Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 22
1.
Clin Neurol Neurosurg ; 239: 108246, 2024 04.
Article En | MEDLINE | ID: mdl-38507988

BACKGROUND: Traumatic subdural effusion (TSDE) may increase progressively or evolve into chronic subdural hematoma. These events, defined as deterioration of the effusion, often require close observation or even surgical treatment. The aim of our study was to develop and validate a nomogram for predicting the possibility of an effusion deteriorating in patients with TSDE based on the available clinical characteristics. METHODS: Clinical data from 78 patients with TSDE were retrospectively analyzed. All patients were admitted from January 2019 to May 2022. Logistic regression was applied to the data to screen for independent predictors of effusion deterioration within six months; then, a predictive nomogram model was established in R language. The consistency, predictive accuracy and clinical utility of the model were evaluated with the C-index, calibration plots, ROC curves and decision curve analysis (DCA). Furthermore, we performed internal validation using a bootstrap approach to assess the effectiveness of the model. RESULTS: Time of effusion after trauma, maximum thickness of the effusion, CT value of the effusion as well as the use of atorvastatin were identified as predictors in the nomogram. The predictive model was well calibrated and demonstrated good discrimination (C-index: 0.893). The AUC of the model was 0.893 (95% CI: 0.824-0.962), and the modified C-index (0.865) indicated excellent performance in the internal validation. In addition, DCA revealed that the nomogram had clinical value. CONCLUSIONS: This predictive model can effectively assess the risk of effusion deterioration in TSDE patients within six months and identify high-risk patients early.


Body Fluids , Subdural Effusion , Humans , Nomograms , Retrospective Studies , Atorvastatin
2.
Sensors (Basel) ; 23(15)2023 Aug 07.
Article En | MEDLINE | ID: mdl-37571787

Soil salinization is a major obstacle to land productivity, crop yield and crop quality in arid areas and directly affects food security. Soil profile salt data are key for accurately determining irrigation volumes. To explore the potential for using Landsat 8 time-series data to monitor soil salinization, 172 Landsat 8 images from 2013 to 2019 were obtained from the Alar Reclamation Area of Xinjiang, northwest China. The multiyear extreme dataset was synthesized from the annual maximum or minimum values of 16 vegetation indices, which were combined with the soil conductivity of 540 samples from soil profiles at 0~0.375 m, 0~0.75 m and 0~1.00 m depths in 30 cotton fields with varying degrees of salinization as investigated by EM38-MK2. Three remote sensing monitoring models for soil conductivity at different depths were constructed using the Cubist method, and digital mapping was carried out. The results showed that the Cubist model of soil profile electrical conductivity from 0 to 0.375 m, 0 to 0.75 m and 0 to 1.00 m showed high prediction accuracy, and the determination coefficients of the prediction set were 0.80, 0.74 and 0.72, respectively. Therefore, it is feasible to use a multiyear extreme value for the vegetation index combined with a Cubist modeling method to monitor soil profile salinization at a regional scale.

3.
Front Surg ; 9: 995986, 2022.
Article En | MEDLINE | ID: mdl-36329982

Primary intraosseous meningiomas (PIOMs) are a rare subset of meningiomas, comprising fewer than 1% of all such tumors. Furthermore, PIOMs presenting as osteogenic lesions that invade both the dura and subcutaneous tissue are extremely rare. Unlike intracranial meningiomas, diagnosing and treating PIOMs are challenges due to their insidious clinical behavior and a lack of clear radiological diagnostic criteria. We report the case of a 60-year-old female with headache and a slightly outward protrusion of the parietal region of the skull. CT showed an osteogenic lesion in the right parietal bone. MR imaging indicated mild to moderate homogeneous enhancement with an intense dural reaction. The suggested clinical diagnosis was lymphoma, so we performed a skull biopsy, which revealed an intraosseous benign meningioma. A precise resection strategy was planned with a neuronavigation system accompanied by a one-step customized titanium mesh cranioplasty. The lesion was completely removed, and pathological analysis confirmed a meningothelial meningioma (WHO Grade I) of intraosseous layer origin invading the dura mater and subcutaneous tissue. This case highlights the need for an initial biopsy when the lesion is difficult to diagnose on imaging. Complete resection should be attempted to minimize the risk of recurrence.

4.
Sensors (Basel) ; 22(16)2022 Aug 16.
Article En | MEDLINE | ID: mdl-36015885

Soil organic carbon (SOC) plays an important role in the global carbon cycle and soil fertility supply. Rapid and accurate estimation of SOC content could provide critical information for crop production, soil management and soil carbon pool regulation. Many researchers have confirmed the feasibility and great potential of visible and near-infrared (Vis-NIR) spectroscopy in evaluating SOC content rapidly and accurately. Here, to evaluate the feasibility of different spectral bands variable selection methods for SOC prediction, we collected a total of 330 surface soil samples from the cotton field in the Alar Reclamation area in the southern part of Xinjiang, which is located in the arid region of northwest China. Then, we estimated the SOC content using laboratory Vis-NIR spectral. The Particle Swarm optimization (PSO), Competitive adaptive reweighted sampling (CARS) and Ant colony optimization (ACO) were adopted to select SOC feature bands. The partial least squares regression (PLSR), random forest (RF) and convolutional neural network (CNN) inversion models were constructed by using full-bands (400-2400 nm) spectra (R) and feature bands, respectively. And we also analyzed the effects of spectral feature band selection methods and modeling methods on the prediction accuracy of SOC. The results indicated that: (1) There are significant differences in the feature bands selected using different methods. The feature bands selected methods substantially reduced the spectral variable dimensionality and model complexity. The models built by the feature bands selected by CARS, PSO and ACO methods showed the different potential of improvement in model accuracy compared with the full-band models. (2) The CNN model had the best performance for predicting SOC. The R2 of the optimal CNN model is 0.90 in the validation, which was improved by 0.05 and 0.04 in comparison with the PLSR and RF model, respectively. (3) The highest prediction accuracy was archived by the CNN model using the feature bands selected by CARS (validation set R2 = 0.90, RMSE = 0.97 g kg-1, RPD = 3.18, RPIQ = 3.11). This study indicated that using the CARS method to select spectral feature bands, combined with the CNN modeling method can well predict SOC content with higher accuracy.


Carbon , Soil , Carbon/analysis , China , Least-Squares Analysis , Soil/chemistry , Spectroscopy, Near-Infrared/methods
5.
Front Immunol ; 13: 993444, 2022.
Article En | MEDLINE | ID: mdl-36685592

Glioblastoma (GBM) is the most common and malignant primary brain tumor, resulting in poor survival despite aggressive therapies. GBM is characterized in part by a highly heterogeneous and immunosuppressive tumor microenvironment (TME) made up predominantly of infiltrating peripheral immune cells. One significant immune cell type that contributes to glioma immune evasion is a population of immunosuppressive, hematopoietic cells, termed myeloid-derived suppressor cells (MDSCs). Previous studies suggest that a potent subset of myeloid cells, expressing monocytic (M)-MDSC markers, distinguished by dual expression of chemokine receptors CCR2 and CX3CR1, utilize CCR2 to infiltrate into the TME. This study evaluated the T cell suppressive function and migratory properties of CCR2+/CX3CR1+ MDSCs. Bone marrow-derived CCR2+/CX3CR1+ cells adopt an immune suppressive cell phenotype when cultured with glioma-derived factors. Recombinant and glioma-derived CCL2 and CCL7 induce the migration of CCR2+/CX3CR1+ MDSCs with similar efficacy. KR158B-CCL2 and -CCL7 knockdown murine gliomas contain equivalent percentages of CCR2+/CX3CR1+ MDSCs compared to KR158B gliomas. Combined neutralization of CCL2 and CCL7 completely blocks CCR2-expressing cell migration to KR158B cell conditioned media. CCR2+/CX3CR1+ cells are also reduced within KR158B gliomas upon combination targeting of CCL2 and CCL7. High levels of CCL2 and CCL7 are also associated with negative prognostic outcomes in GBM patients. These data provide a more comprehensive understanding of the function of CCR2+/CX3CR1+ MDSCs and the role of CCL2 and CCL7 in the recruitment of these immune suppressive cells and further support the significance of targeting this chemokine axis in GBM.


Glioblastoma , Glioma , Myeloid-Derived Suppressor Cells , Animals , Mice , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL7/metabolism , CX3C Chemokine Receptor 1/metabolism , Glioblastoma/pathology , Monocytes/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Tumor Microenvironment
6.
Proc Natl Acad Sci U S A ; 117(2): 1129-1138, 2020 01 14.
Article En | MEDLINE | ID: mdl-31879345

Immunotherapy directed at the PD-L1/PD-1 axis has produced treatment advances in various human cancers. Unfortunately, progress has not extended to glioblastoma (GBM), with phase III clinical trials assessing anti-PD-1 monotherapy failing to show efficacy in newly diagnosed and recurrent tumors. Myeloid-derived suppressor cells (MDSCs), a subset of immunosuppressive myeloid derived cells, are known to infiltrate the tumor microenvironment of GBM. Growing evidence suggests the CCL2-CCR2 axis is important for this process. This study evaluated the combination of PD-1 blockade and CCR2 inhibition in anti-PD-1-resistant gliomas. CCR2 deficiency unmasked an anti-PD-1 survival benefit in KR158 glioma-bearing mice. CD11b+/Ly6Chi/PD-L1+ MDSCs within established gliomas decreased with a concomitant increase in overall CCR2+ cells and MDSCs within bone marrow of CCR2-deficient mice. The CCR2 antagonist CCX872 increased median survival as a monotherapy in KR158 glioma-bearing animals and further increased median and overall survival when combined with anti-PD-1. Additionally, combination of CCX872 and anti-PD-1 prolonged median survival time in 005 GSC GBM-bearing mice. In both models, CCX872 decreased tumor associated MDSCs and increased these cells within the bone marrow. Examination of tumor-infiltrating lymphocytes revealed an elevated population, increased IFNγ expression, indicating enhanced cytolytic activity, as well as decreased expression of exhaustion markers in CD4+ and CD8+ T cells following combination treatment. These data establish that combining CCR2 and PD-1 blockade extends survival in clinically relevant murine glioma models and provides the basis on which to advance this combinatorial treatment toward early-phase human trials.


B7-H1 Antigen/antagonists & inhibitors , Glioma/drug therapy , Myeloid Cells/metabolism , Receptors, CCR2/drug effects , Receptors, CCR2/metabolism , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Chemokine CCL2 , Disease Models, Animal , Gene Knock-In Techniques , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioma/pathology , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , Myeloid-Derived Suppressor Cells/metabolism , Programmed Cell Death 1 Receptor , Receptors, CCR2/genetics , Survival Analysis , Tumor Microenvironment/drug effects
8.
J Mol Med (Berl) ; 94(11): 1255-1265, 2016 11.
Article En | MEDLINE | ID: mdl-27344677

In this study, the role of CX3CR1 in the progression of diabetic retinopathy (DR) was investigated. The retinas of wild-type (WT), CX3CR1 null (CX3CR1gfp/gfp, KO), and heterozygous (CX3CR1+/gfp, Het) mice were compared in the presence and absence of streptozotocin (STZ)-induced diabetes. CX3CR1 deficiency in STZ-KO increased vascular pathology at 4 months of diabetes, as a significant increase in acellular capillaries was observed only in the STZ-KO group. CX3CR1 deficiency and diabetes had similar effects on retinal neurodegeneration measured by an increase in DNA fragmentation. Retinal vascular pathology in STZ-KO mice was associated with increased numbers of monocyte-derived macrophages in the retina. Furthermore, compared to STZ-WT, STZ-KO mice exhibited increased numbers of inflammatory monocytes in the bone marrow and impaired homing of monocytes to the spleen. The induction of retinal IL-10 expression by diabetes was significantly less in KO mice, and when bone marrow-derived macrophages from KO mice were maintained in high glucose, they expressed significantly less IL-10 and more TNF-α in response to LPS stimulation. These findings support that CX3CR1 deficiency accelerates the development of vascular pathology in DR through increased recruitment of proinflammatory myeloid cells that demonstrate reduced expression of anti-inflammatory IL-10. KEY MESSAGES: • CX3CR1 deletion in STZ-diabetic mice accelerated the onset of diabetic retinopathy (DR). • The early onset of DR was associated with increased retinal cell apoptosis. • The early onset of DR was associated with increased recruitment of bone marrow-derived macrophages to the retina. • Bone marrow-derived macrophages from CX3CR1 KO diabetic mice expressed more TNF-α and less IL-10. • The role of IL-10 in protection from progression of DR is highlighted.


CX3C Chemokine Receptor 1/deficiency , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Diabetic Retinopathy/metabolism , Diabetic Retinopathy/pathology , Animals , Apoptosis , Body Weight , Bone Marrow Cells/metabolism , CX3C Chemokine Receptor 1/metabolism , Disease Models, Animal , Gene Deletion , Glycated Hemoglobin/metabolism , Homeostasis , Hypothalamus/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Myeloid Cells/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retina/metabolism , Retina/pathology , Streptozocin
9.
Hypertension ; 67(3): 574-84, 2016 Mar.
Article En | MEDLINE | ID: mdl-26781279

Emerging evidence indicates that differentiation and mobilization of hematopoietic cell are critical in the development and establishment of hypertension and hypertension-linked vascular pathophysiology. This, coupled with the intimate involvement of the hyperactive renin-angiotensin system in hypertension, led us to investigate the hypothesis that chronic angiotensin II (Ang II) infusion affects hematopoietic stem cell (HSC) regulation at the level of the bone marrow. Ang II infusion resulted in increases in hematopoietic stem/progenitor cells (83%) and long-term HSC (207%) in the bone marrow. Interestingly, increases of HSCs and long-term HSCs were more pronounced in the spleen (228% and 1117%, respectively). Furthermore, we observed higher expression of C-C chemokine receptor type 2 in these HSCs, indicating there was increased myeloid differentiation in Ang II-infused mice. This was associated with accumulation of C-C chemokine receptor type 2(+) proinflammatory monocytes in the spleen. In contrast, decreased engraftment efficiency of GFP(+) HSC was observed after Ang II infusion. Time-lapse in vivo imaging and in vitro Ang II pretreatment demonstrated that Ang II induces untimely proliferation and differentiation of the donor HSC resulting in diminished HSC engraftment and bone marrow reconstitution. We conclude that (1) chronic Ang II infusion regulates HSC proliferation, mediated by angiotensin receptor type 1a, (2) Ang II accelerates HSC to myeloid differentiation resulting in accumulation of C-C chemokine receptor type 2(+) HSCs and inflammatory monocytes in the spleen, and (3) Ang II impairs homing and reconstitution potentials of the donor HSCs. These observations highlight the important regulatory roles of Ang II on HSC proliferation, differentiation, and engraftment.


Angiotensin II/metabolism , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Hypertension/pathology , Animals , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Hypertension/physiopathology , Hypertension/therapy , Male , Mice , Mice, Inbred BALB C , Video Recording
10.
Cancer Lett ; 360(1): 60-7, 2015 Apr 28.
Article En | MEDLINE | ID: mdl-25676691

The failure of standard treatment for patients diagnosed with glioblastoma (GBM) coupled with the highly vascularized nature of this solid tumor has led to the consideration of agents targeting VEGF or VEGFRs, as alternative therapeutic strategies for this disease. Despite modest achievements in survival obtained with such treatments, failure to maintain an enduring survival benefit and more invasive relapsing tumors are evident. Our study suggests a potential mechanism by which anti-VEGF/VEGFR therapies regulate the enhanced invasive phenotype through a pathway that involves TGFßR and CXCR4. VEGFR signaling inhibitors (Cediranib and Vandetanib) elevated the expression of CXCR4 in VEGFR-expressing GBM cell lines and tumors, and enhanced the in vitro migration of these lines toward CXCL12. The combination of VEGFR inhibitor and CXCR4 antagonist provided a greater survival benefit to tumor-bearing animals. The upregulation of CXCR4 by VEGFR inhibitors was dependent on TGFß/TGFßR, but not HGF/MET, signaling activity, suggesting a mechanism of crosstalk among VEGF/VEGFR, TGFß/TGFßR, and CXCL12/CXCR4 pathways in the malignant phenotype of recurrent tumors after anti-VEGF/VEGFR therapies. Thus, the combination of VEGFR, CXCR4, and TGFßR inhibitors could provide an alternative strategy to halt GBM progression.


Angiogenesis Inhibitors/pharmacology , Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , Receptors, CXCR4/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Signal Transduction/drug effects , Adult , Aged , Animals , Benzylamines , Brain Neoplasms/enzymology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Cell Movement/drug effects , Cyclams , Female , Glioblastoma/enzymology , Glioblastoma/genetics , Glioblastoma/pathology , Heterocyclic Compounds/pharmacology , Humans , Interleukin-2 Receptor alpha Subunit/deficiency , Interleukin-2 Receptor alpha Subunit/genetics , Male , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Middle Aged , Neoplasm Invasiveness , Receptor Cross-Talk/drug effects , Receptors, CXCR4/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/metabolism , Time Factors , Up-Regulation , Xenograft Model Antitumor Assays
11.
PLoS One ; 8(3): e59750, 2013.
Article En | MEDLINE | ID: mdl-23555768

Glioblastoma (GBM) is the most common primary brain tumor in adults. The poor prognosis and minimally successful treatments of these tumors indicates a need to identify new therapeutic targets. Therapy resistance of GBMs is attributed to heterogeneity of the glioblastoma due to genetic alterations and functional subpopulations. Chemokine receptors CXCR4 and CXCR7 play important roles in progression of various cancers although the specific functions of the CXCL12-CXCR4-CXCR7 axis in GBM are less characterized. In this study we examined the expression and function of CXCR4 and CXCR7 in four primary patient-derived GBM cell lines of the proliferative subclass, investigating their roles in in vitro growth, migration, sphere and tube formation. CXCR4 and CXCR7 cell surface expression was heterogeneous both between and within each cell line examined, which was not reflected by RT-PCR analysis. Variable percentages of CXCR4+CXCR7- (CXCR4 single positive), CXCR4-CXCR7+ (CXCR7 single positive), CXCR4+CXCR7+ (double positive), and CXCR4-CXCR7- (double negative) subpopulations were evident across the lines examined. A subpopulation of slow cell cycling cells was enriched in CXCR4 and CXCR7. CXCR4+, CXCR7+, and CXCR4+/CXCR7+ subpopulations were able to initiate intracranial tumors in vivo. CXCL12 stimulated in vitro cell growth, migration, sphere formation and tube formation in some lines and, depending on the response, the effects were mediated by either CXCR4 or CXCR7. Collectively, our results indicate a high level of heterogeneity in both the surface expression and functions of CXCR4 and CXCR7 in primary human GBM cells of the proliferative subclass. Should targeting of CXCR4 and CXCR7 provide clinical benefits to GBM patients, a personalized treatment approach should be considered given the differential expression and functions of these receptors in GBM.


Brain Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Animals , Apoptosis , Cell Membrane/metabolism , Cell Movement , Cell Proliferation , Chemokine CXCL12/pharmacology , Flow Cytometry , Gene Expression Profiling , Humans , Immunohistochemistry , Mice , Mice, Inbred NOD , Mice, SCID
12.
Am J Physiol Lung Cell Mol Physiol ; 303(10): L912-22, 2012 Nov 15.
Article En | MEDLINE | ID: mdl-23002075

Distal arterioles with limited smooth muscles help maintain the high blood flow and low pressure in the lung circulation. Chronic hypoxia induces lung distal vessel muscularization. However, the molecular events that trigger alveolar hypoxia-induced peripheral endothelium modulation of vessel wall smooth muscle cell (SMC) proliferation and filling of nonmuscular areas are unclear. Here, we investigated the role of CX3CL1/CX3CR1 system in endothelial-SMC cross talk in response to hypoxia. Human lung microvascular endothelial cells responded to alveolar oxygen deficiency by overproduction of the chemokine CX3CL1. The CX3CL1 receptor CX3CR1 is expressed by SMCs that are adjacent to the distal endothelium. Hypoxic release of endothelial CX3CL1 induced SMC phenotypic switching from the contractile to the proliferative state. Inhibition of CX3CR1 prevented CX3CL1 stimulation of SMC proliferation and monolayer expansion. Furthermore, CX3CR1 deficiency attenuated spiral muscle expansion, distal vessel muscularization, and pressure elevation in response to hypoxia. Our findings indicate that the capillary endothelium relies on the CX3CL1-CX3CR1 axis to sense alveolar hypoxia and promote peripheral vessel muscularization. These results have clinical significance in the development of novel therapeutics that target mechanisms of distal arterial remodeling associated with pulmonary hypertension induced by oxygen deficiency that is present in people living at high altitudes and patients with obstructive lung diseases.


Cell Proliferation , Chemokine CX3CL1/metabolism , Myocytes, Smooth Muscle/metabolism , Pulmonary Alveoli/metabolism , Animals , CX3C Chemokine Receptor 1 , Cell Hypoxia , Chemokine CX3CL1/genetics , Endothelial Cells/metabolism , Endothelial Cells/pathology , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Mice , Mice, Transgenic , Myocytes, Smooth Muscle/pathology , Pulmonary Alveoli/blood supply , Pulmonary Alveoli/pathology , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Pulmonary Disease, Chronic Obstructive/physiopathology , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
13.
J Neuroimmunol ; 246(1-2): 10-7, 2012 May 15.
Article En | MEDLINE | ID: mdl-22425022

Glioblastoma multiforme (GBM) is the most malignant brain tumor. Microglia/macrophages are found within human GBM where they likely promote tumor progression. We report that CCL5, CCR1, and CCR5 are expressed in glioblastoma. Individual deletion of CCR1 or CCR5 had little to no effect on survival of tumor bearing mice, or numbers of glioblastoma-infiltrated microglia/macrophages or lymphocytes. CCL5 promoted in vitro migration of wild type, CCR1- or CCR5-deficient microglia/macrophages that was blocked by the dual CCR1/CCR5 antagonist, Met-CCL5. These data suggest that CCL5 functions within the glioblastoma microenvironment through CCR1 and CCR5 in a redundant manner.


Brain Neoplasms/immunology , Cell Movement/immunology , Chemokine CCL5/biosynthesis , Gene Expression Regulation/immunology , Glioblastoma/immunology , Receptors, CCR1/biosynthesis , Receptors, CCR5/biosynthesis , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Survival/genetics , Cell Survival/immunology , Chemokine CCL5/physiology , Glioblastoma/genetics , Glioblastoma/pathology , Humans , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Mice , Microglia/immunology , Microglia/metabolism , Microglia/pathology , Receptors, CCR1/deficiency , Receptors, CCR1/genetics , Receptors, CCR5/deficiency , Receptors, CCR5/genetics
14.
Carcinogenesis ; 32(2): 129-37, 2011 Feb.
Article En | MEDLINE | ID: mdl-21051441

Human glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. The poor prognosis and minimally successful treatments of GBM indicates a need to identify new therapeutic targets. In this study, we examined the role of CXCR3 in glioma progression using the GL261 murine model of malignant glioma. Intracranial GL261 tumors express CXCL9 and CXCL10 in vivo. Glioma-bearing CXCR3-deficient mice had significantly shorter median survival time and reduced numbers of tumor-infiltrated natural killer and natural killer T cells as compared with tumor-bearing wild-type (WT) mice. In contrast, pharmacological antagonism of CXCR3 with NBI-74330 prolonged median survival times of both tumor-bearing WT and CXCR3-deficient mice when compared with vehicle-treated groups. NBI-74330 treatment did not impact tumor infiltration of lymphocytes and microglia. A small percentage of GL261 cells were identified as CXCR3(+), which was similar to the expression of CXCR3 in several grade IV human glioma cell lines (A172, T98G, U87, U118 and U138). When cultured as gliomaspheres (GS), the human and murine lines increased CXCR3 expression; CXCR3 expression was also found in a primary human GBM-derived GS. Additionally, CXCR3 isoform A was expressed by all lines, whereas CXCR3-B was detected in T98G-, U118- and U138-GS cells. CXCL9 or CXCL10 induced in vitro glioma cell growth in GL261- and U87-GS as well as inhibited cell loss in U138-GS cells and this effect was antagonized by NBI-74330. The results suggest that CXCR3 antagonism exerts a direct anti-glioma effect and this receptor may be a potential therapeutic target for treating human GBM.


Brain Neoplasms/pathology , Glioma/pathology , Receptors, CXCR3/physiology , Animals , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Chemokine CXCL10/genetics , Chemokine CXCL10/physiology , Chemokine CXCL9/genetics , Chemokine CXCL9/physiology , Glioma/immunology , Glioma/mortality , Humans , Mice , Mice, Inbred C57BL , RNA, Messenger/analysis , Receptors, CXCR3/antagonists & inhibitors
15.
Differentiation ; 78(1): 35-44, 2009 Jul.
Article En | MEDLINE | ID: mdl-19427096

The Hematopoietic- and neurologic-expressed sequence 1 (Hn1) gene encodes a small protein that is highly conserved among species. Hn1 expression is upregulated in regenerating neural tissues, including the axotomized adult rodent facial motor nerve and dedifferentiating retinal pigment epithelial cells of the Japanese newt. It is also expressed in numerous tissues during embryonic development as well as in regions of the adult brain that exhibit high plasticity. Hn1 has also been reported as a marker for human ovarian carcinoma and it is expressed in high-grade human gliomas. This study was directed toward understanding the function of Hn1 in a murine melanoma cell line. Hn1 mRNA and protein were identified in B16.F10 cells and in tumors formed from these cells. Inhibition of Hn1 protein expression with siRNA increased melanogenesis. Hn1-depleted cells expressed higher levels of the melanogenic proteins tyrosinase and Trp2 and an increased interaction between actin and Rab27a. The in vitro cell growth rate of Hn1-depleted cells was significantly reduced due to G1/S cell cycle arrest. This was consistent with a reduction in the phosphorylation of retinoblastoma protein as well as lower levels of p27 and increased expression of p21. Decreased expression of c-Met, the receptor for hepatocyte growth factor, was also detected in the Hn1-depleted cells, however HGF-dependent stimulation of phosphorylated-ERK was unaffected. Hn1 depletion also led to increased basal levels of phosphorylated p38 MAPK, while basal ERK phosphorylation was reduced. Moreover, Hn1-depleted cells had reduced expression of transcription factors MITF and USF-1, and increased expression of TFE3. These data, coupled with reports on Hn1 expression in regeneration and development, suggest that Hn1 functions as a suppressor of differentiation in cells undergoing repair or proliferation.


Cell Cycle/genetics , Cell Differentiation/genetics , Melanins/genetics , Melanoma, Experimental/genetics , Nerve Tissue Proteins/genetics , Adenoviridae/genetics , Animals , Cell Cycle/physiology , Cell Cycle Proteins , Cell Differentiation/physiology , Cell Line, Tumor , Escherichia coli/genetics , Fluorescent Antibody Technique, Direct , Fluorescent Dyes/metabolism , Genetic Vectors , Immunohistochemistry , In Situ Hybridization , Indoles/metabolism , Melanins/physiology , Melanoma, Experimental/physiopathology , Mice , Microtubule-Associated Proteins , Nerve Tissue Proteins/physiology , Phenotype , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/physiology , Transduction, Genetic
16.
Pathol Oncol Res ; 15(3): 437-44, 2009 Sep.
Article En | MEDLINE | ID: mdl-19145478

The hematopoietic- and neurologic-expressed sequence 1 (Hn1) gene encodes a highly conserved protein that is expressed in developing and regenerating tissues. In this study, Hn1 expression was evaluated in human and murine malignant gliomas. Hn1 mRNA and protein were detected in the murine GL261 glioma cell line and in GL261 brain tumors in vivo. HN1 is also expressed in human U118MG and U87MG cell lines. Evaluation of human brain tumors using an anti-Hn1 polyclonal antibody detected strong immunoreactivity in high-grade (WHO III and IV) malignant gliomas. The rate of GL261 cell proliferation in vitro was unaltered by Hn1 depletion using an anti-Hn1 siRNA. However, tumors established from Hn1-depleted GL261 cells formed significantly smaller volumes than those established from control-treated cells. These data suggest a role for Hn1 in the biology of malignant brain tumors.


Brain Neoplasms/metabolism , Glioma/metabolism , Nerve Tissue Proteins/biosynthesis , Animals , Blotting, Northern , Blotting, Western , Brain Neoplasms/pathology , Cell Cycle Proteins , Cell Line, Tumor , Cell Proliferation , Glioma/pathology , Humans , In Situ Hybridization , Mice , Microtubule-Associated Proteins , RNA, Messenger/analysis , RNA, Small Interfering , Tissue Array Analysis
17.
J Neuroimmunol ; 198(1-2): 98-105, 2008 Jul 31.
Article En | MEDLINE | ID: mdl-18508133

Human glioblastoma multiforme (GBM) is the most malignant form of human brain tumors. A characteristic of GBM is the marked presence of tumor infiltrated microglia/macrophages and lymphocytes. The goal of this study was directed toward understanding the role of the chemokine system CX3CL1 and its receptor CX3CR1 in the GL261 murine model of malignant glioma. In situ hybridization analysis identified CX3CL1 and CX3CR1 expression in GL261 tumors. The impact of CX3CR1 deletion on the growth of intracranial GL261 gliomas and associated immune cell infiltration was evaluated in CX3CR1 gene-disrupted C57BL/6 mice. A slight increase in the tumor growth rate in CX3CR1-/- mice was evident with similar numbers of microglia and CD4+, CD8+, FoxP3+, or Ly49G2+ lymphocytes within tumors established in CX3CR1 +/- and -/- mice. These data indicate that CX3CR1 has little or no effects on either gliomagenesis or the migration of microglia and lymphocytes into GL261 tumors.


Chemokine CX3CL1/metabolism , Glioma/metabolism , Glioma/pathology , Lymphocytes/immunology , Microglia/immunology , Receptors, Chemokine/metabolism , Animals , Antigens, CD/metabolism , Antigens, Ly/metabolism , CX3C Chemokine Receptor 1 , Cell Movement , Disease Models, Animal , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/physiology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lectins, C-Type/metabolism , Lymphocytes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/metabolism , Receptors, Chemokine/deficiency , Receptors, NK Cell Lectin-Like , Survival Analysis
18.
J Neurosci Res ; 82(5): 581-91, 2005 Dec 01.
Article En | MEDLINE | ID: mdl-16267826

Facial nerve axotomy (FNA) is a well-established experimental model of motoneuron regeneration. After peripheral nerve axotomy, a sequence of events including glial activation and axonal regrowth leads to functional recovery of the afflicted pool of motoneurons. Using microarray analysis we identified an increase in the expression of 60 genes (at a false discovery rate of 0.1, genes were significant P < 0.004) within the facial nucleus as a consequence of nerve injury. In situ hybridization analysis validated the increased expression of many of these axotomy-induced genes. One specific gene, encoding a unique primary amino acid sequence, termed hemopoietic- and neurologic-expressed sequence-1 (Hn1), was evaluated more extensively using several additional nerve injury paradigms. Hn1 mRNA was upregulated in injured facial motoneurons in both rats and mice. Sustained upregulation of Hn1 mRNA was evident after nerve resection whereas levels of Hn1 mRNA returned to baseline in animals subjected to nerve crush or nerve transection. Hn1 was also increased in the dorsal motor nucleus and the nucleus ambiguous after vagus nerve axotomy, another regeneration model. No upregulation of Hn1 expression was observed, however, in two nonregeneration models: FNA in newborn rats and rubrospinal tractotomy. Hn1 mRNA was ubiquitous in the developing central nervous system whereas its expression in adult brain was confined to neurons of the hippocampus, cortex and cerebellum. These findings identify Hn1 as a gene associated with nervous system development and nerve regeneration.


Facial Nerve Injuries/genetics , Facial Nerve/metabolism , Nerve Regeneration/genetics , Nerve Tissue Proteins/genetics , Proteins/genetics , Transcriptional Activation/genetics , Animals , Axotomy , Cell Cycle Proteins , Facial Nerve/cytology , Facial Nerve Injuries/metabolism , Gene Expression Regulation/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins , Motor Neurons/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Rhombencephalon/cytology , Rhombencephalon/metabolism , Up-Regulation/physiology , Vagus Nerve/cytology , Vagus Nerve/metabolism , Vagus Nerve Diseases/genetics , Vagus Nerve Diseases/metabolism
19.
Gene Expr Patterns ; 3(4): 459-62, 2003 Aug.
Article En | MEDLINE | ID: mdl-12915312

The RNA-mediated pathogenesis model for the myotonic dystrophies DM1 and DM2 proposes that mutant transcripts from the affected genes sequester a family of double-stranded RNA-binding factors, the muscleblind proteins MBNL1, MBNL2 and MBNL3, in the nucleus. These proteins are homologues of the Drosophila muscleblind proteins that are required for the terminal differentiation of muscle and photoreceptor tissues, and thus nuclear sequestration of the human proteins might impair their normal function in muscle and eye development and maintenance. To examine this model further, we analyzed the expression pattern of the mouse Mbnl1, Mbnl2, and Mbnl3 genes during embryonic development and compared muscleblind gene expression to Dmpk since the RNA pathogenesis model for DM1 requires the coordinate synthesis of mutant Dmpk transcripts and muscleblind proteins. Our studies reveal a striking overlap between the expression of Dmpk and the muscleblind genes during development of the limbs, nervous system and various muscles, including the diaphragm and tongue.


Gene Expression Regulation, Developmental , RNA-Binding Proteins/genetics , Zinc Fingers/genetics , Animals , Antisense Elements (Genetics) , DNA-Binding Proteins , Extremities/embryology , Humans , In Situ Hybridization , Mice , Muscles/embryology , Myoblasts/cytology , Myoblasts/physiology , Myotonin-Protein Kinase , Nervous System/embryology , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA-Binding Proteins/metabolism
20.
Methods ; 29(4): 312-8, 2003 Apr.
Article En | MEDLINE | ID: mdl-12725797

The expression of a number of chemokines and chemokine receptors by cells resident in normal and pathological central nervous system (CNS) tissue has been characterized by in situ hybridization techniques. As a result, our understanding of the role of this cytokine family in neurobiology has been enhanced greatly. Specific methods for detecting chemokine and chemokine receptor mRNAs in situ vary with the number of these genes that have been characterized and encompass approaches widely utilized by other investigators characterizing cell-specific gene expression patterns. We describe methods that our laboratory has used successfully in characterizing chemokine and chemokine receptor expression in the CNS, focusing on protocols that utilize radiolabeled in vitro-transcribed riboprobes for detecting these transcripts. Because general dye-based histological staining methods do not readily differentiate astrocytes and microglia, specific immunohistochemical protocols are required for definitive localization of gene expression to these glial cell types. As such, methods that are compatible with the in situ hybridization procedure are included for staining astrocytes and microglia.


Central Nervous System/metabolism , Chemokines/biosynthesis , In Situ Hybridization/methods , Receptors, Chemokine/biosynthesis , Animals , Astrocytes/metabolism , Benzoxazines , Blotting, Northern/methods , Coloring Agents/pharmacology , Immunohistochemistry , Neuroglia/pathology , Oxazines/pharmacology , Plasmids/metabolism , RNA, Messenger/metabolism , Rats , Transcription, Genetic
...