1
|
Sarallah R, Jahani S, Soltani Khaboushan A, Moaveni AK, Amiri M, Majidi Zolbin M. The role of CXCL12/CXCR4/CXCR7 axis in cognitive impairment associated with neurodegenerative diseases. Brain Behav Immun Health 2025; 43:100932. [PMID: 39834554 PMCID: PMC11743895 DOI: 10.1016/j.bbih.2024.100932] [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: 08/11/2024] [Revised: 12/03/2024] [Accepted: 12/21/2024] [Indexed: 01/22/2025] Open
Abstract
Neurodegenerative diseases, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Multiple Sclerosis (MS), and Amyotrophic Lateral Sclerosis (ALS), are characterized by progressive neuronal loss and cognitive impairment (CI). The: Cysteine-X-cysteine chemokine ligand 12(CXCL12)/CXC chemokine receptor type 4 (CXCR4)/CXC chemokine receptor type 7 (CXCR7) axis has emerged as a critical molecular pathway in the development of CI in these disorders. This review explores the role of this axis in the pathogenesis of CI across these neurodegenerative diseases, synthesizing current evidence and its implications for targeted therapies. In AD, dysregulation of this axis contributes to amyloid-β accumulation and tau hyperphosphorylation, leading to synaptic dysfunction and cognitive decline. PD studies reveal that CXCL12/CXCR4 signaling influences dopaminergic neuron survival and microglial activation, affecting cognitive function. In MS, the axis modulates neuroinflammation and demyelination processes, impacting cognitive performance. ALS research indicates that the CXCL12/CXCR4/CXCR7 pathway is involved in motor neuron degeneration and associated cognitive deficits. Across these diseases, the axis influences neuroinflammation, synaptic plasticity, and neuronal survival through various signaling cascades, including PI3K/AKT, MAPK, and JAK/STAT pathways. Emerging evidence suggests that modulating this axis could provide neuroprotective effects and potentially alleviate cognitive symptoms. This review highlights the potential of the CXCL12/CXCR4/CXCR7 axis as a therapeutic target for addressing CI in neurodegenerative diseases. It also underscores the need for further research to fully elucidate its role and develop effective interventions, potentially leading to improved clinical management strategies for these devastating disorders.
Collapse
Affiliation(s)
| | - Shima Jahani
- MS Research Center Neuroscience Institute, Tehran University of Medical Science, Iran
| | - Alireza Soltani Khaboushan
- Pediatric and Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Medical Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Kian Moaveni
- Pediatric and Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Medical Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Amiri
- Pediatric and Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Medical Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoumeh Majidi Zolbin
- Pediatric and Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children Medical Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Feng F, Li X, Wang W, Dou M, Li S, Jin X, Chu Y, Zhu L. Matrine protects against experimental autoimmune encephalomyelitis through modulating microglial ferroptosis. Biochem Biophys Res Commun 2024; 735:150651. [PMID: 39260333 DOI: 10.1016/j.bbrc.2024.150651] [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: 07/07/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelination neurodegenerative disease of the central nervous system (CNS). Ferroptosis has been implicated in a range of brain disorders, and iron-loaded microglia are frequently found in affected brain regions. However, the molecular mechanisms linking ferroptosis with MS have not been well-defined. The present study seeks to bridge this gap and investigate the impact of matrine (MAT), a herbal medicine with immunomodulatory capacities, on the regulation of oxidative stress and ferroptosis in the CNS of mice with experimental autoimmune encephalomyelitis (EAE), an animal model of MS. CNS of EAE mice contained elevated levels of ferroptosis-related molecules, e.g., MDA, LPCAT3 and PTGS2, but decreased expression of antioxidant molecules, including GSH and SOD, GPX4 and SLC7A11. This pathogenic process was reversed by MAT treatment, together with significant reduction of disease severity and CNS inflammatory demyelination. Furthermore, the expression of PTGS2 and LOX was largely increased in microglia of EAE mice, accompanied with increased production of IL-6 and TNF-α, indicating a proinflammatory phenotype of microglia that undergo oxidative stress/ferroptosis, and their expression was significantly reduced after MAT treatment. Together, our results indicate that ferroptosis/inflammation plays an important role in the pathogenesis of CNS autoimmunity, and inhibiting ferroptosis-induced microglial activation/inflammation could be a novel mechanism underlying the therapeutic effects of MAT on CNS inflammatory demyelination in EAE.
Collapse
Affiliation(s)
- Furui Feng
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xinyu Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Wenbin Wang
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Mengmeng Dou
- Department of Integrated Traditional and Western Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Silu Li
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Xin Jin
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Yaojuan Chu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Lin Zhu
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| |
Collapse
|
3
|
Grandhi TSP, Mebrahtu M, Musso R, Fullman A, Nifong B, Wisdom K, Roh TT, Sender M, Poore D, Macdougall CE, Oren R, Griffin S, Cheng AT, Ekert JE. A microphysiological assay for studying T-cell chemotaxis, trafficking and tumor killing. Biofabrication 2024; 17:015004. [PMID: 39378897 DOI: 10.1088/1758-5090/ad847f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 10/08/2024] [Indexed: 10/10/2024]
Abstract
Tumors in patients non-responsive to immunotherapy harbor a series of barriers that impede the efficacy of effector T-cells. Consequently, therapeutically modulating the chemotaxis machinery to enable effector T cell infiltration and function in the tumor could result in more successful therapeutic outcomes. Complexin-vitromodels allow re-creation ofin-vivotumor complexities in anin-vitrosetting, allowing improved translatability to patient biology at the laboratory scale. We identified a gap in available industrial scale microphysiological (MPS) assays for faster validation of targets and strategies that enable T-cell chemotaxis and effector function within tumor microenvironments. Using a commercially available, 96-chip 2-lane microfluidic assay system, we present a novel, scalable, complexin vitroMPS assay to study 3D T-cell chemotaxis and function within native, extracellular matrix (ECM)-rich multicellular tumor environments. Activated or naïve CD3+ T-cells stained with far-red nuclear stain responded to the chemokine gradients generated within the matrigel-collagen ECM by migrating into the microfluidic channel (∼5 mm horizontal window), in a concentration- and cell type-dependent manner. Furthermore, we observed and tracked chemotaxis and cancer cell killing function of antigen-specific CD4.CD8. chimeric antigen receptor (CAR)-T cells that responded to CXCR3 agonist gradient built through the expansive 5 mm of cancer cell colony containing stroma. The 2-lane assay system yielded useful information regarding donor and dose-dependent differences in CAR-T cell chemotaxis and tumor killing. The scalable assay system allows a granular window into immune cell migration and function in tissue spaces beyond endothelium, addressing a missing gap in studying tissue-specific immune cell chemotaxis and function to bring forward advancements in cancer immunotherapy.
Collapse
Affiliation(s)
| | - Makda Mebrahtu
- Genome Biology, GSK, Collegeville, PA, United States of America
| | - Ryan Musso
- Genome Biology, GSK, Collegeville, PA, United States of America
| | - Alexis Fullman
- Genome Biology, GSK, Collegeville, PA, United States of America
| | - Brady Nifong
- Research Statistics, GSK, Collegeville, PA, United States of America
| | - Katrina Wisdom
- Complex In-Vitro Models, GSK, Collegeville, PA, United States of America
| | - Terrence T Roh
- Complex In-Vitro Models, GSK, Collegeville, PA, United States of America
| | - Matthew Sender
- Chemical Biology, GSK, Collegeville, PA, United States of America
| | - Derek Poore
- Immuno-Oncology and Combinations (IOC), GSK, Collegeville, PA, United States of America
| | | | - Ravit Oren
- Oncology Cell Therapy, GSK, Stevenage, United Kingdom
| | - Sue Griffin
- Oncology Translational Research, GSK, Stevenage, United Kingdom
| | - Aaron T Cheng
- Genome Biology, GSK, Collegeville, PA, United States of America
| | - Jason E Ekert
- Complex In-Vitro Models, GSK, Collegeville, PA, United States of America
| |
Collapse
|
4
|
Pouzol L, Sassi A, Tunis M, Zurbach A, Baumlin N, Gnerre C, Strasser DS, Marrie J, Vezzali E, Martinic MM. ACKR3 Antagonism Enhances the Repair of Demyelinated Lesions Through Both Immunomodulatory and Remyelinating Effects. Neurochem Res 2024; 49:2087-2104. [PMID: 38819698 PMCID: PMC11233362 DOI: 10.1007/s11064-024-04173-1] [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: 01/27/2024] [Revised: 04/16/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Addressing inflammation, demyelination, and associated neurodegeneration in inflammatory demyelinating diseases like multiple sclerosis (MS) remains challenging. ACT-1004-1239, a first-in-class and potent ACKR3 antagonist, currently undergoing clinical development, showed promise in preclinical MS models, reducing neuroinflammation and demyelination. However, its effectiveness in treating established disease and impact on remyelination after the occurrence of demyelinated lesions remain unexplored. This study assessed the therapeutic effect of ACT-1004-1239 in two demyelinating disease models. In the proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE) model, ACT-1004-1239 administered upon the detection of the first signs of paralysis, resulted in a dose-dependent reduction in EAE disease severity, concomitant with diminished immune cell infiltrates in the CNS and reduced demyelination. Notably, efficacy correlated with elevated plasma concentrations of CXCL11 and CXCL12, two pharmacodynamic biomarkers of ACKR3 antagonism. Combining ACT-1004-1239 with siponimod, an approved immunomodulatory treatment for MS, synergistically reduced EAE severity. In the cuprizone-induced demyelination model, ACT-1004-1239 administered after 5 weeks of cuprizone exposure, significantly accelerated remyelination, already quantifiable one week after cuprizone withdrawal. Additionally, ACT-1004-1239 penetrated the CNS, elevating brain CXCL12 concentrations. These results demonstrate that ACKR3 antagonism significantly reduces the severity of experimental demyelinating diseases, even when treatment is initiated therapeutically, after the occurrence of lesions. It confirms the dual mode of action of ACT-1004-1239, exhibiting both immunomodulatory effects by reducing neuroinflammation and promyelinating effects by accelerating myelin repair. The results further strengthen the rationale for evaluating ACT-1004-1239 in clinical trials for patients with demyelinating diseases.
Collapse
Affiliation(s)
- Laetitia Pouzol
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland.
| | - Anna Sassi
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Mélanie Tunis
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Anaïs Zurbach
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Nadège Baumlin
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Carmela Gnerre
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Daniel S Strasser
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Julia Marrie
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Enrico Vezzali
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Marianne M Martinic
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| |
Collapse
|
5
|
Changes in DNA methylation in APOE and ACKR3 genes in multiple sclerosis patients and the relationship with their heavy metal blood levels. Neurotoxicology 2021; 87:182-187. [PMID: 34624384 DOI: 10.1016/j.neuro.2021.09.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 09/12/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease with demyelinated lesions in the central nervous system caused by genetic and environmental factors. DNA methylation as an epigenetic change influenced by environmental factors, including heavy metals has been implemented in MS disease. We investigated the correlation of DNA methylation changes in APOE and ACKR3 genes in MS patients and the possible association with blood concentration of arsenic (As), cadmium (Cd) and lead (Pb) as major heavy metal pollutants. This study included 69 relapsing-remitting multiple sclerosis (RR-MS) patients and 69 age/gender-matched healthy subjects. The HRM real-time PCR method was used to investigate the changes in DNA methylation and heavy metal concentrations were measured by electrothermal atomic absorption spectrometry. Our results showed that the methylation pattern in the ACKR3 gene of the patient group was more hypomethylated, while in the case of the APOE gene, this pattern was more towards hypermethylation compared to healthy subjects. Moreover, the blood levels of As and Cd metals, but not Pb, were significantly higher in the patient group compare to the control group (p ≤ 0.05). The data indicate that the increase in expression of ACKR3 gene by hypomethylation and the decrease in expression of APOE gene via hypermethylation are possibly involved in the onset and progression of inflammatory processes in MS patients. The level of As can also lead to hypomethylation by disrupting the methylation patterns of the ACKR3 gene, resulting in increased expression in MS patients. Finally, we have shown that epigenetic changes can be an important factor in increasing and decreasing the expression of genes involved in the onset and/or progression of inflammatory processes in MS. Furthermore, exposure to heavy metals, especially As, by changing the natural patterns of DNA methylation can be effective in this disease.
Collapse
|
6
|
Pouzol L, Baumlin N, Sassi A, Tunis M, Marrie J, Vezzali E, Farine H, Mentzel U, Martinic MM. ACT-1004-1239, a first-in-class CXCR7 antagonist with both immunomodulatory and promyelinating effects for the treatment of inflammatory demyelinating diseases. FASEB J 2021; 35:e21431. [PMID: 33595155 DOI: 10.1096/fj.202002465r] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 12/26/2022]
Abstract
Current strategies for the treatment of demyelinating diseases such as multiple sclerosis (MS) are based on anti-inflammatory or immunomodulatory drugs. Those drugs have the potential to reduce the frequency of new lesions but do not directly promote remyelination in the damaged central nervous system (CNS). Targeting CXCR7 (ACKR3) has been postulated as a potential therapeutic approach in demyelinating diseases, leading to both immunomodulation by reducing leukocyte infiltrates and promyelination by enhancing myelin repair. ACT-1004-1239 is a potent, selective, insurmountable, and orally available first-in-class CXCR7 receptor antagonist. The effect of ACT-1004-1239 was evaluated in the myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) and the cuprizone-induced demyelination mouse models. In addition, ACT-1004-1239 was assessed in a rat oligodendrocyte precursor cell (OPC) differentiation assay in vitro. In the MOG-induced EAE model, ACT-1004-1239 treatment (10-100 mg/kg, twice daily, orally) showed a significant dose-dependent reduction in disease clinical scores, resulting in increased survival. At the highest dose tested (100 mg/kg, twice daily), ACT-1004-1239 delayed disease onset and significantly reduced immune cell infiltrates into the CNS and plasma neurofilament light chain concentration. Treatment with ACT-1004-1239 dose-dependently increased plasma CXCL12 concentration, which correlated with a reduction of the cumulative disease score. Furthermore, in the cuprizone model, ACT-1004-1239 treatment significantly increased the number of mature myelinating oligodendrocytes and enhanced myelination in vivo. In vitro, ACT-1004-1239 promoted the maturation of OPCs into myelinating oligodendrocytes. These results provide evidence that ACT-1004-1239 both reduces neuroinflammation and enhances myelin repair substantiating the rationale to explore its therapeutic potential in a clinical setting.
Collapse
Affiliation(s)
| | | | - Anna Sassi
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Mélanie Tunis
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | - Julia Marrie
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | - Hervé Farine
- Idorsia Pharmaceuticals Ltd., Allschwil, Switzerland
| | | | | |
Collapse
|
7
|
Richard-Bildstein S, Aissaoui H, Pothier J, Schäfer G, Gnerre C, Lindenberg E, Lehembre F, Pouzol L, Guerry P. Discovery of the Potent, Selective, Orally Available CXCR7 Antagonist ACT-1004-1239. J Med Chem 2020; 63:15864-15882. [PMID: 33314938 DOI: 10.1021/acs.jmedchem.0c01588] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The chemokine receptor CXCR7, also known as ACKR3, is a seven-transmembrane G-protein-coupled receptor (GPCR) involved in various pathologies such as neurological diseases, autoimmune diseases, and cancers. By binding and scavenging the chemokines CXCL11 and CXCL12, CXCR7 regulates their extracellular levels. From an original high-throughput screening campaign emerged hit 3 among others. The hit-to-lead optimization led to the discovery of a novel chemotype series exemplified by the trans racemic compound 11i. This series provided CXCR7 antagonists that block CXCL11- and CXCL12-induced ß-arrestin recruitment. Further structural modifications on the trisubstituted piperidine scaffold of 11i yielded compounds with high CXCR7 antagonistic activities and balanced ADMET properties. The effort described herein culminated in the discovery of ACT-1004-1239 (28f). Biological characterization of ACT-1004-1239 demonstrated that it is a potent, insurmountable antagonist. Oral administration of ACT-1004-1239 in mice up to 100 mg/kg led to a dose-dependent increase of plasma CXCL12 concentration.
Collapse
Affiliation(s)
- Sylvia Richard-Bildstein
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| | - Hamed Aissaoui
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| | - Julien Pothier
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| | - Gabriel Schäfer
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| | - Carmela Gnerre
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| | - Eleanor Lindenberg
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| | - François Lehembre
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| | - Laetitia Pouzol
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| | - Philippe Guerry
- Drug Discovery, Idorsia Pharmaceuticals Ltd., Hegenheimermattweg 91, Allschwil CH-4123, Switzerland
| |
Collapse
|
8
|
CXCR7 suppression modulates macrophage phenotype and function to ameliorate post-myocardial infarction injury. Inflamm Res 2020; 69:523-532. [DOI: 10.1007/s00011-020-01335-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 02/25/2020] [Accepted: 03/07/2020] [Indexed: 12/22/2022] Open
|
9
|
Lounsbury N. Advances in CXCR7 Modulators. Pharmaceuticals (Basel) 2020; 13:ph13020033. [PMID: 32098047 PMCID: PMC7169404 DOI: 10.3390/ph13020033] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/17/2020] [Accepted: 02/19/2020] [Indexed: 12/16/2022] Open
Abstract
CXC chemokine receptor 7 (CXCR7) is a G-protein-coupled receptor that signals through the β-arrestin pathway. Its ligands include interferon-inducible T cell α chemoattractant (CXCL11) and stromal cell-derived factor-1 (CXCL12). It interacts with CXCR4, and the two are associated with various cancers, as well as other disease states such as coronary artery disease, stroke, inflammation and human immunodeficiency virus (HIV). Antibodies and small interfering RNA (siRNA) have shown the utility of antagonists of CXCR7 in these disease states. Although some small molecules were initially reported as antagonists due to their displayed activity, many function as agonists while still producing the desired pharmacologic effects. A potential reason for this contradiction is that effects may be due to elevated extracellular CXCL12 levels.
Collapse
Affiliation(s)
- Nicole Lounsbury
- Department of Pharmaceutical Sciences, Larkin University College of Pharmacy, Miami, FL 33169, USA
| |
Collapse
|
10
|
He W, Kapate N, Shields CW, Mitragotri S. Drug delivery to macrophages: A review of targeting drugs and drug carriers to macrophages for inflammatory diseases. Adv Drug Deliv Rev 2019; 165-166:15-40. [PMID: 31816357 DOI: 10.1016/j.addr.2019.12.001] [Citation(s) in RCA: 154] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 11/28/2019] [Accepted: 12/04/2019] [Indexed: 12/16/2022]
Abstract
Macrophages play a key role in defending against foreign pathogens, healing wounds, and regulating tissue homeostasis. Driving this versatility is their phenotypic plasticity, which enables macrophages to respond to subtle cues in tightly coordinated ways. However, when this coordination is disrupted, macrophages can aid the progression of numerous diseases, including cancer, cardiovascular disease, and autoimmune disease. The central link between these disorders is aberrant macrophage polarization, which misguides their functional programs, secretory products, and regulation of the surrounding tissue microenvironment. As a result of their important and deterministic roles in both health and disease, macrophages have gained considerable attention as targets for drug delivery. Here, we discuss the role of macrophages in the initiation and progression of various inflammatory diseases, summarize the leading drugs used to regulate macrophages, and review drug delivery systems designed to target macrophages. We emphasize strategies that are approved for clinical use or are poised for clinical investigation. Finally, we provide a prospectus of the future of macrophage-targeted drug delivery systems.
Collapse
Affiliation(s)
- Wei He
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Neha Kapate
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - C Wyatt Shields
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
11
|
Palani CD, Fouda AY, Liu F, Xu Z, Mohamed E, Giri S, Smith SB, Caldwell RB, Narayanan SP. Deletion of Arginase 2 Ameliorates Retinal Neurodegeneration in a Mouse Model of Multiple Sclerosis. Mol Neurobiol 2019; 56:8589-8602. [PMID: 31280447 DOI: 10.1007/s12035-019-01691-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022]
Abstract
Optic neuritis is a major clinical feature of multiple sclerosis (MS) and can lead to temporary or permanent vision loss. Previous studies from our laboratory have demonstrated the critical involvement of arginase 2 (A2) in retinal neurodegeneration in models of ischemic retinopathy. The current study was undertaken to investigate the role of A2 in MS-mediated retinal neuronal damage and degeneration. Experimental autoimmune encephalomyelitis (EAE) was induced in wild-type (WT) and A2 knockout (A2-/-) mice. EAE-induced motor deficits, loss of retinal ganglion cells, retinal thinning, inflammatory signaling, and glial activation were studied in EAE-treated WT and A2-/- mice and their respective controls. Increased expression of A2 was observed in WT retinas in response to EAE induction. EAE-induced motor deficits were markedly reduced in A2-/- mice compared with WT controls. Retinal flat mount studies demonstrated a significant reduction in the number of RGCs in WT EAE retinas in comparison with normal control mice. A significant improvement in neuronal survival was evident in retinas of EAE-induced A2-/- mice compared with WT. RNA levels of the proinflammatory molecules CCL2, COX2, IL-1α, and IL-12α were significantly reduced in the A2-/- EAE retinas compared with WT EAE. EAE-induced activation of glia (microglia and Müller cells) was markedly reduced in A2-/- retinas compared with WT. Western blot analyses showed increased levels of phospho-ERK1/2 and reduced levels of phospho-BAD in the WT EAE retina, while these changes were prevented in A2-/- mice. In conclusion, our studies establish EAE as an excellent model to study MS-mediated retinal neuronal damage and suggest the potential value of targeting A2 as a therapy to prevent MS-mediated retinal neuronal injury.
Collapse
Affiliation(s)
- Chithra D Palani
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
| | - Abdelrahman Y Fouda
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA
| | - Fang Liu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
| | - Zhimin Xu
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA
| | - Eslam Mohamed
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
- Department of Immunology, Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Shailedra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI, 48202, USA
| | - Sylvia B Smith
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, 30912, USA
| | - Ruth B Caldwell
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, GA, 30912, USA
- Charlie Norwood VA Medical Center, Augusta, GA, 30904, USA
| | - S Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia, Augusta, GA, 30912, USA.
- Culver Vision Discovery Institute, Augusta University, Augusta, GA, 30912, USA.
- Vascular Biology Center, Augusta University, Augusta, GA, 30912, USA.
- Charlie Norwood VA Medical Center, Augusta, GA, 30904, USA.
| |
Collapse
|
12
|
García-Cuesta EM, Santiago CA, Vallejo-Díaz J, Juarranz Y, Rodríguez-Frade JM, Mellado M. The Role of the CXCL12/CXCR4/ACKR3 Axis in Autoimmune Diseases. Front Endocrinol (Lausanne) 2019; 10:585. [PMID: 31507535 PMCID: PMC6718456 DOI: 10.3389/fendo.2019.00585] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 08/09/2019] [Indexed: 12/19/2022] Open
Abstract
Chemokine receptors are members of the G protein-coupled receptor superfamily. These receptors are intimately involved in cell movement, and thus play a critical role in several physiological and pathological situations that require the precise regulation of cell positioning. CXCR4 is one of the most studied chemokine receptors and is involved in many functions beyond leukocyte recruitment. During embryogenesis, it plays essential roles in vascular development, hematopoiesis, cardiogenesis, and nervous system organization. It has been also implicated in tumor progression and autoimmune diseases and, together with CD4, is one of the co-receptors used by the HIV-1 virus to infect immune cells. In contrast to other chemokine receptors that are characterized by ligand promiscuity, CXCR4 has a unique ligand-stromal cell-derived factor-1 (SDF1, CXCL12). However, this ligand also binds ACKR3, an atypical chemokine receptor that modulates CXCR4 functions and is overexpressed in multiple cancer types. The CXCL12/CXCR4/ACKR3 axis constitutes a potential therapeutic target for a wide variety of inflammatory diseases, not only by interfering with cell migration but also by modulating immune responses. Thus far, only one antagonist directed against the ligand-binding site of CXCR4, AMD3100, has demonstrated clinical relevance. Here, we review the role of this ligand and its receptors in different autoimmune diseases.
Collapse
Affiliation(s)
- Eva M. García-Cuesta
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - César A. Santiago
- Macromolecular X-Ray Crystallography Unit, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Jesús Vallejo-Díaz
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
| | - Yasmina Juarranz
- Department Cell Biology, Research Institute Hospital 12 de Octubre (i+12), Complutense University of Madrid, Madrid, Spain
| | | | - Mario Mellado
- Department of Immunology and Oncology, Centro Nacional de Biotecnología/CSIC, Madrid, Spain
- *Correspondence: Mario Mellado
| |
Collapse
|
13
|
Huang Y, Hu S, Li Y, Xue D, Wu X. Dexmedetomidine, an Alpha 2a Adrenergic Receptor Agonist, Mitigates Experimental Autoimmune Encephalomyelitis by Desensitization of CXCR7 in Microglia. Biochemistry 2018; 57:4197-4205. [PMID: 29897736 DOI: 10.1021/acs.biochem.8b00430] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Yi Huang
- Department of Anesthesiology, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People’s Hospital), Ruian, Zhejiang 325200, China
| | - Shite Hu
- Department of Anesthesiology, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People’s Hospital), Ruian, Zhejiang 325200, China
| | - Yongliang Li
- Department of Anesthesiology, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People’s Hospital), Ruian, Zhejiang 325200, China
| | - Dan Xue
- Department of Plastic Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310000, China
| | - Xiuguo Wu
- Department of Anesthesiology, The Third Affiliated Hospital of Wenzhou Medical University (Ruian People’s Hospital), Ruian, Zhejiang 325200, China
| |
Collapse
|
14
|
Silwedel C, Speer CP, Haarmann A, Fehrholz M, Claus H, Buttmann M, Glaser K. Novel insights into neuroinflammation: bacterial lipopolysaccharide, tumor necrosis factor α, and Ureaplasma species differentially modulate atypical chemokine receptor 3 responses in human brain microvascular endothelial cells. J Neuroinflammation 2018; 15:156. [PMID: 29792190 PMCID: PMC5966865 DOI: 10.1186/s12974-018-1170-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/19/2018] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Atypical chemokine receptor 3 (ACKR3, synonym CXCR7) is increasingly considered relevant in neuroinflammatory conditions, in which its upregulation contributes to compromised endothelial barrier function and may ultimately allow inflammatory brain injury. While an impact of ACKR3 has been recognized in several neurological autoimmune diseases, neuroinflammation may also result from infectious agents, including Ureaplasma species (spp.). Although commonly regarded as commensals of the adult urogenital tract, Ureaplasma spp. may cause invasive infections in immunocompromised adults as well as in neonates and appear to be relevant pathogens in neonatal meningitis. Nonetheless, clinical and in vitro data on Ureaplasma-induced inflammation are scarce. METHODS We established a cell culture model of Ureaplasma meningitis, aiming to analyze ACKR3 variances as a possible pathomechanism in Ureaplasma-associated neuroinflammation. Non-immortalized human brain microvascular endothelial cells (HBMEC) were exposed to bacterial lipopolysaccharide (LPS) or tumor necrosis factor-α (TNF-α), and native as well as LPS-primed HBMEC were cultured with Ureaplasma urealyticum serovar 8 (Uu8) and U. parvum serovar 3 (Up3). ACKR3 responses were assessed via qRT-PCR, RNA sequencing, flow cytometry, and immunocytochemistry. RESULTS LPS, TNF-α, and Ureaplasma spp. influenced ACKR3 expression in HBMEC. LPS and TNF-α significantly induced ACKR3 mRNA expression (p < 0.001, vs. control), whereas Ureaplasma spp. enhanced ACKR3 protein expression in HBMEC (p < 0.01, vs. broth control). Co-stimulation with LPS and either Ureaplasma isolate intensified ACKR3 responses (p < 0.05, vs. LPS). Furthermore, stimulation wielded a differential influence on the receptor's ligands. CONCLUSIONS We introduce an in vitro model of Ureaplasma meningitis. We are able to demonstrate a pro-inflammatory capacity of Ureaplasma spp. in native and, even more so, in LPS-primed HBMEC, underlining their clinical relevance particularly in a setting of co-infection. Furthermore, our data may indicate a novel role for ACKR3, with an impact not limited to auto-inflammatory diseases, but extending to infection-related neuroinflammation as well. AKCR3-induced blood-brain barrier breakdown might constitute a potential common pathomechanism.
Collapse
Affiliation(s)
- Christine Silwedel
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany.
| | - Christian P Speer
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Axel Haarmann
- Department of Neurology, University of Wuerzburg, Josef-Schneider-Str. 11, 97080, Wuerzburg, Germany
| | - Markus Fehrholz
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Heike Claus
- Institute for Hygiene and Microbiology, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| | - Mathias Buttmann
- Department of Neurology, University of Wuerzburg, Josef-Schneider-Str. 11, 97080, Wuerzburg, Germany.,Department of Neurology, Caritas Hospital, Uhlandstr. 7, 97980, Bad Mergentheim, Germany
| | - Kirsten Glaser
- University Children's Hospital, University of Wuerzburg, Josef-Schneider-Str. 2, 97080, Wuerzburg, Germany
| |
Collapse
|
15
|
CD271+, CXCR7+, CXCR4+, and CD133+ Stem/Progenitor Cells and Clinical Characteristics of Acute Ischemic Stroke Patients. Neuromolecular Med 2018; 20:301-311. [PMID: 29744773 PMCID: PMC6097064 DOI: 10.1007/s12017-018-8494-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 05/02/2018] [Indexed: 12/11/2022]
Abstract
Ischemic stroke causes mobilization of various groups of progenitor cells from bone marrow to bloodstream and this correlates with the neurological status of stroke patients. The goal of our study was to identify the activity of chosen progenitor/stem cells in the peripheral blood of acute ischemic stroke patients in the first 7 days after the incident, through associations between the levels of the cells and clinical features of the patients. Thirty-three acute ischemic stroke patients and 15 non-stroke control subjects had their venous blood collected repeatedly in order to assess the levels of the CD45–CD34 + CD271+, the CD45–CD34 + CXCR4+, the CD45–CD34 + CXCR7+, and the CD45–CD34 + CD133+ stem/progenitor cells by means of flow cytometry. The patients underwent repeated neurological and clinical assessments, pulse wave velocity (PWV) assessment on day 5, and MRI on day 1 and 5 ± 2. The levels of the CD45–CD34 + CXCR7+ and the CD45–CD34 + CD271+ cells were lower in the stroke patients compared with the control subjects. Only the CD45–CD34 + CD271+ cells correlated positively with lesion volume in the second MRI. The levels of the CD45–CD34 + CD133+ cells on day 2 correlated negatively with PWV and NIHSS score on day 9. The patients whose PWV was above 10 m/s had significantly higher levels of the CD45–CD34 + CXCR4+ and the CD45–CD34 + CXCR7+ cells on day 1 than those with PWV below 10 m/s. This study discovers possible activity of the CD45–CD34 + CD271+ progenitor/stem cells during the first 7 days after ischemic stroke, suggests associations of the CD45–CD34 + CD133+ cells with the neurological status of stroke patients, and some activity of the CD45–CD34 + CD133+, the CD45–CD34 + CXCR4+, and the CD45–CD34 + CXCR7+ progenitor/stem cells in the process of arterial remodeling.
Collapse
|
16
|
Xiao R, Li S, Cao Q, Wang X, Yan Q, Tu X, Zhu Y, Zhu F. Human endogenous retrovirus W env increases nitric oxide production and enhances the migration ability of microglia by regulating the expression of inducible nitric oxide synthase. Virol Sin 2017; 32:216-225. [PMID: 28656540 PMCID: PMC6598877 DOI: 10.1007/s12250-017-3997-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 05/26/2017] [Indexed: 01/21/2023] Open
Abstract
Human endogenous retrovirus W env (HERV-W env) plays a critical role in many neuropsychological diseases such as schizophrenia and multiple sclerosis (MS). These diseases are accompanied by immunological reactions in the central nervous system (CNS). Microglia are important immunocytes in brain inflammation that can produce a gasotransmitter-nitric oxide (NO). NO not only plays a role in the function of neuronal cells but also participates in the pathogenesis of various neuropsychological diseases. In this study, we reported increased NO production in CHME-5 microglia cells after they were transfected with HERV-W env. Moreover, HERV-W env increased the expression and function of human inducible nitric oxide synthase (hiNOS) and enhanced the promoter activity of hiNOS. Microglial migration was also enhanced. These data revealed that HERV-W env might contribute to increase NO production and microglial migration ability in neuropsychological disorders by regulating the expression of inducible NOS. Results from this study might lead to the identification of novel targets for the treatment of neuropsychological diseases, including neuroinflammatory diseases, stroke, and neurodegenerative diseases.
Collapse
Affiliation(s)
- Ran Xiao
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Shan Li
- Department of Integrated Medicine, Dongfeng Hospital, Hubei University of Medicine, Wuhan, 442000, China
| | - Qian Cao
- Department of Neurology Medicine, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Xiuling Wang
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Qiujin Yan
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Xiaoning Tu
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan, 430071, China
| | - Ying Zhu
- The State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Fan Zhu
- Department of Medical Microbiology, School of Medicine, Wuhan University, Wuhan, 430071, China.
- Hubei Province Key Laboratory of Allergy and Immunology, Wuhan, 430071, China.
| |
Collapse
|
17
|
Salvi V, Sozio F, Sozzani S, Del Prete A. Role of Atypical Chemokine Receptors in Microglial Activation and Polarization. Front Aging Neurosci 2017; 9:148. [PMID: 28603493 PMCID: PMC5445112 DOI: 10.3389/fnagi.2017.00148] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2017] [Accepted: 05/02/2017] [Indexed: 01/07/2023] Open
Abstract
Inflammatory reactions occurring in the central nervous system (CNS), known as neuroinflammation, are key components of the pathogenic mechanisms underlying several neurological diseases. The chemokine system plays a crucial role in the recruitment and activation of immune and non-immune cells in the brain, as well as in the regulation of microglia phenotype and function. Chemokines belong to a heterogeneous family of chemotactic agonists that signal through the interaction with G protein-coupled receptors (GPCRs). Recently, a small subset of chemokine receptors, now identified as “atypical chemokine receptors” (ACKRs), has been described. These receptors lack classic GPCR signaling and chemotactic activity and are believed to limit inflammation through their ability to scavenge chemokines at the inflammatory sites. Recent studies have highlighted a role for ACKRs in neuroinflammation. However, in the CNS, the role of ACKRs seems to be more complex than the simple control of inflammation. For instance, CXCR7/ACKR3 was shown to control T cell trafficking through the regulation of CXCL12 internalization at CNS endothelial barriers. Furthermore, D6/ACKR2 KO mice were protected in a model of experimental autoimmune encephalomyelitis (EAE). D6/ACKR2 KO showed an abnormal accumulation of dendritic cells at the immunization and a subsequent impairment in T cell priming. Finally, CCRL2, an ACKR-related protein, was shown to play a role in the control of the resolution phase of EAE. Indeed, CCRL2 KO mice showed exacerbated, non-resolving disease with protracted inflammation and increased demyelination. This phenotype was associated with increased microglia and macrophage activation markers and imbalanced M1 vs. M2 polarization. This review will summarize the current knowledge on the role of the ACKRs in neuroinflammation with a particular attention to their role in microglial polarization and function.
Collapse
Affiliation(s)
- Valentina Salvi
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy
| | - Francesca Sozio
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy.,IRCCS-Humanitas Clinical and Research CenterRozzano-Milano, Italy
| | - Silvano Sozzani
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy.,IRCCS-Humanitas Clinical and Research CenterRozzano-Milano, Italy
| | - Annalisa Del Prete
- Department of Molecular and Translational Medicine, University of BresciaBrescia, Italy.,IRCCS-Humanitas Clinical and Research CenterRozzano-Milano, Italy
| |
Collapse
|
18
|
Korean Red Ginseng mitigates spinal demyelination in a model of acute multiple sclerosis by downregulating p38 mitogen-activated protein kinase and nuclear factor-κB signaling pathways. J Ginseng Res 2017; 42:436-446. [PMID: 30337803 PMCID: PMC6187097 DOI: 10.1016/j.jgr.2017.04.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 04/12/2017] [Accepted: 04/26/2017] [Indexed: 01/08/2023] Open
Abstract
Background The potential therapeutic values of Korean Red Ginseng extract (KRGE) in autoimmune disorders of nervous system have not been fully investigated. Methods We used an acute experimental autoimmune encephalomyelitis animal model of multiple sclerosis and determined the effects and mechanism of KRGE on spinal myelination. Results Pretreatment with KRGE (100 mg/kg, orally) for 10 days before immunization with myelin basic protein (MBP)68-82 peptide exerted a protective effect against demyelination in the spinal cord, with inhibited recruitment and activation of immune cells including microglia, decreased mRNA expression of detrimental inflammatory mediators (interleukin-6, interferon-γ, and cyclooxygenase-2), but increased mRNA expression of protective inflammatory mediators (insulin-like growth factor β1, transforming growth factor β, and vascular endothelial growth factor-1). These results were associated with significant downregulation of p38 mitogen-activated protein kinase and nuclear factor-κB signaling pathways in microglia/macrophages, T cells, and astrocytes. Conclusion Our findings suggest that KRGE alleviates spinal demyelination in acute experimental autoimmune encephalomyelitis through inhibiting the activation of the p38 mitogen-activated protein kinase/nuclear factor-κB signaling pathway. Therefore, KRGE might be used as a new therapeutic for autoimmune disorders such as multiple sclerosis, although further investigation is needed.
Collapse
|
19
|
Peng H, Zhang H, Zhu H. Blocking CXCR7-mediated adipose tissue macrophages chemotaxis attenuates insulin resistance and inflammation in obesity. Biochem Biophys Res Commun 2016; 479:649-655. [PMID: 27693695 DOI: 10.1016/j.bbrc.2016.09.158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 09/28/2016] [Indexed: 11/27/2022]
Abstract
Adipose tissue macrophages (ATMs) have been considered to have a pivotal role in the chronic inflammation development during obesity. Although chemokine-chemokine receptor interaction has been studied in ATMs infiltration, most chemokine receptors remain incompletely understood and little is known about their mechanism of actions that lead to ATMs chemotaxis and pathogenesis of insulin resistance during obesity. In this study, we reported that CXCR7 expression is upregulated in adipose tissue, and specifically in ATMs during obesity. In addition, CXCL11 or CXCL12-induced ATMs chemotaxis is mediated by CXCR7 in obesity but not leanness, whereas CXCR3 and CXCR4 are not involved. Additional mechanism study shows that NF-κB activation is essential in ATMs chemotaxis, and manipulates chemotaxis of ATMs via CXCR7 expression regulation in obesity. Most importantly, CXCR7 neutralizing therapy dose dependently leads to less infiltration of macrophages into adipose tissue and thus reduces inflammation and improves insulin sensitivity in obesity. In conclusion, these findings demonstrated that blocking CXCR7-mediated ATMs chemotaxis ameliorates insulin resistance and inflammation in obesity.
Collapse
Affiliation(s)
- Hongxia Peng
- Department of Endocrinology, Shangqiu First People's Hospital, Shangqiu 476100, China.
| | - Hu Zhang
- Department of Endocrinology, Shangqiu First People's Hospital, Shangqiu 476100, China
| | - Honglei Zhu
- Health Center of Chengguan Town, Yucheng, Shangqiu 476300, China
| |
Collapse
|
20
|
Double Roles of Macrophages in Human Neuroimmune Diseases and Their Animal Models. Mediators Inflamm 2016; 2016:8489251. [PMID: 27034594 PMCID: PMC4808549 DOI: 10.1155/2016/8489251] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2015] [Revised: 02/21/2016] [Accepted: 02/23/2016] [Indexed: 12/22/2022] Open
Abstract
Macrophages are important immune cells of the innate immune system that are involved in organ-specific homeostasis and contribute to both pathology and resolution of diseases including infections, cancer, obesity, atherosclerosis, and autoimmune disorders. Multiple lines of evidence point to macrophages as a remarkably heterogeneous cell type. Different phenotypes of macrophages exert either proinflammatory or anti-inflammatory roles depending on the cytokines and other mediators that they are exposed to in the local microenvironment. Proinflammatory macrophages secrete detrimental molecules to induce disease development, while anti-inflammatory macrophages produce beneficial mediators to promote disease recovery. The conversion of the phenotypes of macrophages can regulate the initiation, development, and recovery of autoimmune diseases. Human neuroimmune diseases majorly include multiple sclerosis (MS), neuromyelitis optica (NMO), myasthenia gravis (MG), and Guillain-Barré syndrome (GBS) and macrophages contribute to the pathogenesis of these neuroimmune diseases. In this review, we summarize the double roles of macrophage in neuroimmune diseases and their animal models to further explore the mechanisms of macrophages involved in the pathogenesis of these disorders, which may provide a potential therapeutic approach for these disorders in the future.
Collapse
|