1
|
Niazy AA, Lambarte RNA, Sumague TS, Vigilla MGB, Bin Shwish NM, Kamalan R, Daeab EK, Aljehani NM. FTY720 Reduces the Biomass of Biofilms in Pseudomonas aeruginosa in a Dose-Dependent Manner. Antibiotics (Basel) 2024; 13:621. [PMID: 39061303 PMCID: PMC11273553 DOI: 10.3390/antibiotics13070621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Pseudomonas aeruginosa, a nosocomial pathogen, has strong biofilm capabilities, representing the main source of infection in the human body. Repurposing existing drugs has been explored as an alternative strategy to combat emerging antibiotic-resistant pathogens. Fingolimod hydrochloride (FTY720), an immunomodulatory drug for multiple sclerosis, has shown promising antimicrobial effects against some ESKAPE pathogens. Therefore, the effects of FTY720 on the biofilm capabilities of Pseudomonas aeruginosa were investigated in this study. It was determined that FTY720 inhibited the growth of P. aeruginosa PAO1 at 100 µM. The significant reduction in PAO1 cell viability was observed to be dose-dependent. Additional cytotoxicity analysis on human cell lines showed that FTY720 significantly reduced viabilities at sub-inhibitory concentrations of 25-50 µM. Microtiter assays and confocal analysis confirmed reductions in biofilm mass and thickness and the cell survivability ratio in the presence of FTY720. Similarly, virulence production and biofilm-related gene expression (rhlA, rhlB, pilA, pilI, fliC, fliD and algR) were determined. The results demonstrate that pigment production was affected and quantitative real-time PCR analysis showed a variable degree of reduced gene expression in response to FTY720 at 12.5-50 µM. These findings suggest that FTY720 could be repurposed as an alternative antibiofilm agent against Pseudomonas aeruginosa.
Collapse
Affiliation(s)
- Abdurahman A. Niazy
- Department of Oral Medicine and Diagnostic Sciences, College of Dentistry, King Saud University, Riyadh 11545, Saudi Arabia
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Rhodanne Nicole A. Lambarte
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Terrence S. Sumague
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Mary Grace B. Vigilla
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Najla M. Bin Shwish
- Molecular and Cell Biology Laboratory, Prince Naif bin AbdulAziz Health Research Center, College of Dentistry, King Saud University Medical City, King Saud University, Riyadh 11545, Saudi Arabia; (R.N.A.L.); (T.S.S.); (N.M.B.S.)
| | - Ranan Kamalan
- Research Center, College of Dentistry, King Saud University, Riyadh 11451, Saudi Arabia
| | - Eid Khulaif Daeab
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| | - Nami M. Aljehani
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, King Saud University, Riyadh 11433, Saudi Arabia
| |
Collapse
|
2
|
Rezaie J, Chodari L, Mohammadpour-Asl S, Jafari A, Niknam Z. Cell-mediated barriers in cancer immunosurveillance. Life Sci 2024; 342:122528. [PMID: 38408406 DOI: 10.1016/j.lfs.2024.122528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 02/07/2024] [Accepted: 02/21/2024] [Indexed: 02/28/2024]
Abstract
The immune cells within the tumor microenvironment (TME) exert multifaceted functions ranging from tumor-antagonizing or tumor-promoting activities. During the initial phases of tumor development, the tumor-antagonizing immune cells in the TME combat cancer cells in an immune surveillance process. However, with time, cancer cells can evade detection and impede the immune cells' effectiveness through diverse mechanisms, such as decreasing immunogenic antigen presentation on their surfaces and/or secreting anti-immune factors that cause tolerance in TME. Moreover, some immune cells cause immunosuppressive situations and inhibit antitumoral immune responses. Physical and cellular-mediated barriers in the TME, such as cancer-associated fibroblasts, tumor endothelium, the altered lipid composition of tumor cells, and exosomes secreted from cancer cells, also mediate immunosuppression and prevent extravasation of immune cells. Due to successful clinical outcomes of cancer treatment strategies the potential barriers must be identified and addressed. We need to figure out how to optimize cancer immunotherapy strategies, and how to combine therapeutic approaches for maximum clinical benefit. This review provides a detailed overview of various cells and molecules in the TME, their association with escaping from immune surveillance, therapeutic targets, and future perspectives for improving cancer immunotherapy.
Collapse
Affiliation(s)
- Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Leila Chodari
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran; Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Shadi Mohammadpour-Asl
- Department of Physiology, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran; Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Abbas Jafari
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Zahra Niknam
- Neurophysiology Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
3
|
Ino Y, Maruyama M, Shimizu M, Morita R, Sakamoto A, Suzuki H, Sakai A. TSLP in DRG neurons causes the development of neuropathic pain through T cells. J Neuroinflammation 2023; 20:200. [PMID: 37660072 PMCID: PMC10474733 DOI: 10.1186/s12974-023-02882-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/28/2023] [Indexed: 09/04/2023] Open
Abstract
BACKGROUND Peripheral nerve injury to dorsal root ganglion (DRG) neurons develops intractable neuropathic pain via induction of neuroinflammation. However, neuropathic pain is rare in the early life of rodents. Here, we aimed to identify a novel therapeutic target for neuropathic pain in adults by comprehensively analyzing the difference of gene expression changes between infant and adult rats after nerve injury. METHODS A neuropathic pain model was produced in neonatal and young adult rats by spared nerve injury. Nerve injury-induced gene expression changes in the dorsal root ganglion (DRG) were examined using RNA sequencing. Thymic stromal lymphopoietin (TSLP) and its siRNA were intrathecally injected. T cells were examined using immunofluorescence and were reduced by systemic administration of FTY720. RESULTS Differences in changes in the transcriptome in injured DRG between infant and adult rats were most associated with immunological functions. Notably, TSLP was markedly upregulated in DRG neurons in adult rats, but not in infant rats. TSLP caused mechanical allodynia in adult rats, whereas TSLP knockdown suppressed the development of neuropathic pain. TSLP promoted the infiltration of T cells into the injured DRG and organized the expressions of multiple factors that regulate T cells. Accordingly, TSLP caused mechanical allodynia through T cells in the DRG. CONCLUSION This study demonstrated that TSLP is causally involved in the development of neuropathic pain through T cell recruitment.
Collapse
Affiliation(s)
- Yuka Ino
- Department of Anesthesiology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
- Department of Pharmacology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
| | - Motoyo Maruyama
- Department of Pharmacology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
- Division of Laboratory Animal Science, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
| | - Masumi Shimizu
- Department of Microbiology and Immunology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
| | - Rimpei Morita
- Department of Microbiology and Immunology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
| | - Atsuhiro Sakamoto
- Department of Anesthesiology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
| | - Hidenori Suzuki
- Department of Pharmacology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
| | - Atsushi Sakai
- Department of Pharmacology, Nippon Medical School, 1-1-5 Sendagi, Bunkyo-ku, Tokyo, 113-8602 Japan
| |
Collapse
|
4
|
Abbaszadeh S, Nosrati-Siahmazgi V, Musaie K, Rezaei S, Qahremani M, Xiao B, Santos HA, Shahbazi MA. Emerging strategies to bypass transplant rejection via biomaterial-assisted immunoengineering: Insights from islets and beyond. Adv Drug Deliv Rev 2023; 200:115050. [PMID: 37549847 DOI: 10.1016/j.addr.2023.115050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/14/2023] [Accepted: 08/04/2023] [Indexed: 08/09/2023]
Abstract
Novel transplantation techniques are currently under development to preserve the function of impaired tissues or organs. While current technologies can enhance the survival of recipients, they have remained elusive to date due to graft rejection by undesired in vivo immune responses despite systemic prescription of immunosuppressants. The need for life-long immunomodulation and serious adverse effects of current medicines, the development of novel biomaterial-based immunoengineering strategies has attracted much attention lately. Immunomodulatory 3D platforms can alter immune responses locally and/or prevent transplant rejection through the protection of the graft from the attack of immune system. These new approaches aim to overcome the complexity of the long-term administration of systemic immunosuppressants, including the risks of infection, cancer incidence, and systemic toxicity. In addition, they can decrease the effective dose of the delivered drugs via direct delivery at the transplantation site. In this review, we comprehensively address the immune rejection mechanisms, followed by recent developments in biomaterial-based immunoengineering strategies to prolong transplant survival. We also compare the efficacy and safety of these new platforms with conventional agents. Finally, challenges and barriers for the clinical translation of the biomaterial-based immunoengineering transplants and prospects are discussed.
Collapse
Affiliation(s)
- Samin Abbaszadeh
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Vahideh Nosrati-Siahmazgi
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Kiyan Musaie
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands
| | - Saman Rezaei
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Mostafa Qahremani
- Department of Pharmaceutical Biomaterials, School of Pharmacy, Zanjan University of Medical Science, 45139-56184 Zanjan, Iran
| | - Bo Xiao
- State Key Laboratory of Silkworm Genome Biology, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715 China.
| | - Hélder A Santos
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands; Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, 00014 Helsinki, Finland; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| | - Mohammad-Ali Shahbazi
- Department of Biomedical Engineering, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands; W.J. Kolff Institute for Biomedical Engineering and Materials Science, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, the Netherlands.
| |
Collapse
|
5
|
Zhang Y. From gene identifications to therapeutic targets for asthma: Focus on great potentials of TSLP, ORMDL3, and GSDMB. CHINESE MEDICAL JOURNAL PULMONARY AND CRITICAL CARE MEDICINE 2023; 1:139-147. [PMID: 39171126 PMCID: PMC11332877 DOI: 10.1016/j.pccm.2023.08.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Indexed: 08/23/2024]
Abstract
Asthma is a chronic respiratory disease, and clinically, asthma exacerbations remain difficult to treat. The disease is caused by combinations of and interactions between genetic and environmental factors. Genomic and genetic approaches identified many novel genes to treat asthma and brought new insights into the disease. The products of the genes have functional roles in regulating physiological or pathophysiological processes in airway structural cells and immune system cells. Genetic factors also interact with environmental factors such as air pollutants, and bacterial and viral infections to trigger the disease. Thymic stromal lymphopoietin (TSLP), orosomucoid-like 3 (ORMDL3), and gasdermin B (GSDMB) are three genes identified by genetic studies to have a great potential as therapeutic targets of asthma. TSLP is an important driver of type 2 inflammation. ORMDL3 mediates cell stress, sphingolipid synthesis, and viral and bacterial infections. GSDMB regulates cell pyroptosis through its N and C terminals and can bind sulfatides to influence inflammatory response. Investigating inhibitors or modulators for these pathways would bring a new landscape for therapeutics of asthma in future.
Collapse
Affiliation(s)
- Youming Zhang
- National Heart and Lung Institute, Imperial College London, Dovehouse Street, London SW3 6LY, UK
| |
Collapse
|
6
|
Behara M, Goudy S. FTY720 in immuno-regenerative and wound healing technologies for muscle, epithelial and bone regeneration. Front Physiol 2023; 14:1148932. [PMID: 37250137 PMCID: PMC10213316 DOI: 10.3389/fphys.2023.1148932] [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: 01/20/2023] [Accepted: 05/02/2023] [Indexed: 05/31/2023] Open
Abstract
In 2010, the FDA approved the administration of FTY720, S1P lipid mediator, as a therapy to treat relapsing forms of multiple sclerosis. FTY720 was found to sequester pro-inflammatory lymphocytes within the lymph node, preventing them from causing injury to the central nervous system due to inflammation. Studies harnessing the anti-inflammatory properties of FTY720 as a pro-regenerative strategy in wound healing of muscle, bone and mucosal injuries are currently being performed. This in-depth review discusses the current regenerative impact of FTY720 due to its anti-inflammatory effect stratified into an assessment of wound regeneration in the muscular, skeletal, and epithelial systems. The regenerative effect of FTY720 in vivo was characterized in three animal models, with differing delivery mechanisms emerging in the last 20 years. In these studies, local delivery of FTY720 was found to increase pro-regenerative immune cell phenotypes (neutrophils, macrophages, monocytes), vascularization, cell proliferation and collagen deposition. Delivery of FTY720 to a localized wound environment demonstrated increased bone, muscle, and mucosal regeneration through changes in gene and cytokine production primarily by controlling the local immune cell phenotypes. These changes in gene and cytokine production reduced the inflammatory component of wound healing and increased the migration of pro-regenerative cells (neutrophils and macrophages) to the wound site. The application of FTY720 delivery using a biomaterial has demonstrated the ability of local delivery of FTY720 to promote local wound healing leveraging an immunomodulatory mechanism.
Collapse
Affiliation(s)
- Monica Behara
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| | - Steven Goudy
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, United States
- Department of Otolaryngology, Emory University, Atlanta, GA, United States
| |
Collapse
|
7
|
Wieczorek I, Strosznajder RP. Recent Insight into the Role of Sphingosine-1-Phosphate Lyase in Neurodegeneration. Int J Mol Sci 2023; 24:ijms24076180. [PMID: 37047151 PMCID: PMC10093903 DOI: 10.3390/ijms24076180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 04/14/2023] Open
Abstract
Sphingosine-1-phosphate lyase (SPL) is a pyridoxal 5'-phosphate-dependent enzyme involved in the irreversible degradation of sphingosine-1-phosphate (S1P)-a bioactive sphingolipid that modulates a broad range of biological processes (cell proliferation, migration, differentiation and survival; mitochondrial functioning; and gene expression). Although SPL activity leads to a decrease in the available pool of S1P in the cell, at the same time, hexadecenal and phosphoethanolamine, compounds with potential biological activity, are generated. The increased expression and/or activity of SPL, and hence the imbalance between S1P and the end products of its cleavage, were demonstrated in several pathological states. On the other hand, loss-of-function mutations in the SPL encoding gene are a cause of severe developmental impairments. Recently, special attention has been paid to neurodegenerative diseases as the most common pathologies of the nervous system. This review summarizes the current findings concerning the role of SPL in the nervous system with an emphasis on neurodegeneration. Moreover, it briefly discusses pharmacological compounds directed to inhibit its activity.
Collapse
Affiliation(s)
- Iga Wieczorek
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| | - Robert Piotr Strosznajder
- Laboratory of Preclinical Research and Environmental Agents, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 St., 02-106 Warsaw, Poland
| |
Collapse
|
8
|
Ung J, Tan SF, Fox TE, Shaw JJP, Vass LR, Costa-Pinheiro P, Garrett-Bakelman FE, Keng MK, Sharma A, Claxton DF, Levine RL, Tallman MS, Cabot MC, Kester M, Feith DJ, Loughran TP. Harnessing the power of sphingolipids: Prospects for acute myeloid leukemia. Blood Rev 2022; 55:100950. [PMID: 35487785 PMCID: PMC9475810 DOI: 10.1016/j.blre.2022.100950] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/31/2022] [Accepted: 04/04/2022] [Indexed: 11/02/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive, heterogenous malignancy characterized by clonal expansion of bone marrow-derived myeloid progenitor cells. While our current understanding of the molecular and genomic landscape of AML has evolved dramatically and opened avenues for molecularly targeted therapeutics to improve upon standard intensive induction chemotherapy, curative treatments are elusive, particularly in older patients. Responses to current AML treatments are transient and incomplete, necessitating the development of novel treatment strategies to improve outcomes. To this end, harnessing the power of bioactive sphingolipids to treat cancer shows great promise. Sphingolipids are involved in many hallmarks of cancer of paramount importance in AML. Leukemic blast survival is influenced by cellular levels of ceramide, a bona fide pro-death molecule, and its conversion to signaling molecules such as sphingosine-1-phosphate and glycosphingolipids. Preclinical studies demonstrate the efficacy of therapeutics that target dysregulated sphingolipid metabolism as well as their combinatorial synergy with clinically-relevant therapeutics. Thus, increased understanding of sphingolipid dysregulation may be exploited to improve AML patient care and outcomes. This review summarizes the current knowledge of dysregulated sphingolipid metabolism in AML, evaluates how pro-survival sphingolipids promote AML pathogenesis, and discusses the therapeutic potential of targeting these dysregulated sphingolipid pathways.
Collapse
Affiliation(s)
- Johnson Ung
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Su-Fern Tan
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Todd E Fox
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Jeremy J P Shaw
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Experimental Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Luke R Vass
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Experimental Pathology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Pedro Costa-Pinheiro
- Cancer Biology, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Francine E Garrett-Bakelman
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - Michael K Keng
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Arati Sharma
- Penn State Cancer Institute, Hershey, PA, United States of America
| | - David F Claxton
- Penn State Cancer Institute, Hershey, PA, United States of America
| | - Ross L Levine
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America; Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Martin S Tallman
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States of America
| | - Myles C Cabot
- Department of Biochemistry and Molecular Biology, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America; East Carolina Diabetes and Obesity Institute, East Carolina University, Brody School of Medicine, Greenville, NC, United States of America
| | - Mark Kester
- University of Virginia Cancer Center, Charlottesville, VA, United States of America; Department of Pharmacology, University of Virginia School of Medicine, Charlottesville, VA, United States of America
| | - David J Feith
- Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America
| | - Thomas P Loughran
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; Division of Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA, United States of America; University of Virginia Cancer Center, Charlottesville, VA, United States of America.
| |
Collapse
|
9
|
Etemadifar M, Nouri H, Pitzalis M, Idda ML, Salari M, Baratian M, Mahdavi S, Abhari AP, Sedaghat N. Multiple sclerosis disease-modifying therapies and COVID-19 vaccines: a practical review and meta-analysis. J Neurol Neurosurg Psychiatry 2022; 93:986-994. [PMID: 35688629 DOI: 10.1136/jnnp-2022-329123] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 05/10/2022] [Indexed: 12/19/2022]
Abstract
Studies among people with multiple sclerosis (pwMS) receiving disease-modifying therapies (DMTs) have provided adequate evidence for an appraisal of COVID-19 vaccination policies among them. To synthesise the available evidence addressing the effect of MS DMTs on COVID-19 vaccines' immunogenicity and effectiveness, following the Cochrane guidelines, we systematically reviewed all observational studies available in MEDLINE, Scopus, Web of Science, MedRxiv and Google Scholar from January 2021 to January 2022 and extracted their relevant data. Immunogenicity data were then synthesised in a quantitative, and other data in a qualitative manner. Evidence from 28 studies suggests extensively lower B-cell responses in sphingosine-1-phosphate receptor modulator (S1PRM) treated and anti-CD20 (aCD20) treated, and lower T-cell responses in interferon-treated, S1PRM-treated and cladribine-treated pwMS-although most T cell evidence currently comprises of low or very low certainty. With every 10-week increase in aCD20-to-vaccine period, a 1.94-fold (95% CI 1.57 to 2.41, p<0.00001) increase in the odds of seroconversion was observed. Furthermore, the evidence points out that B-cell-depleting therapies may accelerate postvaccination humoral waning, and boosters' immunogenicity is predictable with the same factors affecting the initial vaccination cycle. Four real-world studies further indicate that the comparative incidence/severity of breakthrough COVID-19 has been higher among the pwMS treated with S1PRM and aCD20-unlike the ones treated with other DMTs. S1PRM and aCD20 therapies were the only DMTs reducing the real-world effectiveness of COVID-19 vaccination among pwMS. Hence, it could be concluded that optimisation of humoral immunogenicity and ensuring its durability are the necessities of an effective COVID-19 vaccination policy among pwMS who receive DMTs.
Collapse
Affiliation(s)
- Masoud Etemadifar
- Neurosurgery Research Department, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hosein Nouri
- Neurosurgery Research Department, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran.,Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| | - Maristella Pitzalis
- Institute of Genetic and Biomedical Research (IRGB) of the National Research Council (CNR), Cagliari, Italy
| | - Maria Laura Idda
- Institute of Genetic and Biomedical Research (IRGB) of the National Research Council (CNR), Cagliari, Italy
| | - Mehri Salari
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahshid Baratian
- Clinical Research Developement Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Sepide Mahdavi
- Clinical Research Developement Center, Najafabad Branch, Islamic Azad University, Najafabad, Iran
| | - Amir Parsa Abhari
- Neurosurgery Research Department, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran.,Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| | - Nahad Sedaghat
- Neurosurgery Research Department, Alzahra University Hospital, Isfahan University of Medical Sciences, Isfahan, Iran .,Network of Immunity in Infection, Malignancy, and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Isfahan, Iran
| |
Collapse
|
10
|
Testa S, Haabeth OAW, Blake TR, Del Castillo TJ, Czerwinski DK, Rajapaksa R, Wender PA, Waymouth RM, Levy R. Fingolimod-Conjugated Charge-Altering Releasable Transporters Efficiently and Specifically Deliver mRNA to Lymphocytes In Vivo and In Vitro. Biomacromolecules 2022; 23:2976-2988. [PMID: 35748182 PMCID: PMC10199726 DOI: 10.1021/acs.biomac.2c00469] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Charge-altering releasable transporters (CARTs) are a class of oligonucleotide delivery vehicles shown to be effective for delivery of messenger RNA (mRNA) both in vitro and in vivo. Here, we exploited the chemical versatility of the CART synthesis to generate CARTs containing the small-molecule drug fingolimod (FTY720) as a strategy to increase mRNA delivery and expression in lymphocytes through a specific ligand-receptor interaction. Fingolimod is an FDA-approved small-molecule drug that, upon in vivo phosphorylation, binds to the sphingosine-1-phosphate receptor 1 (S1P1), which is highly expressed on lymphocytes. Compared to its non-fingolimod-conjugated analogue, the fingolimod-conjugated CART achieved superior transfection of activated human and murine T and B lymphocytes in vitro. The higher transfection of the fingolimod-conjugated CARTs was lost when cells were exposed to a free fingolimod before transfection. In vivo, the fingolimod-conjugated CART showed increased mRNA delivery to marginal zone B cells and NK cells in the spleen, relative to CARTs lacking fingolimod. Moreover, fingolimod-CART-mediated mRNA delivery induces peripheral blood T-cell depletion similar to free fingolimod. Thus, we show that functionalization of CARTs with a pharmacologically validated small molecule can increase transfection of a cellular population of interest while conferring some of the targeting properties of the conjugated small molecule to the CARTs.
Collapse
Affiliation(s)
- Stefano Testa
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, United States
| | - Ole A W Haabeth
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, United States
| | - Timothy R Blake
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, United States
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Trevor J Del Castillo
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Debra K Czerwinski
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, United States
| | - Ranjani Rajapaksa
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, United States
| | - Paul A Wender
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
- Department of Chemical and Systems Biology, Stanford University, Stanford, California 94305, United States
| | - Robert M Waymouth
- Department of Chemistry, Stanford University, Stanford, California 94305, United States
| | - Ronald Levy
- Stanford Cancer Institute, Division of Oncology, Department of Medicine, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
11
|
Pournajaf S, Dargahi L, Javan M, Pourgholami MH. Molecular Pharmacology and Novel Potential Therapeutic Applications of Fingolimod. Front Pharmacol 2022; 13:807639. [PMID: 35250559 PMCID: PMC8889014 DOI: 10.3389/fphar.2022.807639] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Fingolimod is a well-tolerated, highly effective disease-modifying therapy successfully utilized in the management of multiple sclerosis. The active metabolite, fingolimod-phosphate, acts on sphingosine-1-phosphate receptors (S1PRs) to bring about an array of pharmacological effects. While being initially recognized as a novel agent that can profoundly reduce T-cell numbers in circulation and the CNS, thereby suppressing inflammation and MS, there is now rapidly increasing knowledge on its previously unrecognized molecular and potential therapeutic effects in diverse pathological conditions. In addition to exerting inhibitory effects on sphingolipid pathway enzymes, fingolimod also inhibits histone deacetylases, transient receptor potential cation channel subfamily M member 7 (TRMP7), cytosolic phospholipase A2α (cPLA2α), reduces lysophosphatidic acid (LPA) plasma levels, and activates protein phosphatase 2A (PP2A). Furthermore, fingolimod induces apoptosis, autophagy, cell cycle arrest, epigenetic regulations, macrophages M1/M2 shift and enhances BDNF expression. According to recent evidence, fingolimod modulates a range of other molecular pathways deeply rooted in disease initiation or progression. Experimental reports have firmly associated the drug with potentially beneficial therapeutic effects in immunomodulatory diseases, CNS injuries, and diseases including Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and even cancer. Attractive pharmacological effects, relative safety, favorable pharmacokinetics, and positive experimental data have collectively led to its testing in clinical trials. Based on the recent reports, fingolimod may soon find its way as an adjunct therapy in various disparate pathological conditions. This review summarizes the up-to-date knowledge about molecular pharmacology and potential therapeutic uses of fingolimod.
Collapse
Affiliation(s)
- Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
12
|
Sphingosine-1-Phosphate Levels Are Higher in Male Patients with Non-Classic Fabry Disease. J Clin Med 2022; 11:jcm11051233. [PMID: 35268324 PMCID: PMC8911241 DOI: 10.3390/jcm11051233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 11/17/2022] Open
Abstract
Fabry disease is an X-linked lysosomal disease in which defects in the alpha-galactosidase A enzyme activity lead to the ubiquitous accumulation of glycosphingolipids. Whereas the classic disease is characterized by neuropathic pain, progressive renal failure, white matter lesions, cerebral stroke, and hypertrophic cardiomyopathy (HCM), the non-classic phenotype, also known as cardiac variant, is almost exclusively characterized by HCM. Circulating sphingosine-1-phosphate (S1P) has controversially been associated with the Fabry cardiomyopathy. We measured serum S1P levels in 41 patients of the FFABRY cohort. S1P levels were higher in patients with a non-classic phenotype compared to those with a classic phenotype (200.3 [189.6−227.9] vs. 169.4 ng/mL [121.1−203.3], p = 0.02). In a multivariate logistic regression model, elevated S1P concentration remained statistically associated with the non-classic phenotype (OR = 1.03; p < 0.02), and elevated lysoGb3 concentration with the classic phenotype (OR = 0.95; p < 0.03). S1P levels were correlated with interventricular septum thickness (r = 0.46; p = 0.02). In a logistic regression model including S1P serum levels, phenotype, and age, age remained the only variable significantly associated with the risk of HCM (OR = 1.25; p = 0.001). S1P alone was not associated with cardiac hypertrophy but with the cardiac variant. The significantly higher S1P levels in patients with the cardiac variant compared to those with classic Fabry suggest the involvement of distinct pathophysiological pathways in the two phenotypes. S1P dosage could allow the personalization of patient management.
Collapse
|
13
|
Ji D, Fleig A, Horgen FD, Feng ZP, Sun HS. Modulators of TRPM7 and its potential as a drug target for brain tumours. Cell Calcium 2021; 101:102521. [PMID: 34953296 DOI: 10.1016/j.ceca.2021.102521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 12/14/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022]
Abstract
TRPM7 is a non-selective divalent cation channel with an alpha-kinase domain. Corresponding with its broad expression, TRPM7 has a role in a wide range of cell functions, including proliferation, migration, and survival. Growing evidence shows that TRPM7 is also aberrantly expressed in various cancers, including brain cancers. Because ion channels have widespread tissue distribution and result in extensive physiological consequences when dysfunctional, these proteins can be compelling drug targets. In fact, ion channels comprise the third-largest drug target type, following enzymes and receptors. Literature has shown that suppression of TRPM7 results in inhibition of migration, invasion, and proliferation in several human brain tumours. Therefore, TRPM7 presents a potential target for therapeutic brain tumour interventions. This article reviews current literature on TRPM7 as a potential drug target in the context of brain tumours and provides an overview of various selective and non-selective modulators of the channel relevant to pharmacology, oncology, and ion channel function.
Collapse
Affiliation(s)
- Delphine Ji
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8
| | - Andrea Fleig
- Center for Biomedical Research at The Queen's Medical Center and John A. Burns School of Medicine and Cancer Center at the University of Hawaii, Honolulu, Hawaii 96813, USA
| | - F David Horgen
- Department of Natural Sciences, Hawaii Pacific University, Kaneohe, Hawaii 96744, USA
| | - Zhong-Ping Feng
- Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8.
| | - Hong-Shuo Sun
- Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8; Department of Physiology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8; Department of Pharmacology, Temerty Faculty of Medicine, University of Toronto, 1 King's College Circle, Toronto, Ontario, Canada M5S 1A8; Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, Ontario, Canada M5S 3M2.
| |
Collapse
|
14
|
Nabi R, Lewin AC, Collantes TM, Chouljenko VN, Kousoulas KG. Intramuscular Vaccination With the HSV-1(VC2) Live-Attenuated Vaccine Strain Confers Protection Against Viral Ocular Immunopathogenesis Associated With γδT Cell Intracorneal Infiltration. Front Immunol 2021; 12:789454. [PMID: 34868077 PMCID: PMC8634438 DOI: 10.3389/fimmu.2021.789454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/25/2021] [Indexed: 11/18/2022] Open
Abstract
Herpes simplex virus type-1 (HSV-1) ocular infection is one of the leading causes of infectious blindness in developed countries. The resultant herpetic keratitis (HK) is caused by an exacerbated reaction of the adaptive immune response that persists beyond virus clearance causing substantial damage to the cornea. Intramuscular immunization of mice with the HSV-1(VC2) live-attenuated vaccine strain has been shown to protect mice against lethal ocular challenge. Herein, we show that following ocular challenge, VC2 vaccinated animals control ocular immunopathogenesis in the absence of neutralizing antibodies on ocular surfaces. Ocular protection is associated with enhanced intracorneal infiltration of γδ T cells compared to mock-vaccinated animals. The observed γδ T cellular infiltration was inversely proportional to the infiltration of neutrophils, the latter associated with exacerbated tissue damage. Inhibition of T cell migration into ocular tissues by the S1P receptors agonist FTY720 produced significant ocular disease in vaccinated mice and marked increase in neutrophil infiltration. These results indicate that ocular challenge of mice immunized with the VC2 vaccine induce a unique ocular mucosal response that leads into the infiltration of γδ T cells resulting in the amelioration of infection-associated immunopathogenesis.
Collapse
MESH Headings
- Animals
- Chemotaxis, Leukocyte
- Cornea/immunology
- Cornea/pathology
- Cornea/virology
- Cytokines/metabolism
- Disease Models, Animal
- Female
- Herpes Simplex Virus Vaccines/administration & dosage
- Herpes Simplex Virus Vaccines/immunology
- Herpesvirus 1, Human/immunology
- Herpesvirus 1, Human/pathogenicity
- Host-Pathogen Interactions
- Injections, Intramuscular
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/virology
- Keratitis, Herpetic/immunology
- Keratitis, Herpetic/pathology
- Keratitis, Herpetic/prevention & control
- Keratitis, Herpetic/virology
- Lymphangiogenesis
- Mice, Inbred BALB C
- Neovascularization, Pathologic
- Neutrophil Infiltration
- Vaccination
- Vaccines, Attenuated/administration & dosage
- Vaccines, Attenuated/immunology
- Mice
Collapse
Affiliation(s)
- Rafiq Nabi
- Department of Pathobiological Science, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Andrew C. Lewin
- Department of Veterinary Clinical Sciences, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Therese M. Collantes
- Department of Pathobiological Science, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Vladimir N. Chouljenko
- Department of Pathobiological Science, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| | - Konstantin G. Kousoulas
- Department of Pathobiological Science, Louisiana State University School of Veterinary Medicine, Baton Rouge, LA, United States
| |
Collapse
|
15
|
Zhang YM. Orosomucoid-like protein 3, rhinovirus and asthma. World J Crit Care Med 2021; 10:170-182. [PMID: 34616654 PMCID: PMC8462028 DOI: 10.5492/wjccm.v10.i5.170] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 04/16/2021] [Accepted: 08/23/2021] [Indexed: 02/06/2023] Open
Abstract
The genetic variants of orosomucoid-like protein 3 (ORMDL3) gene are associated with highly significant increases in the number of human rhinovirus (HRV)-induced wheezing episodes in children. Recent investigations have been focused on the mechanisms of ORMDL3 in rhinovirus infection for asthma and asthma exacerbations. ORMDL3 not only regulates major human rhinovirus receptor intercellular adhesion molecule 1 expression, but also plays pivotal roles in viral infection through metabolisms of ceramide and sphingosine-1-phosphate, endoplasmic reticulum (ER) stress, ER-Golgi interface and glycolysis. Research on the roles of ORMDL3 in HRV infection will lead us to identify new biomarkers and novel therapeutic targets in childhood asthma and viral induced asthma exacerbations.
Collapse
Affiliation(s)
- You-Ming Zhang
- Section of Genomic and Environmental Medicine, National Heart and Lung Institute, Molecular Genetics Group, Division of Respiratory Sciences, Imperial College London, London SW3 6LY, United Kingdom
| |
Collapse
|
16
|
Ssu72 phosphatase directly binds to ZAP-70, thereby providing fine-tuning of TCR signaling and preventing spontaneous inflammation. Proc Natl Acad Sci U S A 2021; 118:2102374118. [PMID: 34452999 DOI: 10.1073/pnas.2102374118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
ZAP-70 is required for the initiation of T cell receptor (TCR) signaling, and Ssu72 is a phosphatase that regulates RNA polymerase II activity in the nucleus. However, the mechanism by which ZAP-70 regulates the fine-tuning of TCR signaling remains elusive. Here, we found that Ssu72 contributed to the fine-tuning of TCR signaling by acting as tyrosine phosphatase for ZAP-70. Affinity purification-mass spectrometry and an in vitro assay demonstrated specific interaction between Ssu72 and ZAP-70 in T cells. Upon TCR stimulation, Ssu72-deficient T cells increased the phosphorylation of ZAP-70 and downstream molecules and exhibited hyperresponsiveness, which was restored by reducing ZAP-70 phosphorylation. In vitro assay demonstrated that recombinant Ssu72 reduced tyrosine phosphorylation of ZAP-70 via phosphatase activity. Cd4-CreSsu72 fl/fl mice showed a defect in the thymic development of invariant natural killer T cells and reductions in CD4+ and CD8+ T cell numbers in the periphery but more CD44hiCD62Llo memory T cells and fewer CD44loCD62Lhi naïve T cells, compared with wild-type mice. Furthermore, Cd4-CreSsu72 fl/fl mice developed spontaneous inflammation at 6 mo. In conclusion, Ssu72 phosphatase regulates the fine-tuning of TCR signaling by binding to ZAP-70 and regulating its tyrosine phosphorylation, thereby preventing spontaneous inflammation.
Collapse
|
17
|
Derakhshani A, Asadzadeh Z, Safarpour H, Leone P, Shadbad MA, Heydari A, Baradaran B, Racanelli V. Regulation of CTLA-4 and PD-L1 Expression in Relapsing-Remitting Multiple Sclerosis Patients after Treatment with Fingolimod, IFNβ-1α, Glatiramer Acetate, and Dimethyl Fumarate Drugs. J Pers Med 2021; 11:jpm11080721. [PMID: 34442365 PMCID: PMC8400811 DOI: 10.3390/jpm11080721] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 06/29/2021] [Accepted: 07/20/2021] [Indexed: 02/06/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic demyelinating disease of the central nervous system (CNS) that is characterized by inflammation which typically results in significant impairment in most patients. Immune checkpoints act as co-stimulatory and co-inhibitory molecules and play a fundamental role in keeping the equilibrium of the immune system. Cytotoxic T-lymphocyte antigen-4 (CTLA-4) and Programmed death-ligand 1 (PD-L1), as inhibitory immune checkpoints, participate in terminating the development of numerous autoimmune diseases, including MS. We assessed the CTLA-4 and PD-L1 gene expression in the different cell types of peripheral blood mononuclear cells of MS patients using single-cell RNA-seq data. Additionally, this study outlines how CTLA-4 and PD-L1 expression was altered in the PBMC samples of relapsing-remitting multiple sclerosis (RRMS) patients compared to the healthy group. Finally, it investigates the impact of various MS-related treatments in the CTLA-4 and PD-L1 expression to restrain autoreactive T cells and stop the development of MS autoimmunity.
Collapse
Affiliation(s)
- Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (A.D.); (Z.A.); (M.A.S.)
- IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (A.D.); (Z.A.); (M.A.S.)
| | - Hossein Safarpour
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
| | - Patrizia Leone
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Mahdi Abdoli Shadbad
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (A.D.); (Z.A.); (M.A.S.)
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz 516615731, Iran
| | - Ali Heydari
- Department of Applied Mathematics, University of California, Merced, CA 95343, USA;
- Health Sciences Research Institute, University of California, Merced, CA 95343, USA
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (A.D.); (Z.A.); (M.A.S.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 516615731, Iran
- Correspondence: (B.B.); (V.R.)
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
- Correspondence: (B.B.); (V.R.)
| |
Collapse
|
18
|
Ziemkiewicz N, Hilliard G, Pullen NA, Garg K. The Role of Innate and Adaptive Immune Cells in Skeletal Muscle Regeneration. Int J Mol Sci 2021; 22:3265. [PMID: 33806895 PMCID: PMC8005179 DOI: 10.3390/ijms22063265] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle regeneration is highly dependent on the inflammatory response. A wide variety of innate and adaptive immune cells orchestrate the complex process of muscle repair. This review provides information about the various types of immune cells and biomolecules that have been shown to mediate muscle regeneration following injury and degenerative diseases. Recently developed cell and drug-based immunomodulatory strategies are highlighted. An improved understanding of the immune response to injured and diseased skeletal muscle will be essential for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Natalia Ziemkiewicz
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, 3507 Lindell Blvd, St. Louis, MO 63103, USA;
| | - Genevieve Hilliard
- Department of Biology, Saint Louis University, St. Louis, MO 63103, USA;
| | - Nicholas A. Pullen
- School of Biological Sciences, College of Natural and Health Sciences, University of Northern Colorado, Greeley, Colorado, CO 80639, USA;
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, 3507 Lindell Blvd, St. Louis, MO 63103, USA;
| |
Collapse
|
19
|
Zhao Z, Ma CL, Gu ZC, Dong Y, Lv Y, Zhong MK. Incidence and Risk of Infection Associated With Fingolimod in Patients With Multiple Sclerosis: A Systematic Review and Meta-Analysis of 8,448 Patients From 12 Randomized Controlled Trials. Front Immunol 2021; 12:611711. [PMID: 33763062 PMCID: PMC7982402 DOI: 10.3389/fimmu.2021.611711] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 02/03/2021] [Indexed: 12/01/2022] Open
Abstract
Background and Aims: There is a controversy regarding whether fingolimod is associated with an increased risk of infection in patients with multiple sclerosis (MS). We performed a systematic review and meta-analysis of data from randomized controlled trials (RCTs) to determine the risk of infection in these patients. Methods: We systematically searched PubMed, EMBASE, the Cochrane Library, and clinicaltrials.gov from inception to April 8, 2020, to identify RCTs that reported the occurrence of infection in patients with MS treated with fingolimod. Relative risks (RRs) and 95% confidence intervals (95% CIs) were calculated using the random-effects model. Results: Twelve RCTs including 8,448 patients were eligible. Compared with the control (placebo and other active treatments), fingolimod significantly increased the risk of infection (RR, 1.16; 95% CI, 1.07–1.27; I2, 81%), regardless of whether the infection was a general infection (RR, 1.14; 95% CI, 1.05–1.25; I2, 78%), or a serious infection (RR, 1.49; 95% CI, 1.06–2.10; I2, 0%). Analyses of subgroups found that fingolimod significantly increased the risk of lower respiratory infection (RR, 1.48; 95% CI, 1.19–1.85; I2, 0%) and herpes virus infection (RR, 1.34; 95% CI, 1.01–1.78; I2, 9%). There appears to be no dose-dependent increase in the risk of infection associated with fingolimod (0.5 mg: RR, 1.15; 95% CI, 1.07–1.25; I2, 91%; 1.25 mg: RR, 1.11; 95% CI, 0.97–1.28; I2, 81%; Pinteraction = 0.66). Conclusions: Compared with a placebo and other active treatments, fingolimod was associated with a 16% increase in the risk of infection, especially lower respiratory infection and herpes virus infection. The risk of infection associated with fingolimod might not be dose related.
Collapse
Affiliation(s)
- Zhao Zhao
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Chun-Lai Ma
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Zhi-Chun Gu
- Department of Pharmacy, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Yue Dong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| | - Yang Lv
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China.,Department of Pharmacy, The First Affiliated Hospital, College of Clinical Medicine of Henan University of Science and Technology, Luoyang, China
| | - Ming-Kang Zhong
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Kingstad-Bakke B, Toy R, Lee W, Pradhan P, Vogel G, Marinaik CB, Larsen A, Gates D, Luu T, Pandey B, Kawaoka Y, Roy K, Suresh M. Polymeric Pathogen-Like Particles-Based Combination Adjuvants Elicit Potent Mucosal T Cell Immunity to Influenza A Virus. Front Immunol 2021; 11:559382. [PMID: 33767689 PMCID: PMC7986715 DOI: 10.3389/fimmu.2020.559382] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 12/21/2020] [Indexed: 12/17/2022] Open
Abstract
Eliciting durable and protective T cell-mediated immunity in the respiratory mucosa remains a significant challenge. Polylactic-co-glycolic acid (PLGA)-based cationic pathogen-like particles (PLPs) loaded with TLR agonists mimic biophysical properties of microbes and hence, simulate pathogen-pattern recognition receptor interactions to safely and effectively stimulate innate immune responses. We generated micro particle PLPs loaded with TLR4 (glucopyranosyl lipid adjuvant, GLA) or TLR9 (CpG) agonists, and formulated them with and without a mucosal delivery enhancing carbomer-based nanoemulsion adjuvant (ADJ). These adjuvants delivered intranasally to mice elicited high numbers of influenza nucleoprotein (NP)-specific CD8+ and CD4+ effector and tissue-resident memory T cells (TRMs) in lungs and airways. PLPs delivering TLR4 versus TLR9 agonists drove phenotypically and functionally distinct populations of effector and memory T cells. While PLPs loaded with CpG or GLA provided immunity, combining the adjuvanticity of PLP-GLA and ADJ markedly enhanced the development of airway and lung TRMs and CD4 and CD8 T cell-dependent immunity to influenza virus. Further, balanced CD8 (Tc1/Tc17) and CD4 (Th1/Th17) recall responses were linked to effective influenza virus control. These studies provide mechanistic insights into vaccine-induced pulmonary T cell immunity and pave the way for the development of a universal influenza and SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Brock Kingstad-Bakke
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Randall Toy
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Woojong Lee
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Pallab Pradhan
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Gabriela Vogel
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Chandranaik B Marinaik
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Autumn Larsen
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Daisy Gates
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Tracy Luu
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Bhawana Pandey
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - Yoshihoro Kawaoka
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| | - Krishnendu Roy
- The Wallace H. Coulter Department of Biomedical Engineering Georgia Institute of Technology and Emory University, Atlanta, GA, United States
| | - M Suresh
- Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
21
|
He Q, Ding G, Zhang M, Nie P, Yang J, Liang D, Bo J, Zhang Y, Liu Y. Trends in the Use of Sphingosine 1 Phosphate in Age-Related Diseases: A Scientometric Research Study (1992-2020). J Diabetes Res 2021; 2021:4932974. [PMID: 33791388 PMCID: PMC7984909 DOI: 10.1155/2021/4932974] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/13/2020] [Accepted: 02/01/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES This study was designed to explore the intellectual landscape of research into the application of sphingosine 1 phosphate (S1P) in age-related diseases and to identify thematic development trends and research frontiers in this area. METHODS Scientometric research was conducted by analyzing bibliographic records retrieved from the Web of Science (WOS) Sci-Expanded Database dated between 1900 and 2020. Countries, institutions, authors, keyword occurrence analysis, and cooperation network analysis were performed using the CiteSpace and VOSviewer software. RESULTS A total of 348 valid records were included in the final dataset, and the number of publications and the frequency of citations have grown rapidly over the last ten years. The USA (n = 175), China (n = 42), and Germany (n = 37) were the three largest contributors to the global publications on S1P and aging, while the Medical University of South Carolina (n = 15), University of California, San Francisco (n = 13), and University of Toronto (n = 13) were the leading institutions in this field. Analysis showed that early studies primarily focused on the mechanism of S1P intervention in AD. While S1P and its relevant metabolites have remained a long-term active area of research, recent studies have focused more on interventions aimed at improving retinal degeneration, cardiomyopathy, multiple sclerosis, and diabetes, among others. CONCLUSIONS It is worth mentioning that this manuscript is the first to describe any bibliometric analysis of S1P and its application in age-related interventions. This study includes a discussion of the (1) historical overview of the topic; (2) main contributors: journals, countries, institutes, funding agencies, and authors; (3) collaboration between institutes and authors; (4) research hot spots and zones; and 5) research trends and frontiers. This will enable scholars to understand the current status of S1P research in age-related diseases.
Collapse
Affiliation(s)
- Qiong He
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Gaofeng Ding
- Department of Second Medical College, Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Mengyuan Zhang
- Department of Plastic Surgery, First Hospital of Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Peng Nie
- Department of Radiotherapy, First Hospital of Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Jing Yang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Dong Liang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Jiaqi Bo
- Department of Second Medical College, Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan, 030001 Shanxi Province, China
| |
Collapse
|
22
|
He Q, Bo J, Shen R, Li Y, Zhang Y, Zhang J, Yang J, Liu Y. S1P Signaling Pathways in Pathogenesis of Type 2 Diabetes. J Diabetes Res 2021; 2021:1341750. [PMID: 34751249 PMCID: PMC8571914 DOI: 10.1155/2021/1341750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 02/06/2023] Open
Abstract
The pathogenesis of type 2 diabetes mellitus (T2DM) is very complicated. The currently well-accepted etiology is the "Ominous Octet" theory proposed by Professor Defronzo. Since presently used drugs for T2DM have limitations and harmful side effects, studies regarding alternative treatments are being conducted. Analyzing the pharmacological mechanism of biomolecules in view of pathogenesis is an effective way to assess new drugs. Sphingosine 1 phosphate (S1P), an endogenous lipid substance in the human body, has attracted increasing attention in the T2DM research field. This article reviews recent study updates of S1P, summarizing its effects on T2DM with respect to pathogenesis, promoting β cell proliferation and inhibiting apoptosis, reducing insulin resistance, protecting the liver and pancreas from lipotoxic damage, improving intestinal incretin effects, lowering basal glucagon levels, etc. With increasing research, S1P may help treat and prevent T2DM in the future.
Collapse
Affiliation(s)
- Qiong He
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Jiaqi Bo
- Department of Second Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Ruihua Shen
- Department of Second Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yan Li
- Department of Second Medical College, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yi Zhang
- Department of Pharmacology, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Jiaxin Zhang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Jing Yang
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| | - Yunfeng Liu
- Department of Endocrinology, First Hospital of Shanxi Medical University, Taiyuan 030001, Shanxi Province, China
| |
Collapse
|
23
|
Dezfuli NK, Adcock IM, Montazami N, Mortaz E, Velayati A. Update on Immunology of COVID-19 Disease and Potential Strategy for Controlling. TANAFFOS 2020; 19:274-290. [PMID: 33959165 PMCID: PMC8088154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 10/12/2020] [Indexed: 11/02/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by a novel form of the coronavirus that caused severe acute respiratory syndrome (SARS). SARS-CoV-2 raised in China and has broadcast to 261 countries globally. SARS-CoV-2 a member of β-coronavirus family and has an almost matching genome sequence to a bat coronavirus, pointing to the bat as the natural host before it was transmitted to humans. SARS-CoV-2 uses the same receptor, angiotensin-converting enzyme 2 (ACE2) as that used by SARS-CoV and principally infects the respiratory tract. The clinical symptoms of COVID-19 patients include fever, cough and fatigue whilst small populations of patients have gastrointestinal symptoms. The old people and people with underlying metabolic and cardiovascular diseases are more affected to infection and have worse outcomes. These may be associated with acute respiratory distress syndrome (ARDS) and a cytokine storm. In this review, we discuss the pathogenesis and clinical characteristics of disease and the pharmacologic approaches that may control COVID-19.
Collapse
Affiliation(s)
- Neda K. Dezfuli
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Dezful University of Medical Sciences, Dezful, Iran
| | - Ian M Adcock
- Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Nooshin Montazami
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Esmaeil Mortaz
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aliakbar Velayati
- Clinical Tuberculosis and Epidemiology Research Center, National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Cytokines and Transgenic Matrix in Autoimmune Diseases: Similarities and Differences. Biomedicines 2020; 8:biomedicines8120559. [PMID: 33271810 PMCID: PMC7761121 DOI: 10.3390/biomedicines8120559] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/16/2020] [Accepted: 11/26/2020] [Indexed: 12/14/2022] Open
Abstract
Autoimmune diseases are increasingly recognized as disease entities in which dysregulated cytokines contribute to tissue-specific inflammation. In organ-specific and multiorgan autoimmune diseases, the cytokine profiles show some similarities. Despite these similarities, the cytokines have different roles in the pathogenesis of different diseases. Altered levels or action of cytokines can result from changes in cell signaling. This article describes alterations in the JAK-STAT, TGF-β and NF-κB signaling pathways, which are involved in the pathogenesis of multiple sclerosis and systemic lupus erythematosus. There is a special focus on T cells in preclinical models and in patients afflicted with these chronic inflammatory diseases.
Collapse
|
25
|
Effects of Immunomodulatory Drug Fingolimod (FTY720) on Chlamydia Dissemination and Pathogenesis. Infect Immun 2020; 88:IAI.00281-20. [PMID: 32868341 DOI: 10.1128/iai.00281-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/24/2020] [Indexed: 12/21/2022] Open
Abstract
Fingolimod (FTY720), an FDA-approved immunomodulatory drug for treating multiple sclerosis, is an agonist of sphingosine-1-phosphate receptor (S1PR), which has been used as a research tool for inhibiting immune cell trafficking. FTY720 was recently reported to inhibit Chlamydia dissemination. Since genital Chlamydia spreading to the gastrointestinal tract correlated with its pathogenicity in the upper genital tract, we evaluated the effect of FTY720 on chlamydial pathogenicity in the current study. Following an intravaginal inoculation, live chlamydial organisms were detected in mouse rectal swabs. FTY720 treatment significantly delayed live organism shedding in the rectal swabs. However, FTY720 failed to block chlamydial spreading to the gastrointestinal tract. The live chlamydial organisms recovered from rectal swabs reached similar levels between mice with or without FTY720 treatment by day 42 in C57BL/6J and day 28 in CBA/J mice, respectively. Thus, genital Chlamydia is able to launch a 2nd wave of spreading via an FTY720-resistant pathway after the 1st wave of spreading is inhibited by FTY720. As a result, all mice developed significant hydrosalpinx. The FTY720-resistant spreading led to stable colonization of chlamydial organisms in the colon. Consistently, FTY720 did not alter the colonization of intracolonically inoculated Chlamydia Thus, we have demonstrated that, following a delay in chlamydial spreading caused by FTY720, genital Chlamydia is able to both spread to the gastrointestinal tract via an FTY720-resistant pathway and maintain its pathogenicity in the upper genital tract. Further characterization of the FTY720-resistant pathway(s) explored by Chlamydia for spreading to the gastrointestinal tract may promote our understanding of Chlamydia pathogenic mechanisms.
Collapse
|
26
|
Guo Y, Gan X, Zhou H, Zhou H, Pu S, Long X, Ren C, Feng T, Tang H. Fingolimod suppressed the chronic unpredictable mild stress-induced depressive-like behaviors via affecting microglial and NLRP3 inflammasome activation. Life Sci 2020; 263:118582. [PMID: 33058911 DOI: 10.1016/j.lfs.2020.118582] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 09/29/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
Depression is a common aspect of the modern lifestyle, and most patients are recalcitrant to the current antidepressants. Fingolimod (FTY720), a sphingosine analogue approved for the treatment of multiple sclerosis, has a significant neuroprotective effect on the central nervous system. The aim of this study was to determine the potential therapeutic effect of FTY720 on the behavior and cognitive function of rats exposed daily to chronic unpredictable mild stress (CUMS), and elucidate the underlying mechanisms. The 42-day CUMS modeling induced depression-like behavior as indicated by the scores of sugar water preference, forced swimming, open field and Morris water maze tests. Mechanistically, CUMS caused significant damage to the hippocampal neurons, increased inflammation and oxidative stress, activated the NF-κB/NLRP3 axis, and skewed microglial polarization to the M1 phenotype. FTY720 not only alleviated neuronal damage and oxidative stress, but also improved the depression-like behavior and cognitive function of the rats. It also inhibited NF-κB activation and blocked NLRP3 inflammasome assembly by down-regulating NLRP3, ACS and caspase-1. Furthermore, FTY720 inhibited the microglial M1 polarization markers iNOS and CD16, and promoted the M2 markers Arg-1 and CD206. This in turn reduced the levels of TNF-α, IL-6 and IL-1β, and increased that of IL-10 in the hippocampus. In conclusion, FTY720 protects hippocampal neurons from stress-induced damage and alleviates depressive symptoms by inhibiting neuroinflammation. Our study provides a theoretical basis for S1P receptor modulation in treating depression.
Collapse
Affiliation(s)
- Yuanxin Guo
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Xiaohong Gan
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Houfeng Zhou
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Hongjing Zhou
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Shiyun Pu
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Xia Long
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Changyu Ren
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Tao Feng
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China
| | - Hongmei Tang
- Department of Pharmacy, Chengdu Fifth People's Hospital, Chengdu, Sichuan 611130, China.
| |
Collapse
|
27
|
Si Y, Tian Q, Zhao F, Kelly SH, Shores LS, Camacho DF, Sperling AI, Andrade MS, Collier JH, Chong AS. Adjuvant-free nanofiber vaccine induces in situ lung dendritic cell activation and T H17 responses. SCIENCE ADVANCES 2020; 6:eaba0995. [PMID: 32821819 PMCID: PMC7413739 DOI: 10.1126/sciadv.aba0995] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 06/25/2020] [Indexed: 05/07/2023]
Abstract
The current paradigm that subunit vaccines require adjuvants to optimally activate innate immunity implies that increased vaccine reactogenicity will invariably be linked to improved immunogenicity. Countering this paradigm, nanoparticulate vaccines have been reported to act as delivery systems for vaccine antigens and induce immunity without the need for exogenous adjuvants or local inflammation; however, the mechanisms underlying the immunogenicity of nanoparticle vaccines are incompletely identified. Here, we show that antigens displayed on self-assembling nanofiber scaffolds and delivered intranasally are presented by CD103+ and CD11b+ lung dendritic cells that up-regulate CD80 and migrate into the draining lymph node (LN). This was accompanied by a nearly exclusive priming and accumulation of antigen-specific TH17 cells occurring independently in both LN and lung. Thus, self-assembling peptide nanofiber vaccines may represent a novel, needle- and adjuvant-free means of eliciting protective immunity against fungal and bacterial infections at skin and mucosal barrier surfaces.
Collapse
Affiliation(s)
- Youhui Si
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Qiaomu Tian
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Fan Zhao
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Sean H. Kelly
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
| | - Lucas S. Shores
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
| | - Daniel F. Camacho
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Anne I. Sperling
- Department of Medicine, The University of Chicago, Chicago, IL 60637, USA
| | - Michael S. Andrade
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
| | - Joel H. Collier
- Biomedical Engineering Department, Duke University, Durham, NC 27708, USA
- Corresponding author. (A.S.C.); (J.H.C.)
| | - Anita S. Chong
- Department of Surgery, The University of Chicago, Chicago, IL 60637, USA
- Corresponding author. (A.S.C.); (J.H.C.)
| |
Collapse
|
28
|
Kipp M. Does Siponimod Exert Direct Effects in the Central Nervous System? Cells 2020; 9:cells9081771. [PMID: 32722245 PMCID: PMC7463861 DOI: 10.3390/cells9081771] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 12/11/2022] Open
Abstract
The modulation of the sphingosine 1-phosphate receptor is an approved treatment for relapsing multiple sclerosis because of its anti-inflammatory effect of retaining lymphocytes in lymph nodes. Different sphingosine 1-phosphate receptor subtypes are expressed in the brain and spinal cord, and their pharmacological effects may improve disease development and neuropathology. Siponimod (BAF312) is a novel sphingosine 1-phosphate receptor modulator that has recently been approved for the treatment of active secondary progressive multiple sclerosis (MS). In this review article, we summarize recent evidence suggesting that the active role of siponimod in patients with progressive MS may be due to direct interaction with central nervous system cells. Additionally, we tried to summarize our current understanding of the function of siponimod and discuss the effects observed in the case of MS.
Collapse
Affiliation(s)
- Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstrasse 9, 18057 Rostock, Germany
| |
Collapse
|
29
|
Abstract
Multiple sclerosis (MS) is an aggravating autoimmune disease that cripples young patients slowly with physical, sensory and cognitive deficits. The break of self-tolerance to neuronal antigens is the key to the pathogenesis of MS, with autoreactive T cells causing demyelination that subsequently leads to inflammation-mediated neurodegenerative events in the central nervous system. The exact etiology of MS remains elusive; however, the interplay of genetic and environmental factors contributes to disease development and progression. Given that genetic variation only accounts for a fraction of risk for MS, extrinsic risk factors including smoking, infection and lack of vitamin D or sunshine, which cause changes in gene expression, contribute to disease development through epigenetic regulation. To date, there is a growing body of scientific evidence to support the important roles of epigenetic processes in MS. In this chapter, the three main layers of epigenetic regulatory mechanisms, namely DNA methylation, histone modification and microRNA-mediated gene regulation, will be discussed, with a particular focus on the role of epigenetics on dysregulated immune responses and neurodegenerative events in MS. Also, the potential for epigenetic modifiers as biomarkers and therapeutics for MS will be reviewed.
Collapse
Affiliation(s)
- Vera Sau-Fong Chan
- Department of Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
- Queen Mary Hospital, Hong Kong SAR, China.
| |
Collapse
|
30
|
Abstract
It has been over 100 years since the 1918 influenza pandemic, one of the most infamous examples of viral immunopathology. Since that time, there has been an inevitable repetition of influenza pandemics every few decades and yearly influenza seasons, which have a significant impact on human health. Recently, noteworthy progress has been made in defining the cellular and molecular mechanisms underlying pathology induced by an exuberant host response to influenza virus infection. Infection with influenza viruses is associated with a wide spectrum of disease, from mild symptoms to severe complications including respiratory failure, and the severity of influenza disease is driven by a complex interplay of viral and host factors. This chapter will discuss mechanisms of infection severity using concepts of disease resistance and tolerance as a framework for understanding the balance between viral clearance and immunopathology. We review mechanistic studies in animal models of infection and correlational studies in humans that have begun to define these factors and discuss promising host therapeutic targets to improve outcomes from severe influenza disease.
Collapse
Affiliation(s)
- David F Boyd
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States
| | - Taylor L Wilson
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States; Department of Microbiology, Immunology, and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Paul G Thomas
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, United States; Department of Microbiology, Immunology, and Biochemistry, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, United States.
| |
Collapse
|
31
|
Li CX, Yang XX, Wang HW, Li XC, Ng KTP, Lo CM, Man K. FTY720 Suppresses Liver Tumor Growth and Metastasis by Reducing Circulating Regulating T Cells and Enhancing the Anti-Tumor Effect of Rapamycin. Onco Targets Ther 2020; 13:4743-4754. [PMID: 32547103 PMCID: PMC7262652 DOI: 10.2147/ott.s234394] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Accepted: 02/25/2020] [Indexed: 01/13/2023] Open
Abstract
Background In this study, we aimed to study the effect of FTY720 treatment in reducing circulating Tregs level and then suppressing liver tumor metastasis after hepatectomy and I/R injury in animal models. Furthermore, we also investigated the synergistic anti-tumor effect of FTY720 combined with rapamycin on hepatocellular carcinoma. Methods The effect of FTY720 on suppressing Tregs mobilization and tumor metastasis after hepatectomy was investigated in an orthotopic liver tumor rat model with hepatectomy and hepatic ischemia/reperfusion (I/R) injury. The synergistic anti-tumor effect of FTY720 combined with rapamycin was further explored both in in vitro functional study and in orthotopic liver tumor mouse model. Results In rat model, hepatic I/R promoted tumor metastasis and increased circulating Tregs after hepatectomy. The treatment of FTY720 reduced liver tumor metastasis and the number of circulating Tregs. Furthermore, FTY720 enhanced the anti-tumor capacity of rapamycin by inhibiting tumor cell proliferation and migration in vitro and reducing tumor growth in vivo through suppressing hepatic stellate cell activation and tumor angiogenesis. Conclusion FTY720 suppressed liver tumor growth and metastasis by reducing the population of circulating Tregs and enhancing the anti-tumor effect of rapamycin. It was suggested that FTY720 single or combined with rapamycin might provide novel insight for suppressing tumor growth and metastasis for HCC patients.
Collapse
Affiliation(s)
- Chang Xian Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, Nanjing, Jiangsu Province, People's Republic of China
| | - Xin Xiang Yang
- Department of Surgery, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Hong Wei Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, Nanjing, Jiangsu Province, People's Republic of China
| | - Xiang Cheng Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Living Donor Liver Transplantation, Nanjing, Jiangsu Province, People's Republic of China
| | - Kevin Tak-Pan Ng
- Department of Surgery, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Chung Mau Lo
- Department of Surgery, The University of Hong Kong, Hong Kong, People's Republic of China
| | - Kwan Man
- Department of Surgery, The University of Hong Kong, Hong Kong, People's Republic of China
| |
Collapse
|
32
|
Ranmuthu CDS, Ranmuthu CKI, Russell JC, Singhania D, Khan WS. Evaluating the Effect of Non-cellular Bioactive Glass-Containing Scaffolds on Osteogenesis and Angiogenesis in in vivo Animal Bone Defect Models. Front Bioeng Biotechnol 2020; 8:430. [PMID: 32478053 PMCID: PMC7240009 DOI: 10.3389/fbioe.2020.00430] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
The use of bone scaffolds to replace injured or diseased bone has many advantages over the currently used autologous and allogeneic options in clinical practice. This systematic review evaluates the current evidence for non-cellular scaffolds containing bioactive glass on osteogenesis and angiogenesis in animal bone defect models. Studies that reported results of osteogenesis via micro-CT and results of angiogenesis via Microfil perfusion or immunohistochemistry were included in the review. A literature search of PubMed, EMBASE and Scopus was carried out in November 2019 from which nine studies met the inclusion and exclusion criteria. Despite the significant heterogeneity in the composition of the scaffolds used in each study, it could be concluded that scaffolds containing bioactive glass improve bone regeneration in these models, both by osteogenic and angiogenic measures. Incorporation of additional elements into the glass network, using additives, and using biochemical factors generally had a beneficial effect. Comparing the different compositions of non-cellular bioactive glass containing scaffolds is however difficult due to the heterogeneity in bioactive glass compositions, fabrication methods and biochemical additives used.
Collapse
Affiliation(s)
| | | | - Jodie C. Russell
- Cambridge Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - Disha Singhania
- Cambridge Clinical School, University of Cambridge, Cambridge, United Kingdom
| | - Wasim S. Khan
- Division of Trauma and Orthopaedics, Department of Surgery, Addenbrooke's Hospital, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
33
|
Markovic M, Ben-Shabat S, Aponick A, Zimmermann EM, Dahan A. Lipids and Lipid-Processing Pathways in Drug Delivery and Therapeutics. Int J Mol Sci 2020; 21:ijms21093248. [PMID: 32375338 PMCID: PMC7247327 DOI: 10.3390/ijms21093248] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 04/28/2020] [Accepted: 05/02/2020] [Indexed: 12/19/2022] Open
Abstract
The aim of this work is to analyze relevant endogenous lipid processing pathways, in the context of the impact that lipids have on drug absorption, their therapeutic use, and utilization in drug delivery. Lipids may serve as biomarkers of some diseases, but they can also provide endogenous therapeutic effects for certain pathological conditions. Current uses and possible clinical benefits of various lipids (fatty acids, steroids, triglycerides, and phospholipids) in cancer, infectious, inflammatory, and neurodegenerative diseases are presented. Lipids can also be conjugated to a drug molecule, accomplishing numerous potential benefits, one being the improved treatment effect, due to joined influence of the lipid carrier and the drug moiety. In addition, such conjugates have increased lipophilicity relative to the parent drug. This leads to improved drug pharmacokinetics and bioavailability, the ability to join endogenous lipid pathways and achieve drug targeting to the lymphatics, inflamed tissues in certain autoimmune diseases, or enable overcoming different barriers in the body. Altogether, novel mechanisms of the lipid role in diseases are constantly discovered, and new ways to exploit these mechanisms for the optimal drug design that would advance different drug delivery/therapy aspects are continuously emerging.
Collapse
Affiliation(s)
- Milica Markovic
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Shimon Ben-Shabat
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
| | - Aaron Aponick
- Department of Chemistry, University of Florida, Gainesville, FL 32603, USA;
| | - Ellen M. Zimmermann
- Department of Medicine, Division of Gastroenterology, University of Florida, Gainesville, FL 32610, USA;
| | - Arik Dahan
- Department of Clinical Pharmacology, School of Pharmacy, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel; (M.M.); (S.B.-S.)
- Correspondence:
| |
Collapse
|
34
|
Chen M, Qiao G, Hylander BL, Mohammadpour H, Wang XY, Subjeck JR, Singh AK, Repasky EA. Adrenergic stress constrains the development of anti-tumor immunity and abscopal responses following local radiation. Nat Commun 2020; 11:1821. [PMID: 32286326 PMCID: PMC7156731 DOI: 10.1038/s41467-020-15676-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 03/18/2020] [Indexed: 12/13/2022] Open
Abstract
The abscopal effect following ionizing radiation therapy (RT) is considered to be a rare event. This effect does occur more frequently when combined with other therapies, including immunotherapy. Here we demonstrate that the frequency of abscopal events following RT alone is highly dependent upon the degree of adrenergic stress in the tumor-bearing host. Using a combination of physiologic, pharmacologic and genetic strategies, we observe improvements in the control of both irradiated and non-irradiated distant tumors, including metastatic tumors, when adrenergic stress or signaling through β-adrenergic receptor is reduced. Further, we observe cellular and molecular evidence of improved, antigen-specific, anti-tumor immune responses which also depend upon T cell egress from draining lymph nodes. These data suggest that blockade of β2 adrenergic stress signaling could be a useful, safe, and feasible strategy to improve efficacy in cancer patients undergoing radiation therapy.
Collapse
MESH Headings
- Adrenergic Agents/pharmacology
- Adrenergic beta-Antagonists/pharmacology
- Animals
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Disease Models, Animal
- Gene Expression Regulation, Neoplastic/drug effects
- Immunity
- Lymph Nodes/pathology
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/radiotherapy
- Radiation, Ionizing
- Receptors, Adrenergic, beta-2/metabolism
- Signal Transduction/drug effects
- Stress, Physiological
Collapse
Affiliation(s)
- Minhui Chen
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Guanxi Qiao
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Bonnie L Hylander
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Hemn Mohammadpour
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Xiang-Yang Wang
- Department of Genetics, Virginia Commonwealth University, Richmond, VI, 23298, USA
| | - John R Subjeck
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Anurag K Singh
- Department of Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
- Department of Radiation Oncology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA
| | - Elizabeth A Repasky
- Department of Immunology, Roswell Park Comprehensive Cancer Center, Elm and Carlton Streets, Buffalo, NY, 14263, USA.
| |
Collapse
|
35
|
Marciniak A, Camp SM, Garcia JGN, Polt R. In silico Docking Studies of Fingolimod and S1P 1 Agonists. Front Pharmacol 2020; 11:247. [PMID: 32210822 PMCID: PMC7076195 DOI: 10.3389/fphar.2020.00247] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 02/24/2020] [Indexed: 11/13/2022] Open
Abstract
The sphingosine-1-phosphate receptor 1 (S1P1), originally the endothelial differentiation gene 1 receptor (EDG-1), is one of five G protein–coupled receptors (GPCRs) S1P1–5 that bind to and are activated by sphingosine-1-phosphate (S1P). The lipid S1P is an intermediate in sphingolipid homeostasis, and S1P1 is a major medical target for immune system modulation; agonism of the receptor produces a myriad of biological responses, including endothelial cell barrier integrity, chemotaxis, lymphocyte trafficking/targeting, angiogenesis, as well as regulation of the cardiovascular system. Use of in silico docking simulations on the crystal structure of S1P1 allows for pinpointing the residues within the receptor’s active site that actively contribute to the binding of S1P, and point to how these specific interactions can be exploited to design more effective synthetic analogs to specifically target S1P1 in the presence of the closely related receptors S1P2, S1P3, S1P4, and S1P5. We examined the binding properties of the endogenous substrate as well as a selection of synthetic sphingosine-derived S1P1 modulators of S1P1 with in silico docking simulations using the software package Molecular Operating Environment® (MOE®). The modeling studies reveal the relevance of phosphorylation, i.e., the presence of a phosphate or phosphonate moiety within the substrate for successful binding to occur, and indicate which residues are responsible for S1P1 binding of the most prominent sphingosine-1-phosphate receptor (S1PR) modulators, including fingolimod and its structural relatives. Furthermore, trends in steric preferences as for the binding of enantiomers to S1P1 could be observed, facilitating future design of receptor-specific substrates to precisely target the active site of S1P1.
Collapse
Affiliation(s)
- Alexander Marciniak
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, United States
| | - Sara M Camp
- Department of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Joe G N Garcia
- Department of Medicine, The University of Arizona, Tucson, AZ, United States
| | - Robin Polt
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, United States
| |
Collapse
|
36
|
Wang Z, Higashikawa K, Yasui H, Kuge Y, Ohno Y, Kihara A, Midori YA, Houkin K, Kawabori M. FTY720 Protects Against Ischemia-Reperfusion Injury by Preventing the Redistribution of Tight Junction Proteins and Decreases Inflammation in the Subacute Phase in an Experimental Stroke Model. Transl Stroke Res 2020; 11:1103-1116. [PMID: 32103462 PMCID: PMC7496052 DOI: 10.1007/s12975-020-00789-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 01/22/2020] [Accepted: 02/12/2020] [Indexed: 12/18/2022]
Abstract
Injury due to brain ischemia followed by reperfusion (I/R) may be an important therapeutic target in the era of thrombectomy. FTY720, a widely known sphingosine-1-phosphate receptor agonist, exerts various neuroprotective effects. The aim of this study was to examine the protective effect of FTY720 with respect to I/R injury, especially focusing on blood-brain barrier (BBB) protection and anti-inflammatory effects. Male rats were subjected to transient ischemia and administered vehicle or 0.5 or 1.5 mg/kg of FTY720 immediately before reperfusion. Positron emission tomography (PET) with [18F]DPA-714 was performed 2 and 9 days after the insult to serially monitor neuroinflammation. Bovine and rat brain microvascular endothelial cells (MVECs) were also subjected to oxygen-glucose deprivation (OGD) and reperfusion, and administered FTY720, phosphorylated-FTY720 (FTY720-P), or their inhibitor. FTY720 dose-dependently reduced cell death, the infarct size, cell death including apoptosis, and inflammation. It also ameliorated BBB disruption and neurological deficits compared to in the vehicle group. PET indicated that FTY720 significantly inhibited the worsening of inflammation in later stages. FTY720-P significantly prevented the intracellular redistribution of tight junction proteins but did not increase their mRNA expression. These results suggest that FTY720 can ameliorate I/R injury by protecting the BBB and regulating neuroinflammation.
Collapse
Affiliation(s)
- Zifeng Wang
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Kei Higashikawa
- Central Institutes of Isotope Science (Laboratory of Integrated Molecular Imaging, Department of Biomedical Imaging, Graduate School of Biomedical Science and Engineering), Hokkaido University, Sapporo, Hokkaido, Japan
| | - Hironobu Yasui
- Central Institutes of Isotope Science (Laboratory of Integrated Molecular Imaging, Department of Biomedical Imaging, Graduate School of Biomedical Science and Engineering), Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yuji Kuge
- Central Institutes of Isotope Science (Laboratory of Integrated Molecular Imaging, Department of Biomedical Imaging, Graduate School of Biomedical Science and Engineering), Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yusuke Ohno
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Akio Kihara
- Laboratory of Biochemistry, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yenari A Midori
- Department of Neurology, University of California, San Francisco and the San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Kiyohiro Houkin
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Graduate School of Medicine, Hokkaido University, Kita 15, Nishi 7, Kita-ku, Sapporo, Hokkaido, 060-8638, Japan.
| |
Collapse
|
37
|
Fingolimod inhibits proliferation and epithelial-mesenchymal transition in sacral chordoma by inactivating IL-6/STAT3 signalling. Biosci Rep 2020; 40:222049. [PMID: 32027356 PMCID: PMC7029154 DOI: 10.1042/bsr20200221] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Revised: 02/05/2020] [Accepted: 02/05/2020] [Indexed: 12/18/2022] Open
Abstract
Purpose: To explore the sensitivity of the immunosuppressive agent fingolimod (FTY720) in chordoma and determine whether it can serve as an appropriate alternate treatment for unresectable tumours in patients after incomplete surgery. Methods: Cell viability assays, colony formation assays and EdU assays were performed to evaluate the sensitivity of chordoma cell lines to FTY720. Transwell invasion assays, wound healing assays, flow cytometry, cell cycle analysis, immunofluorescence analysis, Western blotting analysis and enzyme-linked immunosorbent assays (ELISAs) were performed to evaluate cell invasion, epithelial–mesenchymal transition (EMT) and activation of related pathways after treatment with FTY720. The effect of FTY720 was also evaluated in vivo in a xenograft model. Results: We found that FTY720 inhibited the proliferation, invasion and metastasis of sacral chordoma cells (P < 0.01). FTY720 also inhibited the proliferation of tumour cells in a xenograft model using sacral chordoma cell lines (P < 0.01). The mechanism was related to the EMT and apoptosis of chordoma cells and inactivation of IL-6/STAT3 signalling in vitro and in vivo. Conclusions: Our findings indicate that FTY720 may be an effective therapeutic agent against chordoma. These findings suggest that FTY720 is a novel agent that can treat locally advanced and metastatic chordoma.
Collapse
|
38
|
Shang Z, Sun W, Zhang M, Xu L, Jia X, Zhang R, Fu S. Identification of key genes associated with multiple sclerosis based on gene expression data from peripheral blood mononuclear cells. PeerJ 2020; 8:e8357. [PMID: 32117605 PMCID: PMC7003695 DOI: 10.7717/peerj.8357] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 12/04/2019] [Indexed: 11/20/2022] Open
Abstract
The aim of this study was to identify the potential key candidate genes of multiple sclerosis (MS) and uncover mechanisms in MS. We combined data from the microarray expression profile of three MS stages and performed bioinformatics analysis. Differentially expressed genes (DEGs) were identified among the distinct stages of MS and healthy controls, and a total of 349 shared DEGs were identified. Gene ontology (GO) and pathway enrichment analyses showed that the DEGs were significantly enriched in the biological processes (BPs) of purine-related metabolic processes and signaling, especially the common DEGs, which were enriched in some immunological processes. Most of the DEGs were enriched in signaling pathways associated with the immune system, some immune diseases and infectious disease pathways. Through a protein-protein interaction (PPI) network analysis and a gene expression regulatory network constructed with MS-related miRNAs, we confirmed FOS, TP53, VEGFA, JUN, HIF1A, RB1, PTGS2, CXCL8, OAS2, NFKBIA and OAS1 as candidate genes of MS. Furthermore , we explored the potential SNPs associated with MS by database mining. In conclusion, this study provides the identified genes, SNPs, biological processes, and cellular pathways associated with MS. The uncovered candidate genes may be potential biomarkers involved in the diagnosis and therapy of MS.
Collapse
Affiliation(s)
- Zhenwei Shang
- Harbin Medical University, Laboratory of Medical Genetics, Harbin, China.,Harbin Medical University, Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin, China.,Harbin Medical University, College of Bioinformatics Science and Technology, Harbin, China
| | - Wenjing Sun
- Harbin Medical University, Laboratory of Medical Genetics, Harbin, China.,Harbin Medical University, Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin, China
| | - Mingming Zhang
- Harbin Medical University, Laboratory of Medical Genetics, Harbin, China.,Harbin Medical University, Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin, China.,Harbin Medical University, College of Bioinformatics Science and Technology, Harbin, China
| | - Lidan Xu
- Harbin Medical University, Laboratory of Medical Genetics, Harbin, China.,Harbin Medical University, Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin, China
| | - Xueyuan Jia
- Harbin Medical University, Laboratory of Medical Genetics, Harbin, China.,Harbin Medical University, Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin, China
| | - Ruijie Zhang
- Harbin Medical University, College of Bioinformatics Science and Technology, Harbin, China
| | - Songbin Fu
- Harbin Medical University, Laboratory of Medical Genetics, Harbin, China.,Harbin Medical University, Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China, Ministry of Education, Harbin, China
| |
Collapse
|
39
|
Kemp JA, Keebaugh A, Edson JA, Chow D, Kleinman MT, Chew YC, McCracken AN, Edinger AL, Kwon YJ. Biocompatible Chemotherapy for Leukemia by Acid-Cleavable, PEGylated FTY720. Bioconjug Chem 2020; 31:673-684. [PMID: 31986014 DOI: 10.1021/acs.bioconjchem.9b00822] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Targeting the inability of cancerous cells to adapt to metabolic stress is a promising alternative to conventional cancer chemotherapy. FTY720 (Gilenya), an FDA-approved drug for the treatment of multiple sclerosis, has recently been shown to inhibit cancer progression through the down-regulation of essential nutrient transport proteins, selectively starving cancer cells to death. However, the clinical use of FTY720 for cancer therapy is prohibited because of its capability of inducing immunosuppression (lymphopenia) and bradycardia when phosphorylated upon administration. A prodrug to specifically prevent phosphorylation during circulation, hence avoiding bradycardia and lymphopenia, was synthesized by capping its hydroxyl groups with polyethylene glycol (PEG) via an acid-cleavable ketal linkage. Improved aqueous solubility was also accomplished by PEGylation. The prodrug reduces to fully potent FTY720 upon cellular uptake and induces metabolic stress in cancer cells. Enhanced release of FTY720 at a mildly acidic endosomal pH and the ability to substantially down-regulate cell-surface nutrient transporter proteins in leukemia cells only by an acid-cleaved drug were confirmed. Importantly, the prodrug demonstrated nearly identical efficacy to FTY720 in an animal model of BCR-Abl-driven leukemia without inducing bradycardia or lymphopenia in vivo, highlighting its potential clinical value. The prodrug formulation of FTY720 demonstrates the utility of precisely engineering a drug to avoid undesirable effects by tackling specific molecular mechanisms as well as a financially favorable alternative to new drug development. A multitude of existing cancer therapeutics may be explored for prodrug formulation to avoid specific side effects and preserve or enhance therapeutic efficacy.
Collapse
Affiliation(s)
| | | | | | | | | | - Yap Ching Chew
- Zymo Research Corporation, Irvine, California 92604, United States
| | | | | | | |
Collapse
|
40
|
Wang Z, Kawabori M, Houkin K. FTY720 (Fingolimod) Ameliorates Brain Injury through Multiple Mechanisms and is a Strong Candidate for Stroke Treatment. Curr Med Chem 2020; 27:2979-2993. [PMID: 31785606 PMCID: PMC7403647 DOI: 10.2174/0929867326666190308133732] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 02/12/2019] [Accepted: 02/19/2019] [Indexed: 02/07/2023]
Abstract
FTY720 (Fingolimod) is a known sphingosine-1-phosphate (S1P) receptor agonist that exerts strong anti-inflammatory effects and was approved as the first oral drug for the treatment of multiple sclerosis by the US Food and Drug Administration (FDA) in 2010. FTY720 is mainly associated with unique functional "antagonist" and "agonist" mechanisms. The functional antagonistic mechanism is mediated by the transient down-regulation and degradation of S1P receptors on lymphocytes, which prevents lymphocytes from entering the blood stream from the lymph node. This subsequently results in the development of lymphopenia and reduces lymphocytic inflammation. Functional agonistic mechanisms are executed through S1P receptors expressed on the surface of various cells including neurons, astrocytes, microglia, and blood vessel endothelial cells. These functions might play important roles in regulating anti-apoptotic systems, modulating brain immune and phagocytic activities, preserving the Blood-Brain-Barrier (BBB), and the proliferation of neural precursor cells. Recently, FTY720 have shown receptor-independent effects, including intracellular target bindings and epigenetic modulations. Many researchers have recognized the positive effects of FTY720 and launched basic and clinical experiments to test the use of this agent against stroke. Although the mechanism of FTY720 has not been fully elucidated, its efficacy against cerebral stroke is becoming clear, not only in animal models, but also in ischemic stroke patients through clinical trials. In this article, we review the data obtained from laboratory findings and preliminary clinical trials using FTY720 for stroke treatment.
Collapse
Affiliation(s)
- Zifeng Wang
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Masahito Kawabori
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Kiyohiro Houkin
- Department of Neurosurgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
41
|
Fingolimod (FTY720) improves the functional recovery and myelin preservation of the optic pathway in focal demyelination model of rat optic chiasm. Brain Res Bull 2019; 153:109-121. [DOI: 10.1016/j.brainresbull.2019.08.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 08/09/2019] [Accepted: 08/18/2019] [Indexed: 12/21/2022]
|
42
|
Geraghty JR, Davis JL, Testai FD. Neuroinflammation and Microvascular Dysfunction After Experimental Subarachnoid Hemorrhage: Emerging Components of Early Brain Injury Related to Outcome. Neurocrit Care 2019; 31:373-389. [PMID: 31012056 PMCID: PMC6759381 DOI: 10.1007/s12028-019-00710-x] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Aneurysmal subarachnoid hemorrhage has a high mortality rate and, for those who survive this devastating injury, can lead to lifelong impairment. Clinical trials have demonstrated that cerebral vasospasm of larger extraparenchymal vessels is not the sole contributor to neurological outcome. Recently, the focus of intense investigation has turned to mechanisms of early brain injury that may play a larger role in outcome, including neuroinflammation and microvascular dysfunction. Extravasated blood after aneurysm rupture results in a robust inflammatory response characterized by activation of microglia, upregulation of cellular adhesion molecules, recruitment of peripheral immune cells, as well as impaired neurovascular coupling, disruption of the blood-brain barrier, and imbalances in endogenous vasodilators and vasoconstrictors. Each of these phenomena is either directly or indirectly associated with neuronal death and brain injury. Here, we review recent studies investigating these various mechanisms in experimental models of subarachnoid hemorrhage with special emphasis on neuroinflammation and its effect on microvascular dysfunction. We discuss the various therapeutic targets that have risen from these mechanistic studies and suggest the utility of a multi-targeted approach to preventing delayed injury and improving outcome after subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Joseph R Geraghty
- Department of Neurology and Rehabilitation, College of Medicine, University of Illinois at Chicago, 912 S. Wood St. Suite 174N, Chicago, IL, 60612, USA.
- Medical Scientist Training Program, University of Illinois at Chicago, Chicago, IL, USA.
| | - Joseph L Davis
- Department of Neurology and Rehabilitation, College of Medicine, University of Illinois at Chicago, 912 S. Wood St. Suite 174N, Chicago, IL, 60612, USA
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, College of Medicine, University of Illinois at Chicago, 912 S. Wood St. Suite 174N, Chicago, IL, 60612, USA
| |
Collapse
|