813 research outputs found

    A Novel Role for Osteopontin in Facilitating West Nile Virus Neuroinvasion

    Get PDF
    West Nile virus (WNV) is a positive-sensed, single-stranded RNA flavivirus that can cause human neuroinvasive diseases, including encephalitis, meningitis, and flaccid paralysis. The mechanisms by which WNV enters the central nervous system and the host-factors that are involved in WNV-neuroinvasiveness are not completely understood. Osteopontin (OPN), a multifunctional glycoprotein, has been implicated as a bio-marker for a number of neuroinflammatory diseases. In particular, secreted (s)OPN has been implicated to participate in recruitment of polymorphonuclear neutrophils (PMN) to sites of its expression, while PMNs have been suggested to act as WNV reservoirs. Therefore, sOPN recruitment of PMNs may contribute to neuroinvasive WNV infection via the ‘Trojan horse’ mechanism of viral entry into the brain. Therefore, we hypothesize brain-infiltration of PMNs during neuroinvasive WNV pathogenesis is in part mediated by sOPN. Our results show that sOPN expression was significantly increased in human sera, human neuronal cells line, murine plasma, brain homogenates and primary neuronal supernatant following WNV infection, indicating a role for OPN in WNV pathogenesis. In addition, after challenge with WNV in vivo, Opn-/- mice exhibited a higher (70%) survival rate than wild-type (WT) mice (30%). Consistent with this, qPCR analysis between WT and Opn-/- mice demonstrated comparable levels of viremia; yet, reduced viral burden in the brains of Opn-/-mice compared to WT controls. Analysis of brain- infiltrating leukocytes displayed reduced PMNs and PMN-chemokine expression levels in Opn-/- mice brains. Importantly, intracerebral supplement of recombinant OPN (rOPN) into Opn-/-mice resulted in increased PMN-brain infiltration, increased viral load and reduced overall survival. Together, these data suggest OPN facilitates WNV neuroinvasion in a mouse model

    Heat Shock Protein 40 and Immune Function in Altered Gravity

    Get PDF
    In space, astronauts are more susceptible to pathogens, viral reactivation and immunosuppression, which poses limits to their health and the mission. Interestingly, during space flight, stress-inducible heat shock proteins (HSP) are robustly induced, and the overexpression of HSPs have been implicated in immune dysregulation, therefore HSPs may be critically involved in regulating immune homeostasis. HSP40/DNAJ1 plays a major role in proper protein translation and folding. Its loss of function has been implicated in susceptibility to microbial infection, while its overexpression has been implicated in autoimmunity, collectively suggesting its complicated, but necessary, role in maintaining immunological function. To determine the role of HSP40 during stress-induced altered gravity conditions, wild-type and Hsp40 mutant Drosophila melanogaster were exposed to ground-based chronic hypergravity conditions, followed by quantitative PCR (qPCR) analysis of immune gene expression. In addition, larval hemocytes were collected to determine the functional output in response to E. coli bioparticle phagocytosis. Preliminary data indicates a required role for Hsp40 in strengthening immune function during stress-induced spaceflight in flies. In short, a critical need to evaluate the relationship between HSPs and immune suppression during space flight is necessary. Since space travel may become available to the general public in the not too distant future, and for the possibility of long-term space missions, a more comprehensive evaluation of the molecules responsible for immune dysfunction observed during space flight is required

    Gravity as a Continuum: Effects of Altered Gravity on Drosophila Melanogaster Immunity

    Get PDF
    The impact of spaceflight on immune function is undoubtedly a critical focus in the area of space biology and human health research. Heat shock proteins (Hsp) are an evolutionarily conserved family of proteins that are expressed in response to cellular and physiological stressors, experienced during radiation exposure, confinement, circadian rhythm disruption, and altered gravity (hypergravity experienced at launch/landing and microgravity experienced in-flight). In particular, Hsp70 aids in the folding of proteins, facilitates the movement of proteins across the membranes during signal transductions and can stimulate innate immunity. Since Hsp70 is induced during cellular stress, and can act as a stimulator for innate immunity, we sought to address how a loss of Hsp70 affects immunity, under the stress-inducing model of acute and chronic hypergravity. Moreover, the effects of gravity as a continuum on the induction of Hsps and key immune genes were also assessed to determine if increased cellular stress, via increased gravity (g)-force, contributes to immune dysfunctions. For this, wildtype (W1118) and Hsp70 deficient (Hsp70null) Drosophila melanogaster were subjected to simulated hypergravity at increasing levels of g-force (1.2g, 3g, and 5g) for acute (1hr) and chronic (7-day) timepoints and were compared to 0g \u27non-hypergravity\u27 controls. Following simulation, whole bodies were sex-segregated, RNA was isolated and quantitative (q)PCR was performed to determine differential immune gene expression profiles. Further, functional output of hemocytes were assessed by a phagocytosis assay. Collectively, these studies evaluated the effects of Hsp70 in the context of immunity during acute and chronic hypergravity. Indeed, relevance for this work can directly translate to acute effects of launch/landing gravitational forces upon liftoff (~1.7g) and entry (~3.4g) that astronauts experience. In addition, the effects of chronic cellular stress is directly relevant to the immune health of astronauts on long duration missions, as well. Thus, as we approach the goal of returning to the Moon and landing the first humans on Mars, an evaluation of gravity as a continuum and the stress-inducing effects of altered gravity experienced during spaceflight on astronaut immunity and health are necessary

    Persistent stress during pregnancy influences thymic development in murine offspring

    Get PDF
    Persistent stress during pregnancy influences thymic development in murine offspring Alex Schroeder1*, Cassandra M. Juran1,2, Carol Mitchell1, and Amber M. Paul1,2 1Embry-Riddle Aeronautical University, Department of Human Factors and Behavioral Neurobiology, Daytona Beach, FL 2Blue Marble Space Institute of Science, NASA Ames Research Center, Moffett Field, CA *Presenting Author Thriving in spaceflight presents a unique challenge for humans. Exposure to extreme socioenvironmental stressors, such as altered gravity, ionizing radiation, and social isolation all can affect human biology. It is understood that physiological and psychological stress can disrupt gestation and reproduction processes in humans, as well. Using the mouse model, Chronic Unpredictable Mild Stress (CUMS), we can assess the influence of socioenvironmental stress on various biological systems, including the relationship between immune and reproductive systems. The thymus is an important gland involved in early life immune development and is sensitive to external factors that can disrupt T cell receptor diversity and antigen specificity. Due to this, we assessed retrospective, open-sourced data from GeneLab Open Science Directory (OSD-287). These data consist of thymic transcriptomes of one-day old pups born to dams that were exposed to CUMS for three-weeks. We aim to identify thymic immune pathways and activities that are involved in T cell development. Therefore, we hypothesize that CUMS exposure in dams will impair antigen presentation pathways and T cell tolerance processes in the thymus of pups. In brief, this project will identify processes that engage T cell activity in pup thymic development and examine the consequences of socioenvironmental stress in gestation

    Calnexin Is Necessary for T Cell Transmigration into the Central Nervous System

    Get PDF
    In multiple sclerosis (MS), a demyelinating inflammatory disease of the CNS, and its animal model (experimental autoimmune encephalomyelitis; EAE), circulating immune cells gain access to the CNS across the blood-brain barrier to cause inflammation, myelin destruction, and neuronal damage. Here, we discovered that calnexin, an ER chaperone, is highly abundant in human brain endothelial cells of MS patients. Conversely, mice lacking calnexin exhibited resistance to EAE induction, no evidence of immune cell infiltration into the CNS, and no induction of inflammation markers within the CNS. Furthermore, calnexin deficiency in mice did not alter the development or function of the immune system. Instead, the loss of calnexin led to a defect in brain endothelial cell function that resulted in reduced T cell trafficking across the blood-brain barrier. These findings identify calnexin in brain endothelial cells as a potentially novel target for developing strategies aimed at managing or preventing the pathogenic cascade that drives neuroinflammation and destruction of the myelin sheath in MS

    Interleukin-17A Promotes CD8 T Cell Cytotoxicity to Facilitate West Nile Virus Clearance

    Get PDF
    CD8 T cells are crucial components of immunity and play a vital role in recovery from West Nile virus (WNV) infection. Here, we identify a previously unrecognized function of interleukin-17A (IL-17A) in inducing cytotoxic-mediator gene expression and promoting CD8 T cell cytotoxicity against WNV infection in mice. We find that IL-17A-deficient (Il17a/) mice are more susceptible to WNV infection and develop a higher viral burden than wild-type (WT) mice. Interestingly, the CD8 T cells isolated from Il17a/ mice are less cytotoxic and express lower levels of cytotoxic-mediator genes, which can be restored by supplying recombinant IL-17A in vitro and in vivo. Importantly, treatment of WNV-infected mice with recombinant IL17A, as late as day 6 post infection, significantly reduces the viral burden and increases survival, suggesting a therapeutic potential for IL-17A. In conclusion, we report a novel function of IL-17A in promoting CD8 T cell cytotoxicity, which may have broad implications in other microbial infections and cancers

    Placenta-Expanded Stromal Cell Therapy in a Rodent Model of Simulated Weightlessness

    Get PDF
    Long duration spaceflight poses potential health risks to astronauts during flight and re-adaptation after return to Earth. There is an emerging need for NASA to provide successful and reliable therapeutics for long duration missions when capability for medical intervention will be limited. Clinically relevant, human placenta-derived therapeutic stromal cells (PLX-PAD) are a promising therapeutic alternative. We found that treatment of adult female mice with PLX-PAD near the onset of simulated weightlessness by hindlimb unloading (HU, 30 d) was well-tolerated and partially mitigated decrements caused by HU. Specifically, PLX-PAD treatment rescued HU-induced thymic atrophy, and mitigated HU-induced changes in percentages of circulating neutrophils, but did not rescue changes in the percentages of lymphocytes, monocytes, natural killer (NK) cells, T-cells and splenic atrophy. Further, PLX-PAD partially mitigated HU effects on the expression of select cytokines in the hippocampus. In contrast, PLX-PAD failed to protect bone and muscle from HU-induced effects, suggesting that the mechanisms which regulate the structure of these mechanosensitive tissues in response to disuse are discrete from those that regulate the immune- and central nervous system (CNS). These findings support the therapeutic potential of placenta-derived stromal cells for select physiological deficits during simulated spaceflight. Multiple countermeasures are likely needed for comprehensive protection from the deleterious effects of prolonged spaceflight

    Loss of Glycosaminoglycan Receptor Binding After Mosquito Cell Passage Reduces Chikungunya Virus Infectivity

    Get PDF
    Chikungunya virus (CHIKV) is a mosquito-transmitted alphavirus that can cause fever and chronic arthritis in humans. CHIKV that is generated in mosquito or mammalian cells differs in glycosylation patterns of viral proteins, which may affect its replication and virulence. Herein, we compare replication, pathogenicity, and receptor binding of CHIKV generated in Vero cells (mammal) or C6/36 cells (mosquito) through a single passage. We demonstrate that mosquito cell-derived CHIKV (CHIKVmos) has slower replication than mammalian cell-derived CHIKV (CHIKVvero), when tested in both human and murine cell lines. Consistent with this, CHIKVmos infection in both cell lines produce less cytopathic effects and reduced antiviral responses. In addition, infection in mice show that CHIKVmos produces a lower level of viremia and less severe footpad swelling when compared with CHIKVvero. Interestingly, CHIKVmos has impaired ability to bind to glycosaminoglycan (GAG) receptors on mammalian cells. However, sequencing analysis shows that this impairment is not due to a mutation in the CHIKV E2 gene, which encodes for the viral receptor binding protein. Moreover, CHIKVmos progenies can regain GAG receptor binding capability and can replicate similarly to CHIKVvero after a single passage in mammalian cells. Furthermore, CHIKVvero and CHIKVmos no longer differ in replication when N-glycosylation of viral proteins was inhibited by growing these viruses in the presence of tunicamycin. Collectively, these results suggest that N-glycosylation of viral proteins within mosquito cells can result in loss of GAG receptor binding capability of CHIKV and reduction of its infectivity in mammalian cells

    Artificial Gravity Partially Protects Space-Induced Neurological Deficits in Drosophila Melanogaster

    Get PDF
    Spaceflight poses risks to the central nervous system (CNS), and understanding neurological responses is important for future missions. We report CNS changes in Drosophila aboard the International Space Station in response to spaceflight microgravity (SFmg) and artificially simulated Earth gravity (SF1g) via inflight centrifugation as a countermeasure. While inflight behavioral analyses of SFmg exhibit increased activity, postflight analysis displays significant climbing defects, highlighting the sensitivity of behavior to altered gravity. Multiomics analysis shows alterations in metabolic, oxidative stress and synaptic transmission pathways in both SFmg and SF1g; however, neurological changes immediately postflight, including neuronal loss, glial cell count alterations, oxidative damage, and apoptosis, are seen only in SFmg. Additionally, progressive neuronal loss and a glial phenotype in SF1g and SFmg brains, with pronounced phenotypes in SFmg, are seen upon acclimation to Earth conditions. Overall, our results indicate that artificial gravity partially protects the CNS from the adverse effects of spaceflight

    Beyond Low-Earth Orbit: Characterizing Immune and microRNA Differentials following Simulated Deep Spaceflight Conditions in Mice

    Get PDF
    Spaceflight missions can cause immune system dysfunction in astronauts with little understanding of immune outcomes in deep space. This study assessed immune responses in mice following ground-based, simulated deep spaceflight conditions, compared with data from astronauts on International Space Station missions. For ground studies, we simulated microgravity using the hindlimb unloaded mouse model alone or in combination with acute simulated galactic cosmic rays or solar particle events irradiation. Immune profiling results revealed unique immune diversity following each experimental condition, suggesting each stressor results in distinct circulating immune responses, with clear consequences for deep spaceflight. Circulating plasma microRNA sequence analysis revealed involvement in immune system dysregulation. Furthermore, a large astronaut cohort showed elevated inflammation during low-Earth orbit missions, thereby supporting our simulated ground experiments in mice. Herein, circulating immune biomarkers are defined by distinct deep space irradiation types coupled to simulated microgravity and could be targets for future space health initiatives
    • …
    corecore