1
|
Li D, Wang Z, Wang W, Zheng Z, Wei H, Su Q, Yang M, Zhao Y, Zhang X, Yu X, Zhang P, Shu Y. The therapeutic effect of Yinqiaosan decoction against influenza A virus infection by regulating T cell receptor signaling pathway. Heliyon 2024; 10:e36178. [PMID: 39253133 PMCID: PMC11382312 DOI: 10.1016/j.heliyon.2024.e36178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/08/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
Background Yinqiaosan decoction (YQSD), a traditional Chinese medicinal recipe, has been employed to treat influenza in China for approximately 300 years. Objective Our study aimed to explore the mechanisms of YQSD against influenza via in vivo and in vitro experimental studies. Study design and methods UHPLC-Q-TOF-MS/MS was utilized to examine the substances of the YQSD. The chemical components of YQSD detected by UHPLC-Q-TOF-MS/MS were used for network pharmacology analysis. The antiviral effect of YQSD in vivo was investigated. The potential mechanisms of YQSD in combating influenza, which were predicted from network pharmacology analysis, were validated in vitro. Results By use of UHPLC-Q-TOF-MS/MS, 97 compounds were identified from YQSD. Network pharmacology analysis revealed that the therapeutic effect of YQSD against influenza may be associated with the regulation of T cell receptors (TCR) and Phosphoinositide 3-Kinase (PI3K)- protein kinase B (Akt) signaling pathways. Treatment with YQSD significantly prolonged the mean survival time of the mice and reduced lung injury due to the influenza A virus in vivo. It was discovered that YQSD efficiently inhibited the expression of inflammation-related cytokines. Moreover, YQSD has been found to significantly reduce the expression levels of cluster of differentiation 3 (CD3), monocyte chemoattractant protein-1 (MCP-1), and H1N1 virus nucleoprotein (NP), and prevent the decrease of epithelial cadherin (E-cadherin) protein. In addition, YQSD can inhibit the phosphorylation of the zeta chain of T cell receptor-associated protein kinase 70 (ZAP70) and PI3K proteins in vitro. Conclusion The capacity of YQSD to suppress viral multiplication and inflammatory response by modulating T cell immunity may explain its effect against influenza viral pneumonia, which may involve the regulation of TCR and PI3K signaling pathways.
Collapse
Affiliation(s)
- Danting Li
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Zekun Wang
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Wenlei Wang
- Institute of Translational Medicine &Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Zhihui Zheng
- Institute of Translational Medicine &Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Hailin Wei
- Institute of Translational Medicine &Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Qin Su
- Institute of Translational Medicine &Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Mengmeng Yang
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Yimeng Zhao
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Xinyuan Zhang
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Xiaocong Yu
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
| | - Pinghu Zhang
- Institute of Translational Medicine &Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Medical College, Yangzhou University, Yangzhou, 225009, China
| | - Yachun Shu
- Department of Pharmacy, Affiliated Hospital of Nanjing University of Chinese Medicine & Jiangsu Province Hospital of Chinese Medicine, Nanjing, 210029, China
- Jiangsu Province Seaside Rehabilitation Hospital, Lianyungang, 222042, China
| |
Collapse
|
2
|
Crum RJ, Huckestien BR, Dwyer G, Mathews L, Nascari DG, Hussey GS, Turnquist HR, Alcorn JF, Badylak SF. Mitigation of influenza-mediated inflammation by immunomodulatory matrix-bound nanovesicles. SCIENCE ADVANCES 2023; 9:eadf9016. [PMID: 37205761 PMCID: PMC10198633 DOI: 10.1126/sciadv.adf9016] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 04/14/2023] [Indexed: 05/21/2023]
Abstract
Cytokine storm describes a life-threatening, systemic inflammatory syndrome characterized by elevated levels of proinflammatory cytokines and immune cell hyperactivation associated with multi-organ dysfunction. Matrix-bound nanovesicles (MBV) are a subclass of extracellular vesicle shown to down-regulate proinflammatory immune responses. The objective of this study was to assess the efficacy of MBV in mediating influenza-induced acute respiratory distress syndrome and cytokine storm in a murine model. Intravenous administration of MBV decreased influenza-mediated total lung inflammatory cell density, proinflammatory macrophage frequencies, and proinflammatory cytokines at 7 and 21 days following viral inoculation. MBV decreased long-lasting alveolitis and the proportion of lung undergoing inflammatory tissue repair at day 21. MBV increased the proportion of activated anti-viral CD4+ and CD8+ T cells at day 7 and memory-like CD62L+ CD44+, CD4+, and CD8+ T cells at day 21. These results show immunomodulatory properties of MBV that may benefit the treatment of viral-mediated pulmonary inflammation with applicability to other viral diseases such as SARS-CoV-2.
Collapse
Affiliation(s)
- Raphael J. Crum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brydie R. Huckestien
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Gaelen Dwyer
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Lisa Mathews
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - David G. Nascari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - George S. Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Heth R. Turnquist
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - John F. Alcorn
- Division of Pulmonary Medicine, Department of Pediatrics, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Toebbe JT, Genter MB. An Update on Sphingosine-1-Phosphate and Lysophosphatidic Acid Receptor Transcripts in Rodent Olfactory Mucosa. Int J Mol Sci 2022; 23:ijms23084343. [PMID: 35457160 PMCID: PMC9032240 DOI: 10.3390/ijms23084343] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/01/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
Olfactory neurons connect the external environment and the brain, allowing the translocation of materials from the nasal cavity into the brain. The olfactory system is involved in SARS-CoV-2 infections; early in the pandemic declared in 2020, a loss of the sense of smell was found in many infected patients. Attention has also been focused on the role that the olfactory epithelium appears to play in the entry of the SARS-CoV-2 virus into the brain. Specifically, SARS-CoV-2 enters cells via the angiotensin-converting enzyme 2 protein (ACE2), which is found on supporting cells in the olfactory epithelium. The intranasal administration of sphingosine has been proposed to prevent the binding of SARS-CoV-2 to ACE2. Further, sphingosine-1-phosphate (S1P) receptors appear to facilitate the entry of SARS-CoV-2 into the brain. The goal of these studies was to characterize S1P receptor expression status in rodent olfactory mucosa. The expression of receptors for a related sphingolipid, lysophosphatidic acid (LPA), was also assessed. The results confirm previous reports of S1P1 and S1P3 receptor expression, as well as LPA receptor 1, in mouse olfactory mucosa; moreover, they extend the previous findings to identify additional S1P and LPA receptor transcripts in rat and mouse olfactory mucosa, as well as in cultured olfactory neurons. These findings may enhance the utility of rodent models in identifying agonists and/or antagonists of S1P and LPA receptors that may block the entry of SARS-CoV-2 and other viruses into nasal epithelial cells, and prevent transmission from the nasal cavity into the brain.
Collapse
|
4
|
Bala N, McGurk AI, Zilch T, Rup AN, Carter EM, Leddon SA, Fowell DJ. T cell activation niches-Optimizing T cell effector function in inflamed and infected tissues. Immunol Rev 2021; 306:164-180. [PMID: 34859453 PMCID: PMC9218983 DOI: 10.1111/imr.13047] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 11/06/2021] [Indexed: 12/29/2022]
Abstract
Successful immunity to infection, malignancy, and tissue damage requires the coordinated recruitment of numerous immune cell subsets to target tissues. Once within the target tissue, effector T cells rely on local chemotactic cues and structural cues from the tissue matrix to navigate the tissue, interact with antigen-presenting cells, and release effector cytokines. This highly dynamic process has been "caught on camera" in situ by intravital multiphoton imaging. Initial studies revealed a surprising randomness to the pattern of T cell migration through inflamed tissues, behavior thought to facilitate chance encounters with rare antigen-bearing cells. Subsequent tissue-wide visualization has uncovered a high degree of spatial preference when it comes to T cell activation. Here, we discuss the basic tenants of a successful effector T cell activation niche, taking cues from the dynamics of Tfh positioning in the lymph node germinal center. In peripheral tissues, steady-state microanatomical organization may direct the location of "pop-up" de novo activation niches, often observed as perivascular clusters, that support early effector T cell activation. These perivascular activation niches appear to be regulated by site-specific chemokines that coordinate the recruitment of dendritic cells and other innate cells for local T cell activation, survival, and optimized effector function.
Collapse
Affiliation(s)
- Noor Bala
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Alexander I McGurk
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Tiago Zilch
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Anastasia N Rup
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Evan M Carter
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Scott A Leddon
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| | - Deborah J Fowell
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, USA
| |
Collapse
|
5
|
Myers MA, Smith AP, Lane LC, Moquin DJ, Aogo R, Woolard S, Thomas P, Vogel P, Smith AM. Dynamically linking influenza virus infection kinetics, lung injury, inflammation, and disease severity. eLife 2021; 10:68864. [PMID: 34282728 PMCID: PMC8370774 DOI: 10.7554/elife.68864] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Influenza viruses cause a significant amount of morbidity and mortality. Understanding host immune control efficacy and how different factors influence lung injury and disease severity are critical. We established and validated dynamical connections between viral loads, infected cells, CD8+ T cells, lung injury, inflammation, and disease severity using an integrative mathematical model-experiment exchange. Our results showed that the dynamics of inflammation and virus-inflicted lung injury are distinct and nonlinearly related to disease severity, and that these two pathologic measurements can be independently predicted using the model-derived infected cell dynamics. Our findings further indicated that the relative CD8+ T cell dynamics paralleled the percent of the lung that had resolved with the rate of CD8+ T cell-mediated clearance rapidly accelerating by over 48,000 times in 2 days. This complimented our analyses showing a negative correlation between the efficacy of innate and adaptive immune-mediated infected cell clearance, and that infection duration was driven by CD8+ T cell magnitude rather than efficacy and could be significantly prolonged if the ratio of CD8+ T cells to infected cells was sufficiently low. These links between important pathogen kinetics and host pathology enhance our ability to forecast disease progression, potential complications, and therapeutic efficacy.
Collapse
Affiliation(s)
- Margaret A Myers
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| | - Amanda P Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| | - Lindey C Lane
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| | - David J Moquin
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, United States
| | - Rosemary Aogo
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| | - Stacie Woolard
- Flow Cytometry Core, St. Jude Children's Research Hospital, Memphis, United States
| | - Paul Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, United States
| | - Peter Vogel
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, United States
| | - Amber M Smith
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, United States
| |
Collapse
|
6
|
Pan Y, Gao F, Zhao S, Han J, Chen F. Role of the SphK-S1P-S1PRs pathway in invasion of the nervous system by SARS-CoV-2 infection. Clin Exp Pharmacol Physiol 2021; 48:637-650. [PMID: 33565127 PMCID: PMC8014301 DOI: 10.1111/1440-1681.13483] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 01/29/2021] [Accepted: 02/02/2021] [Indexed: 01/08/2023]
Abstract
Global spread of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is still ongoing. Before an effective vaccine is available, the development of potential treatments for resultant coronavirus disease 2019 (COVID-19) is crucial. One of the disease hallmarks is hyper-inflammatory responses, which usually leads to a severe lung disease. Patients with COVID-19 also frequently suffer from neurological symptoms such as acute diffuse encephalomyelitis, brain injury and psychiatric complications. The metabolic pathway of sphingosine-1-phosphate (S1P) is a dynamic regulator of various cell types and disease processes, including the nervous system. It has been demonstrated that S1P and its metabolic enzymes, regulating neuroinflammation and neurogenesis, exhibit important functions during viral infection. S1P receptor 1 (S1PR1) analogues including AAL-R and RP-002 inhibit pathophysiological responses at the early stage of H1N1 virus infection and then play a protective role. Fingolimod (FTY720) is an S1P receptor modulator and is being tested for treating COVID-19. Our review provides an overview of SARS-CoV-2 infection and critical role of the SphK-S1P-SIPR pathway in invasion of SARS-CoV-2 infection, particularly in the central nervous system (CNS). This may help design therapeutic strategies based on the S1P-mediated signal transduction, and the adjuvant therapeutic effects of S1P analogues to limit or prevent the interaction between the host and SARS-CoV-2, block the spread of the SARS-CoV-2, and consequently treat related complications in the CNS.
Collapse
Affiliation(s)
- Yuehai Pan
- Department of Hand and Foot SurgeryThe Affiliated Hospital of Qingdao UniversityShangdongChina
| | - Fei Gao
- Department of Hand and Foot SurgeryThe Affiliated Hospital of Qingdao UniversityShangdongChina
| | - Shuai Zhao
- Department of AnesthesiologyBonn UniversityBonnGermany
| | - Jinming Han
- Department of Clinical NeuroscienceKarolinska InstitutetStockholmSweden
| | - Fan Chen
- Department of NeurosurgeryThe Affiliated Hospital of Qingdao UniversityShangdongChina
| |
Collapse
|
7
|
Prakash S, Srivastava R, Coulon PG, Dhanushkodi NR, Chentoufi AA, Tifrea DF, Edwards RA, Figueroa CJ, Schubl SD, Hsieh L, Buchmeier MJ, Bouziane M, Nesburn AB, Kuppermann BD, BenMohamed L. Genome-Wide B Cell, CD4 +, and CD8 + T Cell Epitopes That Are Highly Conserved between Human and Animal Coronaviruses, Identified from SARS-CoV-2 as Targets for Preemptive Pan-Coronavirus Vaccines. THE JOURNAL OF IMMUNOLOGY 2021; 206:2566-2582. [PMID: 33911008 DOI: 10.4049/jimmunol.2001438] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/16/2021] [Indexed: 02/06/2023]
Abstract
Over the last two decades, there have been three deadly human outbreaks of coronaviruses (CoVs) caused by SARS-CoV, MERS-CoV, and SARS-CoV-2, which has caused the current COVID-19 global pandemic. All three deadly CoVs originated from bats and transmitted to humans via various intermediate animal reservoirs. It remains highly possible that other global COVID pandemics will emerge in the coming years caused by yet another spillover of a bat-derived SARS-like coronavirus (SL-CoV) into humans. Determining the Ag and the human B cells, CD4+ and CD8+ T cell epitope landscapes that are conserved among human and animal coronaviruses should inform in the development of future pan-coronavirus vaccines. In the current study, using several immunoinformatics and sequence alignment approaches, we identified several human B cell and CD4+ and CD8+ T cell epitopes that are highly conserved in 1) greater than 81,000 SARS-CoV-2 genome sequences identified in 190 countries on six continents; 2) six circulating CoVs that caused previous human outbreaks of the common cold; 3) nine SL-CoVs isolated from bats; 4) nine SL-CoV isolated from pangolins; 5) three SL-CoVs isolated from civet cats; and 6) four MERS strains isolated from camels. Furthermore, the identified epitopes: 1) recalled B cells and CD4+ and CD8+ T cells from both COVID-19 patients and healthy individuals who were never exposed to SARS-CoV-2, and 2) induced strong B cell and T cell responses in humanized HLA-DR1/HLA-A*02:01 double-transgenic mice. The findings pave the way to develop a preemptive multiepitope pan-coronavirus vaccine to protect against past, current, and future outbreaks.
Collapse
Affiliation(s)
- Swayam Prakash
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Ruchi Srivastava
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Pierre-Gregoire Coulon
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Nisha R Dhanushkodi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Aziz A Chentoufi
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Delia F Tifrea
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA
| | - Robert A Edwards
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA
| | - Cesar J Figueroa
- Division of Trauma, Burns, Critical Care, and Acute Care Surgery, Department of Surgery, School of Medicine, University of California Irvine, Irvine, CA
| | - Sebastian D Schubl
- Division of Trauma, Burns, Critical Care, and Acute Care Surgery, Department of Surgery, School of Medicine, University of California Irvine, Irvine, CA
| | - Lanny Hsieh
- Division of Infectious Diseases and Hospitalist Program, Department of Medicine, School of Medicine, University of California Irvine, Irvine, CA
| | - Michael J Buchmeier
- Center for Virus Research, Division of Infectious Disease, School of Medicine, University of California Irvine, Irvine, CA
| | | | - Anthony B Nesburn
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Baruch D Kuppermann
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA
| | - Lbachir BenMohamed
- Laboratory of Cellular and Molecular Immunology, Gavin Herbert Eye Institute, School of Medicine, University of California Irvine, Irvine, CA; .,Center for Virus Research, Division of Infectious Disease, School of Medicine, University of California Irvine, Irvine, CA.,Institute for Immunology, School of Medicine, University of California Irvine, Irvine, CA
| |
Collapse
|
8
|
Melssen MM, Lindsay RS, Stasiak K, Rodriguez AB, Briegel AM, Cyranowski S, Rutkowski MR, Conaway MR, Melief CJM, van der Burg SH, Eyo U, Slingluff CL, Engelhard VH. Differential Expression of CD49a and CD49b Determines Localization and Function of Tumor-Infiltrating CD8 + T Cells. Cancer Immunol Res 2021; 9:583-597. [PMID: 33619119 DOI: 10.1158/2326-6066.cir-20-0427] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 10/26/2020] [Accepted: 02/18/2021] [Indexed: 11/16/2022]
Abstract
CD8+ T-cell infiltration and effector activity in tumors are correlated with better overall survival of patients, suggesting that the ability of T cells to enter and remain in contact with tumor cells supports tumor control. CD8+ T cells express the collagen-binding integrins CD49a and CD49b, but little is known about their function or how their expression is regulated in the tumor microenvironment (TME). Here, we found that tumor-infiltrating CD8+ T cells initially expressed CD49b, gained CD49a, and then lost CD49b over the course of tumor outgrowth. This differentiation sequence was driven by antigen-independent elements in the TME, although T-cell receptor (TCR) stimulation further increased CD49a expression. Expression of exhaustion markers and CD49a associated temporally but not mechanistically. Intratumoral CD49a-expressing CD8+ T cells failed to upregulate TCR-dependent Nur77 expression, whereas CD69 was constitutively expressed, consistent with both a lack of productive antigen engagement and a tissue-resident memory-like phenotype. Imaging T cells in live tumor slices revealed that CD49a increased their motility, especially of those in close proximity to tumor cells, suggesting that it may interfere with T-cell recognition of tumor cells by distracting them from productive engagement, although we were not able to augment productive engagement by short-term CD49a blockade. CD49b also promoted relocalization of T cells at a greater distance from tumor cells. Thus, our results demonstrate that expression of these integrins affects T-cell trafficking and localization in tumors via distinct mechanisms, and suggests a new way in which the TME, and likely collagen, could promote tumor-infiltrating CD8+ T-cell dysfunction.
Collapse
Affiliation(s)
- Marit M Melssen
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia.,Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Robin S Lindsay
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Katarzyna Stasiak
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Anthony B Rodriguez
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Amanda M Briegel
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Salwador Cyranowski
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Melanie R Rutkowski
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| | - Mark R Conaway
- Department of Public Health Sciences, University of Virginia, Charlottesville, Virginia
| | - Cornelis J M Melief
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands.,ISA Pharmaceutical, Leiden, the Netherlands
| | - Sjoerd H van der Burg
- Department of Medical Oncology, Oncode Institute, Leiden University Medical Center, Leiden, the Netherlands
| | - Ukpong Eyo
- Department of Neuroscience, University of Virginia, Charlottesville, Virginia
| | - Craig L Slingluff
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia.,Department of Surgery, University of Virginia, Charlottesville, Virginia
| | - Victor H Engelhard
- Carter Immunology Center, University of Virginia, Charlottesville, Virginia. .,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia
| |
Collapse
|
9
|
Prakash S, Srivastava R, Coulon PG, Dhanushkodi NR, Chentoufi AA, Tifrea DF, Edwards RA, Figueroa CJ, Schubl SD, Hsieh L, Buchmeier MJ, Bouziane M, Nesburn AB, Kuppermann BD, BenMohamed L. Genome-Wide Asymptomatic B-Cell, CD4 + and CD8 + T-Cell Epitopes, that are Highly Conserved Between Human and Animal Coronaviruses, Identified from SARS-CoV-2 as Immune Targets for Pre-Emptive Pan-Coronavirus Vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 33024971 DOI: 10.1101/2020.09.27.316018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Over the last two decades, there have been three deadly human outbreaks of Coronaviruses (CoVs) caused by emerging zoonotic CoVs: SARS-CoV, MERS-CoV, and the latest highly transmissible and deadly SARS-CoV-2, which has caused the current COVID-19 global pandemic. All three deadly CoVs originated from bats, the natural hosts, and transmitted to humans via various intermediate animal reservoirs. Because there is currently no universal pan-Coronavirus vaccine available, two worst-case scenarios remain highly possible: (1) SARS-CoV-2 mutates and transforms into a seasonal "flu-like" global pandemic; and/or (2) Other global COVID-like pandemics will emerge in the coming years, caused by yet another spillover of an unknown zoonotic bat-derived SARS-like Coronavirus (SL-CoV) into an unvaccinated human population. Determining the antigen and epitope landscapes that are conserved among human and animal Coronaviruses as well as the repertoire, phenotype and function of B cells and CD4 + and CD8 + T cells that correlate with resistance seen in asymptomatic COVID-19 patients should inform in the development of pan-Coronavirus vaccines 1 . In the present study, using several immuno-informatics and sequence alignment approaches, we identified several human B-cell, CD4 + and CD8 + T cell epitopes that are highly conserved in: ( i ) greater than 81,000 SARS-CoV-2 human strains identified to date in 190 countries on six continents; ( ii ) six circulating CoVs that caused previous human outbreaks of the "Common Cold"; ( iii ) five SL-CoVs isolated from bats; ( iv ) five SL-CoV isolated from pangolins; ( v ) three SL-CoVs isolated from Civet Cats; and ( vi ) four MERS strains isolated from camels. Furthermore, we identified cross-reactive asymptomatic epitopes that: ( i ) recalled B cell, CD4 + and CD8 + T cell responses from both asymptomatic COVID-19 patients and healthy individuals who were never exposed to SARS-CoV-2; and ( ii ) induced strong B cell and T cell responses in "humanized" Human Leukocyte Antigen (HLA)-DR/HLA-A*02:01 double transgenic mice. The findings herein pave the way to develop a pre-emptive multi-epitope pan-Coronavirus vaccine to protect against past, current, and potential future outbreaks.
Collapse
|
10
|
Meacci E, Garcia-Gil M, Pierucci F. SARS-CoV-2 Infection: A Role for S1P/S1P Receptor Signaling in the Nervous System? Int J Mol Sci 2020; 21:E6773. [PMID: 32942748 PMCID: PMC7556035 DOI: 10.3390/ijms21186773] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 09/07/2020] [Accepted: 09/11/2020] [Indexed: 02/07/2023] Open
Abstract
The recent coronavirus disease (COVID-19) is still spreading worldwide. The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the virus responsible for COVID-19, binds to its receptor angiotensin-converting enzyme 2 (ACE2), and replicates within the cells of the nasal cavity, then spreads along the airway tracts, causing mild clinical manifestations, and, in a majority of patients, a persisting loss of smell. In some individuals, SARS-CoV-2 reaches and infects several organs, including the lung, leading to severe pulmonary disease. SARS-CoV-2 induces neurological symptoms, likely contributing to morbidity and mortality through unknown mechanisms. Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid with pleiotropic properties and functions in many tissues, including the nervous system. S1P regulates neurogenesis and inflammation and it is implicated in multiple sclerosis (MS). Notably, Fingolimod (FTY720), a modulator of S1P receptors, has been approved for the treatment of MS and is being tested for COVID-19. Here, we discuss the putative role of S1P on viral infection and in the modulation of inflammation and survival in the stem cell niche of the olfactory epithelium. This could help to design therapeutic strategies based on S1P-mediated signaling to limit or overcome the host-virus interaction, virus propagation and the pathogenesis and complications involving the nervous system.
Collapse
Affiliation(s)
- Elisabetta Meacci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Viale GB Morgagni 50, 50134 Firenze, Italy;
- Interuniversity Institute of Myology, University of Firenze, 50134 Firenze, Italy
| | - Mercedes Garcia-Gil
- Unit of Physiology, Department of Biology, University of Pisa, via S. Zeno 31, 56127 Pisa, Italy;
- Interdepartmental Research Center “Nutraceuticals and Food for Health”, University of Pisa, 56127 Pisa, Italy
| | - Federica Pierucci
- Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Firenze, Viale GB Morgagni 50, 50134 Firenze, Italy;
- Interuniversity Institute of Myology, University of Firenze, 50134 Firenze, Italy
| |
Collapse
|
11
|
Malo CS, Hickman HD. Tracing Antiviral CD8 + T Cell Responses Using In Vivo Imaging. THE JOURNAL OF IMMUNOLOGY 2020; 203:775-781. [PMID: 31383748 DOI: 10.4049/jimmunol.1900232] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/29/2019] [Indexed: 12/25/2022]
Abstract
Scientists have long valued the power of in vivo observation to answer fundamental biological questions. Over the last 20 years, the application and evolution of intravital microscopy (IVM) has vastly increased our ability to directly visualize immune responses as they are occurring in vivo after infection or immunization. Many IVM strategies employ a strong multiphoton laser that penetrates deeply into the tissues of living, anesthetized mice, allowing the precise tracking of the movement of cells as they navigate complex tissue environments. In the realm of viral infections, IVM has been applied to better understand many critical phases of effector T cell responses, from activation in the draining lymph node, to the execution of effector functions, and finally to the development of tissue-resident memory. In this review, we discuss seminal studies incorporating IVM that have advanced our understanding of the biology of antiviral CD8+ T cells.
Collapse
Affiliation(s)
- Courtney S Malo
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
12
|
Ueki H, Wang IH, Zhao D, Gunzer M, Kawaoka Y. Multicolor two-photon imaging of in vivo cellular pathophysiology upon influenza virus infection using the two-photon IMPRESS. Nat Protoc 2020; 15:1041-1065. [PMID: 31996843 PMCID: PMC7086515 DOI: 10.1038/s41596-019-0275-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/03/2019] [Indexed: 12/14/2022]
Abstract
In vivo two-photon imaging is a valuable technique for studies of viral pathogenesis and host responses to infection in vivo. In this protocol, we describe a methodology for analyzing influenza virus-infected lung in vivo by two-photon imaging microscopy. We describe the surgical procedure, how to stabilize the lung, and an approach to analyzing the data. Further, we provide a database of fluorescent dyes, antibodies, and reporter mouse lines that can be used in combination with a reporter influenza virus (Color-flu) for multicolor analysis. Setup of this model typically takes ~30 min and enables the observation of influenza virus-infected lungs for >4 h during the acute phase of the inflammation and at least 1 h in the lethal phase. This imaging system, which we termed two-photon IMPRESS (imaging pathophysiology research system), is broadly applicable to analyses of other respiratory pathogens and reveals disease progression at the cellular level in vivo.
Collapse
Affiliation(s)
- Hiroshi Ueki
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - I-Hsuan Wang
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Dongming Zhao
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People's Republic of China
| | - Matthias Gunzer
- Institute for Experimental Immunology and Imaging, University Hospital, University Duisburg-Essen, Essen, Germany
| | - Yoshihiro Kawaoka
- Division of Virology, Department of Microbiology and Immunology, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Department of Special Pathogens, International Research Center for Infectious Diseases, Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Department of Pathobiological Sciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA.
| |
Collapse
|
13
|
Marciniak A, Camp SM, Garcia JGN, Polt R. An update on sphingosine-1-phosphate receptor 1 modulators. Bioorg Med Chem Lett 2018; 28:3585-3591. [PMID: 30409535 DOI: 10.1016/j.bmcl.2018.10.042] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/22/2018] [Accepted: 10/25/2018] [Indexed: 12/14/2022]
Abstract
Sphingolipids represent an essential class of lipids found in all eukaryotes, and strongly influence cellular signal transduction. Autoimmune diseases like asthma and multiple sclerosis (MS) are mediated by the sphingosine-1-phosphate receptor 1 (S1P1) to express a variety of symptoms and disease patterns. Inspired by its natural substrate, an array of artificial sphingolipid derivatives has been developed to target this specific G protein-coupled receptor (GPCR) in an attempt to suppress autoimmune disorders. FTY720, also known as fingolimod, is the first oral disease-modifying therapy for MS on the market. In pursuit of improved stability, bioavailability, and efficiency, structural analogues of this initial prodrug have emerged over time. This review covers a brief introduction to the sphingolipid metabolism, the mechanism of action on S1P1, and an updated overview of synthetic sphingosine S1P1 agonists.
Collapse
Affiliation(s)
- Alexander Marciniak
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, United States.
| | - Sara M Camp
- Department of Medicine, The University of Arizona, Tucson, AZ 85724, United States.
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona, Tucson, AZ 85724, United States.
| | - Robin Polt
- Department of Chemistry & Biochemistry, The University of Arizona, Tucson, AZ 85721, United States.
| |
Collapse
|
14
|
Davidson S. Treating Influenza Infection, From Now and Into the Future. Front Immunol 2018; 9:1946. [PMID: 30250466 PMCID: PMC6139312 DOI: 10.3389/fimmu.2018.01946] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 08/07/2018] [Indexed: 12/15/2022] Open
Abstract
Influenza viruses (IVs) are a continual threat to global health. The high mutation rate of the IV genome makes this virus incredibly successful, genetic drift allows for annual epidemics which result in thousands of deaths and millions of hospitalizations. Moreover, the emergence of new strains through genetic shift (e.g., swine-origin influenza A) can cause devastating global outbreaks of infection. Neuraminidase inhibitors (NAIs) are currently used to treat IV infection and act directly on viral proteins to halt IV spread. However, effectivity is limited late in infection and drug resistance can develop. New therapies which target highly conserved features of IV such as antibodies to the stem region of hemagglutinin or the IV RNA polymerase inhibitor: Favipiravir are currently in clinical trials. Compared to NAIs, these treatments have a higher tolerance for resistance and a longer therapeutic window and therefore, may prove more effective. However, clinical and experimental evidence has demonstrated that it is not just viral spread, but also the host inflammatory response and damage to the lung epithelium which dictate the outcome of IV infection. Therapeutic regimens for IV infection should therefore also regulate the host inflammatory response and protect epithelial cells from unnecessary cell death. Anti-inflammatory drugs such as etanercept, statins or cyclooxygenase enzyme 2 inhibitors may temper IV induced inflammation, demonstrating the possibility of repurposing these drugs as single or adjunct therapies for IV infection. IV binds to sialic acid receptors on the host cell surface to initiate infection and productive IV replication is primarily restricted to airway epithelial cells. Accordingly, targeting therapies to the epithelium will directly inhibit IV spread while minimizing off target consequences, such as over activation of immune cells. The neuraminidase mimic Fludase cleaves sialic acid receptors from the epithelium to inhibit IV entry to cells. While type III interferons activate an antiviral gene program in epithelial cells with minimal perturbation to the IV specific immune response. This review discusses the above-mentioned candidate anti-IV therapeutics and others at the preclinical and clinical trial stage.
Collapse
Affiliation(s)
- Sophia Davidson
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia
| |
Collapse
|
15
|
Mrass P, Oruganti SR, Fricke GM, Tafoya J, Byrum JR, Yang L, Hamilton SL, Miller MJ, Moses ME, Cannon JL. ROCK regulates the intermittent mode of interstitial T cell migration in inflamed lungs. Nat Commun 2017; 8:1010. [PMID: 29044117 PMCID: PMC5647329 DOI: 10.1038/s41467-017-01032-2] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 08/14/2017] [Indexed: 12/27/2022] Open
Abstract
Effector T cell migration through tissues can enable control of infection or mediate inflammatory damage. Nevertheless, the molecular mechanisms that regulate migration of effector T cells within the interstitial space of inflamed lungs are incompletely understood. Here, we show T cell migration in a mouse model of acute lung injury with two-photon imaging of intact lung tissue. Computational analysis indicates that T cells migrate with an intermittent mode, switching between confined and almost straight migration, guided by lung-associated vasculature. Rho-associated protein kinase (ROCK) is required for both high-speed migration and straight motion. By contrast, inhibition of Gαi signaling with pertussis toxin affects speed but not the intermittent migration of lung-infiltrating T cells. Computational modeling shows that an intermittent migration pattern balances both search area and the duration of contacts between T cells and target cells. These data identify that ROCK-dependent intermittent T cell migration regulates tissue-sampling during acute lung injury. ROCK is associated with T cell movement in lymph nodes. Here the authors use an LPS lung damage model and two-photon imaging to show that CD8+ T cells in lung tissue engage in ROCK-dependent fast linear migration alternating with bursts of slower confined migration that together optimize contact with target cells.
Collapse
Affiliation(s)
- Paulus Mrass
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Sreenivasa Rao Oruganti
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - G Matthew Fricke
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Justyna Tafoya
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Mathematics, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Janie R Byrum
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA
| | - Lihua Yang
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Samantha L Hamilton
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Mark J Miller
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, St Louis, MO, 63110, USA
| | - Melanie E Moses
- Department of Computer Science, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,Department of Biology, University of New Mexico, 1 University of New Mexico, Albuquerque, NM, 87131, USA.,External Faculty, Santa Fe Institute, 1399 Hyde Park Road, Santa Fe, NM, 87501, USA
| | - Judy L Cannon
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, MSC 08 4660, 1 University of New Mexico, Albuquerque, NM, 87131, USA.
| |
Collapse
|
16
|
Arish M, Alaidarous M, Ali R, Akhter Y, Rub A. Implication of sphingosine-1-phosphate signaling in diseases: molecular mechanism and therapeutic strategies. J Recept Signal Transduct Res 2017; 37:437-446. [PMID: 28758826 DOI: 10.1080/10799893.2017.1358282] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Sphingosine-1-phosphate signaling is emerging as a critical regulator of cellular processes that is initiated by the intracellular production of bioactive lipid molecule, sphingosine-1-phosphate. Binding of sphingosine-1-phosphate to its extracellular receptors activates diverse downstream signaling that play a critical role in governing physiological processes. Increasing evidence suggests that this signaling pathway often gets impaired during pathophysiological and diseased conditions and hence manipulation of this signaling pathway may be beneficial in providing treatment. In this review, we summarized the recent findings of S1P signaling pathway and the versatile role of the participating candidates in context with several disease conditions. Finally, we discussed its possible role as a novel drug target in different diseases.
Collapse
Affiliation(s)
- Mohd Arish
- a Infection and Immunity Lab, Department of Biotechnology , Jamia Millia Islamia (A Central University) , New Delhi , India
| | - Mohammed Alaidarous
- b Department of Medical Laboratory Sciences, College of Applied Medical Sciences , Majmaah University , Al Majmaah , Saudi Arabia
| | - Rahat Ali
- a Infection and Immunity Lab, Department of Biotechnology , Jamia Millia Islamia (A Central University) , New Delhi , India
| | - Yusuf Akhter
- c Centre for Computational Biology & Bioinformatics, School of Life Sciences , Central University of Himachal Pradesh , Shahpur, Kangra , India
| | - Abdur Rub
- a Infection and Immunity Lab, Department of Biotechnology , Jamia Millia Islamia (A Central University) , New Delhi , India.,b Department of Medical Laboratory Sciences, College of Applied Medical Sciences , Majmaah University , Al Majmaah , Saudi Arabia
| |
Collapse
|
17
|
Regulatory T Cells in Skin Facilitate Epithelial Stem Cell Differentiation. Cell 2017; 169:1119-1129.e11. [PMID: 28552347 DOI: 10.1016/j.cell.2017.05.002] [Citation(s) in RCA: 483] [Impact Index Per Article: 60.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 03/22/2017] [Accepted: 04/28/2017] [Indexed: 12/17/2022]
Abstract
The maintenance of tissue homeostasis is critically dependent on the function of tissue-resident immune cells and the differentiation capacity of tissue-resident stem cells (SCs). How immune cells influence the function of SCs is largely unknown. Regulatory T cells (Tregs) in skin preferentially localize to hair follicles (HFs), which house a major subset of skin SCs (HFSCs). Here, we mechanistically dissect the role of Tregs in HF and HFSC biology. Lineage-specific cell depletion revealed that Tregs promote HF regeneration by augmenting HFSC proliferation and differentiation. Transcriptional and phenotypic profiling of Tregs and HFSCs revealed that skin-resident Tregs preferentially express high levels of the Notch ligand family member, Jagged 1 (Jag1). Expression of Jag1 on Tregs facilitated HFSC function and efficient HF regeneration. Taken together, our work demonstrates that Tregs in skin play a major role in HF biology by promoting the function of HFSCs.
Collapse
|
18
|
Mohammed S, Harikumar KB. Sphingosine 1-Phosphate: A Novel Target for Lung Disorders. Front Immunol 2017; 8:296. [PMID: 28352271 PMCID: PMC5348531 DOI: 10.3389/fimmu.2017.00296] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 03/01/2017] [Indexed: 01/11/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is involved in a wide range of cellular processes, which include proliferation, apoptosis, lymphocyte egress, endothelial barrier function, angiogenesis, and inflammation. S1P is produced by two isoenzymes, namely, sphingosine kinase 1 and 2 (SphK1 and 2) and once produced, S1P can act both in an autocrine and paracrine manner. S1P can be dephosphorylated back to sphingosine by two phosphatases (SGPP 1 and 2) or can be irreversibly cleaved by S1P lyase. S1P has a diverse range of functions, which is mediated in a receptor dependent, through G-protein coupled receptors (S1PR1-5) or receptor independent manner, through intracellular targets such as HDACs and TRAF2. The involvement of S1P signaling has been confirmed in various disease conditions including lung diseases. The SphK inhibitors and S1PR modulators are currently under clinical trials for different pathophysiological conditions. There is a significant effort in targeting various components of S1P signaling for several diseases. This review focuses on the ways in which S1P signaling can be therapeutically targeted in lung disorders.
Collapse
Affiliation(s)
- Sabira Mohammed
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , India
| | - K B Harikumar
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology , Thiruvananthapuram , India
| |
Collapse
|
19
|
Hickman HD. New insights into antiviral immunity gained through intravital imaging. Curr Opin Virol 2017; 22:59-63. [PMID: 28081484 DOI: 10.1016/j.coviro.2016.11.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 11/22/2016] [Indexed: 01/25/2023]
Abstract
Viral infections pose an ongoing challenge for mankind. Much of our knowledge of the immune response to viral infections comes from ex vivo analyses of infected animals, which provide important yet static information about events occurring within the host. Recently, a relatively new technique known as intravital microscopy (IVM) has been applied to the study of antiviral immunity. Intravital imaging affords a unique, real-time view of both viral dynamics and the ensuing immune response (along with their interplay) in the living animal. This review details some of the newest observations about the antiviral immune response gained using IVM.
Collapse
Affiliation(s)
- Heather D Hickman
- Laboratory of Viral Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, United States.
| |
Collapse
|
20
|
Lambert Emo K, Hyun YM, Reilly E, Barilla C, Gerber S, Fowell D, Kim M, Topham DJ. Live Imaging of Influenza Infection of the Trachea Reveals Dynamic Regulation of CD8+ T Cell Motility by Antigen. PLoS Pathog 2016; 12:e1005881. [PMID: 27644089 PMCID: PMC5028057 DOI: 10.1371/journal.ppat.1005881] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 08/19/2016] [Indexed: 12/31/2022] Open
Abstract
During a primary influenza infection, cytotoxic CD8+ T cells need to infiltrate the infected airways and engage virus-infected epithelial cells. The factors that regulate T cell motility in the infected airway tissue are not well known. To more precisely study T cell infiltration of the airways, we developed an experimental model system using the trachea as a site where live imaging can be performed. CD8+ T cell motility was dynamic with marked changes in motility on different days of the infection. In particular, significant changes in average cell velocity and confinement were evident on days 8–10 during which the T cells abruptly but transiently increase velocity on day 9. Experiments to distinguish whether infection itself or antigen affect motility revealed that it is antigen, not active infection per se that likely affects these changes as blockade of peptide/MHC resulted in increased velocity. These observations demonstrate that influenza tracheitis provides a robust experimental foundation to study molecular regulation of T cell motility during acute virus infection. Influenza virus infects the cells that line the trachea and lung airways. Virus-specific cytotoxic (cell killing) T cells are the first line of adaptive immunity responsible for elimination of infected cells. We studied the cell movement, or motility, of these T cells responding to infection in the mouse trachea. Multiphoton live imaging was used to observe the cells in real time in intact tissue and measure their movement both quantitatively and qualitatively. The behavior of the CD8+ T cells responding to influenza infection was highly variable depending on the day after infection the imaging was performed. The most dramatic changes occurred after infectious virus was eliminated from the tissue, triggering a substantial shift in cell motility between days 8 and 9. Blocking peptide/MHC complexes with antibodies reversed cell arrest, increased velocities, and reduced confinement, similar to the changes observed from days 8 to 9. This suggested antigen-presentation persists after virus clearance with continued T cell engagement, and that T cell motility in the infected tissue is dynamically regulated by the infection and the presence of antigen-bearing cells in particular. In addition, these studies establish the trachea as a suitable site for live imaging of immune responses to virus infection.
Collapse
Affiliation(s)
- Kris Lambert Emo
- David H. Smith Center for Vaccine Biology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Young-min Hyun
- David H. Smith Center for Vaccine Biology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Emma Reilly
- David H. Smith Center for Vaccine Biology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Christopher Barilla
- David H. Smith Center for Vaccine Biology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Scott Gerber
- Department of Surgery, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Deborah Fowell
- David H. Smith Center for Vaccine Biology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - Minsoo Kim
- David H. Smith Center for Vaccine Biology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
| | - David J. Topham
- David H. Smith Center for Vaccine Biology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- Department of Microbiology & Immunology, University of Rochester School of Medicine and Dentistry, Rochester, New York, United States of America
- * E-mail:
| |
Collapse
|
21
|
Stolp B, Melican K. Microbial pathogenesis revealed by intravital microscopy: pros, cons and cautions. FEBS Lett 2016; 590:2014-26. [PMID: 26938770 DOI: 10.1002/1873-3468.12122] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/15/2016] [Accepted: 02/29/2016] [Indexed: 12/22/2022]
Abstract
Intravital multiphoton imaging allows visualization of infections and pathogenic mechanisms within intact organs in their physiological context. Today, most organs of mice and rats are applicable to in vivo or ex vivo imaging, opening completely new avenues for many researchers. Advances in fluorescent labeling of pathogens and infected cells, as well as improved small animal models for human pathogens, led to the increased application of in vivo imaging in infectious diseases research in recent years. Here, we review the latest literature on intravital or ex vivo imaging of viral and bacterial infections and critically discuss requirements, benefits and drawbacks of applied animal models, labeling strategies, and imaged organs.
Collapse
Affiliation(s)
- Bettina Stolp
- Heidelberg University Hospital, Center of Infectious Diseases, Integrative Virology, Heidelberg, Germany
| | - Keira Melican
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
22
|
Arish M, Husein A, Kashif M, Saleem M, Akhter Y, Rub A. Sphingosine-1-phosphate signaling: unraveling its role as a drug target against infectious diseases. Drug Discov Today 2015; 21:133-142. [PMID: 26456576 DOI: 10.1016/j.drudis.2015.09.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 09/23/2015] [Accepted: 09/29/2015] [Indexed: 12/25/2022]
Abstract
Sphingosine-1-phosphate (S1P) signaling is reported in variety of cell types, including immune, endothelial and cancerous cells. It is emerging as a crucial regulator of cellular processes, such as apoptosis, cell proliferation, migration, differentiation and so on. This signaling pathway is initiated by the intracellular production and secretion of S1P through a cascade of enzymatic reactions. Binding of S1P to different S1P receptors (S1PRs) activates different downstream signaling pathways that regulate the cellular functions differentially depending upon the cell type. An accumulating body of evidence suggests that S1P metabolism and signaling is often impaired during infectious diseases; thus, its manipulation might be helpful in the treatment of such diseases. In this review, we summarize recent advances in our understanding of the S1P signaling pathway and its candidature as a novel drug target against infectious diseases.
Collapse
Affiliation(s)
- Mohd Arish
- Infection and Immunity Lab, Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Atahar Husein
- Infection and Immunity Lab, Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Mohammad Kashif
- Infection and Immunity Lab, Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India
| | - Mohammed Saleem
- Department of Life Sciences, National Institute of Technology, Rourkela, Odisha 769008, India
| | - Yusuf Akhter
- School of Life Sciences, Central University of Himachal Pradesh, Temporary Academic Block, Shahpur, Kangra, HP 176216, India
| | - Abdur Rub
- Infection and Immunity Lab, Department of Biotechnology, Jamia Millia Islamia (A Central University), New Delhi 110025, India.
| |
Collapse
|
23
|
Pichyangkul S, Yongvanitchit K, Limsalakpetch A, Kum-Arb U, Im-Erbsin R, Boonnak K, Thitithayanont A, Jongkaewwattana A, Wiboon-ut S, Mongkolsirichaikul D, Mahanonda R, Spring M, Chuang I, Mason CJ, Saunders DL. Tissue Distribution of Memory T and B Cells in Rhesus Monkeys following Influenza A Infection. THE JOURNAL OF IMMUNOLOGY 2015; 195:4378-86. [PMID: 26408671 DOI: 10.4049/jimmunol.1501702] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 09/02/2015] [Indexed: 01/16/2023]
Abstract
Studies of influenza-specific immune responses in humans have largely assessed systemic responses involving serum Ab and peripheral blood T cell responses. However, recent evidence indicates that tissue-resident memory T (TRM) cells play an important role in local murine intrapulmonary immunity. Rhesus monkeys were pulmonary exposed to 2009 pandemic H1N1 virus at days 0 and 28 and immune responses in different tissue compartments were measured. All animals were asymptomatic postinfection. Although only minimal memory immune responses were detected in peripheral blood, a high frequency of influenza nucleoprotein-specific memory T cells was detected in the lung at the "contraction phase," 49-58 d after second virus inoculation. A substantial proportion of lung nucleoprotein-specific memory CD8(+) T cells expressed CD103 and CD69, phenotypic markers of TRM cells. Lung CD103(+) and CD103(-) memory CD8(+) T cells expressed similar levels of IFN-γ and IL-2. Unlike memory T cells, spontaneous Ab secreting cells and memory B cells specific to influenza hemagglutinin were primarily observed in the mediastinal lymph nodes. Little difference in systemic and local immune responses against influenza was observed between young adult (6-8 y) and old animals (18-28 y). Using a nonhuman primate model, we revealed substantial induction of local T and B cell responses following 2009 pandemic H1N1 infection. Our study identified a subset of influenza-specific lung memory T cells characterized as TRM cells in rhesus monkeys. The rhesus monkey model may be useful to explore the role of TRM cells in local tissue protective immunity after rechallenge and vaccination.
Collapse
Affiliation(s)
- Sathit Pichyangkul
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand;
| | - Kosol Yongvanitchit
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | | | - Utaiwan Kum-Arb
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Rawiwan Im-Erbsin
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Kobporn Boonnak
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Arunee Thitithayanont
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | - Anan Jongkaewwattana
- National Center for Genetic Engineering and Biotechnology, Pathum Thani 12120, Thailand; and
| | - Suwimon Wiboon-ut
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok 10400, Thailand
| | | | | | - Michele Spring
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Ilin Chuang
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - Carl J Mason
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| | - David L Saunders
- Armed Forces Research Institute of Medical Sciences, Bangkok 10400, Thailand
| |
Collapse
|
24
|
Weinger JG, Greenberg ML, Matheu MP, Parker I, Walsh CM, Lane TE, Cahalan MD. Two-photon imaging of cellular dynamics in the mouse spinal cord. J Vis Exp 2015. [PMID: 25742043 DOI: 10.3791/52580] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Two-photon (2P) microscopy is utilized to reveal cellular dynamics and interactions deep within living, intact tissues. Here, we present a method for live-cell imaging in the murine spinal cord. This technique is uniquely suited to analyze neural precursor cell (NPC) dynamics following transplantation into spinal cords undergoing neuroinflammatory demyelinating disorders. NPCs migrate to sites of axonal damage, proliferate, differentiate into oligodendrocytes, and participate in direct remyelination. NPCs are thereby a promising therapeutic treatment to ameliorate chronic demyelinating diseases. Because transplanted NPCs migrate to the damaged areas on the ventral side of the spinal cord, traditional intravital 2P imaging is impossible, and only information on static interactions was previously available using histochemical staining approaches. Although this method was generated to image transplanted NPCs in the ventral spinal cord, it can be applied to numerous studies of transplanted and endogenous cells throughout the entire spinal cord. In this article, we demonstrate the preparation and imaging of a spinal cord with enhanced yellow fluorescent protein-expressing axons and enhanced green fluorescent protein-expressing transplanted NPCs.
Collapse
Affiliation(s)
- Jason G Weinger
- Molecular Biology and Biochemistry, University of California, Irvine
| | | | - Melanie P Matheu
- University of California San Francisco Diabetes Center, University of California, San Francisco
| | - Ian Parker
- Neurobiology and Behavior, University of California, Irvine
| | - Craig M Walsh
- Molecular Biology and Biochemistry, University of California, Irvine
| | | | | |
Collapse
|
25
|
Gendron D, Lemay AM, Tremblay C, Lai LJ, Langlois A, Bernatchez É, Flamand N, Blanchet MR, Don AS, Bossé Y, Bissonnette É, Marsolais D. Treatment with a sphingosine analog after the inception of house dust mite-induced airway inflammation alleviates key features of experimental asthma. Respir Res 2015; 16:7. [PMID: 25645346 PMCID: PMC4330646 DOI: 10.1186/s12931-015-0180-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 01/21/2015] [Indexed: 12/23/2022] Open
Abstract
Background In vivo phosphorylation of sphingosine analogs with their ensuing binding and activation of their cell-surface sphingosine-1-phosphate receptors is regarded as the main immunomodulatory mechanism of this new class of drugs. Prophylactic treatment with sphingosine analogs interferes with experimental asthma by impeding the migration of dendritic cells to draining lymph nodes. However, whether these drugs can also alleviate allergic airway inflammation after its onset remains to be determined. Herein, we investigated to which extent and by which mechanisms the sphingosine analog AAL-R interferes with key features of asthma in a murine model during ongoing allergic inflammation induced by Dermatophagoides pteronyssinus. Methods BALB/c mice were exposed to either D. pteronyssinus or saline, intranasally, once-daily for 10 consecutive days. Mice were treated intratracheally with either AAL-R, its pre-phosphorylated form AFD-R, or the vehicle before every allergen challenge over the last four days, i.e. after the onset of allergic airway inflammation. On day 11, airway responsiveness to methacholine was measured; inflammatory cells and cytokines were quantified in the airways; and the numbers and/or viability of T cells, B cells and dendritic cells were assessed in the lungs and draining lymph nodes. Results AAL-R decreased airway hyperresponsiveness induced by D. pteronyssinus by nearly 70%. This was associated with a strong reduction of IL-5 and IL-13 levels in the airways and with a decreased eosinophilic response. Notably, the lung CD4+ T cells were almost entirely eliminated by AAL-R, which concurred with enhanced apoptosis/necrosis in that cell population. This inhibition occurred in the absence of dendritic cell number modulation in draining lymph nodes. On the other hand, the pre-phosphorylated form AFD-R, which preferentially acts on cell-surface sphingosine-1-phosphate receptors, was relatively impotent at enhancing cell death, which led to a less efficient control of T cell and eosinophil responses in the lungs. Conclusion Airway delivery of the non-phosphorylated sphingosine analog, but not its pre-phosphorylated counterpart, is highly efficient at controlling the local T cell response after the onset of allergic airway inflammation. The mechanism appears to involve local induction of lymphocyte apoptosis/necrosis, while mildly affecting dendritic cell and T cell accumulation in draining lymph nodes.
Collapse
Affiliation(s)
- David Gendron
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada.
| | - Anne-Marie Lemay
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada.
| | - Claudine Tremblay
- Laboratoires Charles River, Services Précliniques, Montréal, Canada.
| | - Laetitia Ja Lai
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada.
| | - Anick Langlois
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada.
| | - Émilie Bernatchez
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada.
| | - Nicolas Flamand
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada. .,Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada.
| | - Marie-Renée Blanchet
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada. .,Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada.
| | - Anthony S Don
- Prince of Wales Clinical School, Faculty of Medicine, University of New South Wales, Sydney, 2052, NSW, Australia.
| | - Ynuk Bossé
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada. .,Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada.
| | - Élyse Bissonnette
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada. .,Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada.
| | - David Marsolais
- Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec (CRIUCPQ), Québec, QC, Canada. .,Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
26
|
Porterfield WB, Prescher JA. Tools for visualizing cell–cell ‘interactomes’. Curr Opin Chem Biol 2015; 24:121-30. [DOI: 10.1016/j.cbpa.2014.11.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2014] [Accepted: 11/06/2014] [Indexed: 11/28/2022]
|
27
|
Hickman HD. Imaging CD8 + T cells during diverse viral infections. INTRAVITAL 2015; 4:e1055425. [PMID: 28243513 PMCID: PMC5226004 DOI: 10.1080/21659087.2015.1055425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Accepted: 05/20/2015] [Indexed: 12/12/2022]
Abstract
CD8+ T cells play a critical role in host defense against pathogens and tumors. Much of our current knowledge of the activation and subsequent effector activities of CD8+ T cells has been gained using ex vivo approaches examining the T cell population en masse for surface phenotype, activation status and the production of effector molecules. Thus, the precise behaviors and diversity of individual CD8+ T cells responding to virus infection in vivo have not been extensively explored, leaving many unanswered questions relevant to the rational design of antiviral vaccines and therapeutics. Recently, intravital multiphoton microscopy (MPM) has been used to image CD8+ T cell priming after infection with disparate viral pathogens ranging from small RNA viruses encoding few proteins to DNA viruses producing hundreds of viral proteins (many immunomodulatory). After priming, effector CD8+ T cells have been visualized in virus-infected tissue, both during primary infection and after transitioning to tissue resident memory cells (TRM). Here, I highlight recent advances in our understanding of antiviral CD8+ T cell responses revealed through intravital MPM.
Collapse
Affiliation(s)
- Heather D Hickman
- Laboratory of Viral Diseases; National Institute of Allergy and Infectious Diseases; National Institutes of Health; Bethesda, MD USA
| |
Collapse
|
28
|
Multiscale Modeling of the Early CD8 T-Cell Immune Response in Lymph Nodes: An Integrative Study. COMPUTATION 2014. [DOI: 10.3390/computation2040159] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
29
|
Two-photon imaging of remyelination of spinal cord axons by engrafted neural precursor cells in a viral model of multiple sclerosis. Proc Natl Acad Sci U S A 2014; 111:E2349-55. [PMID: 24843159 DOI: 10.1073/pnas.1406658111] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neural precursor cells (NPCs) offer a promising approach for treating demyelinating diseases. However, the cellular dynamics that underlie transplanted NPC-mediated remyelination have not been described. Using two-photon imaging of a newly developed ventral spinal cord preparation and a viral model of demyelination, we describe the motility and intercellular interactions of transplanted mouse NPCs expressing green fluorescent protein (GFP) with damaged axons expressing yellow fluorescent protein (YFP). Our findings reveal focal axonal degeneration that occurs in the ventral side of the spinal cord within 1 wk following intracranial instillation with the neurotropic JHM strain of mouse hepatitis virus (JHMV). Axonal damage precedes extensive demyelination and is characterized by swelling along the length of the axon, loss of YFP signal, and transected appearance. NPCs engrafted into spinal cords of JHMV-infected mice exhibited diminished migration velocities and increased proliferation compared with transplanted cells in noninfected mice. NPCs preferentially accumulated within areas of axonal damage, initiated direct contact with axons, and subsequently expressed the myelin proteolipid protein gene, initiating remyelination. These findings indicate that NPCs transplanted into an inflammatory demyelinating microenvironment participate directly in therapeutic outcome through the wrapping of myelin around damaged neurons.
Collapse
|
30
|
Baaten BJG, Cooper AM, Swain SL, Bradley LM. Location, location, location: the impact of migratory heterogeneity on T cell function. Front Immunol 2013; 4:311. [PMID: 24115949 PMCID: PMC3792444 DOI: 10.3389/fimmu.2013.00311] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 09/16/2013] [Indexed: 01/13/2023] Open
Abstract
T cell migration is crucial for an effective adaptive immune response to invading pathogens. Naive and memory T cells encounter pathogen antigens, become activated, and differentiate into effector cells in secondary lymphoid tissues, and then migrate to the site(s) of infection where they exert effector activities that control and eliminate pathogens. To achieve activation, efficient effector function, and good memory formation, T cells must traffic between lymphoid and non-lymphoid tissues within the body. This complex process is facilitated by chemokine receptors, selectins, CD44, and integrins that mediate the interactions of T cells with the environment. The expression patterns of these migration receptors (MR) dictate the tissues into which the effector T cells migrate and enable them to occupy specific niches within the tissue. While MR have been considered primarily to facilitate cell movement, we highlight how the heterogeneity of signaling through these receptors influences the function and fate of T cells in situ. We explore what drives MR expression heterogeneity, how this affects migration, and how this impacts T cell effector function and memory formation.
Collapse
Affiliation(s)
- Bas J G Baaten
- Infectious and Inflammatory Diseases Center, Sanford-Burnham Medical Research Institute , La Jolla, CA , USA
| | | | | | | |
Collapse
|