UFGI publication round-up week 6/12 and 6/19/17

Histopathological and proteomic responses in male Chinese rare minnow (Gobiocypris rarus) indicate hepatotoxicity following benzotriazole exposure.

Author information: Liang X1, Zha J2, Martyniuk CJ3, Wang Z4, Zhao J5.

1School of Ecology and Environment, Inner Mongolia University, Hohhot 010021, China. Electronic address: liangxf@imu.edu.cn.
2Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; Beijing Key Laboratory of Industrial Wastewater Treatment and Reuse, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
3Department of Physiological Sciences and Center for Environmental and Human Toxicology, UF Genetics Institute, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA.
4Key Laboratory of Drinking Water Science and Technology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; State Key Laboratory of Environmental Aquatic Chemistry, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China.
5School of Ecology and Environment, Inner Mongolia University, Hohhot 010021, China.
Journal: Environmental Pollution (Barking, Essex : 1987)

Date of e-pub: June 2017

Abstract: Benzotriazole (BT) and its associated derivatives are used ubiquitously in industrial processes, and can be detected in indoor temperature coolants and in chemicals designed to inhibit corrosion. This chemical has been widely detected in aquatic environments and shows some degree of environmental persistence. Evidence has shown that BT exposure can negatively affect endocrine systems and can result in neurotoxicity in fish. However, no study has examined whether this chemical exhibits hepatotoxicity in fish, and if so, what are the underlying mechanism associated with the damage. To address this knowledge gap, we measured the liver proteome of adult male Chinese rare minnow (Gobiocypris rarus) exposed to either 0.05, 0.5, or 5 mg/L BT for 28 days. Overall, 17 proteins were induced and 9 were reduced in abundance following BT treatment (ratio > 1.5, p < 0.05). Pathway analysis revealed that cellular processes affected by BT included xenobiotic clearance, oxidative stress response, apoptosis, and translation. Moreover, transcripts related to these toxic pathways were also significantly affected by BT. In addition, rare minnows exposed to BT showed signs of hypertrophy of hepatocytes, nuclei pyknosis, and higher levels of cellular vacuolization compared to the controls, thus these early proteomic responses in the liver may be related to pathology (i.e. adverse outcome pathway). Our data demonstrate that BT dysregulates molecular responses in the liver and tissue pathology indicative of damage. This study provides new insight into BT hepatotoxicity in Chinese rare minnow.

 

 

Novel oligodendroglial alpha synuclein viral vector models of multiple system atrophy: studies in rodents and nonhuman primates.

Author information: Mandel RJ1, Marmion DJ2, Kirik D3, Chu Y2, Heindel C4, McCown T4,5, Gray SJ4,6, Kordower JH7,8.

1Gainesville, University of Florida College of Medicine, po 100244, Gainesville, 32610, FL, USA.
2Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Chicago, IL, 60611, USA.
3Department of Experimental Medical Science, Lund University, Lund, Sweden.
4Gene Therapy Center, University of North Carolina, Chapel Hill, NC, USA.
5Department of Psychiatry, University of North Carolina, Chapel Hill, NC, USA.
6Department of Ophthalmology, University of North Carolina, Chapel Hill, NC, USA.
7Department of Neurological Sciences, Rush University Medical Center, 1735 West Harrison Street, Chicago, IL, 60611, USA. jkordowe@rush.edu.
8Van Andel Institute, Grand Rapids, MI, USA. jkordowe@rush.edu.
Journal: Acta Neuropathologica Communications

Date of e-pub: June 2017

Abstract: Multiple system atrophy (MSA) is a horrible and unrelenting neurodegenerative disorder with an uncertain etiology and pathophysiology. MSA is a unique proteinopathy in which alpha-synuclein (α-syn) accumulates preferentially in oligodendroglia rather than neurons. Glial cytoplasmic inclusions (GCIs) of α-syn are thought to elicit changes in oligodendrocyte function, such as reduced neurotrophic support and demyelination, leading to neurodegeneration. To date, only a murine model using one of three promoters exist to study this disease. We sought to develop novel rat and nonhuman primate (NHP) models of MSA by overexpressing α-syn in oligodendroglia using a novel oligotrophic adeno-associated virus (AAV) vector, Olig001. To establish tropism, rats received intrastriatal injections of Olig001 expressing GFP. Histological analysis showed widespread expression of GFP throughout the striatum and corpus callosum with >95% of GFP+ cells co-localizing with oligodendroglia and little to no expression in neurons or astrocytes. We next tested the efficacy of this vector in rhesus macaques with intrastriatal injections of Olig001 expressing GFP. As in rats, we observed a large number of GFP+ cells in gray matter and white matter tracts of the striatum and the corpus callosum, with 90-94% of GFP+ cells co-localizing with an oligodendroglial marker. To evaluate the potential of our vector to elicit MSA-like pathology in NHPs, we injected rhesus macaques intrastriatally with Olig001 expressing the α-syn transgene. Histological analysis 3-months after injection demonstrated widespread α-syn expression throughout the striatum as determined by LB509 and phosphorylated serine-129 α-syn immunoreactivity, all of which displayed as tropism similar to that seen with GFP. As in MSA, Olig001-α-syn GCIs in our model were resistant to proteinase K digestion and caused microglial activation. Critically, demyelination was observed in the white matter tracts of the corpus callosum and striatum of Olig001-α-syn but not Olig001-GFP injected animals, similar to the human disease. These data support the concept that this vector can provide novel rodent and nonhuman primate models of MSA.

 

 

Hydrogen peroxide stimulates exosomal cathepsin B regulation of the receptor for advanced glycation end-products (RAGE).

Author information: Downs CA1, Dang VD2, Johnson NM1, Denslow N2, Alli AA3.

1College of Nursing, Biobehavioral Healthscience Division & College of Medicine, Department of Medicine Division of Translational & Regenerative Medicine, The University of Arizona, Tucson, AZ.
2Department of Physiological Sciences and Center for Environmental and Human Toxicology, University of Florida, Gainesville Florida.
3Department of Physiology and Functional Genomics and Department of Medicine Division of Nephrology, Hypertension, and Renal Transplantation, University of Florida College of Medicine, Gainesville Florida.
Journal: Journal of Cellular Biochemistry

Date of e-pub: June 2017

Abstract: Exosomes are nano-sized vesicles that are secreted into the extracellular environment. These vesicles contain various biological effector molecules that can regulate intracellular signaling pathways in recipient cells. The aim of this study was to examine a correlation between exosomal cathepsin B activity and the receptor for advanced glycation end-products (RAGE). Type 1 alveolar epithelial (R3/1) cells were treated with or without hydrogen peroxide and exosomes isolated from the cell conditioned media were characterized by NanoSight analysis. Lipidomic and proteomic analysis showed exosomes released from R3/1 cells exposed to oxidative stress induced by hydrogen peroxide or vehicle differ in their lipid and protein content, respectively. Cathepsin B activity was detected in exosomes isolated from hydrogen peroxide treated cells. The mRNA and protein expression of RAGE increased in cultured R3/1 cells treated with exosomes containing active cathepsin B while depletion of exosomal cathepsin B attenuated RAGE mRNA and protein expression. These results suggest exosomal cathepsin B regulates RAGE in type 1 alveolar cells under conditions of oxidative stress. This article is protected by copyright. All rights reserved.

 

 

Multi-drug resistant Klebsiella pneumoniae strains circulating in hospital setting: whole-genome sequencing and Bayesian phylogenetic analysis for outbreak investigations.

Author information: Cella E1,2, Ciccozzi M3,4, Presti AL1, Fogolari M5, Azarian T6, Prosperi M7, Salemi M8, Equestre M9, Antonelli F5, Conti A5, Cesaris M5, Spoto S10, Incalzi RA11, Coppola R12, Dicuonzo G5, Angeletti S5.

1Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Rome, Italy.
2Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
3Department of Infectious, Parasitic and Immune-Mediated Diseases, Istituto Superiore di Sanità, Rome, Italy. ciccozzi@iss.it.
4Unit of Clinical Pathology and Microbiology, University Campus Bio-Medico of Rome, Rome, Italy. ciccozzi@iss.it.
5Unit of Clinical Pathology and Microbiology, University Campus Bio-Medico of Rome, Rome, Italy.
6Department of Epidemiology, Center for Communicable Disease Dynamics, Harvard’s T.H. Chan School of Public Health, Boston, MA, USA.
7Department of Epidemiology, University of Florida, Gainesville, FL, USA.
8Department of Pathology, Immunology, and Laboratory Medicine, Emerging Pathogens Institute, University of Florida, Gainesville, FL, USA.
9Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy.
10Internal Medicine Department, University Hospital Campus Bio-Medico, Rome, Italy.
11Unit of Geriatrics, Department of Medicine, University Campus Bio-Medico of Rome, Rome, Italy.
12 Department of Surgery, University Campus Bio-Medico of Rome, Rome, Italy.
Journal: Scientific Reports

Date of e-pub: June 2017

Abstract:  Carbapenems resistant Enterobacteriaceae infections are increasing worldwide representing an emerging public health problem. The application of phylogenetic and phylodynamic analyses to bacterial whole genome sequencing (WGS) data have become essential in the epidemiological surveillance of multi-drug resistant nosocomial pathogens. Between January 2012 and February 2013, twenty-one multi-drug resistant K. pneumoniae strains, were collected from patients hospitalized among different wards of the University Hospital Campus Bio-Medico. Epidemiological contact tracing of patients and Bayesian phylogenetic analysis of bacterial WGS data were used to investigate the evolution and spatial dispersion of K. pneumoniae in support of hospital infection control. The epidemic curve of incident K. pneumoniae cases showed a bimodal distribution of cases with two peaks separated by 46 days between November 2012 and January 2013. The time-scaled phylogeny suggested that K. pneumoniae strains isolated during the study period may have been introduced into the hospital setting as early as 2007. Moreover, the phylogeny showed two different epidemic introductions in 2008 and 2009. Bayesian genomic epidemiology is a powerful tool that promises to improve the surveillance and control of multi-drug resistant pathogens in an effort to develop effective infection prevention in healthcare settings or constant strains reintroduction.

 

 

A novel phylogroup of Pseudomonas cichorii identified following an unusual disease outbreak on tomato.

Author information: Timilsina S1, Adkison H2, Testen AL3, Newberry EA4, Miller SA5, Paret ML6, Minsavage GV7, Goss EM8, Jones JB9, Vallad GE10,11.

1University of Florida, Department of Plant Pathology, Gainesville, Florida, United States ; sujan.timilsina@ufl.edu.
2University of Florida, Plant Pathology , Gulf Coast Research and Education Center , 14625 C.R. 672 , Wimauma, Florida, United States , 33598 ; hadkison@ufl.edu.
3The Ohio State University OARDC, Plant Pathology , 1680 Madison Ave , Wooster, Ohio, United States , 44691 ; testen.2@osu.edu.
4University of Florida, Plant Pathology , PO Box 110680 , Gainesville, Florida, United States , 32611 ; ean06@ufl.edu.
5The Ohio State University, Plant Pathology , 1680 Madison Ave. , Wooster, Ohio, United States , 44691 ; miller.769@osu.edu.
6NFREC, University of Florida , 155 Research Road , Quincy, Florida, United States , 32351 ; paret@ufl.edu.
7University of Florida, Department of Plant Pathology, Gainesville, Florida, United States ; gvmins@ufl.edu.
8University of Florida, Dept. of Plant Pathology & Emerging Pathogens Institute , P.O. Box 110680 , Gainesville, Florida, United States , 32605 ; emgoss@ufl.edu.
9University of Florida, Plant Patholgoy Dept. , 1453 Fifield Hall , Gainesville, Florida, United States , 32611 ; jbjones@ufl.edu.
10University of Florida, Plant Pathology , Gulf Coast Research and Education Center , 14625 C.R. 672 , Wimauma, Florida, United States , 33598.
11United States ; gvallad@ufl.edu.
Journal: Phytopathology

Date of e-pub: June 2017

Abstract: Recently, in Central Florida tomato production fields, tomato foliage and fruit were observed with symptoms similar to bacterial speck. Fluorescent pseudomonads were consistently isolated and the strains were characterized by standard LOPAT tests, pathogenicity tests and genetic characterization using 16S rRNA sequences and multilocus sequence analysis of conserved housekeeping genes. LOPAT test results indicated the strains were likely P. cichorii. These strains were pathogenic on tomato and were also pathogenic on lettuce, the host for the type strain of P. cichorii. Likewise, strains of P. cichorii isolated in Florida since the early 1980s from hosts other than tomato, along with the type strain, were also pathogenic on tomato. Genetic characterization using 16S rRNA and multilocus sequence analysis (MLSA) confirmed that the strains were most closely related to P. cichorii, but they varied significantly from the type strain. The Florida P. cichorii strains formed a separate phylogenetic group along with P. cichorii strains isolated from tomato in Tanzania. These strains were different from the previously described morphotypes and genomovars of P. cichorii. Our results indicate the presence of a genetically distinct group of multi-host pathogenic P. cichorii strains that have been present in Florida since at least the early 1980s.

 

 

Submicroscopic Malaria Infections in Pregnant Women from Six Departments in Haiti.

Author information: Elbadry MA1,2, Tagliamonte MS3, Raccurt CP4, Lemoine JF5, Existe A4, Boncy J4, Weppelman TA1,6, Dame JB2,3, Okech BA1,2.

1Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, USA.
2Emerging Pathogens Institute, University of Florida, Gainesville, USA.
3Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, USA.
4Laboratoire National de Santé Publique, Ministère de la Santé Publique et de la Population, Port au Prince, Haiti.
5Programme National de Contrôle de la Malaria, Ministère de la Santé Publique et de la Population, Port-au-Prince, Haiti.
6Herbert Wertheim College of Medicine, Florida International University, Miami, USA.
Journal: Tropical Medicine & International Health : TM & IH

Date of e-pub: June 2017

Abstract: 
To describe the epidemiology of malaria in pregnancy in Haiti.

Cross-sectional study among pregnant women in 6 departments of Haiti. After obtaining informed consent, whole blood samples and demographic surveys were collected to investigate malaria prevalence, anemia and socio-behavioral risk factors for infection, respectively. 311 pregnant women were screened for Plasmodium falciparum infection using a rapid diagnostic test (RDT), microscopy, and a novel, quantitative reverse-transcriptase polymerase chain reaction method (qRT-PCR).

Overall, 1.2% (4/311) of pregnant women tested positive for malaria infection by both microscopy and RDT. However, using the qRT-PCR, 16.4% (51/311) of pregnant women were positive. The prevalence of malaria infection varied with geographic locations ranging between 0% to 46.4%. Additionally, 53% of pregnant women had some form of anemia; however no significant association was found between anemia and submicroscopic malaria infection. The socio-behavioral risk factors identified to be protective of malaria infection were marital status (P<0.05) and travel within one month prior to screening (P< 0.05).

This study is the first to document the high prevalence of submicroscopic malaria infections among pregnant women in Haiti and identify social and behavioral risk factors for disease transmission. This article is protected by copyright. All rights reserved.

 

 

UBASH3A Mediates Risk for Type 1 Diabetes through Inhibition of T-Cell Receptor-Induced NF-κB Signaling.

Author information: Ge Y1,2, Paisie TK1,2,3, Newman JRB2,4, McIntyre LM2,4, Concannon P5,2.

1Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL.
2Genetics Institute, University of Florida, Gainesville, FL.
3Genetics and Genomics Graduate Program, University of Florida, Gainesville, FL.
4Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL.
5Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL patcon@ufl.edu.
Journal: Diabetes

Date of e-pub: July 2017

Abstract: Although over 40 type 1 diabetes (T1D) risk loci have been mapped in humans, the causative genes and variants for T1D are largely unknown. Here, we investigated a candidate gene in the 21q22.3 risk locus-UBASH3A, which is primarily expressed in T cells where it is thought to play a largely redundant role. Genetic variants in UBASH3A have been shown to be associated with several autoimmune diseases in addition to T1D. However, the molecular mechanism underlying these genetic associations is unresolved. Our study reveals a previously unrecognized role of UBASH3A in human T cells: UBASH3A attenuates the NF-κB signal transduction upon T cell receptor (TCR) stimulation by specifically suppressing the activation of the IKK complex. We identify novel interactions of UBASH3A with non-degradative polyubiquitin chains, TAK1 and NEMO, suggesting that UBASH3A regulates the NF-κB signaling pathway by a ubiquitin-dependent mechanism. Finally, we show that risk alleles at rs11203203 and rs80054410, two T1D-associated variants in UBASH3A, increase UBASH3A expression in human primary CD4+ T cells upon TCR stimulation, inhibiting NF-κB signaling via its effects on the IKK complex and resulting in reduced IL2 gene expression.

 

 

Enzyme activity and substrate specificity of the major cinnamyl alcohol dehydrogenases in sorghum.

Author information: Jun SY1, Walker AM1, Kim H2, Ralph J3, Vermerris W4, Sattler SE5, Kang C6.

1Washington State University CITY: Pullman STATE: WA United States Of America [US].
2U. Wisconsin-Madison CITY: Madison STATE: Wisconsin United States Of America [US].
3U. Wisconsin-Madison CITY: Madison STATE: Wisconsin POSTAL_CODE: 53726-4084 United States Of America [US].
4University of Florida CITY: Gainesville STATE: Florida POSTAL_CODE: 32610 United States Of America [US].
5USDA CITY: Lincoln STATE: Nebraska POSTAL_CODE: 68583-0737 United States Of America [US].
6Washington State University 264 Fulmer, Washington State University CITY: Pullman STATE: WA POSTAL_CODE: WA99164 United States Of America [US] chkang@wsu.edu.
Journal: Plant Physiology

Date of e-pub: June 2017

Abstract: Cinnamyl alcohol dehydrogenase (CAD) catalyzes the final step in monolignol biosynthesis, reducing sinapaldehyde, coniferaldehyde, and p-coumaraldehyde to their corresponding alcohols in an NADPH-dependent manner. Because of its terminal location in monolignol biosynthesis, variation in substrate specificity and activity of CAD can result in significant changes in overall composition and amount of lignin. Our in-depth characterization of two major CAD isoforms, SbCAD2 (Brown midrib6) and SbCAD4, in lignifying tissues of sorghum, a strategic plant for generating renewable chemicals and fuels, indicates their similarity in both structure and activity to Arabidopsis thaliana CAD5 and Populus tremuloides sinapyl alcohol dehydrogenase (SAD), respectively. This first crystal structure of a monocot CAD combined with enzyme kinetic data and a catalytic model supported by site-directed mutagenesis allows full comparison with dicot CADs and elucidates the potential signature sequence for their substrate specificity and activity. The L119W/G301F-SbCAD4 double mutant displayed its substrate-preference in the order coniferaldehyde > p-coumaraldehyde > sinapaldehyde, with higher catalytic efficiency than that of both wild-type SbCAD4 and SbCAD2. As SbCAD4 is the only major CAD isoform in bmr6 mutants, replacing SbCAD4 with L119W/G301F-SbCAD4 in bmr6 plants could produce a phenotype that is more amenable to biomass processing.

 

 

An Immune-Competent Murine Model to Study Elimination of AAV-Transduced Hepatocytes by Capsid-Specific CD8+ T Cells.

Author information: Palaschak B1, Marsic D1, Herzog RW1, Zolotukhin S1, Markusic DM1.

1Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA.
Journal: Molecular Therapy. Methods & Clinical Development

Date of e-pub: June 2017

Abstract: Multiple independent adeno-associated virus (AAV) gene therapy clinical trials for hemophilia B, utilizing different AAV serotypes, have reported a vector dose-dependent loss of circulating factor IX (FIX) protein associated with capsid-specific CD8+ T cell (Cap-CD8) elimination of transduced hepatocytes. Hemophilia B patients who develop transient transaminitis and loss of FIX protein may be stabilized with the immune-suppressive (IS) drug prednisolone, but do not all recover lost FIX expression, whereas some patients fail to respond to IS. We developed the first animal model demonstrating Cap-CD8 infiltration and elimination of AAV-transduced hepatocytes of immune-deficient mice. Here, we extend this model to an immune-competent host where Cap-CD8 transfer to AAV2-F9-treated mice significantly reduced circulating and hepatocyte FIX expression. Further, we studied two high-expressing liver tropic AAV2 variants, AAV2-LiA and AAV2-LiC, obtained from a rationally designed capsid library. Unlike AAV2, Cap-CD8 did not initially reduce circulating FIX levels for either variant. However, FIX levels were significantly reduced in AAV2-LiC-F9-treated, but not AAV2-LiA-F9-treated, mice at the study endpoint. Going forward, the immune-competent model may provide an opportunity to induce immunological memory directed against a surrogate AAV capsid antigen and study recall responses following AAV gene transfer.

 

 

Immune Modulatory Cell Therapy for Hemophilia B Based on CD20-Targeted Lentiviral Gene Transfer to Primary B Cells.

Author information: Wang X1, Herzog RW1, Byrne BJ2, Kumar SRP1, Zhou Q3, Buchholz CJ3, Biswas M1.

1Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA.
2Powell Gene Therapy Center, Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA.
3Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut, 63225 Langen, Germany.
Journal: Molecular Therapy. Methods & Clinical Development

Date of e-pub: June 2017

Abstract: Gene-modified B cells expressing immunoglobulin G (IgG) fusion proteins have been shown to induce tolerance in several autoimmune and other disease models. However, lack of a vector suitable for gene transfer to human B cells has been an obstacle for translation of this approach. To overcome this hurdle, we developed an IgG-human factor IX (hFIX) lentiviral fusion construct that was targeted to specifically transduce cells expressing human CD20 (hCD20). Receptor-specific retargeting by mutating envelope glycoproteins of measles virus (MV)-lentiviral vector (LV) and addition of a single-chain variable fragment specific for hCD20 resulted in gene delivery into primary human and transgenic hCD20 mouse B cells with high specificity. Notably, this protocol neither required nor induced activation of the B cells, as confirmed by minimal activation of inflammatory cytokines. Using this strategy, we were able to demonstrate induction of humoral tolerance, resulting in suppression of antibody formation against hFIX in a mouse model of hemophilia B (HB). In conclusion, transduction of receptor-specific retargeted LV into resting B cells is a promising method to develop B cell therapies for antigen-specific tolerance induction in human disease.

 

 

Plasmacytoid and conventional dendritic cells cooperate in crosspriming AAV capsid-specific CD8+ T cells.

Author information: Rogers GL1, Shirley JL1, Zolotukhin I1, Kumar SRP1, Sherman A1, Perrin GQ1, Hoffman BE1, Srivastava A1, Basner-Tschakarjan E2, Wallet MA3, Terhorst C4, Biswas M1, Herzog RW1.

1Department of Pediatrics, Division of Cellular and Molecular Therapy, University of Florida, Gainesville, FL.
2Center of Molecular and Cellular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA.
3Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL; and.
4Division of Immunology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA.
Journal: Blood

Date of e-pub: June 2017

Abstract: Adeno-associated virus (AAV) is a replication-deficient parvovirus that is extensively used as a gene therapy vector. CD8+ T-cell responses against the AAV capsid protein can, however, affect therapeutic efficacy. Little is known about the in vivo mechanism that leads to the crosspriming of CD8+ T cells against the input viral capsid antigen. In this study, we report that the Toll-like receptor 9 (TLR9)-MyD88 pattern-recognition receptor pathway is uniquely capable of initiating this response. By contrast, the absence of TLR2, STING, or the addition of TLR4 agonist has no effect. Surprisingly, both conventional dendritic cells (cDCs) and plasmacytoid DCs (pDCs) are required for the crosspriming of capsid-specific CD8+ T cells, whereas other antigen-presenting cells are not involved. TLR9 signaling is specifically essential in pDCs but not in cDCs, indicating that sensing of the viral genome by pDCs activates cDCs in trans to cross-present capsid antigen during CD8+ T-cell activation. Cross-presentation and crosspriming depend not only on TLR9, but also on interferon type I signaling, and both mechanisms can be inhibited by administering specific molecules to prevent induction of capsid-specific CD8+ T cells. Thus, these outcomes directly point to therapeutic interventions and demonstrate that innate immune blockade can eliminate unwanted immune responses in gene therapy.

 

Gene Therapy for Leber Hereditary Optic Neuropathy: Low- and Medium-Dose Visual Results.

Author information: Guy J1, Feuer WJ2, Davis JL2, Porciatti V2, Gonzalez PJ2, Koilkonda RD2, Yuan H2, Hauswirth WW3, Lam BL2.

1Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida. Electronic address: JGuy@med.miami.edu.
2Bascom Palmer Eye Institute, University of Miami Miller School of Medicine, Miami, Florida.
3Department of Ophthalmology University of Florida College of Medicine, Gainesville, Florida.
Journal: Ophthalmology

Date of e-pub: June 2017

Abstract: To determine the effects of AAV2(Y444,500,730F)-P1ND4v2 in patients with Leber hereditary optic neuropathy (LHON).

Prospective open-label, unilateral single-dose, intravitreal injection of AAV2(Y444,500,730F)-P1ND4v2 per participant.

Fourteen patients with visual loss and mutated G11778A mitochondrial DNA.

Intravitreal injection with the gene therapy vector AAV2(Y444,500,730F)-P1ND4v2 into 1 eye. Six participants with chronic bilateral visual loss lasting more than 12 months (group 1), 6 participants with bilateral visual loss lasting less than 12 months (group 2), and 2 participants with unilateral visual loss (group 3) were treated. Nine patients had at least 12 months of follow-up. Clinical testing included visual acuity, visual fields, optical coherence tomography, pattern electroretinography, and neuro-ophthalmic examinations. Generalized estimating equation methods were used for longitudinal analyses.
MAIN OUTCOME MEASURE:
Loss of visual acuity.

For groups 1 and 2, month 12 average acuity improvements with treatment relative to baseline were 0.24 logarithm of the minimum angle of resolution (logMAR). Fellow eyes had a 0.09-logMAR improvement. A post hoc comparison found that at month 12, the difference between study eye minus fellow eye improvement in group 2 patients of 0.53 logMAR was greater than that observed in our prior acute natural history patients of 0.21 logMAR (P = 0.053). At month 18, the difference between study eye minus fellow eye improvement in our acute group 2 gene therapy patients of 0.96 was more than that observed in our prior acute natural history patients (0.17 logMAR; P < 0.001). Two patients demonstrated asymptomatic uveitis that resolved without treatment. Optical coherence tomography of treated eyes showed an average temporal retinal nerve fiber layer thickness of 54 μm before injection and 55 μm at month 12. For fellow eyes before injection, it was 56 μm, decreasing to 50 μm at month 12 (P = 0.013). Generalized estimating equations suggested that PERG amplitudes worsened more in treated eyes than in fellow eyes by approximately 0.05 μV (P = 0.009 exchangeable). No difference between eyes in outcomes of other visual function measures was evident.

Allotopic gene therapy for LHON at low and medium doses seems to be safe and does not damage the temporal retinal nerve fiber layer, opening the door next for testing of the high dose.

 

 

Phylogenomic analysis supports multiple instances of polyphyly in the oomycete peronosporalean lineage.

Author information: Ascunce MS1, Huguet-Tapia JC2, Ortiz-Urquiza A3, Keyhani NO3, Braun EL4, Goss EM5.

1Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA; Department of Plant Pathology, University of Florida, Gainesville, Florida, USA.
2Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA.
3Department of Microbiology and Cell Science, University of Florida, Gainesville, Florida, USA.
4Department of Biology and Genetics Institute, University of Florida, Gainesville, Florida, USA. Electronic address: ebraun68@ufl.edu.
5Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA; Department of Plant Pathology, University of Florida, Gainesville, Florida, USA. Electronic address: emgoss@ufl.edu.
Journal: Molecular Phylogenetics and Evolution

Date of e-pub: June 2017

Abstract: The study of biological diversification of oomycetes has been a difficult task for more than a century. Pioneer researchers used morphological characters to describe this heterogeneous group, and physiological and genetic tools expanded knowledge of these microorganisms. However, research on oomycete diversification is limited by conflicting phylogenies. Using whole genomic data from 17 oomycete taxa, we obtained a dataset of 277 core orthologous genes shared among these genomes. Analyses of this dataset resulted in highly congruent and strongly supported estimates of oomycete phylogeny when we used concatenated maximum likelihood and coalescent-based methods; the one important exception was the position of Albugo. Our results supported the position of Phytopythium vexans (formerly in Pythium clade K) as a sister clade to the Phytophthora-Hyaloperonospora clade. The remaining clades comprising Pythium sensu lato formed two monophyletic groups. One group was composed of three taxa that correspond to Pythium clades A, B and C, and the other group contained taxa representing clades F, G and I, in agreement with previous Pythium phylogenies. However, the group containing Pythium clades F, G and I was placed as sister to the Phytophthora-Hyaloperonospora-Phytopythium clade, thus confirming the lack of monophyly of Pythium sensu lato. Multispecies coalescent methods revealed that the white blister rust, Albugo laibachii, could not be placed with a high degree of confidence. Our analyses show that genomic data can resolve the oomycete phylogeny and provide a phylogenetic framework to study the evolution of oomycete lifestyles.

 

 

Comparative transcriptomic analysis of the evolution and development of flower size in Saltugilia (Polemoniaceae).

Author information: Landis JB1,2,3, Soltis DE4,5,6, Soltis PS5,6.

1Department of Biology, University of Florida, Gainesville, FL, 32611, USA. jacob.landis@ucr.edu.
2Florida Museum of Natural History, University of Florida, Gainesville, FL, 32611, USA. jacob.landis@ucr.edu.
3Department of Botany and Plant Sciences, University of California Riverside, 4412 Boyce Hall, 3401 Watkins Drive, Riverside, CA, 92521, USA. jacob.landis@ucr.edu.
4Department of Biology, University of Florida, Gainesville, FL, 32611, USA.
5Florida Museum of Natural History, University of Florida, Gainesville, FL, 32611, USA.
6Genetics Institute, University of Florida, Gainesville, FL, 32610, USA.
Journal: BMC Genomics

Date of e-pub: June 2017

Abstract: Flower size varies dramatically across angiosperms, representing innovations over the course of >130 million years of evolution and contributing substantially to relationships with pollinators. However, the genetic underpinning of flower size is not well understood. Saltugilia (Polemoniaceae) provides an excellent non-model system for extending the genetic study of flower size to interspecific differences that coincide with variation in pollinators.

Using targeted gene capture methods, we infer phylogenetic relationships among all members of Saltugilia to provide a framework for investigating the genetic control of flower size differences via RNA-Seq de novo assembly. Nuclear concatenation and species tree inference methods provide congruent topologies. The inferred evolutionary trajectory of flower size is from small flowers to larger flowers. We identified 4 to 10,368 transcripts that are differentially expressed during flower development, with many unigenes associated with cell wall modification and components of the auxin and gibberellin pathways.

Saltugilia is an excellent model for investigating covarying floral and pollinator evolution. Four candidate genes from model systems (BIG BROTHER, BIG PETAL, GASA, and LONGIFOLIA) show differential expression during development of flowers in Saltugilia, and four other genes (FLOWERING-PROMOTING FACTOR 1, PECTINESTERASE, POLYGALACTURONASE, and SUCROSE SYNTHASE) fit into hypothesized organ size pathways. Together, these gene sets provide a strong foundation for future functional studies to determine their roles in specifying interspecific differences in flower size.

 

 

CD33 Splicing Polymorphism Determines Gemtuzumab Ozogamicin Response in De Novo Acute Myeloid Leukemia: Report From Randomized Phase III Children’s Oncology Group Trial AAML0531.

Author information: Lamba JK1, Chauhan L1, Shin M1, Loken MR1, Pollard JA1, Wang YC1, Ries RE1, Aplenc R1, Hirsch BA1, Raimondi SC1, Walter RB1, Bernstein ID1, Gamis AS1, Alonzo TA1, Meshinchi S1.

1Jatinder K. Lamba, Lata Chauhan, and Miyoung Shin, University of Florida, Gainesville, FL; Michael R. Loken, Hematologics Inc; Rhonda E. Ries, Irwin D. Bernstein, and Soheil Meshinchi, Fred Hutchinson Cancer Research Center; Roland B. Walter and Soheil Meshinchi, University of Washington, Seattle, WA; Jessica A. Pollard, Maine Medical Center, Portland, ME; Jessica A. Pollard, Tufts University, Boston, MA; Yi-Cheng Wang, Children’s Oncology Group, Monrovia; Todd A. Alonzo, University of Southern California, Los Angeles, CA; Richard Aplenc, Children’s Hospital of Philadelphia, Philadelphia, PA; Betsy A. Hirsch, University of Minnesota, Minneapolis, MN; Susana C. Raimondi, St Jude Children’s Research Hospital, Memphis, TN; and Alan S. Gamis, Children’s Mercy Hospitals and Clinics, Kansas City, MO.
Journal: Journal of Clinical Oncology : Official Journal of the American Society of Clinical Oncology

Date of e-pub: June 2017

Abstract: Purpose Gemtuzumab ozogamicin (GO), a CD33-targeted immunoconjugate, is a re-emerging therapy for acute myeloid leukemia (AML). CD33 single nucleotide polymorphism rs12459419 C>T in the splice enhancer region regulates the expression of an alternatively spliced CD33 isoform lacking exon2 (D2-CD33), thus eliminating the CD33 IgV domain, which is the antibody-binding site for GO, as well as diagnostic immunophenotypic panels. We aimed to determine the impact of the genotype of this splicing polymorphism in patients with AML treated with GO-containing chemotherapy. Patients and Methods CD33 splicing single nucleotide polymorphism was evaluated in newly diagnosed patients with AML randomly assigned to receive standard five-course chemotherapy alone (No-GO arm, n = 408) or chemotherapy with the addition of two doses of GO once during induction and once during intensification (GO arm, n = 408) as per the Children’s Oncology Group AAML0531 trial. Results The rs12459419 genotype was CC in 415 patients (51%), CT in 316 patients (39%), and TT in 85 patients (10%), with a minor allele frequency of 30%. The T allele was significantly associated with higher levels of D2-CD33 transcript ( P < 1.0E-6) and with lower diagnostic leukemic cell surface CD33 intensity ( P < 1.0E-6). Patients with the CC genotype had significantly lower relapse risk in the GO arm than in the No-GO arm (26% v 49%; P < .001). However, in patients with the CT or TT genotype, exposure to GO did not influence relapse risk (39% v 40%; P = .85). Disease-free survival was higher in patients with the CC genotype in the GO arm than in the No-GO arm (65% v 46%, respectively; P = .004), but this benefit of GO addition was not seen in patients with the CT or TT genotype. Conclusion Our results suggest that patients with the CC genotype for rs12459419 have a substantial response to GO, making this a potential biomarker for the selection of patients with a likelihood of significant response to GO.

 

 

Redox regulation of a guard cellSNF1-relatedprotein kinase in Brassica napus, an oilseed crop.

Author information: Zhu M1, Zhang T2, Ji W3, Silva-Sanchez C4, Song W5, Assmann SM6, Harmon AC7, Chen S3.

1Department of Biology, Pennsylvania State University, 208 Mueller Laboratory, University Park, 16802-5301, United States.
2Department of Biology, University of Florida, Genetics Institute, Gainesville, Florida, 32610, United States.
3Dept. of Biology, Genetics Institute, University of Florida, GAINSVILLE, Florida, 32611, United States.
44Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, United States.
5Plant Molecular and Cellular Biology Program, University of Florida, Gainesville, Florida, United States.
6Department of Biology, Pennsylvania State University, 208 Mueller Laboratory, University Park, 16802-5301, United States sma3@psu.edu.
7Associate Professor, Dept. of Botany,and Program in Plant Cellular and Molecular Biology,Adjunct Associate Professor, Dept. of Chemistry, University of Florida, PO Box 118526, GAINSVILLE, FL, 32611, United States.
Journal: The Biochemical Journal

Date of e-pub: June 2017

Abstract: Kinase-mediated phosphorylation is a pivotal regulatory process in stomatal responses to stresses. Through a redox proteomics study, a sucrose non-fermenting 1-related protein kinase (SnRK2.4) was identified to be redox regulated in Brassica napus guard cells upon abscisic acid (ABA) treatment. There are six genes encoding SnRK2.4 paralogs in B. napus Here we show that recombinant BnSnRK2.4-1C exhibited autophosphorylation activity and preferentially phosphorylated the N-terminal region of B. napus slow anion channel (BnSLAC1-NT) over generic substrates. The in vitro activity of BnSnRK2.4-1C requires the presence of manganese (Mn2+). Phosphorylation sites of autophosphorylated BnSnRK2.4-1C were mapped, including serine and threonine residues in the activation loop. In vitro BnSnRK2.4-1C autophosphorylation activity was inhibited by oxidants such as H2O2 and recovered by active thioredoxin isoforms, indicating redox regulation of BnSnRK2.4-1C. Thiol-specific isotope tagging followed by mass spectrometry analysis revealed specific cysteine residues responsive to oxidant treatments. The in vivo activity of BnSnRK2.4-1C is inhibited by 15 min of H2O2 treatment. Together, these data indicate that BnSnRK2.4-1C, a SnRK preferentially expressed in guard cells, is redox regulated with potential roles in guard cell signal transduction.

 

 

Rare Copy Number Variants in NRXN1 and CNTN6 Increase Risk for Tourette Syndrome.

Author information: Huang AY1, Yu D2, Davis LK3, Sul JH4, Tsetsos F5, Ramensky V6, Zelaya I1, Ramos EM4, Osiecki L7, Chen JA1, McGrath LM8, Illmann C7, Sandor P9, Barr CL10, Grados M11, Singer HS11, Nöthen MM12, Hebebrand J13, King RA14, Dion Y15, Rouleau G16, Budman CL17, Depienne C18, Worbe Y19, Hartmann A19, Müller-Vahl KR20, Stuhrmann M21, Aschauer H22, Stamenkovic M23, Schloegelhofer M23, Konstantinidis A24, Lyon GJ25, McMahon WM26, Barta C27, Tarnok Z28, Nagy P28, Batterson JR29, Rizzo R30, Cath DC31, Wolanczyk T32, Berlin C33, Malaty IA34, Okun MS34, Woods DW35, Rees E36, Pato CN37, Pato MT37, Knowles JA38, Posthuma D39, Pauls DL7, Cox NJ3, Neale BM40, Freimer NB4, Paschou P5, Mathews CA41, Scharf JM42, Coppola G43; Tourette Syndrome Association International Consortium for Genetics (TSAICG); Gilles de la Tourette Syndrome GWAS Replication Initiative (GGRI).

1Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA; Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA 90095, USA.
2Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
3Division of Genetic Medicine, Vanderbilt Genetics Institute, Vanderbilt University Medical Center, Nashville, TN 37232, USA.
4Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA.
5Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA.
6Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA; Moscow Institute of Physics and Technology, Dolgoprudny, Institusky 9, Moscow 141701, Russian Federation.
7Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA.
8Department of Psychology, University of Denver, Denver, CO 80210, USA.
9Toronto Western Research Institute, University Health Network and Youthdale Treatment Centres, University of Toronto, Toronto, ON M5T 2S8, Canada.
10Krembil Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada.
11Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
12Department of Genomics, Life & Brain Center, University of Bonn, 53127 Bonn, Germany; Institute of Human Genetics, University of Bonn, 53127 Bonn, Germany.
13Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany.
14Yale Child Study Center, Yale University School of Medicine, New Haven, CT 06520, USA.
15University of Montréal, Montréal, QC H3T 1J4, Canada.
16Department of Neurology and Neurosurgery, Montréal Neurological Institute, McGill University, Montréal, QC H3A 2B4, Canada.
17Hofstra Northwell School of Medicine, Hempstead, NY 11549, USA.
18IGBMC, CNRS UMR 7104/INSERM U964/Université de Strasbourg, 67404 Illkirch Cedex, France; Brain and Spine Institute, UPMC/INSERM UMR_S1127, 75013 Paris Cedex 05, France.
19Brain and Spine Institute, UPMC/INSERM UMR_S1127, 75013 Paris Cedex 05, France.
20Clinic of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, 30625 Hannover, Germany.
21Institute of Human Genetics, Hannover Medical School, 30625 Hannover, Germany.
22Department of Psychiatry and Psychotherapy, Medical University Vienna, 1090 Vienna, Austria; Biopsychosocial Corporation, 1090 Vienna, Austria.
23Department of Psychiatry and Psychotherapy, Medical University Vienna, 1090 Vienna, Austria.
24Department of Psychiatry and Psychotherapy, Medical University Vienna, 1090 Vienna, Austria; Center for Mental Health Muldenstrasse, BBRZMed, 4020 Linz, Austria.
25Stanley Institute for Cognitive Genomics, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA.
26Department of Psychiatry, University of Utah, Salt Lake City, UT 84108, USA.
27Institute of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University, 1085 Budapest, Hungary.
28Vadaskert Child and Adolescent Psychiatric Hospital, 1021 Budapest, Hungary.
29Children’s Mercy Hospital, Kansas City, KS 64108, USA.
30Dipartimento di Medicina Clinica e Sperimentale, Università di Catania, 95131 Catania, Italy.
31Department of Psychiatry, University Medical Center Groningen & Drenthe Mental Health Center, 9700 RB Groningen, the Netherlands; Department of Clinical Psychology, Utrecht University, 3584 CS Utrecht, the Netherlands.
32Department of Child Psychiatry, Medical University of Warsaw, 00-001 Warsaw, Poland.
33Penn State University College of Medicine, Hershey, PA 17033, USA.
34Department of Neurology and Center for Movement Disorders and Neurorestoration, University of Florida, Gainesville, FL 32607, USA.
35Marquette University, Milwaukee, WI 53233, USA; University of Wisconsin-Milwaukee, Milwaukee, WI 53211, USA.
36Medical Research Council Centre for Neuropsychiatric Genetics and Genomics, Cardiff University, Cardiff CF24 4HQ, Wales, UK.
37SUNY Downstate Medical Center, Brooklyn, NY 11203, USA.
38Department of Psychiatry & Behavioral Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
39Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, VU University Amsterdam, 1081 HV Amsterdam, the Netherlands.
40Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Analytic and Translational Genetics Unit, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA.
41Department of Psychiatry, Genetics Institute, University of Florida, Gainesville, FL 32611, USA.
42Psychiatric and Neurodevelopmental Genetics Unit, Center for Genomic Medicine, Department of Psychiatry, Massachusetts General Hospital, Boston, MA 02114, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Neurology, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Neurology, Brigham and Women’s Hospital, Boston, MA 02115, USA. Electronic address: jscharf@partners.org.
43Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Department of Psychiatry and Biobehavioral Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA. Electronic address: gcoppola@ucla.edu.
Journal: Neuron

Date of e-pub: June 2017

Abstract: Tourette syndrome (TS) is a model neuropsychiatric disorder thought to arise from abnormal development and/or maintenance of cortico-striato-thalamo-cortical circuits. TS is highly heritable, but its underlying genetic causes are still elusive, and no genome-wide significant loci have been discovered to date. We analyzed a European ancestry sample of 2,434 TS cases and 4,093 ancestry-matched controls for rare (< 1% frequency) copy-number variants (CNVs) using SNP microarray data. We observed an enrichment of global CNV burden that was prominent for large (> 1 Mb), singleton events (OR = 2.28, 95% CI [1.39-3.79], p = 1.2 × 10-3) and known, pathogenic CNVs (OR = 3.03 [1.85-5.07], p = 1.5 × 10-5). We also identified two individual, genome-wide significant loci, each conferring a substantial increase in TS risk (NRXN1 deletions, OR = 20.3, 95% CI [2.6-156.2]; CNTN6 duplications, OR = 10.1, 95% CI [2.3-45.4]). Approximately 1% of TS cases carry one of these CNVs, indicating that rare structural variation contributes significantly to the genetic architecture of TS.

 

 

Ischemia responsive protein 94 is a key mediator of ischemic neuronal injury-induced microglial activation.

Author information: Tikamdas R1, Singhal S1, Zhang P1, Smith JA1, Krause EG1, Stevens SM Jr2, Song S3, Liu B1.

1Departments of Pharmacodynamics, University of Florida.
2Department of Cell Biology, Microbiology and Molecular Biology, College of Arts and Sciences, University of South Florida, Tampa, Florida, USA.
3Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, Florida, USA.
Journal: Journal of Neurochemistry

Date of e-pub: June 2017

Abstract: Neuroinflammation, especially activation of microglia, the key immune cells in the brain, has been proposed to contribute to the pathogenesis of ischemic stroke. However, the dynamics and the potential mediators of microglial activation following ischemic neuronal injury are not well understood. In this study, using oxygen/glucose deprivation and reoxygenation (OGD/R) with neuronal and microglial cell cultures as an in vitro model of ischemic neuronal injury, we set out to identify neuronal factors released from injured neurons that are capable of inducing microglial activation. Conditioned media (CM) from hippocampal and cortical neurons exposed to OGD/R induced significant activation of microglial cells as well as primary microglia, evidenced by upregulation of inducible nitric oxide synthase, increased production of nitrite and reactive oxygen species and increased expression of microglial markers. Mechanistically, neuronal ischemia responsive protein 94 (Irp94) was a key contributor to microglial activation since significant increase in Irp94 was detected in the neuronal CM following ischemic insult and immunodepletion of Irp94 rendered ischemic neuronal CM ineffective in inducing microglial activation. Ischemic insult-augmented oxidative stress was a major facilitator of neuronal Irp94 release and pharmacological inhibition of NADPH oxidase significantly reduced the ischemic injury induced-neuronal reactive oxygen species production and Irp94 release. Taken together, these results indicate that neuronal Irp94 may play a pivotal role in the propagation of ischemic neuronal damage. Continued studies may help identify Irp94 and/or related proteins as potential therapeutic targets and/or diagnostic/prognostic biomarkers for managing ischemia-associated brain disorders. This article is protected by copyright. All rights reserved.

 

 

Determination of metabolic resistance mechanisms in pyrethroid-resistant and fipronil-tolerant brown dog ticks.

Author information: Eiden AL1, Kaufman PE1, Oi FM1, Dark MJ2,3, Bloomquist JR1,3, Miller RJ4.

1Department of Entomology and Nematology, University of Florida, Gainesville, FL, U.S.A.
2Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL, U.S.A.
3Emerging Pathogens Institute, University of Florida, Gainesville, FL, U.S.A.
4U.S. Department of Agriculture-Agricultural Research Service, Cattle Tick Research Laboratory, Edinburg, TX, U.S.A.
Journal: Medical and Veterinary Entomology

Date of e-pub: June 2017

Abstract: Rhipicephalus sanguineus (Latreille) (Ixodida: Ixodidae) is a three-host dog tick found worldwide that is able to complete its’ entire lifecycle indoors. Options for the management of R. sanguineus are limited and its’ control relies largely on only a few acaricidal active ingredients. Previous studies have confirmed permethrin resistance and fipronil tolerance in R. sanguineus populations, commonly conferred by metabolic detoxification or target site mutations. Herein, five strains of permethrin-resistant and three strains of fipronil-tolerant ticks were evaluated for metabolic resistance using synergists to block metabolic enzymes. Synergist studies were completed with triphenyl phosphate (TPP) for esterase inhibition, piperonyl butoxide (PBO) for cytochrome P450 inhibition, and diethyl maleate (DEM) for glutathione-S-transferase inhibition. Additionally, increased esterase activity was confirmed using gel electrophoresis. The most important metabolic detoxification mechanism in permethrin-resistant ticks was increased esterase activity, followed by increased cytochrome P450 activity. The inhibition of metabolic enzymes did not have a marked impact on fipronil-tolerant tick strains.

 

 

Optimal alpha reduces error rates in gene expression studies: a meta-analysis approach.

Author information: Mudge JF1, Martyniuk CJ2, Houlahan JE3.

1Department of Biology, Canadian Rivers Institute, University of New Brunswick, Saint John, NB, E2L 4L5, Canada.
2Center for Environmental and Human Toxicology & Department of Physiological Sciences, UF Genetics Institute, University of Florida, Gainesville, Florida, 32611, USA.
3Department of Biology, Canadian Rivers Institute, University of New Brunswick, Saint John, NB, E2L 4L5, Canada. jeffhoul@unb.ca.
Journal: BMC Bioinformatics

Date of e-pub: June 2017

Abstract: Transcriptomic approaches (microarray and RNA-seq) have been a tremendous advance for molecular science in all disciplines, but they have made interpretation of hypothesis testing more difficult because of the large number of comparisons that are done within an experiment. The result has been a proliferation of techniques aimed at solving the multiple comparisons problem, techniques that have focused primarily on minimizing Type I error with little or no concern about concomitant increases in Type II errors. We have previously proposed a novel approach for setting statistical thresholds with applications for high throughput omics-data, optimal α, which minimizes the probability of making either error (i.e. Type I or II) and eliminates the need for post-hoc adjustments.

A meta-analysis of 242 microarray studies extracted from the peer-reviewed literature found that current practices for setting statistical thresholds led to very high Type II error rates. Further, we demonstrate that applying the optimal α approach results in error rates as low or lower than error rates obtained when using (i) no post-hoc adjustment, (ii) a Bonferroni adjustment and (iii) a false discovery rate (FDR) adjustment which is widely used in transcriptome studies.

We conclude that optimal α can reduce error rates associated with transcripts in both microarray and RNA-seq experiments, but point out that improved statistical techniques alone cannot solve the problems associated with high throughput datasets – these approaches need to be coupled with improved experimental design that considers larger sample sizes and/or greater study replication.

 

 

Spatio-temporal analysis of coding and long noncoding transcripts during maize endosperm development.

Author information: Kim ED1, Xiong Y2, Pyo Y1, Kim DH1, Kang BH3,4, Sung S5.

1Department of Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA.
2Microbiology and Cell Science, University of Florida, Gainesville, FL, 32611, USA.
3Microbiology and Cell Science, University of Florida, Gainesville, FL, 32611, USA. bkang@cuhk.edu.hk.
4School of Life Sciences, State Key Laboratory for Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China. bkang@cuhk.edu.hk.
5Department of Molecular Biosciences and Institute for Cellular and Molecular Biology, University of Texas at Austin, Austin, TX, 78712, USA. sbsung@austin.utexas.edu.
Journal: Scientific Reports

Date of e-pub: June 2017

Abstract: The maize endosperm consists of three major compartmentalized cell types: the starchy endosperm (SE), the basal endosperm transfer cell layer (BETL), and the aleurone cell layer (AL). Differential genetic programs are activated in each cell type to construct functionally and structurally distinct cells. To compare gene expression patterns involved in maize endosperm cell differentiation, we isolated transcripts from cryo-dissected endosperm specimens enriched with BETL, AL, or SE at 8, 12, and 16 days after pollination (DAP). We performed transcriptome profiling of coding and long noncoding transcripts in the three cell types during differentiation and identified clusters of the transcripts exhibiting spatio-temporal specificities. Our analysis uncovered that the BETL at 12 DAP undergoes the most dynamic transcriptional regulation for both coding and long noncoding transcripts. In addition, our transcriptome analysis revealed spatio-temporal regulatory networks of transcription factors, imprinted genes, and loci marked with histone H3 trimethylated at lysine 27. Our study suggests that various regulatory mechanisms contribute to the genetic networks specific to the functions and structures of the cell types of the endosperm.

 

 

ATM, radiation, and the risk of second primary breast cancer.

Author information: Bernstein JL1, Concannon P2; WECARE Study Collaborative Group.

1a Department of Epidemiology and Biostatistics , Memorial Sloan-Kettering Cancer Center , 485 Lexington Avenue, 2nd Floor , New York , NY 10017.
2b Genetics Institute and Department of Pathology, Immunology and Laboratory Medicine , University of Florida , P.O. Box 103610 , Gainesville , FL 32610-3610.
Journal: International Journal of Radiation Biology

Date of e-pub: June 2017

Abstract: It was first suggested more than 40 years ago that heterozygous carriers for the human autosomal recessive disorder Ataxia-Telangiectasia (A-T) might also be at increased risk for cancer. Subsequent studies have identified the responsible gene, Ataxia-Telangiectasia Mutated (ATM), characterized genetic variation at this locus in A-T and a variety of different cancers, and described the functions of the ATM protein with respect to cellular DNA damage responses. However, an overall model of how ATM contributes to cancer risk, and in particular, the role of DNA damage in this process, remains lacking. This review considers these questions in the context of contralateral breast cancer (CBC).

Heterozygous carriers of loss of function mutations in ATM that are A-T causing, are at increased risk of breast cancer. However, examination of a range of genetic variants, both rare and common, across multiple cancers, suggests that ATM may have additional effects on cancer risk that are allele dependent. In the case of CBC, selected common alleles at ATM are associated with a reduced incidence of CBC, while other rare and predicted deleterious variants may act jointly with radiation exposure to increase risk. Further studies that characterize germline and somatic ATM mutations in breast cancer and relate the detected genetic changes to functional outcomes, particularly with regard to radiation responses, are needed to gain a complete picture of the complex relationship between ATM, radiation and breast cancer.

 

 

NOTE: These abstracts were retrieved from the U.S. National Library of Medicine website managed in collaboration with the U.S. National Library of Medicine

X