1
|
Tusnim J, Kutuzov P, Grasman JM. In Vitro Models for Peripheral Nerve Regeneration. Adv Healthc Mater 2024; 13:e2401605. [PMID: 39324286 DOI: 10.1002/adhm.202401605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 08/14/2024] [Indexed: 09/27/2024]
Abstract
Peripheral nerve injury (PNI) resulting in lesions is highly prevalent clinically, but current therapeutic approaches fail to provide satisfactory outcomes in many patients. While peripheral nerves have intrinsic regenerative capacity, the regenerative capabilities of peripheral nerves are often insufficient to restore full functionality. This highlights an unmet need for developing more effective strategies to repair damaged peripheral nerves and improve regenerative success. Consequently, researchers are actively exploring a variety of therapeutic strategies, encompassing the local delivery of trophic factors or bioactive molecules, the design of advanced biomaterials that interact with regenerating axons, and augmentation with nerve guidance conduits or complex prostheses. However, clinical translation of these technologies remains limited, emphasizing the need for continued research on peripheral nerve regeneration modalities that can enhance functional restoration. Experimental models that accurately recapitulate key aspects of peripheral nerve injury and repair biology can accelerate therapeutic development by enabling systematic testing of new techniques. Advancing regenerative therapies for PNI requires bridging the gap between basic science discoveries and clinical application. This review discusses different in vitro models of peripheral nerve injury and repair, including their advantages, limitations, and potential applications.
Collapse
Affiliation(s)
- Jarin Tusnim
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Peter Kutuzov
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Jonathan M Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| |
Collapse
|
2
|
Martin MK, Rodriguez R, Guerrero G, Sheehan GD, Powell R, Klein AH, Bhattacharjee A. Pharmacologically enabling the degradation of NaV1.8 channels to reduce neuropathic pain. Pain 2024:00006396-990000000-00766. [PMID: 39514325 DOI: 10.1097/j.pain.0000000000003470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/14/2024] [Indexed: 11/16/2024]
Abstract
ABSTRACT In phase II clinical trials, NaV1.8 channels were identified as viable targets to treat acute pain. Results were modest, however, and NaV1.8 pore blockers must be given systemically, potentially leading to adverse effects, especially during prolonged use. A local, long-lasting approach is desirable, yet local anesthetics are neither specific nor long-lasting. In lieu of a pore blocker approach, we show a pharmacological method targeting the scaffolding and degradation of NaV1.8 channels, which attenuated neuropathic pain behavior in mice. NaV1.8 channels interact with the WW domain-containing scaffold protein called Magi-1. WW domains are typically found in ubiquitin ligases, and NaV1.8 channels are susceptible to degradation by ubiquitin ligases. Here, we show NaV1.8 and MAGI-1 colocalized in human tissues. We demonstrate that a lipidated peptide derived from the NaV1.8 WW binding domain, at sub-micromolar concentrations, inhibited rodent dorsal root ganglion neuronal firing. The peptide reduced NaV1.8 channel immunoreactivity and tetrodotoxin-resistant currents in human dorsal root ganglion neurons. We found that the lipidated peptide attenuated neuropathic pain behaviors in mice for multiple weeks after a single injection. Our results reveal that the NaV1.8-targeted lipidated peptide provides local and sustained analgesia, serving as a viable alternative to NaV1.8 pore blockers.
Collapse
Affiliation(s)
- Molly K Martin
- Program for Neuroscience, University at Buffalo-The State University of New York, Buffalo, NY, United States
| | - Raider Rodriguez
- Pharmacology and Toxicology, University at Buffalo-The State University of New York, Buffalo, NY, United States. Dr. Powell is now with the Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Giselle Guerrero
- Pharmacology and Toxicology, University at Buffalo-The State University of New York, Buffalo, NY, United States. Dr. Powell is now with the Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Garrett D Sheehan
- Program for Neuroscience, University at Buffalo-The State University of New York, Buffalo, NY, United States
| | - Rasheen Powell
- Pharmacology and Toxicology, University at Buffalo-The State University of New York, Buffalo, NY, United States. Dr. Powell is now with the Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| | - Amanda H Klein
- Program for Neuroscience, University at Buffalo-The State University of New York, Buffalo, NY, United States
- Pharmacology and Toxicology, University at Buffalo-The State University of New York, Buffalo, NY, United States. Dr. Powell is now with the Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
- Department of Pharmacy Practice and Pharmaceutical Sciences, University of Minnesota, Duluth, MN, United States
| | - Arin Bhattacharjee
- Program for Neuroscience, University at Buffalo-The State University of New York, Buffalo, NY, United States
- Pharmacology and Toxicology, University at Buffalo-The State University of New York, Buffalo, NY, United States. Dr. Powell is now with the Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, United States
| |
Collapse
|
3
|
Tyagi S, Higerd-Rusli GP, Akin EJ, Baker CA, Liu S, Dib-Hajj FB, Waxman SG, Dib-Hajj SD. Real-time imaging of axonal membrane protein life cycles. Nat Protoc 2024; 19:2771-2802. [PMID: 38831222 PMCID: PMC11721981 DOI: 10.1038/s41596-024-00997-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 02/12/2024] [Indexed: 06/05/2024]
Abstract
The construction of neuronal membranes is a dynamic process involving the biogenesis, vesicular packaging, transport, insertion and recycling of membrane proteins. Optical imaging is well suited for the study of protein spatial organization and transport. However, various shortcomings of existing imaging techniques have prevented the study of specific types of proteins and cellular processes. Here we describe strategies for protein tagging and labeling, cell culture and microscopy that enable the real-time imaging of axonal membrane protein trafficking and subcellular distribution as they progress through some stages of their life cycle. First, we describe a process for engineering membrane proteins with extracellular self-labeling tags (either HaloTag or SNAPTag), which can be labeled with fluorescent ligands of various colors and cell permeability, providing flexibility for investigating the trafficking and spatiotemporal regulation of multiple membrane proteins in neuronal compartments. Next, we detail the dissection, transfection and culture of dorsal root ganglion sensory neurons in microfluidic chambers, which physically compartmentalizes cell bodies and distal axons. Finally, we describe four labeling and imaging procedures that utilize these enzymatically tagged proteins, flexible fluorescent labels and compartmentalized neuronal cultures to study axonal membrane protein anterograde and retrograde transport, the cotransport of multiple proteins, protein subcellular localization, exocytosis and endocytosis. Additionally, we generated open-source software for analyzing the imaging data in a high throughput manner. The experimental and analysis workflows provide an approach for studying the dynamics of neuronal membrane protein homeostasis, addressing longstanding challenges in this area. The protocol requires 5-7 days and expertise in cell culture and microscopy.
Collapse
Affiliation(s)
- Sidharth Tyagi
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Grant P Higerd-Rusli
- Medical Scientist Training Program, Yale School of Medicine, New Haven, CT, USA
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Elizabeth J Akin
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
- Department of Pharmacology, University of Nevada, Reno School of Medicine, Reno, NV, USA
| | - Christopher A Baker
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Shujun Liu
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Fadia B Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA
| | - Stephen G Waxman
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA.
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA.
| | - Sulayman D Dib-Hajj
- Center for Neuroscience and Regeneration Research, West Haven, CT, USA.
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA.
- Center for Restoration of Nervous System Function, VA Connecticut Healthcare System, West Haven, CT, USA.
| |
Collapse
|
4
|
Kimourtzis G, Rangwani N, Jenkins BJ, Jani S, McNaughton PA, Raouf R. Prostaglandin E2 depolarises sensory axons in vitro in an ANO1 and Nav1.8 dependent manner. Sci Rep 2024; 14:17360. [PMID: 39075089 PMCID: PMC11286870 DOI: 10.1038/s41598-024-67793-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024] Open
Abstract
Prostaglandin E2 (PGE2) is a major contributor to inflammatory pain hyperalgesia, however, the extent to which it modulates the activity of nociceptive axons is incompletely understood. We developed and characterized a microfluidic cell culture model to investigate sensitisation of the axons of dorsal root ganglia neurons. We show that application of PGE2 to fluidically isolated axons leads to sensitisation of their responses to depolarising stimuli. Interestingly the application of PGE2 to the DRG axons elicited a direct and persistent spiking activity propagated to the soma. Both the persistent activity and the membrane depolarisation in the axons are abolished by the EP4 receptor inhibitor and a blocker of cAMP synthesis. Further investigated into the mechanisms of the spiking activity showed that the PGE2 evoked depolarisation was inhibited by Nav1.8 sodium channel blockers but was refractory to the application of TTX or zatebradine. Interestingly, the depolarisation of axons was blocked by blocking ANO1 channels with T16Ainh-A01. We further show that PGE2-elicited axonal responses are altered by the changes in chloride gradient within the axons following treatment with bumetanide a Na-K-2Cl cotransporter NKCC1 inhibitor, but not by VU01240551 an inhibitor of potassium-chloride transporter KCC2. Our data demonstrate a novel role for PGE2/EP4/cAMP pathway which culminates in a sustained depolarisation of sensory axons mediated by a chloride current through ANO1 channels. Therefore, using a microfluidic culture model, we provide evidence for a potential dual function of PGE2 in inflammatory pain: it sensitises depolarisation-evoked responses in nociceptive axons and directly triggers action potentials by activating ANO1 and Nav1.8 channels.
Collapse
Affiliation(s)
- Georgios Kimourtzis
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, UK
| | - Natasha Rangwani
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, UK
| | - Bethan J Jenkins
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, UK
| | - Siddharth Jani
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, UK
| | - Peter A McNaughton
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, UK
| | - Ramin Raouf
- Wolfson Sensory, Pain and Regeneration Centre (SPaRC), Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, UK.
| |
Collapse
|
5
|
Buentello DC, Garcia-Corral M, Trujillo-de Santiago G, Alvarez MM. Neuron(s)-on-a-Chip: A Review of the Design and Use of Microfluidic Systems for Neural Tissue Culture. IEEE Rev Biomed Eng 2024; 17:243-263. [PMID: 36301779 DOI: 10.1109/rbme.2022.3217486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neuron-on-chip (NoC) systems-microfluidic devices in which neurons are cultured-have become a promising alternative to replace or minimize the use of animal models and have greatly facilitated in vitro research. Here, we review and discuss current developments in neuron-on-chip platforms, with a particular emphasis on existing biological models, culturing techniques, biomaterials, and topologies. We also discuss how the architecture, flow, and gradients affect neuronal growth, differentiation, and development. Finally, we discuss some of the most recent applications of NoCs in fundamental research (i.e., studies on the effects of electrical, mechanical/topological, or chemical stimuli) and in disease modeling.
Collapse
|
6
|
Kimourtzis G, Raouf R. A microfluidic model of the first sensory synapse for analgesic target discovery. Mol Pain 2024; 20:17448069241293286. [PMID: 39415077 PMCID: PMC11565614 DOI: 10.1177/17448069241293286] [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: 07/10/2024] [Revised: 09/04/2024] [Accepted: 09/27/2024] [Indexed: 10/18/2024] Open
Abstract
The synaptic connections between dorsal root ganglia (DRG) and dorsal horn (DH) neurons are a crucial relay point for the transmission of painful stimuli. To delineate how synaptic plasticity may modulate the excitability of DH neurons, we have devised a microfluidic co-culture model that recapitulates the first sensory synapse using postnatal mouse sensory neurons. We show that DRG-DH co-cultures characterize salient features of the in vivo physiology of sensory neurons. Immunocytcochemical experiments of the cultured DH neurons show a co-localization of Map2 with VGlut2 and of Map2 with Synapsin 1, corroborating the glutamatergic identity of the DH neurons and further suggesting the potential formation of active synapses in this neuronal set. Fluorometric imaging experiments demonstrate the elicitation of calcium responses in DH neurons following the stimulation of DRG cell bodies or axons. Selective NMDA and AMPA receptor blockade appreciably silences DH neuron responses, suggesting that glutamatergic signaling is maintained in vitro. Last, a surrogate model of peripheral nerve injury is introduced in the form of an axotomy, which results in elevated and prolonged calcium responses of DH neurons. Overall, the microfluidic mouse co-cultures provide a method advancement in the study of periphery-to-center pain signaling, where the potential of utilizing the platform for drug target identification is underscored.
Collapse
Affiliation(s)
- Georgios Kimourtzis
- Wolfson Centre for Age-Related Diseases, Institute of Psychology, Psychiatry & Neuroscience, King’s College London, London, UK
- Department of Neurobiology, Harvard Medical School, Boston, MA, USA
- F.M. Kirby Neurobiology Center, Boston Children’s Hospital, Boston, MA, USA
| | - Ramin Raouf
- Wolfson Centre for Age-Related Diseases, Institute of Psychology, Psychiatry & Neuroscience, King’s College London, London, UK
| |
Collapse
|
7
|
Kandhavivorn W, Glaß H, Herrmannsdörfer T, Böckers TM, Uhlarz M, Gronemann J, Funk RHW, Pietzsch J, Pal A, Hermann A. Restoring Axonal Organelle Motility and Regeneration in Cultured FUS-ALS Motoneurons through Magnetic Field Stimulation Suggests an Alternative Therapeutic Approach. Cells 2023; 12:1502. [PMID: 37296623 PMCID: PMC10252208 DOI: 10.3390/cells12111502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 05/17/2023] [Accepted: 05/22/2023] [Indexed: 06/12/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating motoneuron disease characterized by sustained loss of neuromuscular junctions, degenerating corticospinal motoneurons and rapidly progressing muscle paralysis. Motoneurons have unique features, essentially a highly polarized, lengthy architecture of axons, posing a considerable challenge for maintaining long-range trafficking routes for organelles, cargo, mRNA and secretion with a high energy effort to serve crucial neuronal functions. Impaired intracellular pathways implicated in ALS pathology comprise RNA metabolism, cytoplasmic protein aggregation, cytoskeletal integrity for organelle trafficking and maintenance of mitochondrial morphology and function, cumulatively leading to neurodegeneration. Current drug treatments only have marginal effects on survival, thereby calling for alternative ALS therapies. Exposure to magnetic fields, e.g., transcranial magnetic stimulations (TMS) on the central nervous system (CNS), has been broadly explored over the past 20 years to investigate and improve physical and mental activities through stimulated excitability as well as neuronal plasticity. However, studies of magnetic treatments on the peripheral nervous system are still scarce. Thus, we investigated the therapeutic potential of low frequency alternating current magnetic fields on cultured spinal motoneurons derived from induced pluripotent stem cells of FUS-ALS patients and healthy persons. We report a remarkable restoration induced by magnetic stimulation on axonal trafficking of mitochondria and lysosomes and axonal regenerative sprouting after axotomy in FUS-ALS in vitro without obvious harmful effects on diseased and healthy neurons. These beneficial effects seem to derive from improved microtubule integrity. Thus, our study suggests the therapeutic potential of magnetic stimulations in ALS, which awaits further exploration and validation in future long-term in vivo studies.
Collapse
Affiliation(s)
- Wonphorn Kandhavivorn
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
- Institute of Anatomy, Technische Universität Dresden, D-01307 Dresden, Germany
| | - Hannes Glaß
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, D-01307 Dresden, Germany;
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, D-18147 Rostock, Germany
| | - Thomas Herrmannsdörfer
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
| | - Tobias M. Böckers
- Institute of Anatomy and Cell Biology, University of Ulm, D-89081 Ulm, Germany;
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Ulm, D-89081 Ulm, Germany
| | - Marc Uhlarz
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
| | - Jonas Gronemann
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
| | - Richard H. W. Funk
- Institute of Anatomy, Technische Universität Dresden, D-01307 Dresden, Germany
- Dresden International University, D-01067 Dresden, Germany
| | - Jens Pietzsch
- Department of Radiopharmaceutical and Chemical Biology, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany
- Faculty of Chemistry and Food Chemistry, School of Science, Technische Universität Dresden, D-01069 Dresden, Germany
| | - Arun Pal
- Dresden High Magnetic Field Laboratory (HLD-EMFL), Helmholtz-Zentrum Dresden-Rossendorf, D-01328 Dresden, Germany; (W.K.); (T.H.); (M.U.); (J.G.)
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, D-01307 Dresden, Germany;
| | - Andreas Hermann
- Division for Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, D-01307 Dresden, Germany;
- Translational Neurodegeneration Section “Albrecht Kossel”, Department of Neurology, University Medical Center Rostock, University of Rostock, D-18147 Rostock, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) Rostock/Greifswald, D-18147 Rostock, Germany
- Center for Transdisciplinary Neurosciences Rostock (CTNR), University Medical Center Rostock, University of Rostock, D-18147 Rostock, Germany
| |
Collapse
|
8
|
Suzuki I, Matsuda N, Han X, Noji S, Shibata M, Nagafuku N, Ishibashi Y. Large-Area Field Potential Imaging Having Single Neuron Resolution Using 236 880 Electrodes CMOS-MEA Technology. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023:e2207732. [PMID: 37088859 PMCID: PMC10369302 DOI: 10.1002/advs.202207732] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 03/21/2023] [Indexed: 05/03/2023]
Abstract
The electrophysiological technology having a high spatiotemporal resolution at the single-cell level and noninvasive measurements of large areas provide insights on underlying neuronal function. Here, a complementary metal-oxide semiconductor (CMOS)-microelectrode array (MEA) is used that uses 236 880 electrodes each with an electrode size of 11.22 × 11.22 µm and 236 880 covering a wide area of 5.5 × 5.9 mm in presenting a detailed and single-cell-level neural activity analysis platform for brain slices, human iPS cell-derived cortical networks, peripheral neurons, and human brain organoids. Propagation pattern characteristics between brain regions changes the synaptic propagation into compounds based on single-cell time-series patterns, classification based on single DRG neuron firing patterns and compound responses, axonal conduction characteristics and changes to anticancer drugs, and network activities and transition to compounds in brain organoids are extracted. This detailed analysis of neural activity at the single-cell level using the CMOS-MEA provides a new understanding of the basic mechanisms of brain circuits in vitro and ex vivo, on human neurological diseases for drug discovery, and compound toxicity assessment.
Collapse
Affiliation(s)
- Ikuro Suzuki
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Naoki Matsuda
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Xiaobo Han
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Shuhei Noji
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Mikako Shibata
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Nami Nagafuku
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| | - Yuto Ishibashi
- Department of Electronics, Graduate School of Engineering, Tohoku Institute of Technology, 35-1 Yagiyama Kasumicho, Taihaku-ku, Sendai, Miyagi, 982-8577, Japan
| |
Collapse
|
9
|
Giorgi S, Lamberti A, Butrón L, Gross-Amat O, Alarcón-Alarcón D, Rodríguez-Cañas E, Fernández-Carvajal A, Ferrer-Montiel A. Compartmentalized primary cultures of dorsal root ganglion neurons to model peripheral pathophysiological conditions. Mol Pain 2023; 19:17448069231197102. [PMID: 37578145 PMCID: PMC10521292 DOI: 10.1177/17448069231197102] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/24/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023] Open
Abstract
Neurosensory disorders such as pain and pruritus remain a major health problem greatly impacting the quality of life, and often increasing the risk of mortality. Current pre-clinical models to investigate dysfunction of sensory neurons have shown a limited clinical translation, in part, by failing to mimic the compartmentalized nociceptor anatomy that exhibits a central compartment containing the soma and a peripheral one harboring the axon endings with distinct molecular and cellular environmental composition. Thus, there is a need to validate compartmentalized preclinical neurosensory models for investigating the pathophysiology of peripheral sensory disorders and to test drug candidates. Here, we have addressed this issue and developed a microfluidic-based preclinical nociceptor model and validated it for investigating inflammatory and neuropathic peripheral disorders. We show that this model reproduces the peripheral sensitization and resolution produced by an inflammatory soup and by the chemotherapeutic drug paclitaxel. Furthermore, compartmentalized nociceptor primary cultures were amenable to co-culture with keratinocytes in the axonal compartment. Interaction of axonal endings with keratinocytes modulated neuronal responses, consistent with a crosstalk between both cell types. These findings pave the way towards translational pre-clinical sensory models for skin pathophysiological research and drug development.
Collapse
Affiliation(s)
- Simona Giorgi
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Angela Lamberti
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Laura Butrón
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Olivia Gross-Amat
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - David Alarcón-Alarcón
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Enrique Rodríguez-Cañas
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Asia Fernández-Carvajal
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| | - Antonio Ferrer-Montiel
- Instituto de Investigación, Desarrollo e Innovación en Biotecnología Sanitaria de Elche (IDiBE), Universitas Miguel Hernández, Elche, Spain
| |
Collapse
|
10
|
Mießner H, Seidel J, Smith ESJ. In vitro models for investigating itch. Front Mol Neurosci 2022; 15:984126. [PMID: 36385768 PMCID: PMC9644192 DOI: 10.3389/fnmol.2022.984126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Itch (pruritus) is a sensation that drives a desire to scratch, a behavior observed in many animals. Although generally short-lasting and not causing harm, there are several pathological conditions where chronic itch is a hallmark symptom and in which prolonged scratching can induce damage. Finding medications to counteract the sensation of chronic itch has proven difficult due to the molecular complexity that involves a multitude of triggers, receptors and signaling pathways between skin, immune and nerve cells. While much has been learned about pruritus from in vivo animal models, they have limitations that corroborate the necessity for a transition to more human disease-like models. Also, reducing animal use should be encouraged in research. However, conducting human in vivo experiments can also be ethically challenging. Thus, there is a clear need for surrogate models to be used in pre-clinical investigation of the mechanisms of itch. Most in vitro models used for itch research focus on the use of known pruritogens. For this, sensory neurons and different types of skin and/or immune cells are stimulated in 2D or 3D co-culture, and factors such as neurotransmitter or cytokine release can be measured. There are however limitations of such simplistic in vitro models. For example, not all naturally occurring cell types are present and there is also no connection to the itch-sensing organ, the central nervous system (CNS). Nevertheless, in vitro models offer a chance to investigate otherwise inaccessible specific cell–cell interactions and molecular pathways. In recent years, stem cell-based approaches and human primary cells have emerged as viable alternatives to standard cell lines or animal tissue. As in vitro models have increased in their complexity, further opportunities for more elaborated means of investigating itch have been developed. In this review, we introduce the latest concepts of itch and discuss the advantages and limitations of current in vitro models, which provide valuable contributions to pruritus research and might help to meet the unmet clinical need for more refined anti-pruritic substances.
Collapse
Affiliation(s)
- Hendrik Mießner
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Judith Seidel
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Ewan St. John Smith,
| |
Collapse
|
11
|
In Vitro Sensitive Skin Models: Review of the Standard Methods and Introduction to a New Disruptive Technology. COSMETICS 2022. [DOI: 10.3390/cosmetics9040067] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The skin is a protective organ, able to decode a wide range of tactile, thermal, or noxious stimuli. Some of the sensors belonging to the transient receptor potential (TRP) family, for example, TRPV1, can elicit capsaicin-induced heat pain or histamine-induced itching sensations. The sensory nerve fibers, whose soma is located in the trigeminal or the dorsal root ganglia, are able to carry signals from the skin’s sensory receptors toward the brain via the spinal cord. In some cases, in response to environmental factors, nerve endings might be hyper activated, leading to a sensitive skin syndrome (SSS). SSS affects about 50% of the population and is correlated with small-fiber neuropathies resulting in neuropathic pain. Thus, for cosmetical and pharmaceutical industries developing SSS treatments, the selection of relevant and predictive in vitro models is essential. In this article, we reviewed the different in vitro models developed for the assessment of skin and neuron interactions. In a second part, we presented the advantages of microfluidic devices and organ-on-chip models, with a focus on the first model we developed in this context.
Collapse
|
12
|
Varier P, Raju G, Madhusudanan P, Jerard C, Shankarappa SA. A Brief Review of In Vitro Models for Injury and Regeneration in the Peripheral Nervous System. Int J Mol Sci 2022; 23:816. [PMID: 35055003 PMCID: PMC8775373 DOI: 10.3390/ijms23020816] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Revised: 12/03/2021] [Accepted: 12/05/2021] [Indexed: 02/06/2023] Open
Abstract
Nerve axonal injury and associated cellular mechanisms leading to peripheral nerve damage are important topics of research necessary for reducing disability and enhancing quality of life. Model systems that mimic the biological changes that occur during human nerve injury are crucial for the identification of cellular responses, screening of novel therapeutic molecules, and design of neural regeneration strategies. In addition to in vivo and mathematical models, in vitro axonal injury models provide a simple, robust, and reductionist platform to partially understand nerve injury pathogenesis and regeneration. In recent years, there have been several advances related to in vitro techniques that focus on the utilization of custom-fabricated cell culture chambers, microfluidic chamber systems, and injury techniques such as laser ablation and axonal stretching. These developments seem to reflect a gradual and natural progression towards understanding molecular and signaling events at an individual axon and neuronal-soma level. In this review, we attempt to categorize and discuss various in vitro models of injury relevant to the peripheral nervous system and highlight their strengths, weaknesses, and opportunities. Such models will help to recreate the post-injury microenvironment and aid in the development of therapeutic strategies that can accelerate nerve repair.
Collapse
Affiliation(s)
| | | | | | | | - Sahadev A. Shankarappa
- Centre for Nanosciences & Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham, Kochi 682041, India; (P.V.); (G.R.); (P.M.); (C.J.)
| |
Collapse
|
13
|
Mateus JC, Lopes C, Aroso M, Costa AR, Gerós A, Meneses J, Faria P, Neto E, Lamghari M, Sousa MM, Aguiar P. Bidirectional flow of action potentials in axons drives activity dynamics in neuronal cultures. J Neural Eng 2021; 18. [PMID: 34891149 DOI: 10.1088/1741-2552/ac41db] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 12/10/2021] [Indexed: 12/20/2022]
Abstract
Objective. Recent technological advances are revealing the complex physiology of the axon and challenging long-standing assumptions. Namely, while most action potential (AP) initiation occurs at the axon initial segment in central nervous system neurons, initiation in distal parts of the axon has been reported to occur in both physiological and pathological conditions. The functional role of these ectopic APs, if exists, is still not clear, nor its impact on network activity dynamics.Approach. Using an electrophysiology platform specifically designed for assessing axonal conduction we show here for the first time regular and effective bidirectional axonal conduction in hippocampal and dorsal root ganglia cultures. We investigate and characterize this bidirectional propagation both in physiological conditions and after distal axotomy.Main results.A significant fraction of APs are not coming from the canonical synapse-dendrite-soma signal flow, but instead from signals originating at the distal axon. Importantly, antidromic APs may carry information and can have a functional impact on the neuron, as they consistently depolarize the soma. Thus, plasticity or gene transduction mechanisms triggered by soma depolarization can also be affected by these antidromic APs. Conduction velocity is asymmetrical, with antidromic conduction being slower than orthodromic.Significance.Altogether these findings have important implications for the study of neuronal functionin vitro, reshaping our understanding on how information flows in neuronal cultures.
Collapse
Affiliation(s)
- J C Mateus
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Cdf Lopes
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - M Aroso
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - A R Costa
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - A Gerós
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.,FEUP-Faculdade de Engenharia da Universidade do Porto, Porto, Portugal
| | - J Meneses
- CDRSP-IPL-Centre for Rapid and Sustainable Product Development-Instituto Politécnico de Leiria, Marinha Grande, Portugal.,IBEB-Instituto de Biofísica e Engenharia Biomédica, Faculdade de Ciências, Universidade de Lisboa, Lisboa, Portugal
| | - P Faria
- CDRSP-IPL-Centre for Rapid and Sustainable Product Development-Instituto Politécnico de Leiria, Marinha Grande, Portugal
| | - E Neto
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - M Lamghari
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - M M Sousa
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - P Aguiar
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| |
Collapse
|
14
|
Atmaramani R, Veeramachaneni S, Mogas LV, Koppikar P, Black BJ, Hammack A, Pancrazio JJ, Granja-Vazquez R. Investigating the Function of Adult DRG Neuron Axons Using an In Vitro Microfluidic Culture System. MICROMACHINES 2021; 12:mi12111317. [PMID: 34832729 PMCID: PMC8621475 DOI: 10.3390/mi12111317] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/19/2021] [Accepted: 10/21/2021] [Indexed: 11/16/2022]
Abstract
A critical role of the peripheral axons of nociceptors of the dorsal root ganglion (DRG) is the conduction of all-or-nothing action potentials from peripheral nerve endings to the central nervous system for the perception of noxious stimuli. Plasticity along multiple sites along the pain axis has now been widely implicated in the maladaptive changes that occur in pathological pain states such as neuropathic and inflammatory pain. Notably, increasing evidence suggests that nociceptive axons actively participate through the local expression of ion channels, receptors, and signal transduction molecules through axonal mRNA translation machinery that is independent of the soma component. In this report, we explore the sensitization of sensory neurons through the treatment of compartmentalized axon-like structures spanning microchannels that have been treated with the cytokine IL-6 and, subsequently, capsaicin. These data demonstrate the utility of isolating DRG axon-like structures using microfluidic systems, laying the groundwork for constructing the complex in vitro models of cellular networks that are involved in pain signaling for targeted pharmacological and genetic perturbations.
Collapse
Affiliation(s)
- Rahul Atmaramani
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (R.A.); (S.V.); (L.V.M.); (B.J.B.)
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (P.K.); (J.J.P.)
| | - Srivennela Veeramachaneni
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (R.A.); (S.V.); (L.V.M.); (B.J.B.)
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (P.K.); (J.J.P.)
| | - Liz Valeria Mogas
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (R.A.); (S.V.); (L.V.M.); (B.J.B.)
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (P.K.); (J.J.P.)
| | - Pratik Koppikar
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (P.K.); (J.J.P.)
| | - Bryan J. Black
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (R.A.); (S.V.); (L.V.M.); (B.J.B.)
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (P.K.); (J.J.P.)
| | - Audrey Hammack
- Department of Research, University of Texas at Dallas, Richardson, TX 75080, USA;
| | - Joseph J. Pancrazio
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX 75080, USA; (P.K.); (J.J.P.)
| | - Rafael Granja-Vazquez
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX 75080, USA; (R.A.); (S.V.); (L.V.M.); (B.J.B.)
- Correspondence: ; Tel.: +1-972-883-2138
| |
Collapse
|
15
|
Tong Z, Kwak E, Aguiar A, Peng B, Pouton CW, Voelcker NH, Haynes JM. Compartmentalized microfluidic chambers enable long-term maintenance and communication between human pluripotent stem cell-derived forebrain and midbrain neurons. LAB ON A CHIP 2021; 21:4016-4030. [PMID: 34487130 DOI: 10.1039/d1lc00505g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Compartmentalized microfluidic devices are becoming increasingly popular and have proven to be valuable tools to probe neurobiological functions that are inherently difficult to study using traditional approaches. The ability of microfluidic devices to compartmentalize neurons offers considerable promise for disease modeling and drug discovery. Rodent cortical neurons/neural progenitors are commonly used in such studies but, while these cells mature rapidly, they do not possess the same receptors, ion channels and transport proteins found in human cortical neurons. Human pluripotent stem cell derived neurons offer a human phenotype, but their slow maturation offsets this phenotypic advantage, particularly over long-term culture where overgrowth and subsequent death of neurons may be a problem. In this work, we integrate the use of Matrigel as a 3D cell culture scaffold that enables high cell seeding density over a small fraction of the culture surface. This approach, in an open chamber microfluidic system, enables culture over a five-month period without the use of growth inhibitors. Matrigel was also uniquely utilized to hinder agonist diffusion across microchannels. We demonstrate the development of neuron-to-neuron communication networks by showing that electrical stimulation or the unilateral addition of agonists to one chamber resulted in activation of neurons in the adjacent chamber. Lastly, using a delayed neuron seeding strategy, we show that we can foster essentially one-way communication between separate populations of human forebrain and midbrain dopaminergic neuron containing cultures.
Collapse
Affiliation(s)
- Ziqiu Tong
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
| | - Eunbi Kwak
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
| | - Alita Aguiar
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
| | - Bo Peng
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria, 3168, Australia
| | - Colin W Pouton
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, Victoria, 3168, Australia
- Melbourne Centre for Nanofabrication, Victorian Node of the Australian National Fabrication Facility, Clayton, Victoria, 3168, Australia.
| | - John M Haynes
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville, Victoria, 3052, Australia.
| |
Collapse
|
16
|
Cutaneous innervation in impaired diabetic wound healing. Transl Res 2021; 236:87-108. [PMID: 34029747 PMCID: PMC8380642 DOI: 10.1016/j.trsl.2021.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 05/09/2021] [Accepted: 05/11/2021] [Indexed: 12/11/2022]
Abstract
Type 2 diabetes is associated with several potential comorbidities, among them impaired wound healing, chronic ulcerations, and the requirement for lower extremity amputation. Disease-associated abnormal cellular responses, infection, immunological and microvascular dysfunction, and peripheral neuropathy are implicated in the pathogenesis of the wound healing impairment and the diabetic foot ulcer. The skin houses a dense network of sensory nerve afferents and nerve-derived modulators, which communicate with epidermal keratinocytes and dermal fibroblasts bidirectionally to effect normal wound healing after trauma. However, the mechanisms through which cutaneous innervation modulates wound healing are poorly understood, especially in humans. Better understanding of these mechanisms may provide the basis for targeted treatments for chronic diabetic wounds. This review provides an overview of wound healing pathophysiology with a focus on neural involvement in normal and diabetic wound healing, as well as future therapeutic perspectives to address the unmet needs of diabetic patients with chronic wounds.
Collapse
|
17
|
Prasanna P, Rathee S, Rahul V, Mandal D, Chandra Goud MS, Yadav P, Hawthorne S, Sharma A, Gupta PK, Ojha S, Jha NK, Villa C, Jha SK. Microfluidic Platforms to Unravel Mysteries of Alzheimer's Disease: How Far Have We Come? Life (Basel) 2021; 11:life11101022. [PMID: 34685393 PMCID: PMC8537508 DOI: 10.3390/life11101022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/16/2021] [Accepted: 09/20/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) is a significant health concern with enormous social and economic impact globally. The gradual deterioration of cognitive functions and irreversible neuronal losses are primary features of the disease. Even after decades of research, most therapeutic options are merely symptomatic, and drugs in clinical practice present numerous side effects. Lack of effective diagnostic techniques prevents the early prognosis of disease, resulting in a gradual deterioration in the quality of life. Furthermore, the mechanism of cognitive impairment and AD pathophysiology is poorly understood. Microfluidics exploits different microscale properties of fluids to mimic environments on microfluidic chip-like devices. These miniature multichambered devices can be used to grow cells and 3D tissues in vitro, analyze cell-to-cell communication, decipher the roles of neural cells such as microglia, and gain insights into AD pathophysiology. This review focuses on the applications and impact of microfluidics on AD research. We discuss the technical challenges and possible solutions provided by this new cutting-edge technique to understand disease-associated pathways and mechanisms.
Collapse
Affiliation(s)
- Pragya Prasanna
- School of Applied Sciences, KK University, Nalanda 803115, Bihar, India;
- Correspondence: or (P.P.); (S.K.J.)
| | - Shweta Rathee
- Department of Food Science and Technology, National Institute of Food Technology, Entrepreneurship and Management, Sonipat 131028, Haryana, India;
| | - Vedanabhatla Rahul
- Department of Mechanical Engineering, National Institute of Technology, Rourkela 769008, Odisha, India;
| | - Debabrata Mandal
- Department of Biotechnology, National Institute of Pharmaceutical Education and Research, Hajipur 844101, Bihar, India;
| | | | - Pardeep Yadav
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (P.Y.); (N.K.J.)
| | - Susan Hawthorne
- School of Pharmacy and Pharmaceutical Sciences, Ulster University, Cromore Road, Coleraine, Co., Londonderry BT52 1SA, UK;
| | - Ankur Sharma
- Department of Life Sciences, School of Basic Science and Research (SBSR), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (A.S.); (P.K.G.)
| | - Piyush Kumar Gupta
- Department of Life Sciences, School of Basic Science and Research (SBSR), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (A.S.); (P.K.G.)
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, P.O. Box 17666, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (P.Y.); (N.K.J.)
| | - Chiara Villa
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy;
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology (SET), Sharda University, Greater Noida 201310, Uttar Pradesh, India; (P.Y.); (N.K.J.)
- Correspondence: or (P.P.); (S.K.J.)
| |
Collapse
|
18
|
Murray BO, Flores C, Williams C, Flusberg DA, Marr EE, Kwiatkowska KM, Charest JL, Isenberg BC, Rohn JL. Recurrent Urinary Tract Infection: A Mystery in Search of Better Model Systems. Front Cell Infect Microbiol 2021; 11:691210. [PMID: 34123879 PMCID: PMC8188986 DOI: 10.3389/fcimb.2021.691210] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 05/04/2021] [Indexed: 12/12/2022] Open
Abstract
Urinary tract infections (UTIs) are among the most common infectious diseases worldwide but are significantly understudied. Uropathogenic E. coli (UPEC) accounts for a significant proportion of UTI, but a large number of other species can infect the urinary tract, each of which will have unique host-pathogen interactions with the bladder environment. Given the substantial economic burden of UTI and its increasing antibiotic resistance, there is an urgent need to better understand UTI pathophysiology - especially its tendency to relapse and recur. Most models developed to date use murine infection; few human-relevant models exist. Of these, the majority of in vitro UTI models have utilized cells in static culture, but UTI needs to be studied in the context of the unique aspects of the bladder's biophysical environment (e.g., tissue architecture, urine, fluid flow, and stretch). In this review, we summarize the complexities of recurrent UTI, critically assess current infection models and discuss potential improvements. More advanced human cell-based in vitro models have the potential to enable a better understanding of the etiology of UTI disease and to provide a complementary platform alongside animals for drug screening and the search for better treatments.
Collapse
Affiliation(s)
- Benjamin O. Murray
- Centre for Urological Biology, Department of Renal Medicine, University College London, London, United Kingdom
| | - Carlos Flores
- Centre for Urological Biology, Department of Renal Medicine, University College London, London, United Kingdom
| | - Corin Williams
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Deborah A. Flusberg
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Elizabeth E. Marr
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Karolina M. Kwiatkowska
- Centre for Urological Biology, Department of Renal Medicine, University College London, London, United Kingdom
| | - Joseph L. Charest
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Brett C. Isenberg
- Department of Bioengineering, Charles Stark Draper Laboratory, Inc., Cambridge, MA, United States
| | - Jennifer L. Rohn
- Centre for Urological Biology, Department of Renal Medicine, University College London, London, United Kingdom
| |
Collapse
|
19
|
Sidar B, Jenkins BR, Huang S, Spence JR, Walk ST, Wilking JN. Long-term flow through human intestinal organoids with the gut organoid flow chip (GOFlowChip). LAB ON A CHIP 2019; 19:3552-3562. [PMID: 31556415 PMCID: PMC8327675 DOI: 10.1039/c9lc00653b] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Human intestinal organoids (HIOs) are millimeter-scale models of the human intestinal epithelium and hold tremendous potential for advancing fundamental and applied biomedical research. HIOs resemble the native gut in that they consist of a fluid-filled lumen surrounded by a polarized epithelium and associated mesenchyme; however, their topologically-closed, spherical shape prevents flow through the interior luminal space, making the system less physiological and leading to the buildup of cellular and metabolic waste. These factors ultimately limit experimentation inside the HIOs. Here, we present a millifluidic device called the gut organoid flow chip (GOFlowChip), which we use to "port" HIOs and establish steady-state liquid flow through the lumen for multiple days. This long-term flow is enabled by the use of laser-cut silicone gaskets, which allow liquid in the device to be slightly pressurized, suppressing bubble formation. To demonstrate the utility of the device, we establish separate luminal and extraluminal flow and use luminal flow to remove accumulated waste. This represents the first demonstration of established liquid flow through the luminal space of a gastrointestinal organoid over physiologically relevant time scales. Flow cytometry results reveal that HIO cell viability is unaffected by long-term porting and luminal flow. We expect the real-time, long-term control over luminal and extraluminal contents provided by the GOFlowChip will enable a wide variety of studies including intestinal secretion, absorption, transport, and co-culture with intestinal microorganisms.
Collapse
Affiliation(s)
- Barkan Sidar
- Department of Chemical and Biological Engineering, Montana State University, Bozeman, MT, USA.
| | | | | | | | | | | |
Collapse
|
20
|
Vysokov N, McMahon SB, Raouf R. The role of Na V channels in synaptic transmission after axotomy in a microfluidic culture platform. Sci Rep 2019; 9:12915. [PMID: 31501450 PMCID: PMC6733904 DOI: 10.1038/s41598-019-49214-w] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 08/20/2019] [Indexed: 01/07/2023] Open
Abstract
Voltage gated sodium channels are key players in aberrant pain signaling and sensitization of nociceptors after peripheral nerve injury. The extent to which sodium channel activity after injury contributes to synaptic transmission at the first pain synapse however remains unclear. To investigate the effect of axotomy on synaptic transmission between dorsal root ganglia neurons and dorsal horn neurons, we reconstructed the first pain synapse in a novel microfluidic based compartmentalized cell culture system, which recapitulates the connectivity of peripheral pain signaling. We show that following axotomy of the distal axons, inhibition of NaV1.7 and NaV1.8 sodium channels in incoming presynaptic DRG axons is no longer sufficient to block activation of these axons and the resulting synaptic transmission to dorsal horn neurons. We found that blockade of NaV1.6 activity is highly effective in reducing activation of incoming axons contributing to synaptic transmission after axotomy of DRG neurons. The microfluidic culture system described here offers an in vitro platform to recapitulate and study the first pain synapse.
Collapse
Affiliation(s)
- Nickolai Vysokov
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Stephen B McMahon
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom
| | - Ramin Raouf
- Wolfson Centre for Age-Related Diseases, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, SE1 1UL, United Kingdom.
| |
Collapse
|
21
|
van de Wijdeven R, Ramstad OH, Valderhaug VD, Köllensperger P, Sandvig A, Sandvig I, Halaas Ø. A novel lab-on-chip platform enabling axotomy and neuromodulation in a multi-nodal network. Biosens Bioelectron 2019; 140:111329. [DOI: 10.1016/j.bios.2019.111329] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 05/13/2019] [Accepted: 05/15/2019] [Indexed: 12/29/2022]
|
22
|
Kankala RK, Wang SB, Chen AZ. Microengineered Organ-on-a-chip Platforms towards Personalized Medicine. Curr Pharm Des 2019; 24:5354-5366. [DOI: 10.2174/1381612825666190222143542] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/12/2019] [Indexed: 11/22/2022]
Abstract
Current preclinical drug evaluation strategies that are explored to predict the pharmacological parameters,
as well as toxicological issues, utilize traditional oversimplified cell cultures and animal models. However,
these traditional approaches are time-consuming, and cannot reproduce the functions of the complex biological
tissue architectures. On the other hand, the obtained data from animal models cannot be precisely extrapolated to
humans because it sometimes results in the distinct safe starting doses for clinical trials due to vast differences in
their genomes. To address these limitations, the microengineered, biomimetic organ-on-a-chip platforms fabricated
using advanced materials that are interconnected using the microfluidic circuits, can stanchly reiterate or
mimic the complex tissue-organ level structures including the cellular architecture and physiology, compartmentalization
and interconnectivity of human organ platforms. These innovative and cost-effective systems potentially
enable the prediction of the responses toward pharmaceutical compounds and remarkable advances in
materials and microfluidics technology, which can rapidly progress the drug development process. In this review,
we emphasize the integration of microfluidic models with the 3D simulations from tissue engineering to fabricate
organ-on-a-chip platforms, which explicitly fulfill the demand of creating the robust models for preclinical testing
of drugs. At first, we give a brief overview of the limitations associated with the current drug development pipeline
that includes drug screening methods, in vitro molecular assays, cell culture platforms and in vivo models.
Further, we discuss various organ-on-a-chip platforms, highlighting their benefits and performance in the preclinical
stages. Next, we aim to emphasize their current applications toward pharmaceutical benefits including the
drug screening as well as toxicity testing, and advances in personalized precision medicine as well as potential
challenges for their commercialization. We finally recapitulate with the lessons learned and the outlook highlighting
the future directions for accelerating the clinical translation of delivery systems.
Collapse
Affiliation(s)
- Ranjith Kumar Kankala
- Institute of Biomaterials and Tissue Engineering, Huaqiao University, Xiamen 361021, China
| | - Shi-Bin Wang
- College of Chemical Engineering, Huaqiao University, Xiamen 361021, China
| | - Ai-Zheng Chen
- Fujian Provincial Key Laboratory of Biochemical Technology (Huaqiao University), Xiamen 361021, China
| |
Collapse
|
23
|
Black BJ, Atmaramani R, Plagens S, Campbell ZT, Dussor G, Price TJ, Pancrazio JJ. Emerging neurotechnology for antinoceptive mechanisms and therapeutics discovery. Biosens Bioelectron 2018; 126:679-689. [PMID: 30544081 DOI: 10.1016/j.bios.2018.11.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 11/01/2018] [Accepted: 11/10/2018] [Indexed: 12/20/2022]
Abstract
The tolerance, abuse, and potential exacerbation associated with classical chronic pain medications such as opioids creates a need for alternative therapeutics. Phenotypic screening provides a complementary approach to traditional target-based drug discovery. Profiling cellular phenotypes enables quantification of physiologically relevant traits central to a disease pathology without prior identification of a specific drug target. For complex disorders such as chronic pain, which likely involves many molecular targets, this approach may identify novel treatments. Sensory neurons, termed nociceptors, are derived from dorsal root ganglia (DRG) and can undergo changes in membrane excitability during chronic pain. In this review, we describe phenotypic screening paradigms that make use of nociceptor electrophysiology. The purpose of this paper is to review the bioelectrical behavior of DRG neurons, signaling complexity in sensory neurons, various sensory neuron models, assays for bioelectrical behavior, and emerging efforts to leverage microfabrication and microfluidics for assay development. We discuss limitations and advantages of these various approaches and offer perspectives on opportunities for future development.
Collapse
Affiliation(s)
- Bryan J Black
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA.
| | - Rahul Atmaramani
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Sarah Plagens
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Zachary T Campbell
- Department of Biological Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| | - Joseph J Pancrazio
- Department of Bioengineering, University of Texas at Dallas, 800 W. Campbell Road, Richardson, TX 75080, USA
| |
Collapse
|
24
|
Modeling Host-Pathogen Interactions in the Context of the Microenvironment: Three-Dimensional Cell Culture Comes of Age. Infect Immun 2018; 86:IAI.00282-18. [PMID: 30181350 DOI: 10.1128/iai.00282-18] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Tissues and organs provide the structural and biochemical landscapes upon which microbial pathogens and commensals function to regulate health and disease. While flat two-dimensional (2-D) monolayers composed of a single cell type have provided important insight into understanding host-pathogen interactions and infectious disease mechanisms, these reductionist models lack many essential features present in the native host microenvironment that are known to regulate infection, including three-dimensional (3-D) architecture, multicellular complexity, commensal microbiota, gas exchange and nutrient gradients, and physiologically relevant biomechanical forces (e.g., fluid shear, stretch, compression). A major challenge in tissue engineering for infectious disease research is recreating this dynamic 3-D microenvironment (biological, chemical, and physical/mechanical) to more accurately model the initiation and progression of host-pathogen interactions in the laboratory. Here we review selected 3-D models of human intestinal mucosa, which represent a major portal of entry for infectious pathogens and an important niche for commensal microbiota. We highlight seminal studies that have used these models to interrogate host-pathogen interactions and infectious disease mechanisms, and we present this literature in the appropriate historical context. Models discussed include 3-D organotypic cultures engineered in the rotating wall vessel (RWV) bioreactor, extracellular matrix (ECM)-embedded/organoid models, and organ-on-a-chip (OAC) models. Collectively, these technologies provide a more physiologically relevant and predictive framework for investigating infectious disease mechanisms and antimicrobial therapies at the intersection of the host, microbe, and their local microenvironments.
Collapse
|
25
|
Black BJ, Atmaramani R, Kumaraju R, Plagens S, Romero-Ortega M, Dussor G, Price TJ, Campbell ZT, Pancrazio JJ. Adult mouse sensory neurons on microelectrode arrays exhibit increased spontaneous and stimulus-evoked activity in the presence of interleukin-6. J Neurophysiol 2018; 120:1374-1385. [PMID: 29947589 DOI: 10.1152/jn.00158.2018] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Following inflammation or injury, sensory neurons located in the dorsal root ganglia (DRG) may exhibit increased spontaneous and/or stimulus-evoked activity, contributing to chronic pain. Current treatment options for peripherally mediated chronic pain are highly limited, driving the development of cell- or tissue-based phenotypic (function-based) screening assays for peripheral analgesic and mechanistic lead discovery. Extant assays are often limited by throughput, content, use of tumorigenic cell lines, or tissue sources from immature developmental stages (i.e., embryonic or postnatal). Here, we describe a protocol for culturing adult mouse DRG neurons on substrate-integrated multiwell microelectrode arrays (MEAs). This approach enables multiplexed measurements of spontaneous as well as stimulus-evoked extracellular action potentials from large populations of cells. The DRG cultures exhibit stable spontaneous activity from 9 to 21 days in vitro. Activity is readily evoked by known chemical and physical agonists of sensory neuron activity such as capsaicin, bradykinin, PGE2, heat, and electrical field stimulation. Most importantly, we demonstrate that both spontaneous and stimulus-evoked activity may be potentiated by incubation with the inflammatory cytokine interleukin-6 (IL-6). Acute responsiveness to IL-6 is inhibited by treatment with a MAPK-interacting kinase 1/2 inhibitor, cercosporamide. In total, these findings suggest that adult mouse DRG neurons on multiwell MEAs are applicable to ongoing efforts to discover peripheral analgesic and their mechanisms of action. NEW & NOTEWORTHY This work describes methodologies for culturing spontaneously active adult mouse dorsal root ganglia (DRG) sensory neurons on microelectrode arrays. We characterize spontaneous and stimulus-evoked adult DRG activity over durations consistent with pharmacological interventions. Furthermore, persistent hyperexcitability could be induced by incubation with inflammatory cytokine IL-6 and attenuated with cercosporamide, an inhibitor of the IL-6 sensitization pathway. This constitutes a more physiologically relevant, moderate-throughput in vitro model for peripheral analgesic screening as well as mechanistic lead discovery.
Collapse
Affiliation(s)
- Bryan J Black
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Rahul Atmaramani
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Rajeshwari Kumaraju
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Sarah Plagens
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Mario Romero-Ortega
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| | - Gregory Dussor
- School of Behavioral and Brain Sciences, The University of Texas at Dallas , Richardson, Texas
| | - Theodore J Price
- School of Behavioral and Brain Sciences, The University of Texas at Dallas , Richardson, Texas
| | - Zachary T Campbell
- Department of Biological Sciences, The University of Texas at Dallas, Richardson, Richardson, Texas
| | - Joseph J Pancrazio
- Department of Bioengineering, The University of Texas at Dallas , Richardson, Texas
| |
Collapse
|
26
|
Jocher G, Mannschatz SH, Offterdinger M, Schweigreiter R. Microfluidics of Small-Population Neurons Allows for a Precise Quantification of the Peripheral Axonal Growth State. Front Cell Neurosci 2018; 12:166. [PMID: 29962939 PMCID: PMC6013724 DOI: 10.3389/fncel.2018.00166] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/29/2018] [Indexed: 12/30/2022] Open
Abstract
Neurons are morphologically the most complex cell types and are characterized by a significant degree of axonal autonomy as well as having efficient means of communication between axons and neuronal cell bodies. For studying the response to axonal injury, compartmentalized microfluidic chambers (MFCs) have become the method of choice because they allow for the selective treatment of axons, independently of the soma, in a highly controllable and reproducible manner. A major disadvantage of these devices is the relatively large number of neurons needed for seeding, which makes them impractical to use with small-population neurons, such as sensory neurons of the mouse. Here, we describe a simple approach of seeding and culturing neurons in MFCs that allows for a dramatic reduction of neurons required to 10,000 neurons per device. This technique facilitates efficient experiments with small-population neurons in compartmentalized MFCs. We used this experimental setup to determine the intrinsic axonal growth state of adult mouse sensory neurons derived from dorsal root ganglia (DRG) and even trigeminal ganglia (TG). In combination with a newly developed linear Sholl analysis tool, we have examined the axonal growth responses of DRG and TG neurons to various cocktails of neurotrophins, glial cell line-derived neurotrophic factor (GDNF), ciliary neurotrophic factor (CNTF) and leptin. Precise quantification of axonal outgrowth revealed specific differences in the potency of each combination to promote axonal regeneration and to switch neurons into an intrinsic axonal growth state. This novel experimental setup opens the way to practicable microfluidic analyses of neurons that have previously been largely neglected simply due to insufficient numbers, including sensory neurons, sympathetic neurons and motor neurons.
Collapse
Affiliation(s)
- Georg Jocher
- Biocenter, Division of Neurobiochemistry, Innsbruck Medical University, Innsbruck, Austria
| | - Sidney H Mannschatz
- Biocenter, Division of Neurobiochemistry, Innsbruck Medical University, Innsbruck, Austria
| | - Martin Offterdinger
- Biocenter, Division of Neurobiochemistry, Innsbruck Medical University, Innsbruck, Austria
| | - Rüdiger Schweigreiter
- Biocenter, Division of Neurobiochemistry, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
27
|
Tillmaand EG, Sweedler JV. Integrating Mass Spectrometry with Microphysiological Systems for Improved Neurochemical Studies. ACTA ACUST UNITED AC 2018; 2. [PMID: 30148282 DOI: 10.21037/mps.2018.05.01] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Microphysiological systems, often referred to as "organs-on-chips", are in vitro platforms designed to model the spatial, chemical, structural, and physiological elements of in vivo cellular environments. They enhance the evaluation of complex engineered biological systems and are a step between traditional cell culture and in vivo experimentation. As neurochemists and measurement scientists studying the molecules involved in intercellular communication in the nervous system, we focus here on recent advances in neuroscience using microneurological systems and their potential to interface with mass spectrometry. We discuss a number of examples - microfluidic devices, spheroid cultures, hydrogels, scaffolds, and fibers - highlighting those that would benefit from mass spectrometric technologies to obtain improved chemical information.
Collapse
Affiliation(s)
- Emily G Tillmaand
- Department of Chemistry, the Neuroscience Program and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jonathan V Sweedler
- Department of Chemistry, the Neuroscience Program and the Beckman Institute, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
28
|
Cook AD, Christensen AD, Tewari D, McMahon SB, Hamilton JA. Immune Cytokines and Their Receptors in Inflammatory Pain. Trends Immunol 2018; 39:240-255. [DOI: 10.1016/j.it.2017.12.003] [Citation(s) in RCA: 117] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Revised: 11/28/2017] [Accepted: 12/13/2017] [Indexed: 01/23/2023]
|
29
|
Niemeyer BF, Zhao P, Tuder RM, Benam KH. Advanced Microengineered Lung Models for Translational Drug Discovery. SLAS DISCOVERY 2018; 23:777-789. [PMID: 29447055 DOI: 10.1177/2472555218760217] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Lung diseases impose a significant socioeconomic burden and are a leading cause of morbidity and mortality worldwide. Moreover, respiratory medicine, unlike several other therapeutic areas, faces a disappointingly low number of new approved therapies. This is partly due to lack of reliable in vitro or in vivo models that can reproduce organ-level complexity and pathophysiological responses of human lung. Here, we examine new opportunities in application of recently emerged organ-on-chip technology to model human lung alveolus and small airway in preclinical drug development and biomarker discovery. We also discuss challenges that need to be addressed in coming years to further enhance the physiological and clinical relevance of these microsystems, enable their increased accessibility, and support their leap into personalized medicine.
Collapse
Affiliation(s)
- Brian F Niemeyer
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Peng Zhao
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Rubin M Tuder
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA
| | - Kambez H Benam
- 1 Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, CO, USA.,2 Department of Bioengineering, University of Colorado Denver, Aurora, CO, USA
| |
Collapse
|
30
|
Functional imaging in microfluidic chambers reveals sensory neuron sensitivity is differentially regulated between neuronal regions. Pain 2018; 159:1413-1425. [DOI: 10.1097/j.pain.0000000000001145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Structuring a multi-nodal neural network in vitro within a novel design microfluidic chip. Biomed Microdevices 2018; 20:9. [DOI: 10.1007/s10544-017-0254-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
32
|
Geraili A, Jafari P, Hassani MS, Araghi BH, Mohammadi MH, Ghafari AM, Tamrin SH, Modarres HP, Kolahchi AR, Ahadian S, Sanati-Nezhad A. Controlling Differentiation of Stem Cells for Developing Personalized Organ-on-Chip Platforms. Adv Healthc Mater 2018; 7. [PMID: 28910516 DOI: 10.1002/adhm.201700426] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/01/2017] [Indexed: 01/09/2023]
Abstract
Organ-on-chip (OOC) platforms have attracted attentions of pharmaceutical companies as powerful tools for screening of existing drugs and development of new drug candidates. OOCs have primarily used human cell lines or primary cells to develop biomimetic tissue models. However, the ability of human stem cells in unlimited self-renewal and differentiation into multiple lineages has made them attractive for OOCs. The microfluidic technology has enabled precise control of stem cell differentiation using soluble factors, biophysical cues, and electromagnetic signals. This study discusses different tissue- and organ-on-chip platforms (i.e., skin, brain, blood-brain barrier, bone marrow, heart, liver, lung, tumor, and vascular), with an emphasis on the critical role of stem cells in the synthesis of complex tissues. This study further recaps the design, fabrication, high-throughput performance, and improved functionality of stem-cell-based OOCs, technical challenges, obstacles against implementing their potential applications, and future perspectives related to different experimental platforms.
Collapse
Affiliation(s)
- Armin Geraili
- Department of Chemical and Petroleum Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
| | - Parya Jafari
- Graduate Program in Biomedical Engineering; Western University; London N6A 5B9 ON Canada
- Department of Electrical Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohsen Sheikh Hassani
- Department of Systems and Computer Engineering; Carleton University; 1125 Colonel By Drive Ottawa K1S 5B6 ON Canada
| | - Behnaz Heidary Araghi
- Department of Materials Science and Engineering; Sharif University of Technology; Azadi, Tehran 14588-89694 Iran
| | - Mohammad Hossein Mohammadi
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Mohammad Ghafari
- Department of Stem Cells and Developmental Biology; Cell Science Research Center; Royan Institute for Stem Cell Biology and Technology; Tehran 16635-148 Iran
| | - Sara Hasanpour Tamrin
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Hassan Pezeshgi Modarres
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Ahmad Rezaei Kolahchi
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
| | - Samad Ahadian
- Institute of Biomaterials and Biomedical Engineering; University of Toronto; Toronto ON M5S 3G9 Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario M5S 3E5 Canada
| | - Amir Sanati-Nezhad
- BioMEMS and Bioinspired Microfluidic Laboratory (BioM); Department of Mechanical and Manufacturing Engineering; University of Calgary; 2500 University Drive N.W. Calgary T2N 1N4 AB Canada
- Center for Bioengineering Research and Education; Biomedical Engineering Program; University of Calgary; Calgary T2N 1N4 AB Canada
| |
Collapse
|
33
|
Aberrant plasticity of peripheral sensory axons in a painful neuropathy. Sci Rep 2017; 7:3407. [PMID: 28611388 PMCID: PMC5469767 DOI: 10.1038/s41598-017-03390-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Accepted: 04/27/2017] [Indexed: 12/21/2022] Open
Abstract
Neuronal cells express considerable plasticity responding to environmental cues, in part, through subcellular mRNA regulation. Here we report on the extensive changes in distribution of mRNAs in the cell body and axon compartments of peripheral sensory neurons and the 3' untranslated region (3'UTR) landscapes after unilateral sciatic nerve entrapment (SNE) injury in rats. Neuronal cells dissociated from SNE-injured and contralateral L4 and L5 dorsal root ganglia were cultured in a compartmentalized system. Axonal and cell body RNA samples were separately subjected to high throughput RNA sequencing (RNA-Seq). The injured axons exhibited enrichment of mRNAs related to protein synthesis and nerve regeneration. Lengthening of 3'UTRs was more prevalent in the injured axons, including the newly discovered alternative cleavage and polyadenylation of NaV1.8 mRNA. Alternative polyadenylation was largely independent from the relative abundance of axonal mRNAs; but they were highly clustered in functional pathways related to RNA granule formation in the injured axons. These RNA-Seq data analyses indicate that peripheral nerve injury may result in highly selective mRNA enrichment in the affected axons with 3'UTR alterations potentially contributing to the mechanism of neuropathic pain.
Collapse
|
34
|
Aziz AUR, Geng C, Fu M, Yu X, Qin K, Liu B. The Role of Microfluidics for Organ on Chip Simulations. Bioengineering (Basel) 2017; 4:E39. [PMID: 28952518 PMCID: PMC5590458 DOI: 10.3390/bioengineering4020039] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 05/01/2017] [Accepted: 05/02/2017] [Indexed: 12/19/2022] Open
Abstract
A multichannel three-dimensional chip of a microfluidic cell culture which enables the simulation of organs is called an "organ on a chip" (OC). With the integration of many other technologies, OCs have been mimicking organs, substituting animal models, and diminishing the time and cost of experiments which is better than the preceding conventional in vitro models, which make them imperative tools for finding functional properties, pathological states, and developmental studies of organs. In this review, recent progress regarding microfluidic devices and their applications in cell cultures is discussed to explain the advantages and limitations of these systems. Microfluidics is not a solution but only an approach to create a controlled environment, however, other supporting technologies are needed, depending upon what is intended to be achieved. Microfluidic platforms can be integrated with additional technologies to enhance the organ on chip simulations. Besides, new directions and areas are mentioned for interested researchers in this field, and future challenges regarding the simulation of OCs are also discussed, which will make microfluidics more accurate and beneficial for biological applications.
Collapse
Affiliation(s)
- Aziz Ur Rehman Aziz
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Chunyang Geng
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Mengjie Fu
- Dalian Institute of Maternal and Child Health Care. Dalian 116024, Liaoning Province, China.
| | - Xiaohui Yu
- Dalian Institute of Maternal and Child Health Care. Dalian 116024, Liaoning Province, China.
| | - Kairong Qin
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| | - Bo Liu
- Department of Biomedical Engineering, Dalian University of Technology, Dalian 116024, Liaoning Province, China.
| |
Collapse
|
35
|
Wang S, Wang S, Asgar J, Joseph J, Ro JY, Wei F, Campbell JN, Chung MK. Ca 2+ and calpain mediate capsaicin-induced ablation of axonal terminals expressing transient receptor potential vanilloid 1. J Biol Chem 2017; 292:8291-8303. [PMID: 28360106 DOI: 10.1074/jbc.m117.778290] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 03/28/2017] [Indexed: 01/01/2023] Open
Abstract
Capsaicin is an ingredient in spicy peppers that produces burning pain by activating transient receptor potential vanilloid 1 (TRPV1), a Ca2+-permeable ion channel in nociceptors. Capsaicin has also been used as an analgesic, and its topical administration is approved for the treatment of certain pain conditions. The mechanisms underlying capsaicin-induced analgesia likely involve reversible ablation of nociceptor terminals. However, the mechanisms underlying these effects are not well understood. To visualize TRPV1-lineage axons, a genetically engineered mouse model was used in which a fluorophore is expressed under the TRPV1 promoter. Using a combination of these TRPV1-lineage reporter mice and primary afferent cultures, we monitored capsaicin-induced effects on afferent terminals in real time. We found that Ca2+ influx through TRPV1 is necessary for capsaicin-induced ablation of nociceptive terminals. Although capsaicin-induced mitochondrial Ca2+ uptake was TRPV1-dependent, dissipation of the mitochondrial membrane potential, inhibition of the mitochondrial transition permeability pore, and scavengers of reactive oxygen species did not attenuate capsaicin-induced ablation. In contrast, MDL28170, an inhibitor of the Ca2+-dependent protease calpain, diminished ablation. Furthermore, overexpression of calpastatin, an endogenous inhibitor of calpain, or knockdown of calpain 2 also decreased ablation. Quantitative assessment of TRPV1-lineage afferents in the epidermis of the hind paws of the reporter mice showed that EGTA and MDL28170 diminished capsaicin-induced ablation. Moreover, MDL28170 prevented capsaicin-induced thermal hypoalgesia. These results suggest that TRPV1/Ca2+/calpain-dependent signaling plays a dominant role in capsaicin-induced ablation of nociceptive terminals and further our understanding of the molecular mechanisms underlying the effects of capsaicin on nociceptors.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - Sen Wang
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - Jamila Asgar
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - John Joseph
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - Jin Y Ro
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | - Feng Wei
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201
| | | | - Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland, Baltimore, Maryland 21201.
| |
Collapse
|
36
|
Abstract
Diabetic neuropathy is a common secondary complication of diabetes that impacts on patient's health and well-being. Distal axon degeneration is a key feature of diabetic neuropathy, but the pathological changes which underlie axonal die-back are incompletely understood; despite decades of research a treatment has not yet been identified. Basic research must focus on understanding the complex mechanisms underlying changes that occur in the nervous system during diabetes. To this end, tissue culture techniques are invaluable as they enable researchers to examine the intricate mechanistic responses of cells to high glucose or other factors in order to better understand the pathogenesis of nerve dysfunction. This chapter describes the use of in vitro models to study a wide range of specific cellular effects pertaining to diabetic neuropathy including apoptosis, neurite outgrowth, neurodegeneration, activity, and bioenergetics. We consider problems associated with in vitro modeling and future refinement such as use of induced pluripotent stem cells and microfluidic technology.
Collapse
|
37
|
Curley JL, Sklare SC, Bowser DA, Saksena J, Moore MJ, Chrisey DB. Isolated node engineering of neuronal systems using laser direct write. Biofabrication 2016; 8:015013. [DOI: 10.1088/1758-5090/8/1/015013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
38
|
Retinal synaptic regeneration via microfluidic guiding channels. Sci Rep 2015; 5:13591. [PMID: 26314276 PMCID: PMC4551994 DOI: 10.1038/srep13591] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 07/31/2015] [Indexed: 11/08/2022] Open
Abstract
In vitro culture of dissociated retinal neurons is an important model for investigating retinal synaptic regeneration (RSR) and exploring potentials in artificial retina. Here, retinal precursor cells were cultured in a microfluidic chip with multiple arrays of microchannels in order to reconstruct the retinal neuronal synapse. The cultured retinal cells were physically connected through microchannels. Activation of electric signal transduction by the cells through the microchannels was demonstrated by administration of glycinergic factors. In addition, an image-based analytical method was used to quantify the synaptic connections and to assess the kinetics of synaptic regeneration. The rate of RSR decreased significantly below 100 μM of inhibitor glycine and then approached to a relatively constant level at higher concentrations. Furthermore, RSR was enhanced by chemical stimulation with potassium chloride. Collectively, the microfluidic synaptic regeneration chip provides a novel tool for high-throughput investigation of RSR at the cellular level and may be useful in quality control of retinal precursor cell transplantation.
Collapse
|
39
|
BK Channels Localize to the Paranodal Junction and Regulate Action Potentials in Myelinated Axons of Cerebellar Purkinje Cells. J Neurosci 2015; 35:7082-94. [PMID: 25948259 DOI: 10.1523/jneurosci.3778-14.2015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
In myelinated axons, K(+) channels are clustered in distinct membrane domains to regulate action potentials (APs). At nodes of Ranvier, Kv7 channels are expressed with Na(+) channels, whereas Kv1 channels flank nodes at juxtaparanodes. Regulation of axonal APs by K(+) channels would be particularly important in fast-spiking projection neurons such as cerebellar Purkinje cells. Here, we show that BK/Slo1 channels are clustered at the paranodal junctions of myelinated Purkinje cell axons of rat and mouse. The paranodal junction is formed by a set of cell-adhesion molecules, including Caspr, between the node and juxtaparanodes in which it separates nodal from internodal membrane domains. Remarkably, only Purkinje cell axons have detectable paranodal BK channels, whose clustering requires the formation of the paranodal junction via Caspr. Thus, BK channels occupy this unique domain in Purkinje cell axons along with the other K(+) channel complexes at nodes and juxtaparanodes. To investigate the physiological role of novel paranodal BK channels, we examined the effect of BK channel blockers on antidromic AP conduction. We found that local application of blockers to the axon resulted in a significant increase in antidromic AP failure at frequencies above 100 Hz. We also found that Ni(2+) elicited a similar effect on APs, indicating the involvement of Ni(2+)-sensitive Ca(2+) channels. Furthermore, axonal application of BK channel blockers decreased the inhibitory synaptic response in the deep cerebellar nuclei. Thus, paranodal BK channels uniquely support high-fidelity firing of APs in myelinated Purkinje cell axons, thereby underpinning the output of the cerebellar cortex.
Collapse
|
40
|
Bhatia SN, Ingber DE. Microfluidic organs-on-chips. Nat Biotechnol 2015; 32:760-72. [PMID: 25093883 DOI: 10.1038/nbt.2989] [Citation(s) in RCA: 1999] [Impact Index Per Article: 199.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 07/10/2014] [Indexed: 02/07/2023]
Abstract
An organ-on-a-chip is a microfluidic cell culture device created with microchip manufacturing methods that contains continuously perfused chambers inhabited by living cells arranged to simulate tissue- and organ-level physiology. By recapitulating the multicellular architectures, tissue-tissue interfaces, physicochemical microenvironments and vascular perfusion of the body, these devices produce levels of tissue and organ functionality not possible with conventional 2D or 3D culture systems. They also enable high-resolution, real-time imaging and in vitro analysis of biochemical, genetic and metabolic activities of living cells in a functional tissue and organ context. This technology has great potential to advance the study of tissue development, organ physiology and disease etiology. In the context of drug discovery and development, it should be especially valuable for the study of molecular mechanisms of action, prioritization of lead candidates, toxicity testing and biomarker identification.
Collapse
Affiliation(s)
- Sangeeta N Bhatia
- 1] Department of Electrical Engineering &Computer Science, Koch Institute and Institute for Medical Engineering and Science, Massachusetts Institute of Technology and Broad Institute, Cambridge, Massachusetts, USA. [2] Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Donald E Ingber
- 1] Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA. [2] Vascular Biology Program, Departments of Pathology &Surgery, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA. [3] School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts, USA
| |
Collapse
|
41
|
Tong Z, Segura-Feliu M, Seira O, Homs-Corbera A, Del Río JA, Samitier J. A microfluidic neuronal platform for neuron axotomy and controlled regenerative studies. RSC Adv 2015. [DOI: 10.1039/c5ra11522a] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We have presented here a simple microfluidic approach to model mechanical and synchronized axotomy of a large number of axons to study axonal regeneration, and to facilitate rapid screening and discovery of novel pharmaceutical compounds.
Collapse
Affiliation(s)
- Ziqiu Tong
- Institute for Bioengineering of Catalonia (IBEC)
- 08028 Barcelona
- Spain
| | - Miriam Segura-Feliu
- Institute for Bioengineering of Catalonia (IBEC)
- 08028 Barcelona
- Spain
- Department of Cell Biology
- University of Barcelona
| | - Oscar Seira
- Institute for Bioengineering of Catalonia (IBEC)
- 08028 Barcelona
- Spain
| | - Antoni Homs-Corbera
- Institute for Bioengineering of Catalonia (IBEC)
- 08028 Barcelona
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
- Biomateriales y Nanomedicina (CIBERBBN)
| | - José Antonio Del Río
- Institute for Bioengineering of Catalonia (IBEC)
- 08028 Barcelona
- Spain
- Department of Cell Biology
- University of Barcelona
| | - Josep Samitier
- Institute for Bioengineering of Catalonia (IBEC)
- 08028 Barcelona
- Spain
- Centro de Investigación Biomédica en Red de Bioingeniería
- Biomateriales y Nanomedicina (CIBERBBN)
| |
Collapse
|