1
|
Shui X, Chen J, Fu Z, Zhu H, Tao H, Li Z. Microglia in Ischemic Stroke: Pathogenesis Insights and Therapeutic Challenges. J Inflamm Res 2024; 17:3335-3352. [PMID: 38800598 PMCID: PMC11128258 DOI: 10.2147/jir.s461795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Ischemic stroke is the most common type of stroke, which is the main cause of death and disability on a global scale. As the primary immune cells in the brain that are crucial for preserving homeostasis of the central nervous system microenvironment, microglia have been found to exhibit dual or even multiple effects at different stages of ischemic stroke. The anti-inflammatory polarization of microglia and release of neurotrophic factors may provide benefits by promoting neurological recovery at the lesion in the early phase after ischemic stroke. However, the pro-inflammatory polarization of microglia and secretion of inflammatory factors in the later phase of injury may exacerbate the ischemic lesion, suggesting the therapeutic potential of modulating the balance of microglial polarization to predispose them to anti-inflammatory transformation in ischemic stroke. Microglia-mediated signaling crosstalk with other cells may also be key to improving functional outcomes following ischemic stroke. Thus, this review provides an overview of microglial functions and responses under physiological and ischemic stroke conditions, including microglial activation, polarization, and interactions with other cells. We focus on approaches that promote anti-inflammatory polarization of microglia, inhibit microglial activation, and enhance beneficial cell-to-cell interactions. These targets may hold promise for the creation of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Xinyao Shui
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Jingsong Chen
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Ziyue Fu
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Haoyue Zhu
- Clinical Medical College, Southwest Medical University, Luzhou, People’s Republic of China
| | - Hualin Tao
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| | - Zhaoyinqian Li
- Department of Laboratory Medicine, the Affiliated Hospital of Southwest Medical University, Luzhou, People’s Republic of China
- Sichuan Province Engineering Technology Research Center of Molecular Diagnosis of Clinical Diseases, Luzhou, People’s Republic of China
- Molecular Diagnosis of Clinical Diseases Key Laboratory of Luzhou, Luzhou, People’s Republic of China
| |
Collapse
|
2
|
Namba MD, Xie Q, Park K, Jackson JG, Barker JM. EcoHIV Infection Modulates the Effects of Cocaine Exposure Pattern and Abstinence on Cocaine Seeking and Neuroimmune Protein Expression in Male Mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589615. [PMID: 38659915 PMCID: PMC11042347 DOI: 10.1101/2024.04.15.589615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Cocaine use disorders (CUDs) and human immunodeficiency virus (HIV) remain persistent public health dilemmas throughout the world. One major hurdle for treating CUD is the increase in cocaine craving and seeking behavior that occurs over a protracted period of abstinence, an effect known as the incubation of craving. Little is known about how HIV may modulate this process. Thus, we sought to examine the impact of chronic HIV infection on the incubation of cocaine craving and associated changes in the central and peripheral immune systems. Here, mice were inoculated with EcoHIV, which is a chimeric HIV-1 construct that produces chronic HIV infection in mice. EcoHIV- and sham-infected mice were conditioned with cocaine daily or intermittently in a conditioned place preference (CPP) paradigm, followed by 1 or 21 days of forced abstinence prior to assessing preference for the cocaine-paired chamber. Under both conditioning regimens, sham mice exhibited incubation of cocaine CPP after 21 days of abstinence. EcoHIV-infected mice conditioned daily with cocaine showed enhanced cocaine seeking at both abstinence timepoints, whereas infected mice conditioned intermittently showed a reversal of the incubation effect, with higher cocaine seeking after 1 day of abstinence compared to 21 days. Analysis of corticolimbic CX3CL1-CX3CR1 and glutamate receptor expression revealed alterations in medial prefrontal cortex (mPFC) CX3CL1 and nucleus accumbens (NAc) GluN2A receptors that correlated with cocaine seeking following daily cocaine exposure. Moreover, examination of peripheral immune markers showed that the effect of abstinence and EcoHIV infection on these measures depended on the cocaine exposure regimen. Altogether, these results highlight the importance of cocaine abstinence and exposure pattern as critical variables that modulate HIV-associated neuroimmune outcomes and relapse vulnerability.
Collapse
Affiliation(s)
- Mark D. Namba
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Qiaowei Xie
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
- Graduate Program in Pharmacology and Physiology, College of Medicine, Drexel University
| | - Kyewon Park
- Center for AIDS Research (CFAR), University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua G. Jackson
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| | - Jacqueline M. Barker
- Department of Pharmacology & Physiology, College of Medicine, Drexel University, Philadelphia, PA, USA
| |
Collapse
|
3
|
Zhang C, Zhang Y, Zhuang R, Yang K, Chen L, Jin B, Ma Y, Zhang Y, Tang K. Alterations in CX3CL1 Levels and Its Role in Viral Pathogenesis. Int J Mol Sci 2024; 25:4451. [PMID: 38674036 PMCID: PMC11050295 DOI: 10.3390/ijms25084451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/12/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
CX3CL1, also named fractalkine or neurotactin, is the only known member of the CX3C chemokine family that can chemoattract several immune cells. CX3CL1 exists in both membrane-anchored and soluble forms, with each mediating distinct biological activities. CX3CL1 signals are transmitted through its unique receptor, CX3CR1, primarily expressed in the microglia of the central nervous system (CNS). In the CNS, CX3CL1 acts as a regulator of microglia activation in response to brain disorders or inflammation. Recently, there has been a growing interest in the role of CX3CL1 in regulating cell adhesion, chemotaxis, and host immune response in viral infection. Here, we provide a comprehensive review of the changes and function of CX3CL1 in various viral infections, such as human immunodeficiency virus (HIV), SARS-CoV-2, influenza virus, and cytomegalovirus (CMV) infection, to highlight the emerging roles of CX3CL1 in viral infection and associated diseases.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yun Zhang
- Department of Immunology, The Fourth Military Medical University, Xi’an 710032, China; (C.Z.); (Y.Z.); (R.Z.); (K.Y.); (L.C.); (B.J.); (Y.M.)
| | - Kang Tang
- Department of Immunology, The Fourth Military Medical University, Xi’an 710032, China; (C.Z.); (Y.Z.); (R.Z.); (K.Y.); (L.C.); (B.J.); (Y.M.)
| |
Collapse
|
4
|
Yang Y, Zhao X, Zhu Z, Zhang L. Vascular dementia: A microglia's perspective. Ageing Res Rev 2022; 81:101734. [PMID: 36113763 DOI: 10.1016/j.arr.2022.101734] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 08/28/2022] [Accepted: 09/11/2022] [Indexed: 01/31/2023]
Abstract
Vascular dementia (VaD) is a second most common form of age-related dementia. It is characterized by cognitive impairment associated with vascular pathology, symptoms mainly caused by cerebral damage due to inadequate blood flow to the brain. The pathogenesis of VaD is complex, and a growing body of literature emphasizes on the involvement of microglia in disease development and progression. Here, we review the current knowledge on the role of microglia in regulating neuroinflammation under the pathogenesis of VaD. The commonly used animal and cell models for understanding the disease pathogenesis were summarized. The mechanisms by which microglia contribute to VaD are multifactorial, and we specifically focus on some of the predominant functions of microglia, including chemotaxis, secretory property, phagocytosis, and its crosstalk with other neurovascular unit cells. Finally, potential therapeutic strategies targeting microglia-modulated neuroinflammation are discussed.
Collapse
Affiliation(s)
- Yi Yang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China.
| | - Xinyuan Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Zirui Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China
| | - Lihui Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou 311121, China; Hangzhou Key Laboratory of Medical Neurobiology, Hangzhou Normal University, Hangzhou 311121, China; Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou 311121, China.
| |
Collapse
|
5
|
Daskou M, Mu W, Sharma M, Vasilopoulos H, Heymans R, Ritou E, Rezek V, Hamid P, Kossyvakis A, Sen Roy S, Grijalva V, Chattopadhyay A, Kitchen SG, Fogelman AM, Reddy ST, Kelesidis T. ApoA-I mimetics reduce systemic and gut inflammation in chronic treated HIV. PLoS Pathog 2022; 18:e1010160. [PMID: 34995311 PMCID: PMC8740974 DOI: 10.1371/journal.ppat.1010160] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022] Open
Abstract
Novel therapeutic strategies are needed to attenuate increased systemic and gut inflammation that contribute to morbidity and mortality in chronic HIV infection despite potent antiretroviral therapy (ART). The goal of this study is to use preclinical models of chronic treated HIV to determine whether the antioxidant and anti-inflammatory apoA-I mimetic peptides 6F and 4F attenuate systemic and gut inflammation in chronic HIV. We used two humanized murine models of HIV infection and gut explants from 10 uninfected and 10 HIV infected persons on potent ART, to determine the in vivo and ex vivo impact of apoA-I mimetics on systemic and intestinal inflammation in HIV. When compared to HIV infected humanized mice treated with ART alone, mice on oral apoA-I mimetic peptide 6F with ART had consistently reduced plasma and gut tissue cytokines (TNF-α, IL-6) and chemokines (CX3CL1) that are products of ADAM17 sheddase activity. Oral 6F attenuated gut protein levels of ADAM17 that were increased in HIV-1 infected mice on potent ART compared to uninfected mice. Adding oxidized lipoproteins and endotoxin (LPS) ex vivo to gut explants from HIV infected persons increased levels of ADAM17 in myeloid and intestinal cells, which increased TNF-α and CX3CL1. Both 4F and 6F attenuated these changes. Our preclinical data suggest that apoA-I mimetic peptides provide a novel therapeutic strategy that can target increased protein levels of ADAM17 and its sheddase activity that contribute to intestinal and systemic inflammation in treated HIV. The large repertoire of inflammatory mediators involved in ADAM17 sheddase activity places it as a pivotal orchestrator of several inflammatory pathways associated with morbidity in chronic treated HIV that make it an attractive therapeutic target.
Collapse
Affiliation(s)
- Maria Daskou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - William Mu
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Madhav Sharma
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Hariclea Vasilopoulos
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Rachel Heymans
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Eleni Ritou
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Valerie Rezek
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Philip Hamid
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Athanasios Kossyvakis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Shubhendu Sen Roy
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Victor Grijalva
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Arnab Chattopadhyay
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Scott G. Kitchen
- Department of Medicine, Division of Hematology and Oncology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Alan M. Fogelman
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Srinivasa T. Reddy
- Department of Medicine, Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, California, United States of America
- Molecular Toxicology Interdepartmental Degree Program, University of California Los Angeles, Los Angeles, California, United States of America
| | - Theodoros Kelesidis
- Department of Medicine, Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| |
Collapse
|
6
|
Arabska J, Wysokiński A, Brzezińska-Błaszczyk E, Kozłowska E. Serum Levels and in vitro CX3CL1 (Fractalkine), CXCL8, and IL-10 Synthesis in Phytohemaglutinin-Stimulated and Non-stimulated Peripheral Blood Mononuclear Cells in Subjects With Schizophrenia. Front Psychiatry 2022; 13:845136. [PMID: 35782435 PMCID: PMC9247257 DOI: 10.3389/fpsyt.2022.845136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/28/2022] [Indexed: 11/14/2022] Open
Abstract
INTRODUCTION Although schizophrenia is a severe mental illness, whose etiology is still largely unknown, its pathogenesis may be associated with dysregulation of the immune mechanisms. The present study compares the levels of interleukin (IL)-10, interleukin-8 (CXCL8), and fractalkine (CX3CL1) between schizophrenia patients and healthy controls. It also assesses the ability of peripheral peripheral blood mononuclear cells (PBMCs) to produce these cytokines spontaneously and following mitogen-stimulation. MATERIALS AND METHODS A prospective study was performed of 60 adult schizophrenia patients and 32 controls. CXCL8, IL-10, and fractalkine concentrations were measured in serum and supernatants from cultured PBMCs. Anthropometric (BMI, WHR) and body composition measurements were taken using bioimpedance analysis (BIA) and dual-energy X-ray absorptiometry (DXA). RESULTS AND CONCLUSION The schizophrenia patients demonstrated significantly higher levels of serum CXCL8 (schizophrenia: 13.4 ± 15.7 pg/mL, control: 6.9 ± 4.2 pg/mL, p = 0.001) and lower level of serum fractalkine (schizophrenia: 22.8 ± 9.9 pg/mL, control: 45.4 ± 84.5 pg/mL, p = 0.041). Serum IL-10 levels did not significantly differ. No in vitro synthesis of fractalkine was observed. Neither unstimulated or PHA-stimulated CXCL8 secretion differed between the two groups (p >0.05). The patients not taking mood stabilizers (MS-) demonstrated significantly higher CXCL8 levels than those on mood stabilizers (MS+) (p = 0.03) and control (p < 0.001). In addition, the MS- sub-group demonstrated significantly lower serum fraktalkine than controls (p = 0.009). These effects could be described as pseudo-normalization of CXCL8 and fractalkine in schizophrenia patients taking mood stabilizers.
Collapse
Affiliation(s)
- Jaśmina Arabska
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | - Adam Wysokiński
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | | | - Elżbieta Kozłowska
- Department of Experimental Immunology, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
7
|
Sil S, Periyasamy P, Thangaraj A, Niu F, Chemparathy DT, Buch S. Advances in the Experimental Models of HIV-Associated Neurological Disorders. Curr HIV/AIDS Rep 2021; 18:459-474. [PMID: 34427869 DOI: 10.1007/s11904-021-00570-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Involvement of the central nervous system (CNS) in HIV-1 infection is commonly associated with neurological disorders and cognitive impairment, commonly referred to as HIV-associated neurocognitive disorders (HAND). Severe and progressive neurocognitive impairment is rarely observed in the post-cART era; however, asymptomatic and mild neurocognitive disorders still exist, despite viral suppression. Additionally, comorbid conditions can also contribute to the pathogenesis of HAND. RECENT FINDINGS In this review, we summarize the characterization of HAND, factors contributing, and the functional impairments in both preclinical and clinical models. Specifically, we also discuss recent advances in the animal models of HAND and in in vitro cultures and the potential role of drugs of abuse in this model system of HAND. Potential peripheral biomarkers associated with HAND are also discussed. Overall, this review identifies some of the recent advances in the field of HAND in cell culture studies, animal models, clinical findings, and the limitations of each model system, which can play a key role in developing novel therapeutics in the field.
Collapse
Affiliation(s)
- Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA.
| | - Annadurai Thangaraj
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Divya T Chemparathy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, 68198-5880, USA
| |
Collapse
|
8
|
Caba E, Sherman MD, Farizatto KLG, Alcira B, Wang HW, Giardina C, Shin DG, Sandefur CI, Bahr BA. Excitotoxic stimulation activates distinct pathogenic and protective expression signatures in the hippocampus. J Cell Mol Med 2021; 25:9011-9027. [PMID: 34414662 PMCID: PMC8435451 DOI: 10.1111/jcmm.16864] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 12/20/2022] Open
Abstract
Excitotoxic events underlying ischaemic and traumatic brain injuries activate degenerative and protective pathways, particularly in the hippocampus. To understand opposing pathways that determine the brain's response to excitotoxicity, we used hippocampal explants, thereby eliminating systemic variables during a precise protocol of excitatory stimulation. N‐methyl‐d‐aspartate (NMDA) was applied for 20 min and total RNA isolated one and 24 h later for neurobiology‐specific microarrays. Distinct groups of genes exhibited early vs. delayed induction, with 63 genes exclusively reduced 24‐h post‐insult. Egr‐1 and NOR‐1 displayed biphasic transcriptional modulation: early induction followed by delayed suppression. Opposing events of NMDA‐induced genes linked to pathogenesis and cell survival constituted the early expression signature. Delayed degenerative indicators (up‐regulated pathogenic genes, down‐regulated pro‐survival genes) and opposing compensatory responses (down‐regulated pathogenic genes, up‐regulated pro‐survival genes) generated networks with temporal gene profiles mirroring coexpression network clustering. We then used the expression profiles to test whether NF‐κB, a potent transcription factor implicated in both degenerative and protective pathways, is involved in the opposing responses. The NF‐κB inhibitor MG‐132 indeed altered NMDA‐mediated transcriptional changes, revealing components of opposing expression signatures that converge on the single response element. Overall, this study identified counteracting avenues among the distinct responses to excitotoxicity, thereby suggesting multi‐target treatment strategies and implications for predictive medicine.
Collapse
Affiliation(s)
- Ebru Caba
- Vertex Pharmaceuticals, Cambridge, MA, USA.,Department of Pharmaceutical Sciences and the Neurosciences Program, University of Connecticut, Storrs, CT, USA
| | - Marcus D Sherman
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Karen L G Farizatto
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Britney Alcira
- Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA
| | - Hsin-Wei Wang
- Bioinformatics and Biocomputing Institute, University of Connecticut, Storrs, CT, USA.,Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, USA
| | - Dong-Guk Shin
- Bioinformatics and Biocomputing Institute, University of Connecticut, Storrs, CT, USA.,Department of Computer Science and Engineering, University of Connecticut, Storrs, CT, USA
| | - Conner I Sandefur
- Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Pharmacology and the Cystic Fibrosis and Pulmonary Diseases Research and Treatment Center, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA.,Sandefur Modeling, Pittsboro, NC, USA
| | - Ben A Bahr
- Department of Pharmaceutical Sciences and the Neurosciences Program, University of Connecticut, Storrs, CT, USA.,Biotechnology Research and Training Center, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Biology, University of North Carolina-Pembroke, Pembroke, NC, USA.,Department of Chemistry and Physics, University of North Carolina-Pembroke, Pembroke, NC, USA
| |
Collapse
|
9
|
Dong R, Huang R, Wang J, Liu H, Xu Z. Effects of Microglial Activation and Polarization on Brain Injury After Stroke. Front Neurol 2021; 12:620948. [PMID: 34276530 PMCID: PMC8280287 DOI: 10.3389/fneur.2021.620948] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 05/03/2021] [Indexed: 12/12/2022] Open
Abstract
Stroke is one of the most common causes of death worldwide. The subsequent development of neuroinflammation and brain edema dramatically increases the risks associated with stroke, leading to a substantial increase in mortality. Although considerable progress has been made in improving cerebral perfusion in the acute phase of stroke, effective treatment options for the subacute and chronic phases associated with cerebral infarction are limited. Microglia, the innate immune cells of the central nervous system (CNS), can be activated and polarized to take on different phenotypes in response to stimulations associated with stroke, including pro-inflammatory and anti-inflammatory phenotypes, which affect the prognosis of stroke. Therefore, investigation of the activation and polarizing mechanisms of microglia plays a critical role in treating stroke. The aim of this article was to investigate the significance of microglial phenotype regulation in stroke treatment by summarizing the activation, polarizing mechanisms, and general microglia characteristics.
Collapse
Affiliation(s)
- Rui Dong
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Renxuan Huang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jiaoqi Wang
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Hongyu Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhongxin Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
10
|
Zhang Q, He L, Chen M, Yang H, Cao X, Liu X, Hao Q, Chen Z, Liu T, Wei XE, Rong L. PSD-93 mediates the crosstalk between neuron and microglia and facilitates acute ischemic stroke injury by binding to CX3CL1. J Neurochem 2021; 157:2145-2157. [PMID: 33599284 DOI: 10.1111/jnc.15324] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/23/2022]
Abstract
Post-synaptic density 93 (PSD-93) mediates glutamate excitotoxicity induced by ischemic brain injury, which then induces microglial inflammatory response. However, the underlying mechanisms of how PSD-93 mediates the crosstalk between neurons and microglia in the post-synaptic dense region remain elusive. CX3 chemokine ligand 1 (CX3CL1) is a chemokine specifically expressed in neurons while its receptor CX3CR1 is highly expressed in microglia. In this study, we examined the interaction of PSD-93 and CX3CL1 in the crosstalk between neurons and microglia in acute ischemic stroke. We utilized male C57BL/6 mice to establish the middle cerebral artery occlusion model (MCAO) and designed a fusion small peptide Tat-CX3CL1 (357-395aa) to inhibit PSD-93 and CX3CL1 interaction. The combination peaks of PSD-93 and CX3CL1 at 6 hr after I/R were observed. The binding sites were located at the 420-535 amino acid sequence of PSD-93 and 357-395 amino acid sequence of CX3CL1. Tat-CX3CL1 (357-395aa) could inhibit the interaction of PSD-93 and CX3CL1 and inhibited the pro-inflammatory cytokine IL-1β and TNF-α expression and provided neuroprotection following reperfusion. Together, these data suggest that PSD-93 binds CX3CL1 to activate microglia and initiate neuroinflammation. Specific blockade of PSD-93-CX3CL1 interaction reduces I/R induced neuronal cell death, and provides a new therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Qingxiu Zhang
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Department of Neurology, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Lei He
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Mo Chen
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Hui Yang
- Department of Neurosurgery, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaowei Cao
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiaomei Liu
- Department of Pathogenic Biology and Immunology, Lab of Infection and Immunity, Xuzhou Medical University, Xuzhou, China
| | - Qi Hao
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Zhengwei Chen
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Tengfei Liu
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Xiu-E Wei
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Liangqun Rong
- Department of Neurology, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
11
|
Suresh P, Phasuk S, Liu IY. Modulation of microglia activation and Alzheimer's disease: CX3 chemokine ligand 1/CX3CR and P2X 7R signaling. Tzu Chi Med J 2021; 33:1-6. [PMID: 33505871 PMCID: PMC7821819 DOI: 10.4103/tcmj.tcmj_144_20] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/05/2020] [Accepted: 08/24/2020] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive deficits. Two hallmarks of AD that cause chronic inflammation and lead to neuronal dysfunction and damage are tau tangles and amyloid plaques. Microglial cells, the primary immune cells of the central nervous system, maintain a homeostatic active/inactive state via a bidirectional, dynamic communication with neurons. Several studies have revealed that dysregulated microglial activation leads to AD pathology. Therefore, we reviewed the relationship between AD and two important signaling complexes, CX3 chemokine ligand 1 (CX3CL1)/CX3CR1 and ATP/P2X7R, that play critical roles in the regulation of microglial activation. CX3CL1/CX3CR1 is one important signaling which controls the microglia function. Altering this pathway can have opposite effects on amyloid and tau pathology in AD. Another important molecule is P2X7R which involves in the activation of microglia. Over activation of P2X7R is evident in AD pathogenesis. In this review, we discuss influence of the two signaling pathways at different stages of AD pathology as well as the drug candidates that can modulate CX3CL1/CX3CR1 and ATP/P2X7R.
Collapse
Affiliation(s)
- Pavithra Suresh
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Sarayut Phasuk
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| | - Ingrid Y Liu
- Institute of Medical Sciences, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
12
|
Angelopoulou E, Paudel YN, Shaikh MF, Piperi C. Fractalkine (CX3CL1) signaling and neuroinflammation in Parkinson’s disease: Potential clinical and therapeutic implications. Pharmacol Res 2020; 158:104930. [DOI: 10.1016/j.phrs.2020.104930] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/16/2020] [Accepted: 05/12/2020] [Indexed: 12/14/2022]
|
13
|
Pandur E, Tamási K, Pap R, Varga E, Miseta A, Sipos K. Fractalkine Induces Hepcidin Expression of BV-2 Microglia and Causes Iron Accumulation in SH-SY5Y Cells. Cell Mol Neurobiol 2019; 39:985-1001. [PMID: 31172340 PMCID: PMC6711953 DOI: 10.1007/s10571-019-00694-4] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/29/2019] [Indexed: 02/08/2023]
Abstract
Fractalkine (CX3CL1) is a potent inflammatory mediator of the central nervous system, which is expressed by neurons and regulates microglial functions by binding to fractalkine receptor (CX3CR1). It has been demonstrated that neuroinflammation plays an important role in iron accumulation of the brain leading to neuronal cell death. The major regulator of iron homeostasis is the peptide hormone hepcidin. Hepcidin expression is triggered by inflammatory conditions, which may contribute to the neuronal iron accumulation. In the present study, we established a bilaminar co-culture system of differentiated SH-SY5Y cells and BV-2 microglia as a neuronal model to examine the effect of soluble fractalkine on iron homeostasis of microglia and SH-SY5Y cells. We determined the hepcidin expression of fractalkine-treated microglia which showed significant elevation. We examined the relation between increased hepcidin secretion, the known hepcidin regulators and the signalling pathways controlled by fractalkine receptor. Our data revealed that TMPRSS6 and alpha 1-antitrypsin levels decreased due to fractalkine treatment, as well as the activity of NFκB pathway and the tyrosine phosphorylation of STAT5 factor. Moreover, fractalkine-induced hepcidin production of microglia initiated ferroportin internalisation of SH-SY5Y cells, which contributed to iron accumulation of neurons. Our results demonstrate that soluble form of fractalkine regulates hepcidin expression of BV-2 cells through fractalkine-mediated CX3CR1 internalisation. Moreover, fractalkine indirectly contributes to the iron accumulation of SH-SY5Y cells by activating ferroportin internalisation and by triggering the expressions of divalent metal transporter-1, ferritin heavy chain and mitochondrial ferritin.
Collapse
Affiliation(s)
- Edina Pandur
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2, 7624, Pécs, Hungary.
| | - Kitti Tamási
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2, 7624, Pécs, Hungary
| | - Ramóna Pap
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2, 7624, Pécs, Hungary
| | - Edit Varga
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2, 7624, Pécs, Hungary
| | - Attila Miseta
- Department of Laboratory Medicine, Medical School, University of Pécs, Ifjúság Str. 13, 7624, Pécs, Hungary
| | - Katalin Sipos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Pécs, Rókus Str. 2, 7624, Pécs, Hungary
| |
Collapse
|
14
|
Microglia Mediate HIV-1 gp120-Induced Synaptic Degeneration in Spinal Pain Neural Circuits. J Neurosci 2019; 39:8408-8421. [PMID: 31471472 DOI: 10.1523/jneurosci.2851-18.2019] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 08/23/2019] [Accepted: 08/26/2019] [Indexed: 02/07/2023] Open
Abstract
HIV-1 infection of the nervous system causes various neurological diseases, and synaptic degeneration is likely a critical step in the neuropathogenesis. Our prior studies revealed a significant decrease of synaptic protein, specifically in the spinal dorsal horn of patients with HIV-1 in whom pain developed, suggesting a potential contribution of synaptic degeneration to the pathogenesis of HIV-associated pain. However, the mechanism by which HIV-1 causes the spinal synaptic degeneration is unclear. Here, we identified a critical role of microglia in the synaptic degeneration. In primary cortical cultures (day in vitro 14) and spinal cords of 3- to 5-month-old mice (both sexes), microglial ablation inhibited gp120-induced synapse decrease. Fractalkine (FKN), a microglia activation chemokine specifically expressed in neurons, was upregulated by gp120, and knockout of the FKN receptor CX3CR1, which is predominantly expressed in microglia, protected synapses from gp120-induced toxicity. These results indicate that the neuron-to-microglia intercellular FKN/CX3CR1 signaling plays a role in gp120-induced synaptic degeneration. To elucidate the mechanism controlling this intercellular signaling, we tested the role of the Wnt/β-catenin pathway in regulating FKN expression. Inhibition of Wnt/β-catenin signaling blocked both gp120-induced FKN upregulation and synaptic degeneration, and gp120 stimulated Wnt/β-catenin-regulated FKN expression via NMDA receptors (NMDARs). Furthermore, NMDAR antagonist APV, Wnt/β-catenin signaling suppressor DKK1, or knockout of CX3CR1 alleviated gp120-induced mechanical allodynia in mice, suggesting a critical contribution of the Wnt/β-catenin/FKN/CX3R1 pathway to gp120-induced pain. These findings collectively suggest that HIV-1 gp120 induces synaptic degeneration in the spinal pain neural circuit by activating microglia via Wnt3a/β-catenin-regulated FKN expression in neurons.SIGNIFICANCE STATEMENT Synaptic degeneration develops in the spinal cord dorsal horn of HIV patients with chronic pain, but the patients without the pain disorder do not show this neuropathology, indicating a pathogenic contribution of the synaptic degeneration to the development of HIV-associated pain. However, the mechanism underlying the synaptic degeneration is unclear. We report here that HIV-1 gp120, a neurotoxic protein that is specifically associated with the manifestation of pain in HIV patients, induces synapse loss via microglia. Further studies elucidate that gp120 activates microglia by stimulating Wnt/β-catenin-regulated fractalkine in neuron. The results demonstrate a critical role of microglia in the pathogenesis of HIV-associated synaptic degeneration in the spinal pain neural circuit.
Collapse
|
15
|
Anderson AM, Nguyen ML, Potter M, Rosario D, Kempinska K, Ellis RJ, Diccianni M, Letendre SL. Comparison of bead array and glass nanoreactor multi-analyte platforms for the evaluation of CNS and peripheral inflammatory markers during HIV infection. J Immunol Methods 2018; 465:7-12. [PMID: 30468733 DOI: 10.1016/j.jim.2018.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 12/26/2022]
Abstract
While human immunodeficiency virus (HIV) infection has become a treatable disease with the development of combination antiretroviral therapy (cART), chronic inflammation that affects the central nervous system and other organs is still common. Reliable methods are needed to study HIV-associated inflammatory biomarkers. In this study involving both plasma and cerebrospinal fluid (CSF), we compared multiplex bead array (MBA) to a relatively new technology based on microfluidics and glass nanoreactor (GNR) technology for the measurement of three commonly studied markers from HIV-infected individuals. We found that results correlated between the two platforms for MCP-1 in both fluids as well as for plasma TNFα (all p < .005). However, results between the two platforms did not correlate for CSF TNFα or fractalkine from plasma or CSF. A statistically significant decrease in CSF TNFα over time (p < .0001) was only detectable with the MBA platform, and TNFα on the MBA was the only CSF biomarker to correlate with CSF HIV RNA (rho = 0.71, p < .0001). Meanwhile, the GNR platform was superior in terms of intra-assay fractalkine (FKN) variability and the detection of a significant FKN decrease over time. Additionally, the only significant correlation between blood biomarkers and plasma HIV RNA was with FKN on the GNR platform (rho = 0.38, p = .015). Given the variability in results between platforms, more research is needed on methods to quantitate HIV-associated inflammation.
Collapse
Affiliation(s)
- Albert M Anderson
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States.
| | - Minh Ly Nguyen
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States
| | - Michael Potter
- Department of Medicine, University of California at San Diego School of Medicine, La Jolla, CA, United States; Department of Psychiatry, University of California at San Diego School of Medicine, La Jolla, CA, United States
| | - Debra Rosario
- Department of Medicine, University of California at San Diego School of Medicine, La Jolla, CA, United States; Department of Psychiatry, University of California at San Diego School of Medicine, La Jolla, CA, United States
| | - Katarzyna Kempinska
- Department of Pediatrics, University of California at San Diego School of Medicine, La Jolla, CA, United States
| | - Ronald J Ellis
- Department of Neurosciences, University of California at San Diego School of Medicine, La Jolla, CA, United States
| | - Mitchell Diccianni
- Department of Pediatrics, University of California at San Diego School of Medicine, La Jolla, CA, United States
| | - Scott L Letendre
- Department of Medicine, University of California at San Diego School of Medicine, La Jolla, CA, United States; Department of Psychiatry, University of California at San Diego School of Medicine, La Jolla, CA, United States
| |
Collapse
|
16
|
Abstract
Endothelins were discovered more than thirty years ago as potent vasoactive compounds. Beyond their well-documented cardiovascular properties, however, the contributions of the endothelin pathway have been demonstrated in several neuroinflammatory processes and the peptides have been reported as clinically relevant biomarkers in neurodegenerative diseases. Several studies report that endothelin-1 significantly contributes to the progression of neuroinflammatory processes, particularly during infections in the central nervous system (CNS), and is associated with a loss of endothelial integrity at the blood brain barrier level. Because of the paucity of clinical trials with endothelin-1 antagonists in several infectious and non-infectious neuroinflammatory diseases, it remains an open question whether the 21 amino acid peptide is a mediator/modulator rather than a biomarker of the progression of neurodegeneration. This review focuses on the potential roles of endothelins in the pathology of neuroinflammatory processes, including infectious diseases of viral, bacterial or parasitic origin in which the synthesis of endothelins or its pharmacology have been investigated from the cell to the bedside in several cases, as well as in non-infectious inflammatory processes such as neurodegenerative disorders like Alzheimers Disease or central nervous system vasculitis.
Collapse
|
17
|
Abstract
Human immunodeficiency virus (HIV) infection induces neuronal injuries, with almost 50% of infected individuals developing HIV-associated neurocognitive disorders (HAND). Although highly activate antiretroviral therapy (HAART) has significantly reduced the incidence of severe dementia, the overall prevalence of HAND remains high. Synaptic degeneration is emerging as one of the most relevant neuropathologies associate with HAND. Previous studies have reported critical roles of viral proteins and inflammatory responses in this pathogenesis. Infected cells, including macrophages, microglia and astrocytes, may release viral proteins and other neurotoxins to stimulate neurons and cause excessive calcium influx, overproduction of free radicals and disruption of neurotransmitter hemostasis. The dysregulation of neural circuits likely leads to synaptic damage and loss. Identification of the specific mechanism of the synaptic degeneration may facilitate the development of effective therapeutic approaches to treat HAND.
Collapse
Affiliation(s)
- Wenjuan Ru
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Shao-Jun Tang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
18
|
Perera SS, Wang B, Damian A, Dyer W, Zhou L, Conceicao V, Saksena NK. Retrospective Proteomic Analysis of Cellular Immune Responses and Protective Correlates of p24 Vaccination in an HIV Elite Controller Using Antibody Arrays. ACTA ACUST UNITED AC 2016; 5:microarrays5020014. [PMID: 27600080 PMCID: PMC5003490 DOI: 10.3390/microarrays5020014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 01/14/2016] [Accepted: 01/25/2016] [Indexed: 01/22/2023]
Abstract
Background: HIV p24 is an extracellular HIV antigen involved in viral replication. Falling p24 antibody responses are associated with clinical disease progression and their preservation with non-progressive disease. Stimulation of p24 antibody production by immunization to delay progression was the basis of discontinued p24 vaccine. We studied a therapy-naive HIV+ man from Sydney, Australia, infected in 1988. He received the HIV-p24-virus like particle (VLP) vaccine in 1993, and continues to show vigorous p24 antigen responses (>4% p24-specific CD4+ T cells), coupled with undetectable plasma viremia. We defined immune-protective correlates of p24 vaccination at the proteomic level through parallel retrospective analysis of cellular immune responses to p24 antigen in CD4+ and CD8+ T cells and CD14+ monocytes at viremic and aviremic phases using antibody-array. We found statistically significant coordinated up-regulation by all three cell-types with high fold-changes in fractalkine, ITAC, IGFBP-2, and MIP-1α in the aviremic phase. TECK and TRAIL-R4 were down-regulated in the viremic phase and up-regulated in the aviremic phase. The up-regulation of fractalkine in all three cell-types coincided with protective effect, whereas the dysfunction in anti-apoptotic chemokines with the loss of immune function. This study highlights the fact that induction of HIV-1-specific helper cells together with coordinated cellular immune response (p < 0.001) might be important in immunotherapeutic interventions and HIV vaccine development.
Collapse
Affiliation(s)
- Suneth S Perera
- Department of Medicine, University of Sydney, Sydney 2000, Australia.
| | - Bin Wang
- Department of Medicine, University of Sydney, Sydney 2000, Australia.
| | - Arturo Damian
- Department of Cytogenetics, Children's Hospital at Westmead, Sydney 2000, Australia.
| | - Wayne Dyer
- Australian Red Cross Blood Service, 17 O'Riordan Street, Alexandria NSW 2015 and School of Medical Sciences, (Faculty of Medicine) University of Sydney, Sydney 2000, Australia.
| | - Li Zhou
- Department of Medicine, University of Sydney, Sydney 2000, Australia.
| | - Viviane Conceicao
- Department of Medicine, University of Sydney, Sydney 2000, Australia.
| | - Nitin K Saksena
- Department of Medicine, University of Sydney, Sydney 2000, Australia.
| |
Collapse
|
19
|
Liu W, Jiang L, Bian C, Liang Y, Xing R, Yishakea M, Dong J. Role of CX3CL1 in Diseases. Arch Immunol Ther Exp (Warsz) 2016; 64:371-83. [PMID: 27098399 DOI: 10.1007/s00005-016-0395-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 02/21/2016] [Indexed: 01/04/2023]
Abstract
Chemokines are a family of small 8-10 kDa inducible cytokines. Initially characterized as chemotactic factors, they are now considered to affect not just cellular recruitment. CX3CL1 is a unique chemokine that can exist in a soluble form, as a chemotactic cytokine, or in a membrane-attached form that acts as a binding molecule. Recently, the effects of CX3CL1 on diseases, such as inflammation and cancer, have been supported and confirmed by numerous publications. However, due to its dual effects, CX3CL1 exerts numerous effects on pathophysiological conditions that have both negative and positive consequences on pathogenesis and outcome. This review article summarizes the important scientific and clinical data that now point to a critical role for CX3CL1 in diseases.
Collapse
Affiliation(s)
- WangMi Liu
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Libo Jiang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Chong Bian
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yun Liang
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Rong Xing
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Mumingjiang Yishakea
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jian Dong
- Department of Orthopedic Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
20
|
Reiss CS. Innate Immunity in Viral Encephalitis. NEUROTROPIC VIRAL INFECTIONS 2016. [PMCID: PMC7153449 DOI: 10.1007/978-3-319-33189-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- Carol Shoshkes Reiss
- Departments of Biology and Neural Science, New York University, New York, New York USA
| |
Collapse
|
21
|
CX3CR1 Disruption Differentially Influences Dopaminergic Neuron Degeneration in Parkinsonian Mice Depending on the Neurotoxin and Route of Administration. Neurotox Res 2015; 29:364-80. [PMID: 26403659 DOI: 10.1007/s12640-015-9557-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Revised: 08/03/2015] [Accepted: 08/18/2015] [Indexed: 01/02/2023]
Abstract
Parkinson's disease (PD) is characterized by progressive degeneration of dopaminergic neurons accompanied by an inflammatory reaction. The neuron-derived chemokine fractalkine (CX3CL1) is an exclusive ligand for the receptor CX3CR1 expressed on microglia. The CX3CL1/CX3CR1 signaling is important for sustaining microglial activity. Using a recently developed PD model, in which the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) toxin is delivered intranasally, we hypothesized that CX3CR1 could play a role in neurotoxicity and glial activation. For this, we used CX3CR1 knock-in mice and compared results with those obtained using the classical PD models through intraperitonal MPTP or intrastriatal 6-hydroxydopamine (6-OHDA). The striatum from all genotypes (CX3CR1(+/+), CX3CR1(+/GFP) and CX3CR1-deficient mice) showed a significant dopaminergic depletion after intranasal MPTP inoculation. In contrast to that, we could not see differences in the number of dopaminergic neurons in the substantia nigra of CX3CR1-deficient animals. Similarly, after 6-OHDA infusion, the CX3CR1 deletion decreased the amphetamine-induced turning behavior observed in CX3CR1(+/GFP) mice. After the 6-OHDA inoculation, a minor dopaminergic neuronal loss was observed in the substantia nigra from CX3CR1-deficient mice. Distinctly, a more extensive neuronal cell loss was observed in the substantia nigra after the intraperitoneal MPTP injection in CX3CR1 disrupted animals, corroborating previous results. Intranasal and intraperitoneal MPTP inoculation induced a similar microgliosis in CX3CR1-deficient mice but a dissimilar change in the astrocyte proliferation in the substantia nigra. Nigral astrocyte proliferation was observed only after intraperitoneal MPTP inoculation. In conclusion, intranasal MPTP and 6-OHDA lesion in CX3CR1-deficient mice yield no nigral dopaminergic neuron loss, linked to the absence of astroglial proliferation.
Collapse
|
22
|
Plasma and Cerebrospinal Fluid Biomarkers Predict Cerebral Injury in HIV-Infected Individuals on Stable Combination Antiretroviral Therapy. J Acquir Immune Defic Syndr 2015; 69:29-35. [PMID: 25622053 DOI: 10.1097/qai.0000000000000532] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
OBJECTIVES HIV-associated brain injury persists despite combination antiretroviral therapy, but contributing factors remain poorly understood. We postulated that inflammation-associated biomarkers will be associated with cerebral injury on proton magnetic resonance spectroscopy in chronically HIV-infected subjects. METHODS Five biomarkers were measured in 197 HIV-infected subjects: soluble CD14, MCP-1, IP-10, MIP-1β, and fractalkine. Levels of N-acetyl aspartate (NAA), Choline (Cho), Myoinositol (MI), Glutamate + Glutamine (Glx), and Creatine (Cr) were acquired in the midfrontal cortex (MFC), frontal white matter, and basal ganglia (BG). Predictive models were built through linear regression, and the best models were chosen using the Akaike Information Criterion. RESULTS Increases in plasma or CSF MCP-1 were associated with lower NAA/Cr in the MFC and BG, whereas metabolite changes in the frontal white matter for NAA/Cr, GlxCr, and Cho/Cr were explained almost exclusively by a single factor, sCD14. Plasma and CSF levels of this factor were also significantly associated with Glx/Cr in MFC and BG. Higher CSF FKN was associated with higher NAA/Cr in BG. Best predictors for higher Cho/Cr in BG and MFC were CSF sCD14 and CSF MIP-1β. Plasma and CSF IP-10 were only associated with Cho/Cr in MFC. Of the 3 models that simultaneously accounted for both plasma and CSF, there were more associations between CSF biomarkers and magnetic resonance spectroscopy metabolites. CONCLUSIONS Markers of inflammation and immune activation, in particular MCP-1 and sCD14, predominantly reflecting CNS sources, contribute to the persistence of brain injury in a metabolite and region-dependent manner in chronically HIV-infected patients on stable combination antiretroviral therapy.
Collapse
|
23
|
Cytokines and chemokines as biomarkers of ethanol-induced neuroinflammation and anxiety-related behavior: role of TLR4 and TLR2. Neuropharmacology 2015; 89:352-9. [PMID: 25446779 DOI: 10.1016/j.neuropharm.2014.10.014] [Citation(s) in RCA: 139] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Revised: 09/03/2014] [Accepted: 10/14/2014] [Indexed: 12/23/2022]
Abstract
Recent evidence supports the influence of neuroimmune system activation on behavior. We have demonstrated that ethanol activates the innate immune system by stimulating toll-like receptor 4 (TLR4) signaling in glial cells, which triggers the release of inflammatory mediators and causes neuroinflammation. The present study aimed to evaluate whether the ethanol-induced up-regulation of cytokines and chemokines is associated with anxiety-related behavior, 24 h after ethanol removal, and if TLR4 or TLR2 is involved in these effects. We used WT, TLR4-KO and TLR2-KO mice treated with alcohol for 5 months to show that chronic ethanol consumption increases the levels of cytokines (IL-1β, IL-17, TNF-α) and chemokines (MCP-1, MIP-1α, CX3CL1) in the striatum and serum (MCP-1, MIP-1α, CX3CL1) of WT mice. Alcohol deprivation for 24 h induces IFN-γ levels in the striatum and maintains high levels of some cytokines (IL-1β, IL-17) and chemokines (MIP-1α, CX3CL1) in this brain region. The latter events were associated with an increase in anxiogenic-related behavior, as evaluated by the dark and light box and the elevated plus maze tests. Notably, mice lacking TLR4 or TLR2 receptors are largely protected against ethanol-induced cytokine and chemokine release, and behavioral associated effects during alcohol abstinence. These data support the role of TLR4 and TLR2 responses in neuroinflammation and in anxiogenic-related behavior effects during ethanol deprivation, and also provide evidence that chemokines and cytokines can be biomarkers of ethanol-induced neuroimmune response.
Collapse
|
24
|
Lauro C, Catalano M, Trettel F, Limatola C. Fractalkine in the nervous system: neuroprotective or neurotoxic molecule? Ann N Y Acad Sci 2015; 1351:141-8. [DOI: 10.1111/nyas.12805] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Clotilde Lauro
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti; Sapienza University of Rome; Rome Italy
| | - Myriam Catalano
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti; Sapienza University of Rome; Rome Italy
- IRCCS NeuroMed; Pozzilli Italy
| | - Flavia Trettel
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti; Sapienza University of Rome; Rome Italy
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Istituto Pasteur Fondazione Cenci Bolognetti; Sapienza University of Rome; Rome Italy
- IRCCS NeuroMed; Pozzilli Italy
| |
Collapse
|
25
|
Crowell CS, Malee KM, Yogev R, Muller WJ. Neurologic disease in HIV-infected children and the impact of combination antiretroviral therapy. Rev Med Virol 2014; 24:316-31. [PMID: 24806816 DOI: 10.1002/rmv.1793] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 03/23/2014] [Accepted: 03/24/2014] [Indexed: 11/10/2022]
Abstract
The prevalence of HIV-associated neurocognitive impairment in perinatally HIV-infected children has declined since the introduction of combination antiretroviral therapy (cART). Early initiation of cART in infancy has been shown to positively impact neurodevelopment; however, children continue to be diagnosed with HIV outside of the early infancy period and can experience subtle to severe neurocognitive deficits despite cART. The causes of these neurocognitive deficits despite effective cART are multifactorial and likely include continued viral replication in the CNS, ongoing neuroinflammation, irreversible CNS injury prior to cART initiation, neurotoxic effects of cART, and socioeconomic and psychosocial effects. Many aspects of our understanding of HIV-associated neurocognitive disorders have emerged from research in adult patients, but perinatally HIV-infected children represent a very different population. These children were exposed to HIV during a period of rapid brain development and have lifelong infection and potential lifelong cART exposure. HIV is no longer a rapidly fatal disease, and most HIV-infected children in resource-rich countries are living into adulthood. It is therefore critical to optimize neurocognitive outcomes of these youth. This review summarizes current understanding of the pathogenesis of HIV-associated CNS infection and the impact of cART on neurocognitive function in children and adolescents and discusses important areas for future research.
Collapse
Affiliation(s)
- Claudia S Crowell
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, USA
| | | | | | | |
Collapse
|
26
|
Bertin J, Jalaguier P, Barat C, Roy MA, Tremblay MJ. Exposure of human astrocytes to leukotriene C4 promotes a CX3CL1/fractalkine-mediated transmigration of HIV-1-infected CD4⁺ T cells across an in vitro blood-brain barrier model. Virology 2014; 454-455:128-38. [PMID: 24725939 DOI: 10.1016/j.virol.2014.02.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 12/23/2013] [Accepted: 02/07/2014] [Indexed: 11/18/2022]
Abstract
Eicosanoids, including cysteinylleukotrienes (cysLTs), are found in the central nervous system (CNS) of individuals infected with HIV-1. Few studies have addressed the contribution of cysLTs in HIV-1-associated CNS disorders. We demonstrate that conditioned medium from human astrocytes treated with leukotriene C4 (LTC4) increases the transmigration of HIV-1-infected CD4(+) T cells across an in vitro blood-brain barrier (BBB) model using cultured brain endothelial cells. Additional studies indicate that the higher cell migration is linked with secretion by astrocytes of CX3CL1/fractalkine, a chemokine that has chemoattractant activity for CD4(+) T cells. Moreover, we report that the enhanced cell migration across BBB leads to a more important CD4(+) T cell-mediated HIV-1 transfer toward macrophages. Altogether data presented in the present study reveal the important role that LTC4, a metabolite of arachidonic acid, may play in the HIV-1-induced neuroinvasion, neuropathogenesis and disease progression.
Collapse
Affiliation(s)
- Jonathan Bertin
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada
| | - Pascal Jalaguier
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada
| | - Corinne Barat
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada
| | - Marc-André Roy
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada
| | - Michel J Tremblay
- Axe des Maladies Infectieuses et Immunitaires, Centre de recherche du Centre Hospitalier Universitaire (CHU) de Québec - pavillon CHUL, Canada; Département de Microbiologie-Infectiologie et Immunologie, Faculté de médecine, Université Laval, Québec, Canada.
| |
Collapse
|
27
|
Sheridan GK, Murphy KJ. Neuron-glia crosstalk in health and disease: fractalkine and CX3CR1 take centre stage. Open Biol 2013; 3:130181. [PMID: 24352739 PMCID: PMC3877844 DOI: 10.1098/rsob.130181] [Citation(s) in RCA: 255] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
An essential aspect of normal brain function is the bidirectional interaction and communication between neurons and neighbouring glial cells. To this end, the brain has evolved ligand-receptor partnerships that facilitate crosstalk between different cell types. The chemokine, fractalkine (FKN), is expressed on neuronal cells, and its receptor, CX(3)CR1, is predominantly expressed on microglia. This review focuses on several important functional roles for FKN/CX(3)CR1 in both health and disease of the central nervous system. It has been posited that FKN is involved in microglial infiltration of the brain during development. Microglia, in turn, are implicated in the developmental synaptic pruning that occurs during brain maturation. The abundance of FKN on mature hippocampal neurons suggests a homeostatic non-inflammatory role in mechanisms of learning and memory. There is substantial evidence describing a role for FKN in hippocampal synaptic plasticity. FKN, on the one hand, appears to prevent excess microglial activation in the absence of injury while promoting activation of microglia and astrocytes during inflammatory episodes. Thus, FKN appears to be neuroprotective in some settings, whereas it contributes to neuronal damage in others. Many progressive neuroinflammatory disorders that are associated with increased microglial activation, such as Alzheimer's disease, show disruption of the FKN/CX(3)CR1 communication system. Thus, targeting CX(3)CR1 receptor hyperactivation with specific antagonists in such neuroinflammatory conditions may eventually lead to novel neurotherapeutics.
Collapse
Affiliation(s)
- Graham K Sheridan
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge CB2 3DY, UK
| | | |
Collapse
|
28
|
Hu S, Sheng WS, Rock RB. CB2 receptor agonists protect human dopaminergic neurons against damage from HIV-1 gp120. PLoS One 2013; 8:e77577. [PMID: 24147028 PMCID: PMC3798286 DOI: 10.1371/journal.pone.0077577] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 09/03/2013] [Indexed: 11/18/2022] Open
Abstract
Despite the therapeutic impact of anti-retroviral therapy, HIV-1-associated neurocognitive disorder (HAND) remains a serious threat to AIDS patients, and there currently remains no specific therapy for the neurological manifestations of HIV-1. Recent work suggests that the nigrostriatal dopaminergic area is a critical brain region for the neuronal dysfunction and death seen in HAND and that human dopaminergic neurons have a particular sensitivity to gp120-induced damage, manifested as reduced function (decreased dopamine uptake), morphological changes, and reduced viability. Synthetic cannabinoids inhibit HIV-1 expression in human microglia, suppress production of inflammatory mediators in human astrocytes, and there is substantial literature demonstrating the neuroprotective properties of cannabinoids in other neuropathogenic processes. Based on these data, experiments were designed to test the hypothesis that synthetic cannabinoids will protect dopaminergic neurons against the toxic effects of the HIV-1 protein gp120. Using a human mesencephalic neuronal/glial culture model, which contains dopaminergic neurons, microglia, and astrocytes, we were able to show that the CB1/CB2 agonist WIN55,212-2 blunts gp120-induced neuronal damage as measured by dopamine transporter function, apoptosis and lipid peroxidation; these actions were mediated principally by the CB2 receptor. Adding supplementary human microglia to our cultures enhances gp120-induced damage; WIN55,212-2 is able to alleviate this enhanced damage. Additionally, WIN55,212-2 inhibits gp120-induced superoxide production by purified human microglial cells, inhibits migration of human microglia towards supernatants generated from gp120-stimulated human mesencephalic neuronal/glial cultures and reduces chemokine and cytokine production from the human mesencephalic neuronal/glial cultures. These data suggest that synthetic cannabinoids are capable of protecting human dopaminergic neurons from gp120 in a variety of ways, acting principally through the CB2 receptors and microglia.
Collapse
Affiliation(s)
- Shuxian Hu
- Center for Infectious Diseases and Microbiology Translational Research, Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Wen S. Sheng
- Center for Infectious Diseases and Microbiology Translational Research, Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - R. Bryan Rock
- Center for Infectious Diseases and Microbiology Translational Research, Division of Infectious Diseases and International Medicine, Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
29
|
Réaux-Le Goazigo A, Van Steenwinckel J, Rostène W, Mélik Parsadaniantz S. Current status of chemokines in the adult CNS. Prog Neurobiol 2013; 104:67-92. [PMID: 23454481 DOI: 10.1016/j.pneurobio.2013.02.001] [Citation(s) in RCA: 169] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 02/01/2013] [Accepted: 02/03/2013] [Indexed: 12/13/2022]
Abstract
Chemokines - chemotactic cytokines - are small secreted proteins that attract and activate immune and non-immune cells in vitro and in vivo. It has been suggested that chemokines and their receptors play a role in the central nervous system (CNS), in addition to their well established role in the immune system. We focus here on three chemokines-CXCL12 (C-X-C motif ligand 12), CCL2 (C-C motif ligand 2), and CX3CL1 (C-X-3C motif ligand 1) - and their principal receptors - CXCR4 (C-X-C motif receptor 4), CCR2 (C-C motif receptor 2) and CX3CR1 (C-X-3C motif receptor 1), respectively. We first introduce the classification of chemokines and their G-protein coupled receptors and the main signaling pathways triggered by receptor activation. We then discuss the cellular distribution of CXCL12/CXCR4, CCL2/CCR2 and CX3CL1/CX3CR1 in adult brain and the neurotransmission and neuromodulation effects controlled by these chemokines in the adult CNS. Changes in the expression of CXCL12, CCL2 and CX3CL1 and their respective receptors are also increasingly being implicated in the pathogenesis of CNS disorders, such as Alzheimer's disease, Parkinson's disease, HIV-associated encephalopathy, stroke and multiple sclerosis, and are therefore plausible targets for future pharmacological intervention. The final section thus discusses the role of these chemokines in these pathophysiological states. In conclusion, the role of these chemokines in cellular communication may make it possible: (i) to identify new pathways of neuron-neuron, glia-glia or neuron-glia communications relevant to both normal brain function and neuroinflammatory and neurodegenerative diseases; (ii) to develop new therapeutic approaches for currently untreatable brain diseases.
Collapse
|
30
|
Davis PJ, Glinsky GV, Lin HY, Incerpi S, Davis FB, Mousa SA, Tang HY, Hercbergs A, Luidens MK. Molecular mechanisms of actions of formulations of the thyroid hormone analogue, tetrac, on the inflammatory response. Endocr Res 2013; 38:112-8. [PMID: 23545000 DOI: 10.3109/07435800.2013.778865] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
BACKGROUND Tetraiodothyroacetic acid (tetrac) and its nanoparticulate formulation (Nanotetrac) act at a cell surface receptor to block angiogenesis and tumor cell proliferation. OBJECTIVE The complex anti-angiogenic properties of tetrac and Nanotetrac caused us to search in the literature and in certain of our unpublished mRNA experiments for evidence that these agents affect the early inflammatory response, perhaps through actions on specific cytokines and chemokines. RESULTS AND DISCUSSION Tetrac and Nanotetrac inhibit expression in tumor cells of cytokine genes, e.g., specific interleukins, and chemokine genes, such as fractalkine (CX3CL1), and chemokine receptor genes (CX3CR1) that have been identified as high priority targets in the development of inflammation-suppressant drugs. The possibility is also examined that tetrac formulations have an effect on the function of inflammatory cells.
Collapse
Affiliation(s)
- Paul J Davis
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Albany, New York 12144, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Pivotal role of M-DC8+ monocytes from viremic HIV-infected patients in TNFα overproduction in response to microbial products. Blood 2012; 120:2259-68. [DOI: 10.1182/blood-2012-03-418681] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Abstract
HIV infects activated CD4+ T cells and induces their depletion. Progressive HIV infection leading to AIDS is fueled by chronic immune hyperactivation, mediated by inflammatory cytokines like TNFα. This has been related to intestinal epithelial damage and microbial LPS translocation into the circulation. Using 11-color flow cytometry, cell sorting, and cell culture, we investigated the numbers and TNFα production of fully defined circulating dendritic cell and monocyte populations during HIV-1 infection. In 15 viremic, untreated patients, compared with 8 treated, virologically suppressed patients or to 13 healthy blood donors, circulating CD141 (BDCA-3)+ and CD1c (BDCA-1)+ dendritic cell counts were reduced. Conversely, CD14+CD16++ monocyte counts were increased, particularly those expressing M-DC8, while classical CD14++CD16−M-DC8− monocyte numbers were unchanged. Blood mononuclear cells from viremic patients produced more TNFα in response to LPS than those from virologically suppressed patients. M-DC8+ monocytes were mostly responsible for this overproduction. Moreover, M-DC8+ monocytes differentiated in vitro from classical monocytes using M-CSF and GM-CSF, which is increased in viremic patient's plasma. This M-DC8+ monocyte population, which is involved in the pathogenesis of chronic inflammatory diseases like Crohn disease, might thus be considered as a major actor in the immune hyperactivation fueling HIV infection progression.
Collapse
|
32
|
Hauser KF, Fitting S, Dever SM, Podhaizer EM, Knapp PE. Opiate drug use and the pathophysiology of neuroAIDS. Curr HIV Res 2012; 10:435-52. [PMID: 22591368 PMCID: PMC3431547 DOI: 10.2174/157016212802138779] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Revised: 01/12/2012] [Accepted: 01/14/2012] [Indexed: 11/22/2022]
Abstract
Opiate abuse and HIV-1 have been described as interrelated epidemics, and even in the advent of combined anti-retroviral therapy, the additional abuse of opiates appears to result in greater neurologic and cognitive deficits. The central nervous system (CNS) is particularly vulnerable to interactive opiate-HIV-1 effects, in part because of the unique responses of microglia and astroglia. Although neurons are principally responsible for behavior and cognition, HIV-1 infection and replication in the brain is largely limited to microglia, while astroglia and perhaps glial progenitors can be latently infected. Thus, neuronal dysfunction and injury result from cellular and viral toxins originating from HIV-1 infected/exposed glia. Importantly, subsets of glial cells including oligodendrocytes, as well as neurons, express µ-opioid receptors and therefore can be direct targets for heroin and morphine (the major metabolite of heroin in the CNS), which preferentially activate µ-opioid receptors. This review highlights findings that neuroAIDS is a glially driven disease, and that opiate abuse may act at multiple glial-cell types to further compromise neuron function and survival. The ongoing, reactive cross-talk between opiate drug and HIV-1 co-exposed microglia and astroglia appears to exacerbate critical proinflammatory and excitotoxic events leading to neuron dysfunction, injury, and potentially death. Opiates enhance synaptodendritic damage and a loss of synaptic connectivity, which is viewed as the substrate of cognitive deficits. We especially emphasize that opioid signaling and interactions with HIV-1 are contextual, differing among cell types, and even within subsets of the same cell type. For example, astroglia even within a single brain region are heterogeneous in their expression of µ-, δ-, and κ-opioid receptors, as well as CXCR4 and CCR5, and Toll-like receptors. Thus, defining the distinct targets engaged by opiates in each cell type, and among brain regions, is critical to an understanding of how opiate abuse exacerbates neuroAIDS.
Collapse
Affiliation(s)
- Kurt F Hauser
- Department of Pharmacology and Toxicology, 1217 East Marshall Street, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, USA.
| | | | | | | | | |
Collapse
|
33
|
Porta C, Riboldi E, Totaro MG, Strauss L, Sica A, Mantovani A. Macrophages in cancer and infectious diseases: the 'good' and the 'bad'. Immunotherapy 2012; 3:1185-202. [PMID: 21995571 DOI: 10.2217/imt.11.116] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Macrophages are crucial orchestrators of host defence and tissue homeostasis. Macrophages are heterogeneous and plastic cells that in response to different microenvironmental signals can mount a broad spectrum of different programs of polarized activation. In different pathological contexts including cancer and infectious diseases, macrophages diversity and plasticity may act as a double-edged sword. The elucidation of the molecular mechanisms underlying macrophages recruitment and functional activation allows the identification of valuable targets for the development of innovative therapeutic approaches.
Collapse
Affiliation(s)
- Chiara Porta
- DiSCAFF, University of Piemonte Orientale A. Avogadro, Novara, Italy
| | | | | | | | | | | |
Collapse
|
34
|
Williams DW, Eugenin EA, Calderon TM, Berman JW. Monocyte maturation, HIV susceptibility, and transmigration across the blood brain barrier are critical in HIV neuropathogenesis. J Leukoc Biol 2012; 91:401-15. [PMID: 22227964 DOI: 10.1189/jlb.0811394] [Citation(s) in RCA: 160] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
HIV continues to be a global health crisis with more than 34 million people infected worldwide (UNAIDS: Report on the Global AIDS Epidemic 2010, Geneva, World Health Organization). HIV enters the CNS within 2 weeks of infection and establishes a spectrum of HAND in a large percentage of infected individuals. These neurologic deficits greatly impact the quality of life of those infected with HIV. The establishment of HAND is largely attributed to monocyte transmigration, particularly that of a mature CD14(+)CD16(+) monocyte population, which is more susceptible to HIV infection, across the BBB into the CNS parenchyma in response to chemotactic signals. To enter the CNS, junctional proteins on the monocytes must participate in homo- and heterotypic interactions with those present on BMVECs of the BBB as they transmigrate across the barrier. This transmigration is responsible for bringing virus into the brain and establishing chronic neuroinflammation. While there is baseline trafficking of monocytes into the CNS, the increased chemotactic signals present during HIV infection of the brain promote exuberant monocyte transmigration into the CNS. This review will discuss the mechanisms of monocyte differentiation/maturation, HIV infectivity, and transmigration into the CNS parenchyma that contribute to the establishment of cognitive impairment in HIV-infected individuals. It will focus on markers of monocyte subpopulations, how differentiation/maturation alters HIV infectivity, and the mechanisms that promote their increased transmigration across the BBB into the CNS.
Collapse
Affiliation(s)
- Dionna W Williams
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY, 10461, USA
| | | | | | | |
Collapse
|
35
|
Molecular cloning and characterization of canine fractalkine and its receptor CX3CR1. Vet Immunol Immunopathol 2012; 145:100-9. [DOI: 10.1016/j.vetimm.2011.10.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 10/20/2011] [Accepted: 10/26/2011] [Indexed: 01/22/2023]
|
36
|
Broux B, Pannemans K, Zhang X, Markovic-Plese S, Broekmans T, Eijnde BO, Van Wijmeersch B, Somers V, Geusens P, van der Pol S, van Horssen J, Stinissen P, Hellings N. CX(3)CR1 drives cytotoxic CD4(+)CD28(-) T cells into the brain of multiple sclerosis patients. J Autoimmun 2011; 38:10-9. [PMID: 22123179 DOI: 10.1016/j.jaut.2011.11.006] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2011] [Revised: 11/10/2011] [Accepted: 11/12/2011] [Indexed: 10/25/2022]
Abstract
Immunosenescence, or ageing of the immune system, contributes to the increased morbidity and mortality seen in the elderly population. Premature immunosenescence is shown to occur in a subgroup of patients with autoimmune diseases. One of the main characteristics of immunosenescence is the expansion of CD4(+)CD28(-) T cells in the blood. In this study, we investigate the potential contribution of these cells to disease processes in a subgroup of multiple sclerosis (MS) and rheumatoid arthritis (RA) patients. Characterization of CD4(+)CD28(-) T cells in patients and healthy controls reveals that they have an inflammation-seeking effector-memory T cell phenotype with cytotoxic properties, as they expel cytotoxic granules in response to a polyclonal stimulus or MS-related autoantigens. We identify CX(3)CR1, the fractalkine receptor, as a selective marker to discriminate CD4(+)CD28(-) T cells from their CD4(+)CD28(+) counterparts. CX(3)CR1 expression enables CD4(+)CD28(-) T cells to migrate towards a fractalkine gradient in vitro. In addition, we find increased levels of fractalkine in the cerebrospinal fluid and inflammatory lesions of MS patients. We demonstrate for the first time that CD4(+)CD28(-) T cells accumulate in MS lesions of a subgroup of patients. Moreover, we have indications that these cells are cytotoxic in the target tissue. Overall, our findings suggest that CD4(+)CD28(-) T cells migrate in response to a chemotactic gradient of fractalkine to sites of inflammation, where they contribute to the inflammatory processes in a subgroup of patients with MS and RA.
Collapse
Affiliation(s)
- Bieke Broux
- Hasselt University, Biomedical Research Institute and transnationale Universiteit Limburg, School of Life Sciences, Agoralaan, Building C, 3590 Diepenbeek, Belgium.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Suzuki M, El-Hage N, Zou S, Hahn YK, Sorrell ME, Sturgill JL, Conrad DH, Knapp PE, Hauser KF. Fractalkine/CX3CL1 protects striatal neurons from synergistic morphine and HIV-1 Tat-induced dendritic losses and death. Mol Neurodegener 2011; 6:78. [PMID: 22093090 PMCID: PMC3287119 DOI: 10.1186/1750-1326-6-78] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Accepted: 11/17/2011] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Fractalkine/CX3CL1 and its cognate receptor CX3CR1 are abundantly expressed in the CNS. Fractalkine is an unusual C-X3-C motif chemokine that is important in neuron-microglial communication, a co-receptor for HIV infection, and can be neuroprotective. To assess the effects of fractalkine on opiate-HIV interactive neurotoxicity, wild-type murine striatal neurons were co-cultured with mixed glia from the striata of wild-type or Cx3cr1 knockout mice ± HIV-1 Tat and/or morphine. Time-lapse digital images were continuously recorded at 20 min intervals for up to 72 h using computer-aided microscopy to track the same cells repeatedly. RESULTS Co-exposure to Tat and morphine caused synergistic increases in neuron death, dendritic pruning, and microglial motility as previously reported. Exogenous fractalkine prevented synergistic Tat and morphine-induced dendritic losses and neuron death even though the inflammatory mediator TNF-α remained significantly elevated. Antibody blockade of CX3CR1 mimicked the toxic effects of morphine plus Tat, but did not add to their toxicity; while fractalkine failed to protect wild-type neurons co-cultured with Cx3cr1-/--null glia against morphine and Tat toxicity. Exogenous fractalkine also normalized microglial motility, which is elevated by Tat and morphine co-exposure, presumably limiting microglial surveillance that may lead to toxic effects on neurons. Fractalkine immunofluorescence was expressed in neurons and to a lesser extent by other cell types, whereas CX3CR1 immunoreactivity or GFP fluorescence in cells cultured from the striatum of Cx3cr1-/- (Cx3cr1GFP/GFP) mice were associated with microglia. Immunoblotting shows that fractalkine levels were unchanged following Tat and/or morphine exposure and there was no increase in released fractalkine as determined by ELISA. By contrast, CX3CR1 protein levels were markedly downregulated. CONCLUSIONS The results suggest that deficits in fractalkine-CX3CR1 signaling contribute to the synergistic neurotoxic effects of opioids and Tat. Importantly, exogenous fractalkine can selectively protect neurons from the injurious effects of chronic opioid-HIV-1 Tat co-exposure, and this suggests a potential therapeutic course for neuroAIDS. Although the cellular mechanisms underlying neuroprotection are not certain, findings that exogenous fractalkine reduces microglial motility and fails to protect neurons co-cultured with Cx3cr1-/- mixed glia suggest that fractalkine may act by interfering with toxic microglial-neuron interactions.
Collapse
Affiliation(s)
- Masami Suzuki
- Department of Pharmacology and Toxicology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298-0613, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
|
39
|
Abstract
Microglial activation is an early response to brain ischemia and many other stressors. Microglia continuously monitor and respond to changes in brain homeostasis and to specific signaling molecules expressed or released by neighboring cells. These signaling molecules, including ATP, glutamate, cytokines, prostaglandins, zinc, reactive oxygen species, and HSP60, may induce microglial proliferation and migration to the sites of injury. They also induce a nonspecific innate immune response that may exacerbate acute ischemic injury. This innate immune response includes release of reactive oxygen species, cytokines, and proteases. Microglial activation requires hours to days to fully develop, and thus presents a target for therapeutic intervention with a much longer window of opportunity than acute neuroprotection. Effective agents are now available for blocking both microglial receptor activation and the microglia effector responses that drive the inflammatory response after stroke. Effective agents are also available for targeting the signal transduction mechanisms linking these events. However, the innate immune response can have beneficial as well deleterious effects on outcome after stoke, and a challenge will be to find ways to selectively suppress the deleterious effects of microglial activation after stroke without compromising neurovascular repair and remodeling.
Collapse
Affiliation(s)
- Midori A. Yenari
- Department of Neurology, University of California San Francisco and San Francisco Veterans Affairs Medical Center, 94121 San Francisco, California
| | - Tiina M. Kauppinen
- Department of Neurology, University of California San Francisco and San Francisco Veterans Affairs Medical Center, 94121 San Francisco, California
| | - Raymond A. Swanson
- Department of Neurology, University of California San Francisco and San Francisco Veterans Affairs Medical Center, 94121 San Francisco, California
| |
Collapse
|
40
|
Hu D, Liu J, Keblesh J, Xiong H. Involvement of the 4-aminopyridine-sensitive transient A-type K+ current in macrophage-induced neuronal injury. Eur J Neurosci 2010; 31:214-22. [PMID: 20074219 DOI: 10.1111/j.1460-9568.2009.07063.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Through their capacity to secrete, upon activation, a variety of bioactive molecules, brain macrophages (and resident microglia) play an important role in brain immune and inflammatory responses. To test our hypothesis that activated macrophages induce neuronal injury by enhancing neuronal outward K(+) current, we studied the effects of lipopolysaccharide (LPS)-stimulated human monocyte-derived macrophage (MDM) on neuronal transient A-type K(+) current (I(A)) and resultant neuronal injury in primary rat hippocampal neuronal cultures. Bath application of LPS-stimulated MDM-conditioned media (MCM+) enhanced neuronal I(A) in a concentration-dependent manner. Non-stimulated MCM (MCM-) failed to alter I(A). The enhancement of neuronal I(A) was recapitulated in neurons co-cultured with macrophages. The link of MCM(+)-induced enhancement of I(A) to MCM(+)-associated neuronal injury, as detected by propidium iodide and 4'',6-diamidino-2-phenylindol staining (DAPI) and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, was demonstrated by experimental results showing that addition of I(A) blocker 4-aminopyridine to the cultures protected hippocampal neurons from MCM(+)-induced neuronal injury. Further investigation revealed that glutamate was involved in MCM(+)-induced enhancement of neuronal I(A). These results suggest that during brain inflammation macrophages (and microglia) might mediate neuronal injury via enhancement of neuronal I(A), and that neuronal K(v) channel might be a potential target for the development of therapeutic strategies for some neurodegenerative disorders by which immune and inflammatory responses are believed to be involved in the pathogenesis.
Collapse
Affiliation(s)
- Dehui Hu
- Center for Neurovirology and Neurodegenerative Disorders, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | |
Collapse
|
41
|
Yadav A, Collman RG. CNS inflammation and macrophage/microglial biology associated with HIV-1 infection. J Neuroimmune Pharmacol 2009; 4:430-47. [PMID: 19768553 PMCID: PMC5935112 DOI: 10.1007/s11481-009-9174-2] [Citation(s) in RCA: 151] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Accepted: 09/03/2009] [Indexed: 10/20/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection of the central nervous system (CNS) can result in neurological dysfunction with devastating consequences in a significant proportion of individuals with acquired immune deficiency syndrome. HIV-1 does not infect neurons directly but induces damage indirectly through the accumulation of activated macrophage/microglia (M/M) cells, some of which are infected, that release neurotoxic mediators including both cellular activation products and viral proteins. One mechanism for the accumulation of activated M/M involves the development in infected individuals of an activated peripheral blood monocyte population that traffics through the blood-brain barrier, a process that also serves to carry virus into CNS and establish local infection. A second mechanism involves the release by infected and activated M/M in the CNS of chemotactic mediators that recruit additional monocytes from the periphery. These activated M/M, some of which are infected, release a number of cytokines and small molecule mediators as well as viral proteins that act on bystander cells and in turn activate them, thus amplifying the cascade. These viral proteins and cellular products have neurotoxic properties as well, both directly and through induction of astrocyte dysfunction, which ultimately lead to neuronal injury and death. In patients effectively treated with antiretroviral therapy, frank dementia is now uncommon and has been replaced by milder forms of neurocognitive impairment, with less frequent and more focal neuropathology. This review summarizes key findings that support the critical role and mechanisms of monocyte/macrophage activation and inflammation as a major component for HIV-1 encephalitis or HIV-1 associated dementia.
Collapse
Affiliation(s)
- Anjana Yadav
- Department of Medicine and Center for AIDS Research, University of Pennsylvania School of Medicine, 522 Johnson Pavilion, 36th & Hamilton Walk, Philadelphia, PA 19104, USA
| | | |
Collapse
|
42
|
Expression of fractalkine receptor CX3CR1 on cochlear macrophages influences survival of hair cells following ototoxic injury. J Assoc Res Otolaryngol 2009; 11:223-34. [PMID: 19936834 DOI: 10.1007/s10162-009-0198-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Accepted: 11/02/2009] [Indexed: 12/14/2022] Open
Abstract
The role of innate immunity and macrophage recruitment to the inner ear after hair cell injury is a subject where little is known. In this paper, we demonstrate recruitment of monocytes and macrophages to the inner ear after kanamycin. We also examined the effect of fractalkine receptor (CX3CR1) deletion in kanamycin ototoxicity. We observed more functional and structural damage in CX3CR1 null mice compared to wild-type and heterozygous littermates. In order to determine if increased susceptibility to kanamycin resulted from CX3CR1 deletion from cochlear leukocytes, we created bone marrow chimeras by transplanting CX3CR1-null bone marrow into wild-type mice whose native bone marrow was ablated by lethal irradiation. These mice were then treated with kanamycin sulfate. Auditory brainstem responses (ABR), hair cell counts, and numbers of macrophages recruited to the cochlea were recorded in irradiated mice that received either wild-type, CX3CR1 heterozygous, or CX3CR1 knockout bone marrow. A strong correlation was present between numbers of macrophages and hair cell death in recipients transplanted with CX3CR1 null marrow. No correlation between macrophage number and hair cell loss was present in mice transplanted with wild-type or CX3CR1 heterozygous marrow. We suggest that CX3CR1 plays a role in modulating the detrimental effects of cochlear macrophages after kanamycin ototoxicity. Our data point to the possibility that CX3CR1-deficient cochlear macrophages exacerbate kanamycin ototoxicity while CX3CR1-expressing monocytes do not.
Collapse
|
43
|
Phagocytosis inhibits F-actin-enriched membrane protrusions stimulated by fractalkine (CX3CL1) and colony-stimulating factor 1. Infect Immun 2009; 77:4487-95. [PMID: 19620351 DOI: 10.1128/iai.00530-09] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Cryptococcus neoformans is the only encapsulated human-pathogenic fungus and a facultative intracellular pathogen that can reside in macrophages without host cell lysis. In the present study, we investigated how phagocytosis of C. neoformans affected the macrophage response to chemoattractants such as fractalkine (FKN) (CX3CL1) and colony-stimulating factor 1 (CSF-1). Phagocytosis of immunoglobulin G (IgG)-opsonized C. neoformans and IgG- or C3bi-opsonized sheep erythrocytes was performed using a RAW 264.7 subline (LR5 cells) and bone marrow-derived macrophages (BMM). The chemotactic response to FKN or CSF-1 was quantitated by measurement of the formation of F-actin-enriched membrane protrusions (ruffles), which showed that FKN or CSF-1 stimulated strong transient ruffling in both LR5 cells and BMM. This stimulated cell ruffling was inhibited by phagocytosis in an intracellular-pathogen-number-dependent manner. The inhibition of ruffling was not simply a result of reduced membrane availability since membrane sequestration by sucrose treatment did not inhibit the ruffling response. The phagocytosis process was required to inhibit ruffling as BMM from Fc gamma (-/-) mice that bound C. neoformans but did not ingest it retained the ability to ruffle in response to chemoattractants. These results imply that the inhibition of FKN- or CSF-1-stimulated cell ruffling was a direct consequence of the phagocytosis process. Since cell ruffling is a prelude to chemotaxis, this observation links two functions of macrophages that are critical to host defense, chemotaxis and phagocytosis. Phagocytosis-induced chemotactic suppression may enhance host defense by keeping these antimicrobial effector cells at infected sites and reduce the likelihood of microbial spread by wandering macrophages containing infectious cargo.
Collapse
|
44
|
Singh KK, Spector SA. Host genetic determinants of human immunodeficiency virus infection and disease progression in children. Pediatr Res 2009; 65:55R-63R. [PMID: 19190524 PMCID: PMC2802664 DOI: 10.1203/pdr.0b013e31819dca03] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Increasing data support host genetic factors as an important determinants of human immunodeficiency virus type-1 (HIV-1) susceptibility, mother-to-child transmission (MTCT), and disease progression. Of these genetic mediators, those impacting innate and adaptive immune responses seem to play a critical role in viral infectivity and pathogenesis. During primary infection, CCR5 using virus is predominantly transmitted and polymorphisms that affect the expression of CCR5 alter the risk for MTCT and rate of disease. Chemokines that naturally bind to coreceptors alter infectivity and viral pathogenesis. Additional genes that affect innate immunity including those encoding for MBL2 and those modulating the adaptive immune response including CX3CR1 and human leukocyte antigen types can significantly modify susceptibility and response to HIV-1 infection. As young children develop, the dependence on certain arms of the immune system varies and can alter the effect of genetic variants. Additionally, host genetic factors may alter the response to antiretrovirals. Finally, because HIV-infected children progress more rapidly than adults and have fewer background cofactors, such as drug use and coinfections, the effects of host factors on HIV-1 disease may be more clearly identified. In this review, we summarize available data on the impact of host genetics on MTCT and disease progression of HIV-infected children.
Collapse
Affiliation(s)
- Kumud K Singh
- Department of Pediatrics, University of California, San Diego, La Jolla, California 92093, USA
| | | |
Collapse
|
45
|
New evidences for fractalkine/CX3CL1 involved in substantia nigral microglial activation and behavioral changes in a rat model of Parkinson's disease. Neurobiol Aging 2009; 32:443-58. [PMID: 19368990 DOI: 10.1016/j.neurobiolaging.2009.03.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2008] [Revised: 02/13/2009] [Accepted: 03/05/2009] [Indexed: 11/23/2022]
Abstract
Activated microglia are instrumental to neurodegeneration in Parkinson's disease (PD). Fractalkine, as an exclusive ligand for CX3CR1 expressed on microglia, has recently been reported to be released out by neurons, and induce microglial activation as a neuron-to-glia signal in the spinal cord. However, the role of fractalkine-induced microglial activation in PD remains unknown. In our study, we injected 1-methyl-4-phenylpyridinium (MPP(+)) into unilateral substantia nigra (SN) which induced ipsilateral endogenous fractalkine expression on neuron and observe the increase of CX3CR1 expression in response to MPP(+) by Western blotting analysis. Moreover, pre-administration of anti-CX3CR1 neutralizing antibody which potentially blocked microglial activation can promote rotation behaviors. To further investigate the role of fractalkine in PD, we injected exogenous fractalkine in unilateral SN, and observed microglial activation, dopaminergic cell depletion, and motor dysfunction. All these effects can be totally abolished by cerebroventricular administration of anti-CX3CR1. Intracerebroventricular administration of minocycline, a selective microglia inhibitor, can prevent fractalkine-induced rotation behaviors, and inhibit dopaminergic neurons from degeneration in the way of dose-dependent. Our studies demonstrate that fractalkine-induced microglial activation plays an important role in the development of PD, and provide an evidence of fractalkine and CX3CR1 as new therapeutic targets for PD treatment.
Collapse
|
46
|
Ancuta P, Wang J, Gabuzda D. CD16+ monocytes produce IL-6, CCL2, and matrix metalloproteinase-9 upon interaction with CX3CL1-expressing endothelial cells. J Leukoc Biol 2007; 80:1156-64. [PMID: 17056766 DOI: 10.1189/jlb.0206125] [Citation(s) in RCA: 144] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The CD16+ subset of peripheral blood monocytes (Mo) is expanded dramatically during inflammatory conditions including sepsis, HIV-1 infection, and cancer. CD16+ express high levels of CX3CR1, which mediates arrest onto CX3CL1-expressing endothelial cells (EC) under flow conditions. In contrast, attachment of CD16- Mo onto cytokine-activated EC is independent of CX3CL1. Here, we investigate the ability of CD16+ and CD16- Mo to produce proinflammatory cytokines upon interaction with CX3CL1-expressing HUVEC. We demonstrate that CD16+ but not CD16- Mo produce high levels of IL-6, CCL2, and matrix metalloproteinase (MMP)-9 when cocultured with TNF/IFN-gamma-activated HUVEC or nonactivated HUVEC expressing CX3CL1. Furthermore, supernatants from Mo cocultured with cytokine-activated HUVEC induce neuronal death in vitro. These results suggest that membrane-bound CX3CL1 stimulates production of IL-6, CCL2, and MMP-9 by CD16+ Mo, likely via engagement of CX3CR1. Thus, expansion of CD16+ Mo and their accumulation onto CX3CL1-expressing EC may result in recruitment of Mo and T cell subsets at sites of inflammation in response to CCL2, IL-6-induced cell activation and/or differentiation, and MMP-9-mediated vascular and tissue injury.
Collapse
Affiliation(s)
- Petronela Ancuta
- Department of Cancer Immunology, and AIDS, Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | | | | |
Collapse
|
47
|
Clay CC, Rodrigues DS, Ho YS, Fallert BA, Janatpour K, Reinhart TA, Esser U. Neuroinvasion of fluorescein-positive monocytes in acute simian immunodeficiency virus infection. J Virol 2007; 81:12040-8. [PMID: 17715237 PMCID: PMC2168770 DOI: 10.1128/jvi.00133-07] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Monocytes and macrophages play a central role in the pathogenesis of human immunodeficiency virus (HIV)-associated dementia. They represent prominent targets for HIV infection and are thought to facilitate viral neuroinvasion and neuroinflammatory processes. However, many aspects regarding monocyte brain recruitment in HIV infection remain undefined. The nonhuman primate model of AIDS is uniquely suited for examination of the role of monocytes in the pathogenesis of AIDS-associated encephalitis. Nevertheless, an approach to monitor cell migration from peripheral blood into the central nervous system (CNS) in primates had been lacking. Here, upon autologous transfer of fluorescein dye-labeled leukocytes, we demonstrate the trafficking of dye-positive monocytes into the choroid plexus stromata and perivascular spaces in the cerebra of rhesus macaques acutely infected with simian immunodeficiency virus between days 12 and 14 postinfection (p.i.). Dye-positive cells that had migrated expressed the monocyte activation marker CD16 and the macrophage marker CD68. Monocyte neuroinvasion coincided with the presence of the virus in brain tissue and cerebrospinal fluid and with the induction of the proinflammatory mediators CXCL9/MIG and CCL2/MCP-1 in the CNS. Prior to neuroinfiltration, plasma viral load levels peaked on day 11 p.i. Furthermore, the numbers of peripheral blood monocytes rapidly increased between days 4 and 8 p.i., and circulating monocytes exhibited increased functional capacity to produce CCL2/MCP-1. Our findings demonstrate acute monocyte brain infiltration in an animal model of AIDS. Such studies facilitate future examinations of the migratory profile of CNS-homing monocytes, the role of monocytes in virus import into the brain, and the disruption of blood-cerebrospinal fluid and blood-brain barrier functions in primates.
Collapse
Affiliation(s)
- Candice C Clay
- Department of Pathology and Laboratory Medicine, Research III Building, Room 3400A, University of California-Davis Medical Center, 4645 2nd Avenue, Sacramento, CA 95817, USA
| | | | | | | | | | | | | |
Collapse
|
48
|
Passam AM, Sourvinos G, Krambovitis E, Miyakis S, Stavrianeas N, Zagoreos I, Spandidos DA. Polymorphisms of Cx(3)CR1 and CXCR6 receptors in relation to HAART therapy of HIV type 1 patients. AIDS Res Hum Retroviruses 2007; 23:1026-32. [PMID: 17725420 DOI: 10.1089/aid.2006.0248] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The chemokine polymorphisms CXCR6-3E/K, In1.1T/C, H7 haplotype, CX(3)CR1-V249I, and CX(3)CR1-T280M have been shown to affect the course of HIV infection. We studied their influence on immunologic and virologic response to HAART in a group of 143 HIV-1 patients. We performed Kaplan-Meier analysis using the following end-point criteria: (1) time from HAART initiation to undetectable viral load (VL < 50 copies/ml), (2) maximum duration of viral suppression, (3) time from HAART administration until CD4 elevation above 200 cells/microl for patients with baseline CD4 below 200 cells/microl and above 500 cells/microl for patients with baseline CD4 between 200 and 500 cells/microl, respectively, and (4) time from HAART initiation until CD4 reduction below baseline values. Our results revealed an improved immunologic response to HAART in patients with the CX(3)CR1-249I or CX(3)CR1-280M allele. On the contrary, patients with initial VL suppression due to HAART showed a faster virologic failure in the presence of the CXCR6-3K allele. The In1.1T/C polymorphism and H7 haplotype did not reveal any specific effect on HAART response.
Collapse
Affiliation(s)
- A M Passam
- Department of Virology, Medical School, University of Crete, Heraklion, Crete, Greece
| | | | | | | | | | | | | |
Collapse
|
49
|
Irvine E, Keblesh J, Liu J, Xiong H. Voltage-gated potassium channel modulation of neurotoxic activity in human immunodeficiency virus type-1(HIV-1)-infected macrophages. J Neuroimmune Pharmacol 2007; 2:265-9. [PMID: 18040860 DOI: 10.1007/s11481-007-9072-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2007] [Accepted: 03/15/2007] [Indexed: 11/30/2022]
Abstract
Macrophages play an important role in brain immune and inflammatory responses. They are also critical cells in mediating the pathology of neurodegenerative disorders such as HIV-associated dementia. This is largely through their capacity to secrete a variety of bioactive molecules such as cytokines, leading to neuronal dysfunction and/or death. Accumulating evidence indicates that voltage-gated potassium (Kv) channels play a pivotal role in the modulation of macrophage proliferation, activation, and secretion. Blockade of Kv channels by specific antagonists decreases macrophage cytokine production and ameliorates macrophage-associated neuronal injury. These results suggest that Kv channels might become a potential target for the development of new therapeutic strategies for chronic inflammatory diseases.
Collapse
Affiliation(s)
- Elizabeth Irvine
- Neurophysiology Laboratory, the Center for Neurovirology and Neurodegenerative Disorders, and Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880, USA
| | | | | | | |
Collapse
|
50
|
Cinque P, Brew BJ, Gisslen M, Hagberg L, Price RW. Cerebrospinal fluid markers in central nervous system HIV infection and AIDS dementia complex. HANDBOOK OF CLINICAL NEUROLOGY 2007; 85:261-300. [PMID: 18808988 DOI: 10.1016/s0072-9752(07)85017-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Affiliation(s)
- Paola Cinque
- Clinic of Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | |
Collapse
|