Αρχειοθήκη ιστολογίου

Πέμπτη 29 Ιουνίου 2017

Good (and Bad) Ways to Help a Dog Afraid of Fireworks

You are more important than you think 

-- Read more on ScientificAmerican.com

feed?d=yIl2AUoC8zA feed?d=qj6IDK7rITs feed?i=Bc25yX53IWw:OpJCk7l3tFM:gIN9vFwOq feed?d=l6gmwiTKsz0 feed?d=ZC7T4KBF6Nw feed?d=I9og5sOYxJI feed?d=xQlvkV3S7Ew


from # All Medicine by Alexandros G. Sfakianakis via Alexandros G.Sfakianakis on Inoreader http://ift.tt/2tsD03b
via IFTTT

ILF2 Is a Regulator of RNA Splicing and DNA Damage Response in 1q21-Amplified Multiple Myeloma

Publication date: Available online 29 June 2017
Source:Cancer Cell
Author(s): Matteo Marchesini, Yamini Ogoti, Elena Fiorini, Anil Aktas Samur, Luigi Nezi, Marianna D'Anca, Paola Storti, Mehmet Kemal Samur, Irene Ganan-Gomez, Maria Teresa Fulciniti, Nipun Mistry, Shan Jiang, Naran Bao, Valentina Marchica, Antonino Neri, Carlos Bueso-Ramos, Chang-Jiun Wu, Li Zhang, Han Liang, Xinxin Peng, Nicola Giuliani, Giulio Draetta, Karen Clise-Dwyer, Hagop Kantarjian, Nikhil Munshi, Robert Orlowski, Guillermo Garcia-Manero, Ronald A. DePinho, Simona Colla
Amplification of 1q21 occurs in approximately 30% of de novo and 70% of relapsed multiple myeloma (MM) and is correlated with disease progression and drug resistance. Here, we provide evidence that the 1q21 amplification-driven overexpression of ILF2 in MM promotes tolerance of genomic instability and drives resistance to DNA-damaging agents. Mechanistically, elevated ILF2 expression exerts resistance to genotoxic agents by modulating YB-1 nuclear localization and interaction with the splicing factor U2AF65, which promotes mRNA processing and the stabilization of transcripts involved in homologous recombination in response to DNA damage. The intimate link between 1q21-amplified ILF2 and the regulation of RNA splicing of DNA repair genes may be exploited to optimize the use of DNA-damaging agents in patients with high-risk MM.

Graphical abstract

image

Teaser

Marchesini et al. show that in multiple myeloma the overexpression of ILF2, resulting from chromosome 1q21 amplification, drives resistance to DNA-damaging agents partly by modulating the interaction between YB-1 and the splicing factor U2AF65 to promote the processing and stabilization of transcripts involved in homologous recombination.


from # All Medicine by Alexandros G. Sfakianakis via Alexandros G.Sfakianakis on Inoreader http://ift.tt/2tw42aE
via IFTTT

Endogenous endophthalmitis and osteomyelitis associated with interleukin 17 inhibitor treatment for psoriasis in a patient with diabetes

A 64-year-old man with type 2 diabetes mellitus and plaque psoriasis presented to the emergency room with 3 days of progressive right eye pain and decreased vision. After extensive workup and multidisciplinary team effort, the patient was diagnosed with and treated for unilateral endogenous methicillin-sensitive Staphylococcus aureus endophthalmitis, bacteraemia and osteomyelitis of the foot. The patient had been started on the interleukin 17 (IL-17) inhibitor secukinumab for his treatment-resistant plaque psoriasis 4 weeks prior to presentation. After treatment, his final vision was light perception and the foot infection resolved without sequelae. To our knowledge, this is the first reported case of both endogenous endophthalmitis and osteomyelitis associated with an IL-17 inhibitor.



from # All Medicine by Alexandros G. Sfakianakis via Alexandros G.Sfakianakis on Inoreader http://ift.tt/2triYGu
via IFTTT

Intrinsic and Extrinsic Causes of Malignancies in Primary Immunodeficiency Disorders

Malignancies occur with a higher incidence rate and manifest earlier in life in patients with primary immunodeficiency disorders (PID) than in the general population. However, no universal mechanism of malignancy predisposition in PID can been determined. Despite the strong support for the physiological role of tumor immunosurveillance and increasing success of strategies in immunological tumor therapy, which include checkpoint inhibition, monoclonal antibodies, and engineered T cell antigen receptors, the incidence and pattern of malignancies in PID do not reflect an increased tumor immune escape per se.

from # All Medicine by Alexandros G. Sfakianakis via Alexandros G.Sfakianakis on Inoreader http://ift.tt/2uqeVY2
via IFTTT

An elusive ciliopathy: Joubert syndrome

The police brought a 65-year-old female patient to the EADU after being found 'roaming the streets' in an apparent state of confusion. This was her third admission under the same circumstances during the last 3 years. Neurological examination revealed (1) cognitive impairment, (2) oculomotor apraxia, (3) abnormal cancellation of vestibular ocular reflex, (4) mild ataxia and (5) mild hypotonia. Renal function was abnormal and liver function was normal. No retinal disturbance was found. The head CT on admission was normal for stroke and the lumbar puncture was negative for encephalitis. Her brain MRI showed 'molar tooth sign', suggestive of Joubert syndrome, which was confirmed by genetic testing showing anomalous NPHP1 gene



from # All Medicine by Alexandros G. Sfakianakis via Alexandros G.Sfakianakis on Inoreader http://ift.tt/2s8jeq9
via IFTTT

Depression links to physical health decline in cancer caregivers

People who care for a family member with cancer show a steady decline in their own physical health, a study in the journal Cancer has found.1The researchers found that symptoms of depression were the...

recent?d=yIl2AUoC8zA recent?d=dnMXMwOfBR0 recent?i=tZw_iYxGKEk:d4kybL9yAhk:V_sGLiP recent?d=qj6IDK7rITs recent?i=tZw_iYxGKEk:d4kybL9yAhk:gIN9vFw recent?d=l6gmwiTKsz0 recent?d=7Q72WNTAKBA recent?i=tZw_iYxGKEk:d4kybL9yAhk:F7zBnMy recent?i=tZw_iYxGKEk:d4kybL9yAhk:-BTjWOF


from # All Medicine by Alexandros G. Sfakianakis via Alexandros G.Sfakianakis on Inoreader http://ift.tt/2u5oGLU
via IFTTT

Items dispensed in England rise sharply in 10 years

The number of drugs dispensed in the community has risen sharply in the past 10 years, annual figures of prescribing rates have shown.1Data from NHS Digital show that 1.1 billion items were dispensed...

recent?d=yIl2AUoC8zA recent?d=dnMXMwOfBR0 recent?i=w9TYZgXCbOw:CNQ9NKcqhaw:V_sGLiP recent?d=qj6IDK7rITs recent?i=w9TYZgXCbOw:CNQ9NKcqhaw:gIN9vFw recent?d=l6gmwiTKsz0 recent?d=7Q72WNTAKBA recent?i=w9TYZgXCbOw:CNQ9NKcqhaw:F7zBnMy recent?i=w9TYZgXCbOw:CNQ9NKcqhaw:-BTjWOF


from # All Medicine by Alexandros G. Sfakianakis via Alexandros G.Sfakianakis on Inoreader http://ift.tt/2twlxrc
via IFTTT

Subclinical Hypothyroidism

Subclinical Hypothyroidism:



nejmcp1611144.fp.png_v03


New England Journal of Medicine, Volume 376, Issue 26, Page 2556-2565, June 2017.



from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2u53BRw
via IFTTT

Acute Neuroinflammation Promotes Cell Responses to 1800 MHz GSM Electromagnetic Fields in the rat Cerebral Cortex

Erratum to: Acute Neuroinflammation Promotes Cell Responses to 1800 MHz GSM Electromagnetic Fields in the rat Cerebral Cortex:







Neurotox Res. 2017 Jun 3. doi: 10.1007/s12640-017-9756-3. [Epub ahead of print]

Acute Neuroinflammation Promotes Cell Responses to 1800 MHz GSM Electromagnetic Fields in the Rat Cerebral Cortex.



Lameth J1, Gervais A1, Colin C1, Lévêque P2, Jay TM3, Edeline JM4, Mallat M5.

Author information

1

Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France.

2

Université de Limoges, CNRS, XLIM, UMR 7252, 123 avenue Albert Thomas, F-87000, Limoges, France.

3

Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR_S894 INSERM, Université Paris Descartes, 102-108 rue de la Santé, 75014, Paris, France.

4

Paris Saclay Institute of Neuroscience, Neuro-PSI, UMR 9197 CNRS, Université Paris-Sud, 91405, Orsay cedex, France.

5

Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-Salpêtrière, Bat. ICM, 47 boulevard de l'Hôpital, F-75013, Paris, France. michel.mallat@upmc.fr.

Abstract

Mobile phone communications are conveyed by radiofrequency (RF) electromagnetic fields, including pulse-modulated global system for mobile communications (GSM)-1800 MHz, whose effects on the CNS affected by pathological states remain to be specified. Here, we investigated whether a 2-h head-only exposure to GSM-1800 MHz could impact on a neuroinflammatory reaction triggered by lipopolysaccharide (LPS) in 2-week-old or adult rats. We focused on the cerebral cortex in which the specific absorption rate (SAR) of RF averaged 2.9 W/kg. In developing rats, 24 h after GSM exposure, the levels of cortical interleukin-1ß (IL1ß) or NOX2 NADPH oxidase transcripts were reduced by 50 to 60%, in comparison with sham-exposed animals (SAR = 0), as assessed by RT-qPCR. Adult rats exposed to GSM also showed a 50% reduction in the level of IL1ß mRNA, but they differed from developing rats by the lack of NOX2 gene suppression and by displaying a significant growth response of microglial cell processes imaged in anti-Iba1-stained cortical sections. As neuroinflammation is often associated with changes in excitatory neurotransmission, we evaluated changes in expression and phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the adult cerebral cortex by Western blot analyses. We found that GSM exposure decreased phosphorylation at two residues on the GluA1 AMPAR subunit (serine 831 and 845). The GSM-induced changes in gene expressions, microglia, and GluA1 phosphorylation did not persist 72 h after RF exposure and were not observed in the absence of LPS pretreatment. Together, our data provide evidence that GSM-1800 MHz can modulate CNS cell responses triggered by an acute neuroinflammatory state.

KEYWORDS:

AMPA receptor; Electromagnetic fields; Lipopolysaccharide; Microglia; Mobile phone; Neuroinflammation; Radiofrequency

PMID: 28578480 DOI: 10.1007/s12640-017-9756-3









  • Julie Lameth
  • Annie Gervais
  • Catherine Colin
  • Philippe Lévêque
  • Thérèse M. Jay
  • Jean-Marc Edeline
  • Michel MallatEmail author
  • Julie Lameth
    • 1
  • Annie Gervais
    • 1
  • Catherine Colin
    • 1
  • Philippe Lévêque
    • 2
  • Thérèse M. Jay
    • 3
  • Jean-Marc Edeline
    • 4
  • Michel Mallat
    • 1
    Email author
  1. 1.Sorbonne Universités, UPMC Univ Paris 06, INSERM U.1127, CNRS, Institut du Cerveau et de la Moelle épinière (ICM), Hôpital Pitié-SalpêtrièreParisFrance
  2. 2.Université de Limoges, CNRS, XLIM, UMR 7252LimogesFrance
  3. 3.Physiopathologie des Maladies Psychiatriques, Centre de Psychiatrie et Neurosciences, UMR_S894 INSERM, Université Paris DescartesParisFrance
  4. 4.Paris Saclay Institute of Neuroscience, Neuro-PSI, UMR 9197 CNRS, Université Paris-SudOrsay cedexFrance
ORIGINAL ARTICLE

First Online:03 June 2017

Received:13 February 2017Revised:16 May 2017Accepted:19 May 2017
DOI: 10.1007/s12640-017-9756-3

Cite this article as:Lameth, J., Gervais, A., Colin, C. et al. Neurotox Res (2017). doi:10.1007/s12640-017-9756-3

Abstract

Mobile phone communications are conveyed by radiofrequency (RF) electromagnetic fields, including pulse-modulated global system for mobile communications (GSM)-1800 MHz, whose effects on the CNS affected by pathological states remain to be specified. Here, we investigated whether a 2-h head-only exposure to GSM-1800 MHz could impact on a neuroinflammatory reaction triggered by lipopolysaccharide (LPS) in 2-week-old or adult rats. We focused on the cerebral cortex in which the specific absorption rate (SAR) of RF averaged 2.9 W/kg. In developing rats, 24 h after GSM exposure, the levels of cortical interleukin-1ß (IL1ß) or NOX2 NADPH oxidase transcripts were reduced by 50 to 60%, in comparison with sham-exposed animals (SAR = 0), as assessed by RT-qPCR. Adult rats exposed to GSM also showed a 50% reduction in the level of IL1ß mRNA, but they differed from developing rats by the lack of NOX2 gene suppression and by displaying a significant growth response of microglial cell processes imaged in anti-Iba1-stained cortical sections. As neuroinflammation is often associated with changes in excitatory neurotransmission, we evaluated changes in expression and phosphorylation of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors in the adult cerebral cortex by Western blot analyses. We found that GSM exposure decreased phosphorylation at two residues on the GluA1 AMPAR subunit (serine 831 and 845). The GSM-induced changes in gene expressions, microglia, and GluA1 phosphorylation did not persist 72 h after RF exposure and were not observed in the absence of LPS pretreatment. Together, our data provide evidence that GSM-1800 MHz can modulate CNS cell responses triggered by an acute neuroinflammatory state.

Keywords

Electromagnetic fields Radiofrequency Mobile phone Neuroinflammation Microglia AMPA receptor Lipopolysaccharide 
An erratum to this article is available at http://ift.tt/2tse6Av.

References

  1. Ammari M, Gamez C, Lecomte A, Sakly M, Abdelmelek H, De Seze R (2010) GFAP expression in the rat brain following sub-chronic exposure to a 900 MHz electromagnetic field signal. Int J Radiat Biol 86:367–375. doi:10.3109/09553000903567946CrossRefPubMedGoogle Scholar
  2. Arendash GW, Sanchez-Ramos J, Mori T, Mamcarz M, Lin X, Runfeldt M, Wang L, Zhang G, Sava V, Tan J, Cao C (2010) Electromagnetic field treatment protects against and reverses cognitive impairment in Alzheimer's disease mice. J Alzheimers Dis 19:191–210. doi:10.3233/JAD-2010-1228CrossRefPubMedGoogle Scholar
  3. Banke TG, Bowie D, Lee H, Huganir RL, Schousboe A, Traynelis SF (2000) Control of GluR1 AMPA receptor function by cAMP-dependent protein kinase. J Neurosci 20:89–102PubMedGoogle Scholar
  4. Beason RC, Semm P (2002) Responses of neurons to an amplitude modulated microwave stimulus. Neurosci Lett 333:175–178CrossRefPubMedGoogle Scholar
  5. Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB, Mulinyawe SB, Bohlen CJ, Adil A, Tucker A, Weissman IL, Chang EF, Li G, Grant GA, Hayden Gephart MG, Barres BA (2016) New tools for studying microglia in the mouse and human CNS. Proc Natl Acad Sci U S A 113:E1738–E1746. doi:10.1073/pnas.1525528113CrossRefPubMedPubMedCentralGoogle Scholar
  6. Boisse L, Mouihate A, Ellis S, Pittman QJ (2004) Long-term alterations in neuroimmune responses after neonatal exposure to lipopolysaccharide. J Neurosci 24:4928–4934. doi:10.1523/JNEUROSCI.1077-04.2004CrossRefPubMedGoogle Scholar
  7. Bouji M, Lecomte A, Hode Y, de Seze R, Villegier AS (2012) Effects of 900 MHz radiofrequency on corticosterone, emotional memory and neuroinflammation in middle-aged rats. Exp Gerontol 47:444–451. doi:10.1016/j.exger.2012.03.015CrossRefPubMedGoogle Scholar
  8. Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G, Koeglsperger T, Dake B, Wu PM, Doykan CE, Fanek Z, Liu L, Chen Z, Rothstein JD, Ransohoff RM, Gygi SP, Antel JP, Weiner HL (2014) Identification of a unique TGF-beta-dependent molecular and functional signature in microglia. Nat Neurosci 17:131–143. doi:10.1038/nn.3599CrossRefPubMedGoogle Scholar
  9. Carballo-Quintas M, Martinez-Silva I, Cadarso-Suarez C, Alvarez-Figueiras M, Ares-Pena FJ, Lopez-Martin E (2011) A study of neurotoxic biomarkers, c-fos and GFAP after acute exposure to GSM radiation at 900 MHz in the picrotoxin model of rat brains. Neurotoxicology 32:478–494. doi:10.1016/j.neuro.2011.04.003CrossRefPubMedGoogle Scholar
  10. Caudal D, Godsil BP, Mailliet F, Bergerot D, Jay TM (2010) Acute stress induces contrasting changes in AMPA receptor subunit phosphorylation within the prefrontal cortex, amygdala and hippocampus. PLoS One 5:e15282. doi:10.1371/journal.pone.0015282CrossRefPubMedPubMedCentralGoogle Scholar
  11. Caudal D, Rame M, Jay TM, Godsil BP (2016) Dynamic regulation of AMPAR phosphorylation in vivo following acute behavioral stress. Cell Mol Neurobiol 36:1331–1342. doi:10.1007/s10571-016-0332-9CrossRefPubMedGoogle Scholar
  12. Cheret C, Gervais A, Lelli A, Colin C, Amar L, Ravassard P, Mallet J, Cumano A, Krause KH, Mallat M (2008) Neurotoxic activation of microglia is promoted by a nox1-dependent NADPH oxidase. J Neurosci 28:12039–12051CrossRefPubMedGoogle Scholar
  13. Christensen LB, Woods TA, Carmody AB, Caughey B, Peterson KE (2014) Age-related differences in neuroinflammatory responses associated with a distinct profile of regulatory markers on neonatal microglia. J Neuroinflammation 11:70. doi:10.1186/1742-2094-11-70CrossRefPubMedPubMedCentralGoogle Scholar
  14. Collin A, Perrin A, Cretallaz C, Pla S, Arnaud-Cormos D, Debouzy JC, Leveque P (2016) In vivo setup characterization for pulsed electromagnetic field exposure at 3 GHz. Phys Med Biol 61:5925–5941. doi:10.1088/0031-9155/61/16/5925CrossRefPubMedGoogle Scholar
  15. Court-Kowalski S, Finnie JW, Manavis J, Blumbergs PC, Helps SC, Vink R (2015) Effect of long-term (2 years) exposure of mouse brains to global system for mobile communication (GSM) radiofrequency fields on astrocytic immunoreactivity. Bioelectromagnetics 36:245–250. doi:10.1002/bem.21891CrossRefPubMedGoogle Scholar
  16. Cunningham C, Campion S, Lunnon K, Murray CL, Woods JF, Deacon RM, Rawlins JN, Perry VH (2009) Systemic inflammation induces acute behavioral and cognitive changes and accelerates neurodegenerative disease. Biol Psychiatry 65:304–312. doi:10.1016/j.biopsych.2008.07.024CrossRefPubMedPubMedCentralGoogle Scholar
  17. Dantzer R (2001) Cytokine-induced sickness behavior: where do we stand? Brain Behav Immun 15:7–24. doi:10.1006/brbi.2000.0613CrossRefPubMedGoogle Scholar
  18. Dantzer R, Walker AK (2014) Is there a role for glutamate-mediated excitotoxicity in inflammation-induced depression? J Neural Transm (Vienna) 121:925–932. doi:10.1007/s00702-014-1187-1CrossRefGoogle Scholar
  19. Dantzer R, O'Connor JC, Freund GG, Johnson RW, Kelley KW (2008) From inflammation to sickness and depression: when the immune system subjugates the brain. Nat Rev Neurosci 9:46–56. doi:10.1038/nrn2297CrossRefPubMedPubMedCentralGoogle Scholar
  20. Dent GW, Smith MA, Levine S (1999) The ontogeny of the neuroendocrine response to endotoxin brain. Res Dev Brain Res 117:21–29CrossRefGoogle Scholar
  21. Derkach V, Barria A, Soderling TR (1999) Ca2+/calmodulin-kinase II enhances channel conductance of alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionate type glutamate receptors. Proc Natl Acad Sci U S A 96:3269–3274CrossRefPubMedPubMedCentralGoogle Scholar
  22. Dragicevic N, Bradshaw PC, Mamcarz M, Lin X, Wang L, Cao C, Arendash GW (2011) Long-term electromagnetic field treatment enhances brain mitochondrial function of both Alzheimer's transgenic mice and normal mice: a mechanism for electromagnetic field-induced cognitive benefit? Neuroscience 185:135–149. doi:10.1016/j.neuroscience.2011.04.012CrossRefPubMedGoogle Scholar
  23. Dubreuil D, Jay T, Edeline JM (2003) Head-only exposure to GSM 900-MHz electromagnetic fields does not alter rat's memory in spatial and non-spatial tasks. Behav Brain Res 145:51–61CrossRefPubMedGoogle Scholar
  24. Ehlers MD (2000) Reinsertion or degradation of AMPA receptors determined by activity-dependent endocytic sorting. Neuron 28:511–525CrossRefPubMedGoogle Scholar
  25. Eklind S, Hagberg H, Wang X, Savman K, Leverin AL, Hedtjarn M, Mallard C (2006) Effect of lipopolysaccharide on global gene expression in the immature rat brain. Pediatr Res 60:161–168. doi:10.1203/01.pdr.0000228323.32445.7dCrossRefPubMedGoogle Scholar
  26. Feychting M (2011) Mobile phones, radiofrequency fields, and health effects in children—epidemiological studies. Prog Biophys Mol Biol 107:343–348. doi:10.1016/j.pbiomolbio.2011.09.016CrossRefPubMedGoogle Scholar
  27. Fontainhas AM, Wang M, Liang KJ, Chen S, Mettu P, Damani M, Fariss RN, Li W, Wong WT (2011) Microglial morphology and dynamic behavior is regulated by ionotropic glutamatergic and GABAergic neurotransmission. PLoS One 6:e15973. doi:10.1371/journal.pone.0015973CrossRefPubMedPubMedCentralGoogle Scholar
  28. Frenois F, Moreau M, O'Connor J, Lawson M, Micon C, Lestage J, Kelley KW, Dantzer R, Castanon N (2007) Lipopolysaccharide induces delayed FosB/DeltaFosB immunostaining within the mouse extended amygdala, hippocampus and hypothalamus, that parallel the expression of depressive-like behavior. Psychoneuroendocrinology 32:516–531. doi:10.1016/j.psyneuen.2007.03.005CrossRefPubMedPubMedCentralGoogle Scholar
  29. Gosselin D, Link VM, Romanoski CE, Fonseca GJ, Eichenfield DZ, Spann NJ, Stender JD, Chun HB, Garner H, Geissmann F, Glass CK (2014) Environment drives selection and function of enhancers controlling tissue-specific macrophage identities. Cell 159:1327–1340. doi:10.1016/j.cell.2014.11.023CrossRefPubMedPubMedCentralGoogle Scholar
  30. Gyoneva S, Davalos D, Biswas D, Swanger SA, Garnier-Amblard E, Loth F, Akassoglou K, Traynelis SF (2014) Systemic inflammation regulates microglial responses to tissue damage in vivo. Glia 62:1345–1360. doi:10.1002/glia.22686CrossRefPubMedPubMedCentralGoogle Scholar
  31. Hagberg H, Gressens P, Mallard C (2012) Inflammation during fetal and neonatal life: implications for neurologic and neuropsychiatric disease in children and adults. Ann Neurol 71:444–457. doi:10.1002/ana.22620CrossRefPubMedGoogle Scholar
  32. He K, Song L, Cummings LW, Goldman J, Huganir RL, Lee HK (2009) Stabilization of Ca2+-permeable AMPA receptors at perisynaptic sites by GluR1-S845 phosphorylation. Proc Natl Acad Sci U S A 106:20033–20038. doi:10.1073/pnas.0910338106CrossRefPubMedPubMedCentralGoogle Scholar
  33. Heida JG, Boisse L, Pittman QJ (2004) Lipopolysaccharide-induced febrile convulsions in the rat: short-term sequelae. Epilepsia 45:1317–1329. doi:10.1111/j.0013-9580.2004.13704.xCrossRefPubMedGoogle Scholar
  34. Heneka MT, Kummer MP, Latz E (2014) Innate immune activation in neurodegenerative disease. Nat Rev Immunol 14:463–477. doi:10.1038/nri3705CrossRefPubMedGoogle Scholar
  35. Hirose H, Sasaki A, Ishii N, Sekijima M, Iyama T, Nojima T, Ugawa Y (2010) 1950 MHz IMT-2000 field does not activate microglial cells in vitro. Bioelectromagnetics 31:104–112. doi:10.1002/bem.20532PubMedGoogle Scholar
  36. Hoogland IC, Houbolt C, van Westerloo DJ, van Gool WA, van de Beek D (2015) Systemic inflammation and microglial activation: systematic review of animal experiments. J Neuroinflammation 12:114. doi:10.1186/s12974-015-0332-6CrossRefPubMedPubMedCentralGoogle Scholar
  37. Hristovska I, Pascual O (2016) Deciphering resting microglial morphology and process motility from a synaptic prospect. Front Integr Neurosci 9:73. doi:10.3389/fnint.2015.00073CrossRefPubMedPubMedCentralGoogle Scholar
  38. Incontro S, Ciruela F, Ziff E, Hofmann F, Sanchez-Prieto J, Torres M (2013) The type II cGMP dependent protein kinase regulates GluA1 levels at the plasma membrane of developing cerebellar granule cells. Biochim Biophys Acta 1833:1820–1831. doi:10.1016/j.bbamcr.2013.03.021CrossRefPubMedPubMedCentralGoogle Scholar
  39. Iwasa T, Matsuzaki T, Murakami M, Kinouchi R, Gereltsetseg G, Nakazawa H, Yamamoto S, Kuwahara A, Yasui T, Irahara M (2011) Changes in responsiveness of appetite, leptin and hypothalamic IL-1β and TNF-α to lipopolysaccharide in developing rats. J Neuroimmunol 236(1–2):10–16Google Scholar
  40. Jeong YJ, Kang GY, Kwon JH, Choi HD, Pack JK, Kim N, Lee YS, Lee HJ (2015) 1950 MHz electromagnetic fields ameliorate abeta pathology in Alzheimer's disease mice. Curr Alzheimer Res 12:481–492CrossRefPubMedPubMedCentralGoogle Scholar
  41. Kaplan S, Deniz OG, Onger ME, Turkmen AP, Yurt KK, Aydin I, Altunkaynak BZ, Davis D (2016) Electromagnetic field and brain development. J Chem Neuroanat 75:52–61. doi:10.1016/j.jchemneu.2015.11.005CrossRefPubMedGoogle Scholar
  42. Kettenmann H, Hanisch UK, Noda M, Verkhratsky A (2011) Physiology of microglia. Physiol Rev 91:461–553. doi:10.1152/physrev.00011.2010CrossRefPubMedGoogle Scholar
  43. Klose M, Grote K, Spathmann O, Streckert J, Clemens M, Hansen VW, Lerchl A (2014) Effects of early-onset radiofrequency electromagnetic field exposure (GSM 900 MHz) on behavior and memory in rats. Radiat Res 182:435–447. doi:10.1667/RR13695.1CrossRefPubMedGoogle Scholar
  44. Kondo S, Kohsaka S, Okabe S (2011) Long-term changes of spine dynamics and microglia after transient peripheral immune response triggered by LPS in vivo. Mol Brain 4:27. doi:10.1186/1756-6606-4-27CrossRefPubMedPubMedCentralGoogle Scholar
  45. Konsman JP, Parnet P, Dantzer R (2002) Cytokine-induced sickness behaviour: mechanisms and implications. Trends Neurosci 25:154–159CrossRefPubMedGoogle Scholar
  46. Kumlin T, Iivonen H, Miettinen P, Juvonen A, van Groen T, Puranen L, Pitkaaho R, Juutilainen J, Tanila H (2007) Mobile phone radiation and the developing brain: behavioral and morphological effects in juvenile rats. Radiat Res 168:471–479. doi:10.1667/RR1002.1CrossRefPubMedGoogle Scholar
  47. Kuster N, Torres VB, Nikoloski N, Frauscher M, Kainz W (2006) Methodology of detailed dosimetry and treatment of uncertainty and variations for in vivo studies. Bioelectromagnetics 27:378–391. doi:10.1002/bem.20219CrossRefPubMedGoogle Scholar
  48. Lelli A, Gervais A, Colin C, Cheret C, de Ruiz Almodovar C, Carmeliet P, Krause KH, Boillee S, Mallat M (2013) The NADPH oxidase Nox2 regulates VEGFR1/CSF-1R-mediated microglial chemotaxis and promotes early postnatal infiltration of phagocytes in the subventricular zone of the mouse cerebral cortex. Glia 61:1542–1555. doi:10.1002/glia.22540CrossRefPubMedGoogle Scholar
  49. Leveque P, Dale C, Veyret B, Viart J (2004) Dosimetric analysis of a 900 MHz rat head exposure system. IEE Trans Microw Theory Tech 52:2076–2083CrossRefGoogle Scholar
  50. Li Z, Ma L, Kulesskaya N, Voikar V, Tian L (2014) Microglia are polarized to M1 type in high-anxiety inbred mice in response to lipopolysaccharide challenge. Brain Behav Immun 38:237–248. doi:10.1016/j.bbi.2014.02.008CrossRefPubMedGoogle Scholar
  51. Liu M, Li J, Dai P, Zhao F, Zheng G, Jing J, Wang J, Luo W, Chen J (2015) Microglia activation regulates GluR1 phosphorylation in chronic unpredictable stress-induced cognitive dysfunction. Stress 18:96–106. doi:10.3109/10253890.2014.995085CrossRefPubMedGoogle Scholar
  52. Lopez-Martin E, Relova-Quinteiro JL, Gallego-Gomez R, Peleteiro-Fernandez M, Jorge-Barreiro FJ, Ares-Pena FJ (2006) GSM radiation triggers seizures and increases cerebral c-Fos positivity in rats pretreated with subconvulsive doses of picrotoxin. Neurosci Lett 398:139–144. doi:10.1016/j.neulet.2005.12.082CrossRefPubMedGoogle Scholar
  53. Lu W, Roche KW (2012) Posttranslational regulation of AMPA receptor trafficking and function. Curr Opin Neurobiol 22:470–479. doi:10.1016/j.conb.2011.09.008CrossRefPubMedGoogle Scholar
  54. Lu W, Shi Y, Jackson AC, Bjorgan K, During MJ, Sprengel R, Seeburg PH, Nicoll RA (2009) Subunit composition of synaptic AMPA receptors revealed by a single-cell genetic approach. Neuron 62:254–268. doi:10.1016/j.neuron.2009.02.027CrossRefPubMedPubMedCentralGoogle Scholar
  55. Lu Y, He M, Zhang Y, Xu S, Zhang L, He Y, Chen C, Liu C, Pi H, Yu Z, Zhou Z (2014) Differential pro-inflammatory responses of astrocytes and microglia involve STAT3 activation in response to 1800 MHz radiofrequency fields. PLoS One 9:e108318. doi:10.1371/journal.pone.0108318CrossRefPubMedPubMedCentralGoogle Scholar
  56. Maier SF (2003) Bi-directional immune-brain communication: implications for understanding stress, pain, and cognition. Brain Behav Immun 17:69–85CrossRefPubMedGoogle Scholar
  57. Marino C, Lagroye I, Scarfi MR, Sienkiewicz Z (2011) Are the young more sensitive than adults to the effects of radiofrequency fields? An examination of relevant data from cellular and animal studies. Prog Biophys Mol Biol 107:374–385. doi:10.1016/j.pbiomolbio.2011.09.002CrossRefPubMedGoogle Scholar
  58. Mausset AL, de Seze R, Montpeyroux F, Privat A (2001) Effects of radiofrequency exposure on the GABAergic system in the rat cerebellum: clues from semi-quantitative immunohistochemistry. Brain Res 912:33–46CrossRefPubMedGoogle Scholar
  59. Mausset-Bonnefont AL, Hirbec H, Bonnefont X, Privat A, Vignon J, de Seze R (2004) Acute exposure to GSM 900-MHz electromagnetic fields induces glial reactivity and biochemical modifications in the rat brain. Neurobiol Dis 17:445–454CrossRefPubMedGoogle Scholar
  60. McCusker RH, Kelley KW (2013) Immune-neural connections: how the immune system's response to infectious agents influences behavior. J Exp Biol 216:84–98. doi:10.1242/jeb.073411CrossRefPubMedPubMedCentralGoogle Scholar
  61. Moretti D, Garenne A, Haro E, de Poulletier Gannes F, Lagroye I, Leveque P, Veyret B, Lewis N (2013) In-vitro exposure of neuronal networks to the GSM-1800 signal. Bioelectromagnetics. doi:10.1002/bem.21805
  62. Nittby H, Grafstrom G, Tian DP, Malmgren L, Brun A, Persson BR, Salford LG, Eberhardt J (2008) Cognitive impairment in rats after long-term exposure to GSM-900 mobile phone radiation. Bioelectromagnetics 29:219–232. doi:10.1002/bem.20386CrossRefPubMedGoogle Scholar
  63. Odaci E, Bas O, Kaplan S (2008) Effects of prenatal exposure to a 900 MHz electromagnetic field on the dentate gyrus of rats: a stereological and histopathological study. Brain Res 1238:224–229. doi:10.1016/j.brainres.2008.08.013CrossRefPubMedGoogle Scholar
  64. Oh MC, Derkach VA, Guire ES, Soderling TR (2006) Extrasynaptic membrane trafficking regulated by GluR1 serine 845 phosphorylation primes AMPA receptors for long-term potentiation. J Biol Chem 281:752–758. doi:10.1074/jbc.M509677200CrossRefPubMedGoogle Scholar
  65. Orr AG, Orr AL, Li XJ, Gross RE, Traynelis SF (2009) Adenosine A(2A) receptor mediates microglial process retraction. Nat Neurosci 12:872–878. doi:10.1038/nn.2341CrossRefPubMedPubMedCentralGoogle Scholar
  66. Pall ML (2016) Microwave frequency electromagnetic fields (EMFs) produce widespread neuropsychiatric effects including depression. J Chem Neuroanat 75:43–51. doi:10.1016/j.jchemneu.2015.08.001CrossRefPubMedGoogle Scholar
  67. Petitdant N, Lecomte A, Robidel F, Gamez C, Blazy K, Villegier AS (2016) Cerebral radiofrequency exposures during adolescence: impact on astrocytes and brain functions in healthy and pathologic rat models. Bioelectromagnetics 37:338–350. doi:10.1002/bem.21986CrossRefPubMedGoogle Scholar
  68. Peyman A, Rezazadeh AA, Gabriel C (2001) Changes in the dielectric properties of rat tissue as a function of age at microwave frequencies. Phys Med Biol 46:1617–1629CrossRefPubMedGoogle Scholar
  69. Poulletier de Gannes F, Haro E, Hurtier A, Taxile M, Ruffie G, Billaudel B, Veyret B, Lagroye I (2011) Effect of exposure to the edge signal on oxidative stress in brain cell models. Radiat Res 175:225–230. doi:10.1667/RR2320.1
  70. Qin L, Liu Y, Hong JS, Crews FT (2013) NADPH oxidase and aging drive microglial activation, oxidative stress, and dopaminergic neurodegeneration following systemic LPS administration. Glia 61:855–868. doi:10.1002/glia.22479CrossRefPubMedPubMedCentralGoogle Scholar
  71. Schneider J, Stangassinger M (2014) Nonthermal effects of lifelong high-frequency electromagnetic field exposure on social memory performance in rats. Behav Neurosci 128:633–637. doi:10.1037/a0037299CrossRefPubMedGoogle Scholar
  72. Sekio M, Seki K (2015) Lipopolysaccharide-induced depressive-like behavior is associated with alpha(1)-adrenoceptor dependent downregulation of the membrane GluR1 subunit in the mouse medial prefrontal cortex and ventral tegmental area. Int J Neuropsychopharmacol 18. doi:10.1093/ijnp/pyu005
  73. Shemer A, Erny D, Jung S, Prinz M (2015) Microglia plasticity during health and disease: an immunological perspective. Trends Immunol 36:614–624. doi:10.1016/j.it.2015.08.003CrossRefPubMedGoogle Scholar
  74. Shepherd JD, Huganir RL (2007) The cell biology of synaptic plasticity: AMPA receptor trafficking. Annu Rev Cell Dev Biol 23:613–643. doi:10.1146/annurev.cellbio.23.090506.123516CrossRefPubMedGoogle Scholar
  75. Srinivasan K, Friedman BA, Larson JL, Lauffer BE, Goldstein LD, Appling LL, Borneo J, Poon C, Ho T, Cai F, Steiner P, van der Brug MP, Modrusan Z, Kaminker JS, Hansen DV (2016) Untangling the brain's neuroinflammatory and neurodegenerative transcriptional responses. Nat Commun 7:11295. doi:10.1038/ncomms11295CrossRefPubMedPubMedCentralGoogle Scholar
  76. Stam R (2010) Electromagnetic fields and the blood-brain barrier. Brain Res Rev 65:80–97. doi:10.1016/j.brainresrev.2010.06.001CrossRefPubMedGoogle Scholar
  77. Tay TL, Savage J, Hui CW, Bisht K, Tremblay ME (2016) Microglia across the lifespan: from origin to function in brain development, plasticity and cognition. J Physiol. doi:10.1113/JP272134
  78. Thorlin T, Rouquette JM, Hamnerius Y, Hansson E, Persson M, Bjorklund U, Rosengren L, Ronnback L (2006) Exposure of cultured astroglial and microglial brain cells to 900 MHz microwave radiation. Radiat Res 166:409–421. doi:10.1667/RR3584.1CrossRefPubMedGoogle Scholar
  79. Valdivia A, Duran C, San Martin A (2015) The role of Nox-mediated oxidation in the regulation of cytoskeletal dynamics. Curr Pharm Des 21:6009–6022CrossRefPubMedPubMedCentralGoogle Scholar
  80. Walker FR, Beynon SB, Jones KA, Zhao Z, Kongsui R, Cairns M, Nilsson M (2014) Dynamic structural remodelling of microglia in health and disease: a review of the models, the signals and the mechanisms. Brain Behav Immun 37:1–14. doi:10.1016/j.bbi.2013.12.010CrossRefPubMedGoogle Scholar
  81. Watilliaux A, Edeline JM, Leveque P, Jay TM, Mallat M (2011) Effect of exposure to 1,800 MHz electromagnetic fields on heat shock proteins and glial cells in the brain of developing rats. Neurotox Res 20:109–119. doi:10.1007/s12640-010-9225-8CrossRefPubMedGoogle Scholar
  82. Wes PD, Holtman IR, Boddeke EW, Moller T, Eggen BJ (2016) Next generation transcriptomics and genomics elucidate biological complexity of microglia in health and disease. Glia 64:197–213. doi:10.1002/glia.22866CrossRefPubMedGoogle Scholar
  83. Xu S, Ning W, Xu Z, Zhou S, Chiang H, Luo J (2006) Chronic exposure to GSM 1800-MHz microwaves reduces excitatory synaptic activity in cultured hippocampal neurons. Neurosci Lett 398(3):253–257Google Scholar
  84. Xu S, Zhong M, Zhang L, Zhou Z, Zhang W, Wang Y, Wang X, Li M, Chen Y, Chen C, He M, Zhang G, Yu Z (2010) Exposure to 1800 MHz radiofrequency radiation induces oxidative damage to mitochondrial DNA in primary cultured neurons. Brain Res 1311:189–196CrossRefPubMedGoogle Scholar
  85. Zhang J, Malik A, Choi HB, Ko RW, Dissing-Olesen L, MacVicar BA (2014) Microglial CR3 activation triggers long-term synaptic depression in the hippocampus via NADPH oxidase. Neuron 82:195–207. doi:10.1016/j.neuron.2014.01.043CrossRefPubMedGoogle Scholar

Funding information


Funder Name Grant Number Funding Note
French National Research Program for Environmental and Occupational health of ANSES
  • 2013/2/03 and 2015/2RF/12
Institut National de l'environnement et des risques (INERIS)
  • DRC-13-135967-08716A
program"Investissements d'avenir"
  • ANR-10-IAIHU-06




from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2ts1EAS
via IFTTT

Increased activation of the hippocampus during a Chinese character subvocalization task in adults with cleft lip and palate palatoplasty and speech therapy.

Increased activation of the hippocampus during a Chinese character subvocalization task in adults with cleft lip and palate palatoplasty and speech therapy.: This study aimed to explore brain activation in patients with cleft lip and palate (CLP) using a Chinese character subvocalization task, in which the stimuli were selected from a clinical articulation evaluation test. CLP is a congenital disability. Individuals with CLP usually have articulation disorder caused by abnormal lip and palate structure. Previous studies showed that primary somatosensory and motor areas had a significant difference in activation in patients with CLP. However, whether brain activation was restored to a normal level after palatoplasty and speech rehabilitation is not clear. Two groups, adults after palatoplasty with speech training and age-matched and sex-matched controls, participated in this study. Brain activation during Chinese character subvocalization task and behavioral data were recorded using functional MRI. Patients with CLP responded to the target significantly more slowly compared with the controls, whereas no significant difference in accuracy was found between the groups. Brain activation had similar patterns between groups. Broca's area, Wernicke's area, motor areas, somatosensory areas, and insula in both hemispheres, and the dorsolateral prefrontal cortex and the ventrolateral prefrontal cortex in the right hemisphere were activated in both groups, with no statistically significant difference. Furthermore, the two-sample t-test showed that the hippocampus in the left hemisphere was activated significantly in patients with CLP compared with the controls. The results suggested that the hippocampus might be involved in the language-related neural circuit in patients with CLP and play a role of pronunciation retrieval to help patients with CLP to complete the pronunciation effectively.

(C) 2017 Wolters Kluwer Health | Lippincott Williams & Wilkins




from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2u56mlU
via IFTTT

Effects of Src kinase inhibition on expression of pro-caspase-2 after brain hypoxia

 in a piglet animal model.: Caspase-2 has features of both initiator and effector caspases. Previously, we have shown that brain hypoxia-induced production of caspases 1, 3, 8, and 9 is Src kinase mediated, a nonreceptor intracellular family of kinases. The present study tests the hypothesis that hypoxia results in increased expression of caspase-2 and this effect is mediated by Src kinase. Two to three days old newborn piglets were subjected to normoxia, hypoxia (Hx, FiO2 7%), and Src kinase inhibition (using PP2, 1 mg/kg, intravenous), followed by 30 min of acute hypoxia (Hx+PP2). ATP and phosphocreatine were determined biochemically to verify energy molecule depletion in the hypoxic groups. The cytosolic brain function was isolated and a western blot analysis was carried out using an antibody specific for the caspase-2. The immune-complex band density was expressed as OD/mm2. Caspase-2 expression was increased two-fold in the Hx group. After Src kinase inhibition followed by hypoxia, caspase-2 expression was similar to normoxia levels. We conclude that hypoxia results in increased expression of caspase-2 protein in the cytosolic fraction of the cerebral cortex of the newborn piglets. This increase is mediated by Src kinase.

(C) 2017 Wolters Kluwer Health | Lippincott Williams & Wilkins




from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2u4YIbl
via IFTTT

A magnetoencephalography investigation of coherence source imaging in panic disorder.

A magnetoencephalography investigation of coherence source imaging in panic disorder.: Limbic and frontal structures are largely implicated in panic disorder (PD). Decreased coherence imaging values, as determined by magnetoencephalography (MEG), are suggestive of decreased or inefficient communication among these structures. We have previously demonstrated that coherence source imaging (CSI) values could be similar or higher in some PD patients. The purpose of the current investigation was to replicate these finding in a larger sample. Nine strictly diagnosed PD patients and nine age-matched and sex-matched healthy controls were examined. The CSI-MEG values of 26 frontotemporal regions (FTRs) and 28 extra-frontotemporal regions (ex-FTR; Brodmann areas) were determined for each participant. MEG scans were acquired using a 151-channel whole-head biomagnetometer system. Despite the relatively small sample size, CSI values were significantly lower in a number of FTRs in PD patients. In none of the ex-FTRs (i.e. posterior regions) were there differences between panic and control groups. The above data add to the complexity of understanding the nature of the pathophysiology of PD. Our finding of decreased focal coherence imaging values may reflect decreased excitability in these areas. The preliminary finding could be interpreted as an inhibitory process guarding against the spread of activity in closer hyperexcitable areas as seen in epilepsy. The current data provide evidence for dysfunctional communication within the frontotemporal structures. The findings have implications for the understanding of the neural circuitry underlying PD.

(C) 2017 Wolters Kluwer Health | Lippincott Williams & Wilkins




from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2tw1iKo
via IFTTT

The relationship between cognitive style and event-related potentials during auditory and somatosensory Go/No-go paradigms.

The relationship between cognitive style and event-related potentials during auditory and somatosensory Go/No-go paradigms.: Cognitive style including field dependence/independence (FDI) is an important factor affecting individual personalities. The present study aimed to clarify the relationship between cognitive styles and the characteristics of cognitive processing using event-related potentials. We used the Embedded-Figures Test to assess the cognitive styles [field dependence (FD), field independence (FI)] of the individuals, and individuals performed auditory and somatosensory Go/No-go paradigms. Participants were divided into two groups (FD and FI) on the basis of EFT scores. Reaction times during auditory Go/No-go paradigms were significantly earlier in the FI group than in the FD group, and response variabilities and omission errors were significantly smaller in the FI group than in the FD group. Go-P300 and No-go-P300 amplitudes were significantly larger in the FI group than in the FD group, suggesting that the FI group exhibited greater neural activity for response executive and inhibitory processing. No significant differences were observed in the amplitudes or latencies of auditory N100 and somatosensory N140 components between the FI and FD groups, suggesting that auditory-related and somatosensory-related neural activities were not related to FDI cognitive styles. Our results showed that FDI cognitive styles were related to neural activity for response executive and inhibitory processing.

(C) 2017 Wolters Kluwer Health | Lippincott Williams & Wilkins




from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2u5mm7E
via IFTTT

Dysphagia in Motor Neuron Diseases (MND)

Trends in Research Literature Describing Dysphagia in Motor Neuron Diseases (MND): A Scoping Review:

Abstract

Dysphagia in motor neuron diseases (MNDs) is highly complex, affecting all stages of swallowing and leading to impaired swallowing safety and efficiency. In order to explore the degree to which research is capturing the symptom of dysphagia in MND, we conducted a scoping review of the existing literature. The primary aims of this review were to identify common themes within the literature on dysphagia in MND, explore patterns and trends in research focus, and identify if any imbalances exist between the research themes related to dysphagia description and management. A comprehensive search strategy yielded 1690 unique articles for review. Following relevance screening, a total of 157 articles were included in the synthesis. Relevant data and keywords were extracted from each article and grouped into themes. Frequency estimates were calculated for each theme to identify trends across research literature. Swallowing impairment in MNDs is described in a variety of ways across current research. The most commonly reported theme was Aspiration/Penetration, mentioned in 73.2% of all included articles; a significant imbalance was identified between reports of swallowing safety and efficiency (p = 0.008). The most frequently reported theme related to dysphagia management was Enteral Nutrition, and very few studies have reported on the efficacy of Rehabilitation/Compensatory recommendations. It is suggested that researchers and clinicians remain mindful of imbalances and gaps in research, and aim to characterize dysphagia in MNDs in a comprehensive manner. Further research investigating discrete, measureable changes in swallowing pathophysiology would be beneficial to delineate the key factors contributing to impaired swallowing safety and efficiency.



from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2t7n7Ng
via IFTTT

The clinical impact of 18 F–FDG PET/CT in extracranial pediatric germ cell tumors

The clinical impact of 18 F–FDG PET/CT in extracranial pediatric germ cell tumors:

Abstract



Background

Extracranial germ cell tumors are an uncommon pediatric malignancy with limited information on the clinical impact of 18F–fluorodeoxyglucose (FDG) positron emission tomography/computed tomography (PET/CT) in the literature.




Objective

The purpose of this study was to evaluate and compare the clinical impact on management of 18F–FDG PET/CT with diagnostic computed tomography (CT) in pediatric extracranial germ cell tumor.




Materials and methods

The list of 18F–FDG PET/CT performed for extracranial germ cell tumor between May 2007 and November 2015 was obtained from the nuclear medicine database. 18F–FDG PET/CT and concurrent diagnostic CT were obtained and independently reviewed. Additionally, the patients' charts were reviewed for duration of follow-up and biopsy when available. The impact of 18F–FDG PET/CT compared with diagnostic CT on staging and patient management was demonstrated by chart review, imaging findings and follow-up studies.




Results

During the study period, 9 children (5 males and 4 females; age range: 1.6–17 years, mode age: 14 years) had 11 18F–FDG PET/CT studies for the evaluation of germ cell tumor. Diagnostic CTs were available for comparison in 8 patients (10 18F–FDG PET/CT studies). The average interval between diagnostic CT and PET/CT was 7.2 days (range: 0–37 days). In total, five lesions concerning for active malignancy were identified on diagnostic CT while seven were identified on PET/CT. Overall, 18F–FDG PET/CT resulted in a change in management in 3 of the 9 patients (33%).




Conclusion

18F–FDG PET/CT had a significant impact on the management of pediatric germ cell tumors in this retrospective study. Continued multicenter studies are required secondary to the rarity of this tumor to demonstrate the benefit of 18F–FDG PET/CT in particular clinical scenarios.



from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2snwviH
via IFTTT

Linear Unilateral Basaloid Follicular Hamartoma Following Blaschko's Lines: Two Case Reports and Review of the Literature

Linear Unilateral Basaloid Follicular Hamartoma Following Blaschko's Lines: Two Case Reports and Review of the Literature:

Abstract

Basaloid follicular hamartoma (BFH) is a rare follicular malformation characterized by variable clinical presentations and identical histopathologic features. We present the cases of a 3-month-old boy and an 8-year-old boy with linear unilateral BFH. To the best of our knowledge, only 14 cases of linear unilateral BFH have been described in the English-language literature.


from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2t7cWIJ
via IFTTT

Variability of Delivery of Timolol for the Treatment of Infantile Hemangiomas

Variability of Delivery of Timolol for the Treatment of Infantile Hemangiomas:

Abstract

Background/Objectives

Topical timolol maleate solution or gel-forming solution is used alone or in conjunction with oral propranolol for the treatment of infantile hemangiomas. The consistency of the amount of timolol dispensed has never been evaluated. We evaluated the variability of drug delivery between different brands and formulations of timolol solution and gel-forming solution.

Methods

Five blinded volunteers sequentially dispensed five drops from each of the eight bottles containing timolol 0.5% solution or gel-forming solution. This was repeated three times per user for each bottle. The average amount of timolol dispensed was analyzed according to brand, formulation, and user for variability. The intra- and interuser variability of dispensing both formulations of timolol was also measured.

Results

Our study demonstrates statistically significant differences in the amount of timolol dispensed between timolol solution and gel-forming solution, with the latter closer to manufacturer estimates. Significant differences in the amount of timolol dispensed were noted between users regardless of the formulation or brand. Variability in the amount of timolol dispensed was greater for gel-forming solution than 0.5% solution. Inter- and intrauser variability in the amount of timolol dispensed was greater for gel-forming solution than 0.5% solution.

Conclusion

Statistically significant differences were noted in the amount of timolol dispensed according to formulation, brand, and user. Whether this is clinically significant is unknown given the lack of pharmacokinetic data available for timolol.


from ! Medicine by Alexandros G. Sfakianakis via Αλέξανδρος Σφακιανάκης on Inoreader http://ift.tt/2ssRuvR
via IFTTT

Αναζήτηση αυτού του ιστολογίου

! # Ola via Alexandros G.Sfakianakis on Inoreader