Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(19)2022 Oct 08.
Article in English | MEDLINE | ID: mdl-36230844

ABSTRACT

The current standard for investigating tumors is surgical biopsy, which is costly, invasive, and difficult to perform serially. As an adjunct, circulating tumor cells (CTCs)-cells that have broken away from the primary tumor or metastatic sites-can be obtained from a blood draw and offer the potential for obtaining serial genetic information and serving as biomarkers. Here, we detail the potential for melanoma CTCs to serve as biomarkers and discuss a clinically viable methodology for single-cell CTC isolation and analysis that overcomes previous limitations. We explore the use of melanoma CTC biomarkers by isolating and performing single-cell RNA sequencing on CTCs from melanoma patients. We then compared transcriptional profiles of single melanoma CTCs against A375 cells and peripheral blood mononuclear cells to identify unique genes differentially regulated in circulating melanoma tumor cells. The information that can be obtained via analysis of these CTCs has significant potential in disease tracking.

2.
Cell Immunol ; 378: 104561, 2022 08.
Article in English | MEDLINE | ID: mdl-35738135

ABSTRACT

Our previous studies demonstrated increased serum levels of macrophage migration inhibitory factor (MIF-1) and its homologue, MIF-2, in males during MS progression; and that genetically high-MIF-expressing male subjects with relapsing multiple sclerosis (MS) had a significantly greater risk of conversion to progressive MS than lower-MIF-expressing males and females. However, female MS subjects with severe disease expressed higher levels of CD74, the common MIF-1/MIF-2 receptor, on blood cells. In the murine model of MS, experimental autoimmune encephalomyelitis (EAE), both male and female mice lacking MIF-1 and/or MIF-2 were clinically improved during development of moderately severe disease, thus implicating both homologs as co-pathogenic contributors. The current study using MIF-deficient mice with severe acute EAE revealed a highly significant reduction of EAE scores in MIF-1-deficient females, in contrast to only minor and delayed reduction of clinical signs in MIF-1-deficient males. However, clinical EAE scores and factor expression were strongly suppressed in males and further reduced in females after treatment of WT and MIF-1-, MIF-2- and MIF-1/2-DUAL-deficient female and male mice with a MHCII DRα1-MOG-35-55 molecular construct that competitively inhibits MIF-1 & MIF-2 signaling through CD74 as well as T cell activation. These results suggest sex-dependent differences in which the absence of the MIF-1 and/or MIF-2 genotypes may permit stronger compensatory CD74-dependent EAE-inducing responses in males than in females. However, EAE severity in both sexes could still be reduced nearly to background (a "near cure") with DRα1-MOG-35-55 blockade of compensatory MIF and CD74-dependent factors known to attract peripheral inflammatory cells into the spinal cord tissue.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , MSH Release-Inhibiting Hormone , Macrophage Migration-Inhibitory Factors , Multiple Sclerosis , Animals , Female , Histocompatibility Antigens Class II/immunology , Humans , Intramolecular Oxidoreductases/genetics , Intramolecular Oxidoreductases/metabolism , MSH Release-Inhibiting Hormone/metabolism , MSH Release-Inhibiting Hormone/therapeutic use , Macrophage Migration-Inhibitory Factors/metabolism , Male , Mice , Mice, Inbred C57BL , Spinal Cord
3.
Cell Immunol ; 370: 104439, 2021 12.
Article in English | MEDLINE | ID: mdl-34607646

ABSTRACT

Macrophage migration inhibitory factor (MIF-1) and its homologue d-dopachrome tautomerase (MIF-2) share the common CD74 receptor and function innately to enhance severity of multiple sclerosis (MS) as well as the experimental autoimmune encephalomyelitis (EAE) model for MS. We previously demonstrated that genetically high-MIF-expressing male subjects with relapsing MS had a significantly greater risk of conversion to progressive MS (PMS) than lower-MIF-expressing males. To expand on this observation, we utilized MIF-1, MIF-2, and MIF-1/2-DUAL-deficient male mice to discern if there would be a greater contribution of these inflammatory factors in EAE mice with severe vs. moderate clinical disease signs. As shown previously, mice deficient in either MIF-1 or MIF-2 each had a ∼25% reduction of moderate EAE compared to WT mice, with significant differences in disease onset and trajectory. However, EAE induction in mice deficient in both MIF-1 and MIF-2 genes did not result in a further reduction in EAE severity. This result suggests that the two MIF homologues were likely affecting the same pathogenic pathways such that each could partially compensate for the other but not in an additive or synergistic manner. However, MIF-1-KO, MIF-2-KO, and MIF-1/2-DUAL-KO mice with severe EAE did not exhibit a significant reduction in cumulative EAE scores compared with WT mice, but the MIF-1-KO and, to a lesser extent, MIF-1/2-DUAL-KO mice did show a significant reduction in daily EAE scores over the last 3 days of observation, and MIF-2-KO mice showed a more modest but still consistent reduction over the same span. Furthermore, deletion of MIF-1 resulted in a massive reduction in the expression of EAE- and Complete Freund's Adjuvant-associated inflammatory factors, suggesting delayed involvement of the MIF/CD74 axis in promoting disease expression. To further explore modulation of MIF-1 and MIF-2 effects on EAE, we treated WT mice with moderate EAE using DRα1-mMOG-35-55, an inhibitor of CD74 that blocks both MIF-1 and MIF-2 action. This treatment reduced ongoing moderate EAE severity in excess of 25%, suggesting efficient blockade of the MIF/CD74 axis in disease-enhancing pathways. Moreover, DRα1-mMOG-35-55 treatment of mice with severe EAE strongly reversed EAE- and CFA-associated expression of inflammatory cytokines and chemokines including Tnf, Ccr7, Ccr6, Ccl8, Cxcr3, and Ccl19 in MIF-deficient mouse genotypes, and also exceeded innate MIF-1 and MIF-2 EAE enhancing effects, especially in MIF-1-KO mice. These results illustrate the therapeutic potential of targeting the disease-enhancing MIF/CD74 pathway in male mice with moderate and severe EAE, with implications for treatment of high-MIF-expressing RRMS human males at risk of conversion to progressive MS as well as those that have already transitioned to PMS.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/immunology , Neuroprotective Agents/pharmacology , Recombinant Fusion Proteins/pharmacology , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
4.
Cell Immunol ; 359: 104242, 2021 01.
Article in English | MEDLINE | ID: mdl-33190849

ABSTRACT

Experimental autoimmune encephalomyelitis (EAE) is commonly used as an animal model for evaluating clinical, histological and immunological processes potentially relevant to the human disease multiple sclerosis (MS), for which the mode of disease induction remains largely unknown. An important caveat for interpreting EAE processes in mice is the inflammatory effect of immunization with myelin peptides emulsified in Complete Freund's Adjuvant (CFA), often followed by additional injections of pertussis toxin (Ptx) in some strains to induce EAE. The current study evaluated clinical, histological, cellular (spleen), and chemokine-driven processes in spinal cords of male vs. female C57BL/6 mice that were immunized with mouse (m)MOG-35-55/CFA/Ptx to induce EAE; immunized with saline/CFA/Ptx only (CFA, no EAE); or were untreated (Naïve, no EAE). Analysis of response curves utilized a rigorous and sophisticated methodology to parse and characterize the effects of EAE and adjuvant alone vs. the Naive baseline responses. The results demonstrated stronger pro-inflammatory responses of immune cells and their associated cytokines, chemokines, and receptors in male vs. female CFA and EAE mice that appeared to be offset partially by increased percentages of male anti-inflammatory, regulatory and checkpoint T cell, B cell, and monocyte/macrophage subsets. These sex differences in peripheral immune responses may explain the reduced cellular infiltration and differing chemokine profiles in the Central Nervous System (CNS) of male vs. female CFA immunized mice and the reduced CNS infiltration and demyelination observed in male vs. female EAE groups of mice that ultimately resulted in the same clinical EAE disease severity in both sexes. Our findings suggest EAE disease severity is governed not only by the degree of CNS infiltration and demyelination, but also by the balance of pro-inflammatory vs. regulatory cell types and their secreted cytokines and chemokines.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Freund's Adjuvant/pharmacology , Adjuvants, Immunologic/pharmacology , Adoptive Transfer , Animals , B-Lymphocytes/immunology , Central Nervous System/immunology , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Immunization/methods , Male , Mice , Mice, Inbred C57BL , Multiple Sclerosis/metabolism , Peptide Fragments/immunology , Pertussis Toxin/pharmacology , Sex Characteristics , Sex Factors , Spinal Cord/immunology , T-Lymphocytes/immunology
5.
J Neuroinflammation ; 16(1): 195, 2019 Oct 29.
Article in English | MEDLINE | ID: mdl-31665042

ABSTRACT

BACKGROUND: IL-10 knockout (KO) mice are protected from experimental autoimmune encephalomyelitis (EAE) with low-dose estrogen (E2) treatment similar to wild-type (WT) mice. Previous studies have demonstrated a decrease in tumor necrosis factor in all E2-treated groups, which led to the protection of the mice. METHODS: This study used IL-10 KO mice and WT mice treated either with E2 or sham pellets 7 days prior to induction of EAE. Mice were observed for 21 days post-immunization. The spleen, inguinal lymph nodes, and brain were evaluated by flow cytometry. Spinal cords were evaluated using a cytokine/chemokine array, RT-PCR, and histology. RESULTS: This study demonstrates that E2 treatment induced three heightened regulatory mechanisms that potentially protect IL-10 KO mice from EAE: (1) an increase in programmed death-ligands 1 and 2 on monocytes and macrophages in the periphery and within the CNS; (2) an increase in CD73 in the inflamed CNS, which can increase the production of the anti-inflammatory molecule adenosine; and (3) a decrease in CD4+CD25+FoxP3+ regulatory T cells in the spleen. Together, these factors comprise an alternative compensatory mechanism that significantly downregulates key pro-inflammatory cytokine, chemokine, and chemokine receptor genes which are enhanced in the spinal cord of IL-10 KO mice. This group of E2-treated mice remained asymptomatic after EAE challenge similar to E2-treated WT mice, despite their having more T and B lymphocytes in the brain, and modestly increased demyelination in the spinal cord. CONCLUSION: These results indicate that previously unrecognized compensatory mechanisms of EAE protection are stimulated by E2 in the absence of IL-10, which can provide disease protection comparable to the IL-10-dependent effects induced by E2 in WT mice.


Subject(s)
Drug Implants/administration & dosage , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Estrogens/administration & dosage , Interleukin-10/deficiency , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout
6.
Metab Brain Dis ; 34(6): 1705-1712, 2019 12.
Article in English | MEDLINE | ID: mdl-31410774

ABSTRACT

Inflammation involving migration of immune cells across the damaged blood-brain barrier (BBB), activation of resident innate microglia and production of inflammatory humoral mediators such as cytokines and chemokines play a critical role in the pathogenesis of ischemic stroke. Cell-cell signaling involved in the process also includes checkpoint interaction between programmed death receptor (PD1) and programmed death ligands, PDL1 and PDL2. Based on our previous studies showing reduced MCAO infarct volumes in PDL2 deficient mice, we evaluated the ability of anti-PDL2 mAb to treat MCAO in male and female C57BL/6 mice. We found that anti-PDL2 neutralizing antibody treatment of MCAO significantly reduced infarct volumes in male mice but had no protective effects in female mice even at a 5-fold increased dose of anti-PDL2 mAb. The protection in male mice was likely mediated by reduced percentages in the spleen of PDL2+CD19+ B cells, PDL1+CD4+ T cells and CD86+CD11b+ macrophages in concert with reduced expression of PDL1 and TNFα and continued expression of CD206, in the injured ipsilateral brain hemisphere. The lack of a therapeutic benefit of anti-PDL2 on stroke-induced infarct volumes in female mice was reflected by no detectable reduction in expressed PDL2 or PDL1 and an increased frequency of Th1 and Th17 pro-inflammatory T cell subsets in the spleen, an effect not seen in PDL2 mAb treated males. This result potentially limits the utility of anti-PDL2 mAb therapy in stroke to males but underscores the importance of meeting the STAIR requirements for development of new stroke therapies for both sexes.


Subject(s)
Antibodies, Neutralizing/therapeutic use , Infarction, Middle Cerebral Artery/drug therapy , Programmed Cell Death 1 Ligand 2 Protein/immunology , Stroke/drug therapy , T-Lymphocyte Subsets/immunology , Animals , Disease Models, Animal , Female , Infarction, Middle Cerebral Artery/immunology , Male , Mice , Sex Factors , Stroke/immunology , Treatment Outcome
7.
J Stroke Cerebrovasc Dis ; 27(10): 2746-2754, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30068479

ABSTRACT

BACKGROUND AND PURPOSE: Acid/base and electrolytes could provide clinically valuable information about cerebral infarct core and penumbra. We evaluated associations between acid/base and electrolyte changes and outcomes in 2 rat models of stroke, permanent, and transient middle cerebral artery occlusion. METHODS: Three-month old Sprague-Dawley rats underwent permanent or transient middle cerebral artery occlusion. Pre- and post-middle cerebral artery occlusion venous samples for permanent and transient models provided pH, carbon dioxide, oxygen, glucose, and electrolyte values of ionized calcium, potassium, and sodium. Multiple regression determined predictors of infarct volume from these values, and Kaplan-Meier curve analyzed morality between permanent and transient middle cerebral artery occlusion models. RESULTS: Analysis indicated significant differences in the blood gas and electrolytes between pre- to post-middle cerebral artery occlusion. A decrease in pH and sodium with increases in carbon dioxide, potassium, ionized calcium, and glucose changes were found in both middle cerebral artery occlusion models; while hematocrit and hemoglobin were significant in the transient model. pH and ionized calcium were predictors of infarct volume in the permanent model, as changes in pH and ionized calcium decreased, infarct volume increased. CONCLUSIONS: There are acute changes in acid/base balance and electrolytes during stroke in transient and permanent rodent models. Additionally, we found pH and ionized calcium changes predicted stroke volume in the permanent middle cerebral artery occlusion model. These preliminary findings are novel, and warrant further exploration in human conditions.


Subject(s)
Acid-Base Equilibrium , Infarction, Middle Cerebral Artery/physiopathology , Water-Electrolyte Balance , Animals , Biomarkers/blood , Blood Glucose/metabolism , Calcium/blood , Carbon Dioxide/blood , Disease Models, Animal , Hemoglobins/metabolism , Hydrogen-Ion Concentration , Infarction, Middle Cerebral Artery/blood , Infarction, Middle Cerebral Artery/pathology , Oxygen/blood , Potassium/blood , Rats, Sprague-Dawley , Sodium/blood , Time Factors
8.
J Neuroinflammation ; 15(1): 195, 2018 Jul 03.
Article in English | MEDLINE | ID: mdl-29970193

ABSTRACT

BACKGROUND: Stroke is the fifth leading cause of death and the leading cause of long-term disability in the USA, costing $40.2 billion in direct and indirect costs. Globally, stroke is the second leading cause of death and has a higher prevalence in lower- and middle-income countries compared to high-income countries. The role of the spleen in stroke has been studied in rodent models of stroke and is seen as a major contributor to increased secondary neural injury after stroke. Splenectomy 2 weeks prior to ischemic and hemorrhagic stroke in mice and rats shows decreased infarct volumes. Additionally, the spleen decreases in size following stroke in rodents. Pro-inflammatory mediators are also increased in the spleen and subsequently the brain after stroke. These data in preclinical models of stroke have led stroke neurologists to look at the splenic response in stroke subjects. The outcomes of these studies suggest the spleen is responding in a similar manner in stroke subjects as it is in animal models of stroke. CONCLUSION: Animal models demonstrating the detrimental role of the spleen in stroke are providing strong evidence of how the spleen is responding during stroke in human subjects. This indicates treatments targeting the splenic immune response in animals could provide useful targets and treatments for stroke subjects.


Subject(s)
Spleen/physiology , Stroke/immunology , Stroke/pathology , Animals , Disease Models, Animal , Humans , Mice , Rats , Splenectomy
9.
Metab Brain Dis ; 33(5): 1599-1607, 2018 10.
Article in English | MEDLINE | ID: mdl-29916184

ABSTRACT

A seven day pretreatment course of an oral antibiotic cocktail (Ampicillin, Metronidazole, Neomycin Sulfate, and Vancomycin) was shown to induce changes in peripheral immune regulation and protect mice from signs of experimental autoimmune encephalomyelitis (EAE). To determine if a shorter course of antibiotic pretreatment could also protect the mice from EAE and induce regulatory immune cells, studies were conducted using the same oral antibiotic cocktail for three days. In addition, the CNS was examined to determine the effects of antibiotic pretreatment on EAE disease course and immune modulation within the affected tissue. The shorter three day pretreatment course was also significantly protective against severe EAE in C57BL/6 mice. Moreover, our study found increased frequencies of regulatory cells and a decrease in the frequency of anti-inflammatory macrophages in the spleen of EAE protected mice. Additionally, a chemokine and chemokine receptor array run on mRNA from spinal cords revealed that genes associated with regulatory T cells and macrophage recruitment were strongly upregulated in the antibiotic pretreated mice. Additional RT-PCR data showed genes associated with anti-inflammatory microglia/macrophages were upregulated and pro-inflammatory genes were downregulated. This suggests the macrophages recruited to the spinal cord by chemokines are subsequently polarized toward an anti-inflammatory phenotype. These results lend strong support to the conclusion that a three day course of antibiotic treatment given prior to the induction of severe EAE profoundly protected the mice by inducing regulatory lymphocytes in the periphery and an anti-inflammatory milieu in the affected spinal cord tissue.


Subject(s)
Anti-Bacterial Agents/pharmacology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immunomodulation , Macrophages/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Chemokines/genetics , Down-Regulation/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Lymph Nodes/cytology , Lymph Nodes/drug effects , Macrophages/immunology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Microglia/immunology , Receptors, Chemokine/genetics , Spinal Cord/drug effects , Spinal Cord/physiopathology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Up-Regulation/genetics
10.
Immunology ; 154(2): 169-177, 2018 06.
Article in English | MEDLINE | ID: mdl-29313944

ABSTRACT

Current treatment options for human stroke are limited mainly to the modestly effective infusion of tissue plasminogen activator (tPA), with additional improvement of functional independence and higher rates of angiographic revascularization observed after mechanical thrombectomy. However, new therapeutic strategies that address post-stroke immune-mediated inflammatory responses are urgently needed. Recent studies in experimental stroke have firmly implicated immune mechanisms in the propagation and partial resolution of central nervous system damage after the ischaemic event. A new-found anti-inflammatory role for regulatory B (Breg) cells in autoimmune diseases sparked interest in these cells as potential immunomodulators in stroke. Subsequent studies identified interleukin-10 as a common regulatory cytokine among all five of the currently recognized Breg cell subsets, several of which can be found in the affected brain hemisphere after induction of experimental stroke in mice. Transfer of enriched Breg cell subpopulations into both B-cell-depleted and wild-type mice confirmed their potent immunosuppressive activities in vivo, including recruitment and potentiation of regulatory T cells. Moreover, Breg cell therapy strongly reduced stroke volumes and treatment outcomes in ischaemic mice even when administered 24 hr after induction of experimental stroke, a treatment window far exceeding that of tPA. These striking results suggest that transfer of enriched Breg cell populations could have therapeutic value in human stroke, although considerable clinical challenges remain.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , B-Lymphocytes, Regulatory/immunology , B-Lymphocytes, Regulatory/metabolism , Disease Susceptibility , Stroke/etiology , Stroke/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Cell Communication/immunology , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility/immunology , Humans , Stroke/pathology
11.
J Neuroimmunol ; 310: 51-59, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28778445

ABSTRACT

Sex hormones promote immunoregulatory effects on multiple sclerosis. In the current study we evaluated the composition of the gut microbiota and the mucosal-associated regulatory cells in estrogen or sham treated female mice before and after autoimmune encephalomyelitis (EAE) induction. Treatment with pregnancy levels of estrogen induces changes in the composition and diversity of gut microbiota. Additionally, estrogen prevents EAE-associated changes in the gut microbiota and might promote the enrichment of bacteria that are associated with immune regulation. Our results point to a possible cross-talk between the sex hormones and the gut microbiota, which could promote neuroprotection.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Estrogens/therapeutic use , Intestines/microbiology , Microbiota/drug effects , Mucous Membrane/drug effects , Mucous Membrane/pathology , Animals , Antigens, CD/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/chemically induced , Encephalomyelitis, Autoimmune, Experimental/pathology , Feces/microbiology , Female , Interleukin-10/genetics , Interleukin-10/metabolism , Intestines/drug effects , Leukocytes/drug effects , Lymph Nodes/drug effects , Lymph Nodes/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , RNA, Ribosomal, 16S/genetics , RNA, Ribosomal, 16S/metabolism , Spinal Cord/pathology , Time Factors
12.
Metab Brain Dis ; 32(5): 1747-1754, 2017 10.
Article in English | MEDLINE | ID: mdl-28689297

ABSTRACT

Autoimmune diseases including multiple sclerosis predominantly affect females. Although high levels of sex hormones, particularly estrogen (E2), can reduce proinflammatory immune responses, it remains unclear if a lack of endogenous sex hormones might affect treatment with exogenous sex hormones. Pretreatment with E2 almost completely prevents intact female and male mice from developing clinical and histological signs of experimental autoimmune encephalomyelitis (EAE) by promoting various regulatory immune cell phenotypes. To evaluate the effects of exogenous estrogen in the absence of endogenous sex hormones, the current study compared EAE severity and the emergence of different immunoregulatory cell populations after E2 pretreatment of ovariectomized (OVX) female versus male mice. We found that E2 equally protected both OVX females and males from EAE over a 21 day observation period concomitant with reduced total cell numbers in spleen and spinal cord (males only), but enhanced percentages of CD19+CD5+CD1dhi, CD19+CD138+CD44hi and CD19+Tim-1+ Breg cells, CD8+CD122+ Treg cells and CD11b+CD 206+ARG-1+ anti-inflammatory M2-like monocytes/macrophages in both groups. In contrast, E2 decreased the percentage of CD4+CD25+FoxP3+ Treg cells in OVX females but increased these Treg cells in males and intact female mice. These data suggest that with the exception of CD4+CD25+FoxP3+ Treg cells, E2 protection against EAE promotes highly overlapping immunoregulatory subsets in OVX females and males.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Estradiol/therapeutic use , Animals , Antigens, CD/metabolism , B-Lymphocytes, Regulatory/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Estradiol/metabolism , Female , Macrophages/drug effects , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Ovariectomy , Spinal Cord/pathology , Spleen/pathology , T-Lymphocytes, Regulatory
13.
Cell Immunol ; 318: 49-54, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28606360

ABSTRACT

Stroke is the leading cause of disability in the United States. Sex differences, including smaller infarcts in females and greater involvement of immune-mediated inflammation in males may affect the efficacy of immune-modulating interventions. To address these differences, we sought to identify distinct stroke-modifying mechanisms in female vs. male mice. The current study demonstrated smaller infarcts and increased levels of regulatory CD19+CD5+CD1dhi B10 cells as well as anti-inflammatory CD11b+CD206+ microglia/macrophages in the ipsilateral vs. contralateral hemisphere of female but not male mice undergoing 60min middle cerebral artery occlusion followed by 96h of reperfusion. Moreover, female mice with MCAO had increased total spleen cell numbers but lower B10 levels in spleens. These results elucidate differing sex-dependent regulatory mechanisms that account for diminished stroke severity in females and underscore the need to test immune-modulating therapies for stroke in both males and females.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Brain/immunology , Macrophages/immunology , Sex , Stroke/immunology , Animals , Brain/pathology , Cells, Cultured , Disease Models, Animal , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Cerebral Artery/surgery , Sex Factors
14.
J Neuroimmunol ; 305: 59-67, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28284347

ABSTRACT

Immunoregulatory sex hormones, including estrogen and estriol, may prevent relapses in multiple sclerosis during pregnancy. Our previous studies have demonstrated that regulatory B cells are crucial for estrogen-mediated protection against experimental autoimmune encephalomyelitis (EAE). Herein, we demonstrate an estrogen-dependent induction of alternatively activated (M2) macrophages/microglia that results in an increased frequency of regulatory B cells in the spinal cord of estrogen treated mice with EAE. We further demonstrate that cultured M2-polarized microglia promote the induction of regulatory B cells. Our study suggests that estrogen neuroprotection induces a regulatory feedback loop between M2 macrophages/microglia and regulatory B cells.


Subject(s)
B-Lymphocytes, Regulatory/physiology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Estrogens/therapeutic use , Macrophages/physiology , Microglia/physiology , Spinal Cord/pathology , Animals , Arginase/genetics , Arginase/metabolism , B-Lymphocytes, Regulatory/drug effects , Cells, Cultured , Coculture Techniques , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Female , Gene Expression Regulation/drug effects , Interleukin-10/genetics , Interleukin-10/metabolism , Macrophages/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microglia/drug effects , Myelin-Oligodendrocyte Glycoprotein/toxicity , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Peptide Fragments/toxicity , Spleen/pathology , Time Factors
15.
Mol Neurobiol ; 54(1): 608-622, 2017 01.
Article in English | MEDLINE | ID: mdl-26746670

ABSTRACT

Leukemia inhibitory factor (LIF) has been shown to protect oligodendrocytes from ischemia by upregulating endogenous antioxidants. The goal of this study was to determine whether LIF protects neurons during stroke by upregulating superoxide dismutase 3 (SOD3). Animals were administered phosphate-buffered saline (PBS) or 125 µg/kg LIF at 6, 24, and 48 h after middle cerebral artery occlusion or sham surgery. Neurons were isolated from rat pups on embryonic day 18 and used between 7 and 15 days in culture. Cells were treated with LIF and/or 10 µM Akt inhibitor IV with PBS and 0.1 % DMSO acting as vehicle controls. Neurons transfected with scrambled or SOD3 small interfering RNA (siRNA) were subjected to 24-h ischemia after PBS or LIF treatment. LIF significantly increased superoxide dismutase activity and SOD3 expression in ipsilateral brain tissue compared to PBS. Following 24-h ischemia, LIF reduced cell death and increased SOD3 messenger RNA (mRNA) in vitro compared to PBS. Adding Akt inhibitor IV with LIF counteracted the decrease in cell death. Partially silencing the expression of SOD3 using siRNA prior to LIF treatment counteracted the protective effect of LIF-alone PBS treatment. These results indicate that LIF protects neurons in vivo and in vitro via upregulation of SOD3.


Subject(s)
Cerebral Cortex/enzymology , Leukemia Inhibitory Factor/pharmacology , Neurons/enzymology , Neuroprotective Agents/pharmacology , Superoxide Dismutase/biosynthesis , Up-Regulation/physiology , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Cerebral Cortex/drug effects , Cerebral Cortex/pathology , Dose-Response Relationship, Drug , Male , Neurons/drug effects , Neurons/pathology , Rats , Up-Regulation/drug effects
16.
Acta Neurochir Suppl ; 121: 243-50, 2016.
Article in English | MEDLINE | ID: mdl-26463956

ABSTRACT

Cerebral edema after stroke is associated with poor neurological outcomes. Current therapies are limited to osmotic agents, such as hypertonic saline (HS), which reduce intracranial pressure. Although studies have demonstrated edema reductions following HS, tissue survival has not been thoroughly examined. Additionally, the efficacy of promising pharmacological agents has not been evaluated for synergy with osmotic agents. Conivaptan is an FDA-approved vasopressin receptor antagonist that may exert both osmotic and anti-inflammatory effects. In this study, rats were subjected to middle cerebral artery occlusion prior to treatment with 5 % HS bolus +5 % HS maintenance (HS), conivaptan alone (Con), conivaptan +5 % HS maintenance (Con + HS), or conivaptan +5 % HS bolus +5 % maintenance (Con + HSb). Treatments were initiated at six (Early) or 24 h (Late) following stroke and rats were euthanized at 48 h to evaluate infarct volume, brain edema, and microglia/macrophage activation. Infarct volume and brain edema in the Early HS, Early Con, and Late HS groups were significantly reduced compared with controls. Interestingly, only the Early Con group demonstrated reduced microglia/macrophage activation. These data suggest an anti-inflammatory mechanism for conivaptan and provide support for a multipronged approach combining osmotic agents with compounds that inhibit the neuroinflammatory response to stroke.


Subject(s)
Antidiuretic Hormone Receptor Antagonists/pharmacology , Benzazepines/pharmacology , Brain Edema/metabolism , Brain/drug effects , Infarction, Middle Cerebral Artery/metabolism , Saline Solution, Hypertonic/pharmacology , Animals , Brain/blood supply , Brain/metabolism , Brain/pathology , Brain Edema/pathology , Disease Models, Animal , Fluoresceins , Fluorescent Dyes , Immunohistochemistry , Infarction, Middle Cerebral Artery/pathology , Laser-Doppler Flowmetry , Macrophage Activation/drug effects , Male , Microglia/drug effects , Rats , Rats, Sprague-Dawley
17.
Semin Thromb Hemost ; 41(6): 582-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26270113

ABSTRACT

Patients with inflammatory bowel disease (IBD) exhibit a threefold higher risk for development of systemic thrombosis than the general population. Although the underlying causes of the increased risk for thrombus development remain poorly understood, there is a large body of evidence suggesting that abnormalities in coagulation, fibrinolysis, and platelet function may contribute to this response. Changes in hemostatic biomarkers are consistent with subclinical activation of coagulation system, including tissue factor activation, impaired protein C pathway, enhanced thrombin generation, and diminished fibrinolytic capacity. There is also evidence for an increased production and reactivity of platelets, with an enhanced formation of platelet-platelet and platelet-leukocyte aggregates. The altered coagulation and platelet function, and the predisposition to thrombus formation have also been demonstrated in animal models of IBD. The animal studies have revealed a major role for inflammatory cytokines, including tumor necrosis factor-α, interleukin (IL)-1ß, and IL-6, as mediators of the platelet abnormalities and enhanced thrombus development in experimental IBD. These findings in animal models raise hope for the development of novel therapeutic strategies to reduce thrombosis-related mortality in IBD patients.


Subject(s)
Blood Platelets/pathology , Inflammatory Bowel Diseases/blood , Thrombophilia/blood , Animals , Blood Coagulation Factors/metabolism , Colitis/blood , Colon/blood supply , Cytokines/physiology , Disease Models, Animal , Fibrinolysis , Humans , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/drug therapy , Leukocytes/pathology , Microcirculation , Models, Biological , Nitric Oxide/metabolism , Platelet Activation , Risk , Thrombophilia/etiology , Thrombophilia/prevention & control , Thrombosis/epidemiology , Thrombosis/etiology
18.
Exp Neurol ; 271: 351-7, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26054883

ABSTRACT

OBJECTIVE: The cerebral microvasculature is rendered more vulnerable to thrombus formation following a brief (5.0 min) period of focal ischemia. This study examined the contribution of interleukin-6 (IL-6), a neuroprotective and prothrombotic cytokine produced by the brain, to transient ischemia-induced thrombosis in cerebral arterioles. APPROACH & RESULTS: The middle cerebral artery of C57BL/6J mice was occluded for 5 min, followed by 24h of reperfusion (MCAo/R). Intravital fluorescence microscopy was used to monitor thrombus development in cerebral arterioles induced by light/dye photoactivation. Thrombosis was quantified as the time of onset of platelet aggregation on the vessel wall and the time for complete blood flow cessation. MCAo/R in wild type (WT) mice yielded an acceleration of thrombus formation that was accompanied by increased IL-6 levels in plasma and in post-ischemic brain tissue. The exaggerated thrombosis response to MCAo/R was blunted in WT mice receiving an IL-6 receptor-blocking antibody and in IL-6 deficient (IL-6(-/-)) mice. Bone marrow chimeras, produced by transplanting IL-6(-/-) marrow into WT recipients, did not exhibit protection against MCAo/R-induced thrombosis. CONCLUSIONS: The increased vulnerability of the cerebral vasculature to thrombus development after MCAo/R is mediated by IL-6, which is likely derived from brain cells rather than circulating blood cells. These findings suggest that anti-IL-6 therapy may reduce the likelihood of cerebral thrombus development after a transient ischemic attack.


Subject(s)
Infarction, Middle Cerebral Artery/complications , Interleukin-6/deficiency , Thrombosis/etiology , Thrombosis/metabolism , Animals , Bone Marrow Transplantation , Brain/metabolism , Brain/pathology , Disease Models, Animal , Functional Laterality , Interleukin-6/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Video , Receptors, Interleukin-6/metabolism , Regional Blood Flow , Thrombosis/surgery
19.
J Neuroimmune Pharmacol ; 9(5): 679-89, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25104571

ABSTRACT

The delayed immune response to stroke is responsible for the increased neural injury that continues to occur after the initial ischemic event. This delayed immune response has been linked to the spleen, as splenectomy prior to middle cerebral artery occlusion (MCAO) is neuroprotective. Interferon gamma (IFNγ) is linked to the splenic response, which enhances neural injury following MCAO. IFNγ activates the expression of the inflammatory chemokine interferon-inducible protein 10 (IP-10). This study was designed to determine the role of IFNγ signaling in the inflammatory response following MCAO. Expression of IP-10 increased in the brain and the spleen following MCAO. Splenectomy inhibited the increase of IP-10 in the brain post-MCAO, while recombinant IFNγ administration to splenectomized rats returned IP-10 levels in the brain to levels found in rats after MCAO only. Systemic administration of an IFNγ neutralizing antibody to MCAO-treated rats reduced infarct volume and IP-10 levels in the brain. T cell infiltration was reduced in the MCAO-damaged brains of IFNγ antibody-treated animals relative to those that received isotype control antibodies. Additionally, inhibiting IFNγ signaling with splenectomy or an IFNγ neutralizing antibody blocked the induction of IP-10 expression and decreased neurodegeneration following MCAO. Targeting this pro-inflammatory pathway following stroke could be a promising stroke therapeutic.


Subject(s)
Chemokine CXCL10/biosynthesis , Interferon-gamma/therapeutic use , Neurodegenerative Diseases/metabolism , Signal Transduction/physiology , Stroke/metabolism , Animals , Inflammation Mediators/metabolism , Interferon-gamma/pharmacology , Male , Neurodegenerative Diseases/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Stroke/pathology
20.
Exp Neurol ; 261: 417-23, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25058045

ABSTRACT

OBJECTIVE: While transient ischemic attack (TIA) is a well-known harbinger of ischemic stroke, the mechanisms that link TIA to subsequent strokes remain poorly understood. The overall aim of this study was to determine whether: 1) brief periods of transient cerebral ischemia render this tissue more vulnerable to thrombus development and 2) antiplatelet agents used in TIA patients alter ischemia-induced thrombogenesis. APPROACH & RESULTS: The middle cerebral artery of C57BL/6 mice was occluded for 2.5-10min, followed by reperfusion periods of 1-28days. Intravital microscopy was used to monitor thrombus development in cerebral microvessels induced by light/dye photoactivation. Thrombosis was quantified as the time to platelet aggregation on the vessel wall and the time for complete blood flow cessation. While brief periods of cerebral ischemia were not associated with neurological deficits or brain infarction (evaluated after 1day), it yielded a pronounced and prolonged (up to 28days) acceleration of thrombus formation, compared to control (sham) mice. This prothrombotic phenotype was not altered by pre- and/or post-treatment of mice with either aspirin (A), clopidogrel (C), dipyridamole (D), or atorvastatin (S), or with A+D+S. CONCLUSIONS: The increased vulnerability of the cerebral vasculature to thrombus development after a brief period of transient ischemia can be recapitulated in a murine model. Antiplatelet or antithrombotic agents used in patients with TIA show no benefit in this mouse model of brief transient ischemia.


Subject(s)
Fibrinolytic Agents/therapeutic use , Infarction, Middle Cerebral Artery/physiopathology , Intracranial Thrombosis , Microvessels/physiopathology , Animals , Brain Infarction/etiology , Disease Models, Animal , Drug Therapy, Combination , Fibrinogen/metabolism , Intracranial Thrombosis/drug therapy , Intracranial Thrombosis/etiology , Intracranial Thrombosis/pathology , Mice , Mice, Inbred C57BL , Microscopy, Video , Microvessels/drug effects , Neurologic Examination , Platelet Glycoprotein GPIb-IX Complex/metabolism , Reperfusion Injury/complications , Statistics, Nonparametric , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...