Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Astrid E. Cardona is active.

Publication


Featured researches published by Astrid E. Cardona.


Nature Neuroscience | 2006

Control of microglial neurotoxicity by the fractalkine receptor

Astrid E. Cardona; Erik P. Pioro; Margaret E. Sasse; Volodymyr Kostenko; Sandra M. Cardona; Ineke M. Dijkstra; De Ren Huang; Grahame J. Kidd; Stephen M. Dombrowski; Ranjan Dutta; Jar Chi Lee; Donald N. Cook; Steffen Jung; Sergio A. Lira; Dan R. Littman; Richard M. Ransohoff

Microglia, the resident inflammatory cells of the CNS, are the only CNS cells that express the fractalkine receptor (CX3CR1). Using three different in vivo models, we show that CX3CR1 deficiency dysregulates microglial responses, resulting in neurotoxicity. Following peripheral lipopolysaccharide injections, Cx3cr1−/− mice showed cell-autonomous microglial neurotoxicity. In a toxic model of Parkinson disease and a transgenic model of amyotrophic lateral sclerosis, Cx3cr1−/− mice showed more extensive neuronal cell loss than Cx3cr1+ littermate controls. Augmenting CX3CR1 signaling may protect against microglial neurotoxicity, whereas CNS penetration by pharmaceutical CX3CR1 antagonists could increase neuronal vulnerability.


Nature | 2010

The myeloid cells of the central nervous system parenchyma

Richard M. Ransohoff; Astrid E. Cardona

A microglial cell is both a glial cell of the central nervous system and a mononuclear phagocyte, which belongs to the haematopoietic system and is involved in inflammatory and immune responses. As such, microglia face a challenging task. The neurons of the central nervous system cannot divide and be replenished, and therefore need to be protected against pathogens, which is a key role of the immune system, but without collateral damage. In addition, after physical injury, neural cells need restorative support, which is provided by inflammatory responses. Excessive or chronic inflammatory responses can, however, be harmful. How microglia balance these demands, and how their behaviour can be modified to ameliorate disorders of the central nervous system, is becoming clear.


PLOS ONE | 2010

Selective Chemokine Receptor Usage by Central Nervous System Myeloid Cells in CCR2-Red Fluorescent Protein Knock-In Mice

Noah Saederup; Astrid E. Cardona; Kelsey Croft; Makiko Mizutani; Anne C. Cotleur; Chia Lin Tsou; Richard M. Ransohoff; Israel F. Charo

Background Monocyte subpopulations distinguished by differential expression of chemokine receptors CCR2 and CX3CR1 are difficult to track in vivo, partly due to lack of CCR2 reagents. Methodology/Principal Findings We created CCR2-red fluorescent protein (RFP) knock-in mice and crossed them with CX3CR1-GFP mice to investigate monocyte subset trafficking. In mice with experimental autoimmune encephalomyelitis, CCR2 was critical for efficient intrathecal accumulation and localization of Ly6Chi/CCR2hi monocytes. Surprisingly, neutrophils, not Ly6Clo monocytes, largely replaced Ly6Chi cells in the central nervous system of these mice. CCR2-RFP expression allowed the first unequivocal distinction between infiltrating monocytes/macrophages from resident microglia. Conclusion/Significance These results refine the concept of monocyte subsets, provide mechanistic insight about monocyte entry into the central nervous system, and present a novel model for imaging and quantifying inflammatory myeloid populations.


American Journal of Pathology | 2010

CX3CR1 Deficiency Alters Microglial Activation and Reduces Beta-Amyloid Deposition in Two Alzheimer's Disease Mouse Models

Sungho Lee; Megan E. Konerth; Guixiang Xu; Astrid E. Cardona; Richard M. Ransohoff; Bruce T. Lamb

Microglia, the primary immune effector cells in the brain, continually monitor the tissue parenchyma for pathological alterations and become activated in Alzheimers disease. Loss of signaling between neurons and microglia via deletion of the microglial receptor, CX3CR1, worsens phenotypes in various models of neurodegenerative diseases. In contrast, CX3CR1 deficiency ameliorates pathology in murine stroke models. To examine the role of CX3CR1 in Alzheimers disease-related β-amyloid pathology, we generated APPPS1 and R1.40 transgenic mouse models of Alzheimers disease deficient for CX3CR1. Surprisingly, CX3CR1 deficiency resulted in a gene dose-dependent reduction in β-amyloid deposition in both the APPPS1 and R1.40 mouse models of AD. Immunohistochemical analysis revealed reduced staining for CD68, a marker of microglial activation. Furthermore, quantitative immunohistochemical analysis revealed reduced numbers of microglia surrounding β-amyloid deposits in the CX3CR1-deficient APPPS1 animals. The reduced β-amyloid pathology correlated with reduced levels of TNFα and CCL2 mRNAs, but elevated IL1β mRNA levels, suggesting an altered neuroinflammatory milieu. Finally, to account for these seemingly disparate results, both in vitro and in vivo studies provided evidence that CX3CL1/CX3CR1 signaling alters the phagocytic capacity of microglia, including the uptake of Aβ fibrils. Taken together, these results demonstrate that loss of neuron-microglial fractalkine signaling leads to reduced β-amyloid deposition in mouse models of AD that is potentially mediated by altered activation and phagocytic capability of CX3CR1-deficient microglia.


Journal of Immunology | 2012

The Fractalkine Receptor but Not CCR2 Is Present on Microglia from Embryonic Development throughout Adulthood

Makiko Mizutani; Paula A. Pino; Noah Saederup; Israel F. Charo; Richard M. Ransohoff; Astrid E. Cardona

Microglial cells are difficult to track during development because of the lack of specific reagents for myeloid subpopulations. To further understand how myeloid lineages differentiate during development to create microglial cells, we investigated CX3CR1 and CCR2 transcription unit activation in Cx3cr1+/GFPCCR2+/RFP knockin fluorescent protein reporter mice. The principal findings include: 1) CX3CR1+ cells localized to the aorta–gonad–mesonephros region, and visualized at embryonic day (E)9.0 in the yolk sac and neuroectoderm; 2) at E10.5, CX3CR1 single-positive microglial cells were visualized penetrating the neuroepithelium; and 3) CX3CR1 and CCR2 distinguished infiltrating macrophages from resident surveillant or activated microglia within tissue sections and by flow cytometric analyses. Our results support the contribution of the yolk sac as a source of microglial precursors. We provide a novel model to monitor chemokine receptor expression changes in microglia and myeloid cells early (E8.0–E10.5) in development and during inflammatory conditions, which have been challenging to visualize in mammalian tissues.


The FASEB Journal | 2005

Chronic expression of monocyte chemoattractant protein-1 in the central nervous system causes delayed encephalopathy and impaired microglial function in mice

DeRen Huang; Jerome Wujek; Graham Kidd; Toby T. He; Astrid E. Cardona; Margaret E. Sasse; Erica Stein; Jacqueline Kish; Marie Tani; Israel F. Charo; Amanda E. I. Proudfoot; Barrett J. Rollins; Tracy M. Handel; Richard M. Ransohoff

Increased central nervous system (CNS) levels of monocyte chemoattractant protein 1 [CC chemokine ligand 2 (CCL2) in the systematic nomenclature] have been reported in chronic neurological diseases such as human immunodeficiency virus type 1‐associated dementia, amyotrophic lateral sclerosis, and multiple sclerosis. However, a pathogenic role for CCL2 has not been confirmed, and there is no established model for the effects of chronic CCL2 expression on resident and recruited CNS cells. We report that aged (>6 months) transgenic (tg) mice expressing CCL2 under the control of the human glial fibrillary acidic protein promoter (huGFAP‐CCL2hi tg+ mice) manifested encephalopathy with mild perivascular leukocyte infiltration, impaired blood brain barrier function, and increased CD45‐immunoreactive microglia, which had morphologic features of activation. huGFAP‐CCL2hi tg+ mice lacking CC chemokine receptor 2 (CCR2) were normal, showing that chemokine action via CCR2 was required. Studies of cortical slice preparations using video confocal microscopy showed that microglia in the CNS of huGFAP‐CCL2hi tg+ mice were defective in expressing amoeboid morphology. Treatment with mutant CCL2 peptides, a receptor antagonist and an obligate monomer, also suppressed morphological transformation in this assay, indicating a critical role for CCL2 in microglial activation and suggesting that chronic CCL2 exposure desensitized CCR2 on microglia, which in the CNS of huGFAP‐CCL2hi tg+ mice, did not up‐regulate cell‐surface expression of major histocompatibility complex class II, CD11b, CD11c, or CD40, in contrast to recruited perivascular macrophages that expressed enhanced levels of these markers. These results indicate that huGFAP‐CCL2hi tg+ mice provide a useful model to study how chronic CNS expression of CCL2 alters microglial function and CNS physiology.—Huang, D., Wujek, J., Kidd, G., He, T. T., Cardona, A., Sasse, M. E., Stein, E. J., Kish, J., Tani, M., Charo, I. F., Proudfoot, A. E., Rollins, B. J., Handel, T., Ransohoff, R. M. Chronic expression of monocyte chemoattractant protein‐1 in the central nervous system causes delayed encephalopathy and impaired microglial function in mice. FASEB J. 19, 761–772 (2005)


Nature Protocols | 2006

Isolation of murine microglial cells for RNA analysis or flow cytometry

Astrid E. Cardona; DeRen Huang; Margaret E. Sasse; Richard M. Ransohoff

There is increasing interest in the isolation of adult microglia to study their functions at a morphological and molecular level during normal and neuroinflammatory conditions. Microglia have important roles in brain homeostasis, and in disease states they exert neuroprotective or neurodegenerative functions. To assay expression profiles or functions of microglia, we have developed a method to isolate microglial cells and infiltrating leukocytes from adult mouse brain. This protocol uses a digestion cocktail containing collagenase and dispase, and it involves separation over discontinuous percoll gradients. Isolated cells can be used for RNA analysis, including RNase protection analysis (RPA), quantitative RT-PCR, high-density microarray, proteomic or flow cytometric characterization of cell surface markers or adoptive transfer. Cell isolation can be completed in less than 4 h.


International Review of Neurobiology | 2007

Chemokines and Chemokine Receptors: Multipurpose Players in Neuroinflammation

Richard M. Ransohoff; LiPing Liu; Astrid E. Cardona

Chemokines were detected by virtue of chemotactic effects toward neutrophils in the late 1970s. During subsequent decades, it has become clear that their primordial role in vertebrate biology was to facilitate organogenesis, with particularly important functions in the central nervous system (CNS). In common with other developmentally relevant factors, chemokines and their G-protein-coupled receptors continue to be expressed in the adult CNS as neuromodulators. In our progress toward chemokine receptor blockade for treatment of inflammatory and infectious diseases, the CNS physiology of the chemokine system will need to be a material consideration. In some cases, the dual functions of the chemokine system in the periphery and in the CNS offer unique possibilities for disease treatment.


Brain | 2015

Reactive microglia drive tau pathology and contribute to the spreading of pathological tau in the brain

Nicole Maphis; Guixiang Xu; Olga N. Kokiko-Cochran; Shanya Jiang; Astrid E. Cardona; Richard M. Ransohoff; Bruce T. Lamb; Kiran Bhaskar

Pathological aggregation of tau is a hallmark of Alzheimers disease and related tauopathies. We have previously shown that the deficiency of the microglial fractalkine receptor (CX3CR1) led to the acceleration of tau pathology and memory impairment in an hTau mouse model of tauopathy. Here, we show that microglia drive tau pathology in a cell-autonomous manner. First, tau hyperphosphorylation and aggregation occur as early as 2 months of age in hTauCx3cr1(-/-) mice. Second, CD45(+) microglial activation correlates with the spatial memory deficit and spread of tau pathology in the anatomically connected regions of the hippocampus. Third, adoptive transfer of purified microglia derived from hTauCx3cr1(-/-) mice induces tau hyperphosphorylation within the brains of non-transgenic recipient mice. Finally, inclusion of interleukin 1 receptor antagonist (Kineret®) in the adoptive transfer inoculum significantly reduces microglia-induced tau pathology. Together, our results suggest that reactive microglia are sufficient to drive tau pathology and correlate with the spread of pathological tau in the brain.


Blood | 2008

Scavenging roles of chemokine receptors: chemokine receptor deficiency is associated with increased levels of ligand in circulation and tissues

Astrid E. Cardona; Margaret E. Sasse; LiPing Liu; Sandra M. Cardona; Makiko Mizutani; Carine Savarin; Taofang Hu; Richard M. Ransohoff

In vitro studies have implicated chemokine receptors in consumption and clearance of specific ligands. We studied the role that various signaling chemokine receptors play during ligand homeostasis in vivo. We examined the levels of ligands in serum and CNS tissue in mice lacking chemokine receptors. Compared with receptor-sufficient controls, Cx3cr1(-/-) mice exhibited augmented levels of CX3CL1 both in serum and brain, and circulating levels of CXCL1 and CXCL2 were increased in Cxcr2(-/-) mice. CCR2-deficient mice showed significantly increased amounts of circulating CCL2 compared with wild-type mice. Cxcr3(-/-) mice revealed increased levels of circulating and brain CXCL10 after experimental autoimmune encephalomyelitis (EAE) induction. CCR2-deficient peripheral blood and resident peritoneal cells exhibited reduced binding capacity and biologic responses to the CCR1 ligand CCL3, suggesting that elevated levels of CCR2 ligands had down-regulated CCR1. The results indicate that signaling chemokine receptors clear chemokines from circulation and tissues. These homeostatic functions of signaling chemokine receptors need to be integrated into safety and efficacy calculations when considering therapeutic receptor blockade.

Collaboration


Dive into the Astrid E. Cardona's collaboration.

Top Co-Authors

Avatar

Sandra M. Cardona

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Jenny A. Garcia

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew S. Mendiola

University of Texas at San Antonio

View shared research outputs
Top Co-Authors

Avatar

Kiran Bhaskar

University of New Mexico

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge