoting native-like morphological and transcriptomic phenotypes in PTECs in vitro, which is critical for developing more relevant pre-clinical models of the proximal tubule for drug screening or disease modeling.
Collapse
Affiliation(s)
- Natalie N. Khalil
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Andrew P. Petersen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | | | - Yibu Chen
- USC Libraries Bioinformatics Service, University of Southern California, Los Angeles, California 90089, USA
| | - Kaelyn Takamoto
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Austin C. Kellogg
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | - Elaine Zhelan Chen
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, Los Angeles, California 90089, USA
| | | | - Megan L. McCain
- Author to whom correspondence should be addressed:. Tel.: +1 2138210791. URL:https://livingsystemsengineering.usc.edu
| |
Collapse
|
5
|
Optimization of Primary Human Bronchial Epithelial 3D Cell Culture with Donor-Matched Fibroblasts and Comparison of Two Different Culture Media. Int J Mol Sci 2023; 24:ijms24044113. [PMID: 36835529 PMCID: PMC9965758 DOI: 10.3390/ijms24044113] [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: 12/16/2022] [Revised: 02/11/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
In vitro airway models are increasingly important for pathomechanistic analyses of respiratory diseases. Existing models are limited in their validity by their incomplete cellular complexity. We therefore aimed to generate a more complex and meaningful three-dimensional (3D) airway model. Primary human bronchial epithelial cells (hbEC) were propagated in airway epithelial cell growth (AECG) or PneumaCult ExPlus medium. Generating 3D models, hbEC were airlifted and cultured on a collagen matrix with donor-matched bronchial fibroblasts for 21 days comparing two media (AECG or PneumaCult ALI (PC ALI)). 3D models were characterized by histology and immunofluorescence staining. The epithelial barrier function was quantified by transepithelial electrical resistance (TEER) measurements. The presence and function of ciliated epithelium were determined by Western blot and microscopy with high-speed camera. In 2D cultures, an increased number of cytokeratin 14-positive hbEC was present with AECG medium. In 3D models, AECG medium accounted for high proliferation, resulting in hypertrophic epithelium and fluctuating TEER values. Models cultured with PC ALI medium developed a functional ciliated epithelium with a stable epithelial barrier. Here, we established a 3D model with high in vivo-in vitro correlation, which has the potential to close the translational gap for investigations of the human respiratory epithelium in pharmacological, infectiological, and inflammatory research.
Collapse
|
6
|
Hunter K, Larsen JA, Love HD, Evans RC, Roy S, Zent R, Harris RC, Wilson MH, Fissell WH. Inhibition of Transforming Growth Factor-β Improves Primary Renal Tubule Cell Differentiation in Long-Term Culture. Tissue Eng Part A 2023; 29:102-111. [PMID: 36274231 PMCID: PMC10081716 DOI: 10.1089/ten.tea.2022.0147] [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: 08/05/2022] [Accepted: 10/19/2022] [Indexed: 11/19/2022] Open
Abstract
Patient-oriented applications of cell culture include cell therapy of organ failure like chronic renal failure. Clinical deployment of a cell-based device for artificial renal replacement requires qualitative and quantitative fidelity of a cultured cell to its in vivo counterpart. Active specific apicobasal ion transport reabsorbs 90-99% of the filtered load of salt and water in the kidney. In a bioengineered kidney, tubular transport concentrates wastes and eliminates the need for hemodialysis, but renal tubule cells in culture transport little or no salt and water due to dedifferentiation that mammalian cells undergo in vitro thereby losing important cell-type specific functions. We previously identified transforming growth factor-β (TGF-β) as a signaling pathway necessary for in vitro differentiation of renal tubule cells. Inhibition of TGF-β receptor-1 led to active and inhibitable electrolyte and water transport by primary human renal tubule epithelial cells in vitro. Addition of metformin increased transport, in the context of a transient effect on 5'-AMP-activated kinase phosphorylation. These data motivated us to examine whether increased transport was an idiosyncratic effect of SB431542, probe pathways downstream of TGF-β receptors possibly responsible for the improved differentiation, evaluate whether TGF-β inhibition induced a range of differentiated tubule functions, and to explore crosstalk between the effects of SB431542 and metformin. In this study, we use multiple small-molecule inhibitors of canonical and noncanonical pathways to confirm that inhibition of canonical TGF-β signaling caused the increased apicobasal transport. Hallmarks of proximal tubule cell function, including sodium reabsorption, para-amino hippurate excretion, and glucose uptake increased with TGF-β inhibition, and the specificity of the response was shown using inhibitors of each transport protein. We did not find any evidence of crosstalk between metformin and SB431542. These data suggest that the TGF-β signaling pathway governs multiple features of differentiation in renal proximal tubule cells in vitro. Inhibition of TGF-β by pharmacologic or genome engineering approaches may be a viable approach to enhancing differentiated function of tubule cells in vitro. Impact statement Cell therapy of renal failure requires qualitative and quantitative fidelity between in vitro and in vivo phenotypes, which has been elusive. We show that control of transforming growth factor-β signaling can promote differentiation of renal tubule cells grown in artificial environments. This is a key enabling step for cell therapy of renal failure.
Collapse
Affiliation(s)
- Kuniko Hunter
- Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jaclyn A. Larsen
- School of Medicine, University of Utah, Salt Lake City, Utah, USA
| | - Harold D. Love
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Rachel C. Evans
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Shuvo Roy
- Bioengineering and Therapeutic Sciences, University of California, San Francisco, California, USA
| | - Roy Zent
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Raymond C. Harris
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Matthew H. Wilson
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - William H. Fissell
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
7
|
Uda J, Ashizawa N, Iwanaga T. An evaluation method for uric acid uptake inhibition using primary human proximal tubule epithelial cells treated with insulin. NUCLEOSIDES, NUCLEOTIDES & NUCLEIC ACIDS 2022; 41:724-735. [PMID: 35770496 DOI: 10.1080/15257770.2022.2070204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/07/2022] [Accepted: 04/20/2022] [Indexed: 06/15/2023]
Abstract
The effects of uricosuric agents have been evaluated in vitro with indices of uric acid uptake into human urate transporter 1 (URAT1)-overexpressed oocytes or cells. In the present study, we evaluated a method using primary human renal proximal tubule epithelial cells (RPTECs). Pretreatment of RPTECs with insulin significantly increased the uptake of uric acid into these cells. The uric acid uptake was inhibited in a concentration-dependent manner by the URAT1 inhibitors benzbromarone and dotinurad. Therefore, effects of uricosuric agents can be evaluated by the novel method, which is closer to the physiological system compared with previous methods.
Collapse
Affiliation(s)
- Junichiro Uda
- Medicinal Chemistry Research Department, Research Institute, FUJIYAKUHIN CO., Ltd, Saitama, Japan
| | - Naoki Ashizawa
- Biological Research Department, Research Institute, FUJIYAKUHIN CO., Ltd, Saitama, Japan
| | - Takashi Iwanaga
- Biological Research Department, Research Institute, FUJIYAKUHIN CO., Ltd, Saitama, Japan
| |
Collapse
|
8
|
Ajay AK. Functional Drug Screening using Kidney Cells On-A-Chip: Advances in Disease Modeling and Development of Biomarkers. KIDNEY360 2022; 3:194-198. [PMID: 35373124 PMCID: PMC8967633 DOI: 10.34067/kid.0007172021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 01/13/2022] [Indexed: 01/12/2023]
Affiliation(s)
- Amrendra K. Ajay
- Division of Renal Medicine, Brigham and Women’s Hospital, Boston, Massachusetts,Department of Medicine, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
9
|
Application of Porcine Kidney-Derived Extracellular Matrix as Coating, Hydrogel, and Scaffold Material for Renal Proximal Tubular Epithelial Cell. BIOMED RESEARCH INTERNATIONAL 2022; 2022:2220641. [PMID: 35127940 PMCID: PMC8816548 DOI: 10.1155/2022/2220641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/27/2021] [Indexed: 11/30/2022]
Abstract
Background Human renal proximal tubular epithelial (RPTE) cell is a very useful tool for kidney-related experiments in vitro/ex vivo. However, only a few primary RPTE cells can be obtained through kidney biopsy, the proliferation rate of primary cell is very low, and the cultured cell properties are easily altered in artificial conditions. Thus, RPTE cell usage is very tricky; we applied porcine kidney-derived extracellular matrix (renal ECM) as coating, hydrogel, and scaffold material to increase cell proliferation and maintain cellular properties providing three-dimensional (3D) niche, which can be a valuable cell delivery vehicle. Methods Porcine renal ECM was prepared by decellularization using 1% Triton X-100, solubilized with 0.5 M acetic acid. The final protein concentration was adjusted to 10 μg/μL (pH 7.0). The efficacies as coating, hydrogel, and scaffold materials were analyzed through cell morphology, proliferation rate, renal-associated gene expressions, chemical composition, and microstructure evaluation. The efficacies as a coating material were compared with Matrigel, collagen type 1 (col1), gelatin, fibrinogen, and thrombin. After confirmation of coating effects, the effective concentration range was decided. The efficacies as hydrogel and scaffold materials were compared with hyaluronic acid (HA) and col1, respectively. Results As the coating material, renal ECM showed a higher cell proliferation rate compared to other materials, except for Matrigel. Renal-associated gene expressions were significantly enhanced in the renal ECM than other materials. Coating effect on cell proliferation was dependent on the renal ECM concentration, and the effective concentration ranged from 30 to 100 μg. As the hydrogel material, renal ECM showed a distinct inner cell network morphology and significantly increased renal-associated gene expressions, compared to HA hydrogel. As the scaffold material, renal ECM showed specific amide peaks, enhanced internal porosity, cell proliferation rate, and renal-associated gene expression compared to the col1 scaffold. Conclusions We concluded that renal ECM can be a suitable material for RPTE cell culture and usage. More practically, the coated renal ECM stimulates RPTE cell proliferation, and the hydrogel and scaffold of renal ECM provide useful 3D culture niche and cell delivery vehicles maintaining renal cell properties.
Collapse
|
10
|
Van Ness KP, Cesar F, Yeung CK, Himmelfarb J, Kelly EJ. Microphysiological systems in absorption, distribution, metabolism, and elimination sciences. Clin Transl Sci 2022; 15:9-42. [PMID: 34378335 PMCID: PMC8742652 DOI: 10.1111/cts.13132] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 07/20/2021] [Accepted: 07/22/2021] [Indexed: 12/11/2022] Open
Abstract
The use of microphysiological systems (MPS) to support absorption, distribution, metabolism, and elimination (ADME) sciences has grown substantially in the last decade, in part driven by regulatory demands to move away from traditional animal-based safety assessment studies and industry desires to develop methodologies to efficiently screen and characterize drugs in the development pipeline. The past decade of MPS development has yielded great user-driven technological advances with the collective fine-tuning of cell culture techniques, fluid delivery systems, materials engineering, and performance enhancing modifications. The rapid advances in MPS technology have now made it feasible to evaluate critical ADME parameters within a stand-alone organ system or through interconnected organ systems. This review surveys current MPS developed for liver, kidney, and intestinal systems as stand-alone or interconnected organ systems, and evaluates each system for specific performance criteria recommended by regulatory authorities and MPS leaders that would render each system suitable for evaluating drug ADME. Whereas some systems are more suitable for ADME type research than others, not all system designs were intended to meet the recently published desired performance criteria and are reported as a summary of initial proof-of-concept studies.
Collapse
Affiliation(s)
- Kirk P. Van Ness
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
| | - Francine Cesar
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
| | - Catherine K. Yeung
- Department of PharmacyUniversity of WashingtonSeattleWashingtonUSA
- Kidney Research InstituteUniversity of WashingtonSeattleWashingtonUSA
| | | | - Edward J. Kelly
- Department of PharmaceuticsUniversity of WashingtonSeattleWashingtonUSA
- Kidney Research InstituteUniversity of WashingtonSeattleWashingtonUSA
| |
Collapse
|
11
|
Ebefors K, Lassén E, Anandakrishnan N, Azeloglu EU, Daehn IS. Modeling the Glomerular Filtration Barrier and Intercellular Crosstalk. Front Physiol 2021; 12:689083. [PMID: 34149462 PMCID: PMC8206562 DOI: 10.3389/fphys.2021.689083] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 05/05/2021] [Indexed: 12/11/2022] Open
Abstract
The glomerulus is a compact cluster of capillaries responsible for blood filtration and initiating urine production in the renal nephrons. A trilaminar structure in the capillary wall forms the glomerular filtration barrier (GFB), composed of glycocalyx-enriched and fenestrated endothelial cells adhering to the glomerular basement membrane and specialized visceral epithelial cells, podocytes, forming the outermost layer with a molecular slit diaphragm between their interdigitating foot processes. The unique dynamic and selective nature of blood filtration to produce urine requires the functionality of each of the GFB components, and hence, mimicking the glomerular filter in vitro has been challenging, though critical for various research applications and drug screening. Research efforts in the past few years have transformed our understanding of the structure and multifaceted roles of the cells and their intricate crosstalk in development and disease pathogenesis. In this review, we present a new wave of technologies that include glomerulus-on-a-chip, three-dimensional microfluidic models, and organoids all promising to improve our understanding of glomerular biology and to enable the development of GFB-targeted therapies. Here, we also outline the challenges and the opportunities of these emerging biomimetic systems that aim to recapitulate the complex glomerular filter, and the evolving perspectives on the sophisticated repertoire of cellular signaling that comprise the glomerular milieu.
Collapse
Affiliation(s)
- Kerstin Ebefors
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Emelie Lassén
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Nanditha Anandakrishnan
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Evren U Azeloglu
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Ilse S Daehn
- Division of Nephrology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
12
|
Mihevc M, Petreski T, Maver U, Bevc S. Renal proximal tubular epithelial cells: review of isolation, characterization, and culturing techniques. Mol Biol Rep 2020; 47:9865-9882. [PMID: 33170426 DOI: 10.1007/s11033-020-05977-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/03/2020] [Indexed: 12/23/2022]
Abstract
The kidney is a complex organ, comprised primarily of glomerular, tubular, mesangial, and endothelial cells, and podocytes. The fact that renal cells are terminally differentiated at 34 weeks of gestation is the main obstacle in regeneration and treatment of acute kidney injury or chronic kidney disease. Furthermore, the number of chronic kidney disease patients is ever increasing and with it the medical community should aim to improve existing and develop new methods of renal replacement therapy. On the other hand, as polypharmacy is on the rise, thought should be given into developing new ways of testing drug safety. A possible way to tackle these issues is with isolation and culture of renal cells. Several protocols are currently described to isolate the desired cells, of which the most isolated are the proximal tubular epithelial cells. They play a major role in water homeostasis, acid-base control, reabsorption of compounds, and secretion of xenobiotics and endogenous metabolites. When exposed to ischemic, toxic, septic, or obstructive conditions their death results in what we clinically perceive as acute kidney injury. Additionally, due to renal cells' limited regenerative potential, the profibrotic environment inevitably leads to chronic kidney disease. In this review we will focus on human proximal tubular epithelial cells. We will cover human kidney culture models, cell sources, isolation, culture, immortalization, and characterization subdivided into morphological, phenotypical, and functional characterization.
Collapse
Affiliation(s)
- Matic Mihevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia
| | - Tadej Petreski
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia
| | - Uroš Maver
- Faculty of Medicine, Institute of Biomedical Sciences, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
| | - Sebastjan Bevc
- Department of Nephrology, Clinic for Internal Medicine, University Medical Centre Maribor, Ljubljanska ulica 5, 2000, Maribor, Slovenia.
- Department of Pharmacology, Faculty of Medicine, University of Maribor, Taborska ulica 8, 2000, Maribor, Slovenia.
| |
Collapse
|
13
|
Wallstabe L, Göttlich C, Nelke LC, Kühnemundt J, Schwarz T, Nerreter T, Einsele H, Walles H, Dandekar G, Nietzer SL, Hudecek M. ROR1-CAR T cells are effective against lung and breast cancer in advanced microphysiologic 3D tumor models. JCI Insight 2019; 4:126345. [PMID: 31415244 DOI: 10.1172/jci.insight.126345] [Citation(s) in RCA: 140] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 08/08/2019] [Indexed: 02/02/2023] Open
Abstract
Solid tumors impose immunologic and physical barriers to the efficacy of chimeric antigen receptor (CAR) T cell therapy that are not reflected in conventional preclinical testing against singularized tumor cells in 2-dimensional culture. Here, we established microphysiologic three-dimensional (3D) lung and breast cancer models that resemble architectural and phenotypical features of primary tumors and evaluated the antitumor function of receptor tyrosine kinase-like orphan receptor 1-specific (ROR1-specific) CAR T cells. 3D tumors were established from A549 (non-small cell lung cancer) and MDA-MB-231 (triple-negative breast cancer) cell lines on a biological scaffold with intact basement membrane (BM) under static and dynamic culture conditions, which resulted in progressively increasing cell mass and invasive growth phenotype (dynamic > static; MDA-MB-231 > A549). Treatment with ROR1-CAR T cells conferred potent antitumor effects. In dynamic culture, CAR T cells actively entered arterial medium flow and adhered to and infiltrated the tumor mass. ROR1-CAR T cells penetrated deep into tumor tissue and eliminated multiple layers of tumor cells located above and below the BM. The microphysiologic 3D tumor models developed in this study are standardized, scalable test systems that can be used either in conjunction with or in lieu of animal testing to interrogate the antitumor function of CAR T cells and to obtain proof of concept for their safety and efficacy before clinical application.
Collapse
Affiliation(s)
| | - Claudia Göttlich
- Tissue Engineering and Regenerative Medicine, Universitätsklinikum Würzburg, Würzburg, Germany.,Fraunhofer Institute for Silicate Research, Translational Center Regenerative Therapies, Würzburg, Germany
| | - Lena C Nelke
- Tissue Engineering and Regenerative Medicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Johanna Kühnemundt
- Tissue Engineering and Regenerative Medicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Thomas Schwarz
- Tissue Engineering and Regenerative Medicine, Universitätsklinikum Würzburg, Würzburg, Germany.,Fraunhofer Institute for Silicate Research, Translational Center Regenerative Therapies, Würzburg, Germany
| | | | | | - Heike Walles
- Tissue Engineering and Regenerative Medicine, Universitätsklinikum Würzburg, Würzburg, Germany.,Fraunhofer Institute for Silicate Research, Translational Center Regenerative Therapies, Würzburg, Germany
| | - Gudrun Dandekar
- Tissue Engineering and Regenerative Medicine, Universitätsklinikum Würzburg, Würzburg, Germany.,Fraunhofer Institute for Silicate Research, Translational Center Regenerative Therapies, Würzburg, Germany
| | - Sarah L Nietzer
- Tissue Engineering and Regenerative Medicine, Universitätsklinikum Würzburg, Würzburg, Germany
| | | |
Collapse
|
14
|
Petrosyan A, Cravedi P, Villani V, Angeletti A, Manrique J, Renieri A, De Filippo RE, Perin L, Da Sacco S. A glomerulus-on-a-chip to recapitulate the human glomerular filtration barrier. Nat Commun 2019; 10:3656. [PMID: 31409793 PMCID: PMC6692336 DOI: 10.1038/s41467-019-11577-z] [Citation(s) in RCA: 113] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2018] [Accepted: 07/23/2019] [Indexed: 12/24/2022] Open
Abstract
In this work we model the glomerular filtration barrier, the structure responsible for filtering the blood and preventing the loss of proteins, using human podocytes and glomerular endothelial cells seeded into microfluidic chips. In long-term cultures, cells maintain their morphology, form capillary-like structures and express slit diaphragm proteins. This system recapitulates functions and structure of the glomerulus, including permselectivity. When exposed to sera from patients with anti-podocyte autoantibodies, the chips show albuminuria proportional to patients' proteinuria, phenomenon not observed with sera from healthy controls or individuals with primary podocyte defects. We also show its applicability for renal disease modeling and drug testing. A total of 2000 independent chips were analyzed, supporting high reproducibility and validation of the system for high-throughput screening of therapeutic compounds. The study of the patho-physiology of the glomerulus and identification of therapeutic targets are also feasible using this chip.
Collapse
Affiliation(s)
- Astgik Petrosyan
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Paolo Cravedi
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Valentina Villani
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Andrea Angeletti
- Department of Experimental, Diagnostic and Specialty Medicine, Nephrology, Dialysis and Renal Transplant Unit, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Joaquin Manrique
- Nephrology Service, Complejo Hospitalario de Navarra, Pamplona, Spain
| | | | - Roger E De Filippo
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, CA, USA
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Laura Perin
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Stefano Da Sacco
- GOFARR Laboratory for Organ Regenerative Research and Cell Therapeutics in Urology, Saban Research Institute, Division of Urology, Children's Hospital Los Angeles, Los Angeles, CA, USA.
- Department of Urology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
15
|
Love HD, Ao M, Jorgensen S, Swearingen L, Ferrell N, Evans R, Gewin L, Harris RC, Zent R, Roy S, Fissell WH. Substrate Elasticity Governs Differentiation of Renal Tubule Cells in Prolonged Culture. Tissue Eng Part A 2019; 25:1013-1022. [PMID: 30484388 DOI: 10.1089/ten.tea.2018.0182] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
IMPACT STATEMENT Successful clinical tissue engineering requires functional fidelity of the cultured cell to its in vivo counterpart, but this has been elusive in renal tissue engineering. Typically, renal proximal tubule cells in culture have a flattened morphology and do not express key transporters essential to their function. In this article, we show for the first time that in vitro substrate mechanical properties dictate differentiation of cultured renal proximal tubule cells. Remarkably, this effect was only discernable after 4 weeks in culture, longer than usually reported for this cell type. These results demonstrate a new tunable parameter to optimize cell differentiation in renal tissue engineering.
Collapse
Affiliation(s)
- Harold D Love
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Mingfang Ao
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Seiver Jorgensen
- 2College of Arts and Science, Vanderbilt University, Nashville, Tennessee
| | - Lindsey Swearingen
- 2College of Arts and Science, Vanderbilt University, Nashville, Tennessee
| | - Nicholas Ferrell
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Rachel Evans
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Leslie Gewin
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Raymond C Harris
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Roy Zent
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shuvo Roy
- 3Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, California
| | - William H Fissell
- 1Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| |
Collapse
|
16
|
Apoptosis signal-regulating kinase 1 inhibition attenuates cardiac hypertrophy and cardiorenal fibrosis induced by uremic toxins: Implications for cardiorenal syndrome. PLoS One 2017; 12:e0187459. [PMID: 29107962 PMCID: PMC5673193 DOI: 10.1371/journal.pone.0187459] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 10/22/2017] [Indexed: 01/16/2023] Open
Abstract
Intracellular accumulation of protein-bound uremic toxins in the setting of cardiorenal syndrome leads to adverse effects on cardiorenal cellular functions, where cardiac hypertrophy and cardiorenal fibrosis are the hallmarks. In this study, we sought to determine if Apoptosis Signal-Regulated Kinase 1 (ASK1), an upstream regulator of cellular stress response, mediates cardiac hypertrophy and cardiorenal fibrosis induced by indoxyl sulfate (IS) and p-cresol sulfate (PCS) in vitro, and whether ASK1 inhibition is beneficial to ameliorate these cellular effects. PCS augmented cardiac myocyte hypertrophy and fibroblast collagen synthesis (as determined by 3H-leucine and 3H-proline incorporation, respectively), similar to our previous finding with IS. IS and PCS also increased collagen synthesis of proximal tubular cells and renal mesangial cells. Pro-hypertrophic (α-skeletal muscle actin and β-MHC) and pro-fibrotic genes (TGF-β1 and ctgf) were induced by both IS and PCS. Western blot analyses revealed the activation of ASK1 and downstream mitogen activated protein kinases (MAPKs) (p38MAPK and ERK1/2) as well as nuclear factor-kappa B (NF-κB) by IS and PCS. ASK1, OAT1/3, ERK1/2 and p38MAPK inhibitors suppressed all these effects. In summary, IS and PCS exhibit pro-hypertrophic and pro-fibrotic properties, at least in part, via the activation of ASK1 and its downstream pathways. ASK1 inhibitor is an effective therapeutic agent to alleviate protein-bound uremic toxin-induced cardiac hypertrophy and cardiorenal fibrosis in vitro, and may be translated further for cardiorenal syndrome therapy.
Collapse
|
17
|
Abstract
AbstractAn analysis of biological effects induced by environmental toxins and exposure-related evaluation of potential risks for health and environment represent central tasks in classical biomonitoring. While epidemiological data and population surveys are clearly the methodological frontline of this scientific field, cellbased in vitro assays provide information on toxin-affected cellular pathways and mechanisms, and are important sources for the identification of relevant biomarkers. This review provides an overview on currently available in vitro methods based on cultured cells, as well as some limitations and considerations that are of specific interest in the context of environmental toxicology. Today, a large number of different endpoints can be determined to pinpoint basal and specific toxicological cellular effects. Technological progress and increasingly refined protocols are extending the possibilities of cell-based in vitro assays in environmental toxicology and promoting their increasingly important role in biomonitoring.
Collapse
|
18
|
Kremer A, Ribitsch I, Reboredo J, Dürr J, Egerbacher M, Jenner F, Walles H. Three-Dimensional Coculture of Meniscal Cells and Mesenchymal Stem Cells in Collagen Type I Hydrogel on a Small Intestinal Matrix—A Pilot Study Toward Equine Meniscus Tissue Engineering. Tissue Eng Part A 2017; 23:390-402. [DOI: 10.1089/ten.tea.2016.0317] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Affiliation(s)
- Antje Kremer
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center Wuerzburg ‘Regenerative therapies,’ Wuerzburg Branch of the Fraunhofer IGB, Wuerzburg, Germany
| | - Iris Ribitsch
- Vienna Equine Tissue Engineering and Regenerative Medicine, Equine Clinic, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Jenny Reboredo
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center Wuerzburg ‘Regenerative therapies,’ Wuerzburg Branch of the Fraunhofer IGB, Wuerzburg, Germany
| | - Julia Dürr
- Department of Pathobiology, Institute of Histology & Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Monika Egerbacher
- Department of Pathobiology, Institute of Histology & Embryology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Florien Jenner
- Vienna Equine Tissue Engineering and Regenerative Medicine, Equine Clinic, University of Veterinary Medicine Vienna, Vienna, Austria
- Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Heike Walles
- Department of Tissue Engineering and Regenerative Medicine (TERM), University Hospital Wuerzburg, Wuerzburg, Germany
- Translational Center Wuerzburg ‘Regenerative therapies,’ Wuerzburg Branch of the Fraunhofer IGB, Wuerzburg, Germany
| |
Collapse
|
19
|
Chang SY, Weber EJ, Ness KV, Eaton DL, Kelly EJ. Liver and Kidney on Chips: Microphysiological Models to Understand Transporter Function. Clin Pharmacol Ther 2016; 100:464-478. [PMID: 27448090 DOI: 10.1002/cpt.436] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Revised: 07/13/2016] [Accepted: 07/15/2016] [Indexed: 12/19/2022]
Abstract
Because of complex cellular microenvironments of both the liver and kidneys, accurate modeling of transport function has remained a challenge, leaving a dire need for models that can faithfully recapitulate both the architecture and cell-cell interactions observed in vivo. The study of hepatic and renal transport function is a fundamental component of understanding the metabolic fate of drugs and xenobiotics; however, there are few in vitro systems conducive for these types of studies. For both the hepatic and renal systems, we provide an overview of the location and function of the most significant phase I/II/III (transporter) of enzymes, and then review current in vitro systems for the suitability of a transporter function study and provide details on microphysiological systems that lead the field in these investigations. Microphysiological modeling of the liver and kidneys using "organ-on-a-chip" technologies is rapidly advancing in transport function assessment and has emerged as a promising method to evaluate drug and xenobiotic metabolism. Future directions for the field are also discussed along with technical challenges encountered in complex multiple-organs-on-chips development.
Collapse
Affiliation(s)
- S Y Chang
- Department of Occupational and Environmental Health Sciences, University of Washington, Seattle, Washington, USA
| | - E J Weber
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - Kp Van Ness
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - D L Eaton
- Department of Occupational and Environmental Health Sciences, University of Washington, Seattle, Washington, USA
| | - E J Kelly
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
20
|
Nietzer S, Baur F, Sieber S, Hansmann J, Schwarz T, Stoffer C, Häfner H, Gasser M, Waaga-Gasser AM, Walles H, Dandekar G. Mimicking Metastases Including Tumor Stroma: A New Technique to Generate a Three-Dimensional Colorectal Cancer Model Based on a Biological Decellularized Intestinal Scaffold. Tissue Eng Part C Methods 2016; 22:621-35. [PMID: 27137941 PMCID: PMC4943469 DOI: 10.1089/ten.tec.2015.0557] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Tumor models based on cancer cell lines cultured two-dimensionally (2D) on plastic lack histological complexity and functionality compared to the native microenvironment. Xenogenic mouse tumor models display higher complexity but often do not predict human drug responses accurately due to species-specific differences. We present here a three-dimensional (3D) in vitro colon cancer model based on a biological scaffold derived from decellularized porcine jejunum (small intestine submucosa+mucosa, SISmuc). Two different cell lines were used in monoculture or in coculture with primary fibroblasts. After 14 days of culture, we demonstrated a close contact of human Caco2 colon cancer cells with the preserved basement membrane on an ultrastructural level as well as morphological characteristics of a well-differentiated epithelium. To generate a tissue-engineered tumor model, we chose human SW480 colon cancer cells, a reportedly malignant cell line. Malignant characteristics were confirmed in 2D cell culture: SW480 cells showed higher vimentin and lower E-cadherin expression than Caco2 cells. In contrast to Caco2, SW480 cells displayed cancerous characteristics such as delocalized E-cadherin and nuclear location of β-catenin in a subset of cells. One central drawback of 2D cultures—especially in consideration of drug testing—is their artificially high proliferation. In our 3D tissue-engineered tumor model, both cell lines showed decreased numbers of proliferating cells, thus correlating more precisely with observations of primary colon cancer in all stages (UICC I-IV). Moreover, vimentin decreased in SW480 colon cancer cells, indicating a mesenchymal to epithelial transition process, attributed to metastasis formation. Only SW480 cells cocultured with fibroblasts induced the formation of tumor-like aggregates surrounded by fibroblasts, whereas in Caco2 cocultures, a separate Caco2 cell layer was formed separated from the fibroblast compartment beneath. To foster tissue generation, a bioreactor was constructed for dynamic culture approaches. This induced a close tissue-like association of cultured tumor cells with fibroblasts reflecting tumor biopsies. Therapy with 5-fluorouracil (5-FU) was effective only in 3D coculture. In conclusion, our 3D tumor model reflects human tissue-related tumor characteristics, including lower tumor cell proliferation. It is now available for drug testing in metastatic context—especially for substances targeting tumor–stroma interactions.
Collapse
Affiliation(s)
- Sarah Nietzer
- 1 Institute of Tissue Engineering and Regenerative Medicine (TERM), University Hospital of the Julius-Maximilians University , Würzburg, Germany
| | - Florentin Baur
- 1 Institute of Tissue Engineering and Regenerative Medicine (TERM), University Hospital of the Julius-Maximilians University , Würzburg, Germany
| | - Stefan Sieber
- 1 Institute of Tissue Engineering and Regenerative Medicine (TERM), University Hospital of the Julius-Maximilians University , Würzburg, Germany
| | - Jan Hansmann
- 1 Institute of Tissue Engineering and Regenerative Medicine (TERM), University Hospital of the Julius-Maximilians University , Würzburg, Germany
| | - Thomas Schwarz
- 1 Institute of Tissue Engineering and Regenerative Medicine (TERM), University Hospital of the Julius-Maximilians University , Würzburg, Germany
| | - Carolin Stoffer
- 1 Institute of Tissue Engineering and Regenerative Medicine (TERM), University Hospital of the Julius-Maximilians University , Würzburg, Germany
| | - Heide Häfner
- 2 Translational Center Würzburg "Regenerative Therapies in Oncology and Musculoskeletal Disease, " Fraunhofer Institute Interfacial Engineering and Biotechnology IGB , Würzburg, Germany
| | - Martin Gasser
- 3 Department of Surgery I, Molecular Oncology and Immunology, University Hospital of the Julius-Maximilians University , Würzburg, Germany
| | - Ana Maria Waaga-Gasser
- 3 Department of Surgery I, Molecular Oncology and Immunology, University Hospital of the Julius-Maximilians University , Würzburg, Germany
| | - Heike Walles
- 1 Institute of Tissue Engineering and Regenerative Medicine (TERM), University Hospital of the Julius-Maximilians University , Würzburg, Germany .,2 Translational Center Würzburg "Regenerative Therapies in Oncology and Musculoskeletal Disease, " Fraunhofer Institute Interfacial Engineering and Biotechnology IGB , Würzburg, Germany
| | - Gudrun Dandekar
- 1 Institute of Tissue Engineering and Regenerative Medicine (TERM), University Hospital of the Julius-Maximilians University , Würzburg, Germany .,2 Translational Center Würzburg "Regenerative Therapies in Oncology and Musculoskeletal Disease, " Fraunhofer Institute Interfacial Engineering and Biotechnology IGB , Würzburg, Germany
| |
Collapse
|
21
|
Batchelder CA, Martinez ML, Duru N, Meyers FJ, Tarantal AF. Three Dimensional Culture of Human Renal Cell Carcinoma Organoids. PLoS One 2015; 10:e0136758. [PMID: 26317980 PMCID: PMC4552551 DOI: 10.1371/journal.pone.0136758] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 08/07/2015] [Indexed: 12/22/2022] Open
Abstract
Renal cell carcinomas arise from the nephron but are heterogeneous in disease biology, clinical behavior, prognosis, and response to systemic therapy. Development of patient-specific in vitro models that efficiently and faithfully reproduce the in vivo phenotype may provide a means to develop personalized therapies for this diverse carcinoma. Studies to maintain and model tumor phenotypes in vitro were conducted with emerging three-dimensional culture techniques and natural scaffolding materials. Human renal cell carcinomas were individually characterized by histology, immunohistochemistry, and quantitative PCR to establish the characteristics of each tumor. Isolated cells were cultured on renal extracellular matrix and compared to a novel polysaccharide scaffold to assess cell-scaffold interactions, development of organoids, and maintenance of gene expression signatures over time in culture. Renal cell carcinomas cultured on renal extracellular matrix repopulated tubules or vessel lumens in renal pyramids and medullary rays, but cells were not observed in glomeruli or outer cortical regions of the scaffold. In the polysaccharide scaffold, renal cell carcinomas formed aggregates that were loosely attached to the scaffold or free-floating within the matrix. Molecular analysis of cell-scaffold constructs including immunohistochemistry and quantitative PCR demonstrated that individual tumor phenotypes could be sustained for up to 21 days in culture on both scaffolds, and in comparison to outcomes in two-dimensional monolayer cultures. The use of three-dimensional scaffolds to engineer a personalized in vitro renal cell carcinoma model provides opportunities to advance understanding of this disease.
Collapse
Affiliation(s)
- Cynthia A. Batchelder
- California National Primate Research Center, University of California Davis, Davis, CA, United States of America
| | - Michele L. Martinez
- California National Primate Research Center, University of California Davis, Davis, CA, United States of America
| | - Nadire Duru
- California National Primate Research Center, University of California Davis, Davis, CA, United States of America
| | - Frederick J. Meyers
- Department of Internal Medicine, School of Medicine, University of California Davis, Davis, CA, United States of America
- Comprehensive Cancer Center, University of California Davis, Davis, CA, United States of America
| | - Alice F. Tarantal
- California National Primate Research Center, University of California Davis, Davis, CA, United States of America
- Comprehensive Cancer Center, University of California Davis, Davis, CA, United States of America
- Department of Pediatrics, School of Medicine, University of California Davis, Davis, CA, United States of America
- Department of Cell Biology and Human Anatomy, School of Medicine, University of California Davis, Davis, CA, United States of America
- * E-mail:
| |
Collapse
|