1
|
Bibi A, Yu Z, Cui L, Yang G. Harnessing monocyte dynamics for treatment of multiple sclerosis; insights from experimental model studies. IMMUNOTHERAPY ADVANCES 2025; 5:ltaf003. [PMID: 40342728 PMCID: PMC12059560 DOI: 10.1093/immadv/ltaf003] [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: 09/08/2024] [Accepted: 03/27/2025] [Indexed: 05/11/2025] Open
Abstract
Monocytes are central to the innate immune system's response to infection or injury. In murine, these cells are classified into distinct subsets: classical monocytes, defined by elevated Ly6C expression (Ly6Chi), intermediate monocytes (Ly6Cint), and non-classical inflammatory monocytes, characterized by low Ly6C expression (Ly6Clow). Monocytes recruited to tissues differentiate into macrophages, which can be pro-inflammatory or anti-inflammatory, thereby influencing disease processes and outcomes. The principal function of classical monocytes is the mediation of pro-inflammatory reactions, whereas non-classical monocytes are associated with repair and anti-inflammatory processes, patrolling the lumen of the vessels. Growing evidence highlights the importance of monocytes in multiple sclerosis (MS), an autoimmune and neurodegenerative disease of the central nervous system (CNS). Recent studies indicate that modulation of the innate immune system, focusing specifically on the shift from Ly6Chi to Ly6Clow monocytes, is an effective therapeutic strategy for neurodegenerative diseases, such as Alzheimer's and MS. This transition is crucial for switching the immune response from inflammation to tissue repair and inflammation resolution, emphasizing the plasticity of monocytes and their potential as targets in MS. This review differs from prior studies in that it focuses solely on animal models of MS, which either directly perturb or study monocytes, or where therapeutic approaches mediate their protective effects through monocytes. Such details permit a subtle comprehension of monocyte dynamics in the context of MS.
Collapse
Affiliation(s)
- Aqsa Bibi
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250061, China
| | - Zhenjiang Yu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250061, China
| | - Lv Cui
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Guiwen Yang
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan 250061, China
| |
Collapse
|
2
|
Doghish AS, Elazazy O, Mohamed HH, Mansour RM, Ghanem A, Faraag AHI, Elballal MS, Elrebehy MA, Elesawy AE, Abdel Mageed SS, Mohammed OA, Nassar YA, Abulsoud AI, Raouf AA, Abdel-Reheim MA, Rashad AA, Elawady AS, Elsisi AM, Alsalme A, Ali MA. The role of miRNAs in multiple sclerosis pathogenesis, diagnosis, and therapeutic resistance. Pathol Res Pract 2023; 251:154880. [PMID: 37832353 DOI: 10.1016/j.prp.2023.154880] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023]
Abstract
In recent years, microRNAs (miRNAs) have gained increased attention from researchers around the globe. Although it is twenty nucleotides long, it can modulate several gene targets simultaneously. Their mal expression is a signature of various pathologies, and they provide the foundation to elucidate the molecular mechanisms of each pathology. Among the debilitating central nervous system (CNS) disorders with a growing prevalence globally is the multiple sclerosis (MS). Moreover, the diagnosis of MS is challenging due to the lack of disease-specific biomarkers, and the diagnosis mainly depends on ruling out other disabilities. MS could adversely affect patients' lives through its progression, and only symptomatic treatments are available as therapeutic options, but an exact cure is yet unavailable. Consequently, this review hopes to further the study of the biological features of miRNAs in MS and explore their potential as a therapeutic target.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt.
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Hend H Mohamed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Reda M Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt; Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed H I Faraag
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Osama A Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Yara A Nassar
- Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt; Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Ahmed Amr Raouf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Ahmed A Rashad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Alaa S Elawady
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Ahmed Mohammed Elsisi
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt; Department of Biochemistry, Faculty of Pharmacy, Sinai University, Al-Arish, Egypt
| | - Ali Alsalme
- Chemistry Department, College of Science, King Saud University, Riyadh 1145, Saudi Arabia
| | - Mohamed A Ali
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| |
Collapse
|
3
|
Diebold M, Fehrenbacher L, Frosch M, Prinz M. How myeloid cells shape experimental autoimmune encephalomyelitis: At the crossroads of outside-in immunity. Eur J Immunol 2023; 53:e2250234. [PMID: 37505465 DOI: 10.1002/eji.202250234] [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: 02/02/2023] [Revised: 05/21/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE) is an animal model of central nervous system (CNS) autoimmunity. It is most commonly used to mimic aspects of multiple sclerosis (MS), a demyelinating disorder of the human brain and spinal cord. The innate immune response displays one of the core pathophysiological features linked to both the acute and chronic stages of MS. Hence, understanding and targeting the innate immune response is essential. Microglia and other CNS resident MUs, as well as infiltrating myeloid cells, diverge substantially in terms of both their biology and their roles in EAE. Recent advances in the field show that antigen presentation, as well as disease-propagating and regulatory interactions with lymphocytes, can be attributed to specific myeloid cell types and cell states in EAE lesions, following a distinct temporal pattern during disease initiation, propagation and recovery. Furthermore, single-cell techniques enable the assessment of characteristic proinflammatory as well as beneficial cell states, and identification of potential treatment targets. Here, we discuss the principles of EAE induction and protocols for varying experimental paradigms, the composition of the myeloid compartment of the CNS during health and disease, and systematically review effects on myeloid cells for therapeutic approaches in EAE.
Collapse
Affiliation(s)
- Martin Diebold
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Luca Fehrenbacher
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Maximilian Frosch
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, University Medical Center Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| |
Collapse
|
4
|
Cashion JM, Young KM, Sutherland BA. How does neurovascular unit dysfunction contribute to multiple sclerosis? Neurobiol Dis 2023; 178:106028. [PMID: 36736923 DOI: 10.1016/j.nbd.2023.106028] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 01/17/2023] [Accepted: 01/30/2023] [Indexed: 02/04/2023] Open
Abstract
Multiple sclerosis is an inflammatory demyelinating disease of the central nervous system (CNS) and the most common non-traumatic cause of neurological disability in young adults. Multiple sclerosis clinical care has improved considerably due to the development of disease-modifying therapies that effectively modulate the peripheral immune response and reduce relapse frequency. However, current treatments do not prevent neurodegeneration and disease progression, and efforts to prevent multiple sclerosis will be hampered so long as the cause of this disease remains unknown. Risk factors for multiple sclerosis development or severity include vitamin D deficiency, cigarette smoking and youth obesity, which also impact vascular health. People with multiple sclerosis frequently experience blood-brain barrier breakdown, microbleeds, reduced cerebral blood flow and diminished neurovascular reactivity, and it is possible that these vascular pathologies are tied to multiple sclerosis development. The neurovascular unit is a cellular network that controls neuroinflammation, maintains blood-brain barrier integrity, and tightly regulates cerebral blood flow, matching energy supply to neuronal demand. The neurovascular unit is composed of vessel-associated cells such as endothelial cells, pericytes and astrocytes, however neuronal and other glial cell types also comprise the neurovascular niche. Recent single-cell transcriptomics data, indicate that neurovascular cells, particular cells of the microvasculature, are compromised within multiple sclerosis lesions. Large-scale genetic and small-scale cell biology studies also suggest that neurovascular dysfunction could be a primary pathology contributing to multiple sclerosis development. Herein we revisit multiple sclerosis risk factors and multiple sclerosis pathophysiology and highlight the known and potential roles of neurovascular unit dysfunction in multiple sclerosis development and disease progression. We also evaluate the suitability of the neurovascular unit as a potential target for future disease modifying therapies for multiple sclerosis.
Collapse
Affiliation(s)
- Jake M Cashion
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia.
| |
Collapse
|
5
|
Attfield KE, Jensen LT, Kaufmann M, Friese MA, Fugger L. The immunology of multiple sclerosis. Nat Rev Immunol 2022; 22:734-750. [PMID: 35508809 DOI: 10.1038/s41577-022-00718-z] [Citation(s) in RCA: 168] [Impact Index Per Article: 56.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2022] [Indexed: 12/11/2022]
Abstract
Our incomplete understanding of the causes and pathways involved in the onset and progression of multiple sclerosis (MS) limits our ability to effectively treat this complex neurological disease. Recent studies explore the role of immune cells at different stages of MS and how they interact with cells of the central nervous system (CNS). The findings presented here begin to question the exclusivity of an antigen-specific cause and highlight how seemingly distinct immune cell types can share common functions that drive disease. Innovative techniques further expose new disease-associated immune cell populations and reinforce how environmental context is critical to their phenotype and subsequent role in disease. Importantly, the differentiation of immune cells into a pathogenic state is potentially reversible through therapeutic manipulation. As such, understanding the mechanisms that provide plasticity to causal cell types is likely key to uncoupling these disease processes and may identify novel therapeutic targets that replace the need for cell ablation.
Collapse
Affiliation(s)
- Kathrine E Attfield
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK
| | - Lise Torp Jensen
- Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Max Kaufmann
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institut für Neuroimmunologie und Multiple Sklerose, Zentrum für Molekulare Neurobiologie Hamburg, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Lars Fugger
- Oxford Centre for Neuroinflammation, Nuffield Department of Clinical Neurosciences, Oxford University Hospitals, University of Oxford, Oxford, UK.
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
6
|
Camacho-Toledano C, Machín-Díaz I, Calahorra L, Cabañas-Cotillas M, Otaegui D, Castillo-Triviño T, Villar LM, Costa-Frossard L, Comabella M, Midaglia L, García-Domínguez JM, García-Arocha J, Ortega MC, Clemente D. Peripheral myeloid-derived suppressor cells are good biomarkers of the efficacy of fingolimod in multiple sclerosis. J Neuroinflammation 2022; 19:277. [PMCID: PMC9675277 DOI: 10.1186/s12974-022-02635-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/30/2022] [Indexed: 11/21/2022] Open
Abstract
Background The increasing number of treatments that are now available to manage patients with multiple sclerosis (MS) highlights the need to develop biomarkers that can be used within the framework of individualized medicine. Fingolimod is a disease-modifying treatment that belongs to the sphingosine-1-phosphate receptor modulators. In addition to inhibiting T cell egress from lymph nodes, fingolimod promotes the immunosuppressive activity of myeloid-derived suppressor cells (MDSCs), whose monocytic subset (M-MDSCs) can be used as a biomarker of disease severity, as well as the degree of demyelination and extent of axonal damage in the experimental autoimmune encephalomyelitis (EAE) model of MS. In the present study, we have assessed whether the abundance of circulating M-MDSCs may represent a useful biomarker of fingolimod efficacy in EAE and in the clinical context of MS patients. Methods Treatment with vehicle or fingolimod was orally administered to EAE mice for 14 days in an individualized manner, starting the day when each mouse began to develop clinical signs. Peripheral blood from EAE mice was collected previous to treatment and human peripheral blood mononuclear cells (PBMCs) were collected from fingolimod to treat MS patients’ peripheral blood. In both cases, M-MDSCs abundance was analyzed by flow cytometry and its relationship with the future clinical affectation of each individual animal or patient was assessed. Results Fingolimod-treated animals presented a milder EAE course with less demyelination and axonal damage, although a few animals did not respond well to treatment and they invariably had fewer M-MDSCs prior to initiating the treatment. Remarkably, M-MDSC abundance was also found to be an important and specific parameter to distinguish EAE mice prone to better fingolimod efficacy. Finally, in a translational effort, M-MDSCs were quantified in MS patients at baseline and correlated with different clinical parameters after 12 months of fingolimod treatment. M-MDSCs at baseline were highly representative of a good therapeutic response to fingolimod, i.e., patients who met at least two of the criteria used to define non-evidence of disease activity-3 (NEDA-3) 12 months after treatment. Conclusion Our data indicate that M-MDSCs might be a useful predictive biomarker of the response of MS patients to fingolimod. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-022-02635-3.
Collapse
Affiliation(s)
- Celia Camacho-Toledano
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Isabel Machín-Díaz
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Leticia Calahorra
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - María Cabañas-Cotillas
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - David Otaegui
- grid.432380.eMultiple Sclerosis Unit, Biodonostia Health Institute, 20014 Donostia-San Sebastián, Spain
| | - Tamara Castillo-Triviño
- grid.432380.eMultiple Sclerosis Unit, Biodonostia Health Institute, 20014 Donostia-San Sebastián, Spain ,grid.414651.30000 0000 9920 5292Neurology Department, Hospital Universitario Donostia, San Sebastián, Spain
| | - Luisa María Villar
- grid.411347.40000 0000 9248 5770Immunology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Lucienne Costa-Frossard
- grid.411347.40000 0000 9248 5770Immunology Department, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain ,grid.411347.40000 0000 9248 5770Multiple Sclerosis Unit, Neurology, Ramón y Cajal University Hospital, Madrid, Spain
| | - Manuel Comabella
- grid.411083.f0000 0001 0675 8654Neurology-Neuroimmunology Service, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Luciana Midaglia
- grid.411083.f0000 0001 0675 8654Neurology-Neuroimmunology Service, Centre d’Esclerosi Múltiple de Catalunya (Cemcat), Institut de Recerca Vall d’Hebron, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - José Manuel García-Domínguez
- grid.410526.40000 0001 0277 7938Multiple Sclerosis Unit, Department of Neurology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Jennifer García-Arocha
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - María Cristina Ortega
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| | - Diego Clemente
- grid.414883.20000 0004 1767 1847Neuroimmuno-Repair Group, Hospital Nacional de Parapléjicos-SESCAM, Finca La Peraleda s/n, 45071 Toledo, Spain
| |
Collapse
|
7
|
Ansari MA, Nadeem A, Attia SM, Bakheet SA, Shahid M, Rehman MU, Alanazi MM, Alhamed AS, Ibrahim KE, Albekairi NA, Ahmad SF. CCR1 antagonist J-113863 corrects the imbalance of pro- and anti-inflammatory cytokines in a SJL/J mouse model of relapsing-remitting multiple sclerosis. Immunobiology 2022; 227:152245. [PMID: 35868215 DOI: 10.1016/j.imbio.2022.152245] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2021] [Revised: 06/20/2022] [Accepted: 07/06/2022] [Indexed: 11/17/2022]
Abstract
Multiple sclerosis (MS), an immune-mediated and neurodegenerative disorder of the central nervous system (CNS), is characterized by infiltrating myelin-reactive T lymphocytes and demyelinating lesions. Experimental autoimmune encephalomyelitis (EAE) is a well-established animal model used to study MS. To explore the impact of chemokine receptor CCR1 blockade in EAE and the underlying mechanisms, we used CCR1 antagonist J-113863 in PLP139-151-induced EAE in SJL/J mice. Following EAE induction, mice were treated with J-113863 (10 mg/kg) daily from day 14 until day 25. We investigated the effect of J-113863 on expression levels of GM-CSF, IL-6, IL-10, IL-27 in CD4+ spleen cells, using flow cytometry. We also analyzed the effect of J-113863 on GM-CSF, IL-6, IL-10, IL-27 mRNA and protein expression levels using RT-PCR and Western blot analysis in brain tissues. J-113863 treatment decreased the populations of CD4+GM-CSF+ and CD4+IL-6+ cells and increased CD4+IL-27+ and CD4+IL-10+ cells in the spleen. J-113863 had a suppressive effect on the mRNA and protein expression levels of GM-CSF, and IL-6 in the brain tissue. On the other hand, J-113863 treatment increased the mRNA and protein expression of IL-10 and IL-27 in the brain tissue. Our results highlighted J-113863's potential role in suppressing pro-inflammatory expression and up-regulating anti-inflammatory mediators, which could represent a beneficial alternative approach to MS treatment.
Collapse
Affiliation(s)
- Mushtaq A Ansari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sabry M Attia
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Saleh A Bakheet
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mudassar Shahid
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Muneeb U Rehman
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M Alanazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah S Alhamed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khalid E Ibrahim
- Department of Zoology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Norah A Albekairi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia.
| |
Collapse
|
8
|
Abstract
The CNS vasculature tightly regulates the passage of circulating molecules and leukocytes into the CNS. In the neuroinflammatory disease multiple sclerosis (MS), these regulatory mechanisms fail, and autoreactive T cells invade the CNS via blood vessels, leading to neurological deficits depending on where the lesions are located. The region-specific mechanisms directing the development of such lesions are not well understood. In this study, we investigated whether pericytes regulate CNS endothelial cell permissiveness toward leukocyte trafficking into the brain parenchyma. By using a pericyte-deficient mouse model, we show that intrinsic changes in the brain vasculature due to absence of pericytes facilitate the neuroinflammatory cascade and can influence the localization of the neuroinflammatory lesions. Pericytes regulate the development of organ-specific characteristics of the brain vasculature such as the blood–brain barrier (BBB) and astrocytic end-feet. Whether pericytes are involved in the control of leukocyte trafficking in the adult central nervous system (CNS), a process tightly regulated by CNS vasculature, remains elusive. Using adult pericyte-deficient mice (Pdgfbret/ret), we show that pericytes limit leukocyte infiltration into the CNS during homeostasis and autoimmune neuroinflammation. The permissiveness of the vasculature toward leukocyte trafficking in Pdgfbret/ret mice inversely correlates with vessel pericyte coverage. Upon induction of experimental autoimmune encephalomyelitis (EAE), pericyte-deficient mice die of severe atypical EAE, which can be reversed with fingolimod, indicating that the mortality is due to the massive influx of immune cells into the brain. Additionally, administration of anti-VCAM-1 and anti–ICAM-1 antibodies reduces leukocyte infiltration and diminishes the severity of atypical EAE symptoms of Pdgfbret/ret mice, indicating that the proinflammatory endothelium due to absence of pericytes facilitates exaggerated neuroinflammation. Furthermore, we show that the presence of myelin peptide-specific peripheral T cells in Pdgfbret/ret;2D2tg mice leads to the development of spontaneous neurological symptoms paralleled by the massive influx of leukocytes into the brain. These findings indicate that intrinsic changes within brain vasculature can promote the development of a neuroinflammatory disorder.
Collapse
|
9
|
Fani Maleki A, Cisbani G, Laflamme N, Prefontaine P, Plante MM, Baillargeon J, Rangachari M, Gosselin J, Rivest S. Selective Immunomodulatory and Neuroprotective Effects of a NOD2 Receptor Agonist on Mouse Models of Multiple Sclerosis. Neurotherapeutics 2021; 18:889-904. [PMID: 33479802 PMCID: PMC8423880 DOI: 10.1007/s13311-020-00998-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/18/2020] [Indexed: 12/13/2022] Open
Abstract
The significance of monocytes has been demonstrated in multiple sclerosis (MS). One of the therapeutic challenges is developing medications that induce mild immunomodulation that is solely targeting specific monocyte subsets without affecting microglia. Muramyl dipeptide (MDP) activates the NOD2 receptor, and systemic MDP administrations convert Ly6Chigh into Ly6Clow monocytes. Here, we report selective immunomodulatory and therapeutic effects of MDP on cuprizone and experimental autoimmune encephalomyelitis (EAE) mouse models of MS. MDP treatment exerted various therapeutic effects in EAE, including delaying EAE onset and reducing infiltration of leukocytes into the CNS before EAE onset. Of great interest is the robust beneficial effect of the MDP treatment in mice already developing the disease several days after EAE onset. Finally, we found that the NOD2 receptor plays a critical role in MDP-mediated EAE resistance. Our results demonstrate that MDP is beneficial in both early and progressive phases of EAE. Based on these results, and upon comprehensive basic and clinical research, we anticipate developing NOD2 agonist-based medications for MS in the future.
Collapse
MESH Headings
- Acetylmuramyl-Alanyl-Isoglutamine/pharmacology
- Acetylmuramyl-Alanyl-Isoglutamine/therapeutic use
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/therapeutic use
- Animals
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Freund's Adjuvant/toxicity
- Immunomodulating Agents/pharmacology
- Immunomodulating Agents/therapeutic use
- Male
- Mice
- Mice, Inbred C57BL
- Monocytes/drug effects
- Monocytes/immunology
- Multiple Sclerosis/chemically induced
- Multiple Sclerosis/immunology
- Multiple Sclerosis/prevention & control
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Nod2 Signaling Adaptor Protein/agonists
- Peptide Fragments/toxicity
Collapse
Affiliation(s)
- Adham Fani Maleki
- Neuroscience Laboratory, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada
| | - Giulia Cisbani
- Neuroscience Laboratory, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada
| | - Nataly Laflamme
- Neuroscience Laboratory, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada
| | - Paul Prefontaine
- Neuroscience Laboratory, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada
| | - Marie-Michele Plante
- Neuroscience Laboratory, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada
| | - Joanie Baillargeon
- Neuroscience Laboratory, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada
| | - Manu Rangachari
- Neuroscience Laboratory, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada
| | - Jean Gosselin
- Laboratory of Innate Immunity, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU of Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, 2705 Laurier Boul., Quebec City, QC, G1V 4G2, Canada.
| |
Collapse
|
10
|
Li Z, Chen J, Lei L, Jiang N, Zhu Y, Jia Y, Zhuo Y, Su W. Laquinimod Inhibits Inflammation-Induced Angiogenesis in the Cornea. Front Med (Lausanne) 2020; 7:598056. [PMID: 33244468 PMCID: PMC7683777 DOI: 10.3389/fmed.2020.598056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 10/13/2020] [Indexed: 12/02/2022] Open
Abstract
Background: Inflammation-induced angiogenesis plays a critical role in many eye diseases, and abnormal angiogenesis inhibition is regarded as a therapeutic approach. Here, we examined the effects of laquinimod on inflammatory corneal angiogenesis. Methods: Mouse model of corneal neovascularization was induced by NaOH. Laquinimod or control vehicle were topically applied to alkali-treated eyes twice a day for 10 days. Corneal neovascularization, infiltrating inflammatory cells, and the levels of chemokines, pro-inflammatory cytokines were assessed. RAW cells and human umbilical vein endothelial cells were used in vitro to further explore the underlying mechanisms of the effects of laquinimod on inflammation-induced angiogenesis. Results: Topical administration of laquinimod to the injured corneas dramatically inhibited alkali-induced corneal neovascularization and decreased inflammatory cell (such as macrophage) infiltration in a corneal injury mouse model. Laquinimod significantly downregulated the expression of chemokines (monocyte chemotactic protein-1 and macrophage inflammatory protein-1), pro-inflammatory cytokines (interleukin-1β and tumor necrosis factor-alpha), vascular endothelial growth factor, nucleotide-binding oligomerization domain-like receptor family pyrin domain-containing 3 and apoptosis-associated speck-like protein containing C-terminal caspase-recruitment domain adaptor protein in both injured corneas and RAW cells. In vitro, laquinimod also dramatically inhibited the proliferation, migration and tube formation of human umbilical vein endothelial cells. Conclusion: Laquinimod inhibits inflammation-induced angiogenesis in the cornea. These results suggest that laquinimod is a potential new therapeutic option for corneal neovascularization and other angiogenesis-associated diseases.
Collapse
Affiliation(s)
- Zuohong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Jianping Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Lei Lei
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Nan Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.,Department of Pediatric Ophthalmology, Guangzhou Children's Hospital and Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Yanling Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yu Jia
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Colombo E, Pascente R, Triolo D, Bassani C, De Angelis A, Ruffini F, Ottoboni L, Comi G, Martino G, Farina C. Laquinimod Modulates Human Astrocyte Function and Dampens Astrocyte-Induced Neurotoxicity during Inflammation. Molecules 2020; 25:E5403. [PMID: 33218208 PMCID: PMC7699283 DOI: 10.3390/molecules25225403] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/06/2020] [Accepted: 11/16/2020] [Indexed: 11/18/2022] Open
Abstract
Astrocytes greatly participate to inflammatory and neurotoxic reactions occurring in neurodegenerative diseases and are valuable pharmacological targets to support neuroprotection. Here we used human astrocytes generated from reprogrammed fibroblasts as a cellular model to study the effect of the compound Laquinimod and its active metabolite de-Laquinimod on astrocyte functions and the astrocyte-neuron interaction. We show that human iAstrocytes expressed the receptor for the inflammatory mediator IL1 and responded to it via nuclear translocation of NFκB, an event that did not occur if cells were treated with Laquinimod, indicating a direct anti-inflammatory activity of the drug on the human astrocyte. Similarly, while exposure to IL1 downregulated glial glutamate transporters GLAST and GLT1, treatment with Laquinimod supported maintenance of physiological levels of these proteins despite the inflammatory milieu. Laquinimod also induced nuclear translocation of the aryl hydrocarbon receptor (AHR), suggesting that drug action was mediated by activation of the AHR pathway. However, the drug was effective despite AHR inhibition via CH223191, indicating that AHR signaling in the astrocyte is dispensable for drug responses. Finally, in vitro experiments with rat spinal neurons showed that laquinimod did not exert neuroprotection directly on the neuron but dampened astrocyte-induced neurodegeneration. Our findings indicate that fibroblast-derived human astrocytes represent a suitable model to study astrocyte-neuron crosstalk and demonstrate indirect, partial neuroprotective efficacy for laquinimod.
Collapse
Affiliation(s)
- Emanuela Colombo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| | - Rosaria Pascente
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| | - Daniela Triolo
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| | - Claudia Bassani
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| | - Anthea De Angelis
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| | - Francesca Ruffini
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| | - Linda Ottoboni
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| | - Giancarlo Comi
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| | - Gianvito Martino
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
- Faculty of Medicine and Surgery, Vita-Salute San Raffaele University, 20132 Milan, Italy
| | - Cinthia Farina
- Institute of Experimental Neurology (INSpe), Division of Neuroscience, IRCCS San Raffaele Hospital, 20132 Milan, Italy; (E.C.); (R.P.); (D.T.); (C.B.); (A.D.A.); (F.R.); (L.O.); (G.C.); (G.M.)
| |
Collapse
|
12
|
Engel S, Jolivel V, Kraus SHP, Zayoud M, Rosenfeld K, Tumani H, Furlan R, Kurschus FC, Waisman A, Luessi F. Laquinimod dampens IL-1β signaling and Th17-polarizing capacity of monocytes in patients with MS. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 8:8/1/e908. [PMID: 33203651 PMCID: PMC7676421 DOI: 10.1212/nxi.0000000000000908] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 09/23/2020] [Indexed: 01/16/2023]
Abstract
OBJECTIVE To assess the impact of laquinimod treatment on monocytes and to investigate the underlying immunomodulatory mechanisms in MS. METHODS In this cross-sectional study, we performed in vivo and in vitro analyses of cluster of differentiation (CD14+) monocytes isolated from healthy donors (n = 15), untreated (n = 13), and laquinimod-treated patients with MS (n = 14). Their frequency and the expression of surface activation markers were assessed by flow cytometry and the viability by calcein staining. Cytokine concentrations in the supernatants of lipopolysaccharide (LPS)-stimulated monocytes were determined by flow cytometry. The messenger ribonucleic acid (mRNA) expression level of genes involved in cytokine expression was measured by quantitative PCR. The LPS-mediated nuclear factor kappa-light-chain-enhancer of activated B-cell (NF-κB) activation was determined by the quantification of the phosphorylation level of the p65 subunit. Laquinimod-treated monocytes were cocultured with CD4+ T cells, and the resulting cytokine production was analyzed by flow cytometry after intracellular cytokine staining. The interleukin (IL)-17A concentration of the supernatant was assessed by ELISA. RESULTS Laquinimod did not alter the frequency or viability of circulating monocytes, but led to an upregulation of CD86 expression. LPS-stimulated monocytes of laquinimod-treated patients with MS secreted less IL-1β following a downregulation of IL-1β gene expression. Phosphorylation levels of the NF-κB p65 subunit were reduced after laquinimod treatment, indicating a laquinimod-associated inhibition of the NF-κB pathway. T cells primed with laquinimod-treated monocytes differentiated significantly less into IL-17A-producing T helper (Th)-17 cells. CONCLUSIONS Our findings suggest that inhibited NF-κB signaling and downregulation of IL-1β expression in monocytes contributes to the immunomodulatory effects of laquinimod and that the impairment of Th17 polarization might mediate its disease-modifying activity in MS.
Collapse
Affiliation(s)
- Sinah Engel
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Valérie Jolivel
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Stefan H-P Kraus
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Morad Zayoud
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Karolina Rosenfeld
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Hayrettin Tumani
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Roberto Furlan
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Florian C Kurschus
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Ari Waisman
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Luessi
- From the Department of Neurology (S.E., V.J., S.H.-P.K., K.R., F.L.), Focus Program Translational Neuroscience (FTN), University Medical Center of the Johannes Gutenberg University of Mainz, Germany; Biopathology of Myelin (V.J.), Neuroprotection and Therapeutic Strategy, INSERM U1119, Fédération de Médecine Translationnelle de Strasbourg (FMTS), University of Strasbourg, France; Institute for Molecular Medicine (M.Z., F.C.K., A.W.), University Medical Centre of the Johannes Gutenberg University of Mainz, Germany; Sheba Cancer Research Center (M.Z.), Chaim Sheba Academic Medical Center, Ramat Gan, Israel; Department of Neurology (H.T.), University of Ulm, Germany and Specialty Clinic of Neurology Dietenbronn, Schwendi, Germany; Clinical Neuroimmunology Unit (R.F.), San Raffaele Scientific Institute, Milan, Italy; and Department of Dermatology (F.C.K.), Heidelberg University Hospital, Heidelberg, Germany.
| |
Collapse
|
13
|
Jiang N, Li Z, Li Z, Zhang Y, Yu Z, Wan P, Zhu Y, Li Y, Su W, Zhuo Y. Laquinimod exerts anti-inflammatory and antiapoptotic effects in retinal ischemia/reperfusion injury. Int Immunopharmacol 2020; 88:106989. [PMID: 33182069 DOI: 10.1016/j.intimp.2020.106989] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 08/22/2020] [Accepted: 09/07/2020] [Indexed: 11/15/2022]
Abstract
Retinal ischemia/reperfusion (I/R) occurs in various vision disabled ocular diseases, involved in acute glaucoma, diabetic retinopathy, ischemic optic neuropathy, hypertensive retinopathy and retinal vascular occlusion. Laquinimod (LQ), a new type of immunosuppressant, has been reported to exert anti-inflammatory effects on autoimmune diseases. This research aims to investigate the protective effect of LQ on I/R damage by focusing on inhibiting dysregulated neuroinflammation and neuronal apoptosis. In our study, mice were treated with LQ after high intraocular pressure (IOP)-induced retinal I/R injury. The data showed that LQ significantly attenuated high IOP-induced retinal ganglion cell (RGC) death and inner plexiform layer (IPL) thinning and inhibited microglial activation. The results of qRT-PCR, flow cytometry and Luminex multiplex assays demonstrated the anti-inflammatory action of LQ in BV2 cells stimulated with lipopolysaccharide (LPS). In addition, primary RGC apoptosis induced by oxygen-glucose deprivation/reperfusion (OGD/R) was also directly suppressed by LQ. Importantly, LQ inhibited the expression of cleaved caspase-8 and the downstream NLRP3 inflammasome and IL-1β. In conclusion, our findings offer the first evidence that LQ treatment prevents retinal I/R damage. Furthermore, LQ could directly inhibit RGC apoptosis. Caspase-8 activation and subsequent inflammation can also be suppressed by LQ, which suggests that LQ may act through inhibiting the caspase-8 pathway. This study demonstrates a new mechanism of LQ and provides beneficial preclinical data for the clinical application of LQ.
Collapse
Affiliation(s)
- Nan Jiang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zhidong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Zuohong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yingying Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Ziyu Yu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Peixing Wan
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yingting Zhu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
14
|
Hou X, Chen G, Bracamonte-Baran W, Choi HS, Diny NL, Sung J, Hughes D, Won T, Wood MK, Talor MV, Hackam DJ, Klingel K, Davogustto G, Taegtmeyer H, Coppens I, Barin JG, Čiháková D. The Cardiac Microenvironment Instructs Divergent Monocyte Fates and Functions in Myocarditis. Cell Rep 2020; 28:172-189.e7. [PMID: 31269438 PMCID: PMC6813836 DOI: 10.1016/j.celrep.2019.06.007] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 05/07/2019] [Accepted: 06/03/2019] [Indexed: 12/24/2022] Open
Abstract
Two types of monocytes, Ly6Chi and Ly6Clo, infiltrate the heart in murine experimental autoimmune myocarditis (EAM). We discovered a role for cardiac fibroblasts in facilitating monocyte-to-macrophage differentiation of both Ly6Chi and Ly6Clo cells, allowing these macrophages to perform divergent functions in myocarditis progression. During the acute phase of EAM, IL-17A is highly abundant. It signals through cardiac fibroblasts to attenuate efferocytosis of Ly6Chi monocyte-derived macrophages (MDMs) and simultaneously prevents Ly6Clo monocyte-to-macrophage differentiation. We demonstrated an inverse clinical correlation between heart IL-17A levels and efferocytic receptor expressions in humans with heart failure (HF). In the absence of IL-17A signaling, Ly6Chi MDMs act as robust phagocytes and are less proinflammatory, whereas Ly6Clo monocytes resume their differentiation into MHCII+ macrophages. We propose that MHCII+Ly6Clo MDMs are associated with the reduction of cardiac fibrosis and prevention of the myocarditis sequalae. Hou et al. show that cardiac fibroblasts facilitate infiltrating Ly6Chi and Ly6Clo monocytes to become macrophages. IL-17A trans-signaling through cardiac fibroblasts increases MerTK shedding and promotes a pro-inflammatory and pro-tissue remodeling gene expression profile in Ly6Chi monocyte-derived macrophages. Paradoxically, IL-17A signaling through cardiac fibroblasts can substantially inhibit Ly6Clo monocyte-to-macrophage differentiation.
Collapse
Affiliation(s)
- Xuezhou Hou
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Guobao Chen
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | | | - Hee Sun Choi
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Nicola L Diny
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jungeun Sung
- Institute of Genetic Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - David Hughes
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Taejoon Won
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Megan Kay Wood
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Monica V Talor
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - David Joel Hackam
- Division of General Pediatric Surgery, Johns Hopkins University and Bloomberg Children's Center, Johns Hopkins Hospital, Baltimore, MD 21218, USA
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University of Tübingen, 72076 Tübingen, Germany
| | - Giovanni Davogustto
- Department of Internal Medicine, Division of Cardiovascular Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Heinrich Taegtmeyer
- Department of Internal Medicine, Division of Cardiovascular Medicine, McGovern Medical School at The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Isabelle Coppens
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jobert G Barin
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| |
Collapse
|
15
|
Hemmati S, Sadeghi MA, Yousefi-Manesh H, Eslamiyeh M, Vafaei A, Foroutani L, Donyadideh G, Dehpour A, Rezaei N. Protective Effects of Leukadherin1 in a Rat Model of Targeted Experimental Autoimmune Encephalomyelitis (EAE): Possible Role of P47phox and MDA Downregulation. J Inflamm Res 2020; 13:411-420. [PMID: 32821147 PMCID: PMC7423460 DOI: 10.2147/jir.s258991] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 07/22/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Reactive oxygen and nitrogen species (ROS and RNS) are involved in pathologic mechanisms underlying demyelination and exacerbation in multiple sclerosis (MS) lesions. P47phox is the most important subunit of an ROS-producing enzyme (NADPH oxidase) which is reportedly upregulated in MS plaques due to the intense activity of infiltrated immune cells and resident microglia. Leukadherin1 is a specific CD11b/CD18 agonist that inhibits signaling and transmigration of inflammatory cells to sites of injury. Based on this mechanism, we evaluated therapeutic effects of leukadherin1 in an animal model of targeted experimental autoimmune encephalomyelitis (EAE) through focal injection of inflammatory cytokines to the spinal cord. METHODS For model induction, Lewis rats were first immunized with 15µg MOG 1-125 emulsion. Twenty days later, animals were subjected to stereotaxic injection of IFNγ and TNFα to the specific spinal area (T8). One day after injection, all animals presented EAE clinical signs, and their behaviors were monitored for eight days through open-field locomotion and grid-walking tests. Leukadherin1-treated animals received daily intraperitoneal injections of 1mg/kg of the drug. The specific spinal tissues were extracted on day 5 in order to measure nitric oxide (NO), malon di-aldehyde (MDA), and TNFα concentrations alongside P47phox real-time PCR analysis. In addition, spinal sections were prepared for immunohistochemical (IHC) observation of infiltrated leukocytes and activated microglia. RESULTS Leukadherin1 exhibited promising improvements in EAE clinical scores and behavioral tests. Demyelination, CD45+ leukocyte infiltration, and Iba1+ microglia activation were reduced in spinal tissues of leukadherin1-treated animals. Furthermore, P47phox expression levels, MDA, and NO amounts were decreased in treated animals. However, TNFα concentrations did not differ following treatment. CONCLUSION Based on our results, we suggest that leukadherin1 may be used as a novel therapeutic agent in tackling the clinical challenge of multiple sclerosis, especially during the acute phase of the disease. This effect was possibly mediated through decreased leukocyte infiltration and oxidative stress.
Collapse
Affiliation(s)
- Sara Hemmati
- Molecular Medicine Interest Group (MMIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Sadeghi
- Molecular Medicine Interest Group (MMIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hasan Yousefi-Manesh
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Ali Vafaei
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Laleh Foroutani
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - AhmadReza Dehpour
- Experimental Medicine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children’s Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| |
Collapse
|
16
|
Lu L, Qi S, Chen Y, Luo H, Huang S, Yu X, Luo Q, Zhang Z. Targeted immunomodulation of inflammatory monocytes across the blood-brain barrier by curcumin-loaded nanoparticles delays the progression of experimental autoimmune encephalomyelitis. Biomaterials 2020; 245:119987. [DOI: 10.1016/j.biomaterials.2020.119987] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 03/16/2020] [Accepted: 03/18/2020] [Indexed: 02/08/2023]
|
17
|
Biernacki T, Sandi D, Bencsik K, Vécsei L. Kynurenines in the Pathogenesis of Multiple Sclerosis: Therapeutic Perspectives. Cells 2020; 9:cells9061564. [PMID: 32604956 PMCID: PMC7349747 DOI: 10.3390/cells9061564] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/11/2022] Open
Abstract
Over the past years, an increasing amount of evidence has emerged in support of the kynurenine pathway’s (KP) pivotal role in the pathogenesis of several neurodegenerative, psychiatric, vascular and autoimmune diseases. Different neuroactive metabolites of the KP are known to exert opposite effects on neurons, some being neuroprotective (e.g., picolinic acid, kynurenic acid, and the cofactor nicotinamide adenine dinucleotide), while others are toxic to neurons (e.g., 3-hydroxykynurenine, quinolinic acid). Not only the alterations in the levels of the metabolites but also disturbances in their ratio (quinolinic acid/kynurenic acid) have been reported in several diseases. In addition to the metabolites, the enzymes participating in the KP have been unearthed to be involved in modulation of the immune system, the energetic upkeep of neurons and have been shown to influence redox processes and inflammatory cascades, revealing a sophisticated, intertwined system. This review considers various methods through which enzymes and metabolites of the kynurenine pathway influence the immune system, the roles they play in the pathogenesis of neuroinflammatory diseases based on current evidence with a focus on their involvement in multiple sclerosis, as well as therapeutic approaches.
Collapse
Affiliation(s)
- Tamás Biernacki
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - Dániel Sandi
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - Krisztina Bencsik
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
| | - László Vécsei
- Department of Neurology, Faculty of General Medicine, Albert Szent-Györgyi Clinical Centre, University of Szeged, H-6725 Szeged, Hungary; (T.B.); (D.S.); (K.B.)
- MTA—SZTE Neuroscience Research Group, H-6725 Szeged, Hungary
- Interdisciplinary Excellence Center, University of Szeged, H-6720 Szeged, Hungary
- Correspondence: ; Tel.: +36-62-545-356; Fax: +36-62-545-597
| |
Collapse
|
18
|
Jiang T, Zhang W, Wang Z. Laquinimod Protects Against TNF-α-Induced Attachment of Monocytes to Human Aortic Endothelial Cells (HAECs) by Increasing the Expression of KLF2. DRUG DESIGN DEVELOPMENT AND THERAPY 2020; 14:1683-1691. [PMID: 32440094 PMCID: PMC7222522 DOI: 10.2147/dddt.s243666] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 02/06/2020] [Indexed: 01/01/2023]
Abstract
Introduction As a worldwide health issue, the treatment and prevention of atherosclerosis present an important goal. Increased levels of proinflammatory cytokines such as TNF-α-associated chronic inflammatory response cause endothelial cells to lose their ability to regulate vascular function. Lipid-laden immune cells are recruited to the endothelium where they adhere to the endothelial wall and invade the intimal space, thereby leading to the development of atherosclerotic lesions, fatty plaques, and thickening of the arterial wall. In the present study, for the first time, we investigated the effects of laquinimod, an immunomodulatory agent used for the treatment of multiple sclerosis, on human aortic endothelial in a TNF-α-induced atherosclerotic microenvironment. At present, the mechanism of action of laquinimod is not well defined. Methods The effects of laquinimod on the gene expression of IL-6, MCP-1, VCAM-1, E-selectin, and KLF2 were measured by real-time PCR. ELISA assay was used to determine protein secretion and expression. Phosphorylation of ERK5 and the protein level of KLF2 were measured by Western blot analysis. The attachment of monocytes to endothelial cells was assayed by calcein-AM staining and fluorescent microscopy. Results Our findings demonstrate that laquinimod reduced the expression of key inflammatory cytokines and chemokines, including IL-6, MCP-1, and HMGB1. We further demonstrate that laquinimod significantly reduced the attachment of monocytes to endothelial cells, which is mediated through reduced expression of the cellular adhesion molecules VCAM-1 and E-selectin. Here, we found that laquinimod could significantly increase the expression of KLF2 through activation of ERK5 signaling. The results of our KLF2 knockdown experiment confirm that the effects of laquinimod observed in vitro are dependent on KLF2 expression. Conclusion Together, these findings suggest a potential antiatherosclerotic capacity of laquinimod. Further research will elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Tiechao Jiang
- Department of Cardiovascular Medicine, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China.,Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China.,Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Wenhao Zhang
- Department of Cardiovascular Medicine, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China
| | - Zhongyu Wang
- Department of Cardiovascular Medicine, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China.,Jilin Provincial Precision Medicine Key Laboratory for Cardiovascular Genetic Diagnosis, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China.,Jilin Provincial Engineering Laboratory for Endothelial Function and Genetic Diagnosis of Cardiovascular Disease, The Third Hospital of Jilin University, Changchun 130033, People's Republic of China
| |
Collapse
|
19
|
Xie P, Dan F, Yu G, Ruan W, Yu H. Laquinimod Mitigated IL-1β-Induced Impairment of the Cartilage Extracellular Matrix in Human ATDC5 Chondrocytes. Chem Res Toxicol 2020; 33:933-939. [PMID: 32191437 DOI: 10.1021/acs.chemrestox.9b00482] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
To date, a safe and reliable treatment of osteoarthritis (OA) has not yet been announced. Inflammatory response and degradation of the articular extracellular matrix (ECM) induced by IL-1β are important pathological characteristics of OA. Laquinimod is a quinoline-3-carboxamide and a novel oral immunomodulatory compound in clinical use. However, whether laquinimod has a beneficial effect in OA is not known. In our research, we found that laquinimod could ameliorate IL-1β-induced generation of ROS and improve mitochondrial function by increasing mitochondrial membrane potential (ΔΨm). Furthermore, treatment with laquinimod suppressed IL-1β-induced production of TNF-α and IL-6. Notably, laquinimod prevented the degradation of type II collagen by inhibiting MMP-3 and MMP-13. Meanwhile, the presence of laquinimod attenuated the reduction in aggrecan by mediating ADAMTS-4 and ADAMTS-5. Mechanistically, laquinimod ameliorated IL-1β-induced inflammation and degeneration of ECM by suppressing the activation of NF-κB. Taken together, our findings reveal that laquinimod possesses a beneficial effect against IL-1β insults in human chondrocytes, implying an important role of laquinimod in OA.
Collapse
Affiliation(s)
- Peng Xie
- Department of Bone and Joint Trauma, Hanzhong Central Hospital, Shaanxi Province 723000, China
| | - Feng Dan
- Department of Bone and Joint Trauma, Hanzhong Central Hospital, Shaanxi Province 723000, China
| | - Guoyong Yu
- Department of Bone and Joint Trauma, Hanzhong Central Hospital, Shaanxi Province 723000, China
| | - Wenhui Ruan
- Department of Bone and Joint Trauma, Hanzhong Central Hospital, Shaanxi Province 723000, China
| | - Hong Yu
- Department of Bone and Joint Trauma, Hanzhong Central Hospital, Shaanxi Province 723000, China
| |
Collapse
|
20
|
Machado CA, Silva ACSE, de Miranda AS, Cordeiro TME, Ferreira RN, de Souza LC, Teixeira AL, de Miranda AS. Immune-Based Therapies for Traumatic Brain Injury: Insights from Pre-Clinical Studies. Curr Med Chem 2019; 27:5374-5402. [PMID: 31291871 DOI: 10.2174/0929867326666190710173234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/24/2019] [Accepted: 05/22/2019] [Indexed: 12/18/2022]
Abstract
Traumatic Brain Injury (TBI) is a major public health problem. It is the leading cause of death and disability, especially among children and young adults. The neurobiology basis underlying TBI pathophysiology remains to be fully revealed. Over the past years, emerging evidence has supported the hypothesis that TBI is an inflammatory based condition, paving the way for the development of potential therapeutic targets. There is no treatment capable to prevent or minimize TBIassociated outcomes. Therefore, the search for effective therapies is a priority goal. In this context, animal models have become valuable tools to study molecular and cellular mechanisms involved in TBI pathogenesis as well as novel treatments. Herein, we discuss therapeutic strategies to treat TBI focused on immunomodulatory and/or anti-inflammatory approaches in the pre-clinical setting.
Collapse
Affiliation(s)
- Caroline Amaral Machado
- Laboratorio de Neurobiologia, Departamento de Morfologia, Instituto de Ciencias Biologicas, UFMG, Brazil
| | - Ana Cristina Simões E Silva
- Laboratorio Interdisciplinar de Investigacao Medica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Amanda Silva de Miranda
- Departamento de Quimica, Instituto de Ciencias Exatas, Universidade Federal de Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Thiago Macedo E Cordeiro
- Laboratorio Interdisciplinar de Investigacao Medica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Rodrigo Novaes Ferreira
- Laboratorio de Neurobiologia, Departamento de Morfologia, Instituto de Ciencias Biologicas, UFMG, Brazil
| | - Leonardo Cruz de Souza
- Laboratorio Interdisciplinar de Investigacao Medica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| | - Antônio Lúcio Teixeira
- Neuropsychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, University of Texas Health Science Center, Houston, United States
| | - Aline Silva de Miranda
- Laboratorio Interdisciplinar de Investigacao Medica (LIIM), Faculdade de Medicina, Universidade Federal de Minas Gerais (UFMG), Brazil
| |
Collapse
|
21
|
Hirsutanol A Attenuates Lipopolysaccharide-Mediated Matrix Metalloproteinase 9 Expression and Cytokines Production and Improves Endotoxemia-Induced Acute Sickness Behavior and Acute Lung Injury. Mar Drugs 2019; 17:md17060360. [PMID: 31213027 PMCID: PMC6627105 DOI: 10.3390/md17060360] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 06/11/2019] [Accepted: 06/14/2019] [Indexed: 12/15/2022] Open
Abstract
Activated human monocytes/macrophages, which increase the levels of matrix metalloproteinases (MMPs) and pro-inflammatory cytokines, are the essential mechanisms for the progression of sepsis. In the present study, we determined the functions and mechanisms of hirsutanolA (HA), which is isolated from the red alga-derived marine fungus Chondrostereum sp. NTOU4196, on the production of pro-inflammatory mediators produced from lipopolysaccharide (LPS)-treated THP-1 cells. Our results showed that HA suppressed LPS-triggered MMP-9-mediated gelatinolysis and expression of protein and mRNA in a concentration-dependent manner without effects on TIMP-1 activity. Also, HA significantly attenuated the levels of TNF-α, IL-6, and IL-1β from LPS-treated THP-1 cells. Moreover, HA significantly inhibited LPS-mediated STAT3 (Tyr705) phosphorylation, IκBα degradation and ERK1/2 activation in THP-1 cells. In an LPS-induced endotoxemia mouse model, studies indicated that HA pretreatment improved endotoxemia-induced acute sickness behavior, including acute motor deficits and anxiety-like behavior. HA also attenuated LPS-induced phospho-STAT3 and pro-MMP-9 activity in the hippocampus. Notably, HA reduced pathologic lung injury features, including interstitial tissue edema, infiltration of inflammatory cells and alveolar collapse. Likewise, HA suppressed the induction of phospho-STAT3 and pro-MMP-9 in lung tissues. In conclusion, our results provide pharmacological evidence that HA could be a useful agent for treating inflammatory diseases, including sepsis.
Collapse
|
22
|
Abstract
Multiple sclerosis treatment faces tremendous changes owing to the approval of new medications, some of which are available as oral formulations. Until now, the four orally available medications, fingolimod, dimethylfumarate (BG-12), teriflunomide, and cladribine have received market authorization, whereas laquinimod is still under development. Fingolimod is a sphingosine-1-phosphate inhibitor, which is typically used as escalation therapy and leads to up to 60% reduction of the annualized relapse rate, but might also have neuroprotective properties. In addition, there are three more specific S1P agonists in late stages of development: siponimod, ponesimod, and ozanimod. Dimethylfumarate has immunomodulatory and cytoprotective functions and is used as baseline therapy. Teriflunomide, the active metabolite of the rheumatoid arthritis medication leflunomide, targets the dihydroorotate dehydrogenase, thus inhibiting the proliferation of lymphocytes by depletion of pyrimidines. Here we will review the mechanisms of action, clinical trial data, as well as data about safety and tolerability of the compounds.
Collapse
Affiliation(s)
- Simon Faissner
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| | - Ralf Gold
- Department of Neurology, St. Josef-Hospital, Ruhr-University Bochum, 44791 Bochum, Germany
| |
Collapse
|
23
|
Spiegelstein O, Mimrod D, Rabinovich L, Eyal E, Sprenger C, Elgart A, Samara E, Morganroth J. A Thorough QT/QTc Study With Laquinimod, a Novel Immunomodulator in Development for Multiple Sclerosis and Huntington Disease. Clin Pharmacol Drug Dev 2018; 8:49-59. [DOI: 10.1002/cpdd.442] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/20/2017] [Indexed: 11/08/2022]
Affiliation(s)
- Ofer Spiegelstein
- Clinical Pharmacology and Pharmacometrics; Teva Pharmaceutical Industries Ltd; Netanya Israel
| | - Dorit Mimrod
- Project Leadership; Teva Pharmaceutical Industries Ltd; Netanya Israel
| | - Laura Rabinovich
- Clinical Pharmacology and Pharmacometrics; Teva Pharmaceutical Industries Ltd; Frazer PA USA
| | - Eli Eyal
- Global Biostatistics Unit; TEVA Pharmaceutical Industries Ltd; Netanya Israel
| | | | - Anna Elgart
- Clinical Pharmacology and Pharmacometrics; Teva Pharmaceutical Industries Ltd; Netanya Israel
| | | | | |
Collapse
|
24
|
Tahvili S, Törngren M, Holmberg D, Leanderson T, Ivars F. Paquinimod prevents development of diabetes in the non-obese diabetic (NOD) mouse. PLoS One 2018; 13:e0196598. [PMID: 29742113 PMCID: PMC5942776 DOI: 10.1371/journal.pone.0196598] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
Quinoline-3-carboxamides (Q compounds) are immunomodulatory compounds that have shown efficacy both in autoimmune disease and cancer. We have in here investigated the impact of one such compound, paquinimod, on the development of diabetes in the NOD mouse model for type I diabetes (T1D). In cohorts of NOD mice treated with paquinimod between weeks 10 to 20 of age and followed up until 40 weeks of age, we observed dose-dependent reduction in incidence of disease as well as delayed onset of disease. Further, in contrast to untreated controls, the majority of NOD mice treated from 15 weeks of age did not develop diabetes at 30 weeks of age. Importantly, these mice displayed significantly less insulitis, which correlated with selectively reduced number of splenic macrophages and splenic Ly6Chi inflammatory monocytes at end point as compared to untreated controls. Collectively, these results demonstrate that paquinimod treatment can significantly inhibit progression of insulitis to T1D in the NOD mouse. We propose that the effect of paquinimod on disease progression may be related to the reduced number of these myeloid cell populations. Our finding also indicates that this compound could be a candidate for clinical development towards diabetes therapy in humans.
Collapse
Affiliation(s)
- Sahar Tahvili
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Dan Holmberg
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Tomas Leanderson
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
- Active Biotech AB, Lund, Sweden
| | - Fredrik Ivars
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
25
|
Katsumoto A, Miranda AS, Butovsky O, Teixeira AL, Ransohoff RM, Lamb BT. Laquinimod attenuates inflammation by modulating macrophage functions in traumatic brain injury mouse model. J Neuroinflammation 2018; 15:26. [PMID: 29382353 PMCID: PMC5791334 DOI: 10.1186/s12974-018-1075-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 01/22/2018] [Indexed: 12/11/2022] Open
Abstract
Background Traumatic brain injury (TBI) is a critical public health and socio-economic problem worldwide. A growing body of evidence supports the involvement of inflammatory events in TBI. It has been reported that resident microglia and infiltrating monocytes promote an inflammatory reaction that leads to neuronal death and eventually behavioral and cognitive impairment. Currently, there is no effective treatment for TBI and the development of new therapeutic strategies is a scientific goal of highest priority. Laquinimod, an orally administered neuroimmunomodulator initially developed for the treatment of multiple sclerosis, might be a promising neuroprotective therapy for TBI. Herein, we aim to investigate the hypothesis that laquinimod will reduce the central nervous system (CNS) damage caused by TBI. Methods To test our hypothesis, Ccr2rfp/+Cx3cr1gfp/+ mice were submitted to a moderate TBI induced by fluid percussion. Sham controls were submitted only to craniotomy. Mice were treated daily by oral gavage with laquinimod (25 mg/kg) 7 days before and 3 days after TBI. The brains of mice treated or not treated with laquinimod were collected at 3 and 120 days post injury, and brain morphological changes, axonal injury, and neurogenesis were evaluated by microscopy analysis. We also isolated microglia from infiltrating monocytes, and the expression of immune gene mRNAs were analyzed by employing a quantitative NanoString nCounter technique. Results Laquinimod prevented ventricle enlargement caused by TBI in the long term. Immunohistochemical analyses revealed decreased axonal damage and restored neurogenesis in the laquinimod-treated TBI group at early stage (3 days post injury). Notably, laquinimod inhibited the monocytes infiltration to the brain. Hierarchial clustering demonstrated that the microglial gene expression from the TBI group treated with laquinimod resembles the sham group more than the TBI-water control group. Conclusions Administration of laquinimod reduced lesion volume and axonal damage and restored neurogenesis after TBI. Laquinimod might be a potential therapy strategy to improve TBI long-term prognosis. Electronic supplementary material The online version of this article (10.1186/s12974-018-1075-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Atsuko Katsumoto
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA. .,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W 15th St, Indianapolis, IN, 46202, USA.
| | - Aline S Miranda
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA.,Laboratory of Neurobiology, Department of Morphology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Oleg Butovsky
- Center of Neurologic Diseases, Brigham and Women's Hospital, Boston, MA, USA
| | - Antônio L Teixeira
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Richard M Ransohoff
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Bruce T Lamb
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA. .,Stark Neurosciences Research Institute, Indiana University School of Medicine, 320 W 15th St, Indianapolis, IN, 46202, USA.
| |
Collapse
|
26
|
Acetylcholine-producing NK cells attenuate CNS inflammation via modulation of infiltrating monocytes/macrophages. Proc Natl Acad Sci U S A 2017; 114:E6202-E6211. [PMID: 28696300 DOI: 10.1073/pnas.1705491114] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The nonneural cholinergic system of immune cells is pivotal for the maintenance of immunological homeostasis. Here we demonstrate the expression of choline acetyltransferase (ChAT) and cholinergic enzymes in murine natural killer (NK) cells. The capacity for acetylcholine synthesis by NK cells increased markedly under inflammatory conditions such as experimental autoimmune encephalomyelitis (EAE), in which ChAT expression escalated along with the maturation of NK cells. ChAT+ and ChAT- NK cells displayed distinctive features in terms of cytotoxicity and chemokine/cytokine production. Transfer of ChAT+ NK cells into the cerebral ventricles of CX3CR1-/- mice reduced brain and spinal cord damage after EAE induction, and decreased the numbers of CNS-infiltrating CCR2+Ly6Chi monocytes. ChAT+ NK cells killed CCR2+Ly6Chi monocytes directly via the disruption of tolerance and inhibited the production of proinflammatory cytokines. Interestingly, ChAT+ NK cells and CCR2+Ly6Chi monocytes formed immune synapses; moreover, the impact of ChAT+ NK cells was mediated by α7-nicotinic acetylcholine receptors. Finally, the NK cell cholinergic system up-regulated in response to autoimmune activation in multiple sclerosis, perhaps reflecting the severity of disease. Therefore, this study extends our understanding of the nonneural cholinergic system and the protective immune effect of acetylcholine-producing NK cells in autoimmune diseases.
Collapse
|
27
|
Hussain RZ, Miller-Little WA, Lambracht-Washington D, Jaramillo TC, Takahashi M, Zhang S, Fu M, Cutter GR, Hayardeny L, Powell CM, Rosenberg RN, Stüve O. Laquinimod has no effects on brain volume or cellular CNS composition in the F1 3xTg-AD/C3H mouse model of Alzheimer's disease. J Neuroimmunol 2017; 309:100-110. [PMID: 28601278 DOI: 10.1016/j.jneuroim.2017.05.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Laquinimod is an anti-inflammatory agent with good central nervous system (CNS) bioavailability, and neuroprotective and myelorestorative properties. A clinical trial in patients with multiple sclerosis demonstrated that laquinimod significantly reduced loss of brain volume. The cellular substrate or molecular events underlying that treatment effect are unknown. In this study, we aimed to explore laquinimod's potential effects on brain volume, animal behavior, cellular numbers and composition of CNS-intrinsic cells and mononuclear cells within the CNS, amyloid beta (Aβ) accumulation and tau phosphorylation in the F1 3xTg-AD/C3H mouse model of Alzheimer's disease. METHODS Utilizing a dose response study design, four months old F1 3xTg-AD/C3H mice were treated for 10months between ages 4 and 14months with laquinimod (5, 10, or 25mg/kg), or PBS administered by oral gavage. Brain volumes were measured in a 7 Tesla magnetic resonance imager (MRI) at ages 4 and 14months. Behavioral testing included locomotor and rearing activity and the Morris water maze task. Cell numbers and immunophenotypes were assessed by multiparameter flow cytometry. Aβ deposition and tau phosphorylation were determined by immunohistochemistry. RESULTS In the F1 3xTg-AD/C3H animal model of AD, there was no detectable reduction of brain volume over a period of 10months of treatment, as there was not brain atrophy in any of the placebo or treatment groups. Laquinimod had no detectable effects on most neurobehavioral outcomes. The number or composition of CNS intrinsic cells and mononuclear subsets isolated from the CNS were not altered by laquinimod. CONCLUSION This is the first demonstration that there are no age-associated brain volume changes in the F1 3xTg-AD/C3H mouse model of Alzheimer's disease. Consequently, laquinimod had no effect on that outcome of this study. Most secondary outcomes on the effects of laquinimod on behavior and the number and composition of CNS-intrinsic cells and mononuclear cells within the CNS were also negative.
Collapse
Affiliation(s)
- Rehana Z Hussain
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - William A Miller-Little
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Doris Lambracht-Washington
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Tom C Jaramillo
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Masaya Takahashi
- Department of Radiology, University of Texas Southwestern Medical Center at Dallas, USA; Advanced Imaging Center, University of Texas Southwestern Medical Center at Dallas, USA
| | - Shanrong Zhang
- Advanced Imaging Center, University of Texas Southwestern Medical Center at Dallas, USA
| | - Min Fu
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Gary R Cutter
- Department of Biostatistics, University of Alabama at Birmingham, USA
| | | | - Craig M Powell
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Roger N Rosenberg
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA
| | - Olaf Stüve
- Department of Neurology and Neurotherapeutics, the University of Texas Southwestern Medical Center at Dallas, USA; Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Germany; Neurology Section, VA North Texas Health Care System, Medical Service Dallas, VA Medical Center, USA.
| |
Collapse
|
28
|
Laquinimod enhances central nervous system barrier functions. Neurobiol Dis 2017; 102:60-69. [PMID: 28235673 DOI: 10.1016/j.nbd.2017.02.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 12/22/2016] [Accepted: 02/20/2017] [Indexed: 12/30/2022] Open
Abstract
Laquinimod is currently being tested as a therapeutic drug in multiple sclerosis. However, its exact mechanism of action is still under investigation. Tracking of fluorescently-tagged encephalitogenic T cells during experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis, revealed that laquinimod significantly reduces the invasion of pathogenic effector T cells into the CNS tissue. T-cell activation, differentiation and amplification within secondary lymphoid organs after immunization with myelin antigen, their migratory capacity and re-activation within the nervous tissue were either only mildly affected or remained unchanged. Instead, laquinimod directly impacted the functionality of the CNS vasculature. The expression of tight junction proteins p120 and ZO-1 in human brain endothelial cells was up-regulated upon laquinimod treatment, resulting in a significant increase in the transendothelial electrical resistance of confluent monolayers of brain endothelial cells. Similarly, expression of the adhesion molecule activated leukocyte cell adhesion molecule (ALCAM) and inflammatory chemokines CCL2 and IP-10 was suppressed, leading to a significant reduction in the migration of memory TH1 and TH17 lymphocytes across the blood brain barrier (BBB). Our data indicate that laquinimod exerts its therapeutic effects by tightening the BBB and limiting parenchymal invasion of effector T cells, thereby reducing CNS damage.
Collapse
|
29
|
Laquinimod arrests experimental autoimmune encephalomyelitis by activating the aryl hydrocarbon receptor. Proc Natl Acad Sci U S A 2016; 113:E6145-E6152. [PMID: 27671624 DOI: 10.1073/pnas.1607843113] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Laquinimod is an oral drug currently being evaluated for the treatment of relapsing, remitting, and primary progressive multiple sclerosis and Huntington's disease. Laquinimod exerts beneficial activities on both the peripheral immune system and the CNS with distinctive changes in CNS resident cell populations, especially astrocytes and microglia. Analysis of genome-wide expression data revealed activation of the aryl hydrocarbon receptor (AhR) pathway in laquinimod-treated mice. The AhR pathway modulates the differentiation and function of several cell populations, many of which play an important role in neuroinflammation. We therefore tested the consequences of AhR activation in myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) using AhR knockout mice. We demonstrate that the pronounced effect of laquinimod on clinical score, CNS inflammation, and demyelination in EAE was abolished in AhR-/- mice. Furthermore, using bone marrow chimeras we show that deletion of AhR in the immune system fully abrogates, whereas deletion within the CNS partially abrogates the effect of laquinimod in EAE. These data strongly support the idea that AhR is necessary for the efficacy of laquinimod in EAE and that laquinimod may represent a first-in-class drug targeting AhR for the treatment of multiple sclerosis and other neurodegenerative diseases.
Collapse
|
30
|
Berg J, Mahmoudjanlou Y, Duscha A, Massa MG, Thöne J, Esser C, Gold R, Haghikia A. The immunomodulatory effect of laquinimod in CNS autoimmunity is mediated by the aryl hydrocarbon receptor. J Neuroimmunol 2016; 298:9-15. [DOI: 10.1016/j.jneuroim.2016.06.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 06/08/2016] [Indexed: 01/16/2023]
|
31
|
Abstract
Discussions of multiple sclerosis (MS) pathophysiology tend to focus on T cells and B cells of the adaptive immune response. The innate immune system is less commonly considered in this context, although dendritic cells, monocytes, macrophages and microglia - collectively referred to as myeloid cells - have prominent roles in MS pathogenesis. These populations of myeloid cells function as antigen-presenting cells and effector cells in neuroinflammation. Furthermore, a vicious cycle of interactions between T cells and myeloid cells exacerbates pathology. Several disease-modifying therapies are now available to treat MS, and insights into their mechanisms of action have largely focused on the adaptive immune system, but these therapies also have important effects on myeloid cells. In this Review, we discuss the evidence for the roles of myeloid cells in MS and the experimental autoimmune encephalomyelitis model of MS, and consider how interactions between myeloid cells and T cells and/or B cells promote MS pathology. Finally, we discuss the direct and indirect effects of existing MS medications on myeloid cells.
Collapse
Affiliation(s)
- Manoj K Mishra
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Drive, Calgary, Alberta, T2N 4N1, Canada
| |
Collapse
|
32
|
Lovelace MD, Varney B, Sundaram G, Franco NF, Ng ML, Pai S, Lim CK, Guillemin GJ, Brew BJ. Current Evidence for a Role of the Kynurenine Pathway of Tryptophan Metabolism in Multiple Sclerosis. Front Immunol 2016; 7:246. [PMID: 27540379 PMCID: PMC4972824 DOI: 10.3389/fimmu.2016.00246] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 06/10/2016] [Indexed: 12/13/2022] Open
Abstract
The kynurenine pathway (KP) is the major metabolic pathway of the essential amino acid tryptophan (TRP). Stimulation by inflammatory molecules, such as interferon-γ (IFN-γ), is the trigger for induction of the KP, driving a complex cascade of production of both neuroprotective and neurotoxic metabolites, and in turn, regulation of the immune response and responses of brain cells to the KP metabolites. Consequently, substantial evidence has accumulated over the past couple of decades that dysregulation of the KP and the production of neurotoxic metabolites are associated with many neuroinflammatory and neurodegenerative diseases, including Parkinson’s disease, AIDS-related dementia, motor neurone disease, schizophrenia, Huntington’s disease, and brain cancers. In the past decade, evidence of the link between the KP and multiple sclerosis (MS) has rapidly grown and has implicated the KP in MS pathogenesis. KP enzymes, indoleamine 2,3-dioxygenase (IDO-1) and tryptophan dioxygenase (highest expression in hepatic cells), are the principal enzymes triggering activation of the KP to produce kynurenine from TRP. This is in preference to other routes such as serotonin and melatonin production. In neurological disease, degradation of the blood–brain barrier, even if transient, allows the entry of blood monocytes into the brain parenchyma. Similar to microglia and macrophages, these cells are highly responsive to IFN-γ, which upregulates the expression of enzymes, including IDO-1, producing neurotoxic KP metabolites such as quinolinic acid. These metabolites circulate systemically or are released locally in the brain and can contribute to the excitotoxic death of oligodendrocytes and neurons in neurological disease principally by virtue of their agonist activity at N-methyl-d-aspartic acid receptors. The latest evidence is presented and discussed. The enzymes that control the checkpoints in the KP represent an attractive therapeutic target, and consequently several KP inhibitors are currently in clinical trials for other neurological diseases, and hence may make suitable candidates for MS patients. Underpinning these drug discovery endeavors, in recent years, several advances have been made in how KP metabolites are assayed in various biological fluids, and tremendous advancements have been made in how specimens are imaged to determine disease progression and involvement of various cell types and molecules in MS.
Collapse
Affiliation(s)
- Michael D Lovelace
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Bianca Varney
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Gayathri Sundaram
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Nunzio F Franco
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research , Sydney, NSW , Australia
| | - Mei Li Ng
- Faculty of Medicine, Sydney Medical School, University of Sydney , Sydney, NSW , Australia
| | - Saparna Pai
- Sydney Medical School, University of Sydney , Sydney, NSW , Australia
| | - Chai K Lim
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University , Sydney, NSW , Australia
| | - Gilles J Guillemin
- Neuroinflammation Group, Faculty of Medicine and Health Sciences, Macquarie University , Sydney, NSW , Australia
| | - Bruce J Brew
- Applied Neurosciences Program, Peter Duncan Neurosciences Research Unit, St Vincent's Centre for Applied Medical Research, Sydney, NSW, Australia; Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia; Department of Neurology, St Vincent's Hospital, Sydney, NSW, Australia
| |
Collapse
|
33
|
Deronic A, Tahvili S, Leanderson T, Ivars F. The anti-tumor effect of the quinoline-3-carboxamide tasquinimod: blockade of recruitment of CD11b(+) Ly6C(hi) cells to tumor tissue reduces tumor growth. BMC Cancer 2016; 16:440. [PMID: 27400708 PMCID: PMC4939705 DOI: 10.1186/s12885-016-2481-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Revised: 06/17/2016] [Accepted: 07/04/2016] [Indexed: 12/15/2022] Open
Abstract
Background Previous work has demonstrated immunomodulatory, anti-tumor, anti-metastatic and anti-angiogenic effects of the small molecule quinoline-3-carboxamide tasquinimod in pre-clinical cancer models. To better understand the anti-tumor effects of tasquinimod in transplantable tumor models, we have evaluated the impact of the compound both on recruitment of myeloid cells to tumor tissue and on tumor-induced myeloid cell expansion as these cells are known to promote tumor development. Methods Mice bearing subcutaneous 4 T1 mammary carcinoma tumors were treated with tasquinimod in the drinking water. A BrdU-based flow cytometry assay was utilized to assess the impact of short-term tasquinimod treatment on myeloid cell recruitment to tumors. Additionally, long-term treatment was performed to study the anti-tumor effect of tasquinimod as well as its effects on splenic myeloid cells and their progenitors. Myeloid cell populations were also immune-depleted by in vivo antibody treatment. Results Short-term tasquinimod treatment did not influence the proliferation of splenic Ly6Chi and Ly6Ghi cells, but instead reduced the influx of Ly6Chi cells to the tumor. Treatment with tasquinimod for various periods of time after tumor inoculation revealed that the anti-tumor effect of this compound mainly operated during the first few days of tumor growth. Similar to tasquinimod treatment, antibody-mediated depletion of Ly6Chi cells within that same time frame, caused reduced tumor growth, thereby confirming a significant role for these cells in tumor development. Additionally, long-term tasquinimod treatment reduced the splenomegaly and expansion of splenic myeloid cells during a later phase of tumor development. In this phase, tasquinimod normalized the tumor-induced alterations in myeloerythroid progenitor cells in the spleen but had only limited impact on the same populations in the bone marrow. Conclusions Our results indicate that tasquinimod treatment reduces tumor growth by operating early after tumor inoculation and that this effect is at least partially caused by reduced recruitment of Ly6Chi cells to tumor tissue. Long-term treatment also reduces the number of splenic myeloid cells and myeloerythroid progenitors, but these effects did not influence established rapidly growing tumors. Electronic supplementary material The online version of this article (doi:10.1186/s12885-016-2481-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adnan Deronic
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Tomas Leanderson
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Fredrik Ivars
- Immunology group, Section for Immunology, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| |
Collapse
|
34
|
Hainke U, Thomas K, Ziemssen T. Laquinimod in the treatment of relapsing remitting multiple sclerosis. Expert Opin Drug Metab Toxicol 2016; 12:701-9. [PMID: 27089834 DOI: 10.1080/17425255.2016.1179279] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
INTRODUCTION Laquinimod is a new once-daily oral administrable agent, which is under investigation in a phase 3 clinical trial for relapsing remitting multiple sclerosis (RRMS) and in a phase 2 clinical trial for primary progressive MS (PPMS). AREAS COVERED The pharmacokinetic, pharmacodynamic and the safety profiles of laquinimod are covered in this review. In preclinical studies, the ability to prevent both experimental autoimmune encephalomyelitis and experimental autoimmune neuritis has been demonstrated. Reduced cell infiltration, demyelination, axonal damage and a shift of T-helper cell responses have been shown. Accordingly, in human studies, a decrease of pro-inflammatory and an increase of anti-inflammatory cytokines have been measured and a significant reduction of disease progression and a decrease in brain volume loss has been demonstrated. During all clinical studies a favorable safety profile was observed for 0.6mg laquinimod. New information about cardiovascular events is prompting the discontinuation of higher dosing regimens in both ongoing trials. EXPERT OPINION Laquinimod is a first in class oral agent with high potential to reduce disease progression in RRMS and PPMS. Owing to its favorable safety profile, a combination with 0.6mg laquinimod and other disease modifying therapies could be an option in future MS therapy.
Collapse
Affiliation(s)
- Undine Hainke
- a Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus , Dresden University of Technology , Dresden , Germany
| | - Katja Thomas
- a Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus , Dresden University of Technology , Dresden , Germany
| | - Tjalf Ziemssen
- a Department of Neurology, Multiple Sclerosis Center, Center of Clinical Neuroscience, University Hospital Carl Gustav Carus , Dresden University of Technology , Dresden , Germany
| |
Collapse
|
35
|
Demyelination induces transport of ribosome-containing vesicles from glia to axons: evidence from animal models and MS patient brains. Mol Biol Rep 2016; 43:495-507. [DOI: 10.1007/s11033-016-3990-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 04/19/2016] [Indexed: 01/30/2023]
|
36
|
Bravo B, Gallego MI, Flores AI, Bornstein R, Puente-Bedia A, Hernández J, de la Torre P, García-Zaragoza E, Perez-Tavarez R, Grande J, Ballester A, Ballester S. Restrained Th17 response and myeloid cell infiltration into the central nervous system by human decidua-derived mesenchymal stem cells during experimental autoimmune encephalomyelitis. Stem Cell Res Ther 2016; 7:43. [PMID: 26987803 PMCID: PMC4797118 DOI: 10.1186/s13287-016-0304-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/29/2016] [Accepted: 03/02/2016] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Multiple sclerosis is a widespread inflammatory demyelinating disease. Several immunomodulatory therapies are available, including interferon-β, glatiramer acetate, natalizumab, fingolimod, and mitoxantrone. Although useful to delay disease progression, they do not provide a definitive cure and are associated with some undesirable side-effects. Accordingly, the search for new therapeutic methods constitutes an active investigation field. The use of mesenchymal stem cells (MSCs) to modify the disease course is currently the subject of intense interest. Decidua-derived MSCs (DMSCs) are a cell population obtained from human placental extraembryonic membranes able to differentiate into the three germ layers. This study explores the therapeutic potential of DMSCs. METHODS We used the experimental autoimmune encephalomyelitis (EAE) animal model to evaluate the effect of DMSCs on clinical signs of the disease and on the presence of inflammatory infiltrates in the central nervous system. We also compared the inflammatory profile of spleen T cells from DMSC-treated mice with that of EAE control animals, and the influence of DMSCs on the in vitro definition of the Th17 phenotype. Furthermore, we analyzed the effects on the presence of some critical cell types in central nervous system infiltrates. RESULTS Preventive intraperitoneal injection of DMSCs resulted in a significant delay of external signs of EAE. In addition, treatment of animals already presenting with moderate symptoms resulted in mild EAE with reduced disease scores. Besides decreased inflammatory infiltration, diminished percentages of CD4(+)IL17(+), CD11b(+)Ly6G(+) and CD11b(+)Ly6C(+) cells were found in infiltrates of treated animals. Early immune response was mitigated, with spleen cells of DMSC-treated mice displaying low proliferative response to antigen, decreased production of interleukin (IL)-17, and increased production of the anti-inflammatory cytokines IL-4 and IL-10. Moreover, lower RORγT and higher GATA-3 expression levels were detected in DMSC-treated mice. DMSCs also showed a detrimental influence on the in vitro definition of the Th17 phenotype. CONCLUSIONS DMSCs modulated the clinical course of EAE, modified the frequency and cell composition of the central nervous system infiltrates during the disease, and mediated an impairment of Th17 phenotype establishment in favor of the Th2 subtype. These results suggest that DMSCs might provide a new cell-based therapy for the control of multiple sclerosis.
Collapse
Affiliation(s)
- Beatriz Bravo
- />Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Marta I. Gallego
- />Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Mammary Gland Pathology, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Ana I. Flores
- />Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Rafael Bornstein
- />Hospital Central de Cruz Roja, Servicio de Hematología y Hemoterapia, Avenida de Reina Victoria 24, 28003 Madrid, Spain
| | - Alba Puente-Bedia
- />Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Javier Hernández
- />Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Paz de la Torre
- />Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Elena García-Zaragoza
- />Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Mammary Gland Pathology, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Raquel Perez-Tavarez
- />Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Histology Core Unit, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Jesús Grande
- />Grupo de Medicina Regenerativa, Instituto de Investigación Hospital 12 de Octubre, Avda. Córdoba s/n, 28041 Madrid, Spain
| | - Alicia Ballester
- />Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| | - Sara Ballester
- />Instituto de Salud Carlos III, Unidad Funcional de Investigación en Enfermedades Crónicas, Laboratory of Gene Regulation, Carretera de Majadahonda-Pozuelo Km 2, 28220 Madrid, Spain
| |
Collapse
|
37
|
Thöne J, Linker RA. Laquinimod in the treatment of multiple sclerosis: a review of the data so far. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1111-8. [PMID: 27042003 PMCID: PMC4798201 DOI: 10.2147/dddt.s55308] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Laquinimod (ABR-215062) is a new orally available carboxamide derivative, which is currently developed for relapsing remitting (RR) and chronic progressive (CP) forms of multiple sclerosis (MS; RRMS or CPMS) as well as neurodegenerative diseases. Its mechanism of action may comprise immunomodulatory effects on T-cells, monocytes, and dendritic cells as well as neuroprotective effects with prominent actions on astrocytes. Laquinimod was tested in Phase II and III clinical trials in RRMS at different dosages ranging from 0.1 to 0.6 mg/day. The compound was well tolerated, yet at the dosages tested only led to moderate effects on the reduction of relapse rates as primary study endpoint in Phase III trials. In contrast, significant effects on brain atrophy and disease progression were observed. While there were no significant safety signals in the clinical trials, the Committee for Medicinal Products for Human Use (CHMP) refused marketing authorization for RRMS based on the assessment of the risk–benefit ratio with regard to data from animal studies. At present, the compound is further tested in RRMS as well as CPMS and Huntington’s disease at different concentrations. Results from these trials will further inform about the clinical benefit of laquinimod in patient cohorts with a persisting, but still insufficiently met need for safe and at the same time effective oral compounds with neuroprotective effects.
Collapse
Affiliation(s)
- Jan Thöne
- Department of Neurology, University Hospital Essen, Essen, Germany
| | - Ralf A Linker
- Department of Neurology, University Hospital of Erlangen, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
38
|
Jiang W, St-Pierre S, Roy P, Morley BJ, Hao J, Simard AR. Infiltration of CCR2+Ly6Chigh Proinflammatory Monocytes and Neutrophils into the Central Nervous System Is Modulated by Nicotinic Acetylcholine Receptors in a Model of Multiple Sclerosis. THE JOURNAL OF IMMUNOLOGY 2016; 196:2095-108. [PMID: 26810225 DOI: 10.4049/jimmunol.1501613] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/20/2015] [Indexed: 12/18/2022]
Abstract
Myeloid cells, including proinflammatory monocytes and neutrophils, have important roles in the pathology of multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). These cells infiltrate the CNS in the early stages of disease development and contribute to the inflammatory response that is associated with symptom severity. It is thus crucial to identify and understand new mechanisms that can regulate the CNS infiltration of proinflammatory myeloid cells. Nicotinic acetylcholine receptors (nAChRs) have been increasingly studied for their immune-regulatory properties. In this study, we assessed the ability of nicotine, an nAChR ligand, to modulate proinflammatory myeloid cell numbers within the bone marrow, spleen, blood, and CNS of EAE mice. We found that nicotine significantly inhibits the infiltration of proinflammatory monocytes and neutrophils into the CNS at time points where these cells are known to play critical roles in disease pathology. In contrast, nicotine does not affect the expansion of other monocytes. We also show that nicotine exerts these effects by acting on α7 and α9 nAChR subtypes. Finally, mRNA transcript levels for CCL2 and CXCL2, chemokines involved in the chemotaxis of proinflammatory monocytes and neutrophils, respectively, are reduced in the brain of nicotine-treated EAE mice before the massive infiltration of these cells. Taken together, our data provide evidence that nAChRs can regulate proinflammatory cell infiltration into the CNS, which could be of significant value for the treatment of neuroinflammatory disorders.
Collapse
Affiliation(s)
- Wei Jiang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, Heping District 300052, China
| | - Stéphanie St-Pierre
- Département de Chimie et Biochimie, Université de Moncton, Moncton, New Brunswick E1A 3E9, Canada
| | - Patrick Roy
- Département de Chimie et Biochimie, Université de Moncton, Moncton, New Brunswick E1A 3E9, Canada
| | | | - Junwei Hao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, Heping District 300052, China;
| | - Alain R Simard
- Département de Chimie et Biochimie, Université de Moncton, Moncton, New Brunswick E1A 3E9, Canada; Centre de Formation Médicale du Nouveau-Brunswick, Moncton, New Brunswick E1A 3E9, Canada
| |
Collapse
|
39
|
Constantinescu SE, Constantinescu CS. Laquinimod (ABR-215062) for the treatment of relapsing multiple sclerosis. Expert Rev Clin Pharmacol 2015; 9:49-57. [DOI: 10.1586/17512433.2016.1108189] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
40
|
Andre R, Carty L, Tabrizi SJ. Disruption of immune cell function by mutant huntingtin in Huntington's disease pathogenesis. Curr Opin Pharmacol 2015; 26:33-8. [PMID: 26461267 DOI: 10.1016/j.coph.2015.09.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 09/15/2015] [Accepted: 09/16/2015] [Indexed: 12/28/2022]
Abstract
Innate immune dysfunction is increasingly recognised as a key characteristic of neurodegenerative disease. In the fatal inherited neurological disorder, Huntington's disease, altered innate immune cell function and increased inflammation are observed in the brain and the periphery of disease gene carriers many years before symptom onset, suggesting a potentially early and important role in disease pathogenesis. This is due, at least in part, to the intrinsic effects of the disease-causing protein, mutant huntingtin, expressed in innate immune cells themselves. Understanding whether such innate immune dysfunction in Huntington's disease can be targeted to slow the onset and/or the progression of the disease has significant therapeutic implications and is the subject of much current research.
Collapse
Affiliation(s)
- Ralph Andre
- UCL Institute of Neurology, Department of Neurodegenerative Disease, London, UK
| | - Lucy Carty
- UCL Institute of Neurology, Department of Neurodegenerative Disease, London, UK
| | - Sarah J Tabrizi
- UCL Institute of Neurology, Department of Neurodegenerative Disease, London, UK.
| |
Collapse
|
41
|
Abstract
The mononuclear phagocytes (dendritic cells and macrophages) are closely related immune cells with central roles in anti-infectious defense and maintenance of organ integrity. The canonical function of dendritic cells is the activation of T cells, whereas macrophages remove apoptotic cells and microbes by phagocytosis. In the kidney, these cell types form an intricate system of mononuclear phagocytes that surveys against injury and infection and contributes to organ homeostasis and tissue repair but may also promote progression of CKD. This review summarizes the general functions and classification of dendritic cells and macrophages in the immune system and recapitulates why overlapping definitions and historically separate research have created controversy about their tasks. Their roles in acute kidney disease, CKD, and renal transplantation are described, and therapeutic strategy to modify these cells for therapeutic purposes is discussed.
Collapse
Affiliation(s)
- Christina K Weisheit
- Institute of Experimental Immunology, University Clinic, Rheinische Friedrich-Wilhelms University, Bonn, Germany and Clinic for Anesthesiology and Intensive Care, University Clinic, Rheinische Friedrich-Wilhelms University, Bonn, Germany; and
| | - Daniel R Engel
- Institute of Experimental Immunology, University Clinic, Rheinische Friedrich-Wilhelms University, Bonn, Germany and Institute for Experimental Immunology and Imaging, University Duisburg-Essen and University Hospital Essen, Essen, Germany
| | - Christian Kurts
- Institute of Experimental Immunology, University Clinic, Rheinische Friedrich-Wilhelms University, Bonn, Germany and
| |
Collapse
|
42
|
Kan QC, Pan QX, Zhang XJ, Chu YJ, Liu N, Lv P, Zhang GX, Zhu L. Matrine ameliorates experimental autoimmune encephalomyelitis by modulating chemokines and their receptors. Exp Mol Pathol 2015; 99:212-9. [DOI: 10.1016/j.yexmp.2015.06.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 06/15/2015] [Indexed: 11/30/2022]
|
43
|
Stasiolek M, Linker RA, Hayardeny L, Bar Ilan O, Gold R. Immune parameters of patients treated with laquinimod, a novel oral therapy for the treatment of multiple sclerosis: results from a double-blind placebo-controlled study. IMMUNITY INFLAMMATION AND DISEASE 2015; 3:45-55. [PMID: 26029365 PMCID: PMC4444148 DOI: 10.1002/iid3.42] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2014] [Revised: 10/21/2014] [Accepted: 10/22/2014] [Indexed: 11/11/2022]
Abstract
Laquinimod is a novel orally administered drug for the treatment of relapsing remitting multiple sclerosis (RRMS). In this immunological substudy of the phase III Assessment of Oral Laquinimod in Preventing Progression of MS (ALLEGRO) trial, we performed an ex vivo and in vitro analysis of effects exerted by laquinimod on peripheral blood immune cell populations from RRMS patients with a special focus on monocyte phenotype and function. Approximately 100 patients were enrolled following a standardized protocol. Half of the patients received laquinimod and the other half received placebo. Peripheral blood samples were collected prior to commencement of therapy and after 1, 3, 6, 12, and 24 months of continuous therapy. Main lymphocytic and antigen presenting cell fractions were analyzed in peripheral blood mononuclear cells (PBMCs) ex vivo by flow cytometry. The proliferative response of PBMCs to mitogen or recall antigen was assessed in culture experiments. Untouched monocytes were sorted magnetically and cultured under pro-inflammatory conditions. PBMC analysis showed no significant differences of investigated lymphocytic and antigen presenting cell populations over time within each group, or between the two groups. However, the detailed in vitro analysis of monocytes demonstrated a lower level of CD86 expression on monocytes stimulated with LPS in laquinimod patients beginning from the 1st month of treatment. Upon pro-inflammatory stimulation, monocytes obtained from laquinimod treated patients tended to secrete lower levels of the proinflammatory chemokines CCL2 or CCL5. Taken together, in this prospective study, we demonstrate immune modulation but no immunosuppressive biological activity of laquinimod in a large group of MS patients.
Collapse
Affiliation(s)
- Mariusz Stasiolek
- Department of Neurology, Polish Mother's Memorial Hospital-Research Institute Lodz, Poland
| | - Ralf A Linker
- Department of Neurology, Ruhr-University Bochum St. Josef-Hospital, Bochum, Germany
| | - Liat Hayardeny
- Teva Innovative Research and Development Group, Teva Pharmaceutical Industries Ltd 5 Bazel Street, Petah, Tiqva, 49131, Israel
| | - Oren Bar Ilan
- Teva Innovative Research and Development Group, Teva Pharmaceutical Industries Ltd 5 Bazel Street, Petah, Tiqva, 49131, Israel
| | - Ralf Gold
- Department of Neurology, Ruhr-University Bochum St. Josef-Hospital, Bochum, Germany
| |
Collapse
|
44
|
Wild EJ, Tabrizi SJ. Targets for future clinical trials in Huntington's disease: what's in the pipeline? Mov Disord 2014; 29:1434-45. [PMID: 25155142 PMCID: PMC4265300 DOI: 10.1002/mds.26007] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 07/28/2014] [Accepted: 07/30/2014] [Indexed: 01/08/2023] Open
Abstract
The known genetic cause of Huntington's disease (HD) has fueled considerable progress in understanding its pathobiology and the development of therapeutic approaches aimed at correcting specific changes linked to the causative mutation. Among the most promising is reducing expression of mutant huntingtin protein (mHTT) with RNA interference or antisense oligonucleotides; human trials are now being planned. Zinc-finger transcriptional repression is another innovative method to reduce mHTT expression. Modulation of mHTT phosphorylation, chaperone upregulation, and autophagy enhancement represent attempts to alter cellular homeostasis to favor removal of mHTT. Inhibition of histone deacetylases (HDACs) remains of interest; recent work affirms HDAC4 as a target but questions the assumed centrality of its catalytic activity in HD. Phosphodiesterase inhibition, aimed at restoring synaptic function, has progressed rapidly to human trials. Deranged cellular signaling provides several tractable targets, but specificity and complexity are challenges. Restoring neurotrophic support in HD remains a key potential therapeutic approach. with several approaches being pursued, including brain-derived neurotrophic factor (BDNF) mimesis through tyrosine receptor kinase B (TrkB) agonism and monoclonal antibodies. An increasing understanding of the role of glial cells in HD has led to several new therapeutic avenues, including kynurenine monooxygenase inhibition, immunomodulation by laquinimod, CB2 agonism, and others. The complex metabolic derangements in HD remain under study, but no clear therapeutic strategy has yet emerged. We conclude that many exciting therapeutics are progressing through the development pipeline, and combining a better understanding of HD biology in human patients, with concerted medicinal chemistry efforts, will be crucial for bringing about an era of effective therapies.
Collapse
Affiliation(s)
- Edward J Wild
- Department of Neurodegenerative Disease, UCL Institute of Neurology, National Hospital for Neurology & NeurosurgeryQueen Square, London, WC1N 3BG, UK
| | - Sarah J Tabrizi
- Department of Neurodegenerative Disease, UCL Institute of Neurology, National Hospital for Neurology & NeurosurgeryQueen Square, London, WC1N 3BG, UK
| |
Collapse
|
45
|
Ashhurst TM, van Vreden C, Niewold P, King NJC. The plasticity of inflammatory monocyte responses to the inflamed central nervous system. Cell Immunol 2014; 291:49-57. [PMID: 25086710 PMCID: PMC7094263 DOI: 10.1016/j.cellimm.2014.07.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Accepted: 07/01/2014] [Indexed: 12/24/2022]
Abstract
Over the last three decades it has become increasingly clear that monocytes, originally thought to have fixed, stereotypic responses to foreign stimuli, mediate exquisitely balanced protective and pathogenic roles in disease and immunity. This balance is crucial in core functional organs, such as the central nervous system (CNS), where minor changes in neuronal microenvironments and the production of immune factors can result in significant disease with fatal consequences or permanent neurological sequelae. Viral encephalitis and multiple sclerosis are examples of important human diseases in which the pathogenic contribution of monocytes recruited from the bone marrow plays a critical role in the clinical expression of disease, as they differentiate into macrophage or dendritic cells in the CNS to carry out effector functions. While antigen-specific lymphocyte populations are central to the adaptive immune response in both cases, in viral encephalitis a prominent macrophage infiltration may mediate immunopathological damage, seizure induction, and death. However, the autoimmune response to non-replicating, non-infectious, but abundant, self antigen has a different disease progression, associated with differentiation of significant numbers of infiltrating monocytes into dendritic cells in the CNS. Whilst a predominant presence of macrophages or dendritic cells in the inflamed CNS in viral encephalitis or multiple sclerosis is well described, the way in which the inflamed CNS mobilizes monocytes in the bone marrow to migrate to the CNS and the key drivers that lead to these specific differentiation pathways in vivo are not well understood. Here we review the current understanding of factors facilitating inflammatory monocyte generation, migration and entry into the brain, as well as their differentiation towards macrophages or dendritic cells in viral and autoimmune disease in relation to their respective disease outcomes.
Collapse
Affiliation(s)
- Thomas Myles Ashhurst
- Viral Immunopathology Laboratory, Discipline of Pathology, Bosch Institute and The Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Medical Sciences, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Caryn van Vreden
- Viral Immunopathology Laboratory, Discipline of Pathology, Bosch Institute and The Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Medical Sciences, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Paula Niewold
- Viral Immunopathology Laboratory, Discipline of Pathology, Bosch Institute and The Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Medical Sciences, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| | - Nicholas Jonathan Cole King
- Viral Immunopathology Laboratory, Discipline of Pathology, Bosch Institute and The Marie Bashir Institute for Infectious Diseases and Biosecurity, School of Medical Sciences, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
46
|
Kelland EE, Gilmore W, Hayardeny L, Weiner LP, Lund BT. In vitro assessment of the direct effect of laquinimod on basic functions of human neural stem cells and oligodendrocyte progenitor cells. J Neurol Sci 2014; 346:66-74. [PMID: 25125045 DOI: 10.1016/j.jns.2014.07.058] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 06/19/2014] [Accepted: 07/28/2014] [Indexed: 12/21/2022]
Abstract
Laquinimod is an orally active immunomodulatory small molecule that has shown clear clinical benefit in trials for relapsing-remitting multiple sclerosis and in experimental rodent models that emulate multiple sclerosis (MS). Studies in healthy mice, and in mice with experimental autoimmune encephalomyelitis, have demonstrated that laquinimod is capable of entering the central nervous system. It is therefore important to determine if laquinimod is capable of a direct influence on basic functions of neural stem cells (NSC) or oligodendrocyte progenitor cells (OPC)--cells critical for myelin repair in MS. In order to address this question, a series of experiments was conducted to determine the effect of exogenous laquinimod on viability, proliferation, migration and differentiation of human NSC and OPC in vitro. These data show, for the first time in cells of human origin, that direct, short-term interaction between laquinimod and NSC or OPC, in an isolated in vitro setting, is not detrimental to the basic cellular function of these cells.
Collapse
Affiliation(s)
- Eve E Kelland
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA.
| | - Wendy Gilmore
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Liat Hayardeny
- Pharmacology Unit, Global Innovative R&D, Teva Pharmaceutical Industries, Netanya, Israel
| | - Leslie P Weiner
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA; Department of Molecular Microbiology and Immunology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Brett T Lund
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
47
|
Abstract
Multiple sclerosis (MS), an inflammatory disease affecting the central nervous system, is considered to exhibit an important neurodegenerative component as well. Laquinimod is an orally administered quinoline-3-carboxamide under development for the treatment of MS. In vitro and animal studies have revealed various mechanisms by which laquinimod may exert its effects on the immune and nervous systems. These include effects on the innate immune system that promote the differentiation of anti-inflammatory/regulatory T cells, the activation of microglia cells, an increase in the expression of brain-derived neurotrophic factor, as well as the prevention of inflammation-induced excitotoxicity. Two phase III studies revealed the clinical benefits of laquinimod in patients with relapsing-remitting MS and exhibited a benign safety profile for this drug. Ongoing clinical trials will help to define the optimal dose and indication for laquinimod in MS. This article reviews current experimental and clinical evidence on the role of laquinimod in patients with this disabling disease.
Collapse
Affiliation(s)
- Bernd C Kieseier
- Department of Neurology, Heinrich-Heine-University, Moorenstraße 5, 40225 Düsseldorf, Germany
| |
Collapse
|
48
|
Laquinimod exerts strong clinical and immunomodulatory effects in Lewis rat experimental autoimmune neuritis. J Neuroimmunol 2014; 274:38-45. [PMID: 25005118 DOI: 10.1016/j.jneuroim.2014.06.012] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Revised: 06/03/2014] [Accepted: 06/17/2014] [Indexed: 11/23/2022]
Abstract
Laquinimod is an immunomodulatory drug with neuroprotective potential. We used the animal model of experimental autoimmune neuritis (EAN) in the Lewis rat to study the effects of laquinimod treatment. After immunization with the neuritogenic peptide aa 53-78 of P2 myelin protein, preventive therapy with 12.5mg/kg laquinimod once daily inhibited neuritis in clinical and electrophysiological terms. Histology corroborated a lower degree of inflammatory lesions and demyelination in the sciatic nerve. The proportion of FoxP3-positive regulatory T cells in the peripheral lymph nodes of treated rats remained unchanged. We conclude that laquinimod may represent a therapeutic option in human autoimmune neuropathies.
Collapse
|
49
|
Mishra MK, Wang J, Keough MB, Fan Y, Silva C, Sloka S, Hayardeny L, Brück W, Yong VW. Laquinimod reduces neuroaxonal injury through inhibiting microglial activation. Ann Clin Transl Neurol 2014; 1:409-22. [PMID: 25356411 PMCID: PMC4184669 DOI: 10.1002/acn3.67] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 04/19/2014] [Accepted: 04/22/2014] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Laquinimod is an emerging oral medication for multiple sclerosis (MS) that reduces brain atrophy and progression of disability in two Phase III clinical trials. The mechanism of these effects is unclear. Persistent activation of microglia occurs in MS and contributes to injury. Thus, we investigated whether laquinimod alters properties of microglia in culture and in experimental autoimmune encephalomyelitis (EAE), and whether this reduces neurodegeneration. METHODS Microglia were cultured from human brains. EAE was induced in mice. RESULTS The activation of human microglia increased levels of several pro- and anti-inflammatory cytokines and these elevations were attenuated by pretreatment with laquinimod. Laquinimod prevented the decline in activated microglia of miR124a, a microRNA implicated in maintaining microglia quiescence, and reduced the activity of several signaling pathways (Jun-N-terminal kinase, ribosomal S6 kinase, and AKT/protein kinase B) in activated microglia. In EAE, axonal injury correlated with accumulation of microglia/macrophages in the spinal cord. EAE mice treated with laquinimod before onset of clinical signs subsequently had reduced microglia/macrophage density and axonal injury. Remarkably, when laquinimod treatment was initiated well into the disease course, the progressive demyelination, and axonal loss was halted. Besides inflammatory molecules associated with microglia, the level of inducible nitric oxide (NO) synthase capable of producing free radical toxicity was attenuated by laquinimod in EAE mice. Finally, in coculture where microglia activation caused neuronal death, laquinimod decreased NO levels, and neurotoxicity. INTERPRETATION Laquinimod is a novel inhibitor of microglial activation that lowers microglia-induced neuronal death in culture and axonal injury/loss in EAE.
Collapse
Affiliation(s)
- Manoj Kumar Mishra
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Janet Wang
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Michael B Keough
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Yan Fan
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Claudia Silva
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| | - Scott Sloka
- Grand River Hospital Kitchener, Ontario, Canada
| | | | - Wolfgang Brück
- Department of Neuropathology, University Medical Center Göttingen, Germany
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary Calgary, Alberta, Canada
| |
Collapse
|
50
|
Lourenço EV, Wong M, Hahn BH, Palma-Diaz MF, Skaggs BJ. Laquinimod delays and suppresses nephritis in lupus-prone mice and affects both myeloid and lymphoid immune cells. Arthritis Rheumatol 2014; 66:674-85. [PMID: 24574228 DOI: 10.1002/art.38259] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 10/24/2013] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Lupus nephritis depends on autoantibody deposition and activation of multiple immune cell types that promote kidney inflammation, including lymphocytes and monocyte/macrophages. Laquinimod, currently in clinical trials for multiple sclerosis and lupus nephritis, reduces infiltration of inflammatory cells into the spinal cord in experimental autoimmune encephalomyelitis. Activated monocyte/macrophages infiltrate the kidneys during nephritis in systemic lupus erythematosus (SLE). We undertook this study to determine whether using laquinimod to reduce monocyte/macrophage-driven tissue damage as well as to alter lymphocytes in SLE nephritis could have greater therapeutic benefit than current treatments that primarily affect lymphocytes, such as mycophenolate mofetil (MMF). METHODS To test laquinimod efficacy, we used the (NZB × NZW)F1 mouse model of SLE, in which disease manifests as nephritis. Preventive and therapeutic studies were performed to determine whether laquinimod could prevent or delay nephritis, as measured by proteinuria, serum creatinine, survival, and renal pathology. Spleen and kidney leukocyte populations and suppression assays were analyzed by flow cytometry. RESULTS Laquinimod prevented or delayed lupus manifestations at levels equal to or better than MMF. Laquinimod treatment was associated with reduced numbers of monocyte/macrophages, dendritic cells, and lymphocytes, as well as with induction of myeloid-derived suppressor cells in spleens and kidneys. Laquinimod suppressed macrophage-secreted tumor necrosis factor α and induced production of interleukin-10 (IL-10). In addition, laquinimod suppressed interferon-γ and IL-17 production by lymphocytes and down-regulated expression of activation/costimulatory markers on antigen-presenting cells. CONCLUSION The effects of laquinimod on myeloid and lymphoid cells may contribute to improvements in (NZB × NZW)F1 mouse survival, proteinuria, and glomerulonephritis. Future development of laquinimod as a therapeutic agent for lupus nephritis is promising.
Collapse
|