Scientific Program

Conference Series Ltd invites all the participants across the globe to attend 2nd International Congress on Neuroimmunology and Therapeutics Atlanta, Georgia, USA.

Day 2 :

  • Clinical Neuroimmunology
    Psychoneuroimmunology
    Applied Neurobiology
    Neuropharmacology
    Antibody-mediated disorders
Location: ANDIAMO
Speaker

Chair

Denis Gris

University of Sherbrooke , Canada.

Biography:

Dianne Lorton has completed her PhD in Neurosciences from Indiana State University in affiliation with Indiana University School of Medicine. She has completed her Post-doctoral training in Pharmacology from Duke University and in Neuroimmunology from the University of Rochester. She is currently an Assistant Professor at Kent State University in the College of Arts and Sciences. She has published over 70 papers (manuscripts, reviews, and book chapters) on neuroimmunology focusing on sympathetic nervous system regulation of immunity

Abstract:

Statement of the Problem: In 80% of patients, major life stressors precede onset of autoimmune diseases, including rheumatoid arthritis (RA) linking stress pathway activation to disease onset. We examined the contribution of high sympathetic nervous system activity to RA onset using the adjuvant-induced (AA) arthritis model in Lewis rats.

 

Methodology: Rats were immunized with complete Freund’s adjuvant to induce AA. From day (D) 12 (disease onset) through D28, rats were treated with vehicle or 2 mg/kg/day moxonidine, an imidazoline receptor-1 agonist that acts centrally to reduce SNS tone. Disease outcome was assessed using dorsoplantar widths and X-ray analysis. Cytokines critical for inflammation and CD4+ Th cell development (interleukin (IL)-1beta, IL-10, tumor necrosis factor (TNF)-alpha, IL-6, IL-2, IL-4, IFN-gamma, and tumor growth factor (TGF)-beta) were assessed in spleen, draining lymph node (DLN) and peripheral blood mononuclear cells (PBMCs) by enzyme-linked immunoassays. 

Speaker
Biography:

Born 1929 in Shanghai in an intellectual family. My father held the idea of “Save our country with medicine”, which determined my live as a student of medical school and a scientist in medical field.

 

In 1953 I was recruited by People’s Liberation Army to be a teacher in Dept. of Anatomy of the Fourth Military Medical University, a school just established and located in Xi’an, a very back-woods city. The university had little, if any, research facilities. In the Dept. of Anatomy there was only one old Zeiss bronze monocular microscope, probably a little better than the one Cajal had used. I was the only one in Dept. of Anatomy who knew Neuroanatomy. Hodology is an important research field in Neuroanatomy , the best tracing technique was Nauta’s staining invented in mid-50’s. I am the first one in China who mastered this technique and published a paper in early 90’s in Journal of Anatomy (Chinese), then the best journal in China available. Then came a national turmoil of so called “Cultural Revolution”which revolved all cultures away. We were forbidden to do scientific research, even not allowed to go to libraries. The “Cultural Revolution” was finally proved a conspiracy and was crushed down in 1986. It spoiled 20 of by best years.

Abstract:

Throughout my life I have been focused my research on three fields:

1. Direct neural regulation of mammalian anterior pituitary. A student of mine accidentally found many nerve fibers in the anterior pituitary of a monkey”. Since beside the gland cells there are abundant blood vessels in the anterior pituitary, therefore, the first thing is to clarified what do the nerve fibers innervate in the gland. It turned to be that the nerve fibers innervate both of them, but mainly the gland cell. A series of studies were then followed. To make the story short, the golden criterion was that our electron microscopic study found typical synapses between the terminals of the nerve fibers and the gland cells. We then put forth a hypothesis: Neuro-humoral dual regulation of mammalian anterior pituitary.

2. Brain control of immunity

3. Early neural surgery of spinal cord contusion, a surgery I myself designed. The operations were followed by rehabilitation of three months. The outcome was beyond imagination. In 30 ASIA grade-A patients, the optimal operation time window was 4-14 days. There were13 cases in the window, 85% were able to walk with a pair of crunchers, or a stick, or without any support.

4. A small but very popular staining technique. The glucose oxidase-DAB-nickel method in peroxidase histochemistriry of the nervous system. 1988. It has been cited more than 1,000 times and is still popular today.

Zhihai lei

Nanjing Agricultural University, China

Title: A role of Neuromedin U in Pig Immune Regulation
Biography:

Zhihai Lei is a Professor working at Nanjing Agricultural University (NAU), Nanjing, Jiangsu, China. His research fields are nueroanatomy and neuroendocrine of domestic animals and pay more attention to roles of newly discovered neuropeptides in reproduction and immune regulations. 

Abstract:

Statement of the Problem: Neuromedin U (NMU) is a conserved mammalian neuropeptide discovered in the 1980s and found in two forms, NMU-25 and NMU-8. Wide distribution of NMU in animal organs suggests that NMU is involved in multiple physiological functions, including immune regulation. However, the role of NMU in pig immune regulation has not been reported.

 

Methodology & Theoretical Orientation: To study the effect of NMU on pig immune regulation, we cloned and detected the expression of NMU and its receptors in pig lymphatic organs and immune cells. We also investigated the effect of NMU on cytokine secretion after injection of (0, 5, 15, 45 nmol) NMU into the intracerebral ventricle (i.c.v) of 16 pigs (n=4 for each group) and the effect of (0.1~1000 nM) NMU on cytokine secretion in cultured dendritic cells and natural kill (NK) cells using ELISA and RIA methods.

 

Findings: NMU and its receptors were expressed in lymphatic organs, cultured dendritic cells and NK cells. NMU stimulated IL-1β, IL-6, IL-8, TNF-α and IL-10 secretion (P<0.05) post-injection in a time- and dose-dependent manner, compared with the control group. NMU increased IL-8, IL-6 and IL-13 secretion and reduced IL-10 secretion (P<0.05) in cultured dendritic cells. NMU enhanced the killing activity of cultured NK cells, stimulated IFN-γ secretion via PLC, PI3K and MEK signal pathways and inhibited IL-10 secretion (P<0.05) in NK cells in atime- and dose-dependent manner.

 

Conclusion & Significance: This study suggests that NMU has the role in pig immune regulation through its effect on cytokine secretion and increasing killing activity of NK cells.

Speaker
Biography:

Grace Y Sun has completed her BS in Chemistry from Seattle Pacific University and PhD in Biochemistry from Oregon State University. After holding a Research Scientist position in Cleveland, she was recruited to the Biochemistry Department in University of Missouri. As a Neuroscientist, she is recognized for research on brain lipids and signaling pathways related to neuro-inflammation and aging. Her research experience included serving as Program Project Director of the MU Alzheimer’s disease research program and as a Project Leader in the MU Center for Phytonutrient and Phytochemistry. Besides training a large number of graduate students and post doctorates, she is also experienced in organizing national and international symposia and meetings. She has published over 300 articles in highly regarded journals. Among the many awards, the most notable one is the MU System President’s Award for Sustained Excellence in 2012, which was the highest award recognizing faculty achievement at the university.

Abstract:

Microglia is the major immune active cell in the brain, are known to play multiple functional roles for maintaining brain health. Besides ability to migrate to sites of injury and scavenge cellular debris, these cells also become activated in response to stimuli, including cytokines, chemokines and toxins. Upon activation, these cells release pro-inflammatory and neurotrophic factors, this can alter the environment of neighboring cells. Microglia activation has been implicated in the acute and chronic inflammation in a number of neurological diseases, including Alzheimer’s disease, stroke and traumatic brain injury. Some noted pro-inflammatory events include activation of cytosolic phospholipase A2 (cPLA2) and stimulation of the NF-kB transcriptional pathway. In our studies, we have used the bacteria endotoxins (lipopolysaccharides, LPS) to stimulate the Toll-like receptors in microglia and activate both cPLA2 and the NF-kB pathway, leading to the release of prostaglandins, pro-inflammatory cytokines, reactive oxygen species (ROS), and nitric oxide (NO). A number of botanical polyphenols have been shown to inhibit the oxidative/inflammatory responses elicited by LPS. Interestingly, some phytochemicals not only can mitigate the NF-kB responses but also stimulate the anti-oxidant pathway involving the Nrf2 and activation of the Antioxidant Response Element (ARE). The Nrf2/ARE transcription pathway is known to play an important role in the synthesis of a large number of antioxidant enzymes including heme oxygenase-1 (HO-1). Although more studies are needed, there is evidence that activation of MAPKs may play an important role in mediating the cross-talk between the NF-kB and Nrf2 pathways. Future studies to uncover regulation of these pathways in microglia will be important in the development of novel therapeutic strategies to fight against neurological diseases and aging.

Speaker
Biography:

D Gris is a Head of Neuroimmunology Laboratory at the University of Sherbrooke, Canada. He has graduated from Dr. Weaver’s Laboratory in University of Western Ontario where he studied inflammation after spinal cord injury. He has completed his Post-doctoral studies from the University of North Carolina at Chapel Hills, NC, USA where he began to investigate role of NLRs in neurodegeneration. He is investigating novel computerized methods of evaluating behavioral outcomes of neuro-inflammation. His main interest is to discover novel anti-inflammatory pathways within the central nervous system and use this knowledge to design therapies for neurological diseases including multiple sclerosis amyotrophic lateral sclerosis autism and epilepsy.

Abstract:

Many members of NLR family of proteins play an important role in human diseases including diabetes, Crohn’s disease, cancer, etc. NLRs bind multiple proteins inside cells, thus redirecting molecular signaling. We concentrated on the role of anti-inflammatory NLRs in multiple sclerosis. Using state-of the art automated behavioral platform we demonstrate that NLRX1 and NLRP12 inhibit progression of the diseases in a mouse model of MS. We observed reduced inflammation and improve biochemical and behavioral outcomes of the disease. Furthermore, NLRX1 acts at the level of mitochondria promoting DRP1 dependent mitochondrial fission. In inflammatory cells such as microglia and astrocytes, this results in inhibition of assembly of proinflammatory pathways including type I interferon and NFkB. Accordingly; we observed reduction in the expression of iNOS, cytokines including IL-1beta and TNF-alpha during microglial activation. In neurons, NLRX1 effect results in inhibition of necrosis and increased viability. Using N2A cell line, we demonstrated that NLRX1 protects cells from rotenone toxicity. We demonstrated that NLRX1 over-expressing cells were more viable and the ration of apoptosis to necrosis was shifted to necrosis in cells that lacked NLRX1. We confirmed profound role of NLRs by generating mice that spontaneously develop multiple sclerosis-like disease. In conclusion, both NLRX1 and NLP12 decrease inflammatory responses in the CNS and, therefore, present as a target for treatments in neurodegenerative diseases.

Biography:

Biswadev Bishayi has completed his BSc in Physiology in 1990 from Midnapore College and MSc in Human Physiology from Vidyasagar University West Bengal, India in 1992. After qualifying NET-CSIR examination he joined Indian Institute of Chemical Biology as JRF and completed his PhD from Jadavpore University, West Bengal, India in 1999. He has joined as Lecturer in the Department of Physiology, University of Calcutta in 1997 and promoted to Professor in 2012. After being awarded the Biotechnology Overseas Associateship from the DBT he did his Postdoctoral research in Boston University Medical School, USA. The main focus of his research is host-pathogen interaction, role of cytokines in inflammatory diseases as well as neuro-endocrine immune interactions in relation to Staphylococcus aureus infection. He has regularly published papers in national and international reputed journals.

Abstract:

A reciprocal regulation exists between the central nervous and immune systems where the CNS signals the immune system via hormonal and neuronal pathways and the immune system signals the CNS through cytokines. The hypothalamic pituitary adrenal (HPA) axis regulates the immune system via glucocorticoid hormones (GC). Neuroendocrine regulation of immune function is essential for survival during stress or infection and to modulate inflammatory disease. Glucocorticoids have multiple effects on immune cells but insensitivity of immune cells to this hormone during chronic stress and persistent inflammation has been the cause of several neuroimmunological disorders. In a mouse model of restraint stress and infection with Gram negative (E. coli) or Gram positive (S. aureus) or injection of bacterial endotoxin (LPS) several inflammatory parameters and behavioral changes were studied. Increased resistance of mice to infections that correlated with their behavioral changes in an open field test (OFT) and on an elevated plus maze (EPM) has been observed. Reduced acetyl cholinesterase (AChE) activity in the hypothalamus indirectly suggested that more acetylcholine molecules must be playing their anti-inflammatory role to modulate the activity of the HPA axis. Exogenous administration of pro-inflammatory cytokine (IL-6) or an anti-inflammatory cytokine (IL-10) along with ciprofloxacin (an antibiotic having secondary anti-inflammatory role apart from its primary anti-bacterial effect) to stressed and infected mice, down regulated the inflammatory mediators and also effectively helped in bacterial killing. Thus this work provides new insight into the therapeutic strategy that can be adopted when considering treatment against chronic stress and infection induced neuroimmunological disorders.

Speaker
Biography:

Monique F Stins has her expertise in Blood Brain Barrier research and is seeking to dissect underlying molecular relations between Blood brain barrier activation and neurological damage. This may lead to identification of pharmacologic targets for therapeutic intervention to improve neurological outcomes of infections with neurotropic microbes. She is using various models of the blood brain barrier in microbial disease to target these pathogenic mechanisms. She has experience teaching students at levels ranging from middle and high school to undergraduate college and upper level college courses.

Abstract:

Cerebral malaria (CM) is a serious complication of Plasmodium falciparum infection, especially in young children, but also in non-immune travelers and military personnel visiting or sent out to malaria endemic regions. Clinically, CM features symptoms such as seizures and reversible coma. Without proper supportive care often leads to death, despite anti-malarial chemotherapy.  Upon clearance of the infection, patients are often left with neurologic sequelae, such as seizures, learning and behavioral disorders such as ADHD in African children. Post malaria syndromes can also include psychotic or acute confusional episodes and tremors. Sequestration of Plasmodium infected erythrocytes (PRBC) in high endothelial venules is a hallmark of CM. It is unclear as to how these intracellular parasites, while confined to the lumen of the brains vasculature induce neurological dysfunction.  In CM, the blood brain barrier (BBB) lies at the interface of the events occurring in blood and brain. The BBB is part of the neurovascular unit (NVU), a concept that emphasizes homeostatic interactions between its components to ensure optimal functioning of the central nervous system. It is hypothesized that activation of the BBB endothelium disturbs the homeostasis between the astroglial and neuronal components of the NVU leading to neurological dysfunction. Using in vitro models for the human BBB, PRBC increase ICAM-1 expression and decrease the barrier function of monolayers in a dose dependent manner. Microarray and gene ontology (GO) analysis indicated a predominance of the NFB mediated pro-inflammatory responses among the host signaling pathways. RT-PCR and protein analysis confirmed the increase in transcripts and directional release of both cytokines and chemokines in various in vitro models. Basal directed BBB secretions caused dose-dependent abnormal astroneuronal morphology and cell death. Determination of the underlying pathogenesis of observed BBB activation and astroneuronal effects may lead to development of adjunctive neurotherapeutics to ameliorate neurologic sequelae.

Fiaz Mohammed

College of Medicine and Health Sciences, Ethiopia

Title: Neuropharmacology of Aggressive Behaviour
Biography:

Abstract:

Background: Aggression has been recognized as a near-universal behavior with substantial influence on and implications for human and animal social systems. It can be a significant problem since escalated aggression may result in serious damage to others and can reveal itself as a symptom of psycho- pathologies. It is often considered to be the product of environmental stressors, health problems, pain, psychiatric morbidity or behavioral stressors. Psychopharmacology is a one of field of medicine that addresses the use of medications to correct or alleviate common mental illnesses. It is a scientific discipline that utilizes drugs to increase our knowledge and understanding of the workings of the mind.

 

Objective: The objective of this study is to review and discuss the advancements of neural mechanisms and neuropharmacological agents related to aggression.

 

Methods: Systematic review method was used for this study.

 

Results & Discussion: An imbalance in neurochemicals is thought to be the primary reason for psychological problems and seizure activity. The understanding of each neurotransmitter's functions and responsibilities explains why a decrease or increase in normal levels would cause elevated aggression. Psychopharmacology emphasizes drug induced changes in mood, thinking and behavior. Among its aim is the utilization of medicinal substances for the restoration and maintenance of mental health, the prevention of mental breakdowns, and the exploration of the reaches of the mind. Its basic purpose is to help disease, but to change the personality, improve human nature, or cure bad habits. Developing novel therapies to reduce or prevent violent outbursts is a key objective of psychopharmacological research on aggression.

 

Conclusion & Recommendation: Despite considerable progress, therapeutic possibilities for controlling or coping with, pathological aggressive behavior remain far from being satisfactory. Consequently, studying both the fundamental causes of aggression and dysfunctions and the modification of behavior by pharmacologic interventions is surmountable.

 

Biography:

Luis Venegas S has completed his under graduation from Universidad de Antofagasta and has completed his PhD from Universidad Andres Bello in 2016. His Doctoral thesis was on the thyroid hormones during the gestation in the development and function of blood brain barrier. Currently, he is pursuing his Post-doctoral studies from Pontificia Universidad Catolica de Chile.

Abstract:

Hypothyroidism is a thyroid hormone deficiency condition. Around the world, 7% of pregnant women suffer maternal hypothyroidism. It causes low intellectual quotient (IQ), and mental retardation in the offspring. Gestational hypothyroidism increases the intensity of experimental autoimmune encephalomyelitis (EAE), an experimental model for multiple sclerosis (MS). Given that the immune system is important for learning, we propose the following hypothesis: “The offspring gestated in hypothyroidism will affect the pattern of inflammatory cytokines in the serum that will alter learning, the BBB permeability and the migration of the immune system to the CNS”. To evaluate this maternal hypothyroidism was induced to pregnant mice. The learning capacity was analyzed and the serum levels of cytokines were analyzed by ELISA and multiplex in the offspring. The permeability of BBB was analyzed by Evans Blue (EB) extravasation in the CNS and the integrity of BBB was analyzed by immunofluorescence. The migration of immune cells to the CNS was analyzed by FACS. We found that the offspring gestated in hypothyroidism has impaired learning and low levels of IL-4, high basal levels of TNFα and IL-17, high EB extravasation and a higher number of CD4+ T cells in the CNS with an altered expression of BBB proteins. In conclusion, our data support that maternal hypothyroidism have altered cognition, BBB permeability and the presence of the immune system in the CNS. Together with an altered pattern of cytokines in the serum that are important for learning and for altering the BBB.

Yeon Sun Lee

University of Arizona,USA

Title: Neuroexcitatory effects of dynorphin A
Speaker
Biography:

 

Yeon Sun Lee is an Expert in peptide drug discovery area like novel peptide and peptidomimetic ligands for the treatment of pain and neuronal disease. She has been conducting research in identifying key structural features for target receptors including opioid receptors. The goal of her research is to develop new class of ligands modulating serious side effects that are caused by long term administration of opioids to treat chronic pain states, while retaining their high efficacies. Her studies represent a new approach: Drug design for pathological conditions and multifunctional ligands.

Abstract:

Statement of the Problem: Dynorphin A is an endogenous ligand showing neuro-inhibitory effects via opioid related mechanism. However, chronic pain, nerve injury, and inflammation often result in the up regulation of dynorphin A in the spinal column neurotransmitter pathway to the brain and cause neuroexcitatory effects such as motor impairments and hypersensitivity via non-opioid mechanism. The purpose of this research is to develop dynorphin A antagonists to modulate the adverse neuroexcitatory effects under pathological conditions.

 

Methodology & Theoretical Orientation: Systematic structure-activity relationship studies on the non-opioid dynorphin A fragment, dyn A-(2-13) were performed to identify the key structural features for the interaction with the bradykinin receptors, and then structures were refined by examining the effects of different substituents to obtain an antagonist activity. Lead ligands showing a high affinity at the bradykinin receptors were advanced to test their metabolic stability in plasma and anti-hyperalgesic effects in animals.

 

Findings: Our studies have discovered a novel mechanism of neurotransmission related to pain in which the bradykinin receptors are upregulated and dynorphin A peptides have neuroexcitatory effects that result in hyperalgesia. Our studies have also shown that lead ligand LYS1044 blocks dynorphin A-induced hyperalgesia and motor impairments in naïve animals and inhibits thermal hyperalgesia and mechanical hypersensitivity in a dose-dependent manner in nerve-injured animals. Importantly, the ability of LYS1044 is limited to the CNS and thus can avoid serious cardiovascular effects caused by blocking peripheral bradykinin receptors. However, the ligand showed low metabolic stability in plasma and thus, to improve the stability various modifications were performed. As the result, we could identify a highly stable ligand with retained high affinity at the bradykinin receptors.

 

Conclusion & Significance: This study presents a new class of ligands based on the unanticipated pathophysiological interaction between the endogenous opioid ligand dynorphin A and bradykinin receptors for the treatment of chronic pain without the toxicities associated with current treatments for these maladies (Figure 1).

Biography:

Dr. Oz is a scientist with expertise in inflammatory and infectious disease, immune-modulations, pathogenesis, innate and mucosal Immunity, cytokines, chemokines and receptors’ modifications, inflammatory and neuropathic pain and behavioral modifications, ROS, micronutrient,  gastrointestinal complications and drug discoveries.

Guest and lead Editor for Special Issues in journals: Nutrients (infectious and inflammatory Diseases); Antioxidants (Antioxidants, Microbiome and Gut Health), Canadian J Infectious Diseases and Medical Microbiology (Environmental Health – How Does It Relate to Parasites?); Gastroenterology Research and Practice (Gastrointestinal Inflammation and Repair: Role of Microbiome, Infection, and Nutrition), Mediators of Inflammation (Inflammatory, Infectious and Nutrition 

Abstract:

Statement of the Problem: Toxoplasma a ubiquitous organism is one of the most important sources of congenital infection and foodborne diseases. An estimated 1.5 billion people are globally infected frequently with unknown lifelong health complications. Organisms cause severe immune-inflammatory reaction in vital organs with the surge of chemokines and cytokines. Following acute phase, the organisms lodge in cyst forms predominantly in brain and muscles for the life pending to become reactivated by immunosuppression.  Organisms are transmitted mainly by consumption of contaminated animal products (meat, milk and dairy) with cysts, congenital and breast feed from actively infected mom, organ transplantation or by sexual transmission.  Cats are only definitive host which can release environmental resistance forms (oocysts) in feces which contaminate vegetations and water and orally taken to infect humans and animals.  Toxoplasmosis in congenital infection or immunodeficient individuals is manifested with growth retardation, encephalomyelitis, intracranial calcifications, hydrocephalus, neurological, mental illnesses, and seizures, retinochoroiditis, visual and auditory inflammatory disorders, cardiovascular abnormalities, gastroenteritis, myositis and pain.  Toxoplasma is a neurotropic organism which bypasses brain barrier to infect neurons and glial cells and to cause mild to severe behavioral modifications.  Recent studies reveal a mind alteration and sexual attraction in Toxoplasma infected subjects including schizophrenic behaviors in animals as well as humans.  As, fearless infected rodents seek cats are eaten up to keep the sexual life cycle continued. Toxoplasma impairs the limbic brain neurons responsible for instinct defensive behavior and judgment activity adjacent to limbic regions of sexual desire. Yet there is no safe and effective approved therapy for congenital and chronic infection or a vaccine available to prevent toxoplasmosis. Different aspects of maternal and fetal toxoplasmosis will be discussed including neuroinflammatory and behavioral alteration.  

Biography:

Xiaoming Hu has her expertise in neuroinflammatory responses after ischemic brain injury and neurodegeneration. Her core research interests are to explore the immune-related mechanisms and to identify novel immunotherapies for stroke. One of her research directions is to explore the effect of regulatory T cell on stroke and to elucidate the underlying mechanisms. She is among the first who demonstrated neuroprotective effects of this specialized T cell subpopulation in cerebral ischemia (Li et al., Ann Neurol 2013; 74:458; Li et al., Stroke 2013; 44: 3509; Li et al., Stroke 2014; 45: 857). Dr. Hu also investigates the mechanism for microglia/macrophage polarization and explores the exogenous signals that induce the phenotype shift. The identification of these critical signaling molecules brings hope to therapies that promote healthy microglia/macrophage responses even under pathological conditions. Dr. Hu has published more than 50 peer-reviewed manuscripts in this research area.

Abstract:

Delayed thrombolytic treatment with recombinant tissue plasminogen activator (tPA) may exacerbate blood-brain barrier (BBB) breakdown after ischemic stroke and lead to lethal hemorrhagic transformation (HT). This study assessed the protective effect of regulatory T cell (Treg) transfer on tPA-induced HT and investigated the underlying mechanisms of protection. We used murine suture and embolic middle cerebral artery occlusion models of stroke to investigate the therapeutic potential of adoptive Treg transfer against tPA-induced HT. The results showed that delayed administration of tPA (10mg/kg) reproducibly resulted in hemorrhage in the ischemic territory 1d after MCAO. Intravenous administration of Tregs (2×106/mouse) immediately after tPA treatment almost completely abolished this hemorrhage and improved sensorimotor deficits after stroke. Tregs dramatically reduced BBB disruption in tPA-infused stroke mice, as measured by Evans blue or fluorescent tracer (cadaverine) leakage and IgG extravasation. Consistent with these observations, Treg treatment mitigated tight junction damage in tPA-treated stroke mice, as revealed by Western immunoblotting and electron microscopy. Mechanistic studies demonstrated that Tregs almost completely abolished the tPA-induced elevation of matrix metalloproteinase 9 (MMP9) and CCL2 after stroke. Using MMP9 and CCL2 knockout mice, we discovered that both molecules partially contributed to the protective actions of Tregs. In an in vitro endothelial cell-based model of the blood-brain barrier, we confirmed that Tregs inhibited tPA-induced endothelial expression of CCL2 and preserved blood-brain barrier integrity after an ischemic challenge. Lentivirus-mediated CCL2 knockdown in endothelial cells completely abolished the blood-brain barrier protective effect of Tregs. Altogether, our data suggest that Treg adoptive transfer after thrombolytic treatment alleviates hemorrhage in stroke victims. Treg-afforded protection in the tPA-treated stroke model may be mediated by two inhibitory mechanisms involving CCL2 and MMP9. Thus, Treg adoptive transfer may be useful as a cell-based therapy to improve the efficacy and safety of thrombolytic treatment for ischemic stroke

Biography:

Dr. Bielekova received an M.D. degree in 1993 from Comenius University in Bratislava, Slovakia. After a medical internship at SUNY Downstate, Medical Center in Brooklyn and a neurology residency at Boston University, she did a 3 year postdoctoral research fellowship at the NIH/NINDS/Neuroimmunology Branch (NIB). She remained at NIB for additional 5 years as a staff physician, focusing on development of novel therapies for multiple sclerosis (MS). In 2005, she became associate professor of neurology with tenure and director of the Waddell Center for MS at University of Cincinnati. In 2008, she moved back to NINDS as an investigator. Her laboratory is studying mechanisms of immunoregulation and immune-mediated central nervous system (CNS) tissue injury in MS and other neuroimmunological diseases with a long-term goal of developing effective therapies. In addition, Dr. Bielekova is a principal investigator on several innovative protocols including adaptively-designed Phase I/II clinical trials.

 

Abstract:

Because of inadequate efficacy of immunomodulatory treatments, primary- (PPMS) and secondary-progressive multiple sclerosis (SPMS) were thought to have significantly less intrathecal inflammation than relapsing-remitting (RRMS) MS.  Using functional assays and innovative combinatorial cerebrospinal fluid (CSF) biomarkers, we demonstrated that both progressive MS subgroups have on average identical amount of intrathecal inflammation to RRMS. Instead, significantly greater level of compartmentalization of immune responses to central nervous system (CNS) tissue and greater terminal differentiation of intrathecal immune responses were characteristic of progressive MS. This makes progressive MS inflammation inaccessible to systemically-administered large molecules (e.g. monoclonal antibodies; mAb), while small molecules that may penetrate CNS are ineffective, because they predominantly target cells in proliferation cycle. Thus, in the placebo-controlled, Phase II clinical trial (RIVITALISE trial, clinicaltrials.gov identifier NCT01212094) we investigated whether intrathecal (IT) administration of mAb, such as rituximab can effectively inhibit intrathecal inflammation in SPMS. The trial was stopped for futility, after prospectively-acquired CSF biomarkers convincingly demonstrated that IT-administered rituximab decreased intrathecal inflammation only by approximately 10%. Mechanistic studies revealed following reasons for decreased efficacy of rituximab in the intrathecal, as compared to systemic compartments: 1. Due to active transport of antibodies from CSF to blood, achievable CSF concentrations of rituximab did not fully saturate CD20 on intrathecal B cells. 2. CSF lacks lytic complement, which results in decreased complement-dependent cytotoxicity (CDC). 3. The predominant cellular subtype of natural killer (NK) cells in CSF are CD56bright NK cells, which have low expression of Fc receptor and thus decreased levels of antibody-dependent cellular cytotoxicity (ADCC).

In conclusion, novel immunomodulatory agents with high CNS penetrance, not dependent on CDC or ADCC and effective on proliferation-quiescent immune cells will be necessary to successfully inhibit intrathecal inflammation in progressive MS.

 

Biography:

Interest in the medical use of cannabinoids has greatly increased in the last few years. Our previous studies have shown that inhibition of the minor endocannabinoid 2-AG hydrolytic enzyme alpha, beta-hydrolase domain 6 (ABHD6) significantly reduces neuroinflammation and exerts neuroprotection in animal models of traumatic brain injury and multiple sclerosis. Unlike inhibition of the principle 2-AG hydrolytic enzyme, monoacylglycerol lipase (MAGL), which leads to 2-AG overload and cannabinoid receptor desensitization, selective inhibition of ABHD6 is thought to provide therapeutic benefits without producing cannabimimetic side effects.  Accumulating evidence suggests that neuroinflammation is a major contributing factor to the pathogenesis of neuropathic pain. In this study we sought to determine the potential therapeutic effect of a selective ABHD6 inhibitor WWL70 in the management of neuropathic pain. In the murine model of neuropathic pain induced by chronic constriction (CCI) of the sciatic nerve, we found that WWL70 treatment significantly alleviated CCI-induced thermal hyperalgesia and mechanical allodynia in the ipsilateral paw at 3 and 7 days post-injury. Treatment with WWL70 also significantly attenuated the increased inflammatory response characterized by the downregulation of TNF-α, IL-6, IL-1β, MCP-1 in the ipsilateral sciatic nerve, DRG and spinal cord of CCI mice. Co-administration of cannabinoid receptor antagonists did not affect the effect of WWL70, suggesting that WWL70’s anti-nociceptive effect is independent on cannabinoid signaling pathways. Treatment with WWL70 reduced the expression of COX-2 and PGES2 and the production of PGE2 in the injured sciatic nerve. This result suggested that interference with the eicosanoid signaling pathway contributes, at least in part, to the therapeutic mechanisms of WWL70 on CCI induced neuropathic pain.  

Abstract: