1
|
Goto A, Yamamoto S, Igari T, Matsumoto SI, Chisaki I, Iida K, Nakayama M, Oda A, Kakoi Y, Uchida A, Miyata K, Nishikawa M, Nagata T, Kusuhara H, Yokota T, Hirabayashi H. Quantitative Model Analysis and Simulation of Pharmacokinetics and Metastasis-Associated Lung Adenocarcinoma 1 RNA Knockdown Effect After Systemic Administration of Cholesterol-Conjugated DNA/RNA Heteroduplex Oligonucleotide Crossing Blood-Brain Barrier of Mice. J Pharmacol Exp Ther 2023; 384:197-204. [PMID: 36273821 DOI: 10.1124/jpet.122.001331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/07/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022] Open
Abstract
The cholesterol-conjugated heteroduplex oligonucleotide (Chol-HDO) is a double-stranded complex; it comprises an antisense oligonucleotide (ASO) and its complementary strand with a cholesterol ligand. Chol-HDO is a powerful tool for achieving target RNA knockdown in the brains of mice after systemic injection. Here, a quantitative model analysis was conducted to characterize the relationship between the pharmacokinetics (PK) and pharmacodynamics (PD), non-coding RNA metastasis-associated lung adenocarcinoma 1 (Malat1) RNA, of Chol-HDO, in a time-dependent manner. The established PK model could describe regional differences in the observed brain concentration-time profiles. Incorporating the PD model enabled the unique knockdown profiles in the brain to be explained in terms of the time delay after single dosing and enhancement following repeated dosing. Moreover, sensitivity analysis of PK exposure/persistency, target RNA turnover, and knockdown potency identified key factors for the efficient and sustained target RNA knockdown in the brain. The simulation of an adequate dosing regimen quantitatively supported the benefit of Chol-HDO in terms of achieving a suitable dosing interval. This was achieved via sufficient and sustained brain exposure and subsequent strong and sustained target RNA knockdown in the brain, even after systemic injection. The present study provides new insights into drug discoveries and development strategies for HDO in patients with neurogenic disorders. SIGNIFICANCE STATEMENT: The quantitative model analysis presented here characterized the PK/PD relationship of Chol-HDO, enabled its simulation under various conditions or assumptions, and identified key factors for efficient and sustained RNA knockdown, such as PK exposure and persistency. Chol-HDO appears to be an efficient drug delivery system for the systemic administration of desired drugs to brain targets.
Collapse
Affiliation(s)
- Akihiko Goto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Syunsuke Yamamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Tomoko Igari
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Shin-Ichi Matsumoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Ikumi Chisaki
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Koichi Iida
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Miyu Nakayama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Akira Oda
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Yuuichi Kakoi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Akio Uchida
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Kenichi Miyata
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Makiya Nishikawa
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Tetsuya Nagata
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Hiroyuki Kusuhara
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Takanori Yokota
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| | - Hideki Hirabayashi
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.G., S.Y., T.I., S.M., I.C., K.I., Mi.N., H.H.); Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.O., Y.K.); Asia New Chemical Entity Production Laboratories, Takeda Pharmaceutical Company Limited, Kanagawa, Japan (A.U., K.M.); Laboratory of Biopharmaceutics, Faculty of Pharmaceutical Sciences, Tokyo University of Science, Chiba, Japan (Ma.N.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (H.K.); and Department of Neurology and Neurologic Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan. (T.N., T.Y.)
| |
Collapse
|
2
|
Quaranta DV, Weaver RR, Baumann KK, Fujimoto T, Williams LM, Kim HC, Logsdon AF, Omer M, Reed MJ, Banks WA, Erickson MA. Transport of the Proinflammatory Chemokines C-C Motif Chemokine Ligand 2 (MCP-1) and C-C Motif Chemokine Ligand 5 (RANTES) across the Intact Mouse Blood-Brain Barrier Is Inhibited by Heparin and Eprodisate and Increased with Systemic Inflammation. J Pharmacol Exp Ther 2023; 384:205-223. [PMID: 36310035 PMCID: PMC9827507 DOI: 10.1124/jpet.122.001380] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/26/2022] [Accepted: 10/17/2022] [Indexed: 01/12/2023] Open
Abstract
One important function of the vascular blood-brain barrier (BBB) is to facilitate neuroimmune communication. The BBB fulfills this function, in part, through its ability to transport cytokines and chemokines. C-C motif chemokine receptor 2 (CCL2) (MCP-1) and C-C motif chemokine receptor 5 (CCL5) (RANTES) are proinflammatory chemokines that mediate neuroimmune responses to acute insults and aspects of brain injury and neurodegenerative diseases; however, a blood-to-brain transport system has not been evaluated for either chemokine in vivo. Therefore, we determined whether CCL2 and CCL5 in blood can cross the intact BBB and enter the brain. Using CD-1 mice, we found that 125I-labeled CCL2 and CCL5 crossed the BBB and entered the brain parenchyma. We next aimed to identify the mechanisms of 125I-CCL2 and 125I-CCL5 transport in an in situ brain perfusion model. We found that both heparin and eprodisate inhibited brain uptake of 125I-CCL2 and 125I-CCL5 in situ, whereas antagonists of their receptors, CCR2 or CCR5, respectively, did not, suggesting that heparan sulfates at the endothelial surface mediate BBB transport. Finally, we showed that CCL2 and CCL5 transport across the BBB increased following a single injection of 0.3 mg/kg lipopolysaccharide. These data demonstrate that CCL2 and CCL5 in the brain can derive, in part, from the circulation, especially during systemic inflammation. Further, binding to the BBB-associated heparan sulfate is a mechanism by which both chemokines can cross the intact BBB, highlighting a novel therapeutic target for treating neuroinflammation. SIGNIFICANCE STATEMENT: Our work demonstrates that C-C motif chemokine ligand 2 (CCL2) and C-C motif chemokine ligand 5 (CCL5) can cross the intact blood-brain barrier and that transport is robustly increased during inflammation. These data suggest that circulating CCL2 and CCL5 can contribute to brain levels of each chemokine. We further show that the transport of both chemokines is inhibited by heparin and eprodisate, suggesting that CCL2/CCL5-heparan sulfate interactions could be therapeutically targeted to limit accumulation of these chemokines in the brain.
Collapse
Affiliation(s)
- Daniel V Quaranta
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - Riley R Weaver
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - Kristen K Baumann
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - Takashi Fujimoto
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - Lindsey M Williams
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - Hyung Chan Kim
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - Aric F Logsdon
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - Mohamed Omer
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - May J Reed
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - William A Banks
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| | - Michelle A Erickson
- Geriatric Research Education and Clinical Center, VA Puget Sound Healthcare System, Seattle, Washington (D.V.Q., R.R.W., K.K.B., T.F., L.M.W., H.C.K., A.F.L., M.O., M.J.R., W.A.B., M.A.E.); Department of Neurosurgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan (T.F.); and Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington (H.C.K., A.F.L., M.J.R., W.A.B., M.A.E.)
| |
Collapse
|
3
|
Errede M, Annese T, Petrosino V, Longo G, Girolamo F, de Trizio I, d'Amati A, Uccelli A, Kerlero de Rosbo N, Virgintino D. Microglia-derived CCL2 has a prime role in neocortex neuroinflammation. Fluids Barriers CNS 2022; 19:68. [PMID: 36042496 PMCID: PMC9429625 DOI: 10.1186/s12987-022-00365-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/03/2022] [Indexed: 11/12/2022] Open
Abstract
Background In myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE), several areas of demyelination are detectable in mouse cerebral cortex, where neuroinflammation events are associated with scarce inflammatory infiltrates and blood–brain barrier (BBB) impairment. In this condition, the administration of mesenchymal stem cells (MSCs) controls neuroinflammation, attenuating astrogliosis and promoting the acquisition of stem cell traits by astrocytes. To contribute to the understanding of the mechanisms involved in the pathogenesis of EAE in gray matter and in the reverting effects of MSC treatment, the neocortex of EAE-affected mice was investigated by analyzing the cellular source(s) of chemokine CCL2, a molecule involved in immune cell recruitment and BBB-microvessel leakage. Methods The study was carried out by immunohistochemistry (IHC) and dual RNAscope IHC/in situ hybridization methods, using astrocyte, NG2-glia, macrophage/microglia, and microglia elective markers combined with CCL2. Results The results showed that in EAE-affected mice, hypertrophic microglia are the primary source of CCL2, surround the cortex neurons and the damaged BBB microvessels. In EAE-affected mice treated with MSCs, microgliosis appeared diminished very soon (6 h) after treatment, an observation that was long-lasting (tested after 10 days). This was associated with a reduced CCL2 expression and with apparently preserved/restored BBB features. In conclusion, the hallmark of EAE in the mouse neocortex is a condition of microgliosis characterized by high levels of CCL2 expression. Conclusions This finding supports relevant pathogenetic and clinical aspects of the human disease, while the demonstrated early control of neuroinflammation and BBB permeability exerted by treatment with MSCs may have important therapeutic implications. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00365-5.
Collapse
Affiliation(s)
- Mariella Errede
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Tiziana Annese
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.,Department of Medicine and Surgery, LUM University, Casamassima Bari, Italy
| | - Valentina Petrosino
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy
| | - Giovanna Longo
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Francesco Girolamo
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Ignazio de Trizio
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy
| | - Antonio d'Amati
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.,Department of Emergency and Organ Transplantation, Pathology Section, University of Bari School of Medicine, Bari, Italy
| | - Antonio Uccelli
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, Genoa, Italy.,IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nicole Kerlero de Rosbo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy.,TomaLab, Institute of Nanotechnology, Consiglio Nazionale Delle Ricerche (CNR), Rome, Italy
| | - Daniela Virgintino
- Department of Basic Medical Sciences, Neuroscience, and Sensory Organs, University of Bari School of Medicine, Piazza Giulio Cesare, Policlinics, 70124, Bari, Italy.
| |
Collapse
|
4
|
Talsma AD, Niemi JP, Pachter JS, Zigmond RE. The primary macrophage chemokine, CCL2, is not necessary after a peripheral nerve injury for macrophage recruitment and activation or for conditioning lesion enhanced peripheral regeneration. J Neuroinflammation 2022; 19:179. [PMID: 35820932 PMCID: PMC9277969 DOI: 10.1186/s12974-022-02497-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 05/23/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Peripheral nerve injuries stimulate the regenerative capacity of injured neurons through a neuroimmune phenomenon termed the conditioning lesion (CL) response. This response depends on macrophage accumulation in affected dorsal root ganglia (DRGs) and peripheral nerves. The macrophage chemokine CCL2 is upregulated after injury and is allegedly required for stimulating macrophage recruitment and pro-regenerative signaling through its receptor, CCR2. In these tissues, CCL2 is putatively produced by neurons in the DRG and Schwann cells in the distal nerve. METHODS Ccl2fl/fl mice were crossed with Advillin-Cre, P0-Cre, or both to create conditional Ccl2 knockouts (CKOs) in sensory neurons, Schwann cells, or both to hypothetically remove CCL2 and macrophages from DRGs, nerves or both. CCL2 was localized using Ccl2-RFPfl/fl mice. CCL2-CCR2 signaling was further examined using global Ccl2 KOs and Ccr2gfp knock-in/knock-outs. Unilateral sciatic nerve transection was used as the injury model, and at various timepoints, chemokine expression, macrophage accumulation and function, and in vivo regeneration were examined using qPCR, immunohistochemistry, and luxol fast blue staining. RESULTS Surprisingly, in all CKOs, DRG Ccl2 gene expression was decreased, while nerve Ccl2 was not. CCL2-RFP reporter mice revealed CCL2 expression in several cell types beyond the expected neurons and Schwann cells. Furthermore, macrophage accumulation, myelin clearance, and in vivo regeneration were unaffected in all CKOs, suggesting CCL2 may not be necessary for the CL response. Indeed, Ccl2 global knockout mice showed normal macrophage accumulation, myelin clearance, and in vivo regeneration, indicating these responses do not require CCL2. CCR2 ligands, Ccl7 and Ccl12, were upregulated after nerve injury and perhaps could compensate for the absence of Ccl2. Finally, Ccr2gfp knock-in/knock-out animals were used to differentiate resident and recruited macrophages in the injured tissues. Ccr2gfp/gfp KOs showed a 50% decrease in macrophages in the distal nerve compared to controls with a relative increase in resident macrophages. In the DRG there was a small but insignificant decrease in macrophages. CONCLUSIONS CCL2 is not necessary for macrophage accumulation, myelin clearance, and axon regeneration in the peripheral nervous system. Without CCL2, other CCR2 chemokines, resident macrophage proliferation, and CCR2-independent monocyte recruitment can compensate and allow for normal macrophage accumulation.
Collapse
Affiliation(s)
- Aaron D Talsma
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Jon P Niemi
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA
| | - Joel S Pachter
- Department of Immunology, University of Connecticut Health Center, Farmington, CT, 06030-6125, USA
| | - Richard E Zigmond
- Department of Neurosciences, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH, 44106-4975, USA.
| |
Collapse
|
5
|
Fazia T, Nova A, Gentilini D, Beecham A, Piras M, Saddi V, Ticca A, Bitti P, McCauley JL, Berzuini C, Bernardinelli L. Investigating the Causal Effect of Brain Expression of CCL2, NFKB1, MAPK14, TNFRSF1A, CXCL10 Genes on Multiple Sclerosis: A Two-Sample Mendelian Randomization Approach. Front Bioeng Biotechnol 2020; 8:397. [PMID: 32432099 PMCID: PMC7216783 DOI: 10.3389/fbioe.2020.00397] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
Multiple Sclerosis (MS) exhibits considerable heterogeneity in phenotypic expression, course, prognosis and response to therapy. This suggests this disease involves multiple, as yet poorly understood, causal mechanisms. In this work we assessed the possible causal link between gene expression level of five selected genes related to the pro-inflammatory NF-κB signaling pathway (i.e., CCL2, NFKB1, MAPK14, TNFRSF1A, CXCL10) in ten different brain tissues (i.e., cerebellum, frontal cortex, hippocampus, medulla, occipital cortex, putamen, substantia nigra, thalamus, temporal cortex and intralobular white matter) and MS. We adopted a two-stage Mendelian Randomization (MR) approach for the estimation of the causal effects of interest, based on summary-level data from 20 multiplex Sardinian families and data provided by the United Kingdom Brain Expression Consortium (UKBEC). Through Radial-MR and Cochrane's Q statistics we identified and removed genetic variants which are most likely to be invalid instruments. To estimate the total causal effect, univariable MR was carried out separately for each gene and brain region. We used Inverse-Variance Weighted estimator (IVW) as main analysis and MR-Egger Regression estimator (MR-ER) and Weighted Median Estimator (WME) as sensitivity analysis. As these genes belong to the same pathway and thus they can be closely related, we also estimated their direct causal effects by applying IVW and MR-ER within a multivariable MR (MVMR) approach using set of genetic instruments specific and common (composite) to each multiple exposures represented by the expression of the candidate genes. Univariate MR analysis showed a significant positive total causal effect for CCL2 and NFKB1 respectively in medulla and cerebellum. MVMR showed a direct positive causal effect for NFKB1 and TNFRSF1A, and a direct negative causal effect for CCL2 in cerebellum; while in medulla we observed a direct positive causal effect for CCL2. Since in general we observed a different magnitude for the gene specific causal effect we hypothesize that in cerebellum and medulla the effect of each gene expression is direct but also mediated by the others. These results confirm the importance of the involvement of NF-κB signaling pathway in brain tissue for the development of the disease and improve our understanding in the pathogenesis of MS.
Collapse
Affiliation(s)
- Teresa Fazia
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Andrea Nova
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Davide Gentilini
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
- Bioinformatics and Statistical Genomics Unit, Istituto Auxologico Italiano IRCCS, Milan, Italy
- Molecular Biology Laboratory, Istituto Auxologico Italiano IRCCS, Milan, Italy
| | - Ashley Beecham
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL, United States
| | - Marialuisa Piras
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Valeria Saddi
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Anna Ticca
- Divisione di Neurologia, Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Pierpaolo Bitti
- Centro di Tipizzazione Tissutale, S.I.T., Presidio Ospedaliero S. Francesco, ASL Numero 3 Nuoro, Nuoro, Italy
| | - Jacob L. McCauley
- John P. Hussman Institute for Human Genomics, Miller School of Medicine, University of Miami, Miami, FL, United States
- Dr. John T. Macdonald Foundation Department of Human Genetics, Miller School of Medicine, Miami, FL, United States
| | - Carlo Berzuini
- Centre for Biostatistics, University of Manchester, Manchester, United Kingdom
| | - Luisa Bernardinelli
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| |
Collapse
|
6
|
Shemer A, Jung S. Differential roles of resident microglia and infiltrating monocytes in murine CNS autoimmunity. Semin Immunopathol 2015; 37:613-23. [PMID: 26240063 DOI: 10.1007/s00281-015-0519-z] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/24/2015] [Indexed: 12/19/2022]
Abstract
Macrophages can be of dual origin. Most tissue-resident macrophage compartments are generated before birth and subsequently maintain themselves independently from each other locally in healthy tissue. Under inflammatory conditions, these cells can however be complemented by macrophages derived from acute monocyte infiltrates. Due to the lack of suitable experimental systems, differential functional contributions of central nervous system (CNS)-resident microglia and monocyte-derived macrophages (MoMF) to CNS inflammation, such as experimental autoimmune encephalomyelitis (EAE), the mouse model of multiple sclerosis (MS), remain poorly understood. Here, we will review recent progress in this field that suggest distinct roles of microglia and MoMF in disease induction and progression, capitalizing on novel transgenic mouse models. The latter finding could have major implications for the rationale development of therapeutic approaches to the management of brain inflammation and MS therapy.
Collapse
Affiliation(s)
- Anat Shemer
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Steffen Jung
- Department of Immunology, Weizmann Institute of Science, Rehovot, 76100, Israel.
| |
Collapse
|
7
|
p38α (MAPK14) critically regulates the immunological response and the production of specific cytokines and chemokines in astrocytes. Sci Rep 2014; 4:7405. [PMID: 25502009 PMCID: PMC4264013 DOI: 10.1038/srep07405] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 11/19/2014] [Indexed: 02/08/2023] Open
Abstract
In CNS lesions, “reactive astrocytes” form a prominent cellular response. However, the nature of this astrocyte immune activity is not well understood. In order to study astrocytic immune responses to inflammation and injury, we generated mice with conditional deletion of p38α (MAPK14) in GFAP+ astrocytes. We studied the role of p38α signaling in astrocyte immune activation both in vitro and in vivo, and simultaneously examined the effects of astrocyte activation in CNS inflammation. Our results showed that specific subsets of cytokines (TNFα, IL-6) and chemokines (CCL2, CCL4, CXCL1, CXCL2, CXCL10) are critically regulated by p38α signaling in astrocytes. In an in vivo CNS inflammation model of intracerebral injection of LPS, we observed markedly attenuated astrogliosis in conditional GFAPcre p38α−/− mice. However, GFAPcre p38α−/− mice showed marked upregulation of CCL2, CCL3, CCL4, CXCL2, CXCL10, TNFα, and IL-1β compared to p38αfl/fl cohorts, suggesting that in vivo responses to LPS after GFAPcre p38α deletion are complex and involve interactions between multiple cell types. This finding was supported by a prominent increase in macrophage/microglia and neutrophil recruitment in GFAPcre p38α−/− mice compared to p38αfl/fl controls. Together, these studies provide important insights into the critical role of p38α signaling in astrocyte immune activation.
Collapse
|
8
|
Kim RY, Hoffman AS, Itoh N, Ao Y, Spence R, Sofroniew MV, Voskuhl RR. Astrocyte CCL2 sustains immune cell infiltration in chronic experimental autoimmune encephalomyelitis. J Neuroimmunol 2014; 274:53-61. [PMID: 25005117 DOI: 10.1016/j.jneuroim.2014.06.009] [Citation(s) in RCA: 121] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/09/2014] [Accepted: 06/17/2014] [Indexed: 02/09/2023]
Abstract
Chemokine (C-C motif) ligand 2 (CCL2), initially identified as monocyte chemoattractant protein-1 (MCP-1), recruits immune cells to the central nervous system (CNS) during autoimmune inflammation. CCL2 can be expressed by multiple cell types, but which cells are responsible for CCL2 function during acute and chronic phases of autoimmune disease is not known. We determined the role of CCL2 in astrocytes in vivo during experimental autoimmune encephalomyelitis (EAE) by using Cre-loxP gene deletion. Mice with a conditional gene deletion of CCL2 from astrocytes had less severe EAE late in disease while having a similar incidence and severity of disease at onset as compared to wild type (WT) control littermates. EAE mice devoid of CCL2 in astrocytes had less macrophage and T cell inflammation in the white matter of the spinal cord and less diffuse activation of astrocytes and microglia in both white and gray matter as well as less axonal loss and demyelination, compared to WT littermates. These findings demonstrate that CCL2 in astrocytes plays an important role in the continued recruitment of immune cells and activation of glial cells in the CNS during chronic EAE, thereby suggesting a novel cell specific target for neuroprotective treatments of chronic neuroinflammatory diseases.
Collapse
Affiliation(s)
- Roy Y Kim
- Molecular Cellular and Integrative Physiology Interdepartmental Ph.D. Program, University of California, Los Angeles; Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles
| | - Alexandria S Hoffman
- Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles
| | - Noriko Itoh
- Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles
| | - Yan Ao
- Department of Neurobiology, University of California, Los Angeles
| | - Rory Spence
- Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles
| | | | - Rhonda R Voskuhl
- Multiple Sclerosis Program, Department of Neurology, University of California, Los Angeles.
| |
Collapse
|
9
|
Kooij G, Kroon J, Paul D, Reijerkerk A, Geerts D, van der Pol SMA, van Het Hof B, Drexhage JA, van Vliet SJ, Hekking LHP, van Buul JD, Pachter JS, de Vries HE. P-glycoprotein regulates trafficking of CD8(+) T cells to the brain parenchyma. Acta Neuropathol 2014; 127:699-711. [PMID: 24429546 DOI: 10.1007/s00401-014-1244-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2013] [Accepted: 01/02/2014] [Indexed: 01/13/2023]
Abstract
The trafficking of cytotoxic CD8(+) T lymphocytes across the lining of the cerebral vasculature is key to the onset of the chronic neuro-inflammatory disorder multiple sclerosis. However, the mechanisms controlling their final transmigration across the brain endothelium remain unknown. Here, we describe that CD8(+) T lymphocyte trafficking into the brain is dependent on the activity of the brain endothelial adenosine triphosphate-binding cassette transporter P-glycoprotein. Silencing P-glycoprotein activity selectively reduced the trafficking of CD8(+) T cells across the brain endothelium in vitro as well as in vivo. In response to formation of the T cell-endothelial synapse, P-glycoprotein was found to regulate secretion of endothelial (C-C motif) ligand 2 (CCL2), a chemokine that mediates CD8(+) T cell migration in vitro. Notably, CCL2 levels were significantly enhanced in microvessels isolated from human multiple sclerosis lesions in comparison with non-neurological controls. Endothelial cell-specific elimination of CCL2 in mice subjected to experimental autoimmune encephalomyelitis also significantly diminished the accumulation of CD8(+) T cells compared to wild-type animals. Collectively, these results highlight a novel (patho)physiological role for P-glycoprotein in CD8(+) T cell trafficking into the central nervous system during neuro-inflammation and illustrate CCL2 secretion as a potential link in this mechanism.
Collapse
Affiliation(s)
- Gijs Kooij
- Blood-Brain Barrier Research Group, Department of Molecular Cell Biology and Immunology, Neuroscience Campus Amsterdam, VU University Medical Center, P.O. Box 7057, 1007 MB, Amsterdam, The Netherlands,
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Gruol DL, Vo K, Bray JG, Roberts AJ. CCL2-ethanol interactions and hippocampal synaptic protein expression in a transgenic mouse model. Front Integr Neurosci 2014; 8:29. [PMID: 24772072 PMCID: PMC3983522 DOI: 10.3389/fnint.2014.00029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2014] [Accepted: 03/12/2014] [Indexed: 12/20/2022] Open
Abstract
Chronic exposure to ethanol produces a number of detrimental effects on behavior. Neuroadaptive changes in brain structure or function underlie these behavioral effects and may be transient or persistent in nature. Central to the functional changes are alterations in the biology of neuronal and glial cells of the brain. Recent data show that ethanol induces glial cells of the brain to produce elevated levels of neuroimmune factors including CCL2, a key innate immune chemokine. Depending on the conditions of ethanol exposure, the upregulated levels of CCL2 can be transient or persistent and outlast the period of ethanol exposure. Importantly, results indicate that the upregulated levels of CCL2 may lead to CCL2-ethanol interactions that mediate or regulate the effects of ethanol on the brain. Glial cells are in close association with neurons and regulate many neuronal functions. Therefore, effects of ethanol on glial cells may underlie some of the effects of ethanol on neurons. To investigate this possibility, we are studying effects of chronic ethanol on hippocampal synaptic function in a transgenic mouse model that expresses elevated levels of CCL2 in the brain through enhanced glial expression, a situation know to occur in alcoholics. Both CCL2 and ethanol have been reported to alter synaptic function in the hippocampus. In the current study, we determined if interactions are evident between CCL2 and ethanol at the level of hippocampal synaptic proteins. Two ethanol exposure paradigms were used; the first involved ethanol exposure by drinking and the second involved ethanol exposure in a paradigm that combines drinking plus ethanol vapor. The first paradigm does not produce dependence on ethanol, whereas the second paradigm is commonly used to produce ethanol dependence. Results show modest effects of both ethanol exposure paradigms on the level of synaptic proteins in the hippocampus of CCL2 transgenic mice compared with their non-transgenic littermate controls, consistent with ethanol-CCL2 interactions. No evidence of toxic effects of CCL2 or CCL2-ethanol interactions was observed. Taken together, these results support the idea that ethanol induced astrocyte production of CCL2 can result in neuroadaptive changes that interact with the actions of ethanol.
Collapse
Affiliation(s)
- Donna L Gruol
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| | - Khanh Vo
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| | - Jennifer G Bray
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| | - Amanda J Roberts
- Molecular and Cellular Neuroscience Department, The Scripps Research Institute La Jolla, CA, USA
| |
Collapse
|
11
|
Shrestha B, Ge S, Pachter JS. Resolution of central nervous system astrocytic and endothelial sources of CCL2 gene expression during evolving neuroinflammation. Fluids Barriers CNS 2014; 11:6. [PMID: 24589378 PMCID: PMC3944978 DOI: 10.1186/2045-8118-11-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/26/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The chemokine CCL2 is a critical mediator of neuroinflammation in diseases such as multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). CCL2 drives mononuclear cell infiltration into the central nervous system (CNS), alters expression and distribution of microvascular endothelial tight junction proteins, and disrupts the blood-brain and blood-spinal cord barriers. Immunohistochemistry has consistently revealed astrocytes to be a source of this chemokine during neuroinflammation, while providing less uniform evidence that CNS endothelial cells may also express CCL2. Moreover, the relative contributions of these cell types to the CNS pool of CCL2 during MS/EAE are unclear and the aim of this study was to investigate this further. METHODS CCL2 gene expression was determined by qRT-PCR in different populations of CNS cells at different times following EAE induced by immunization with MOG35-55 peptide and adjuvants, or after injection with adjuvants alone. CNS cells types were isolated by two different protocols: bulk isolation to yield crude microvascular and parenchymal fractions (containing astrocytes, other glia, and neurons), or laser capture microdissection (LCM) to acquire more precisely microvascular endothelial cells, astrocytes or other parenchymal cells. RESULTS Both CNS microvessel and parenchymal populations prepared by crude bulk isolation showed up-regulation of CCL2 mRNA following MOG immunization or injection of adjuvants alone. More exact dissection by LCM revealed microvascular endothelial cells and astrocytes to be the specific sources of CCL2 gene induction following MOG immunization, while only astrocytes showed elevated CCL2 mRNA in response to just adjuvants. Astrocytes displayed the greatest degree of stimulation of CCL2 gene expression following EAE induction. CONCLUSIONS High-precision LCM affirmed both microvascular endothelial cells and astrocytes as the major CNS sources of CCL2 gene expression during EAE. Given the high accessibility of the CNS microvascular endothelium, endothelial-derived CCL2 could prove a viable target for therapeutic intervention in neuroinflammatory disease.
Collapse
Affiliation(s)
- Bandana Shrestha
- Blood–brain Barrier Laboratory, Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Shujun Ge
- Blood–brain Barrier Laboratory, Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| | - Joel S Pachter
- Blood–brain Barrier Laboratory, Department of Cell Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06030, USA
| |
Collapse
|
12
|
Paul D, Ge S, Lemire Y, Jellison ER, Serwanski DR, Ruddle NH, Pachter JS. Cell-selective knockout and 3D confocal image analysis reveals separate roles for astrocyte-and endothelial-derived CCL2 in neuroinflammation. J Neuroinflammation 2014; 11:10. [PMID: 24444311 PMCID: PMC3906899 DOI: 10.1186/1742-2094-11-10] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 12/28/2013] [Indexed: 12/30/2022] Open
Abstract
Background Expression of chemokine CCL2 in the normal central nervous system (CNS) is nearly undetectable, but is significantly upregulated and drives neuroinflammation during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis which is considered a contributing factor in the human disease. As astrocytes and brain microvascular endothelial cells (BMEC) forming the blood–brain barrier (BBB) are sources of CCL2 in EAE and other neuroinflammatory conditions, it is unclear if one or both CCL2 pools are critical to disease and by what mechanism(s). Methods Mice with selective CCL2 gene knockout (KO) in astrocytes (Astro KO) or endothelial cells (Endo KO) were used to evaluate the respective contributions of these sources to neuroinflammation, i.e., clinical disease progression, BBB damage, and parenchymal leukocyte invasion in a myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE model. High-resolution 3-dimensional (3D) immunofluorescence confocal microscopy and colloidal gold immuno-electron microscopy were employed to confirm sites of CCL2 expression, and 3D immunofluorescence confocal microscopy utilized to assess inflammatory responses along the CNS microvasculature. Results Cell-selective loss of CCL2 immunoreactivity was demonstrated in the respective KO mice. Compared to wild-type (WT) mice, Astro KO mice showed reduced EAE severity but similar onset, while Endo KO mice displayed near normal severity but significantly delayed onset. Neither of the KO mice showed deficits in T cell proliferation, or IL-17 and IFN-γ production, following MOG35-55 exposure in vitro, or altered MOG-major histocompatibility complex class II tetramer binding. 3D confocal imaging further revealed distinct actions of the two CCL2 pools in the CNS. Astro KOs lacked the CNS leukocyte penetration and disrupted immunostaining of CLN-5 at the BBB seen during early EAE in WT mice, while Endo KOs uniquely displayed leukocytes stalled in the microvascular lumen. Conclusions These results point to astrocyte and endothelial pools of CCL2 each regulating different stages of neuroinflammation in EAE, and carry implications for drug delivery in neuroinflammatory disease.
Collapse
Affiliation(s)
| | - Shujun Ge
- Department of Cell Biology, Blood-brain Barrier Laboratory, 263 Farmington Ave,, Farmington CT 06030, USA.
| | | | | | | | | | | |
Collapse
|
13
|
Estrogen mediates neuroprotection and anti-inflammatory effects during EAE through ERα signaling on astrocytes but not through ERβ signaling on astrocytes or neurons. J Neurosci 2013; 33:10924-33. [PMID: 23804112 DOI: 10.1523/jneurosci.0886-13.2013] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Estrogens can signal through either estrogen receptor α (ERα) or β (ERβ) to ameliorate experimental autoimmune encephalomyelitis (EAE), the most widely used mouse model of multiple sclerosis (MS). Cellular targets of estrogen-mediated neuroprotection are still being elucidated. Previously, we demonstrated that ERα on astrocytes, but not neurons, was critical for ERα ligand-mediated neuroprotection in EAE, including decreased T-cell and macrophage inflammation and decreased axonal loss. Here, we determined whether ERβ on astrocytes or neurons could mediate neuroprotection in EAE, by selectively removing ERβ from either of these cell types using Cre-loxP gene deletion. Our results demonstrated that, even though ERβ ligand treatment was neuroprotective in EAE, this neuroprotection was not mediated through ERβ on either astrocytes or neurons and did not involve a reduction in levels of CNS inflammation. Given the differential neuroprotective and anti-inflammatory effects mediated via ERα versus ERβ on astrocytes, we looked for molecules within astrocytes that were affected by signaling through ERα, but not ERβ. We found that ERα ligand treatment, but not ERβ ligand treatment, decreased expression of the chemokines CCL2 and CCL7 by astrocytes in EAE. Together, our data show that neuroprotection in EAE mediated via ERβ signaling does not require ERβ on either astrocytes or neurons, whereas neuroprotection in EAE mediated via ERα signaling requires ERα on astrocytes and reduces astrocyte expression of proinflammatory chemokines. These findings reveal important cellular differences in the neuroprotective mechanisms of estrogen signaling through ERα and ERβ in EAE.
Collapse
|
14
|
Chemokine CCL2 induces apoptosis in cortex following traumatic brain injury. J Mol Neurosci 2013; 51:1021-9. [PMID: 23934512 DOI: 10.1007/s12031-013-0091-8] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 07/15/2013] [Indexed: 12/19/2022]
Abstract
The chemokine C-C motif ligand 2 (CCL2) is an important mediator of neuroinflammation. Released in response to acute injury, ischemia, and neurodegenerative disease, CCL2 binds primarily to the G-protein-coupled chemokine C-C motif receptor 2 (CCR2) to recruit inflammatory cells to sites of tissue damage. Inflammation is thought to have both beneficial and deleterious consequences following traumatic brain injury (TBI), so we investigated CCL2-CCR2 signaling during the post-TBI period to assess possible neurodegenerative and protective actions. Local TBI in adult rat cortex was induced by Feeney's weight-drop method, and the expression of CCL2 and CCR2 in the tissue around the contusion site was measured by real-time quantitative PCR. Both CCL2 and CCR2 mRNA levels were increased markedly for at least 10 days after injury, peaking on day 3. The CCL2 protein was mainly co-localized with the astroglial marker glial fibrillary acidic protein and CCR2 protein with the neuronal nuclear marker NeuN as revealed by double immunofluorescence staining. A selective CCR2 antagonist, RS504393, reduced TUNEL staining, a marker of apoptosis, and improved performance in the Morris water maze 3 days post-TBI, suggesting that CCL2-CCR2 signaling has deleterious effects on neuronal survival and learning. Targeting the CCL2-CCR2 pathway may provide a novel therapeutic approach for the treatment of TBI.
Collapse
|
15
|
Jensen CJ, Massie A, De Keyser J. Immune players in the CNS: the astrocyte. J Neuroimmune Pharmacol 2013; 8:824-39. [PMID: 23821340 DOI: 10.1007/s11481-013-9480-6] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Accepted: 05/26/2013] [Indexed: 12/20/2022]
Abstract
In the finely balanced environment of the central nervous system astrocytes, the most numerous cell type, play a role in regulating almost every physiological system. First found to regulate extracellular ions and pH, they have since been shown to regulate neurotransmitter levels, cerebral blood flow and energy metabolism. There is also growing evidence for an essential role of astrocytes in central immunity, which is the topic of this review. In the healthy state, the central nervous system is potently anti-inflammatory but under threat astrocytes readily respond to pathogens and to both sterile and pathogen-induced cell damage. In response, astrocytes take on some of the roles of immune cells, releasing cyto- and chemokines to influence effector cells, modulating the blood-brain barrier and forming glial scars. To date, much of the data supporting a role for astrocytes in immunity have been obtained from in vitro systems; however data from experimental models and clinical samples support the suggestion that astrocytes perform similar roles in more complex environments. This review will discuss some aspects of the role of astrocytes in central nervous system immunity.
Collapse
Affiliation(s)
- Cathy J Jensen
- Department of Neurology, Universitair Ziekenhuis Brussel, Center for Neurosciences, Vrije Universiteit Brussel-VUB, Brussels, Belgium.
| | | | | |
Collapse
|
16
|
The CCL2 synthesis inhibitor bindarit targets cells of the neurovascular unit, and suppresses experimental autoimmune encephalomyelitis. J Neuroinflammation 2012; 9:171. [PMID: 22788993 PMCID: PMC3488971 DOI: 10.1186/1742-2094-9-171] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Accepted: 07/12/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Production of the chemokine CCL2 by cells of the neurovascular unit (NVU) drives critical aspects of neuroinflammation. Suppression of CCL2 therefore holds promise in treating neuroinflammatory disease. Accordingly, we sought to determine if the compound bindarit, which inhibits CCL2 synthesis, could repress the three NVU sources of CCL2 most commonly reported in neuroinflammation--astrocytes, microglia and brain microvascular endothelial cells (BMEC)--as well as modify the clinical course of neuroinflammatory disease. METHODS The effect of bindarit on CCL2 expression by cultured murine astrocytes, microglia and BMEC was examined by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Bindarit action on mouse brain and spinal cord in vivo was similarly investigated by qRT-PCR following LPS injection in mice. And to further gauge the potential remedial effects of bindarit on neuroinflammatory disease, its impact on the clinical course of experimental autoimmune encephalomyelitis (EAE) in mice was also explored. RESULTS Bindarit repressed CCL2 expression by all three cultured cells, and antagonized upregulated expression of CCL2 in both brain and spinal cord in vivo following LPS administration. Bindarit also significantly modified the course and severity of clinical EAE, diminished the incidence and onset of disease, and evidenced signs of disease reversal. CONCLUSION Bindarit was effective in suppressing CCL2 expression by cultured NVU cells as well as brain and spinal cord tissue in vivo. It further modulated the course of clinical EAE in both preventative and therapeutic ways. Collectively, these results suggest that bindarit might prove an effective treatment for neuroinflammatory disease.
Collapse
|
17
|
Carson MJ. Molecular Mechanisms and Consequences of Immune and Nervous System Interactions. BASIC NEUROCHEMISTRY 2012. [PMCID: PMC7149717 DOI: 10.1016/b978-0-12-374947-5.00033-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
This chapter provides an overview on the multiple mechanisms by which the nervous system regulates and directs immune function towards what is needed and tolerated by the nervous system. The immune system plays two essential roles necessary for the survival of complex organisms, including tissue homeostasis and tissue defense against pathogens. These immune functions are essential to maintain the functions of all organs in the body and are studied as a part of the general field of immunology. The immune system is divided into two arms: a rapid “innate” immune response system and a slow “adaptive” immune response system. The receptors for the types of “alarm” signals detected by the innate immune system are preformed and stably encoded in the genome. By contrast, the receptors for the types of signals that trigger the adaptive immune system are in part stochastically generated and in part shaped by the types and frequency of the pathogens encountered.
Collapse
|
18
|
Lyck R, Martinelli R. Mechanisms of T-cell migration across the BBB. FUTURE NEUROLOGY 2011. [DOI: 10.2217/fnl.11.18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Under physiological conditions, the highly specialized BBB strictly limits the entrance of immune cells into the CNS. By contrast, in the course of neuroinflammation such as that observed in multiple sclerosis, circulating T cells readily breach the BBB and initiate a cascade of events culminating in disease onset. Lymphocyte extravasation across the BBB occurs through a sequential multistep process, orchestrated by chemokines and cell adhesion molecules that precisely regulate the dynamic interaction of T cells with the endothelial cells forming the BBB. In this article, we will discuss the molecular players triggering the sophisticated process of T-cell migration across the BBB during pathological conditions.
Collapse
Affiliation(s)
- Ruth Lyck
- Theodor Kocher Institute, University of Bern, Freiestrasse 1, 3012 Bern, Switzerland
| | - Roberta Martinelli
- Department of Medicine, Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Avenue, Boston, MA 02215, USA
| |
Collapse
|
19
|
Semple BD, Frugier T, Morganti-Kossmann MC. CCL2 modulates cytokine production in cultured mouse astrocytes. J Neuroinflammation 2010; 7:67. [PMID: 20942978 PMCID: PMC2964657 DOI: 10.1186/1742-2094-7-67] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2010] [Accepted: 10/14/2010] [Indexed: 12/30/2022] Open
Abstract
Background The chemokine CCL2 (also known as monocyte chemoattractant protein-1, or MCP-1) is upregulated in patients and rodent models of traumatic brain injury (TBI), contributing to post-traumatic neuroinflammation and degeneration by directing the infiltration of blood-derived macrophages into the injured brain. Our laboratory has previously reported that Ccl2-/- mice show reduced macrophage accumulation and tissue damage, corresponding to improved motor recovery, following experimental TBI. Surprisingly, Ccl2-deficient mice also exhibited delayed but exacerbated secretion of key proinflammatory cytokines in the injured cortex. Thus we sought to further characterise CCL2's potential ability to modulate immunoactivation of astrocytes in vitro. Methods Primary astrocytes were isolated from neonatal wild-type and Ccl2-deficient mice. Established astrocyte cultures were stimulated with various concentrations of lipopolysaccharide (LPS) and interleukin (IL)-1β for up to 24 hours. Separate experiments involved pre-incubation with mouse recombinant (r)CCL2 prior to IL-1β stimulation in wild-type cells. Following stimulation, cytokine secretion was measured in culture supernatant by immunoassays, whilst cytokine gene expression was quantified by real-time reverse transcriptase polymerase chain reaction. Results LPS (0.1-100 μg/ml; 8 h) induced the significantly greater secretion of five key cytokines and chemokines in Ccl2-/- astrocytes compared to wild-type cells. Consistently, IL-6 mRNA levels were 2-fold higher in Ccl2-deficient cells. IL-1β (10 and 50 ng/ml; 2-24 h) also resulted in exacerbated IL-6 production from Ccl2-/- cultures. Despite this, treatment of wild-type cultures with rCCL2 alone (50-500 ng/ml) did not induce cytokine/chemokine production by astrocytes. However, pre-incubation of wild-type astrocytes with rCCL2 (250 ng/ml, 12 h) prior to stimulation with IL-1β (10 ng/ml, 8 h) significantly reduced IL-6 protein and gene expression. Conclusions Our data indicate that astrocytes are likely responsible for the exacerbated cytokine response seen in vivo post-injury in the absence of CCL2. Furthermore, evidence that CCL2 inhibits cytokine production by astrocytes following IL-1β stimulation, suggests a novel, immunomodulatory role for this chemokine in acute neuroinflammation. Further investigation is required to determine the physiological relevance of this phenomenon, which may have implications for therapeutics targeting CCL2-mediated leukocyte infiltration following TBI.
Collapse
Affiliation(s)
- Bridgette D Semple
- Department of Medicine, Monash University, National Trauma Research Institute, The Alfred Hospital, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
20
|
Roberts TK, Eugenin EA, Morgello S, Clements JE, Zink MC, Berman JW. PrPC, the cellular isoform of the human prion protein, is a novel biomarker of HIV-associated neurocognitive impairment and mediates neuroinflammation. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1848-60. [PMID: 20724601 DOI: 10.2353/ajpath.2010.091006] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Of the 33 million people infected with the human immunodeficiency virus (HIV) worldwide, 40-60% of individuals will eventually develop neurocognitive sequelae that can be attributed to the presence of HIV-1 in the central nervous system (CNS) and its associated neuroinflammation despite antiretroviral therapy. PrP(C) (protease resistant protein, cellular isoform) is the nonpathological cellular isoform of the human prion protein that participates in many physiological processes that are disrupted during HIV-1 infection. However, its role in HIV-1 CNS disease is unknown. We demonstrate that PrP(C) is significantly increased in both the CNS of HIV-1-infected individuals with neurocognitive impairment and in SIV-infected macaques with encephalitis. PrP(C) is released into the cerebrospinal fluid, and its levels correlate with CNS compromise, suggesting it is a biomarker of HIV-associated neurocognitive impairment. We show that the chemokine (c-c Motif) Ligand-2 (CCL2) increases PrP(C) release from CNS cells, while HIV-1 infection alters PrP(C) release from peripheral blood mononuclear cells. Soluble PrP(C) mediates neuroinflammation by inducing astrocyte production of both CCL2 and interleukin 6. This report presents the first evidence that PrP(C) dysregulation occurs in cognitively impaired HIV-1-infected individuals and that PrP(C) participates in the pathogenesis of HIV-1-associated CNS disease.
Collapse
Affiliation(s)
- Toni K Roberts
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | | | | | | | |
Collapse
|
21
|
Macdonald JA, Murugesan N, Pachter JS. Endothelial cell heterogeneity of blood-brain barrier gene expression along the cerebral microvasculature. J Neurosci Res 2010; 88:1457-74. [PMID: 20025060 DOI: 10.1002/jnr.22316] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The blood-brain barrier (BBB) refers to the network of microvessels that selectively restricts the passage of substances between the circulation and the central nervous system (CNS). This microvascular network is comprised of arterioles, capillaries and venules, yet the respective contribution of each of these to the BBB awaits clarification. In this regard, it has been postulated that brain microvascular endothelial cells (BMEC) from these different tributaries might exhibit considerable heterogeneity in form and function, with such diversity underlying unique roles in physiological and pathophysiological processes. Means to begin exploring such endothelial differences in situ, free from caveats associated with cell isolation and culturing procedures, are crucial to comprehending the nature and treatment of CNS diseases with vascular involvement. Here, the recently validated approach of immuno-laser capture microdissection (immuno-LCM) coupled to quantitative real-time PCR (qRT-PCR) was used to analyze gene expression patterns of BMEC retrieved in situ from either capillaries or venules. From profiling 87 genes known to play a role in BBB function and/or be enriched in isolated brain microvessels, results imply that most BBB properties reside in both segments, but that capillaries preferentially express some genes related to solute transport, while venules tend toward higher expression of an assortment of genes involved in inflammatory-related tasks. Fuller appreciation of such heterogeneity will be critical for efficient therapeutic targeting of the endothelium and the management of CNS disease.
Collapse
Affiliation(s)
- Jennifer A Macdonald
- Blood-Brain Barrier Laboratory, Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 07070, USA
| | | | | |
Collapse
|