1
|
Alver CG, Dominguez-Bendala J, Agarwal A. Engineered tools to study endocrine dysfunction of pancreas. BIOPHYSICS REVIEWS 2024; 5:041303. [PMID: 39449867 PMCID: PMC11498943 DOI: 10.1063/5.0220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 09/04/2024] [Indexed: 10/26/2024]
Abstract
Pancreas, a vital organ with intricate endocrine and exocrine functions, is central to the regulation of the body's glucose levels and digestive processes. Disruptions in its endocrine functions, primarily regulated by islets of Langerhans, can lead to debilitating diseases such as diabetes mellitus. Murine models of pancreatic dysfunction have contributed significantly to the understanding of insulitis, islet-relevant immunological responses, and the optimization of cell therapies. However, genetic differences between mice and humans have severely limited their clinical translational relevance. Recent advancements in tissue engineering and microfabrication have ushered in a new era of in vitro models that offer a promising solution. This paper reviews the state-of-the-art engineered tools designed to study endocrine dysfunction of the pancreas. Islet on a chip devices that allow precise control of various culture conditions and noninvasive readouts of functional outcomes have led to the generation of physiomimetic niches for primary and stem cell derived islets. Live pancreatic slices are a new experimental tool that could more comprehensively recapitulate the complex cellular interplay between the endocrine and exocrine parts of the pancreas. Although a powerful tool, live pancreatic slices require more complex control over their culture parameters such as local oxygenation and continuous removal of digestive enzymes and cellular waste products for maintaining experimental functionality over long term. The combination of islet-immune and slice on chip strategies can guide the path toward the next generation of pancreatic tissue modeling for better understanding and treatment of endocrine pancreatic dysfunctions.
Collapse
Affiliation(s)
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Ashutosh Agarwal
- Author to whom correspondence should be addressed:. Tel.: +1 305 243-8925
| |
Collapse
|
2
|
Payasi A, Yadav MK, Chaudhary S, Aggarwal A. Evaluating nephrotoxicity reduction in a novel polymyxin B formulation: insights from a 3D kidney-on-a-chip model. Antimicrob Agents Chemother 2024; 68:e0021924. [PMID: 39225483 PMCID: PMC11459911 DOI: 10.1128/aac.00219-24] [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: 02/09/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
This study aimed to assess the nephrotoxicity associated with VRP-034 (novel formulation of polymyxin B [PMB]) compared to marketed PMB in a three-dimensional (3D) kidney-on-a-chip model. To model the human kidney proximal tubule for analysis, tubular structures were established using 23 triple-channel chips seeded with RPTEC/hTERT1 cells. These cells were exposed to VRP-034 or PMB at seven concentrations (1-200 µM) over 12, 24, and 48 h. A suite of novel kidney injury biomarkers, cell health, and inflammatory markers were quantitatively assessed in the effluent. Additionally, caspase and cytochrome C levels were measured, and cell viability was evaluated using calcein AM and ethidium homodimer-1 (EthD-1). Exposure to marketed PMB resulted in significantly elevated levels (P < 0.05) of four key biomarkers (KIM-1, cystatin C, clusterin, and OPN) compared to VRP-034, particularly at clinically relevant concentrations of ≥10 µM. At 25 µM, all biomarkers demonstrated a significant increase (P < 0.05) with marketed PMB exposure compared to VRP-034. Inflammatory markers (interleukin-6 and interleukin-8) increased significantly (P < 0.05) with marketed PMB at concentrations of ≥5 µM, relative to VRP-034. VRP-034 displayed superior cell health outcomes, exhibiting lower lactate dehydrogenase release, while ATP levels remained comparable. Morphological analysis revealed that marketed PMB induced more severe damage, disrupting tubular integrity. Both treatments activated cytochrome C, caspase-3, caspase-8, caspase-9, and caspase-12 in a concentration-dependent manner; however, caspase activation was significantly reduced (P < 0.05) with VRP-034. This study demonstrates that VRP-034 significantly reduces nephrotoxicity compared to marketed PMB within a 3D microphysiological system, suggesting its potential to enable the use of full therapeutic doses of PMB with an improved safety profile, addressing the need for less nephrotoxic polymyxin antibiotics.
Collapse
Affiliation(s)
- Anurag Payasi
- Department of Cell Culture, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| | - Manoj Kumar Yadav
- Department of Cell Culture, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| | | | - Anmol Aggarwal
- Department of Pipeline Strategy, Venus Medicine Research Centre, Baddi, Himachal Pradesh, India
| |
Collapse
|
3
|
Alver CG, Drabbe E, Ishahak M, Agarwal A. Roadblocks confronting widespread dissemination and deployment of Organs on Chips. Nat Commun 2024; 15:5118. [PMID: 38879554 PMCID: PMC11180125 DOI: 10.1038/s41467-024-48864-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 05/16/2024] [Indexed: 06/19/2024] Open
Abstract
Organ on Chip platforms hold significant promise as alternatives to animal models or traditional cell cultures, both of which poorly recapitulate human pathophysiology and human level responses. Within the last 15 years, we have witnessed seminal scientific developments from academic laboratories, a flurry of startups and investments, and a genuine interest from pharmaceutical industry as well as regulatory authorities to translate these platforms. This Perspective identifies several fundamental design and process features that may act as roadblocks that prevent widespread dissemination and deployment of these systems, and provides a roadmap to help position this technology in mainstream drug discovery.
Collapse
Affiliation(s)
- Charles G Alver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Emma Drabbe
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
| | - Matthew Ishahak
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
4
|
Kantawala B, Emir Hamitoglu A, Nohra L, Abdullahi Yusuf H, Jonathan Isaac K, Shariff S, Nazir A, Soju K, Yenkoyan K, Wojtara M, Uwishema O. Microengineered neuronal networks: enhancing brain-machine interfaces. Ann Med Surg (Lond) 2024; 86:3535-3542. [PMID: 38846893 PMCID: PMC11152794 DOI: 10.1097/ms9.0000000000002130] [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: 11/23/2023] [Accepted: 04/05/2024] [Indexed: 06/09/2024] Open
Abstract
The brain-machine interface (BMI), a crucial conduit between the human brain and computers, holds transformative potential for various applications in neuroscience. This manuscript explores the role of micro-engineered neuronal networks (MNNs) in advancing BMI technologies and their therapeutic applications. As the interdisciplinary collaboration intensifies, the need for innovative and user-friendly BMI technologies becomes paramount. A comprehensive literature review sourced from reputable databases (PubMed Central, Medline, EBSCOhost, and Google Scholar) aided in the foundation of the manuscript, emphasizing the pivotal role of MNNs. This study aims to synthesize and analyze the diverse facets of MNNs in the context of BMI technologies, contributing insights into neural processes, technological advancements, therapeutic potentials, and ethical considerations surrounding BMIs. MNNs, exemplified by dual-mode neural microelectrodes, offer a controlled platform for understanding complex neural processes. Through case studies, we showcase the pivotal role of MNNs in BMI innovation, addressing challenges, and paving the way for therapeutic applications. The integration of MNNs with BMI technologies marks a revolutionary stride in neuroscience, refining brain-computer interactions and offering therapeutic avenues for neurological disorders. Challenges, ethical considerations, and future trends in BMI research necessitate a balanced approach, leveraging interdisciplinary collaboration to ensure responsible and ethical advancements. Embracing the potential of MNNs is paramount for the betterment of individuals with neurological conditions and the broader community.
Collapse
Affiliation(s)
- Burhan Kantawala
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Neuroscience Laboratory, Cobrain Centre, Yerevan State Medical University named after Mkhitar Heratsi, Yerevan, Armenia
| | - Ali Emir Hamitoglu
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Faculty of Medicine, Namik Kemal University, Tekirdag, Turkey
| | - Lea Nohra
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Faculty of Medical Science, Lebanese University, Beirut, Lebanon
| | - Hassan Abdullahi Yusuf
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- College of Health science, Faculty of Clinical Sciences Bayero University Kano, Nigeria
| | - Kirumira Jonathan Isaac
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Faculty of Clinical Medicine and Dentistry, Kampala International University, Uganda
| | - Sanobar Shariff
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Neuroscience Laboratory, Cobrain Centre, Yerevan State Medical University named after Mkhitar Heratsi, Yerevan, Armenia
| | - Abubakar Nazir
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Department of Medicine, King Edward Medical University, Pakistan
| | - Kevin Soju
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
- Faculty of Medicine, Christian Medical College, Ludhiana, India
| | - Konstantin Yenkoyan
- Neuroscience Laboratory, Cobrain Centre, Yerevan State Medical University named after Mkhitar Heratsi, Yerevan, Armenia
- Department of Biochemistry, Yerevan State Medical University named after Mkhitar Heratsi, Yerevan, Armenia
| | - Magda Wojtara
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
| | - Olivier Uwishema
- Oli Health Magazine Organization, Research and Education, Kigali, Rwanda
| |
Collapse
|
5
|
Alver CG, Álvarez-Cubela S, Altilio I, Hutchison E, Warrner E, Viso ME, Vitale G, Oliver D, Pastori RL, Dominguez-Bendala J, Agarwal A. SliceChip: a benchtop fluidic platform for organotypic culture and serial assessment of human and rodent pancreatic slices. LAB ON A CHIP 2024; 24:1557-1572. [PMID: 38205530 PMCID: PMC10939771 DOI: 10.1039/d3lc00850a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Enzymatically isolated pancreatic islets are the most commonly used ex vivo testbeds for diabetes research. Recently, precision-cut living slices of human pancreas are emerging as an exciting alternative because they maintain the complex architecture of the endocrine and exocrine tissues, and do not suffer from the mechanical and chemical stress of enzymatic isolation. We report a fluidic pancreatic SliceChip platform with dynamic environmental controls that generates a warm, oxygenated, and bubble-free fluidic pathway across singular immobilized slices with continuous deliver of fresh media and the ability to perform repeat serial perfusion assessments. A degasser ensures the system remains bubble-free while systemic pressurization with compressed oxygen ensures slice medium remains adequately oxygenated. Computational modeling of perfusion and oxygen dynamics within SliceChip guide the system's physiomimetic culture conditions. Maintenance of the physiological glucose dependent insulin secretion profile across repeat perfusion assessments of individual pancreatic slices kept under physiological oxygen levels demonstrated the culture capacity of our platform. Fluorescent images acquired every 4 hours of transgenic murine pancreatic slices were reliably stable and recoverable over a 5 day period due to the inclusion of a 3D-printed bioinert metallic anchor that maintained slice position within the SliceChip. Our slice on a chip platform has the potential to expand the useability of human pancreatic slices for diabetes pathogenesis and the development of new therapeutic approaches, while also enabling organotypic culture and assessment of other tissue slices such as brain and patient tumors.
Collapse
Affiliation(s)
- Charles G Alver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Silvia Álvarez-Cubela
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Isabella Altilio
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Emily Hutchison
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Emma Warrner
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Mariana E Viso
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Giana Vitale
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - David Oliver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Ricardo L Pastori
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Juan Dominguez-Bendala
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Diabetes Research Institute, University of Miami Miller School of Medicine, Miami, FL, USA.
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, Miami, FL, USA
| |
Collapse
|
6
|
Xie Z, Chen M, Lian J, Wang H, Ma J. Glioblastoma-on-a-chip construction and therapeutic applications. Front Oncol 2023; 13:1183059. [PMID: 37503321 PMCID: PMC10368971 DOI: 10.3389/fonc.2023.1183059] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 05/16/2023] [Indexed: 07/29/2023] Open
Abstract
Glioblastoma (GBM) is the most malignant type of primary intracranial tumor with a median overall survival of only 14 months, a very poor prognosis and a recurrence rate of 90%. It is difficult to reflect the complex structure and function of the GBM microenvironment in vivo using traditional in vitro models. GBM-on-a-chip platforms can integrate biological or chemical functional units of a tumor into a chip, mimicking in vivo functions of GBM cells. This technology has shown great potential for applications in personalized precision medicine and GBM immunotherapy. In recent years, there have been efforts to construct GBM-on-a-chip models based on microfluidics and bioprinting. A number of research teams have begun to use GBM-on-a-chip models for the investigation of GBM progression mechanisms, drug candidates, and therapeutic approaches. This review first briefly discusses the use of microfluidics and bioprinting technologies for GBM-on-a-chip construction. Second, we classify non-surgical treatments for GBM in pre-clinical research into three categories (chemotherapy, immunotherapy and other therapies) and focus on the use of GBM-on-a-chip in research for each category. Last, we demonstrate that organ-on-a-chip technology in therapeutic field is still in its initial stage and provide future perspectives for research directions in the field.
Collapse
Affiliation(s)
| | | | | | | | - Jingyun Ma
- *Correspondence: Hongcai Wang, ; Jingyun Ma,
| |
Collapse
|
7
|
McDuffie D, Alver CG, Suthar B, Helm M, Oliver D, Burgess RA, Barr D, Thomas E, Agarwal A. Acrylic-based culture plate format perfusion device to establish liver endothelial-epithelial interface. LAB ON A CHIP 2023; 23:3106-3119. [PMID: 37313651 PMCID: PMC10351567 DOI: 10.1039/d3lc00382e] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Microphysiological Systems (MPSs) or organs-on-chips, are microfluidic devices used to model human physiology in vitro. Polydimethylsiloxane (PDMS) is the most widely used material for organs-on-chips due to its established fabrication methods and biocompatibility properties. However, non-specific binding of small molecules limits PDMS for drug screening applications. Here, we designed a novel acrylic-based MPS to capture the physiological architecture that is observed universally in tissues across the body: the endothelial-epithelial interface (EEI). To reconstruct the EEI biology, we designed a membrane-based chip that features endothelial cells on the underside of the membrane exposed to mechanical shear from the path of media flow, and epithelial cells on the opposite side of the membrane protected from flow, as they are in vivo. We used a liver model with a hepatic progenitor cell line and human umbilical vein endothelial cells to assess the biological efficacy of the MPS. We computationally modeled the physics that govern the function of perfusion through the MPS. Empirically, efficacy was measured by comparing differentiation of the hepatic progenitor cells between the MPS and 2D culture conditions. We demonstrated that the MPS significantly improved hepatocyte differentiation, increased extracellular protein transport, and raised hepatocyte sensitivity to drug treatment. Our results strongly suggest that physiological perfusion has a profound effect on proper hepatocyte function, and the modular chip design motivates opportunities for future study of multi-organ interactions.
Collapse
Affiliation(s)
- Dennis McDuffie
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Charles G Alver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Bhumi Suthar
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - Madeline Helm
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - David Oliver
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
| | - R Alan Burgess
- Department of Pathology & Laboratory Medicine, Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - David Barr
- Department of Pathology & Laboratory Medicine, Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Emmanuel Thomas
- Department of Pathology & Laboratory Medicine, Schiff Center for Liver Diseases, University of Miami Miller School of Medicine, Miami, FL, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, University of Miami, Coral Gables, FL, USA.
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, USA
- Desai Sethi Urology Institute, University of Miami Miller School of Medicine, USA
| |
Collapse
|
8
|
Nguyen VVT, Gkouzioti V, Maass C, Verhaar MC, Vernooij RWM, van Balkom BWM. A systematic review of kidney-on-a-chip-based models to study human renal (patho-)physiology. Dis Model Mech 2023; 16:dmm050113. [PMID: 37334839 DOI: 10.1242/dmm.050113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 05/04/2023] [Indexed: 06/21/2023] Open
Abstract
As kidney diseases affect ∼10% of the world population, understanding the underlying mechanisms and developing therapeutic interventions are of high importance. Although animal models have enhanced knowledge of disease mechanisms, human (patho-)physiology may not be adequately represented in animals. Developments in microfluidics and renal cell biology have enabled the development of dynamic models to study renal (patho-)physiology in vitro. Allowing inclusion of human cells and combining different organ models, such as kidney-on-a-chip (KoC) models, enable the refinement and reduction of animal experiments. We systematically reviewed the methodological quality, applicability and effectiveness of kidney-based (multi-)organ-on-a-chip models, and describe the state-of-the-art, strengths and limitations, and opportunities regarding basic research and implementation of these models. We conclude that KoC models have evolved to complex models capable of mimicking systemic (patho-)physiological processes. Commercial chips and human induced pluripotent stem cells and organoids are important for KoC models to study disease mechanisms and assess drug effects, even in a personalized manner. This contributes to the Reduction, Refinement and Replacement of animal models for kidney research. A lack of reporting of intra- and inter-laboratory reproducibility and translational capacity currently hampers implementation of these models.
Collapse
Affiliation(s)
- Vivian V T Nguyen
- Department of Nephrology and Hypertension, UMC Utrecht, 3584CX Utrecht, The Netherlands
| | - Vasiliki Gkouzioti
- Department of Nephrology and Hypertension, UMC Utrecht, 3584CX Utrecht, The Netherlands
| | | | - Marianne C Verhaar
- Department of Nephrology and Hypertension, UMC Utrecht, 3584CX Utrecht, The Netherlands
| | - Robin W M Vernooij
- Department of Nephrology and Hypertension, UMC Utrecht, 3584CX Utrecht, The Netherlands
- Julius Center for Health Sciences and Primary Care, UMC Utrecht, Utrecht University, 3584CX Utrecht, The Netherlands
| | - Bas W M van Balkom
- Department of Nephrology and Hypertension, UMC Utrecht, 3584CX Utrecht, The Netherlands
| |
Collapse
|
9
|
Hou C, Gu Y, Yuan W, Zhang W, Xiu X, Lin J, Gao Y, Liu P, Chen X, Song L. Application of microfluidic chips in the simulation of the urinary system microenvironment. Mater Today Bio 2023; 19:100553. [PMID: 36747584 PMCID: PMC9898763 DOI: 10.1016/j.mtbio.2023.100553] [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: 11/22/2022] [Revised: 01/01/2023] [Accepted: 01/16/2023] [Indexed: 01/22/2023] Open
Abstract
The urinary system, comprising the kidneys, ureters, bladder, and urethra, has a unique mechanical and fluid microenvironment, which is essential to the urinary system growth and development. Microfluidic models, based on micromachining and tissue engineering technology, can integrate pathophysiological characteristics, maintain cell-cell and cell-extracellular matrix interactions, and accurately simulate the vital characteristics of human tissue microenvironments. Additionally, these models facilitate improved visualization and integration and meet the requirements of the laminar flow environment of the urinary system. However, several challenges continue to impede the development of a tissue microenvironment with controllable conditions closely resemble physiological conditions. In this review, we describe the biochemical and physical microenvironment of the urinary system and explore the feasibility of microfluidic technology in simulating the urinary microenvironment and pathophysiological characteristics in vitro. Moreover, we summarize the current research progress on adapting microfluidic chips for constructing the urinary microenvironment. Finally, we discuss the current challenges and suggest directions for future development and application of microfluidic technology in constructing the urinary microenvironment in vitro.
Collapse
Affiliation(s)
- Changhao Hou
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Yubo Gu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Wei Yuan
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Wukai Zhang
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xianjie Xiu
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Jiahao Lin
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| | - Yue Gao
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Peichuan Liu
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiang Chen
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, Department of Micro/Nano Electronics, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Lujie Song
- Department of Urology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Shanghai Eastern Institute of Urologic Reconstruction, Shanghai, China
| |
Collapse
|
10
|
Fu A, Chang M, Zhu H, Liu H, Wu D, Zeng H. Air-blood barrier (ABB) on a chip. Trends Analyt Chem 2023. [DOI: 10.1016/j.trac.2023.116919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
11
|
Mansouri M, Ahmed A, Ahmad SD, McCloskey MC, Joshi IM, Gaborski TR, Waugh RE, McGrath JL, Day SW, Abhyankar VV. The Modular µSiM Reconfigured: Integration of Microfluidic Capabilities to Study In Vitro Barrier Tissue Models under Flow. Adv Healthc Mater 2022; 11:e2200802. [PMID: 35953453 PMCID: PMC9798530 DOI: 10.1002/adhm.202200802] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/01/2022] [Indexed: 01/28/2023]
Abstract
Microfluidic tissue barrier models have emerged to address the lack of physiological fluid flow in conventional "open-well" Transwell-like devices. However, microfluidic techniques have not achieved widespread usage in bioscience laboratories because they are not fully compatible with traditional experimental protocols. To advance barrier tissue research, there is a need for a platform that combines the key advantages of both conventional open-well and microfluidic systems. Here, a plug-and-play flow module is developed to introduce on-demand microfluidic flow capabilities to an open-well device that features a nanoporous membrane and live-cell imaging capabilities. The magnetic latching assembly of this design enables bi-directional reconfiguration and allows users to conduct an experiment in an open-well format with established protocols and then add or remove microfluidic capabilities as desired. This work also provides an experimentally-validated flow model to select flow conditions based on the experimental needs. As a proof-of-concept, flow-induced alignment of endothelial cells and the expression of shear-sensitive gene targets are demonstrated, and the different phases of neutrophil transmigration across a chemically stimulated endothelial monolayer under flow conditions are visualized. With these experimental capabilities, it is anticipated that both engineering and bioscience laboratories will adopt this reconfigurable design due to the compatibility with standard open-well protocols.
Collapse
Affiliation(s)
- Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Adeel Ahmed
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - S. Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Molly C. McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Indranil M. Joshi
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Thomas R. Gaborski
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Richard E. Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - James L. McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Steven W. Day
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Vinay V. Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| |
Collapse
|
12
|
McCloskey MC, Kasap P, Ahmad SD, Su SH, Chen K, Mansouri M, Ramesh N, Nishihara H, Belyaev Y, Abhyankar VV, Begolo S, Singer BH, Webb KF, Kurabayashi K, Flax J, Waugh RE, Engelhardt B, McGrath JL. The Modular µSiM: A Mass Produced, Rapidly Assembled, and Reconfigurable Platform for the Study of Barrier Tissue Models In Vitro. Adv Healthc Mater 2022; 11:e2200804. [PMID: 35899801 PMCID: PMC9580267 DOI: 10.1002/adhm.202200804] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 07/11/2022] [Indexed: 01/27/2023]
Abstract
Advanced in vitro tissue chip models can reduce and replace animal experimentation and may eventually support "on-chip" clinical trials. To realize this potential, however, tissue chip platforms must be both mass-produced and reconfigurable to allow for customized design. To address these unmet needs, an extension of the µSiM (microdevice featuring a silicon-nitride membrane) platform is introduced. The modular µSiM (m-µSiM) uses mass-produced components to enable rapid assembly and reconfiguration by laboratories without knowledge of microfabrication. The utility of the m-µSiM is demonstrated by establishing an hiPSC-derived blood-brain barrier (BBB) in bioengineering and nonengineering, brain barriers focused laboratories. In situ and sampling-based assays of small molecule diffusion are developed and validated as a measure of barrier function. BBB properties show excellent interlaboratory agreement and match expectations from literature, validating the m-µSiM as a platform for barrier models and demonstrating successful dissemination of components and protocols. The ability to quickly reconfigure the m-µSiM for coculture and immune cell transmigration studies through addition of accessories and/or quick exchange of components is then demonstrated. Because the development of modified components and accessories is easily achieved, custom designs of the m-µSiM shall be accessible to any laboratory desiring a barrier-style tissue chip platform.
Collapse
Affiliation(s)
- Molly C McCloskey
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Pelin Kasap
- Theodor Kocher Institute, University of Bern, Bern, 3012, Switzerland
- Graduate School of Cellular and Biomedical Sciences (GCB), University of Bern, Bern, 3012, Switzerland
| | - S Danial Ahmad
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Shiuan-Haur Su
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kaihua Chen
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Mehran Mansouri
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | - Natalie Ramesh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Hideaki Nishihara
- Theodor Kocher Institute, University of Bern, Bern, 3012, Switzerland
| | - Yury Belyaev
- Microscopy Imaging Center, University of Bern, Bern, 3012, Switzerland
| | - Vinay V Abhyankar
- Department of Biomedical Engineering, Rochester Institute of Technology, Rochester, NY, 14623, USA
| | | | - Benjamin H Singer
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kevin F Webb
- Optics & Photonics Research Group, Department of Electrical and Electronic Engineering, University of Nottingham, Nottingham, NG7 2RD, UK
| | - Katsuo Kurabayashi
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Jonathan Flax
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Richard E Waugh
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| | - Britta Engelhardt
- Theodor Kocher Institute, University of Bern, Bern, 3012, Switzerland
| | - James L McGrath
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, 14627, USA
| |
Collapse
|
13
|
Patel SN, Mathews CE, Chandler R, Stabler CL. The Foundation for Engineering a Pancreatic Islet Niche. Front Endocrinol (Lausanne) 2022; 13:881525. [PMID: 35600597 PMCID: PMC9114707 DOI: 10.3389/fendo.2022.881525] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 03/30/2022] [Indexed: 12/01/2022] Open
Abstract
Progress in diabetes research is hindered, in part, by deficiencies in current experimental systems to accurately model human pathophysiology and/or predict clinical outcomes. Engineering human-centric platforms that more closely mimic in vivo physiology, however, requires thoughtful and informed design. Summarizing our contemporary understanding of the unique and critical features of the pancreatic islet can inform engineering design criteria. Furthermore, a broad understanding of conventional experimental practices and their current advantages and limitations ensures that new models address key gaps. Improving beyond traditional cell culture, emerging platforms are combining diabetes-relevant cells within three-dimensional niches containing dynamic matrices and controlled fluidic flow. While highly promising, islet-on-a-chip prototypes must evolve their utility, adaptability, and adoptability to ensure broad and reproducible use. Here we propose a roadmap for engineers to craft biorelevant and accessible diabetes models. Concurrently, we seek to inspire biologists to leverage such tools to ask complex and nuanced questions. The progenies of such diabetes models should ultimately enable investigators to translate ambitious research expeditions from benchtop to the clinic.
Collapse
Affiliation(s)
- Smit N. Patel
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Clayton E. Mathews
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida, Gainesville, FL, United States
- Diabetes Institute, University of Florida, Gainesville, FL, United States
| | - Rachel Chandler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
| | - Cherie L. Stabler
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL, United States
- Diabetes Institute, University of Florida, Gainesville, FL, United States
| |
Collapse
|
14
|
Kumar V, Madhurakkat Perikamana SK, Tata A, Hoque J, Gilpin A, Tata PR, Varghese S. An In Vitro Microfluidic Alveolus Model to Study Lung Biomechanics. Front Bioeng Biotechnol 2022; 10:848699. [PMID: 35252157 PMCID: PMC8895303 DOI: 10.3389/fbioe.2022.848699] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 01/25/2022] [Indexed: 01/18/2023] Open
Abstract
The gas exchange units of the lung, the alveoli, are mechanically active and undergo cyclic deformation during breathing. The epithelial cells that line the alveoli contribute to lung function by reducing surface tension via surfactant secretion, which is highly influenced by the breathing-associated mechanical cues. These spatially heterogeneous mechanical cues have been linked to several physiological and pathophysiological states. Here, we describe the development of a microfluidically assisted lung cell culture model that incorporates heterogeneous cyclic stretching to mimic alveolar respiratory motions. Employing this device, we have examined the effects of respiratory biomechanics (associated with breathing-like movements) and strain heterogeneity on alveolar epithelial cell functions. Furthermore, we have assessed the potential application of this platform to model altered matrix compliance associated with lung pathogenesis and ventilator-induced lung injury. Lung microphysiological platforms incorporating human cells and dynamic biomechanics could serve as an important tool to delineate the role of alveolar micromechanics in physiological and pathological outcomes in the lung.
Collapse
Affiliation(s)
- Vardhman Kumar
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | | | - Aleksandra Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
| | - Jiaul Hoque
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States
| | - Anna Gilpin
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
| | - Purushothama Rao Tata
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, United States
- Regeneration Next, Duke University, Durham, NC, United States
| | - Shyni Varghese
- Department of Biomedical Engineering, Duke University, Durham, NC, United States
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, NC, United States
- Department of Mechanical Engineering and Material Science, Duke University, Durham, NC, United States
- *Correspondence: Shyni Varghese,
| |
Collapse
|
15
|
Nguyen N, Thurgood P, Sekar NC, Chen S, Pirogova E, Peter K, Baratchi S, Khoshmanesh K. Microfluidic models of the human circulatory system: versatile platforms for exploring mechanobiology and disease modeling. Biophys Rev 2021; 13:769-786. [PMID: 34777617 DOI: 10.1007/s12551-021-00815-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/22/2021] [Indexed: 02/07/2023] Open
Abstract
The human circulatory system is a marvelous fluidic system, which is very sensitive to biophysical and biochemical cues. The current animal and cell culture models do not recapitulate the functional properties of the human circulatory system, limiting our ability to fully understand the complex biological processes underlying the dysfunction of this multifaceted system. In this review, we discuss the unique ability of microfluidic systems to recapitulate the biophysical, biochemical, and functional properties of the human circulatory system. We also describe the remarkable capacity of microfluidic technologies for exploring the complex mechanobiology of the cardiovascular system, mechanistic studying of cardiovascular diseases, and screening cardiovascular drugs with the additional benefit of reducing the need for animal models. We also discuss opportunities for further advancement in this exciting field.
Collapse
Affiliation(s)
- Ngan Nguyen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Peter Thurgood
- School of Engineering, RMIT University, Melbourne, Australia
| | - Nadia Chandra Sekar
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | - Sheng Chen
- School of Engineering, RMIT University, Melbourne, Australia
| | - Elena Pirogova
- School of Engineering, RMIT University, Melbourne, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, Australia.,Department of Cardiometabolic Health, The University of Melbourne, Parkville, Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Australia
| | | |
Collapse
|
16
|
Williams S, Charest J, Pollak M, Subramanian BK. Bioengineering Strategies To Develop Podocyte Culture Systems. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:938-948. [PMID: 34541902 PMCID: PMC9419930 DOI: 10.1089/ten.teb.2021.0154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Unraveling the complex behavior of healthy and disease podocytes by analyzing the changes in their unique arrangement of foot processes, slit diaphragm and the 3D morphology is a long-standing goal in kidney-glomerular research. The complexities surrounding the podocytes' accessibility in animal models and growing evidence of differences between humans and animal systems have compelled researchers to look for alternate approaches to study podocyte behaviors. With the advent of bioengineered models, an increasingly powerful and diverse set of tools is available to develop novel podocyte culture systems. This review discusses the pertinence of various culture models of podocytes to study podocyte mechanisms in both normal physiology and disease conditions. While no one in vitro system comprehensively recapitulates podocytes' in vivo architecture, we emphasize how the existing systems can be exploited to answer targeted questions on podocyte structure and function. We highlight the distinct advantages and limitations of using these models to study podocyte behaviors and screen therapeutics. Finally, we discuss various considerations and potential engineering strategies for developing next-generation complex 3D culture models for studying podocyte behaviors in vitro.
Collapse
Affiliation(s)
- Sarah Williams
- Beth Israel Deaconess Medical Center, 1859, Boston, Massachusetts, United States;
| | - Joseph Charest
- Draper Laboratory, Biomedical Engineering, 555 Technology Square, Cambridge, Massachusetts, United States, 02139;
| | - Martin Pollak
- Beth Israel Deaconess Medical Center, 1859, Boston, Massachusetts, United States;
| | | |
Collapse
|
17
|
Neves ER, Harley BAC, Pedron S. Microphysiological systems to study tumor-stroma interactions in brain cancer. Brain Res Bull 2021; 174:220-229. [PMID: 34166771 PMCID: PMC8324563 DOI: 10.1016/j.brainresbull.2021.06.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 05/17/2021] [Accepted: 06/15/2021] [Indexed: 12/15/2022]
Abstract
Brain tumors still lack effective treatments, and the mechanisms of tumor progression and therapeutic resistance are unclear. Multiple parameters affect cancer prognosis (e.g., type and grade, age, location, size, and genetic mutations) and election of suitable treatments is based on preclinical models and clinical data. However, most candidate drugs fail in human trials due to inefficacy. Cell lines and tissue culture plates do not provide physiologically relevant environments, and animal models are not able to adequately mimic characteristics of disease in humans. Therefore, increasing technological advances are focusing on in vitro and computational modeling to increase the throughput and predicting capabilities of preclinical systems. The extensive use of these therapeutic agents requires a more profound understanding of the tumor-stroma interactions, including neural tissue, extracellular matrix, blood-brain barrier, astrocytes and microglia. Microphysiological brain tumor models offer physiologically relevant vascularized 'minitumors' that can help deciphering disease mechanisms, accelerating the drug discovery and predicting patient's response to anticancer treatments. This article reviews progress in tumor-on-a-chip platforms that are designed to comprehend the particular roles of stromal cells in the brain tumor microenvironment.
Collapse
Affiliation(s)
- Edward R Neves
- Department of Chemical and Biomolecular Engineering, Carl R Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Brendan A C Harley
- Department of Chemical and Biomolecular Engineering, Carl R Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Sara Pedron
- Department of Chemical and Biomolecular Engineering, Carl R Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
18
|
Archer F, Bobet-Erny A, Gomes M. State of the art on lung organoids in mammals. Vet Res 2021; 52:77. [PMID: 34078444 PMCID: PMC8170649 DOI: 10.1186/s13567-021-00946-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/04/2021] [Indexed: 02/08/2023] Open
Abstract
The number and severity of diseases affecting lung development and adult respiratory function have stimulated great interest in developing new in vitro models to study lung in different species. Recent breakthroughs in 3-dimensional (3D) organoid cultures have led to new physiological in vitro models that better mimic the lung than conventional 2D cultures. Lung organoids simulate multiple aspects of the real organ, making them promising and useful models for studying organ development, function and disease (infection, cancer, genetic disease). Due to their dynamics in culture, they can serve as a sustainable source of functional cells (biobanking) and be manipulated genetically. Given the differences between species regarding developmental kinetics, the maturation of the lung at birth, the distribution of the different cell populations along the respiratory tract and species barriers for infectious diseases, there is a need for species-specific lung models capable of mimicking mammal lungs as they are of great interest for animal health and production, following the One Health approach. This paper reviews the latest developments in the growing field of lung organoids.
Collapse
Affiliation(s)
- Fabienne Archer
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France.
| | - Alexandra Bobet-Erny
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France
| | - Maryline Gomes
- UMR754, IVPC, INRAE, EPHE, Univ Lyon, Université Claude Bernard Lyon 1, 69007, Lyon, France
| |
Collapse
|