Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 9(1): 19162, 2019 12 16.
Article in English | MEDLINE | ID: mdl-31844087

ABSTRACT

Human cerebral malaria (HCM), a severe encephalopathy associated with Plasmodium falciparum infection, has a 20-30% mortality rate and predominantly affects African children. The mechanisms mediating HCM-associated brain injury are difficult to study in human subjects, highlighting the urgent need for non-invasive ex vivo human models. HCM elevates the systemic levels of free heme, which damages the blood-brain barrier and neurons in distinct regions of the brain. We determined the effects of heme on induced pluripotent stem cells (iPSCs) and a three-dimensional cortical organoid system and assessed apoptosis and differentiation. We evaluated biomarkers associated with heme-induced brain injury, including a pro-inflammatory chemokine, CXCL-10, and its receptor, CXCR3, brain-derived neurotrophic factor (BDNF) and a receptor tyrosine-protein kinase, ERBB4, in the organoids. We then tested the neuroprotective effect of neuregulin-1 (NRG-1) against heme treatment in organoids. Neural stem and mature cells differentially expressed CXCL-10, CXCR3, BDNF and ERBB4 in the developing organoids and in response to heme-induced neuronal injury. The organoids underwent apoptosis and structural changes that were attenuated by NRG-1. Thus, cortical organoids can be used to model heme-induced cortical brain injury associated with HCM pathogenesis as well as for testing agents that reduce brain injury and neurological sequelae.


Subject(s)
Brain Injuries/pathology , Cerebral Cortex/pathology , Malaria, Cerebral/pathology , Models, Biological , Organoids/pathology , Apoptosis , Brain-Derived Neurotrophic Factor/metabolism , Cell Differentiation , Cells, Cultured , Chemokine CXCL12/metabolism , Heme , Humans , Induced Pluripotent Stem Cells/pathology , Inflammation/pathology , Neuregulin-1/metabolism , Receptor, ErbB-4 , Receptors, CXCR3/metabolism , Umbilical Cord/cytology
2.
J Neuroinflammation ; 11: 9, 2014 Jan 17.
Article in English | MEDLINE | ID: mdl-24433482

ABSTRACT

BACKGROUND: Cerebral Malaria (CM) is a diffuse encephalopathy caused by Plasmodium falciparum infection. Despite availability of antimalarial drugs, CM-associated mortality remains high at approximately 30% and a subset of survivors develop neurological and cognitive disabilities. While antimalarials are effective at clearing Plasmodium parasites they do little to protect against CM pathophysiology and parasite-induced brain inflammation that leads to seizures, coma and long-term neurological sequelae in CM patients. Thus, there is urgent need to explore therapeutics that can reduce or prevent CM pathogenesis and associated brain inflammation to improve survival. Neuregulin-1 (NRG-1) is a neurotrophic growth factor shown to protect against brain injury associated with acute ischemic stroke (AIS) and neurotoxin exposure. However, this drug has not been tested against CM-associated brain injury. Since CM-associated brain injuries and AIS share similar pathophysiological features, we hypothesized that NRG-1 will reduce or prevent neuroinflammation and brain damage as well as improve survival in mice with late-stage experimental cerebral malaria (ECM). METHODS: We tested the effects of NRG-1 on ECM-associated brain inflammation and mortality in P. berghei ANKA (PbA)-infected mice and compared to artemether (ARM) treatment; an antimalarial currently used in various combination therapies against malaria. RESULTS: Treatment with ARM (25 mg/kg/day) effectively cleared parasites and reduced mortality in PbA-infected mice by 82%. Remarkably, NRG-1 therapy (1.25 ng/kg/day) significantly improved survival against ECM by 73% despite increase in parasite burden within NRG-1-treated mice. Additionally, NRG-1 therapy reduced systemic and brain pro-inflammatory factors TNFalpha, IL-6, IL-1alpha and CXCL10 and enhanced anti-inflammatory factors, IL-5 and IL-13 while decreasing leukocyte accumulation in brain microvessels. CONCLUSIONS: This study suggests that NRG-1 attenuates ECM-associated brain inflammation and injuries and may represent a novel supportive therapy for the management of CM.


Subject(s)
Antimalarials/therapeutic use , Encephalitis/drug therapy , Malaria, Cerebral/drug therapy , Malaria, Cerebral/mortality , Neuregulin-1/therapeutic use , Neuroprotective Agents/therapeutic use , Animals , Artemether , Artemisinins/therapeutic use , Behavior, Animal/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/pathology , Brain/parasitology , Brain/pathology , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Encephalitis/etiology , Encephalitis/pathology , Endothelium/drug effects , Endothelium/pathology , Leukocytes/drug effects , Leukocytes/pathology , Malaria, Cerebral/complications , Mice , Mice, Inbred C57BL , Neuregulin-1/metabolism , Plasmodium berghei/physiology
3.
PLoS One ; 8(4): e60898, 2013.
Article in English | MEDLINE | ID: mdl-23630573

ABSTRACT

Despite appropriate anti-malarial treatment, cerebral malaria (CM)-associated mortalities remain as high as 30%. Thus, adjunctive therapies are urgently needed to prevent or reduce such mortalities. Overproduction of CXCL10 in a subset of CM patients has been shown to be tightly associated with fatal human CM. Mice with deleted CXCL10 gene are partially protected against experimental cerebral malaria (ECM) mortality indicating the importance of CXCL10 in the pathogenesis of CM. However, the direct effect of increased CXCL10 production on brain cells is unknown. We assessed apoptotic effects of CXCL10 on human brain microvascular endothelial cells (HBVECs) and neuroglia cells in vitro. We tested the hypothesis that reducing overexpression of CXCL10 with a synthetic drug during CM pathogenesis will increase survival and reduce mortality. We utilized atorvastatin, a widely used synthetic blood cholesterol-lowering drug that specifically targets and reduces plasma CXCL10 levels in humans, to determine the effects of atorvastatin and artemether combination therapy on murine ECM outcome. We assessed effects of atorvastatin treatment on immune determinants of severity, survival, and parasitemia in ECM mice receiving a combination therapy from onset of ECM (day 6 through 9 post-infection) and compared results with controls. The results indicate that CXCL10 induces apoptosis in HBVECs and neuroglia cells in a dose-dependent manner suggesting that increased levels of CXCL10 in CM patients may play a role in vasculopathy, neuropathogenesis, and brain injury during CM pathogenesis. Treatment of ECM in mice with atorvastatin significantly reduced systemic and brain inflammation by reducing the levels of the anti-angiogenic and apoptotic factor (CXCL10) and increasing angiogenic factor (VEGF) production. Treatment with a combination of atorvastatin and artemether improved survival (100%) when compared with artemether monotherapy (70%), p<0.05. Thus, adjunctively reducing CXCL10 levels and inflammation by atorvastatin treatment during anti-malarial therapy may represent a novel approach to treating CM patients.


Subject(s)
Antimalarials/pharmacology , Chemokine CXCL10/antagonists & inhibitors , Malaria, Cerebral/drug therapy , Malaria, Cerebral/metabolism , Animals , Antimalarials/therapeutic use , Apoptosis/drug effects , Artemether , Artemisinins/pharmacology , Atorvastatin , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Caspases/metabolism , Cell Count , Chemokine CXCL10/blood , Chemokine CXCL10/genetics , Chemokine CXCL10/pharmacology , Disease Models, Animal , Drug Interactions , Endothelial Cells/cytology , Endothelial Cells/drug effects , Gene Expression Regulation/drug effects , Heme Oxygenase-1/metabolism , Heptanoic Acids/pharmacology , Heptanoic Acids/therapeutic use , Humans , Leukocytes/cytology , Leukocytes/drug effects , Malaria, Cerebral/immunology , Malaria, Cerebral/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Models, Molecular , Neuroglia/cytology , Neuroglia/drug effects , Nucleic Acid Conformation , Pyrroles/pharmacology , Pyrroles/therapeutic use , Survival Analysis
4.
J Steroid Biochem Mol Biol ; 135: 7-14, 2013 May.
Article in English | MEDLINE | ID: mdl-23201171

ABSTRACT

Vitamin D hormone (1,25-dihydroxyvitamin D) is involved in innate immunity and induces host defense peptides in epithelial cells, suggesting its involvement in mucosal defense against infections. Chlamydia trachomatis is a major cause of bacterial sexually transmitted disease worldwide. We tested the hypothesis that the vitamin D endocrine system would attenuate chlamydial infection. Vitamin D receptor knock-out mice (VDR(-/-)) and wild-type mice (VDR(+/+)) were infected with 10(3) inclusion forming units of Chlamydia muridarum and cervical epithelial cells (HeLa cells) were infected with C. muridarum at multiplicity of infection 5:1 in the presence and absence of 1,25-dihydroxyvitamin D3. VDR(-/-) mice exhibited significantly higher bacterial loading than wild-type VDR(+/+) mice (P<0.01) and cleared the chlamydial infection in 39 days, compared with 18 days for VDR(+/+) mice. Monocytes and neutrophils were more numerous in the uterus and oviduct of VDR(-/-) mice than in VDR(+/+) mice (P<0.05) at d 45 after infection. Pre-treatment of HeLa cells with 10nM or 100nM 1,25-dihydroxyvitamin D3 decreased the infectivity of C. muridarum (P<0.001). Several differentially expressed protein spots were detected by proteomic analysis of chlamydial-infected HeLa cells pre-treated with 1,25-dihydroxyvitamin D3. Leukocyte elastase inhibitor (LEI), an anti-inflammatory protein, was up-regulated. Expression of LEI in the ovary and oviduct of infected VDR(+/+) mice was greater than that of infected VDR(-/-) mice. We conclude that the vitamin D endocrine system reduces the risk for prolonged chlamydial infections through regulation of several proteins and that LEI is involved in its anti-inflammatory activity.


Subject(s)
Calcitriol/pharmacology , Chlamydia muridarum/pathogenicity , Chlamydiaceae Infections/metabolism , Receptors, Calcitriol/physiology , Animals , Bacterial Load , Cell Line, Tumor , Chlamydiaceae Infections/immunology , Chlamydiaceae Infections/microbiology , Chlamydiaceae Infections/pathology , Female , Gene Expression Regulation , HeLa Cells , Humans , Leukocyte Elastase/antagonists & inhibitors , Mice , Mice, Inbred C57BL , Mice, Knockout , Proteome , Receptors, Calcitriol/deficiency , Receptors, Calcitriol/genetics , Serpins/metabolism
5.
PLoS One ; 7(3): e34280, 2012.
Article in English | MEDLINE | ID: mdl-22479586

ABSTRACT

BACKGROUND: The mortality of severe malaria [cerebral malaria (CM), severe malaria anemia (SMA), acute lung injury (ALI) and acute respiratory distress syndrome (ARDS)] remains high despite the availability associated with adequate treatments. Recent studies in our laboratory and others have revealed a hitherto unknown correlation between chemokine CXCL10/CXCR3, Heme/HO-1 and STAT3 and cerebral malaria severity and mortality. Although Heme/HO-1 and CXCL10/CXCR3 interactions are directly involved in the pathogenesis of CM and fatal disease, the mechanism dictating how Heme/HO-1 and CXCL10/CXCR3 are expressed and regulated under these conditions is still unknown. We therefore tested the hypothesis that these factors share common signaling pathways and may be mutually regulated. METHODS: We first clarified the roles of Heme/HO-1, CXCL10/CXCR3 and STAT3 in CM pathogenesis utilizing a well established experimental cerebral malaria mouse (ECM, P. berghei ANKA) model. Then, we further determined the mechanisms how STAT3 regulates HO-1 and CXCL10 as well as mutual regulation among them in CRL-2581, a murine endothelial cell line. RESULTS: The results demonstrate that (1) STAT3 is activated by P. berghei ANKA (PBA) infection in vivo and Heme in vitro. (2) Heme up-regulates HO-1 and CXCL10 production through STAT3 pathway, and regulates CXCL10 at the transcriptional level in vitro. (3) HO-1 transcription is positively regulated by CXCL10. (4) HO-1 regulates STAT3 signaling. CONCLUSION: Our data indicate that Heme/HO-1, CXCL10/CXCR3 and STAT3 molecules as well as related signaling pathways play very important roles in the pathogenesis of severe malaria. We conclude that these factors are mutually regulated and provide new opportunities to develop potential novel therapeutic targets that could be used to supplement traditional prophylactics and treatments for malaria and improve clinical outcomes while reducing malaria mortality. Our ultimate goal is to develop novel therapies targeting Heme or CXCL10-related biological signaling molecules associated with development of fatal malaria.


Subject(s)
Gene Expression Regulation , Heme/chemistry , Malaria/metabolism , STAT3 Transcription Factor/metabolism , Animals , Brain/pathology , Chemokine CXCL10/metabolism , Chemokines/metabolism , Female , Heme Oxygenase-1/metabolism , Humans , Kidney/pathology , Lung/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Biological , Plasmodium berghei/metabolism , Receptors, CXCR3/metabolism , Signal Transduction
6.
Cells Tissues Organs ; 171(2-3): 188-98, 2002.
Article in English | MEDLINE | ID: mdl-12097841

ABSTRACT

Anabolic agents are useful tools for probing the mechanisms by which muscle fibers perceive and respond to disuse. beta(2)-Adrenergic agonists exert protective, and/or reparative, effects on atrophying muscle tissue. The effects of one such agent, clenbuterol (Cb), were examined on muscle mass, total protein content, and myofibrillar protein content in selected hindlimb muscles [adductor longus (ADL), extensor digitorum longus (EDL), plantaris (PLAN), soleus (SOL)] of mature male rats, under different loading conditions. Pair-fed rats were divided into four experimental groups: vehicle- and Cb-treated nonsuspended, vehicle- and Cb-treated hindlimb suspended (HLS). Experiments lasted 14 days, during which the rats received subcutaneous injections of 1 mg/kg Cb or 1 ml/kg vehicle. HLS induced significant atrophy in all muscles, except the EDL, in a generally fiber type-related pattern. However, myofibrillar protein content was affected in a more regional pattern. Cb treatment of nonsuspended rats induced hypertrophy in all muscles, in a generally uniform pattern. However, myofibrillar protein content was affected in a more fiber type-related pattern. Cb treatment of HLS rats reduced or eliminated HLS-induced atrophy in all muscles, in a muscle-specific pattern. Overall, the ADL and SOL were most susceptible to HLS-induced atrophy. The PLAN had the greatest magnitude of Cb-induced sparing of atrophy. The results show that, in mature male rats, Cb exerts anabolic effects that are load-dependent and muscle-specific. Responses to this drug cannot be reliably predicted by fiber-type composition alone.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Clenbuterol/pharmacology , Hindlimb Suspension , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Animals , Humans , Male , Muscular Atrophy/physiopathology , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...