Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where George T. Kannarkat is active.

Publication


Featured researches published by George T. Kannarkat.


Journal of Parkinson's disease | 2013

The role of innate and adaptive immunity in Parkinson's disease.

George T. Kannarkat; Jeremy M. Boss; Malú G. Tansey

In recent years, inflammation has become implicated as a major pathogenic factor in the onset and progression of Parkinsons disease. Understanding the precise role for inflammation in PD will likely lead to understanding of how sporadic disease arises. In vivo evidence for inflammation in PD includes microglial activation, increased expression of inflammatory genes in the periphery and in the central nervous system (CNS), infiltration of peripheral immune cells into the CNS, and altered composition and phenotype of peripheral immune cells. These findings are recapitulated in various animal models of PD and are reviewed herein. Furthermore, we examine the potential relevance of PD-linked genetic mutations to altered immune function and the extent to which environmental exposures that recapitulate these phenotypes, which may lead to sporadic PD through similar mechanisms. Given the implications of immune system involvement on disease progression, we conclude by reviewing the evidence supporting the potential efficacy of immunomodulatory therapies in PD prevention or treatment. There is a clear need for additional research to clarify the role of immunity and inflammation in this chronic, neurodegenerative disease.


npj Parkinson's disease | 2015

Common Genetic Variant Association with Altered HLA Expression, Synergy with Pyrethroid Exposure, and Risk for Parkinson's Disease: An Observational and Case-Control Study.

George T. Kannarkat; Da Cook; Jae Lee; Jianjun Chang; Jaegwon Chung; E Sandy; Kc Paul; Beate Ritz; J Bronstein; Stewart A. Factor; Jeremy M. Boss; MariadeLourdes Tansey

Background:The common noncoding single-nucleotide polymorphism (SNP) rs3129882 in HLA-DRA is associated with risk for idiopathic Parkinson’s disease (PD). The location of the SNP in the major histocompatibility complex class II (MHC-II) locus implicates regulation of antigen presentation as a potential mechanism by which immune responses link genetic susceptibility to environmental factors in conferring lifetime risk for PD.Aims:The aim of this study was to determine the effect of this SNP on the MHC-II locus and its synergy with pesticide exposure.Methods:For immunophenotyping, blood cells from 81 subjects were analyzed by quantitative reverse transcription-PCR and flow cytometry. A case–control study was performed on a separate cohort of 962 subjects to determine association of pesticide exposure and the SNP with risk of PD.Results:Homozygosity for G at this SNP was associated with heightened baseline expression and inducibility of MHC class II molecules in B cells and monocytes from peripheral blood of healthy controls and PD patients. In addition, exposure to a commonly used class of insecticide, pyrethroids, synergized with the risk conferred by this SNP (odds ratio=2.48, P=0.007), thereby identifying a novel gene–environment interaction that promotes risk for PD via alterations in immune responses.Conclusions:In sum, these novel findings suggest that the MHC-II locus may increase susceptibility to PD through presentation of pathogenic, immunodominant antigens and/or a shift toward a more pro-inflammatory CD4+ T-cell response in response to specific environmental exposures, such as pyrethroid exposure through genetic or epigenetic mechanisms that modulate MHC-II gene expression.


Human Molecular Genetics | 2015

The G2019S LRRK2 mutation increases myeloid cell chemotactic responses and enhances LRRK2 binding to actin-regulatory proteins

Mark S. Moehle; João Paulo Lima Daher; Travis D. Hull; Ravindra Boddu; Hisham Abdelmotilib; James A. Mobley; George T. Kannarkat; Malú G. Tansey; Andrew B. West

The Leucine rich repeat kinase 2 (LRRK2) gene is genetically and biochemically linked to several diseases that involve innate immunity. LRRK2 protein is highly expressed in phagocytic cells of the innate immune system, most notably in myeloid cells capable of mounting potent pro-inflammatory responses. Knockdown of LRRK2 protein in these cells reduces pro-inflammatory responses. However, the effect of LRRK2 pathogenic mutations that cause Parkinsons disease on myeloid cell function is not clear but could provide insight into LRRK2-linked disease. Here, we find that rats expressing G2019S LRRK2 have exaggerated pro-inflammatory responses and subsequent neurodegeneration after lipopolysaccharide injections in the substantia nigra, with a marked increase in the recruitment of CD68 myeloid cells to the site of injection. While G2019S LRRK2 expression did not affect immunological homeostasis, myeloid cells expressing G2019S LRRK2 show enhanced chemotaxis both in vitro in two-chamber assays and in vivo in response to thioglycollate injections in the peritoneum. The G2019S mutation enhanced the association between LRRK2 and actin-regulatory proteins that control chemotaxis. The interaction between G2019S LRRK2 and actin-regulatory proteins can be blocked by LRRK2 kinase inhibitors, although we did not find evidence that LRRK2 phosphorylated these interacting proteins. These results suggest that the primary mechanism of G2019S LRRK2 with respect to myeloid cell function in disease may be related to exaggerated chemotactic responses.


PLOS ONE | 2013

Critical Role of Regulator G-Protein Signaling 10 (RGS10) in Modulating Macrophage M1/M2 Activation

Jae-Kyung Lee; Jaegwon Chung; George T. Kannarkat; Malú G. Tansey

Regulator of G protein signaling 10 (RGS10), a GTPase accelerating protein (GAP) for G alpha subunits, is a negative regulator of NF-κB in microglia. Here, we investigated the role of RGS10 in macrophages, a closely related myeloid-derived cell type. Features of classical versus alternative activation were assessed in Rgs10-/- peritoneal and bone marrow-derived macrophages upon LPS or IL-4 treatments, respectively. Our results showed that Rgs10-/- macrophages produced higher levels of pro-inflammatory cytokines including TNF, IL-1β and IL-12p70 in response to LPS treatment and exerted higher cytotoxicity on dopaminergic MN9D neuroblastoma cells. We also found that Rgs10-/- macrophages displayed a blunted M2 phenotype upon IL-4 priming. Specifically, Rgs10-/- macrophages displayed lower YM1 and Fizz1 mRNA levels as measured by QPCR compared to wild type macrophages upon IL-4 treatment and this response was not attributable to differences in IL-4 receptor expression. Importantly, phagocytic activities of Rgs10-/- macrophages were blunted in response to IL-4 priming and/or LPS treatments. However, there was no difference in chemotaxis between Rgs10-/- and WT macrophages. Our data indicate that Rgs10-/- macrophages displayed dysregulated M1 responses along with blunted M2 alternative activation responses, suggesting that RGS10 plays an important role in determining macrophage activation responses.


Neurobiology of Disease | 2017

Peripheral administration of the soluble TNF inhibitor XPro1595 modifies brain immune cell profiles, decreases beta-amyloid plaque load, and rescues impaired long-term potentiation in 5xFAD mice

Kathryn P. MacPherson; Pradoldej Sompol; George T. Kannarkat; Jianjun Chang; Lindsey Sniffen; Mary E. Wildner; Christopher M. Norris; Malú G. Tansey

Clinical and animal model studies have implicated inflammation and peripheral immune cell responses in the pathophysiology of Alzheimers disease (AD). Peripheral immune cells including T cells circulate in the cerebrospinal fluid (CSF) of healthy adults and are found in the brains of AD patients and AD rodent models. Blocking entry of peripheral macrophages into the CNS was reported to increase amyloid burden in an AD mouse model. To assess inflammation in the 5xFAD (Tg) mouse model, we first quantified central and immune cell profiles in the deep cervical lymph nodes and spleen. In the brains of Tg mice, activated (MHCII+, CD45high, and Ly6Chigh) myeloid-derived CD11b+ immune cells are decreased while CD3+ T cells are increased as a function of age relative to non-Tg mice. These immunological changes along with evidence of increased mRNA levels for several cytokines suggest that immune regulation and trafficking patterns are altered in Tg mice. Levels of soluble Tumor Necrosis Factor (sTNF) modulate blood-brain barrier (BBB) permeability and are increased in CSF and brain parenchyma post-mortem in AD subjects and Tg mice. We report here that in vivo peripheral administration of XPro1595, a novel biologic that sequesters sTNF into inactive heterotrimers, reduced the age-dependent increase in activated immune cells in Tg mice, while decreasing the overall number of CD4+ T cells. In addition, XPro1595 treatment in vivo rescued impaired long-term potentiation (LTP) measured in brain slices in association with decreased Aβ plaques in the subiculum. Selective targeting of sTNF may modulate brain immune cell infiltration, and prevent or delay neuronal dysfunction in AD. SIGNIFICANCE STATEMENT Immune cells and cytokines perform specialized functions inside and outside the brain to maintain optimal brain health; but the extent to which their activities change in response to neuronal dysfunction and degeneration is not well understood. Our findings indicate that neutralization of sTNF reduced the age-dependent increase in activated immune cells in Tg mice, while decreasing the overall number of CD4+ T cells. In addition, impaired long-term potentiation (LTP) was rescued by XPro1595 in association with decreased hippocampal Aβ plaques. Selective targeting of sTNF holds translational potential to modulate brain immune cell infiltration, dampen neuroinflammation, and prevent or delay neuronal dysfunction in AD.


npj Parkinson's disease | 2017

LRRK2 levels in immune cells are increased in Parkinson’s disease

Da Cook; George T. Kannarkat; A.F Cintron; Laura M. Butkovich; Kyle B. Fraser; Jianjun Chang; N. Grigoryan; Stewart A. Factor; Andrew B. West; Jeremy M. Boss; MariadeLourdes Tansey

Mutations associated with leucine-rich repeat kinase 2 are the most common known cause of Parkinson’s disease. The known expression of leucine-rich repeat kinase 2 in immune cells and its negative regulatory function of nuclear factor of activated T cells implicates leucine-rich repeat kinase 2 in the development of the inflammatory environment characteristic of Parkinson’s disease. The aim of this study was to determine the expression pattern of leucine-rich repeat kinase 2 in immune cell subsets and correlate it with the immunophenotype of cells from Parkinson’s disease and healthy subjects. For immunophenotyping, blood cells from 40 Parkinson’s disease patients and 32 age and environment matched-healthy control subjects were analyzed by flow cytometry. Multiplexed immunoassays were used to measure cytokine output of stimulated cells. Leucine-rich repeat kinase 2 expression was increased in B cells (p = 0.0095), T cells (p = 0.029), and CD16+ monocytes (p = 0.01) of Parkinson’s disease patients compared to healthy controls. Leucine-rich repeat kinase 2 induction was also increased in monocytes and dividing T cells in Parkinson’s disease patients compared to healthy controls. In addition, Parkinson’s disease patient monocytes secreted more inflammatory cytokines compared to healthy control, and cytokine expression positively correlated with leucine-rich repeat kinase 2 expression in T cells from Parkinson’s disease but not healthy controls. Finally, the regulatory surface protein that limits T-cell activation signals, CTLA-4 (cytotoxic T-lymphocyte-associated protein 4), was decreased in Parkinson’s disease compared to HC in T cells (p = 0.029). In sum, these findings suggest that leucine-rich repeat kinase 2 has a regulatory role in immune cells and Parkinson’s disease. Functionally, the positive correlations between leucine-rich repeat kinase 2 expression levels in T-cell subsets, cytokine expression and secretion, and T-cell activation states suggest that targeting leucine-rich repeat kinase 2 with therapeutic interventions could have direct effects on immune cell function.Immune system regulation: A known suspectHigh levels of leucine-rich repeat kinase 2 (LRRK2) in immune cells disrupt immune system function in patients with Parkinson’s disease (PD). Mutations in LRRK2 are the most common genetic cause of PD. Although LRRK2 is found in many immune cells, research efforts have focussed on determining its effects on neuronal function. Malu G. Tansey at Emory University, USA, and colleagues compared the levels and function of LRKK2 in immune cells from 40 late-onset PD patients and 32 age- and environment-matched healthy controls. The cells from PD patients had higher levels of LRKK2 protein and produced more pro-inflammatory molecules in response to stimulation than the control cells. As exacerbated inflammatory responses are known to aggravate neurodegeneration, monitoring LRKK2 levels may aid the assessment of disease progression in both inherited and sporadic cases of PD.


Journal of Neuroinflammation | 2016

RGS10 deficiency ameliorates the severity of disease in experimental autoimmune encephalomyelitis

Jae-Kyung Lee; George T. Kannarkat; Jaegwon Chung; Hyun Joon Lee; Kareem Graham; MariadeLourdes Tansey

BackgroundRegulator of G-protein signaling (RGS) family proteins, which are GTPase accelerating proteins (GAPs) that negatively regulate G-protein-coupled receptors (GPCRs), are known to be important modulators of immune cell activation and function. Various single-nucleotide polymorphisms in RGS proteins highly correlate with increased risk for multiple sclerosis (MS), an autoimmune, neurodegenerative disorder. An in-depth search of the gene expression omnibus profile database revealed higher levels of RGS10 and RGS1 transcripts in peripheral blood mononuclear cells (PBMCs) in MS patients, suggesting potential functional roles for RGS proteins in MS etiology and/or progression.MethodsTo define potential roles for RGS10 in regulating autoimmune responses, we evaluated RGS10-null and wild-type (WT) mice for susceptibility to experimental autoimmune encephalomyelitis (EAE), a widely studied model of MS. Leukocyte distribution and functional responses were assessed using biochemical, immunohistological, and flow cytometry approaches.ResultsRGS10-null mice displayed significantly milder clinical symptoms of EAE with reduced disease incidence and severity, as well as delayed onset. We observed fewer CD3+ T lymphocytes and CD11b+ myeloid cells in the central nervous system (CNS) tissues of RGS10-null mice with myelin oligodendrocyte protein (MOG)35–55-induced EAE. Lymph node cells and splenocytes of immunized RGS10-null mice demonstrated decreased proliferative and cytokine responses in response to in vitro MOG memory recall challenge. In adoptive recipients, transferred myelin-reactive RGS10-null Th1 cells (but not Th17 cells) induced EAE that was less severe than their WT counterparts.ConclusionsThese data demonstrate a critical role for RGS10 in mediating autoimmune disease through regulation of T lymphocyte function. This is the first study ever conducted to elucidate the function of RGS10 in effector lymphocytes in the context of EAE. The identification of RGS10 as an important regulator of inflammation might open possibilities for the development of more specific therapies for MS.


Neurobiology of Aging | 2015

Age-related changes in regulator of G-protein signaling (RGS)-10 expression in peripheral and central immune cells may influence the risk for age-related degeneration

George T. Kannarkat; Jae-Kyung Lee; Chenere P. Ramsey; Jaegwon Chung; Jianjun Chang; Isadora Porter; Danielle Oliver; Kennie R. Shepherd; Malú G. Tansey

Inflammation in the aging brain increases risk for neurodegenerative disease. In humans, the regulator of G-protein signaling-10 (RGS10) locus has been associated with age-related maculopathy. Chronic peripheral administration of lipopolysaccharide in the RGS10-null mice induces nigral dopaminergic (DA) degeneration, suggesting that RGS10 modulates neuroimmune interactions and may influence susceptibility to neurodegeneration. Because age is the strongest risk factor for neurodegenerative disease, we assessed whether RGS10 expression changes with age and whether aged RGS10-null mice have altered immune cell profiles. Loss of RGS10 in aged mice does not alter the regulation of nigral DA neurons but does alter B-cell, monocyte, microglial, and CD4+ T-cell populations and inflammatory cytokine levels in the cerebrospinal fluid. These results suggest that loss of RGS10 is associated with an age-dependent dysregulation of peripheral and central immune cells rather than dysregulation of DA neuron function.


Archive | 2014

Role of the Innate and Adaptive Immune System in the Pathogenesis of PD

George T. Kannarkat; Malú G. Tansey

Inflammation has recently been recognized as a significant feature of Parkinson’s disease (PD) pathophysiology, yet its role in limiting or promoting this neurodegenerative multisystem disease is not completely understood. Determining the role of inflammation in PD will help understand how sporadic disease arises. Microglial activation, increased expression of inflammatory genes, infiltration of peripheral immune cells into the central nervous system, and altered distributions of peripheral immune cells make up the in vivo evidence for inflammation in PD. Inflammatory hallmarks are also found in animal models of PD and are reviewed herein. Furthermore, we examine how PD-linked genetic mutations may lead to altered immune function and the extent to which these phenotypes are recapitulated by environmental exposures that may lead to sporadic PD through similar mechanisms. Given the implications of the involvement of the immune system on disease progression, we conclude by considering the potential for translation of immunomodulatory therapies with demonstrated efficacy in preclinical models or other immune-related conditions to the PD clinic. A clear need exists for additional research to understand the role of inflammation and innate and adaptive immunity in the pathophysiology of PD.


Journal of clinical & cellular immunology | 2016

Pesticide-induced Immunotoxicity May Underlie Synergistic Gene-environment Interactions that Increase Parkinson's Disease Risk for High-risk Genotype Individuals at rs3129882 in the HLA-DRA Gene

George T. Kannarkat; Malú G. Tansey

The Human Leukocyte Antigen (HLA) gene loci contains immune system-related genes involved in antigen presentation and in recent years, certain genetic variants in HLA genes have been associated with increased risk for late-onset Parkinson’s disease (PD), the second most common neurodegenerative disorder, in combination with pyrethroid pesticide exposure. The mechanisms behind this interaction are currently under investigation. Evidence that immune responses may confer and/or modulate risk and progression of PD is mounting. Therefore, a clear understanding of how the immune system plays a role in this and other neurodegenerative disorders will be critical for successful development of disease biomarkers, and therapeutic interventions to delay or ameliorate the course of the disease.

Collaboration


Dive into the George T. Kannarkat's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Andrew B. West

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Catherine Kopil

Michael J. Fox Foundation

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge