Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Fluids Barriers CNS ; 20(1): 74, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37858244

ABSTRACT

The meninges surround the brain and spinal cord, affording physical protection while also serving as a niche of neuroimmune activity. Though possessing stromal qualities, its complex cellular and extracellular makeup has yet to be elaborated, and it remains unclear whether the meninges vary along the neuroaxis. Hence, studies were carried-out to elucidate the protein composition and structural organization of brain and spinal cord meninges in normal, adult Biozzi ABH mice. First, shotgun, bottom-up proteomics was carried-out. Prominent proteins at both brain and spinal levels included Type II collagen and Type II keratins, representing extracellular matrix (ECM) and cytoskeletal categories, respectively. While the vast majority of total proteins detected was shared between both meningeal locales, more were uniquely detected in brain than in spine. This pattern was also seen when total proteins were subdivided by cellular compartment, except in the case of the ECM category where brain and spinal meninges each had near equal number of unique proteins, and Type V and type III collagen registered exclusively in the spine. Quantitative analysis revealed differential expression of several collagens and cytoskeletal proteins between brain and spinal meninges. High-resolution immunofluorescence and immunogold-scanning electronmicroscopy on sections from whole brain and spinal cord - still encased within bone -identified major proteins detected by proteomics, and highlighted their association with cellular and extracellular elements of variously shaped arachnoid trabeculae. Western blotting aligned with the proteomic and immunohistological analyses, reinforcing differential appearance of proteins in brain vs spinal meninges. Results could reflect regional distinctions in meninges that govern protective and/or neuroimmune functions.


Subject(s)
Meninges , Proteomics , Mice , Animals , Mice, Biozzi , Meninges/metabolism , Spinal Cord/metabolism , Brain
2.
J Plant Res ; 136(1): 139-156, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36520245

ABSTRACT

Aster tataricus (L.) is an important medicinal plant in China. Its roots are rich in flavonoids, the main medicinal components. However, the molecular basis of flavonoid biosynthesis in the roots of A. tataricus remains unclear. In this study, the content of total flavonoid of A. tataricus roots at different developmental stages was measured first, and the results showed that the content of total flavonoid gradually decreased from September to November, which may be caused by the stagnation of A. tataricus growth due to the decrease in temperature after September. Then, an integrated analysis of transcriptome and metabolome was conducted on five developing stages of A. tataricus roots to identify flavonoid compositions and potential genes involved in flavonoid biosynthesis. A total of 80 flavonoid metabolites, of which 75% were flavonols and flavonoids, were identified in metabolomic analyses, among which isorhamnetin, kaempferol, quercetin, and myricetin were the main skeletons of these flavonoids. Cluster analysis divided these 80 flavonoids into 3 clusters. The compounds in cluster I mainly accumulated in S1, S3, and S5. In cluster II, the relative content of the flavonoid metabolites showed an upward trend from S2 to S4. In cluster III, the flavonoids decreased from S1 to S5. A total of 129 structural genes, including 43 PAL, 23 4CL, 9 C4H, 4 CHS, 18 CHI, 3 F3H, 5 F3'H, 1 F3'5'H, 21 FLS, and 2 FSII, and 65 transcription factors, including 22 AP2/ERF, 7 bHLH, 5 bZIP, 8 MYB, 11 NAC, and 12 WRKY, showed significant correlation with total flavonoid content. Eighteen genes (7 4CL, 5 C4H, 2 CHI, 1 F3H, and 3 FLS) and 30 genes (5 PAL, 9 4CL, 1 C4H, 2 CHI, 1 F3H, 1 DFR, 7 3AT, 1 BZ1, and 3 UGT79B1) were identified as key structural genes for kaempferol and anthocyanins biosynthesis, respectively. Our study provides valuable information for understanding the mechanism of flavonoid biosynthesis in A. tataricus root.


Subject(s)
Kaempferols , Transcriptome , Anthocyanins , Flavonoids/metabolism , Metabolomics , Gene Expression Regulation, Plant
3.
J Neuroinflammation ; 18(1): 296, 2021 Dec 21.
Article in English | MEDLINE | ID: mdl-34933669

ABSTRACT

BACKGROUND: Tight junctions (TJs) are membrane specializations characteristic of barrier-forming membranes, which function to seal the aqueous pathway between endothelial cells or epithelial cells and, thereby, obstruct intercellular solute and cellular movement. However, previous work from our laboratory found that claudin-5 (CLN-5), a TJ protein prominent at the blood-brain barrier (BBB), was also detected, ectopically, on leukocytes (CLN-5+) in the blood and central nervous system (CNS) of mice with experimental autoimmune encephalomyelitis (EAE), a neuroinflammatory, demyelinating disease that is a model for multiple sclerosis. CLN-5 was further shown to be transferred from endothelial cells to circulating leukocytes during disease, prompting consideration this action is coupled to leukocyte transendothelial migration (TEM) into the CNS by fostering transient interactions between corresponding leukocyte and endothelial junctional proteins at the BBB. METHODS: To begin clarifying the significance of CLN-5+ leukocytes, flow cytometry was used to determine their appearance in the blood and CNS during EAE. RESULTS: Flow cytometric analysis revealed CLN-5+ populations among CD4 and CD8 T cells, B cells, monocytes and neutrophils, and these appeared with varying kinetics and to different extents in both blood and CNS. CLN-5 levels on circulating T cells further correlated highly with activation state. And, the percentage of CLN-5+ cells among each of the subtypes analyzed was considerably higher in CNS tissue than in blood, consistent with the interpretation that CLN-5+ leukocytes gain preferred access to the CNS. CONCLUSION: Several leukocyte subtypes variably acquire CLN-5 in blood before they enter the CNS, an event that may represent a novel mechanism to guide leukocytes to sites for paracellular diapedesis across the BBB.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/pathology , Claudin-5/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Leukocytes/pathology , Animals , Blood-Brain Barrier/metabolism , Claudin-5/blood , Claudin-5/metabolism , Female , Flow Cytometry , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases/genetics , Neuroinflammatory Diseases/metabolism , Neuroinflammatory Diseases/pathology , Tight Junction Proteins/metabolism
4.
Fluids Barriers CNS ; 16(1): 18, 2019 Jul 01.
Article in English | MEDLINE | ID: mdl-31256757

ABSTRACT

BACKGROUND: Immune cell trafficking into the CNS is considered to contribute to pathogenesis in MS and its animal model, EAE. Disruption of the blood-brain barrier (BBB) is a hallmark of these pathologies and a potential target of therapeutics. Human embryonic stem cell-derived mesenchymal stem/stromal cells (hES-MSCs) have shown superior therapeutic efficacy, compared to bone marrow-derived MSCs, in reducing clinical symptoms and neuropathology of EAE. However, it has not yet been reported whether hES-MSCs inhibit and/or repair the BBB damage associated with neuroinflammation that accompanies EAE. METHODS: BMECs were cultured on Transwell inserts as a BBB model for all the experiments. Disruption of BBB models was induced by TNF-α, a pro-inflammatory cytokine that is a hallmark of acute and chronic neuroinflammation. RESULTS: Results indicated that hES-MSCs reversed the TNF-α-induced changes in tight junction proteins, permeability, transendothelial electrical resistance, and expression of adhesion molecules, especially when these cells were placed in direct contact with BMEC. CONCLUSIONS: hES-MSCs and/or products derived from them could potentially serve as novel therapeutics to repair BBB disturbances in MS.


Subject(s)
Blood-Brain Barrier/metabolism , Embryonic Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Blood-Brain Barrier/cytology , Blood-Brain Barrier/drug effects , Cell Line, Transformed , Embryonic Stem Cells/drug effects , Humans , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Models, Biological
5.
Neurobiol Dis ; 108: 159-172, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28844788

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) induced by active immunization of C57BL/6 mice with peptide from myelin oligodendrocyte protein (MOG35-55), is a neuroinflammatory, demyelinating disease widely recognized as an animal model of multiple sclerosis (MS). Typically, EAE presents with an ascending course of paralysis, and inflammation that is predominantly localized to the spinal cord. Recent studies have further indicated that inflammation - in both MS and EAE - might initiate within the meninges and propagate from there to the underlying parenchyma. However, the patterns of inflammation within the respective meningeal and parenchymal compartments along the length of the spinal cord, and the progression with which these patterns develop during EAE, have yet to be detailed. Such analysis could hold key to identifying factors critical for spreading, as well as constraining, inflammation along the neuraxis. To address this issue, high-resolution 3-dimensional (3D) confocal microscopy was performed to visualize, in detail, the sequence of leukocyte infiltration at distinct regions of the spinal cord. High quality virtual slide scanning for imaging the entire spinal cord using epifluorescence was further conducted to highlight the directionality and relative degree of inflammation. Meningeal inflammation was found to precede parenchymal inflammation at all levels of the spinal cord, but did not develop equally or simultaneously throughout the subarachnoid space (SAS) of the meninges. Instead, meningeal inflammation was initially most obvious in the caudal SAS, from which it progressed to the immediate underlying parenchyma, paralleling the first signs of clinical disease in the tail and hind limbs. Meningeal inflammation could then be seen to extend in the caudal-to-rostral direction, followed by a similar, but delayed, trajectory of parenchymal inflammation. To additionally determine whether the course of ascending paralysis and leukocyte infiltration during EAE is reflected in differences in inflammatory gene expression by meningeal and parenchymal microvessels along the spinal cord, laser capture microdissection (LCM) coupled with gene expression profiling was performed. Expression profiles varied between these respective vessel populations at both the cervical and caudal levels of the spinal cord during disease progression, and within each vessel population at different levels of the cord at a given time during disease. These results reinforce a significant role for the meninges in the development and propagation of central nervous system inflammation associated with MS and EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Meninges/immunology , Parenchymal Tissue/immunology , Animals , Cervical Vertebrae , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Gene Expression , Inflammation/pathology , Inflammation/physiopathology , Leukocytes/immunology , Leukocytes/pathology , Lumbar Vertebrae , Meninges/pathology , Mice, Inbred C57BL , Microvessels/immunology , Microvessels/pathology , Myelin-Oligodendrocyte Glycoprotein , Parenchymal Tissue/pathology , Peptide Fragments , Spinal Cord/immunology , Spinal Cord/pathology
6.
J Neuroinflammation ; 13(1): 292, 2016 11 16.
Article in English | MEDLINE | ID: mdl-27852330

ABSTRACT

BACKGROUND: The mechanism of leukocyte transendothelial migration (TEM) across the highly restrictive blood-brain barrier (BBB) remains enigmatic, with paracellular TEM thought to require leukocytes to somehow navigate the obstructive endothelial tight junctions (TJs). Transient interactions between TJ proteins on the respective leukocyte and endothelial surfaces have been proposed as one mechanism for TEM. Given the expanding role of extracellular vesicles (EVs) in intercellular communication, we investigated whether EVs derived from brain microvascular endothelial cells (BMEC) of the BBB may play a role in transferring a major TJ protein, claudin-5 (CLN-5), to leukocytes as a possible basis for such a mechanism during neuroinflammation. METHODS: High-resolution 3D confocal imaging was used to highlight CLN-5 immunoreactivity in the central nervous system (CNS) and on leukocytes of mice with the neuroinflammatory condition experimental autoimmune encephalomyelitis (EAE). Both Western blotting of circulating leukocytes from wild-type mice and fluorescence imaging of leukocyte-associated eGFP-CLN-5 in the blood and CNS of endothelial-targeted, Tie-2-eGFP-CLN-5 transgenic mice were used to confirm the presence of CLN-5 protein on these cells. EVs were isolated from TNF-α-stimulated BMEC cultures and blood plasma of Tie-2-eGFP-CLN-5 mice with EAE and evaluated for CLN-5 protein by Western blotting and fluorescence-activated cell sorting (FACS), respectively. Confocal imaging and FACS were used to detect binding of endothelial-derived EVs from these two sources to leukocytes in vitro. Serial electron microscopy (serial EM) and 3D contour-based surface reconstruction were employed to view EV-like structures at the leukocyte:BBB interface in situ in inflamed CNS microvessels. RESULTS: A subpopulation of leukocytes immunoreactive for CLN-5 on their surface was seen to infiltrate the CNS of mice with EAE and reside in close apposition to inflamed vessels. Confocal imaging of immunostained samples and Western blotting established the presence of CLN-5+ leukocytes in blood as well, implying these cells are present prior to TEM. Moreover, imaging of inflamed CNS vessels and the associated perivascular cell infiltrates from Tie-2-eGFP-CLN-5 mice with EAE revealed leukocytes bearing the eGFP label, further supporting the hypothesis CLN-5 is transferred from endothelial cells to circulating leukocytes in vivo. Western blotting of BMEC-derived EVs, corresponding in size to both exosomes and microvesicles, and FACS analysis of plasma-derived EVs from Tie-2-eGFP-CLN-5 mice with EAE validated expression of CLN-5 by EVs of endothelial origin. Confocal imaging and FACS further revealed both PKH-67-labeled EVs from cultured BMECs and eGFP-CLN-5+ EVs from plasma of Tie-2-eGFP-CLN-5 mice with EAE can bind to leukocytes. Lastly, serial EM and 3D contour-based surface reconstruction revealed a close association of EV-like structures between the marginating leukocytes and BMECs in situ during EAE. CONCLUSIONS: During neuroinflammation, CLN-5+ leukocytes appear in the CNS, and both CLN-5+ leukocytes and CLN-5+ EVs are detected in the blood. As endothelial cells transfer CLN-5+ to leukocytes in vivo, and EVs released from BMEC bind to leukocytes in vitro, EVs may serve as the vehicles to transfer CLN-5 protein at sites of leukocyte:endothelial contact along the BBB. This action may be a prelude to facilitate TEM through the formation of temporary TJ protein bridges between these two cell types.


Subject(s)
Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelial Cells/pathology , Endothelium, Vascular/metabolism , Extracellular Vesicles/metabolism , Membrane Glycoproteins/metabolism , Animals , Cells, Cultured , Central Nervous System/diagnostic imaging , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelial Cells/ultrastructure , Endothelium, Vascular/ultrastructure , Extracellular Vesicles/ultrastructure , Female , Leukocytes/metabolism , Lysosomal Membrane Proteins , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/immunology , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/immunology , Peptide Fragments/toxicity
7.
Plant Biotechnol J ; 14(7): 1604-18, 2016 07.
Article in English | MEDLINE | ID: mdl-26806316

ABSTRACT

MtPAR is a proanthocyanidin (PA) biosynthesis regulator; the mechanism underlying its promotion of PA biosynthesis is not fully understood. Here, we showed that MtPAR promotes PA production by a direct repression of biosynthesis of isoflavones, the major flavonoids in legume, and by redirecting immediate precursors, such as anthocyanidins, flux into PA pathway. Ectopic expression of MtPAR repressed isoflavonoid production by directly binding and suppressing isoflavone biosynthetic genes such as isoflavone synthase (IFS). Meanwhile, MtPAR up-regulated PA-specific genes and decreased the anthocyanin levels without altering the expression of anthocyanin biosynthetic genes. MtPAR may shift the anthocyanidin precursor flux from anthocyanin pathway to PA biosynthesis. MtPAR complemented PA-deficient phenotype of Arabidopsis tt2 mutant seeds, demonstrating their similar action on PA production. We showed the direct interactions between MtPAR, MtTT8 and MtWD40-1 proteins from Medicago truncatula and Glycine max, to form a ternary complex to trans-activate PA-specific ANR gene. Finally, MtPAR expression in alfalfa (Medicago sativa) hairy roots and whole plants only promoted the production of small amount of PAs, which was significantly enhanced by co-expression of MtPAR and MtLAP1. Transcriptomic and metabolite profiling showed an additive effect between MtPAR and MtLAP1 on the production of PAs, supporting that efficient PA production requires more anthocyanidin precursors. This study provides new insights into the role and mechanism of MtPAR in partitioning precursors from isoflavone and anthocyanin pathways into PA pathways for a specific promotion of PA production. Based on this, a strategy by combining MtPAR and MtLAP1 co-expression to effectively improve metabolic engineering performance of PA production in legume forage was developed.


Subject(s)
Isoflavones/metabolism , Medicago truncatula/metabolism , Metabolic Engineering/methods , Plant Proteins/genetics , Proanthocyanidins/biosynthesis , Anthocyanins/metabolism , Gene Expression Regulation, Plant , Medicago truncatula/genetics , Metabolic Networks and Pathways/genetics , Oxygenases/genetics , Oxygenases/metabolism , Plant Proteins/metabolism , Plant Proteins/physiology , Plants, Genetically Modified/metabolism
8.
Fluids Barriers CNS ; 11(1): 6, 2014 Mar 04.
Article in English | MEDLINE | ID: mdl-24589378

ABSTRACT

BACKGROUND: The chemokine CCL2 is a critical mediator of neuroinflammation in diseases such as multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). CCL2 drives mononuclear cell infiltration into the central nervous system (CNS), alters expression and distribution of microvascular endothelial tight junction proteins, and disrupts the blood-brain and blood-spinal cord barriers. Immunohistochemistry has consistently revealed astrocytes to be a source of this chemokine during neuroinflammation, while providing less uniform evidence that CNS endothelial cells may also express CCL2. Moreover, the relative contributions of these cell types to the CNS pool of CCL2 during MS/EAE are unclear and the aim of this study was to investigate this further. METHODS: CCL2 gene expression was determined by qRT-PCR in different populations of CNS cells at different times following EAE induced by immunization with MOG35-55 peptide and adjuvants, or after injection with adjuvants alone. CNS cells types were isolated by two different protocols: bulk isolation to yield crude microvascular and parenchymal fractions (containing astrocytes, other glia, and neurons), or laser capture microdissection (LCM) to acquire more precisely microvascular endothelial cells, astrocytes or other parenchymal cells. RESULTS: Both CNS microvessel and parenchymal populations prepared by crude bulk isolation showed up-regulation of CCL2 mRNA following MOG immunization or injection of adjuvants alone. More exact dissection by LCM revealed microvascular endothelial cells and astrocytes to be the specific sources of CCL2 gene induction following MOG immunization, while only astrocytes showed elevated CCL2 mRNA in response to just adjuvants. Astrocytes displayed the greatest degree of stimulation of CCL2 gene expression following EAE induction. CONCLUSIONS: High-precision LCM affirmed both microvascular endothelial cells and astrocytes as the major CNS sources of CCL2 gene expression during EAE. Given the high accessibility of the CNS microvascular endothelium, endothelial-derived CCL2 could prove a viable target for therapeutic intervention in neuroinflammatory disease.

9.
J Neuroinflammation ; 11: 10, 2014 Jan 21.
Article in English | MEDLINE | ID: mdl-24444311

ABSTRACT

BACKGROUND: Expression of chemokine CCL2 in the normal central nervous system (CNS) is nearly undetectable, but is significantly upregulated and drives neuroinflammation during experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis which is considered a contributing factor in the human disease. As astrocytes and brain microvascular endothelial cells (BMEC) forming the blood-brain barrier (BBB) are sources of CCL2 in EAE and other neuroinflammatory conditions, it is unclear if one or both CCL2 pools are critical to disease and by what mechanism(s). METHODS: Mice with selective CCL2 gene knockout (KO) in astrocytes (Astro KO) or endothelial cells (Endo KO) were used to evaluate the respective contributions of these sources to neuroinflammation, i.e., clinical disease progression, BBB damage, and parenchymal leukocyte invasion in a myelin oligodendrocyte glycoprotein peptide (MOG35-55)-induced EAE model. High-resolution 3-dimensional (3D) immunofluorescence confocal microscopy and colloidal gold immuno-electron microscopy were employed to confirm sites of CCL2 expression, and 3D immunofluorescence confocal microscopy utilized to assess inflammatory responses along the CNS microvasculature. RESULTS: Cell-selective loss of CCL2 immunoreactivity was demonstrated in the respective KO mice. Compared to wild-type (WT) mice, Astro KO mice showed reduced EAE severity but similar onset, while Endo KO mice displayed near normal severity but significantly delayed onset. Neither of the KO mice showed deficits in T cell proliferation, or IL-17 and IFN-γ production, following MOG35-55 exposure in vitro, or altered MOG-major histocompatibility complex class II tetramer binding. 3D confocal imaging further revealed distinct actions of the two CCL2 pools in the CNS. Astro KOs lacked the CNS leukocyte penetration and disrupted immunostaining of CLN-5 at the BBB seen during early EAE in WT mice, while Endo KOs uniquely displayed leukocytes stalled in the microvascular lumen. CONCLUSIONS: These results point to astrocyte and endothelial pools of CCL2 each regulating different stages of neuroinflammation in EAE, and carry implications for drug delivery in neuroinflammatory disease.


Subject(s)
Astrocytes/pathology , Chemokine CCL2/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelium/pathology , Imaging, Three-Dimensional , Microscopy, Confocal , Animals , Central Nervous System/pathology , Chemokine CCL2/deficiency , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Mice , Mice, Knockout , Microvessels/pathology , Myelin-Oligodendrocyte Glycoprotein , Peptide Fragments
10.
Microvasc Res ; 86: 1-10, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23261753

ABSTRACT

Tight junctions (TJs) feature critically in maintaining the integrity of the blood-brain barrier (BBB), and undergo significant disruption during neuroinflammatory diseases. Accordingly, the expression and distribution of CLN-5, a prominent TJ protein in central nervous system (CNS) microvessels and BBB determinant, has been shown to parallel physiological and pathophysiological changes in microvascular function. However, efforts to quantify CLN-5 within the CNS microvasculature in situ, by using conventional two-dimensional immunohistochemical analysis of thin sections, are encumbered by the tortuosity of capillaries and distorted diameters of inflamed venules. Herein, we describe a novel contour-based 3D image visualization and quantification method, employing high-resolution confocal z-stacks from thick immunofluorescently-stained thoraco-lumbar spinal cord cryosections, to analyze CLN-5 along the junctional regions of different-sized CNS microvascular segments. Analysis was performed on spinal cords of both healthy mice, and mice experiencing experimental autoimmune encephalomyelitis (EAE), an animal model of the neuroinflammatory disease multiple sclerosis. Results indicated that, under normal conditions, the density of CLN-5 staining (CLN-5 intensity/ endothelial surface area) was greatest in the capillaries and smaller venules, and least in the larger venules. This heterogeneity in junctional CLN-5 staining was exacerbated during EAE, as spinal venules revealed a significant loss of junctional CLN-5 staining that was associated with focal leukocyte extravasation, while adjacent capillaries exhibited neither CLN-5 loss nor infiltrating leukocytes. However, despite only venules displaying these behaviors, both capillaries and venules evidenced leakage of IgG during disease, further underscoring the heterogeneity of the inflammatory response in CNS microvessels. This method should be readily adaptable to analyzing other junctional proteins of the CNS and peripheral microvasculature, and serve to highlight their role(s) in health and disease.


Subject(s)
Blood-Brain Barrier , Claudin-5/analysis , Encephalomyelitis, Autoimmune, Experimental/pathology , Endothelium, Vascular/chemistry , Imaging, Three-Dimensional/methods , Microscopy, Confocal/methods , Microvessels/chemistry , Spinal Cord/blood supply , Tight Junctions/chemistry , Animals , Capillaries/chemistry , Capillaries/ultrastructure , Capillary Permeability , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/immunology , Endothelium, Vascular/ultrastructure , Female , Immunoglobulin G/metabolism , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence , Multiple Sclerosis , Tight Junctions/ultrastructure , Venules/chemistry , Venules/ultrastructure
11.
Fluids Barriers CNS ; 9(1): 15, 2012 Aug 07.
Article in English | MEDLINE | ID: mdl-22870943

ABSTRACT

BACKGROUND: There is increasing awareness that, aside from producing cerebrospinal fluid, the choroid plexus (CP) might be a key regulator of immune activity in the central nervous system (CNS) during neuroinflammation. Specifically, the CP has recently been posited to control entry of sentinel T cells into the uninflamed CNS during the early stages of neuroinflammatory diseases, like multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). As the CP is compartmentalized into a stromal core containing fenestrated capillaries devoid of typical blood-brain barrier properties, surrounded by a tight junction-expressing choroidal epithelium, each of these compartments might mount unique responses that instigate the neuroinflammatory process. METHODS: To discern responses of the respective CP stromal capillary and choroidal epithelial tissues during evolving neuroinflammation, we investigated morphology and in situ expression of 93 immune-related genes during early stages of EAE induced by immunization with myelin oligodendrocyte glycoprotein peptide (MOG35-55). Specifically, 3-D immunofluorescent imaging was employed to gauge morphological changes, and laser capture microdissection was coupled to an Immune Panel TaqMan Low Density Array to detail alterations in gene expression patterns at these separate CP sites on days 9 and 15 post-immunization (p.i.). To resolve CP effects due to autoimmunity against MOG peptide, from those due to complete Freund's adjuvant (CFA) and pertussis toxin (PTX) included in the immunization, analysis was performed on MOG-CFA/PTX-treated, CFA/PTX-treated, and naïve cohorts. RESULTS: The CP became swollen and displayed significant molecular changes in response to MOG-CFA/PTX immunization. Both stromal capillary and choroidal epithelial tissues mounted vigorous, yet different, changes in expression of numerous genes over the time course analyzed - including those encoding adhesion molecules, cytokines, chemokines, statins, interleukins, T cell activation markers, costimulatory molecules, cyclooxygenase, pro-inflammatory transcription factors and pro-apoptotic markers. Moreover, CFA/PTX-treatment, alone, resulted in extensive, though less robust, alterations in both CP compartments. CONCLUSIONS: MOG-CFA/PTX immunization significantly affects CP morphology and stimulates distinct expression patterns of immune-related genes in CP stromal capillary and epithelial tissues during evolving EAE. CFA/PTX treatment, alone, causes widespread gene alterations that could prime the CP to unlock the CNS to T cell infiltration during neuroinflammatory disease.

12.
J Neuroinflammation ; 9: 171, 2012 Jul 12.
Article in English | MEDLINE | ID: mdl-22788993

ABSTRACT

BACKGROUND: Production of the chemokine CCL2 by cells of the neurovascular unit (NVU) drives critical aspects of neuroinflammation. Suppression of CCL2 therefore holds promise in treating neuroinflammatory disease. Accordingly, we sought to determine if the compound bindarit, which inhibits CCL2 synthesis, could repress the three NVU sources of CCL2 most commonly reported in neuroinflammation--astrocytes, microglia and brain microvascular endothelial cells (BMEC)--as well as modify the clinical course of neuroinflammatory disease. METHODS: The effect of bindarit on CCL2 expression by cultured murine astrocytes, microglia and BMEC was examined by quantitative reverse transcription polymerase chain reaction (qRT-PCR). Bindarit action on mouse brain and spinal cord in vivo was similarly investigated by qRT-PCR following LPS injection in mice. And to further gauge the potential remedial effects of bindarit on neuroinflammatory disease, its impact on the clinical course of experimental autoimmune encephalomyelitis (EAE) in mice was also explored. RESULTS: Bindarit repressed CCL2 expression by all three cultured cells, and antagonized upregulated expression of CCL2 in both brain and spinal cord in vivo following LPS administration. Bindarit also significantly modified the course and severity of clinical EAE, diminished the incidence and onset of disease, and evidenced signs of disease reversal. CONCLUSION: Bindarit was effective in suppressing CCL2 expression by cultured NVU cells as well as brain and spinal cord tissue in vivo. It further modulated the course of clinical EAE in both preventative and therapeutic ways. Collectively, these results suggest that bindarit might prove an effective treatment for neuroinflammatory disease.


Subject(s)
Chemokine CCL2/antagonists & inhibitors , Chemokine CCL2/biosynthesis , Drug Delivery Systems/methods , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Indazoles/administration & dosage , Propionates/administration & dosage , Amino Acid Sequence , Animals , Cells, Cultured , Female , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Spinal Cord/drug effects , Spinal Cord/metabolism
13.
Proc Natl Acad Sci U S A ; 109(5): 1766-71, 2012 Jan 31.
Article in English | MEDLINE | ID: mdl-22307644

ABSTRACT

MtPAR (Medicago truncatula proanthocyanidin regulator) is an MYB family transcription factor that functions as a key regulator of proanthocyanidin (PA) biosynthesis in the model legume Medicago truncatula. MtPAR expression is confined to the seed coat, the site of PA accumulation. Loss-of-function par mutants contained substantially less PA in the seed coat than the wild type, whereas levels of anthocyanin and other specialized metabolites were normal in the mutants. In contrast, massive accumulation of PAs occurred when MtPAR was expressed ectopically in transformed hairy roots of Medicago. Transcriptome analysis of par mutants and MtPAR-expressing hairy roots, coupled with yeast one-hybrid analysis, revealed that MtPAR positively regulates genes encoding enzymes of the flavonoid-PA pathway via a probable activation of WD40-1. Expression of MtPAR in the forage legume alfalfa (Medicago sativa) resulted in detectable levels of PA in shoots, highlighting the potential of this gene for biotechnological strategies to increase PAs in forage legumes for reduction of pasture bloat in ruminant animals.


Subject(s)
Medicago truncatula/metabolism , Plant Proteins/physiology , Proanthocyanidins/biosynthesis , Transcription Factors/physiology , Gene Expression Regulation, Plant , Genes, Plant , Medicago truncatula/genetics , Molecular Sequence Data , Mutation , Plant Proteins/genetics , Transcription Factors/genetics
14.
Methods Mol Biol ; 755: 385-94, 2011.
Article in English | MEDLINE | ID: mdl-21761321

ABSTRACT

Laser-capture microdissection (LCM) allows for retrieval of distinct populations of cells from their closely surrounding neighbors in situ. As such, LCM is highly advantageous for investigating gene expression along the central nervous system (CNS) microvascular endothelium, a tissue that shows both -considerable segmental and regional heterogeneity. Combining immunohistochemical staining of CNS microvascular endothelial cells with immunofluorescent staining of perivascular astrocytes or smooth muscle cells, immune-guided LCM, immuno-LCM, may be coupled to downstream qRT-PCR to probe varied expression of the endothelium along the CNS microvascular tree during health and disease. Immuno-LCM/qRT-PCR has been used to highlight contributions of the respective segments of the CNS microvasculature to the blood-brain barrier (BBB), and can be employed to examine changes in BBB gene expression -during pathology.


Subject(s)
Brain/blood supply , Endothelium, Vascular/cytology , Lasers , Microdissection/methods , Microvessels/cytology , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Cryopreservation/methods , Endothelium, Vascular/metabolism , Fluorescent Antibody Technique/methods , Gene Expression Profiling/methods , Mice , Microvessels/metabolism , RNA/genetics , RNA/isolation & purification , Tissue Fixation/methods
15.
Planta ; 233(4): 843-55, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21221632

ABSTRACT

Roots of kudzu (Pueraria lobata) are a rich source of isoflavone O- and C-glycosides. Although O-glycosylation of (iso)flavonoids has been well characterized at the molecular level, no plant isoflavonoid C-glycosyltransferase genes have yet been isolated. To address the biosynthesis of kudzu isoflavonoids, we generated 6,365 high-quality expressed sequence tags (ESTs) from a subtraction cDNA library constructed using RNA from roots that differentially accumulate puerarin. The ESTs were clustered into 722 TCs and 3,913 singletons, from which 15 family I glycosyltransferases (UGTs) were identified. Hierarchical clustering analysis of the expression patterns of these UGTs with isoflavone synthase (IFS) in a range of tissues identified UGTs with potential functions in isoflavone glycosylation. The open reading frames of these UGTs were expressed in E. coli for functional analysis, and one was shown to preferentially glycosylate isoflavones at the 7-O-position. In addition, ESTs corresponding to chalcone synthase, chalcone reductase, chalcone isomerase (CHI) and 2-hydroxyisoflavanone dehydratase were identified. Recombinant CHI proteins had high activities with both 6'-deoxy- and 6'-hydroxy chalcones, typical of Type II CHIs. Establishment of this EST database and identification of genes associated with kudzu isoflavone biosynthesis and glycosylation provide a new resource for metabolic engineering of bioactive kudzu isoflavones.


Subject(s)
Genomics/methods , Isoflavones/biosynthesis , Pueraria/genetics , Pueraria/metabolism , Biosynthetic Pathways , Chromatography, High Pressure Liquid , Cluster Analysis , Expressed Sequence Tags , Gene Expression Regulation, Plant , Gene Library , Glycosyltransferases/genetics , Glycosyltransferases/metabolism , Isoflavones/chemistry , Molecular Sequence Data , Plant Roots/metabolism , Plant Stems/metabolism , Pueraria/enzymology , Substrate Specificity
16.
J Mol Neurosci ; 39(1-2): 269-83, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19340610

ABSTRACT

While the expression of the C-C chemokine ligand 2 (CCL2) in the central nervous system (CNS) is associated with numerous neuroinflammatory conditions, the critical cellular sources of this chemokine, which is responsible for disease processes-as well as associated pathogenic mechanisms, remain unresolved. As the potential for anti-CCL2 therapeutics in treating neuroinflammatory disease is likely to be contingent upon effective drug delivery to the source(s) and/or target(s) of CCL2 action in the CNS, tools to highlight the course of CCL2 action during neuroinflammation are imperative. In response to this need, we used the Cre/loxP and FLP-FRT recombination system to develop the first two, cell-conditional CCL2 knockout mice-separately targeting CCL2 gene elimination to astrocytes and endothelial cells, both of which have been considered to play crucial though undefined roles in neuroinflammatory disease. Specifically, mice containing a floxed CCL2 allele were intercrossed with GFAP-Cre or Tie2-Cre transgenic mice to generate mice with CCL2-deficient astrocytes (astrocyte KO) or endothelial cells (endothelial KO), respectively. Polymerase chain reaction, reverse transcription polymerase chain reaction/quantitative reverse transcriptase polymerase chain reaction, and enzyme-linked immunosorbent assay of CCL2 gene, RNA, and protein, respectively, from cultured astrocytes and brain microvascular endothelial cells (BMEC) established the efficiency and specificity of the CCL2 gene deletions and a CCL2 null phenotype in these CNS cells. Effective cell-conditional knockout of CCL2 was also confirmed in an in vivo setting, wherein astrocytes and BMEC were retrieved by immune-guided laser capture microdissection from their in situ positions in the brains of mice experiencing acute, lipopolysaccharide-mediated endotoxemia to induce CCL2 gene expression. In vivo analysis further revealed apparent cross-talk between BMEC and astrocytes regarding the regulation of astrocyte CCL2 expression. Use of astrocyte KO and endothelial KO mice should prove critical in elaborating the pathogenic mechanisms of and optimizing the treatments for neuroinflammatory disease.


Subject(s)
Astrocytes/metabolism , Central Nervous System , Chemokine CCL2 , Endothelial Cells/metabolism , Inflammation/physiopathology , Animals , Astrocytes/cytology , Central Nervous System/metabolism , Central Nervous System/physiopathology , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , E-Selectin/genetics , E-Selectin/metabolism , Endothelial Cells/cytology , Genotype , Mice , Mice, Knockout , Mice, Transgenic , P-Selectin/genetics , P-Selectin/metabolism
17.
J Neurochem ; 104(5): 1219-32, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18289346

ABSTRACT

The means by which the chemokine CCL2 produced in the brain parenchyma can recruit leukocytes lying behind the highly impervious endothelium of the blood-brain barrier (BBB) has remained a paradox. As other chemokines have been evidenced to stimulate their own synthesis and release by peripheral microvascular endothelial cells, and/or undergo transcytosis in the abluminal-to-luminal direction, we determined whether CCL2 experiences similar fates across brain microvascular endothelial cells (BMEC). Using cultured BMEC as a paradigm of the BBB, it was observed that exogenous unlabeled CCL2 actually depressed the release of endogenous CCL2, and further caused diminished CCL2 mRNA levels in these cells. On the other hand, exogenous (125)I-labeled CCL2 exhibited transport across BMEC in a manner that was sensitive to temperature, competition by excess unlabeled CCL2 but not unlabeled CCL3, knockdown of caveolin-1/caveolae, and elimination of the cognate CCL2 receptor CCR2. These results implied a facet of CCL2 transport by a transcellular mechanism partly involving binding of CCL2 to CCR2, and subsequent transfer to caveolae vesicles for transcytosis. This notion was supported by double-label immuno-electronmicroscopy, which revealed co-localization of caveolin-1 with exogenous CCL2, during this chemokine's transit across BMEC. Collectively, these findings provide a rationale by which CCL2, deposited on the abluminal side of the brain microvasculature during inflammatory episodes, can be relayed across the BBB to foster leukocyte recruitment.


Subject(s)
Brain/blood supply , Brain/metabolism , Capillary Permeability/physiology , Cell Membrane/metabolism , Chemokine CCL2/metabolism , Endothelium, Vascular/metabolism , Animals , Biological Transport, Active/physiology , Blood-Brain Barrier/metabolism , Brain/cytology , Brain/drug effects , Cells, Cultured , Chemokine CCL2/pharmacology , Chemotaxis, Leukocyte/physiology , Endothelium, Vascular/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microcirculation/cytology , Microcirculation/metabolism , Microcirculation/physiology
18.
Blood ; 109(4): 1515-23, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17023578

ABSTRACT

Recent evidence from this laboratory indicated that reduced expression of caveolin-1 accompanied the diminished expression of tight junction (TJ)-associated proteins occludin and zonula occludens-1 (ZO-1) following stimulation of brain microvascular endothelial cells (BMECs) with the chemokine CCL2 (formerly called MCP-1). Because attenuated caveolin-1 levels have also been correlated with heightened permeability of other endothelia, the objective of this study was to test the hypothesis that reduced caveolin-1 expression is causally linked to the action of CCL2 on BMEC junctional protein expression and barrier integrity. This was achieved using adenovirus to nondestructively deliver caveolin-1 siRNA (Ad-siCav-1) to BMEC monolayers, which model the blood-brain barrier (BBB). Treatment with siRNA reduced the caveolin-1 protein level as well as occludin and ZO-1. Additionally, occludin exhibited dissociation from the cytoskeletal framework. These changes were attended by comparable alterations in adherens junction (AJ)-associated proteins, VE-cadherin and beta-catenin, increased BMEC paracellular permeability, and facilitated the ability of CCL2 to stimulate monocytic transendothelial migration. Furthermore, treating BMECs with cavtratin, a synthetic cell-permeable peptide encoding the caveolin-1 scaffolding domain, antagonized effects of both Ad-siCav-1 and CCL2. These results collectively highlight caveolin-1 loss as a critical step in CCL2-induced modulation of BMEC junctional protein expression and integrity, and possibly serve a crucial role in regulating inflammation at the BBB.


Subject(s)
Brain/blood supply , Caveolin 1/physiology , Connexins/genetics , Endothelium, Vascular/cytology , Gene Expression Regulation , Animals , Blood-Brain Barrier , Capillaries/cytology , Capillary Permeability , Chemokine CCL2 , Endothelial Cells , Mice
19.
J Neuroimmunol ; 177(1-2): 209-14, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16806499

ABSTRACT

The isolation and culture of spinal cord microvascular endothelial cells (SCMEC), which form the blood-spinal cord barrier (BSCB), is described. Though morphologically similar to brain microvascular endothelial cells (BMEC) that form the blood-brain barrier (BBB), SCMEC express reduced amounts of several prominent BBB proteins, including tight junction-associated proteins ZO-1 and occludin, adherens junction-associated proteins beta-catenin and VE-cadherin, and the efflux transporter P-glycoprotein. These distinguishing features may reflect more widespread differences between the BBB and BSCB that impact physiological and pathophysiological processes.


Subject(s)
Cell Separation/methods , Endothelial Cells/cytology , Microcirculation/cytology , Spinal Cord/blood supply , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adherens Junctions/metabolism , Animals , Antigens, CD/metabolism , Blood-Brain Barrier/cytology , Blood-Brain Barrier/metabolism , Cadherins/metabolism , Cell Culture Techniques/methods , Cells, Cultured , Endothelial Cells/physiology , Membrane Proteins/metabolism , Mice , Microcirculation/physiology , Occludin , Phosphoproteins/metabolism , Spinal Cord/cytology , Tight Junctions/metabolism , Zonula Occludens-1 Protein , beta Catenin/metabolism
20.
Microvasc Res ; 70(1-2): 53-64, 2005 Jul.
Article in English | MEDLINE | ID: mdl-15927208

ABSTRACT

While the expression of chemokine receptors by endothelial cells is now well established, little is known of the function of these receptors at this cellular locale. However, given that chemokines are instrumental in directing leukocytes to specific parenchymal sites, one possibility is that endothelial chemokine receptors play a role in the process of leukocyte extravasation. To test this hypothesis, we investigated the contribution of CCR2, the major cognate receptor for the chemokine CCL2 (formerly known as MCP-1), to CCL2-stimulated transendothelial migration of macrophages (mØ) across cultured brain microvascular endothelial cells (BMEC). Specifically, we prepared both BMEC and mØ from wild-type (WT) mice and mice deficient in CCR2; i.e., CCR2 (-/-), and compared the ability of WT and CCR2 (-/-) BMEC to support CCL2-stimulated transendothelial migration of WT and CCR2 (-/-) mØ. In response to CCL2, WT mØ, but not CCR2 (-/-) mØ, were stimulated to migrate across WT BMEC, consistent with the recognized obligatory role for CCR2 in mediating CCL2-stimulated responses. Remarkably, however, neither WT nor CCR2 (-/-) mØ were stimulated by CCL2 to migrate across CCR2 (-/-) BMEC. In contrast, both types of mØ were able to migrate similarly across both types of BMEC in response to another chemokine--CCL3 (formerly known as MIP-1alpha)--which utilizes receptors other than CCR2. Lastly, CCL2-induced mØ transendothelial migration was blocked by treatment of WT BMEC with pertussis toxin, suggesting that CCR2 is functionally coupled to the inhibitory G protein Galphai, much as it is in other cell types. These results highlight a heretofore-unrecognized role for endothelial CCR2 in mediating transendothelial migration.


Subject(s)
Brain/blood supply , Chemokine CCL2/pharmacology , Chemotaxis, Leukocyte/drug effects , Endothelium, Vascular/metabolism , Macrophages, Peritoneal/drug effects , Receptors, Chemokine/metabolism , Animals , Cells, Cultured , Chemokine CCL3 , Chemokine CCL4 , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Macrophage Inflammatory Proteins/pharmacology , Macrophages, Peritoneal/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Pertussis Toxin/pharmacology , Receptors, Chemokine/deficiency , Receptors, Chemokine/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...