1
|
Nelson A, McMullen N, Gebremeskel S, De Antueno R, Mackenzie D, Duncan R, Johnston B. Fusogenic vesicular stomatitis virus combined with natural killer T cell immunotherapy controls metastatic breast cancer. Breast Cancer Res 2024; 26:78. [PMID: 38750591 PMCID: PMC11094881 DOI: 10.1186/s13058-024-01818-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Accepted: 03/30/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Metastatic breast cancer is a leading cause of cancer death in woman. Current treatment options are often associated with adverse side effects and poor outcomes, demonstrating the need for effective new treatments. Immunotherapies can provide durable outcomes in many cancers; however, limited success has been achieved in metastatic triple negative breast cancer. We tested whether combining different immunotherapies can target metastatic triple negative breast cancer in pre-clinical models. METHODS Using primary and metastatic 4T1 triple negative mammary carcinoma models, we examined the therapeutic effects of oncolytic vesicular stomatitis virus (VSVΔM51) engineered to express reovirus-derived fusion associated small transmembrane proteins p14 (VSV-p14) or p15 (VSV-p15). These viruses were delivered alone or in combination with natural killer T (NKT) cell activation therapy mediated by adoptive transfer of α-galactosylceramide-loaded dendritic cells. RESULTS Treatment of primary 4T1 tumors with VSV-p14 or VSV-p15 alone increased immunogenic tumor cell death, attenuated tumor growth, and enhanced immune cell infiltration and activation compared to control oncolytic virus (VSV-GFP) treatments and untreated mice. When combined with NKT cell activation therapy, oncolytic VSV-p14 and VSV-p15 reduced metastatic lung burden to undetectable levels in all mice and generated immune memory as evidenced by enhanced in vitro recall responses (tumor killing and cytokine production) and impaired tumor growth upon rechallenge. CONCLUSION Combining NKT cell immunotherapy with enhanced oncolytic virotherapy increased anti-tumor immune targeting of lung metastasis and presents a promising treatment strategy for metastatic breast cancer.
Collapse
Affiliation(s)
- Adam Nelson
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, B3H 4R2, Halifax, NS, Canada
| | - Nichole McMullen
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
| | - Simon Gebremeskel
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, B3H 4R2, Halifax, NS, Canada
| | - Roberto De Antueno
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
| | - Duncan Mackenzie
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
| | - Roy Duncan
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
- Beatrice Hunter Cancer Research Institute, B3H 4R2, Halifax, NS, Canada
- Department of Biochemistry and Molecular Biology, Dalhousie University, B3H 4R2, Halifax, NS, Canada
- Department of Pediatrics, Dalhousie University, B3H 4R2, Halifax, NS, Canada
| | - Brent Johnston
- Department of Microbiology and Immunology, Dalhousie University, B3H 4R2, Halifax, NS, Canada.
- Beatrice Hunter Cancer Research Institute, B3H 4R2, Halifax, NS, Canada.
- Department of Pathology, Dalhousie University, B3H 4R2, Halifax, NS, Canada.
| |
Collapse
|
2
|
Engevik KA, Scribano FJ, Gebert JT, Perry JL, Crawford SE, Hyser JM. Distribution of P2Y and P2X purinergic receptor expression within the intestine. Am J Physiol Gastrointest Liver Physiol 2024; 326:G107-G119. [PMID: 37987757 PMCID: PMC11208031 DOI: 10.1152/ajpgi.00108.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 11/06/2023] [Accepted: 11/16/2023] [Indexed: 11/22/2023]
Abstract
Nucleotides are potent extracellular signaling molecules during homeostasis, infection, and injury due to their ability to activate purinergic receptors. The nucleotide ATP activates P2X receptors (P2RXs), whereas the nucleotides ADP, ATP, UTP, and UDP-glucose selectively activate different P2Y receptors (P2RYs). Several studies have established crucial roles for P2 receptors during intestinal inflammatory and infectious diseases, yet the most extensive characterization of purinergic signaling has focused on immune cells and the central and enteric nervous systems. As epithelial cells serve as the first barrier against irritants and infection, we hypothesized that the gut epithelium may express multiple purinergic receptors that respond to extracellular nucleotide signals. Using the Human Protein Atlas and Gut Cell Survey, we queried single-cell RNA sequencing (RNAseq) data for the P2 purinergic receptors in the small and large intestines. In silico analysis reveals robust mRNA expression of P2RY1, P2RY2, P2RY11, and P2RX4 throughout the gastrointestinal tract. Human intestinal organoids exhibited a similar expression pattern with a prominent expression of P2RY1, P2RY2, and P2RX4, but this purinergic receptor repertoire was not conserved in T84, Caco2, and HT29 intestinal epithelial cell lines. Finally, P2YR1 and P2YR2 agonists elicited robust calcium responses in human intestinal organoids, but calcium responses were weaker or absent in the cell lines. These findings suggest that the gastrointestinal epithelia respond to extracellular purinergic signaling via P2RY1, P2RY2, P2RY11, and P2RX4 receptors and highlight the benefit of using intestinal organoids as a model of intestinal purinergic signaling.NEW & NOTEWORTHY Several studies have revealed crucial roles for P2 receptors during inflammatory and infectious diseases, however, these have largely been demonstrated in immune cells and the enteric nervous system. Although epithelial cells serve as the first barrier against infection and inflammation, the role of purinergic signaling within the gastrointestinal tract remains largely unknown. This work expands our knowledge of purinergic receptor distribution and relative expression along the intestine.
Collapse
Affiliation(s)
- Kristen A Engevik
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States
| | - Francesca J Scribano
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States
| | - J Thomas Gebert
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States
| | - Jacob L Perry
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States
| | - Sue E Crawford
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States
| | - Joseph M Hyser
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States
| |
Collapse
|
3
|
Perry JL, Scribano FJ, Gebert JT, Engevik KA, Ellis JM, Hyser JM. Host IP 3R channels are dispensable for rotavirus Ca 2+ signaling but critical for intercellular Ca 2+ waves that prime uninfected cells for rapid virus spread. mBio 2024; 15:e0214523. [PMID: 38112482 PMCID: PMC10790754 DOI: 10.1128/mbio.02145-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Accepted: 11/15/2023] [Indexed: 12/21/2023] Open
Abstract
IMPORTANCE Many viruses exploit host Ca2+ signaling to facilitate their replication; however, little is known about how Ca2+ signals from different host and viral channels contribute to the overall dysregulation of Ca2+ signaling or promote virus replication. Using cells lacking IP3R, a host ER Ca2+ channel, we delineated intracellular Ca2+ signals within virus-infected cells and intercellular Ca2+ waves (ICWs), which increased Ca2+ signaling in neighboring, uninfected cells. In infected cells, IP3R was dispensable for rotavirus-induced Ca2+ signaling and replication, suggesting the rotavirus NSP4 viroporin supplies these signals. However, IP3R-mediated ICWs increase rotavirus replication kinetics and spread, indicating that the Ca2+ signals from the ICWs may prime nearby uninfected cells to better support virus replication upon eventual infection. This "pre-emptive priming" of uninfected cells by exploiting host intercellular pathways in the vicinity of virus-infected cells represents a novel mechanism for viral reprogramming of the host to gain a replication advantage.
Collapse
Affiliation(s)
- Jacob L. Perry
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Francesca J. Scribano
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - John T. Gebert
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Kristen A. Engevik
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Jenna M. Ellis
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Joseph M. Hyser
- Alkek Center for Metagenomic and Microbiome Research, Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
4
|
Scribano FJ, Engevik KA, Gebert JT, Hyser JM. Live-Cell Fluorescence Imaging for Virus-Host Interactions. Methods Mol Biol 2024; 2751:33-46. [PMID: 38265708 DOI: 10.1007/978-1-0716-3617-6_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024]
Abstract
Recent technological advances in microscopy have facilitated novel approaches to investigate host-pathogen interactions. In particular, improvements in both microscope hardware and engineered biosensors have helped to overcome barriers to live-cell imaging with fluorescence microscopy. Live fluorescent microscopy allows for the detection of discrete signaling events and protein localization, improving our ability to assess the effects of pharmacologic agents, microbes, or infection with high temporal resolution. Here we describe a protocol for long-term live-cell fluorescence imaging of virus infected cell lines.
Collapse
Affiliation(s)
- Francesca J Scribano
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kristen A Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - J Thomas Gebert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA.
- Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
5
|
Perry JL, Scribano FJ, Gebert JT, Engevik KA, Ellis JM, Hyser JM. The Inositol Trisphosphate Receptor (IP 3 R) is Dispensable for Rotavirus-induced Ca 2+ Signaling and Replication but Critical for Paracrine Ca 2+ Signals that Prime Uninfected Cells for Rapid Virus Spread. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.09.552719. [PMID: 37609335 PMCID: PMC10441394 DOI: 10.1101/2023.08.09.552719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Rotavirus is a leading cause of viral gastroenteritis. A hallmark of rotavirus infection is an increase in cytosolic Ca 2+ caused by the nonstructural protein 4 (NSP4). NSP4 is a viral ion channel that releases Ca 2+ from the endoplasmic reticulum (ER) and the increase in Ca 2+ signaling is critical for rotavirus replication. In addition to NSP4 itself, host inositol 1,4,5- trisphosphate receptor (IP 3 R) ER Ca 2+ channels may contribute to rotavirus-induced Ca 2+ signaling and by extension, virus replication. Thus, we set out to determine the role of IP 3 R Ca 2+ signaling during rotavirus infection using IP 3 R-knockout MA104-GCaMP6s cells (MA104- GCaMP6s-IP 3 R-KO), generated by CRISPR/Cas9 genome editing. Live Ca 2+ imaging showed that IP 3 R-KO did not reduce Ca 2+ signaling in infected cells but eliminated rotavirus-induced intercellular Ca 2+ waves (ICWs) and therefore the increased Ca 2+ signaling in surrounding, uninfected cells. Further, MA104-GCaMP6s-IP 3 R-TKO cells showed similar rotavirus susceptibility, single-cycle replication, and viral protein expression as parental MA104- GCaMP6s cells. However, MA104-GCaMP6s-IP 3 R-TKO cells exhibited significantly smaller rotavirus plaques, decreased multi-round replication kinetics, and delayed virus spread, suggesting that rotavirus-induced ICW Ca 2+ signaling stimulates virus replication and spread. Inhibition of ICWs by blocking the P2Y1 receptor also resulted in decreased rotavirus plaque size. Conversely, exogenous expression of P2Y1 in LLC-MK2-GCaMP6s cells, which natively lack P2Y1 and rotavirus ICWs, rescued the generation of rotavirus-induced ICWs and enabled plaque formation. In conclusion, this study shows that NSP4 Ca 2+ signals fully support rotavirus replication in individual cells; however, IP 3 R is critical for rotavirus-induced ICWs and virus spread by priming Ca 2+ -dependent pathways in surrounding cells. Importance Many viruses exploit host Ca 2+ signaling to facilitate their replication; however, little is known about how distinct types of Ca 2+ signals contribute to the overall dysregulation of Ca 2+ signaling or promote virus replication. Using cells lacking IP 3 R, a host ER Ca 2+ channel, we could differentiate between intracellular Ca 2+ signals within virus-infected cells and intercellular Ca 2+ waves (ICWs), which increase Ca 2+ signaling in neighboring, uninfected cells. In infected cells, IP 3 R was dispensable for rotavirus-induced Ca 2+ signaling and replication, suggesting the rotavirus NSP4 viroporin supplies these signals. However, IP 3 R-mediated ICWs increase rotavirus replication kinetics and spread, indicating that the Ca 2+ signals from the ICWs may prime nearby uninfected cells to better support virus replication upon eventual infection. This "pre-emptive priming" of uninfected cells by exploiting host intercellular pathways in the vicinity of virus-infected cells represents a novel mechanism for viral reprogramming of the host to gain a replication advantage.
Collapse
|
6
|
Abstract
Rotavirus (RV), the most common cause of gastroenteritis in children, carries a high economic and health burden worldwide. RV encodes six structural proteins and six nonstructural proteins (NSPs) that play different roles in viral replication. NSP4, a multifunctional protein involved in various viral replication processes, has two conserved N-glycosylation sites; however, the role of glycans remains elusive. Here, we used recombinant viruses generated by a reverse genetics system to determine the role of NSP4 N-glycosylation during viral replication and pathogenesis. The growth rate of recombinant viruses that lost one glycosylation site was as high as that of the wild-type virus. However, a recombinant virus that lost both glycosylation sites (glycosylation-defective virus) showed attenuated replication in cultured cell lines. Specifically, replications of glycosylation-defective virus in MA104 and HT29 cells were 10- and 100,000-fold lower, respectively, than that of the wild-type, suggesting that N-glycosylation of NSP4 plays a critical role in RV replication. The glycosylation-defective virus showed NSP4 mislocalization, delay of cytosolic Ca2+ elevation, and less viroplasm formation in MA104 cells; however, these impairments were not observed in HT29 cells. Further analysis revealed that assembly of glycosylation-defective virus was severely impaired in HT29 cells but not in MA104 cells, suggesting that RV replication mechanism is highly cell type dependent. In vivo mouse experiments also showed that the glycosylation-defective virus was less pathogenic than the wild-type virus. Taken together, the data suggest that N-glycosylation of NSP4 plays a vital role in viral replication and pathogenicity. IMPORTANCE Rotavirus is the main cause of gastroenteritis in young children and infants worldwide, contributing to 128,500 deaths each year. Here, we used a reverse genetics approach to examine the role of NSP4 N-glycosylation. An N-glycosylation-defective virus showed attenuated and cell-type-dependent replication in vitro. In addition, mice infected with the N-glycosylation-defective virus had less severe diarrhea than mice infected with the wild type. These results suggest that N-glycosylation affects viral replication and pathogenesis. Considering the reduced pathogenicity in vivo and the high propagation rate in MA104 cells, this glycosylation-defective virus could be an ideal live attenuated vaccine candidate.
Collapse
|
7
|
Smertina E, Carroll AJ, Boileau J, Emmott E, Jenckel M, Vohra H, Rolland V, Hands P, Hayashi J, Neave MJ, Liu JW, Hall RN, Strive T, Frese M. Lagovirus Non-structural Protein p23: A Putative Viroporin That Interacts With Heat Shock Proteins and Uses a Disulfide Bond for Dimerization. Front Microbiol 2022; 13:923256. [PMID: 35923397 PMCID: PMC9340658 DOI: 10.3389/fmicb.2022.923256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Accepted: 06/16/2022] [Indexed: 12/03/2022] Open
Abstract
The exact function(s) of the lagovirus non-structural protein p23 is unknown as robust cell culture systems for the Rabbit haemorrhagic disease virus (RHDV) and other lagoviruses have not been established. Instead, a range of in vitro and in silico models have been used to study p23, revealing that p23 oligomerizes, accumulates in the cytoplasm, and possesses a conserved C-terminal region with two amphipathic helices. Furthermore, the positional homologs of p23 in other caliciviruses have been shown to possess viroporin activity. Here, we report on the mechanistic details of p23 oligomerization. Site-directed mutagenesis revealed the importance of an N-terminal cysteine for dimerization. Furthermore, we identified cellular interactors of p23 using stable isotope labeling with amino acids in cell culture (SILAC)-based proteomics; heat shock proteins Hsp70 and 110 interact with p23 in transfected cells, suggesting that they ‘chaperone’ p23 proteins before their integration into cellular membranes. We investigated changes to the global transcriptome and proteome that occurred in infected rabbit liver tissue and observed changes to the misfolded protein response, calcium signaling, and the regulation of the endoplasmic reticulum (ER) network. Finally, flow cytometry studies indicate slightly elevated calcium concentrations in the cytoplasm of p23-transfected cells. Taken together, accumulating evidence suggests that p23 is a viroporin that might form calcium-conducting channels in the ER membranes.
Collapse
Affiliation(s)
- Elena Smertina
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
- Centre for Invasive Species Solutions, Canberra, ACT, Australia
| | - Adam J. Carroll
- RSB/RSC Joint Mass Spectrometry Facility, Research School of Chemistry, Australian National University, Acton, ACT, Australia
| | - Joseph Boileau
- RSB/RSC Joint Mass Spectrometry Facility, Research School of Chemistry, Australian National University, Acton, ACT, Australia
| | - Edward Emmott
- Centre for Proteome Research, Department of Biochemistry & Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Maria Jenckel
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
| | - Harpreet Vohra
- Imaging and Cytometry Facility, John Curtin School of Medical Research, Acton, ACT, Australia
| | - Vivien Rolland
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
| | - Philip Hands
- Agriculture and Food, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
| | - Junna Hayashi
- Research School of Chemistry, Australian National University, Acton, ACT, Australia
| | - Matthew J. Neave
- Australian Centre for Disease Preparedness, Commonwealth Scientific and Industrial Research Organisation, Geelong, VIC, Australia
| | - Jian-Wei Liu
- Land and Water, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
| | - Robyn N. Hall
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
- Centre for Invasive Species Solutions, Canberra, ACT, Australia
| | - Tanja Strive
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
- Centre for Invasive Species Solutions, Canberra, ACT, Australia
| | - Michael Frese
- Health and Biosecurity, Commonwealth Scientific and Industrial Research Organisation, Canberra, ACT, Australia
- Faculty of Science and Technology, University of Canberra, Canberra, ACT, Australia
- *Correspondence: Michael Frese,
| |
Collapse
|
8
|
Li S, Kim HY, Lee DJ, Park SH, Otsu K, Harada H, Jung YS, Jung HS. Inhibition of L-type voltage-gated calcium channel-mediated Ca 2+ influx suppresses the collective migration and invasion of ameloblastoma. Cell Prolif 2022; 55:e13305. [PMID: 35794842 PMCID: PMC9628225 DOI: 10.1111/cpr.13305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/15/2022] [Accepted: 06/20/2022] [Indexed: 11/28/2022] Open
Abstract
Objectives Ameloblastoma (AM) has been known as a benign but locally invasive tumour with high recurrence rates. Invasive behaviour of the AM results in destruction of the adjacent jawbone and the non‐detectable remnants during surgery, interrupting the complete elimination of cancer cells. Methods To explore novel targets for the tumour cell invasion, a transcriptomic analysis between AM and odontogenic keratocyst were performed through next‐generation sequencing in detail. Results Enrichment of CACNA1C gene (encoding Cav1.2) in AM, a subunit of the L‐type voltage‐gated calcium channel (VGCC) was observed for the first time. The expression and channel activity of Cav1.2 was confirmed by immunostaining and calcium imaging in the patient samples or primary cells. Verapamil, L‐type VGCC blocker revealed suppression of the Ca2+‐induced cell aggregation and collective invasion of AM cells in vitro. Furthermore, the effect of verapamil in suppressing AM invasion into the adjacent bone was confirmed through orthotopic xenograft model specifically. Conclusion Taken together, Cav1.2 maybe considered to be a therapeutic candidate to decrease the collective migration and invasion of AM.
Collapse
Affiliation(s)
- Shujin Li
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Hyun-Yi Kim
- NGeneS Inc, Ansan-si, Gyeonggi-do, South Korea
| | - Dong-Joon Lee
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, South Korea
| | - Sung-Ho Park
- Department of Oral & Maxillofacial Surgery, Yonsei University College of Dentistry, Seoul, South Korea
| | - Keishi Otsu
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University, Iwate, Japan
| | - Hidemitsu Harada
- Division of Developmental Biology and Regenerative Medicine, Department of Anatomy, Iwate Medical University, Iwate, Japan
| | - Young-Soo Jung
- Department of Oral & Maxillofacial Surgery, Yonsei University College of Dentistry, Seoul, South Korea
| | - Han-Sung Jung
- Division in Anatomy and Developmental Biology, Department of Oral Biology, Taste Research Center, Oral Science Research Center, BK21 FOUR Project, Yonsei University College of Dentistry, Seoul, South Korea
| |
Collapse
|
9
|
Huang W, Wu T, Xie C, Rayner CK, Priest C, Ebendorff‐Heidepriem H, Zhao J(T. Sensing Intra‐ and Extra‐Cellular Ca 2+ in the Islet of Langerhans. ADVANCED FUNCTIONAL MATERIALS 2022; 32. [DOI: 10.1002/adfm.202106020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Indexed: 12/19/2024]
Abstract
AbstractCalcium ions (Ca2+) take part in intra‐ and inter‐cellular signaling to mediate cellular functions. Sensing this ubiquitous messenger is instrumental in disentangling the specific functions of cellular sub‐compartments and/or intercellular communications. In this review, the authors first describe intra‐ and inter‐cellular Ca2+ signaling in relation to insulin secretion from the pancreatic islets, and then outline the development of diverse sensors, for example, chemically synthesized indicators, genetically encoded proteins, and ion‐selective microelectrodes, for intra‐ and extra‐cellular sensing of Ca2+. Particular emphasis is placed on emerging approaches in this field, such as low‐affinity Ca2+ indicators and unique Ca2+‐responsive composite materials. The authors conclude by remarking on the challenges and opportunities for further developments in this field, which may facilitate a more comprehensive understanding of Ca2+ signaling within and outside the islets, and its relevance in health and disease.
Collapse
Affiliation(s)
- Weikun Huang
- Adelaide Medical School Centre of Research Excellence in Translating Nutritional Science to Good Health The University of Adelaide Adelaide South Australia 5005 Australia
- Institute for Photonics and Advanced Sensing School of Physical Sciences ARC Centre of Excellence for Nanoscale BioPhotonics University of Adelaide Adelaide South Australia 5005 Australia
| | - Tongzhi Wu
- Adelaide Medical School Centre of Research Excellence in Translating Nutritional Science to Good Health The University of Adelaide Adelaide South Australia 5005 Australia
| | - Cong Xie
- Adelaide Medical School Centre of Research Excellence in Translating Nutritional Science to Good Health The University of Adelaide Adelaide South Australia 5005 Australia
| | - Christopher K. Rayner
- Adelaide Medical School Centre of Research Excellence in Translating Nutritional Science to Good Health The University of Adelaide Adelaide South Australia 5005 Australia
| | - Craig Priest
- Australian National Fabrication Facility and Future Industries Institute UniSA STEM University of South Australia Mawson Lakes South Australia 5095 Australia
| | - Heike Ebendorff‐Heidepriem
- Institute for Photonics and Advanced Sensing School of Physical Sciences ARC Centre of Excellence for Nanoscale BioPhotonics University of Adelaide Adelaide South Australia 5005 Australia
| | - Jiangbo (Tim) Zhao
- Institute for Photonics and Advanced Sensing School of Physical Sciences ARC Centre of Excellence for Nanoscale BioPhotonics University of Adelaide Adelaide South Australia 5005 Australia
- Department of Engineering Faculty of Science and Engineering University of Hull Hull HU6 7RX UK
| |
Collapse
|
10
|
Bohmwald K, Gálvez NMS, Andrade CA, Mora VP, Muñoz JT, González PA, Riedel CA, Kalergis AM. Modulation of Adaptive Immunity and Viral Infections by Ion Channels. Front Physiol 2021; 12:736681. [PMID: 34690811 PMCID: PMC8531258 DOI: 10.3389/fphys.2021.736681] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 09/10/2021] [Indexed: 12/15/2022] Open
Abstract
Most cellular functions require of ion homeostasis and ion movement. Among others, ion channels play a crucial role in controlling the homeostasis of anions and cations concentration between the extracellular and intracellular compartments. Calcium (Ca2+) is one of the most relevant ions involved in regulating critical functions of immune cells, allowing the appropriate development of immune cell responses against pathogens and tumor cells. Due to the importance of Ca2+ in inducing the immune response, some viruses have evolved mechanisms to modulate intracellular Ca2+ concentrations and the mobilization of this cation through Ca2+ channels to increase their infectivity and to evade the immune system using different mechanisms. For instance, some viral infections require the influx of Ca2+ through ionic channels as a first step to enter the cell, as well as their replication and budding. Moreover, through the expression of viral proteins on the surface of infected cells, Ca2+ channels function can be altered, enhancing the pathogen evasion of the adaptive immune response. In this article, we review those ion channels and ion transporters that are essential for the function of immune cells. Specifically, cation channels and Ca2+ channels in the context of viral infections and their contribution to the modulation of adaptive immune responses.
Collapse
Affiliation(s)
- Karen Bohmwald
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Nicolás M. S. Gálvez
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Catalina A. Andrade
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina P. Mora
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - José T. Muñoz
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo A. González
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Claudia A. Riedel
- Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Millennium Institute on Immunology and Immunotherapy, Universidad Andres Bello, Santiago, Chile
| | - Alexis M. Kalergis
- Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Millennium Institute on Immunology and Immunotherapy, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
11
|
Ihekweazu FD, Engevik MA, Ruan W, Shi Z, Fultz R, Engevik KA, Chang-Graham AL, Freeborn J, Park ES, Venable S, Horvath TD, Haidacher SJ, Haag AM, Goodwin A, Schady DA, Hyser JM, Spinler JK, Liu Y, Versalovic J. Bacteroides ovatus Promotes IL-22 Production and Reduces Trinitrobenzene Sulfonic Acid-Driven Colonic Inflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:704-719. [PMID: 33516788 PMCID: PMC8027925 DOI: 10.1016/j.ajpath.2021.01.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/31/2020] [Accepted: 01/06/2021] [Indexed: 02/06/2023]
Abstract
The intestinal microbiota influences the development and function of the mucosal immune system. However, the exact mechanisms by which commensal microbes modulate immunity is not clear. We previously demonstrated that commensal Bacteroides ovatus ATCC 8384 reduces mucosal inflammation. Herein, we aimed to identify immunomodulatory pathways employed by B. ovatus. In germ-free mice, mono-association with B. ovatus shifted the CD11b+/CD11c+ and CD103+/CD11c+ dendritic cell populations. Because indole compounds are known to modulate dendritic cells, B. ovatus cell-free supernatant was screened for tryptophan metabolites by liquid chromatography-tandem mass spectrometry and larger quantities of indole-3-acetic acid were detected. Analysis of cecal and fecal samples from germ-free and B. ovatus mono-associated mice confirmed that B. ovatus could elevate indole-3-acetic acid concentrations in vivo. Indole metabolites have previously been shown to stimulate immune cells to secrete the reparative cytokine IL-22. Addition of B. ovatus cell-free supernatant to immature bone marrow-derived dendritic cells stimulated IL-22 secretion. The ability of IL-22 to drive repair in the intestinal epithelium was confirmed using a physiologically relevant human intestinal enteroid model. Finally, B. ovatus shifted the immune cell populations in trinitrobenzene sulfonic acid-treated mice and up-regulated colonic IL-22 expression, effects that correlated with decreased inflammation. Our data suggest that B. ovatus-produced indole-3-acetic acid promotes IL-22 production by immune cells, yielding beneficial effects on colitis.
Collapse
Affiliation(s)
- Faith D Ihekweazu
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, Texas.
| | - Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Wenly Ruan
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Section of Gastroenterology, Hepatology, and Nutrition, Texas Children's Hospital, Houston, Texas
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Robert Fultz
- Department of Neuroscience, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston, Texas
| | - Kristen A Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | | | - Jasmin Freeborn
- Division of Gastroenterology, Department of Pediatrics, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas
| | - Evelyn S Park
- Division of Gastroenterology, Department of Pediatrics, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas
| | - Susan Venable
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Thomas D Horvath
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Sigmund J Haidacher
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Anthony M Haag
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Annie Goodwin
- Department of Pediatrics, The University of Texas Medical Branch at Galveston, Galveston, Texas
| | - Deborah A Schady
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Jennifer K Spinler
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Yuying Liu
- Division of Gastroenterology, Department of Pediatrics, The University of Texas Health Science Center at Houston McGovern Medical School, Houston, Texas
| | - James Versalovic
- Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| |
Collapse
|
12
|
Forro C, Caron D, Angotzi GN, Gallo V, Berdondini L, Santoro F, Palazzolo G, Panuccio G. Electrophysiology Read-Out Tools for Brain-on-Chip Biotechnology. MICROMACHINES 2021; 12:124. [PMID: 33498905 PMCID: PMC7912435 DOI: 10.3390/mi12020124] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/18/2021] [Accepted: 01/19/2021] [Indexed: 02/07/2023]
Abstract
Brain-on-Chip (BoC) biotechnology is emerging as a promising tool for biomedical and pharmaceutical research applied to the neurosciences. At the convergence between lab-on-chip and cell biology, BoC couples in vitro three-dimensional brain-like systems to an engineered microfluidics platform designed to provide an in vivo-like extrinsic microenvironment with the aim of replicating tissue- or organ-level physiological functions. BoC therefore offers the advantage of an in vitro reproduction of brain structures that is more faithful to the native correlate than what is obtained with conventional cell culture techniques. As brain function ultimately results in the generation of electrical signals, electrophysiology techniques are paramount for studying brain activity in health and disease. However, as BoC is still in its infancy, the availability of combined BoC-electrophysiology platforms is still limited. Here, we summarize the available biological substrates for BoC, starting with a historical perspective. We then describe the available tools enabling BoC electrophysiology studies, detailing their fabrication process and technical features, along with their advantages and limitations. We discuss the current and future applications of BoC electrophysiology, also expanding to complementary approaches. We conclude with an evaluation of the potential translational applications and prospective technology developments.
Collapse
Affiliation(s)
- Csaba Forro
- Tissue Electronics, Fondazione Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53-80125 Naples, Italy; (C.F.); (F.S.)
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | - Davide Caron
- Enhanced Regenerative Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (D.C.); (V.G.)
| | - Gian Nicola Angotzi
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (G.N.A.); (L.B.)
| | - Vincenzo Gallo
- Enhanced Regenerative Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (D.C.); (V.G.)
| | - Luca Berdondini
- Microtechnology for Neuroelectronics, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (G.N.A.); (L.B.)
| | - Francesca Santoro
- Tissue Electronics, Fondazione Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci, 53-80125 Naples, Italy; (C.F.); (F.S.)
| | - Gemma Palazzolo
- Enhanced Regenerative Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (D.C.); (V.G.)
| | - Gabriella Panuccio
- Enhanced Regenerative Medicine, Fondazione Istituto Italiano di Tecnologia, Via Morego, 30-16163 Genova, Italy; (D.C.); (V.G.)
| |
Collapse
|
13
|
Chang-Graham AL, Perry JL, Engevik MA, Engevik KA, Scribano FJ, Gebert JT, Danhof HA, Nelson JC, Kellen JS, Strtak AC, Sastri NP, Estes MK, Britton RA, Versalovic J, Hyser JM. Rotavirus induces intercellular calcium waves through ADP signaling. Science 2020; 370:370/6519/eabc3621. [PMID: 33214249 PMCID: PMC7957961 DOI: 10.1126/science.abc3621] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 10/16/2020] [Indexed: 01/14/2023]
Abstract
Rotavirus causes severe diarrheal disease in children by broadly dysregulating intestinal homeostasis. However, the underlying mechanism(s) of rotavirus-induced dysregulation remains unclear. We found that rotavirus-infected cells produce paracrine signals that manifested as intercellular calcium waves (ICWs), observed in cell lines and human intestinal enteroids. Rotavirus ICWs were caused by the release of extracellular adenosine 5'-diphosphate (ADP) that activated P2Y1 purinergic receptors on neighboring cells. ICWs were blocked by P2Y1 antagonists or CRISPR-Cas9 knockout of the P2Y1 receptor. Blocking the ADP signal reduced rotavirus replication, inhibited rotavirus-induced serotonin release and fluid secretion, and reduced diarrhea severity in neonatal mice. Thus, rotavirus exploited paracrine purinergic signaling to generate ICWs that amplified the dysregulation of host cells and altered gastrointestinal physiology to cause diarrhea.
Collapse
Affiliation(s)
- Alexandra L. Chang-Graham
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Jacob L. Perry
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Melinda A. Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, USA,Department of Pathology, Texas Children’s Hospital, USA
| | - Kristen A. Engevik
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Francesca J. Scribano
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - J. Thomas Gebert
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Heather A. Danhof
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Joel C. Nelson
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA
| | - Joseph S. Kellen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Alicia C. Strtak
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - Narayan P. Sastri
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA
| | - Mary K. Estes
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA,Department of Medicine, Gastroenterology and Hepatology, Baylor College of Medicine, USA
| | - Robert A. Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, USA,Department of Pathology, Texas Children’s Hospital, USA
| | - Joseph M. Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, USA,Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, USA,Corresponding author. Correspondence and requests for materials should be addressed to J.H.
| |
Collapse
|
14
|
Ion Channels as Therapeutic Targets for Viral Infections: Further Discoveries and Future Perspectives. Viruses 2020; 12:v12080844. [PMID: 32756358 PMCID: PMC7472218 DOI: 10.3390/v12080844] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 12/11/2022] Open
Abstract
Ion channels play key roles in almost all facets of cellular physiology and have emerged as key host cell factors for a multitude of viral infections. A catalogue of ion channel-blocking drugs have been shown to possess antiviral activity, some of which are in widespread human usage for ion channel-related diseases, highlighting new potential for drug repurposing. The emergence of ion channel–virus interactions has also revealed the intriguing possibility that channelopathies may explain some commonly observed virus induced pathologies. This field is rapidly evolving and an up-to-date summary of new discoveries can inform future perspectives. We herein discuss the role of ion channels during viral lifecycles, describe the recently identified ion channel drugs that can inhibit viral infections, and highlight the potential contribution of ion channels to virus-mediated disease.
Collapse
|
15
|
Engevik MA, Danhof HA, Chang-Graham AL, Spinler JK, Engevik KA, Herrmann B, Endres BT, Garey KW, Hyser JM, Britton RA, Versalovic J. Human intestinal enteroids as a model of Clostridioides difficile-induced enteritis. Am J Physiol Gastrointest Liver Physiol 2020; 318:G870-G888. [PMID: 32223302 PMCID: PMC7272722 DOI: 10.1152/ajpgi.00045.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Clostridioides difficile is an important nosocomial pathogen that produces toxins to cause life-threatening diarrhea and colitis. Toxins bind to epithelial receptors and promote the collapse of the actin cytoskeleton. C. difficile toxin activity is commonly studied in cancer-derived and immortalized cell lines. However, the biological relevance of these models is limited. Moreover, no model is available for examining C. difficile-induced enteritis, an understudied health problem. We hypothesized that human intestinal enteroids (HIEs) express toxin receptors and provide a new model to dissect C. difficile cytotoxicity in the small intestine. We generated biopsy-derived jejunal HIE and Vero cells, which stably express LifeAct-Ruby, a fluorescent label of F-actin, to monitor actin cytoskeleton rearrangement by live-cell microscopy. Imaging analysis revealed that toxins from pathogenic C. difficile strains elicited cell rounding in a strain-dependent manner, and HIEs were tenfold more sensitive to toxin A (TcdA) than toxin B (TcdB). By quantitative PCR, we paradoxically found that HIEs expressed greater quantities of toxin receptor mRNA and yet exhibited decreased sensitivity to toxins when compared with traditionally used cell lines. We reasoned that these differences may be explained by components, such as mucins, that are present in HIEs cultures, that are absent in immortalized cell lines. Addition of human-derived mucin 2 (MUC2) to Vero cells delayed cell rounding, indicating that mucus serves as a barrier to toxin-receptor binding. This work highlights that investigation of C. difficile infection in that HIEs can provide important insights into the intricate interactions between toxins and the human intestinal epithelium.NEW & NOTEWORTHY In this article, we developed a novel model of Clostridioides difficile-induced enteritis using jejunal-derived human intestinal enteroids (HIEs) transduced with fluorescently tagged F-actin. Using live-imaging, we identified that jejunal HIEs express high levels of TcdA and CDT receptors, are more sensitive to TcdA than TcdB, and secrete mucus, which delays toxin-epithelial interactions. This work also optimizes optically clear C. difficile-conditioned media suitable for live-cell imaging.
Collapse
Affiliation(s)
- Melinda A. Engevik
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Heather A. Danhof
- 3Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, Texas,4Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | | | - Jennifer K. Spinler
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Kristen A. Engevik
- 3Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, Texas,4Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - Beatrice Herrmann
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Bradley T. Endres
- 5Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas
| | - Kevin W. Garey
- 5Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas
| | - Joseph M. Hyser
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| | - Robert A. Britton
- 3Alkek Center for Metagenomic and Microbiome Research, Baylor College of Medicine, Houston, Texas,4Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas
| | - James Versalovic
- 1Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas,2Department of Pathology, Texas Children’s Hospital, Houston, Texas
| |
Collapse
|
16
|
Tomaro-Duchesneau C, LeValley SL, Roeth D, Sun L, Horrigan FT, Kalkum M, Hyser JM, Britton RA. Discovery of a bacterial peptide as a modulator of GLP-1 and metabolic disease. Sci Rep 2020; 10:4922. [PMID: 32188864 PMCID: PMC7080827 DOI: 10.1038/s41598-020-61112-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
Early work in rodents highlighted the gut microbiota’s importance in metabolic disease, including Type II Diabetes Mellitus (T2DM) and obesity. Glucagon-like peptide-1 (GLP-1), an incretin secreted by L-cells lining the gastrointestinal epithelium, has important functions: promoting insulin secretion, insulin sensitivity, and β-cell mass, while inhibiting gastric emptying and appetite. We set out to identify microbial strains with GLP-1 stimulatory activity as potential metabolic disease therapeutics. Over 1500 human-derived strains were isolated from healthy individuals and screened for GLP-1 modulation by incubating bacterial cell-free supernatants with NCI H716 L-cells. Approximately 45 strains capable of increasing GLP-1 were discovered. All GLP-1 positive strains were identified as Staphylococcus epidermidis by 16S rRNA sequencing. Mass spectrometry analysis identified a 3 kDa peptide, Hld (delta-toxin), present in GLP-1 positive supernatants but absent in GLP-1 neutral supernatants. Studies in NCI-H716 cells and human jejunal enteroids engineered to make more enteroendocrine cells demonstrated that Hld alone is sufficient to enhance GLP-1 secretion. When administered in high-fat-fed mice, Hld-producing S. epidermidis significantly reduced markers associated with obesity and T2DM. Further characterization of Hld suggests GLP-1 stimulatory action of Hld occurs via calcium signaling. The presented results identify a novel host-microbe interaction which may ultimately lead to the development of a microbial peptide-based therapeutic for metabolic disease.
Collapse
Affiliation(s)
- Catherine Tomaro-Duchesneau
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Stephanie L LeValley
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Daniel Roeth
- Department of Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Liang Sun
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Frank T Horrigan
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, United States of America
| | - Markus Kalkum
- Department of Molecular Imaging and Therapy, Beckman Research Institute of the City of Hope, Duarte, California, United States of America
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America
| | - Robert A Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, United States of America.
| |
Collapse
|
17
|
High-Throughput Fluorescence Assays for Ion Channels and GPCRs. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:27-72. [DOI: 10.1007/978-3-030-12457-1_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
18
|
In Vivo Two-photon Calcium Imaging in Dendrites of Rabies Virus-labeled V1 Corticothalamic Neurons. Neurosci Bull 2019; 36:545-553. [PMID: 31808041 DOI: 10.1007/s12264-019-00452-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 10/04/2019] [Indexed: 01/20/2023] Open
Abstract
Monitoring neuronal activity in vivo is critical to understanding the physiological or pathological functions of the brain. Two-photon Ca2+ imaging in vivo using a cranial window and specific neuronal labeling enables real-time, in situ, and long-term imaging of the living brain. Here, we constructed a recombinant rabies virus containing the Ca2+ indicator GCaMP6s along with the fluorescent protein DsRed2 as a baseline reference to ensure GCaMP6s signal reliability. This functional tracer was applied to retrogradely label specific V1-thalamus circuits and detect spontaneous Ca2+ activity in the dendrites of V1 corticothalamic neurons by in vivo two-photon Ca2+ imaging. Notably, we were able to record single-spine spontaneous Ca2+ activity in specific circuits. Distinct spontaneous Ca2+ dynamics in dendrites of V1 corticothalamic neurons were found for different V1-thalamus circuits. Our method can be applied to monitor Ca2+ dynamics in specific input circuits in vivo, and contribute to functional studies of defined neural circuits and the dissection of functional circuit connections.
Collapse
|
19
|
Rotavirus Calcium Dysregulation Manifests as Dynamic Calcium Signaling in the Cytoplasm and Endoplasmic Reticulum. Sci Rep 2019; 9:10822. [PMID: 31346185 PMCID: PMC6658527 DOI: 10.1038/s41598-019-46856-8] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 07/04/2019] [Indexed: 02/08/2023] Open
Abstract
Like many viruses, rotavirus (RV) dysregulates calcium homeostasis by elevating cytosolic calcium ([Ca2+]cyt) and decreasing endoplasmic reticulum (ER) stores. While an overall, monophasic increase in [Ca2+]cyt during RV infection has been shown, the nature of the RV-induced aberrant calcium signals and how they manifest over time at the single-cell level have not been characterized. Thus, we generated cell lines and human intestinal enteroids (HIEs) stably expressing cytosolic and/or ER-targeted genetically-encoded calcium indicators to characterize calcium signaling throughout RV infection by time-lapse imaging. We found that RV induces highly dynamic [Ca2+]cyt signaling that manifest as hundreds of discrete [Ca2+]cyt spikes, which increase during peak infection. Knockdown of nonstructural protein 4 (NSP4) attenuates the [Ca2+]cyt spikes, consistent with its role in dysregulating calcium homeostasis. RV-induced [Ca2+]cyt spikes were primarily from ER calcium release and were attenuated by inhibiting the store-operated calcium entry (SOCE) channel Orai1. RV-infected HIEs also exhibited prominent [Ca2+]cyt spikes that were attenuated by inhibiting SOCE, underlining the relevance of these [Ca2+]cyt spikes to gastrointestinal physiology and role of SOCE in RV pathophysiology. Thus, our discovery that RV increases [Ca2+]cyt by dynamic calcium signaling, establishes a new, paradigm-shifting understanding of the spatial and temporal complexity of virus-induced calcium signaling.
Collapse
|
20
|
Engevik MA, Luk B, Chang-Graham AL, Hall A, Herrmann B, Ruan W, Endres BT, Shi Z, Garey KW, Hyser JM, Versalovic J. Bifidobacterium dentium Fortifies the Intestinal Mucus Layer via Autophagy and Calcium Signaling Pathways. mBio 2019; 10:e01087-19. [PMID: 31213556 PMCID: PMC6581858 DOI: 10.1128/mbio.01087-19] [Citation(s) in RCA: 157] [Impact Index Per Article: 31.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/11/2019] [Indexed: 02/07/2023] Open
Abstract
Much remains unknown about how the intestinal microbiome interfaces with the protective intestinal mucus layer. Bifidobacterium species colonize the intestinal mucus layer and can modulate mucus production by goblet cells. However, select Bifidobacterium strains can also degrade protective glycans on mucin proteins. We hypothesized that the human-derived species Bifidobacterium dentium would increase intestinal mucus synthesis and expulsion, without extensive degradation of mucin glycans. In silico data revealed that B. dentium lacked the enzymes necessary to extensively degrade mucin glycans. This finding was confirmed by demonstrating that B. dentium could not use naive mucin glycans as primary carbon sources in vitro To examine B. dentium mucus modulation in vivo, Swiss Webster germfree mice were monoassociated with live or heat-killed B. dentium Live B. dentium-monoassociated mice exhibited increased colonic expression of goblet cell markers Krüppel-like factor 4 (Klf4), Trefoil factor 3 (Tff3), Relm-β, Muc2, and several glycosyltransferases compared to both heat-killed B. dentium and germfree counterparts. Likewise, live B. dentium-monoassociated colon had increased acidic mucin-filled goblet cells, as denoted by Periodic Acid-Schiff-Alcian Blue (PAS-AB) staining and MUC2 immunostaining. In vitro, B. dentium-secreted products, including acetate, were able to increase MUC2 levels in T84 cells. We also identified that B. dentium-secreted products, such as γ-aminobutyric acid (GABA), stimulated autophagy-mediated calcium signaling and MUC2 release. This work illustrates that B. dentium is capable of enhancing the intestinal mucus layer and goblet cell function via upregulation of gene expression and autophagy signaling pathways, with a net increase in mucin production.IMPORTANCE Microbe-host interactions in the intestine occur along the mucus-covered epithelium. In the gastrointestinal tract, mucus is composed of glycan-covered proteins, or mucins, which are secreted by goblet cells to form a protective gel-like structure above the epithelium. Low levels of mucin or alterations in mucin glycans are associated with inflammation and colitis in mice and humans. Although current literature links microbes to the modulation of goblet cells and mucins, the molecular pathways involved are not yet fully understood. Using a combination of gnotobiotic mice and mucus-secreting cell lines, we have identified a human-derived microbe, Bifidobacterium dentium, which adheres to intestinal mucus and secretes metabolites that upregulate the major mucin MUC2 and modulate goblet cell function. Unlike other Bifidobacterium species, B. dentium does not extensively degrade mucin glycans and cannot grow on mucin alone. This work points to the potential of using B. dentium and similar mucin-friendly microbes as therapeutic agents for intestinal disorders with disruptions in the mucus barrier.
Collapse
Affiliation(s)
- Melinda A Engevik
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Berkley Luk
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Alexandra L Chang-Graham
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Anne Hall
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Beatrice Herrmann
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Wenly Ruan
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Bradley T Endres
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Zhongcheng Shi
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| | - Kevin W Garey
- Department of Pharmacy Practice and Translational Research, University of Houston College of Pharmacy, Houston, Texas, USA
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - James Versalovic
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, Texas, USA
- Department of Pathology, Texas Children's Hospital, Houston, Texas, USA
| |
Collapse
|
21
|
Chang-Graham AL, Danhof HA, Engevik MA, Tomaro-Duchesneau C, Karandikar UC, Estes MK, Versalovic J, Britton RA, Hyser JM. Human Intestinal Enteroids With Inducible Neurogenin-3 Expression as a Novel Model of Gut Hormone Secretion. Cell Mol Gastroenterol Hepatol 2019; 8:209-229. [PMID: 31029854 PMCID: PMC6664234 DOI: 10.1016/j.jcmgh.2019.04.010] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 04/15/2019] [Accepted: 04/16/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Enteroendocrine cells (EECs) are specialized epithelial cells that produce molecules vital for intestinal homeostasis, but because of their limited numbers, in-depth functional studies have remained challenging. Human intestinal enteroids (HIEs) that are derived from intestinal crypt stem cells are biologically relevant in an in vitro model of the intestinal epithelium. HIEs contain all intestinal epithelial cell types; however, similar to the intestine, HIEs spontaneously produce few EECs, which limits their study. METHODS To increase the number of EECs in HIEs, we used lentivirus transduction to stably engineer jejunal HIEs with doxycycline-inducible expression of neurogenin-3 (NGN3), a transcription factor that drives EEC differentiation (tetNGN3-HIEs). We examined the impact of NGN3 induction on EECs by quantifying the increase in the enterochromaffin cells and other EEC subtypes. We functionally assessed secretion of serotonin and EEC hormones in response to norepinephrine and rotavirus infection. RESULTS Treating tetNGN3-HIEs with doxycycline induced a dose-dependent increase of chromogranin A (ChgA)-positive and serotonin-positive cells, showing increased enterochromaffin cell differentiation. Despite increased ChgA-positive cells, other differentiated cell types of the epithelium remained largely unchanged by gene expression and immunostaining. RNA sequencing of doxycycline-induced tetNGN3-HIEs identified increased expression of key hormones and enzymes associated with several other EEC subtypes. Doxycycline-induced tetNGN3-HIEs secreted serotonin, monocyte chemoattractant protein-1, glucose-dependent insulinotropic peptide, peptide YY, and ghrelin in response to norepinephrine and rotavirus infection, further supporting the presence of multiple EEC types. CONCLUSIONS We have combined HIEs and inducible-NGN3 expression to establish a flexible in vitro model system for functional studies of EECs in enteroids and advance the molecular and physiological investigation of EECs.
Collapse
Affiliation(s)
- Alexandra L Chang-Graham
- Department of Molecular Virology and Microbiology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas
| | - Heather A Danhof
- Department of Molecular Virology and Microbiology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas
| | - Melinda A Engevik
- Department of Pathology and Immunology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Catherine Tomaro-Duchesneau
- Department of Molecular Virology and Microbiology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas
| | - Umesh C Karandikar
- Department of Molecular Virology and Microbiology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas
| | - Mary K Estes
- Department of Molecular Virology and Microbiology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas; Department of Medicine, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas
| | - James Versalovic
- Department of Pathology and Immunology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Texas Children's Hospital, Houston, Texas
| | - Robert A Britton
- Department of Molecular Virology and Microbiology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas.
| | - Joseph M Hyser
- Department of Molecular Virology and Microbiology, Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas.
| |
Collapse
|
22
|
Stauffer W, Sheng H, Lim HN. EzColocalization: An ImageJ plugin for visualizing and measuring colocalization in cells and organisms. Sci Rep 2018; 8:15764. [PMID: 30361629 PMCID: PMC6202351 DOI: 10.1038/s41598-018-33592-8] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/01/2018] [Indexed: 02/07/2023] Open
Abstract
Insight into the function and regulation of biological molecules can often be obtained by determining which cell structures and other molecules they localize with (i.e. colocalization). Here we describe an open source plugin for ImageJ called EzColocalization to visualize and measure colocalization in microscopy images. EzColocalization is designed to be easy to use and customize for researchers with minimal experience in quantitative microscopy and computer programming. Features of EzColocalization include: (i) tools to select individual cells and organisms from images; (ii) filters to select specific types of cells and organisms based on physical parameters and signal intensity; (iii) heat maps and scatterplots to visualize the localization patterns of reporters; (iv) multiple metrics to measure colocalization for two or three reporters; (v) metric matrices to systematically measure colocalization at multiple combinations of signal intensity thresholds; and (vi) data tables that provide detailed information on each cell in a sample. These features make EzColocalization well-suited for experiments with low reporter signal, complex patterns of localization, and heterogeneous populations of cells and organisms.
Collapse
Affiliation(s)
- Weston Stauffer
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA, USA
| | - Huanjie Sheng
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA, USA
| | - Han N Lim
- Department of Integrative Biology, University of California Berkeley, Berkeley, CA, USA.
- Atomwise Inc., San Francisco, CA, USA.
| |
Collapse
|
23
|
Assessment of cytosolic free calcium changes during ceramide-induced cell death in MDA-MB-231 breast cancer cells expressing the calcium sensor GCaMP6m. Cell Calcium 2018; 72:39-50. [PMID: 29748132 DOI: 10.1016/j.ceca.2018.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Revised: 02/12/2018] [Accepted: 02/19/2018] [Indexed: 12/17/2022]
Abstract
Alterations in Ca2+ signaling can regulate key cancer hallmarks such as proliferation, invasiveness and resistance to cell death. Changes in the regulation of intracellular Ca2+ and specific components of Ca2+ influx are a feature of several cancers and/or cancer subtypes, including the basal-like breast cancer subtype, which has a poor prognosis. The development of genetically encoded calcium indicators, such as GCaMP6, represents an opportunity to measure changes in intracellular free Ca2+ during processes relevant to breast cancer progression that occur over long periods (e.g. hours), such as cell death. This study describes the development of a MDA-MB-231 breast cancer cell line stably expressing GCaMP6m. The cell line retained the key features of this aggressive basal-like breast cancer cell line. Using this model, we defined alterations in relative cytosolic free Ca2+ ([Ca2+]CYT) when the cells were treated with C2-ceramide. Cell death was measured simultaneously via assessment of propidium iodide permeability. Treatment with ceramide produced delayed and heterogeneous sustained increases in [Ca2+]CYT. Where cell death occurred, [Ca2+]CYT increases preceded cell death. The sustained increases in [Ca2+]CYT were not related to the rapid morphological changes induced by ceramide. Silencing of the plasma membrane Ca2+ ATPase isoform 1 (PMCA1) was associated with an augmentation in ceramide-induced increases in [Ca2+]CYT and also cell death. This work demonstrates the utility of GCaMP6 Ca2+ indicators for investigating [Ca2+]CYT changes in breast cancer cells during events relevant to tumor progression, which occur over hours rather than minutes.
Collapse
|
24
|
Mathews MA, Camp AJ, Murray AJ. Reviewing the Role of the Efferent Vestibular System in Motor and Vestibular Circuits. Front Physiol 2017; 8:552. [PMID: 28824449 PMCID: PMC5539236 DOI: 10.3389/fphys.2017.00552] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 07/17/2017] [Indexed: 12/31/2022] Open
Abstract
Efferent circuits within the nervous system carry nerve impulses from the central nervous system to sensory end organs. Vestibular efferents originate in the brainstem and terminate on hair cells and primary afferent fibers in the semicircular canals and otolith organs within the inner ear. The function of this efferent vestibular system (EVS) in vestibular and motor coordination though, has proven difficult to determine, and remains under debate. We consider current literature that implicate corollary discharge from the spinal cord through the efferent vestibular nucleus (EVN), and hint at a potential role in overall vestibular plasticity and compensation. Hypotheses range from differentiating between passive and active movements at the level of vestibular afferents, to EVS activation under specific behavioral and environmental contexts such as arousal, predation, and locomotion. In this review, we summarize current knowledge of EVS circuitry, its effects on vestibular hair cell and primary afferent activity, and discuss its potential functional roles.
Collapse
Affiliation(s)
- Miranda A Mathews
- Sensory Systems and Integration Laboratory, Bosch Institute, Discipline of Biomedical Science, University of SydneySydney, NSW, Australia
| | - Aaron J Camp
- Sensory Systems and Integration Laboratory, Bosch Institute, Discipline of Biomedical Science, University of SydneySydney, NSW, Australia
| | - Andrew J Murray
- Sainsbury Wellcome Centre for Neural Circuits and Behaviour, University College LondonLondon, United Kingdom
| |
Collapse
|
25
|
Bassett JJ, Monteith GR. Genetically Encoded Calcium Indicators as Probes to Assess the Role of Calcium Channels in Disease and for High-Throughput Drug Discovery. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 79:141-171. [PMID: 28528667 DOI: 10.1016/bs.apha.2017.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The calcium ion (Ca2+) is an important signaling molecule implicated in many cellular processes, and the remodeling of Ca2+ homeostasis is a feature of a variety of pathologies. Typical methods to assess Ca2+ signaling in cells often employ small molecule fluorescent dyes, which are sometimes poorly suited to certain applications such as assessment of cellular processes, which occur over long periods (hours or days) or in vivo experiments. Genetically encoded calcium indicators are a set of tools available for the measurement of Ca2+ changes in the cytosol and subcellular compartments, which circumvent some of the inherent limitations of small molecule Ca2+ probes. Recent advances in genetically encoded calcium sensors have greatly increased their ability to provide reliable monitoring of Ca2+ changes in mammalian cells. New genetically encoded calcium indicators have diverse options in terms of targeting, Ca2+ affinity and fluorescence spectra, and this will further enhance their potential use in high-throughput drug discovery and other assays. This review will outline the methods available for Ca2+ measurement in cells, with a focus on genetically encoded calcium sensors. How these sensors will improve our understanding of the deregulation of Ca2+ handling in disease and their application to high-throughput identification of drug leads will also be discussed.
Collapse
Affiliation(s)
- John J Bassett
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia
| | - Gregory R Monteith
- School of Pharmacy, The University of Queensland, Brisbane, QLD, Australia; Mater Research, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
26
|
Mossman K. Methods related to molecular virology. Methods 2015; 90:1-2. [PMID: 26589248 DOI: 10.1016/j.ymeth.2015.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Affiliation(s)
- Karen Mossman
- Department of Biochemistry and Biomedical Sciences, McMaster Immunology Research Center, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine, McMaster Immunology Research Center, Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|