Figura 1. Esquema del número de pacientes reclutados y que hayan finalizado el estudio.

ABSTRACTS PART 3:

Abstracts PhD Programme in Pharmacology and Physiology

e00095  Visfatin/Nampt: A new therapeutic target in the vascular dysfunction associated with ageing and metabolic diseases?

e00096 The noncanonical notch ligand dlk1 regulates renal inflammation.

e00097 Angiotensin-(1-7)/Mas axis attenuates endothelial cell senescence by Nrf2 activation.

e00098 The biological drug anakinra prevents endothelial senescence and vascular smooth muscle cell inflammation elicited by interleukin- 1β.

e00099 Pharmacogenetic algorithm for acenocoumarol dosing improve anticoagulation control: a multicenter randomized clinical trial.

e00100  New chiral melatonin-derivatives as multitarget directed ligands to treat Alzheimer’s disease.

e00101 Generation and potential applications of an X-linked dyskeratosis congenita model in human hematopoietic stem cells.

e00102 Effect of antidepressants of clinical use on neuronal nicotinic acetylcholine receptors.

e00103 Multitarget compounds for the treatment of neurodegenerative diseases: Nrf2-EpRE pathway as key target.

e00104 Oxidative stress is linked to lifetime cardiovascular risk stratification in young/middle age individuals.

e00105 Loss of NLRP6 expression increases the severity of kidney injury.

e00106 PD-L1 is overexpressed on endotoxin tolerant septic patients via HIF1α imparing the adaptive immune response.

e00107 Alterations in the stimulus-secretion coupling related to aging in the murine model of accelerated senescence SAMP8.

e00108 NLRP3 inflammasome inhibition improves motor and behavioral
outcome in a mouse model of traumatic brain injury.

e00109 Control of inflammation by microglial heme-oxygenase- 1 is differentially regulated with aging.

e00110 Neuroprotective effects of new compounds directed to PP2A, a promising therapeutic target for Alzheimer’s disease.

e00111 Galectin-3 and Monocyte Chemoattractant Protein-1, as new biomarkers for patients with diabetes and high risk of cardiovascular diseases.

e00112 Huntington overexpression is associated to altered excitability and exocytosis in chromaffin cells from the R6/1 mouse model of Huntington’s disease.

e00113 Implication of Interleukin-17A in renal dysfunction progression due to arterial pressure changes.

e00114 NLRP3 inflammasome inhibition reduces infarct volume, Blood-Brain-Barrier breakdown and inflammation in cerebral ischemia.

e00115 Bisphenol A induces autophagy and oxidative stress in experimental chronic kidney injury and in tubular cells.

e00116 Clopidogrel response is defined by CYP2C19 metabolizer status in patients undergoing percutaneous neurointervention procedure.

e00117 Role of the mitochondrial Na+/Ca2+ exchanger in NLRP3 inflammasome activation.

e00118 Mitochondria function and morphology alterations precede neurosecretion impairment in chromaffin cells of the SOD1G93A mouse model of amyotrophic lateral sclerosis.

e00119 Vascular damage in obesity associated to pge2 derived mpges-1 through aldosterone/mineralocorticoid-receptor route.

 

Abstracts PhD Programme in Clinical and Health Psychology

e00120 Assessing individual change without knowing the test properties: Item bootstrapping.

e00121 Physical Activity as a Potential Protective Factor in People at Increased Genetic Risk for Alzheimer’s Disease.

e00122 The impact of subjective well-being on mortality.

Other PhD Programmes

e00123 PPRV effects on the differentiation of sheep monocyte-derived dendritic cells.

e00124 Palbociclib radiosensitizes colon and lung cancer cell lines in a p53-dependent manner.

e00125 Erk5 pathway is a new indirect target of sorafenib.

e00126 Mitochondrial activity plays a critical role in multiple myeloma resistance.

e00127 From stem cells to unique neurons: Specification of the Drosophila melanogaster Orcokinin A neurons.

e00127 From stem cells to unique neurons: Specification of the Drosophila melanogaster Orcokinin A neurons.

e00088 A comprehensive study of Trichomonas vaginalis infection: a step forward to understand the pathobiology of isolates from Madrid, Spain.

Abstracts PhD Programme in Pharmacology and Physiology:

e00095
Visfatin/Nampt: A new therapeutic target in the vascular dysfunction associated with ageing and metabolic diseases?.

Ramos-Gonzalez Mariella1,2, Vallejo Susana1,2, San Hipólito-Luengo Alvaro1, Romero Alejandra1, Valencia Inés1, Cercas Elena1,2, Romacho T1, Peiró Concepción1,2, Sánchez-Ferrer CF1,2.

*Corresponding author:
Ramos-Gonzalez Mariella, affiliations, Madrid, Spain. E-mail: Mariella.ramos@inv.uam.es

Details of affiliation

1Universidad Autónoma de Madrid, School of Medicine, Department of Pharmacology and Therapeutics, Arzobispo Morcillo 4, 28029 Madrid, Spain.
2Instituto de Investigación Sanitaria Hospital Universitario de La Paz (IdiPAZ), Madrid, Spain

Funding

Competing Interests:

Keywords: visfatin, IL-1β, endothelial dysfunction.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00095

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Ramos-Gonzalez Mariella, Vallejo Susana, San Hipólito-Luengo Alvaro, Romero Alejandra, Valencia Inés, Cercas Elena, Romacho T, Peiró Concepción, Sánchez-Ferrer CF. Visfatin/Nampt: A new therapeutic target in the vascular dysfunction associated with ageing and metabolic diseases? IBJ Plus 2018 (S2):e00095 doi: 10.24217/2531-0151.18v1s2.00095.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Vascular ageing, obesity, and type 2 diabetes mellitus are associated with high circulating levels of proinflammatory adipokines, as IL-1β or visfatin/nicotinamide phosphoribosyltranferase (Nampt). We investigated whether
the ex vivo treatment and the in vivo infusion of these adipokines in mice can produce vascular alterations, analyzing the
signaling mechanisms involved.

Material and methods: Vascular reactivity. Mesenteric arteries from control mice were pre-incubated ex vivo with
visfatin/Nampt (50 ng/mL) and/or its specific inhibitor FK866 (10 μmol/L), IL-1β (5 ng/mL) and/or IL-1-receptor antagonist
anakinra (100 μg/mL), as well as with apocynin (NADPH oxidase inhibitor, 10 μmol/L), SQ-29,548 (thromboxane A2
receptor antagonist 10 μmol/L) or anakinra (100 μg / mL). Osmotic mini-pumps with visfatin/Nampt (100 ng/kg/day) and/
or FK866 (2.4 mg/kg/day), as well as IL-1β (12 mg/kg/day) were implanted in mice for 7 days. Some mice received also
anakinra (100 mg/kg/day, 3 doses i.p.). Microvessels from mice with visfatin/Nampt infusion were pre-incubated with
apocynin, SQ-29,548 or anakinra. Vascular reactivity was studied by a small vessel myograph, inducing vasoconstriction
with noradrenaline (NA; 3μM), and endothelium-dependent relaxations with cumulative concentrations of acetylcholine
(ACh; 10nM – 10μM). On the other hand, aortic tissues from these animals were homogenized to detect and analyze
NF-κB phosphorylated (p-p65), NF-κΒ (p65) and β-actin protein expression.

Results. Ex vivo treatment with visfatin/Nampt impaired endothelial relaxations in isolated microvessels from control
mice, which was blocked by FK866, apocynin and SQ-29,548. In vivo infusion of visfatin/Nampt induced a similar effect,
which was prevented by FK866, apocynin, SQ-29,548, or anakinra. In vivo IL-1β produced endothelial dysfunction
antagonized by anakinra. NF-kB (p-p65) signaling increased in aortic tissue from mice visfatin/Nampt infusion, which
was reduced in the presence of FK866.

Discussion. In vivo infusion of visfatin/Nampt and IL-1β produce endothelial dysfunction, similar to the produced by the
same adipokines ex vivo, which was mediated by a FK866-sensitive enzymatic activity, involving NADPH oxidase-derived
superoxide anions and thromboxane A2 receptors. The blockade of IL-1 receptors with anakinra abolished the in vivo
effects of visfatin/Nampt but not the ex vivo actions. Visfatin/Nampt produces a FK866-sensitive activation of NF-κB in
the aortic tissue. We conclude visfatin/Nampt is not only a biomarker but also a mediator of the cardiovascular damage
and premature ageing associated to metabolic alterations and, therefore, can become a pharmacological target for the
prevention of those diseases.

e00096
The noncanonical notch ligand dlk1 regulates renal inflammation.

Laura Marquez Exposito1, Carolina Lavoz2, Sandra Rayego Mateos1, Raul Rodrigues Díez3, Marta Fierro Fernández4, Raquel Rodrigues Díez5, Jorge Laborda6, Sergio Mezzano7, Santiago Lamas4, Marta Ruiz Ortega1

*Corresponding author:
Laura Marquez Exposito, Instituto de Investigación Sanitaria – Fundación Jiménez Díaz, Nephrology, Madrid,SPAIN // UAM, Facultad de medicina, Madrid, Spain. E-mail: laura.marqueze@quironsalud.es , laura.marquez@estudiante.uam.es

Details of affiliation

1Instituto de Investigación Sanitaria – Fundación Jiménez Díaz, Nephrology, Madrid,SPAIN// UAM, Facultad de medicina, Madrid, Spain
2Universidad Austral de Chile, Division of Nephrology, School of Medicine, Valdivia, CHILE
3Instituto de Investigación- Hospital universitario La Paz, IDIPAZ, Madrid, SPAIN
4Centro de Biología Molecular Severo Ochoa, Immunology and molecular biology, Madrid, SPAIN
5Universidad Autónoma de Madrid,Pharmacology, Madrid, SPAIN
6University of Castilla La Mancha, Spanish National Research Council (CSIC), Biochemistry and Molecular Biology Branch-Department of Inorganic and
Organic Chemistry and Biochemistry, Albacete, SPAIN
7Universidad Austral de Chile, Nephrology, Valdivia, CHILE

Funding

This work was supported by the Instituto de Salud Carlos III and Fondos FEDER European Union (PI014/0041; Red de
Investigación Renal REDinREN; RD16/0009), Sociedad Española de Nefrologia, Comunidad Autónoma de Madrid (B2017/BMD-3751
NOVELREN-CM).

Competing Interests:

No conflicts of interest.

Keywords: DLK, Jagged, Notch, inflammation, renal damage.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00096

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Laura Marquez Exposito, Carolina Lavoz, Sandra Rayego Mateos, Raul Rodrigues Díez, Marta Fierro Fernández, Raquel
Rodrigues Díez, Jorge Laborda, Sergio Mezzano, Santiago Lamas, Marta Ruiz Ortega. The noncanonical notch ligand dlk1 regulates
renal inflammation. IBJ Plus 2018 (S2):e00096 doi: 10.24217/2531-0151.18v1s2.00096.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

INTRODUCTION AND AIMS: The main goal of this work is to study the role of the notch signaling pathway non-canonical
ligand DLK1 in experimental renal damage. Notch signaling pathway is highly activated during embryonic development,
but it is inhibited in adult tissues. DLK1 is suggested to be a Notch pathway endogenous inhibitor in vitro; however, there
are no studies about its role in vivo.

METHODS: Progressive renal damage model of unilateral ureteral obstruction was done in wild-type and dlk1-null mice
of SvJ-129 genetic background. These animals were sacrificed after 2, 5, 10 and 14 days of obstruction. Left kidney was
obstructed, whilst right kidney was used as control (contralateral).

RESULTS: Non-canonical ligands DLK1 and DLK2 (DLK1 homolog) were analysed. From 5 days of obstruction, both
genes increased their expression in WT vs contralateral (dlk1) and in obstructed kidneys of dlk1-null mice vs WT (dlk2).
Obstructed kidneys of dlk1-null mice presented an increase in NICD, fragment of Notch receptor which is translocated
to the nucleus and activates the effector genes, hes and hey. Moreover, a significant increase of hes-1 gene expression
levels was observed in obstructed kidneys of dlk1-null mice when compared to obstructed WT kidneys at 14 days, as well
as hey- 1 at 5 days. The evaluation of renal damage through PAS tinction revealed a significant increase in inflammatory
infiltrate as focal aggregates in obstructed kidneys of transgenic mice at 14 days. These aggregates were associated to
a significative increase of CD3+, CD4+, F4/80+ infiltrating cells and neutrophils, as well as Th17 lymphocytes. Studying
the possible inflammatory mechanism, it was observed that p-IκBα was increased in damaged kidneys of dlk1-null mice
when compared to their littermates at 14 days. In addition, a significant increase in ccl-2 gene expression was observed
in obstructed kidneys of transgenic mice from 10 days, following 14 days. It is important to remark the increase of the
Th17 response in these damaged kidneys of dlk1-null mice when compared to the WT ones, as we demonstrated here by
an augment of IL17A renal production and the transcription factors that are implicated in this immune response: RORγt
and STAT3.

CONCLUSIONS: The deletion of the non-canonical ligand DLK1 from Notch pathway involves the overactivation of this
pathway in a renal experimental damage. This confirms that DLK1 acts as an endogenous antagonist on Notch receptor in
pathological processes in the kidney. Notch activation in dlk1 absence is associated in an increase of renal inflammatory
infiltrate and in an activation of Th17 immune response, demonstrating the importance of Notch pathway in renal
inflammatory processes.

e00097
Angiotensin-(1-7)/Mas axis attenuates endothelial cell senescence by Nrf2 activation.

Romero A1, San Hipólito A1, Ramos-Gonzalez M1, Villalobos LA1, Romacho T1 , Vallejo S1, Cercas E1, Valencia I1, Sanz Mj2, Erusalimsky JD3, Sánchez-
Ferrer CF1, Peiró C1.

*Corresponding author:
Alejandra Romero Martínez, Department of pharmacology, Universidad Autónoma de Madrid, Madrid, Spain.
E-mail: alejandraromero1993@hotmail.es

Details of affiliation

1Department of Pharmacology, School of Medicine, Universidad Autónoma de Madrid, Madrid, Spain.
2Department of Pharmacology, School of Medicine, Universidad de Valencia, Valencia, Spain.
3Cardiff School of Health Sciences, Cardiff Metropolitan University, Cardiff, United Kingdom.

Funding

Competing Interests:

Angiotensin-(1-7), endothelium, senescence.

Keywords: DLK, Jagged, Notch, inflammation, renal damage.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00097

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Romero A, San Hipólito A, Ramos-Gonzalez M, Villalobos LA, Romacho T, Vallejo S, Cercas E, Valencia I, Sanz Mj, Erusalimsky
JD, Sánchez-Ferrer CF, Peiró C. Angiotensin-(1-7)/Mas axis attenuates endothelial cell senescence by Nrf2 activation. IBJ Plus 2018
(S2):e00097 doi: 10.24217/2531-0151.18v1s2.00097.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Endothelial senescence is one of the major mechanisms contributing to vascular ageing, a complex
process associated with vascular inflammation, endothelial dysfunction and atherosclerosis. Angiotensin (Ang)-(1-7) is a
heptapeptide of the renin-angiotensin system (RAS), considered as physiological antagonist of Ang II. Little is known on
the capacity of Ang-(1-7) to protect against vascular ageing. In this study, we tested whether Ang-(1-7) could mitigate the
senescence of cultured human umbilical vein endothelial cells (HUVEC) induced by Ang II or interleukin (IL)-1. We further
aimed to identify protective cellular pathways activated by Ang-(1-7), with particular focus on the antioxidant and antiinflammatory
nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2)/heme oxygenase (HO)-1 axis.

Methods: Cultured HUVEC were stimulated with Ang II (100 nM) or IL-1β (2,5 ng/ml) for 18 h. Cell senescence was
quantified by positive senescence-associated β-galactosidase (SA-β-gal+) staining. Vascular cell adhesion molecule-1
(VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) were quantified by flow cytometry, while leukocyteendothelium
adhesion was determined using a flow chamber assay. Nrf2 and HO-1 were determined by Western blot.

Results: Both Ang II and IL-1β enhanced the fraction of SA-β-gal+ cells, together with increased expression of ICAM-1
and VCAM-1, resulting in a higher leukocyte adhesion to HUVEC monolayers. Ang-(1-7) (100 nM) attenuated all these
actions through a mechanism that was prevented by the Mas receptor antagonist A779 (1 μM). Ang-(1-7) enhanced
Nrf2 and HO-1 levels. Indeed, the Nrf2 activator sulforaphane (1 μM) mimicked the effects of Ang-(1-7) on the SA-β-gal+
cells fraction. Interestingly, the HO-1 inhibitor Sn protoporphyrin (1 μM) dampened the anti-senescence action of both
Ang- (1-7).

Conclusions: Ang-(1-7) counteracts endothelial cell senescence triggered by both RAS-dependent and -independent
stimuli. Nrf2/HO-1 pathway seems to be on the basis of these protective properties of Ang-(1-7). Overall, the Ang-(1-7)/
Mas axis arises as a novel pharmacological tool to attenuate endothelial senescence and vascular aging.

e00098
The biological drug anakinra prevents endothelial senescence and vascular smooth muscle cell inflammation elicited by interleukin-
1β.

Álvaro San Hipólito-Luengo1, Alejandra Romero1, Mariella Ramos-González1, Inés Valencia1, Susana Vallejo1, Tania Romacho1, Elena Cercas1, Carlos F. Sánchez-Ferrer1, Concepción Peiró1

*Corresponding author:
Álvaro San Hipólito-Luengo, Department of Pharmacology, Universidad Autónoma de Madrid, Madrid, Spain. E-mail: alvaro.sanhipolito@uam.es

Details of affiliation

1Department of Pharmacology, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.

Funding

Competing Interests:

Keywords: IL-1β, Endothelial senescence, Inflammation

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00098

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Álvaro San Hipólito-Luengo, Alejandra Romero, Mariella Ramos-González, Inés Valencia, Susana Vallejo, Tania Romacho, Elena
Cercas, Carlos F. Sánchez-Ferrer, Concepción Peiró. The biological drug anakinra prevents endothelial senescence and vascular smooth
muscle cell inflammation elicited by interleukin-1β. IBJ Plus 2018 (S2):e00098 doi: 10.24217/2531-0151.18v1s2.00098.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Vascular complications are the main cause of mortality in patients suffering from type 2 diabetes mellitus
(T2DM). In these patients, vascular damage is associated with enhanced inflammation and premature vascular ageing,
among other. The cytokine interleukin (IL)-1β is also an adipokine overexpressed by the adipose tissue in the context of
T2DM. Our goal was to determine the direct impact of IL-1β on endothelial cell senescence and vascular smooth muscle
cell inflammation and to pharmacological interfere with the action of the cytokine.

Materials and Methods: In this study, we used cell cultures of human aortic smooth muscle cells (HASMC) and human
umbilical vein endothelial cells (HUVEC). The Griess method and a lucigenin-derived chemiluminescence assay were
used to determine the activation of NF-κB and NADPH oxidase in HASMCs, respectively, while the levels of iNOS were
determined by western blot. Regarding HUVECs, endothelial senescence was determined using a β-galactosidase staining
kit, while a double indirect immunofluorescence was used to determine total and telomeric DNA damage.

Results: IL-1β (2.5 ng/ml) promoted cell senescence in human umbilical vein endothelial cells (HUVEC), as determined by
the number of cells exhibiting positive senescence-associated β-galactosidase staining (SA-β-gal+). This was accompanied
by increased total and telomeric DNA damage. The biological drug anakinra (0.01 to 1 μg/ml) was able to prevent the
increase in SA-β-gal+ elicited by IL-1β in a concentration-dependent manner.
Moreover, IL-1β (10 ng/ml) promoted the activation of human vascular smooth muscle cell (HASMC) in terms of NADPH
oxidase and NF-κB activity and inducible nitric oxide synthase expression. In a high glucose environment (22 mM vs 5.5
mM) the pro-inflammatory cascade activated by IL-1β was significantly exaggerated. Anakinra 1 μg/ml) prevented not
only the pro-inflammatory action of IL-1β but also the exacerbation observed under high glucose conditions.

Conclusion: IL-1β may be a direct player in promoting vascular damage associated to T2DM by favoring vascular ageing
and inflammation, which is turn potentiated by high glucose concentrations. Biological IL-1β blockers, such as anakinra,
may be useful as pharmacological tools to treat or delay vascular complications.

e00099
Pharmacogenetic algorithm for acenocoumarol dosing improve anticoagulation control: a multicenter randomized clinical trial.

H.Y. Tong1, A. M. Borobia1,2, M.A. Rodriguez Dávila1, N. Ruiz-Giménez3, A. Lorenzo1, M. González Viñolis1, O. Madridano4, S. Fabra1, A. López Parra5, E. Arroyo Pardo5, C. Baeza5, P. Llamas Sillero6, A. Carcas Sansuán1,2 and PGX-ACE Investigators Group.

*Corresponding author:
Hoi Y. Tong. Clinical Pharmacology Department. La Paz University Hospital. Madrid (SPAIN). hoi.tong@idipaz.es

Details of affiliation

1La Paz University Hospital, IdiPAZ, Madrid, Spain
2School of Medicine. Autonomous University de Madrid, Madrid, Spain.
3La Princesa University Hospital, Madrid, Spain.
4Infanta Sofía University Hospital, Madrid, Spain.
5Complutense University of Madrid, Madrid, Spain
6Fundación Jiménez Díaz University Hospital, Madrid, Spain

Funding

The Ministry of Heatlh (project TRA-010).

Competing Interests:

Antonio J. Carcas is the Editor-in-Chief of Ibjournals.com and Alberto M. Borobia is the executive deputy editor.

Keywords: acenocoumarol; pharmacogenetics, clinical trial

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00099

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: H.Y. Tong, A. M. Borobia, M.A. Rodriguez Dávila, N. Ruiz-Giménez, A. Lorenzo, M. González Viñolis, O. Madridano, S. Fabra,
A. López Parra, E. Arroyo Pardo, C. Baeza, P. Llamas Sillero, A. Carcas Sansuán, and PGX-ACE Investigators Group. Pharmacogenetic
algorithm for acenocoumarol dosing improve anticoagulation control: a multicenter randomized clinical trial. IBJ Plus 2018 (S2):e00099
doi: 10.24217/2531-0151.18v1s2.00099.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Background: It has been demonstrated that there is a strong association between genetic polymorphisms and
acenocoumarol dose requirements. Our group designed and validated a pharmacogenetic dosing algorithm for
acenocoumarol, indicated in patients with thromboembolic disease who are going to start treatment with this drug.

Material and methods: The design of this multicenter, single blind, randomized clinical trial has been published (Trials,
2012;13:239; PMID: 23237631). Patients were randomized to one of the two arms: common clinical practice or following
an individualized pharmacogenetic algorithm. The main endpoint was: percentage of patients with INR within the
therapeutic range on day 7 after initiation of acenocoumarol treatment. The variables included in the algorithm were:
demographic, clinical and pharmacogenetic variables (VKORC1, CYP2C9, CYP4F2 and ApoE). The follow-up was three
months since the beginning of acenocoumarol treatment.

Results: 149 patients were recruited, (144 patients conformed the ITT population). Mean age was 59.42 (±18.53) years
old and 77 (55.5%) were males. In the experimental group, 34 of 72 patients (47.22%) and 14 of 64 patients (21.8%) in
the control group had INR value within therapeutic range (p=0.023). No statistically significant differences were found in
the time to achieve therapeutic INR values (p=0.286) or in the INR mean during the last two months of study (p=0.1796).
When classified by phenotype (low, intermediate and high stable dose requirement) the INR at day 3 and 7 in both
groups were:

Conclusions: Better anticoagulant control was obtained at day 7 of the beginning of the drug with the pharmacogenetic
algorithm. Difference is observed in the value of INR between both groups, being more evident in the groups of extreme
dose requirement.

e00100
New chiral melatonin-derivatives as multitarget directed ligands to treat Alzheimer’s disease.

S. Abril1,2, P. Michalska1,2, I. Buendia2, A.M Briones1, J.C. Menéndez3, M. Salaices Sánchez1, R. León*1,2

*Corresponding author:
Rafael León, Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, 28006, Madrid. E-mail: rafael.leon@inv.uam.es

Details of affiliation

1Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, 28029 Madrid,
Spain E-mail: sheila.abril@uam.es
2Instituto de Investigación Sanitaria del Hospital Universitario de la Princesa, 28006, Madrid, Spain
3Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense
de Madrid, 28040, Madrid, Spain

Funding

We are grateful for financial support from European Commission-ERC, People (Marie Curie Actions) FP7 under REA grant
agreement no. PCIG11-GA-2012-322156; Spanish Ministry of Health (Instituto de Salud Carlos III) (grant PI14/00372) Bayer A.G.,
“From Targets to Novel Drugs” program (grant 2015-03-1282) and Fundación FIPSE (grant12-00001344-15) to R.L.; S.A. and P.M.
thanks MECD for FPU fellowships (FPU14/05224 and FPU13/03737). I.B. thanks MECD for Juan de la Cierva contract. R.L. gratefully
thanks Dr. Wolf for sharing the cellular line AREc32. We also gratefully acknowledge the continued support of Instituto-Fundación
Teófilo Hernando, Madrid, Spain.

Competing Interests:

The authors declare no competing financial interest.

Keywords: Medicinal chemistry, multitarget directed ligands, Alzheimer’s disease

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00100

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: S. Abril, P. Michalska, I. Buendia, A.M Briones, J.C. Menéndez, M. Salaices Sánchez, R. León. New chiral melatonin derivatives
as multitarget directed ligands to treat Alzheimer’s Disease. IBJ Plus 2018 (S2):e00100 doi: 10.24217/2531-0151.18v1s2.00100.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Alzheimer’s disease (AD) is the most prevalent neurodegenerative disease (NDD) with around 35 million
people currently affected worldwide, according to World Health Organization statistics [1]. As there is not an effective
therapy for AD and NDDs, considered as multifactorial diseases [2], the new paradigm of multitarget directed ligands,
seems promising. In this context, we designed our compounds towards control neuro-inflammation and oxidative
stress, by combining the induction of the Nrf2 transcription factor, restoration of cholinergic function and free radical
scavenging effect, which would promote the neuroprotection.

Materials and methods: A seven step convergent-lineal enantioselective synthesis was performed to obtain the new
molecules. The pharmacological studies included evaluation as free radical scavengers using the ORAC test; as Nrf2
inducers employing a luminescent method in the AREc32 cell line; as acetylcholinesterase inhibitors with Ellman’s method,
and as neuroprotective agents against okadaic acid in cell line SH-SY5Y, an in vitro model of Tau hyperphosphorylation.
Results: A library of fifteen compounds has been obtained with very good yields and enantiomeric excesses. All of them
are excellent free radical scavengers and inhibitors of acetylcholinesterase around the 10 μM range. Furthermore, some
of them are Nrf2 inducers and neuroprotectants.

Conclusion: We have obtained a new family of melatonin chiral derivatives, Six out of fifteen compounds showed all the
desired properties to a promising degree. Therefore, we plan to continue and complete their pharmacological profile to
choose our lead compound.

References:
[1] http://www.who.int/mediacentre/factsheets/fs362/en/
[2] A. Cavalli et al. J. Med. Chem. 2008, 51, 347- 372

e00101
Generation and potential applications of an X-linked dyskeratosis congenita model in human hematopoietic stem cells.

Carrascoso-Rubio C.1,2, Zittersteijn H.A.1,2, Pintado-Berninches L.4, Fernández-Varas B.3, Lozano ML.1,2, Manguan-Garcia C.3, Sastre L.3, Bueren J.A.1,2, Perona R.3†*, Guenechea G.1,2†*

*Corresponding authors: Guillermo Güenechea Amurrio, PhD (g.guenetxea@ciemat.es), Division of Hematopoietic Innovative Therapies, CIEMAT/
CIBERER/IIS-FJD, Avenida Complutense, 40; 28040; Madrid (Spain) and Rosario Perona Abellón, Professor (rperona@iib.uam.es), Instituto de
Investigaciones Biomédicas Alberto Sols, CSIC7UAM, Calle Arturo Duperier, 4; 28029; Madrid (Spain).
†These authors supervised equally this work.

Details of affiliation

1Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas (CIEMAT) and Centro de
Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII). Madrid, 28040, Spain
2Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD/UAM). Madrid, 28040, Spain.
3Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM) and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERERISCIII).
Madrid, 28029. Spain.
4Instituto de Investigaciones Biomédicas Alberto Sols (CSIC/UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER-ISCIII)
and Advanced Medical Projects SL (AMP). Madrid, 28029. Spain.

Funding

This work was supported by grants from “Ministerio de Economía, Comercio y Competitividad y Fondo Europeo de
Desarrollo Regional (FEDER)” (SAF2015-68073-R) and CIBERER is an initiative of the “Instituto de Salud Carlos III” and “Fondo Europeo
de Desarrollo Regional (FEDER)”.

Competing Interests:

The authors declare no competing financial interest.

Keywords: Dyskeratosis congenita, bone marrow failure syndromes, hematopoietic stem cells, short hairpin RNA and DKC1 gene.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00101

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Carrascoso-Rubio C, Zittersteijn HA, Pintado-Berninches L, Fernández-Varas B, Lozano ML, Manguan-Garcia C, Sastre L,
Bueren JA, Perona R, Guenechea, G. Generation and potential applications of an X-linked dyskeratosis congenita model in human
hematopoietic stem cells. IBJ Plus 2018 (S2):e00101 doi: 10.24217/2531-0151.18v1s2.00101.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: X-linked dyskeratosis congenita (X-DC) is a low prevalent inherited bone marrow failure syndrome caused
by mutations in the DKC1 gene. This gene encodes for the dyskerin nucleolar protein, which is part of the telomerase
complex. These mutations impair telomerase activity leading to premature telomere length attrition. For this reason X-DC
is also classified as a telomere biology disorder. The major cause of premature death in 80% of the DC patients is bone
marrow failure (BMF). To date, the only curative treatment for BMF is hematopoietic stem cell (HSC) transplantation.
However, the difficulties to find compatible donors, risks derived from conditioning regimes and graft versus host disease
postulate that gene therapy may constitute a promising alternative in treating DC patients.

Materials and methods: Due to the complications related to the use of primary HSCs from DC patients for experimental
studies, this work is focused on the generation of X-DC-like CD34+ cells by lentiviral delivery of DKC1 short hairpin RNAs
(shRNAs) to knock down the expression of dyskerin of umbilical cord blood derived HSCs.

Results: Three shRNAs were selected, among a library of seven shRNAs, based on the efficacy to inhibit DKC1 gene
expression. Interfered CD34+ cells showed a downregulated TERC expression, a reduced telomerase activity, a decreased
cell expansion, as well as an impaired clonogenic and hematopoietic reconstitution potential in NSG mice. Moreover,
an upregulation in p21 expression was observed in DKC1-interfered CD34+ cells, consistent with an increased rate of cell
senescence induction and DNA damage.

Conclusions: Development of X-DC-like CD34+ cells will facilitate the understanding of the HSC defects characteristic of
X-DC and contribute to the development of new therapeutic strategies for the treatment of the BMF in X-DC patients.

e00102
Effect of antidepressants of clinical use on neuronal nicotinic acetylcholine receptors.

Isabel Gameiro-Ros1, Carmen Nanclares1, Andrés M. Baraibar1, Alicia Muñoz-Montero1, Iris Álvarez-Merz1, Inés Colmena1, Jesús Miguel Hernández- Guijo1, Luis Gandía1*.

*Corresponding author:
Luis Gandía, Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, C/
Arzobispo Morcillo, 4, 28029, Madrid, Spain. E-mail: luis.gandia@uam.es

Details of affiliation

1Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid

Funding

SAF 2016-78892-R.

Competing Interests:

No competing interests are declared

Keywords: depression, antidepressant, cholinergic theory, neuronal nicotinic acetylcholine receptor, chromaffin cell.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00102

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Isabel Gameiro-Ros, Carmen Nanclares, Andrés M. Baraibar, Alicia Muñoz-Montero, Iris Álvarez-Merz, Inés Colmena, Jesús
Miguel Hernández-Guijo, Luis Gandía. Effect of antidepressants of clinical use on neuronal nicotinic acetylcholine receptors. IBJ Plus
2018 (S2):e00102 doi: 10.24217/2531-0151.18v1s2.00102.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Depression is the most common affective disorder and the main cause of disability. The current
treatment of this pathology is based on the monoaminergic theory, which attributes its origin to a deficit in monoamine
neurotransmitters, mainly serotonin and noradrenaline. Despite current antidepressants are effective, a 30% of patients
do not respond to this therapeutic strategy. Besides, although monoamine levels are increased after a few days of
treatment, the onset of the therapeutic effect takes at least two weeks to appear. To shed light on these questions, we
based our work in the alternative “cholinergic theory” of depression, that considers the overactivity of the cholinergic
system observed in depression as one of its main causes. Thus, we wanted to exploit this field by studying the effect of
several antidepressants on cholinergic neurotransmission, to determine whether they possess an additional mechanism
of action.

Materials and methods: For this study, five antidepressants, one from each group of current clinical use was selected.
Their effect on nAChRs was studied on bovine chromaffin cells (BCCs), that secrete catecholamines upon physiological
nAChR activation by ACh. In this experimental model we assessed the effect of these compounds on: the secretion of
catecholamines measured by amperometry in cell populations, the intracellular calcium levels measured by fluorescence
in cell populations, and the nicotinic and calcium currents in single cell measured by patch-clamp techniques.

Results: Some of the antidepressants blocked the catecholamine release in BCCs populations when they were
physiologically stimulated with ACh, but not with a depolarizing solution of high potassium. Besides, the calcium entry
in BBCs upon ACh stimulation was blocked in their presence in a concentration dependent manner, an effect that was
not observed when they were stimulated with high potassium. Finally, some of the studied compounds were able to
concentration-dependently block the nicotinic inward current elicited by ACh in single BCCs, while their effect on calcium
currents, that would be related to an activity on voltage-dependent calcium channels (VDCCs), cannot be related to an
interaction with subtypes of these channels involved in catecholamine secretion.

Conclusions: These results indicate that some of the studied antidepressants are able to specifically block the nAChRs in
BCCs, with no effect on VDCCs involved in catecholamine secretion, and thus conferring them an additional mechanism of
action. Our study might help to a better understanding of the therapeutic effect of these antidepressants, and eventually
contribute to the development of more effective drugs for depression.

e00103
Multitarget compounds for the treatment of neurodegenerative diseases: Nrf2-EpRE pathway as key target.

Patrycja Michalska1, 2, Izaskun Buendia1,2, Pablo Duarte1, 2, Enrique Luengo1, Cristina Fernández-Mendívil1, Jose A. Morales3, Ana Pérez Castillo3, Manuela García-López1, Rafael León1, 2

*Corresponding author:
Rafael León, rafael.leon@inv.uam.es

Details of affiliation

1Instituto Teófilo Hernando y Departamento de Farmacología y Terapéutica, Facultad de Medicina. Universidad Autónoma de Madrid, 28029 Madrid,
España
2Instituto de Investigación Sanitaria, Servicio de Farmacología Clínica, Hospital Universitario de la Princesa, 28006 Madrid, España
3 Instituto de Investigaciones Biomédicas “Alberto Sols”, Universidad Autónoma de Madrid, 28029 Madrid, España

Funding

IS Carlos III (Contrato Miguel Servet II a RL, proyecto CPII16/00014 y proyecto PI14/00372 a RL),). Ministerio de Educación, Cultura y Deporte (MECD) FPU13/03737 a P.M, FPU16/03977 a P.D.

Competing Interests:

None declared

Keywords: Nrf2-EpRE, oxidative stress, neuroinflammation, multitarget compounds.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00103

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Patrycja Michalska, Izaskun Buendia, Pablo Duarte, Enrique Luengo, Cristina Fernández-Mendívil, Jose A. Morales, Ana Pérez
Castillo, Manuela García-López, Rafael León. Multitarget compounds for the treatment of neurodegenerative diseases: Nrf2-EpRE
pathway as key target. IBJ Plus 2018 (S2):e00103 doi: 10.24217/2531-0151.18v1s2.00103.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Neurodegenerative diseases (NDDs) share several pathophysiological mechanisms such as presence of
aberrant protein aggregates, mitochondrial dysfunction, oxidative stress and neuroinflammation. Given the role of
these factors in the development of NDDs, compounds able to regulate them simultaneously are of great interest.At
physiological conditions, the main route to combat oxidative stress and neuroinflammation is the Nrf2/EpRE pathway.
Besides, it has been observed, that the Nrf2 factor is deregulated in neurodegenerative disorders1. Based on this
hypothesis we carried out the design, synthesis and pharmacological evaluation of a new family of Nrf2 inducers with
complementary activities for the treatment of NDDs2.

Materials and methods: The biological evaluation of the compounds was carried out by the use of bioluminescence
and fluorescence techniques, to determine the Nrf2 inducing and antioxidant capacity of the compounds, respectively.
Also, its capacity for neuroprotection in cell lines and immunomodulatory capacity in primary glia cultures was studied.
Besides, inmunofluorescence techniques were used to study the pharmacological properties.

Results: Newly developed compounds were able to induce the Nrf2-EpRE pathway, and also demonstrated a scavenger
and anti-inflammatory effect. Besides the compounds showed an interesting neuroprotective effect against in vitro and
ex vivo model models of oxidative stress and toxicity induced by protein aggregation.

Conclusions: Compounds targeting the Nrf2-EpRE pathway present interesting properties for the treatment of
neurodegenerative diseases. Altogether, results obtained indicate these compounds should be further evaluated in in
vivo models of NDDs.
(1) Ramsey, et al. J. Neuropathol. Exp. Neurol. 2007, 66: 75–85.
(2) Buendia, et al. Future Med. Chem. 2015, 15: 1961-9.

e00104
Oxidative stress is linked to lifetime cardiovascular risk stratification in young/middle age individuals.

Elena Rodríguez-Sánchez1, José Alberto Navarro-García1, Jennifer Aceves-Ripoll1, Gloria Álvarez-Llamas2, Eva Calvo3, Martha Cabrera3, María G. Barderas4, Luis M. Ruilope1,5, Gema Ruiz-Hurtado1,5

Details of affiliation

1Laboratorio Traslacional Cardiorenal. Instituto de Investigación imas12. Hospital Universitario 12 de Octubre, Madrid
2Departamento de Inmunología, IIS-Fundación Jiménez Díaz, Madrid
3Ibermutuamur, Madrid
4Departamento de Fisiopatología Vascular, Hospital Nacional de Parapléjicos, SESCAM, Toledo
5Unidad de Hipertensión. Instituto de Investigación imas12. Hospital 12 de Octubre, Madrid.

Funding

Competing Interests:

Keywords:

Abstract

NOT AVAILABLE AT THIS MOMENT

e00106
Loss of NLRP6 expression increases the severity of kidney injury.

Lara Valiño-Rivas1,2, Leticia Perez-Cuarental1,2, Gabriel Nuñez3, Ana B Sanz1,2, Alberto Ortiz1,2, Maria Dolores Sanchez-Niño1,2.

*Corresponding author:
Lara Valiño-Rivas, IIS-Fundación Jimenez Diaz-Universidad Autónoma de Madrid and Fundación Renal Íñigo Alvarez de Toledo-IRSIN, Madrid, Spain
E-mail: lara.valino@fjd.es

Details of affiliation

1IIS-Fundación Jimenez Diaz-Universidad Autónoma de Madrid and Fundación Renal Íñigo Alvarez de Toledo-IRSIN, Madrid, Spain
2REDINREN, Madrid, Spain
3Department of Pathology, University of Michigan Medical School, Ann Arbor

Funding

PI15/00288, PI16/02057, ISCIII-RETIC REDINREN RED16/009.

Competing Interests:

Salary support: PFIS FI14/00398.

Keywords:

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00105

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Lara Valiño-Rivas, Leticia Perez-Cuarental, Gabriel Nuñez, Ana B Sanz, Alberto Ortiz, Maria Dolores Sanchez-Niño. Loss of
NLRP6 expression increases the severity of kidney injury. IBJ Plus 2018 (S2):e00105 doi: 10.24217/2531-0151.18v1s2.00105.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Nlrp6 is a nucleotide-binding oligomerization domain-like receptor (NLR) that forms atypical inflammasomes. Nlrp6
protects the gut epithelium, regulating the interaction with microbiota. However, the expression and function of Nlrp6
in the kidney, a sterile environment, have not been characterized.

In a transcriptomics array of murine acute kidney injury (AKI) induced by a folic acid overdose, Nlrp6 and Naip3 were the
only significantly downregulated NLR genes (fold-change 0.55 and 0.80, respectively). The functional implications of Nlrp6
downregulation were explored in mice and in cultured murine tubular cells. Nlrp6 was expressed by the healthy murine
and human kidney tubular epithelium, and expression was reduced in mice during AKI induced by a folic acid overdose
and in chronic kidney injury induced by unilateral ureteral obstruction. Low kidney Nlrp6 was observed in human kidney
injury. Genetic Nlrp6 deficiency resulted in upregulation of kidney ERK1/2 and p38 MAP kinase phosphorylation, more
severe AKI and more severe kidney fibrosis in mice. Nlrp6 downregulation induced by specific siRNA in cultured tubular
cells resulted in upregulation of ERK1/2 and p38 phosphorylation and chemokine mRNA expression and downregulation
of the nephroprotective gene Klotho. MAP kinase inhibition prevented the inflammatory response in Nlrp6-deficient
cells.

In conclusion, Nlrp6 has a role in suppressing sterile inflammation during kidney injury.

e00106
PD-L1 is overexpressed on endotoxin tolerant septic patients via HIF1α imparing the adaptive immune response.

José Avendaño-Ortiz1,2,3, Charbel Maroun-Eid4, Alejandro Martín-Quirós4, Víctor Toledano1,2,3, Carolina Cubillos-Zapata1,2,3, Aníbal Varela-Serrano1,2,3,
Emilio Llanos-Gonzalez1,2, Roberto Lozano-Rodríguez1,2, Enrique Álvarez5, Luis A. Aguirre1,2,3, Enrique Hernández-Jiménez1,2,3, and Eduardo López-
Collazo1,2,3.

*Corresponding author:
Eduardo López-Collazo. Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain.
E-mail: elopezc@salud.madrid.org

Details of affiliation

1Innate Immunity Group, IdiPAZ, La Paz University Hospital, Madrid, Spain.
2Tumor Immunology Lab, IdiPAZ, La Paz University Hospital, Madrid, Spain.
3Center for Biomedical Research Network, CIBEres
4Emergency Department, IdiPAZ, La Paz University Hospital, Madrid, Spain.
5EMPIREO, Madrid, Spain.

Funding

This work was supported by grants from the “Instituto de Salud Carlos III” (ISCiii), “Fondos de Investigaciónes Sanitarias” (FIS)
and FEDER (PI14/01234 and PIE15/00065) to ELC and grant from “Comunidad de Madrid”PEJ15/BIO/AI-0021 to JAO.

Competing Interests:

All authors: No reported conflicts of interests.

Keywords: Monocytes, sepsis, PD-L1,HIF α, endotoxin tolerance

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00106

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: José Avendaño-Ortiz, Charbel Maroun-Eid, Alejandro Martín-Quirós, Víctor Toledano, Carolina Cubillos-Zapata, Aníbal
Varela-Serrano, Emilio Llanos-Gonzalez, Roberto Lozano-Rodríguez, Enrique Álvarez, Luis A. Aguirre, Enrique Hernández-Jiménez,
and Eduardo López-Collazo. PD-L1 is overexpressed on endotoxin tolerant septic patients via HIF1α imparing the adaptive immune
response. IBJ Plus 2018 (S2):e00106 doi: 10.24217/2531-0151.18v1s2.00106.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Background: Endotoxin tolerance (ET) is an important feature of sepsis-associated immunosuppresion, however, a clear
underlaying mechanism is still lacking and its implication in the outcome of patients is controversial, as some authors
suggest it to be protective and others to be detrimental.

Materials and methods: We performed a prospective study classifying at admission 48 patients with sepsis into 3
subgroups according to their ex vivo response to lipopolysaccharide. We evaluated the differences between groups
in both clinical parameters and immune response assays. To check the endotoxin tolerance implication in these
observations, we studied these immune features in a well stablished in vitro model.

Results: We observed a worse APACHE II, qSOFA and clinical outcome in the non-responder/tolerant subgroup.
Futhermore, the monocyte PD-L1 expression was upregulated in these patients corresponding with reduced T lymphocyte
proliferation. PD-L1 knockdown on monocytes and PD-1 blocking assays reverted the impaired T cell response. HIF1α,
which was showed to be overexpressed under ET, controlled PD-L1 expression on septic patients’ and in the in vitro
model.

Conclusions. These findings give a new role of HIF1α explanation controlling PD-L1 overexpression observed in sepsis
patients. Moreover, classifying patients according to the ex vivo lipopolysaccharide response, could be considered an
interesting field of study directed toward a precision medicine and more personalized therapies in sepsis.

e00107
Alterations in the stimulus-secretion coupling related to aging in the murine model of accelerated senescence SAMP8.

Andrés M. Baraibar1, Carmen Nanclares1, Inés Colmena1, Isabel Gameiro-Ros1, Iris Álvarez-Merz1, Alicia Muñoz-Montero1, Jesús M. Hernández-
Guijo1, Luis Gandía1..

*Corresponding author:
Luis Gandía, Instituto Teófilo Hernando, Madrid, Spain. E-mail: luis.gandia@uam.es

Details of affiliation

1Instituto Teófilo Hernando, Department of Pharmacology and Therapeutics, Faculty of Medicine (Universidad Autónoma de Madrid), Madrid, Spain.

Funding

SAF2013-44108-P and SAF2016-78892-R.

Competing Interests:

No competing interests are declared.

Keywords: exocytosis, chromaffin cell, aging, Alzheimer

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00107

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Andrés M. Baraibar, Carmen Nanclares, Inés Colmena, Isabel Gameiro-Ros, Iris Álvarez-Merz, Alicia Muñoz-Montero, Jesús
M. Hernández-Guijo, Luis Gandía. Alterations in the stimulus-secretion coupling related to aging in the murine model of accelerated
senescence SAMP8. IBJ Plus 2018 (S2):e00107 doi: 10.24217/2531-0151.18v1s2.00107.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: The nervous system is especially vulnerable to aging. Its vulnerability is manifested by the existence of
neurodegenerative pathologies like Alzheimer’s disease (AD), Parkinson’s disease (PD) or Amyotrophic Lateral Sclerosis
(ALS). During these diseases, alterations of neurotransmitter systems has been reported. Although, numerous changes
can also be observed in many individuals during non-pathological aging. Our working hypothesis suggests that,
with the progression of age, alterations in the stimulus-secretion coupling can occur, compromising the release of
neurotransmitters and causing cognitive deficits.

Materials and Methods: In this work, mice of the senescence-prone strain 8 (SAMP8) have been used which show agerelated
behavioural deterioration such as deficits in learning and memory, emotional disorders and altered circadian
rhythm, being therefore used as a model of spontaneously occurring Alzheimer’s disease. In parallel, their resistant
senescence (SAMR1) brothers have been used as a control. We used the chromaffin cell as a model of neurosecretion in
SAMP8 and SAMR1 mice at 2, 6 and 12 months of age. By means of the patch-clamp technique, we have studied the ionic
currents involved in the secretory process of catecholamines and in the transmission of the nerve impulse (nicotinic,
Na+, Ca2+ and K+ currents). We have also assessed cell excitability by measuring membrane potential and spontaneous
and triggered action potentials. Moreover, we have studied the release of the neurotransmitters by the amperometric
technique using K+ as stimuli. Finally, we have used the Y-maze to evaluate the cognitive behaviour of the mice.

Results: We have observed that there is an increase in all ionic currents with the age in both, SAMP8 and SAMR1 mice,
but this increase occurs before and more remarkable in SAMP8 mice. Regarding membrane potential and spontaneous
and triggered action potentials we have observed that there is a hyperpolarization of membrane potential in both types
of mice during the aging, moreover, the depolarization that ACh produces is lower throughout the age and again, this
phenomenon occurs before and is more remarkable in SAMP8 mice. Furthermore, the number of action potentials
generated at rest and those produced by depolarizing pulses decreases during the aging, being higher the amplitude and
posthyperpolarization area. Although, the number of action potentials evoked by ACh increases with aging due the less
depolarization being more remarkable in SAMP8 mice.
Regarding the release of the neurotransmitters we have seen that when we stimulate with K+ there is an increase in the
catecholamine secretion with the aging, happening before in SAMP8 mice. Moreover, this increase in the catecholamine
release is accompanied by changes of secretory spikes.
Finally, with Y-maze, we have seen that these alterations in the release of neurotransmitters are associated with a
cognitive deficit, since the SAMP8 mice explore all the arms equally, whereas the R1 always explore more the new arm.

Conclusion: We have found that during the aging the mechanisms of exocytosis of neurotransmitters is altered. These
alterations could be correlated with the changes that occurs in some neurodegenerative diseases and also causing
cognitive deficits.

e00108
NLRP3 inflammasome inhibition improves motor and behavioral outcome in a mouse model of traumatic brain injury.

Víctor Farré-Alins1,2,3, Alejandra Palomino-Antolín1,2,3, Paloma Narros1,2,3, Juliana Martins Rosa1,2,3, Cristina Sánchez Carabias4, Miguel Sáez Alegre5,
Alfonso Lagares4, Borja Jesús Hernández-García5, Javier Egea1,2,3*.

*Corresponding author:
Javier Egea, Hospital Universitario Santa Cristina, Madrid, Spain. E-mail: javier.egea@inv.uam.es

Details of affiliation

1Instituto de Investigación Sanitaria Princesa (ISS-IP), Hospital Universitario Santa Cristina, Madrid, Spain.
2Departamento de Farmacología y Terapéutica, Facultad de Medicina, Madrid, Spain.
3Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.
4Servicio de Neurocirugía, Hospital Universitario 12 de Octubre, Madrid, Spain.
5Servicio de Neurocirugía, Hospital Universitario La Paz, Madrid, Spain.

Funding

This study is funded by Fundación Mutua Madrileña, the program Miguel Servet (CP14/00008) of IS Carlos III, and Fondo de
Investigaciones Sanitarias (FIS) (ISCIII/FEDER) (PI16/00735).

Competing Interests:

The author declares no conflict of interest

Keywords: traumatic brain injury, inflammation, inflammasome, interleukin-1 beta, MCC950.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00108

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Víctor Farré-Alins, Alejandra Palomino-Antolín, Paloma Narros, Juliana Martins Rosa, Cristina Sánchez Carabias, Miguel Sáez
Alegre, Alfonso Lagares, Borja Jesús Hernández-García, Javier Egea. NLRP3 inflammasome inhibition improves motor and behavioral
outcome in a mouse model of traumatic brain injury. IBJ Plus 2018 (S2):e00108 doi: 10.24217/2531-0151.18v1s2.00108.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: traumatic brain injury (TBI) is one of the first causes of death and disability in young adult population in our
society. Recently, TBI has been called “the silent epidemic”, since it is a frequent serious problem and the consequences
of the pathology are unknown. In this context, neuroinflammation that takes place after TBI plays a key role in the
development of secondary lesions, which can become chronic. Thus, the aim of this study is to analyze the effect of
different compounds that can modulate the inflammatory response in in vitro models of inflammation, as well as in an
in vivo model of TBI.

Material and methods: we have evaluated the effect of MCC950 (inhibitor of inflammasome and IL-1β release) on the
release of two proinflammatory cytokines (IL-1β and TNF-α) in primary glial cultures after stimulation with LPS and ATP.
In order to evaluate the potential beneficial effects of the compounds, we used the mice TBI model called “Closed Head
Injury”. After the induction of the lesion and subsequent resuscitation, we assessed the neurological functions of the
animal 1 hour and 24 hour after TBI using Neurological Severity Score (NSS) scale. It consists of the evaluation of different
motor and behavioral parameters with a maximum score of 10 points, considering a severe trauma the value of 7-8
points. The treated groups were injected intraperitoneally with MCC950 (3 mg/kg or 10 mg/kg) immediately following
the 1-hour test.

Results: in primary glial cultures, MCC950 reduces IL-1 β release but not TNF-α. In the TBI model, we obtained a score
of 7 after 1 hour, while after 24 hours it decreased to 5 in non-treated animals. Both dosages decreased the value
obtained at 24 hours, even though it was only significant at 3 mg/kg (value of 3.8). Furthermore, we analyzed the edema
measuring the content of water in the brain, as well as the blood brain barrier (BBB) impairment using Evans Blue dye.
We obtained a reduction of water content and a decrease of BBB permeability at 3 mg/kg and 10 mg/kg.

Conclusions: although an extensive evaluation of molecular parameters and biomarkers related with inflammation is
needed, treatment with MCC950 improves motor and behavioral conditions that correlate with a reduction of brain
edema and a decrease in BBB permeability. Therefore, inflammation could be a potential target to treat detrimental
effects of traumatic brain injury.

e00109
Control of inflammation by microglial heme-oxygenase- 1 is differentially regulated with aging.

Cristina Fernández-Mendívil1, Enrique Luengo1, Paula Trigo-Alonso1, Izaskun Buendia1,2, Manuela G. Lopez1,2.

*Corresponding author:
Manuela G. López 1,2, Madrid, Spain. E-mail: cristina.fernandezm@uam.es

Details of affiliation

1Institute Teofilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma de Madrid. Spain.
2Hospital Universitario La Princesa. Madrid. Spain.

Funding

SAF2015-63936R

Competing Interests:

None

Keywords: HO-1; inflammation; aging

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00109

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Cristina Fernández-Mendívil, Enrique Luengo, Paula Trigo-Alonso, Izaskun Buendia, Manuela G. Lopez. Control of inflammation by microglial heme-oxygenase- 1 is differentially regulated with aging. IBJ Plus 2018 (S2):e00109 doi: 10.24217/2531- 0151.18v1s2.00109.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Neurodegenerative diseases (NDDs) share pathological mechanisms such as oxidative stress, protein
aggregation or chronic inflammation, processes in which microglial cells have a pivotal role. Microglia are the cells of
the innate immune system of the central nervous system. It has been proven that microglial heme-oxygenase 1 (HO-1)
enzyme has anti-inflammatory, antioxidant and neuroprotective effects. However, in patients with Alzheimer’s disease
and during aging, the expression and activity of HO-1 is increased compared to adult and healthy subjects. Also, there
are ferrous deposits derived from the metabolism of HO-1, which can be cytotoxic. Therefore, our aim was to understand
the role of HO-1 under inflammatory conditions, both in adult and aged wild type (WT) mice and LysMCreHmox1ΔΔ (KO
HO-1) mice, which lack the HO-1 enzyme in microglial cells.

Method and Results: Initially, 3-month-old mice were treated i.p. with 0.5 mg/Kg of LPS. The results of behavioral
parameters (locomotion and social interaction) showed that the LysMCreHmox1ΔΔ animals treated with LPS presented a
worse behavioral profile than the WT mice at 4 and 8 hours after the injection of the inflammatory stimulus. In addition,
the absence of microglial HO-1 was related with a higher release of pro-inflammatory cytokines (TNF-α and IL-1β) and
with an increase in pro-oxidant enzymes. In addition, WT mice treated 2 hours before the injection of LPS with 20
mg/Kg of zinc protoporphyrin (ZnPP), as a pharmacological tool to inhibit the activity of total HO-1, provided similar
results to the ones observed in LysMCreHmox1ΔΔ animals. To study the implications of the absence of HO-1 in the
inflammatory response in aged animals, 15-months-old mice treated with 20 mg/Kg of ZnPP or LysMCreHmox1ΔΔ were
used. The results of behavioral parameters and biochemical analysis showed that WT animals treated with ZnPP and
LysMCreHmox1ΔΔ mice subjected to LPS presented a better anti-inflammatory response compared to their respective
controls treated with LPS.

Conclusion: Taken all together, these results highlight the importance of microglial HO-1 in resolving inflammation with
age; its absence in young animals is related to a higher inflammatory profile, while its absence in aged mice seems
to be beneficial. Therefore, regulation of HO-1 to resolve the inflammatory state that underlies many NDDs is tightly
dependent on age.

Acknowledgments: The Spanish MINECO (ref. SAF2015-63936R) supported this work and it was possible thanks to the
FPU scholarship granted by the Ministry of Education, Culture and Sports (ref. FPU15/03269). We also appreciate the
continuous support of Teófilo Hernando’s Foundation Institute.

e00110
Neuroprotective effects of new compounds directed to PP2A, a promising therapeutic target for Alzheimer’s disease.

Raquel L. Arribas1, Rocío Lajarín Cuesta1, Cristóbal de los Ríos Salgado1,2

*Corresponding author:
Raquel L. Arribas, Departamento de Farmacología, UAM, Madrid, Spain. E-mail: raquel.lopezarribas@uam.es

Details of affiliation

1Instituto Teófilo Hernando, Universidad Autónoma de Madrid. C/ Arzobispo Morcillo, 4, 28029. Madrid, Spain.
2Instituto de Investigación Sanitaria of Hospital Universitario de la Princesa. C/ Diego de León, 62, 28006. Madrid, Spain.

Funding

We thank Instituto Fundación Teófilo Hernando for its continuous support. R.L.A.and R.L.-C. thanks Universidad Autónoma
de Madrid for predoctoral fellowships. This study was supported by Proyectos de Investigación en Salud from Instituto de Salud Carlos
III (Madrid, Spain, PI13/00789; PI16/01041) to C.d.l.R.

Competing Interests:

The authors declare no competing financial interests.

Keywords: tau hyperphosphorylation, PP2A, okadaic acid, neuroprotection

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00110

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Raquel L. Arribas, Rocío Lajarín Cuesta, Cristóbal de los Ríos Salgado. Neuroprotective effects of new compounds directed to PP2A, a promising therapeutic target for Alzheimer’s disease. IBJ Plus 2018 (S2):e00110 doi: 10.24217/2531-0151.18v1s2.00110

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Alzheimer’s disease (AD) is a progressive neurological disease that causes a progressive memory loss.
Main histopathological hallmarks of AD are senile plaques and neurofibrillary tangles, generated by aggregation of the
microtubule associated protein tau. In addition to these pathological characteristics, there are other alterations, such as
oxidative stress or loss of cholinergic transmission, among many others. One of the most promising approaches in AD
treatment is to inhibit neurofibrillary degeneration produced by an abnormal tau hyperphosphorylation. In this sense,
serine/threonine phosphoprotein phosphatase 2A (PP2A) is the major phosphatase in brain that accounts for over 70% of
tau dephosphorylation. It has been shown that PP2A activity is significantly decreased in post-mortem AD brains, partly
due to the increase of endogenous inhibitors that bind the PP2A catalytic subunit C.

Hypothesis: Okadaic acid (OA) is a natural toxin capable of inhibiting PP2A, leading to tau hyperphosphorylation. Our
working hypothesis is based on antagonizing the inhibitory effects of OA on PP2A by designing analogues of OA, capable
to bind to PP2A but without exerting inhibition, and thus preventing the attachment of endogenous inhibitors.

Material y Methods: The compounds synthesized, analogues to C19-C27 OA fragment, have been pharmacologically
studied, evaluating them in several in vitro models of AD, such as: tau hyperphosphorylation induced by OA, oxidative
stress caused by the toxic cocktail rotenone and oligomycin A or the glutamate induced excitotoxicity, all of them by the
MTT method. Furthermore, we have measured the cellular phosphatase activity by the pNPP method. In order to carry
out these objectives, we have used SH-SY5Y neuroblastoma cells and rat embryonic cortical neurons. Finally, we confirm
our theory by docking studies.

Results and conclusions: Our molecules are not toxic in SH-SY5Y cells or in cortical neurons, and they are capable of
reducing the neurotoxicity induced by OA. Some of them also showed good profile in the cell stress model induced by
R/O A in SH-SY5Y cells, and in the glutamate-induced excitotoxicity in cortical neurons. The new compounds maintained
the serine/threonine phosphatase activity, depressed by the action of two PP2A inhibitors: OA and citostatin. Molecular
docking studies indicated that compound ITH12680 is capable of binding to PP2A similarly to OA, but it does not interact
with the catalytic site, thus confirming our starting hypothesis. Taking into account these results, we conclude that our
compounds could have potential indication for the treatment of neurodegenerative diseases based on the maintenance of
PP2A activity.

e00111
Galectin-3 and Monocyte Chemoattractant Protein-1, as new biomarkers for patients with diabetes and high risk of cardiovascular diseases.

Lorenzo-Almorós A1, Aceña A2, Pello AM2, Carda R2, López-Castillo M2, Tuñón J2, Lorenzo Ó1,3.

Details of affiliation

1Renal, Vascular and Diabetes laboratory. Instituto de Investigaciones Sanitarias-Fundación Jiménez Díaz. Universidad Autónoma, Madrid, Spain.
2Department of Cardiology, Fundación Jiménez Díaz, Madrid, Spain.
3Spanish Biomedical Research Centre in Diabetes and Associated Metabolic Disorders Network (CIBERDEM)

Funding

This project has been supported by Laboratorios Esteve.

Competing Interests:

Authors declare that there are no conflicts of interest.

Keywords: diabetes mellitus, myocardial infarction, serum biomarkers, galectin-3

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00111

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Lorenzo-Almorós A, Aceña A, Pello AM, Carda R, López-Castillo M, Tuñón J, Lorenzo Ó. Galectin-3 and Monocyte Chemoattractant Protein-1, as new biomarkers for patients with diabetes and high risk of cardiovascular diseases. IBJ Plus 2018 (S2):e00111 doi:
10.24217/2531-0151.18v1s2.00111.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Coronary artery disease (CAD) is a leading cause of death in developed countries. Also, type-II diabetes
mellitus (DM2) has been recognized as an independent cardiovascular risk factor. In this sense, new specific biomarkers
may allow clinicians to early diagnose CAD to classify high-risk patients and to initiate appropriated treatments.

Material and Methods: Plasma samples from 989 patients with stable coronary artery disease (SCAD) were analysed
6 months after an Acute Coronary Syndrome [non-ST elevation acute coronary syndrome (NSTEACS) or ST elevation
myocardial infarction (STEMI)] by enzyme-linked immunosorbent assays (ELISA). Galectin-3, pro-protein convertase
subtilisin/kexin type 9 (PCSK-9), lipoprotein-A (LpA) and monocyte chemoattractant protein-1 (MCP-1) plasma levels
were quantified in parallel to reference biomarkers such as N-terminal fragment of brain natriuretic peptide (NT-proBNP)
and high sensitivity C-reactive protein (hsCRP), as well as the lipid profile (LDL-c, HDL-c, total cholesterol, triglycerides).
Values were compared in patients with DM2 (fasting glucose ≥ 126 mg/dl or anti-diabetic treatment) (n=237) and
without DM2 (n=752).

Results: DM2 patients with SCAD showed higher levels of galectin-3 (8.30 [6.44-10.48] vs 7.76 [5.94-9.77]; p=0.042)
and MCP-1 (144.53 [112.13-194.94] vs 132.83 [105.33-173.30]; p=0.009) as well as CRP (1.40 [0.56-4.42] vs 1.13 [0.304-
2.985]; p=0.005). Total cholesterol (140.0 [121.0-157.0] vs 145.0 [126.0-166.0] mg/dl; p<0.001), LDL (73.0 [61.0-88.0] vs
79.0 [65.0-94.0] mg/dl; p=0.003), HDL (38.0 [33.0-44.0] vs 42.0 [36.0-48.8] mg/dl; p<0.001) were lower in patients with
DM2 than in those without this disorder. Opposite to this, triglycerides were higher in DM2 patients (113.0 [81.0-162.5]
vs 97.0 [75.0-135.0] mg/dl; p<0.001). NT-proBNP, high sensitivity troponin I, PCSK-9 and Lp(a) levels were unchanged.

Conclusion: Galectin-3, a β-galactoside-binding lectin involved in cardiovascular remodelling, and MCP-1, a
chemoattractant factor implicated in vascular inflammation and atherothrombosis, may serve as biomarkers to detect
inflammation in DM2 patients with high risk of cardiovascular disease as ACS.

e00112
Huntingtin overexpression is associated to altered excitability and exocytosis in chromaffin cells from the R6/1 mouse model of Huntington’s disease.

Carmen Martínez-Ramírez1, Andrés M. Baraibar1, Carmen Nanclares1, Iago Méndez-López1, Luis Gandía1, María José Casarejos2, Antonio G. García1.

*Corresponding author:
Antonio G. García: E-mail: antonio.garcia@ifth.es

Details of affiliation

¹Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Spain
²Instituto de Investigación Sanitaria, Hospital Universitario Ramón y Cajal, Madrid, Spain

Funding

SAF2016-78892-R (Ministerio de Economía y competitividad) and Instituto Teófilo Hernando

Competing Interests:

The authors declare no competing financial interest

Keywords: Huntington’s disease, R6/1 mice, chromaffin cells, exocytosis

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00112

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Carmen Martínez-Ramírez, Andrés M. Baraibar, Carmen Nanclares, Iago Méndez-López, Luis Gandía, María José Casarejos, Antonio G. García. Huntingtin overexpression is associated to altered excitability and exocytosis in chromaffin cells from the R6/1 mouse model of Huntington’s disease. IBJ Plus 2018 (S2):e00112 doi: 10.24217/2531-0151.18v1s2.00112.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Adrenal medullary chromaffin cells (CCs) from transgenic mouse models of Alzheimer’s disease (AD) and
amyotrophic lateral sclerosis (ALS) have been reported to undergo functional changes of cell excitability, ion currents,
cytosolic calcium transients ([Ca2+]c), the quantal release of catecholamines and the kinetics of the exocytotic fusion
pore, with respect their wild type (WT) counterparts. In this frame, we planned this investigation trying to answer these
questions in the slow-developing Huntington’s disease (HD) R6/1 mouse model at 2-months of age (absence of motor
symptoms, pre-disease stage) and at 7-months of age (motor deficits present, disease stage).

Material and methods: We here present a thorough investigation on the functional changes undergone by CCs from
2-months and 7-months aged WT and R6/1 mouse model of Huntington’s disease (HD), stimulated with acetylcholine
(ACh) or K+. To study this, we used different approaches such as immunohistochemistry assay, patch clamp and
amperometry techniques.

Results: With respect WT cells, some of the changes were already observed in HD mice at 2 months; however, they were
more pronounced at 7 months, when motor deficits were clearly established. They were as follows: (i) nuclear huntingtin
overexpression as nuclear aggregates; (ii) smaller CC size with decreased dopamine β-hydroxylase expression, indicating
lesser number of chromaffin secretory vesicles; (iii) reduced adrenal tissue catecholamine content; (iv) membrane
hyperpolarisation with reduced ACh-evoked action potentials; (v) reduced Na+ and Ca2+ currents; (vi) reduced [Ca2+]c
transients with faster clearance; (vii) diminished quantal secretion with smaller vesicle quantal size; (viii) slower kinetics
of the exocytotic fusion pore, pore expansion, and faster closure.

Conclusion: These profound changes demonstrate that the altered neurotransmitter release occurring in the brain of
HD patients may also occur in peripheral sympathetic-like adrenal CCs. As in central neurons, these peripheral functional
changes may be attributed to the pathological accumulation of mutated huntingtin in CCs. The drastic reduction of the
quantal release of neurotransmitter indicates that in human HD patients, their response to stress may be hampered by
the drastic decrease of catecholamine release from adrenal medullary CCs.

e00113
Implication of Interleukin-17A in renal dysfunction progression due to arterial pressure changes.

Macarena Orejudo del Río1,2, Raúl Rodrigues-Diez3, Raquel Rodrigues-Díez4, Carolina Lavoz5, Marta Ruiz-Ortega1,2.

*Corresponding author:
Macarena Orejudo del Río. Cellular Biology in Renal Diseases Laboratory. IIS-Fundación Jiménez Díaz. Medicine Department, School of Medicine,
Universidad Autónoma de Madrid Madrid, Spain.
E-mail: macarena.orejudo@estudiante.uam.es

Details of affiliation

1Cellular Biology in Renal Diseases Laboratory. IIS-Fundación Jiménez Díaz. Madrid, Spain.
2Medicine Department, School of Medicine, Universidad Autónoma de Madrid. Spain.
3Nephrology Department, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain.
4Pharmacology Department, School of Medicine, Universidad Autónoma de Madrid. Spain.
5Division of Nephrology, School of Medicine, Universidad Austral, Valdivia, Chile.

Funding

This work was supported by the Instituto de Salud Carlos III and Fondo Europeo de Desarrollo Regional grant PI017/00119
and Sociedad Española de Nefrología. Macarena Orejudo was a Conchita Rábago Foundation Felow.

Competing Interests:

The authors declare that they have not a conflict of interest.

Keywords: Interleukin-17A, hypertension, inflammation

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00113

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Macarena Orejudo del Río, Raúl Rodrigues-Diez, Raquel Rodrigues-Díez, Carolina Lavoz, Marta Ruiz-Ortega. Implication of Interleukin-17A in renal dysfunction progression due to arterial pressure changes. IBJ Plus 2018 (S2):e00113 doi: 10.24217/2531-0151.18v1s2.00113.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Objectives: Hypertension is a vascular disease that damages several organs. In the kidneys, the disorder related to this
damage is called hypertensive nephropathy. This disease involves an interstitial and periglomerular fibrosis, tubular
atrophy and inflammation. There are several cytokines, chemokines and immune system cells engaged in repairing
tissues. Th17 lymphocytes and their main effector cytokine, Interleukin-17A (IL-17A), take part in the response against
extracellular pathogens, autoimmune and chronic inflammatory diseases. Our aim was to study the participation of IL-
17A in renal damage and its relation with arterial pressure elevation.

Methods. The presence of IL-17A positive cells was evaluated in renal biopsies of hypertensive nephropathy patients
and in experimental models of hypertension: systemic infusion of Angiotensin II (AngII) in Wistar rats and in C57BL/6
mice (dose of 100 or 1000 ng/kg/min, respectively, during 15 days). The effect of IL-17A in the kidney was studied using
two approaches: 1) neutralizing antibody against IL-17A (100 μg/mouse intraperitoneally every 4 days) in the AngIIinfusion
model in mice and 2) a model of systemic administration of IL-17A in mice at a dose of 1.5 ng/g mouse.

Results. IL-17A positive cells were found in renal biopsies of hypertensive patients, as well as in the kidneys of hypertensive
models. Accordingly, IL-17A renal levels were also elevated in hypertensive animals compared to controls (evaluated by
ELISA). IL-17A systemic administration in mice increased systolic pressure (117 ± 4 mm Hg vs 88 ± 3 mm Hg; n=7 p≤0.05, at
15 days compared to controls). These mice also had elevated renal kallikrein-1 levels, observed by immunohistochemistry
and quantitative PCR, compared to controls. Interestingly, renal gene expression levels of markers Kim-1 and N-gal
were upregulated in IL-17A-treated compared to control mice. The evaluation of renal lesions showed the presence of
inflammatory cells in the kidneys of IL-17A-treated mice, including CD3, CD4 lymphocytes and neutrophils. Moreover,
upregulation of proinflammatory factors was also found. In the model of AngII-induced hypertension, IL-17A blockade
decreased renal inflammatory cell infiltration and proinflammatory factors overexpression.

Conclusions. The presence of IL-17A positive cells in the kidney of hypertensive patients suggests that these cells could
be involved in the pathogenesis of hypertension. The experimental studies presented here show that elevated IL-17A
circulating levels induce an elevation of blood pressure and contributes to the inflammatory process in the kidney and,
therefore, this cytokine participates in renal dysfunction associated to hypertension.

e00114
NLRP3 inflammasome inhibition reduces infarct volume, Blood-Brain-Barrier breakdown and inflammation in cerebral ischemia.

Alejandra Palomino-Antolín1,2, Víctor Farré-Alins1,2, Paloma Narros1,2, Juliana Martins Rosa, Ana Isabel Casas3, Harald HHW Schmidt3, Javier Egea1,2.

*Corresponding author:
Javier Egea, Madrid, Spain. Email: javier.egea@inv.uam.es

Details of affiliation

1Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Hospital de La Princesa (IIS-IP), Madrid.
2Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid.
3Department of Pharmacology & Personalised Medicine, CARIM, Maastricht University, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands.

Funding

Fundación Mutua Madrileña, el Programa Miguel Servet (CP14/00008) del IS Carlos III, y el Fondo de Investigaciones
Sanitarias (FIS) (ISCIII/FEDER) (PI16/00735) .

Competing Interests:

No conflicts of interest

Keywords: cerebral ischemia, NLRP3 inflammasome, blood-brain barrier.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00114

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Alejandra Palomino-Antolín, Víctor Farré-Alins, Paloma Narros, Juliana Martins Rosa, Ana Isabel Casas, Harald HHW Schmidt, Javier Egea. NLRP3 inflammasome inhibition reduces infarct volume, Blood-Brain-Barrier breakdown and inflammation in cerebral ischemia. IBJ Plus 2018 (S2):e00114 doi: 10.24217/2531-0151.18v1s2.00114.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Cerebral ischemia is the third cause of death and the main cause of adult disability worldwide. Currently,
intravenous tissue plasminogen activator (tPA) is the only pharmacological treatment for acute ischemic stroke. However,
only 3% of patients benefit from tPA administration, due to its limited therapeutic window and the risk of intracerebral
hemorrhage. NLRP3 inflammasome, a key component of the innate immune system, has a critical role in inflammation
damage in ischemic injury. NLRP3 inflammasome activation leads to caspase-1-dependent cleavage of pro-IL-1β to
active IL-1β and pyroptosis.

Material and Methods: In this study, we wanted to investigate the role of NLRP3 in post-ischemic inflammation, using
MCC950, a potent inhibitor of NLRP3 inflammasome. For that purpose, we used transient middle cerebral artery
occlusion (tMCAO) during 1 hour in mice as a model of cerebral ischemia. Furthermore, we analyzed the blood brain
barrier (BBB) permeability using Evans Blue dye 24 hours after reperfusion. Finally, we wanted to know whether MCC950
could be acting on tight junction of endothelial cells.

Results: Administration of MCC950 1h after reperfusion reduced infarct volume in a dose-dependent manner (1, 3,
10 mg/kg; 53,23% ,50,57%, 107,87%, respectively). As a clinical outcome parameter, MCC950 at 3 mg/kg improved
neuro-motor function and reduced expression of different pro-inflammatory cytokines (IL-1β and TNF-α) and NLRP3
inflammasome component. We observed that tMCAO produced BBB disruption that was improved in animals treated
with MCC950 3 mg/kg. In MCC950-treated animals, we observed a functional recovery of endothelial proteins that
forms the tight junctions of BBB (VE-cadherina, Cd31, ZO-1).

Conclusion: From these results we can conclude that i) inhibition of NLRP3 inflammasome with MCC950 significantly
reduces infarct volume and improve neuro-motor function, and ii) MCC950 preserves BBB integrity through stabilization
of the tight junctions. Hence, the inhibition of NLRP3 may be a promising target in cerebral ischemia.

e00115
Bisphenol A induces autophagy and oxidative stress in experimental
chronic kidney injury and in tubular cells.

Alberto Ruiz Priego1, Sandra Rayego Mateos1, Enrique Bosch Panadero1, Sebastian Mas Fontao1, Alberto Ortiz Ardúan1, Marta Ruiz Ortega1, Emilio
González Parra2.

*Corresponding author:
Sandra Rayego Mateos, Instituto de Investigación Sanitaria Fundación Jiménez Díaz. E-mail: srayego@quironsalud.es

Details of affiliation

1Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Laboratorio de nefrología, Madrid, SPAIN.
2Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Servicio de Nefrología, Madrid, SPAIN.

Funding

Competing Interests:

Keywords: Chronic Kidney Disease, Autophagy, Bisphenol A.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00115

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Alberto Ruiz Priego, Sandra Rayego Mateos, Enrique Bosch Panadero, Sebastian Mas Fontao, Alberto Ortiz Ardúan, Marta Ruiz Ortega, Emilio González Parra. Bisphenol A induces autophagy and oxidative stress in experimental chronic kidney injury and in tubular cells. IBJ Plus 2018 (S2):e00115 doi: 10.24217/2531-0151.18v1s2.00115.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction and Aims: Uremic toxins accumulated as a result of chronic kidney disease (CKD) contribute to the
complications of the disease and its progression. . Bisphenol A (BPA) is a ubiquitous environmental toxin accumulated
in CKD. Autophagy is a degradative lysosomal process that eliminates misfolded and protein aggregates or damaged
organelles to maintain intracellular homeostasis and cellular integrity. Recent evidences suggest that autophagy
is implicated in tubular and glomerular cell damage in renal diseases. Nevertheless, the role of autophagy on
pathophysiology of renal disease is still unknown. The aim of this study is to explore the role of autophagy as mechanism
of BPA toxicity on experimental chronic kidney injury.

Methods: The role of BPA in chronic kidney injury was studied in a experimental model of subtotal nephrectomy in mouse
c57bl/6. Some animals were injected with BPA (120mg/kg/day) intraperitoneally during 5 weeks. Furthermore, in vitro
studies were developed in human proximal tubular epithelial cells (HK-2) stimulated with BPA at different concentrations
(50 μM, 100 μM and 200 μM). Gene Expression were measured by qRT-PCR and proteins levels were analyzed by several
techniques such as western blot and immunohistochemistry.

Results: We observed that in subtotal nephrectomized mice, the BPA exposure during 5 weeks led to oxidative stress,
autophagy and inflammation by increasing the gene expression of specific autophagy markers such as Atg5, Atg7, LC3B
and Beclin. On the other hand, BPA exposure in mice increased the expression of oxidative stress and Nrf2 target genes as
Hemo-oxygenase 1 (HO-1) and NAD(P)H dehydrogenase [quinone] 1 (NQO-1). Additionally, in cell culture the stimulation
with BPA increased in a dose dependent manner the gene expression of proinflamatory factors as CCL-2, CCL-5 and IL-6
and the autophagy markers LC3B and Beclin.

Conclusions: These data suggests that BPA causes oxidative stress, inflammation, and autophagy in experimental chronic
kidney injury developping a fundamental role into CKD progression.

e00116
Clopidogrel response is defined by CYP2C19 metabolizer status in patients undergoing percutaneous neurointervention procedure.

Miriam Saiz-Rodríguez1, Daniel Romero-Palacián1, Carlos Villalobos-Vilda1, José Luis Caniego2, Carmen Belmonte1, 3, Dora Koller1, Eduardo Bárcena2,
María Talegón1, Francisco Abad-Santos1, 3, 4.

*Corresponding author:
Miriam Saiz-Rodríguez, Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de
Madrid (UAM), Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain. E-mail: miriam.saiz@salud.madrid.org

Details of affiliation

1Clinical Pharmacology Department, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid (UAM),
Instituto de Investigación Sanitaria la Princesa (IP), Madrid, Spain
2Department of Radiology, Hospital Universitario de la Princesa, Universidad Autónoma de Madrid (UAM), Madrid, Spain
3UICEC Hospital Universitario de la Princesa, Plataforma SCReN (Spanish Clinical Reseach Network), Instituto de Investigación Sanitaria la Princesa (IP),
Madrid, Spain.
4Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain

Funding

Saiz-Rodriguez M was co-financed by Consejería de Educación, Juventud y Deporte from Comunidad de Madrid and Fondo
Social Europeo. D. Koller is co-financed by the H2020 Marie Sklodowska-Curie Innovative Training Network 721236 grant.

Competing Interests:

F. Abad-Santos has been consultant or investigator in clinical trials sponsored by the following pharmaceutical
companies: Abbott, Alter, Chemo, Cinfa, FAES, Farmalíder, Ferrer, GlaxoSmithKline, Galenicum, Gilead, Janssen-Cilag, Kern, Normon,
Novartis, Servier, Silverpharma, Teva, and Zambon. The remaining authors declare no conflicts of interest

Keywords: CYP2C19; phenotype, antiplatelet; clopidogrel; neurointervention, haemorrhage, ischemia

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00116

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Miriam Saiz-Rodríguez, Daniel Romero-Palacián, Carlos Villalobos-Vilda, José Luis Caniego, Carmen Belmonte, Dora Koller,
Eduardo Bárcena, María Talegón, Francisco Abad-Santos. Clopidogrel response is defined by CYP2C19 metabolizer status in patients
undergoing percutaneous neurointervention procedure. IBJ Plus 2018 (S2):e00116 doi: 10.24217/2531-0151.18v1s2.00116.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Clopidogrel is a widely prescribed thienopyridine prodrug which inhibits platelet aggregation. It is
prescribed to prevent atherothrombotic and thromboembolic events in patients who are given a stent implant in carotid,
vertebral or cranial arteries. CYP2C19 is the most studied enzyme involved in clopidogrel metabolism. The most common
CYP2C19 no function polymorphisms (*2 and *3) have been associated with hyporesponse to clopidogrel, showing lower
levels of the active metabolite. On the contrary, the presence of the increased function allele (*17) has demonstrated
enhanced platelet inhibition and clopidogrel hyperresponse.

Methods: This observational retrospective study assessed antiplatelet response and clinical events after clopidogrel
treatment in patients who underwent percutaneous neurointervention, related to CYP2C19 metabolizer status (normal
(NM), intermediate/poor (IM-PM) and ultra-rapid (UM); inferred from *2, *3 and *17 allele determination by real-time
PCR).

Results: One hundred twenty-three patients were analysed (59 men and 64 women, mean age 64 years), of which 83%
had cardiovascular risk factors. The most common type of intervention was angioplasty with stent (60.2%). According to
the aggregation value, 58.7% of the patients were responders to clopidogrel; moreover, 4.1% required dose reduction
and 12.2% change of treatment. Related to their genotype, 32 (26%) patients were classified as IM-PM; 53 patients
(43.1%) as NM and 38 patients (30.9%) as UM. CYP2C19 IM-PM had higher aggregation value (201.1 vs 137.6 NM, 149.4
UM, p<0.05) and lower response rate (37.5% vs. 69.8% NM, 61.1% UM), along with higher treatment change rate (25%
vs. 5.7% NM, 10.5% UM). Moreover, 20% of the patients suffered from a subsequent clinical event. The highest ischemic
events incidence occurred in NM (11.3% vs. 6.3% IM, 10.5% UM; p=0.358) and haemorrhagic events in UM (13.2%
vs. 0% IM and 3.8% NM; p=0.041). No differences found regarding ischemic events’ onset time, while haemorrhagic
events’ frequency in UM was higher with shorter onset time (p=0.047). Additionally, 53% of the patients were receiving
concomitant treatment with proton-pump inhibitors (PPIs), which showed significantly higher aggregation value when
compared to those not receiving PPI concomitant treatment (178.1 vs. 134.4; p=0.009).

Conclusion: CYP2C19 no function and increased function alleles defined clopidogrel response. CYP2C19 genotyping
and platelet reactivity quantification help to determine whether a patient could be at risk of ischemic or haemorrhagic
event. CYP2C19 UM patients have increased bleeding risk after percutaneous neurointervention. This is the first study
to associate CYP2C19 with clinical outcomes in this cohort of patients. Therapeutic recommendations should include an
alternative therapeutic option in IM-PM or UM patients.

e00117
Role of the mitochondrial Na+/Ca2+ exchanger in NLRP3 inflammasome activation.

Paloma Narros Fernández1,2, Alejandra Palomino Antolín1,2, Víctor Farré Alins1,2, Juliana M. Rosa1,2, Cristóbal de los Ríos1,2, Javier Egea1,2.

*Corresponding author:
Javier Egea, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain. E-mail: javer.egea@inv.uam.es

Details of affiliation

¹Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP),Madrid, Spain.
²Instituto Teófilo Hernando, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid

Funding

This study has been financed by Miguel Servet Program (CP14/00008), by the “Fondo de Investigaciones Sanitarias” (FIS)
(ISCIII/FEDER) (PI16/00735), and by the Mutua Madrileña Foundation.

Competing Interests:

The authors declare no conflict of interests.

Keywords: NLRP3 inflammasome, mitochondrial Na+/Ca2+ exchanger (NCLX), mitochondria, inflammation, Reactive oxygen species (ROS).

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00117

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Miriam Saiz-Rodríguez, Daniel Romero-Palacián, Carlos Villalobos-Vilda, José Luis Caniego, Carmen Belmonte, Dora Koller,
Eduardo Bárcena, María Talegón, Francisco Abad-Santos. Clopidogrel response is defined by CYP2C19 metabolizer status in patients
undergoing percutaneous neurointervention procedure. IBJ Plus 2018 (S2):e00116 doi: 10.24217/2531-0151.18v1s2.00116.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: The pathophysiology of multiple neuroinflammatory diseases involve the activation of NLRP3
inflammasome. The inflammatory response triggered by the inflammasome can be activated by different danger stimuli
in the cell. Several studies propose mitochondria as key elements in the activation of this inflammatory signalling
pathway, through the production of reactive oxygen species (ROS) and mitochondrial dysfunction. However, the
exact mechanisms operating in this process are poorly understood. Previous results have shown that inhibition of the
mitochondrial Na+/Ca2+ exchanger (NCLX) by the benzothiazepine CGP37157 exerts a protective effect in several in vitro
models of neurodegeneration. Moreover, NCLX inhibition reduces ROS induced by hypoxia. Since mitochondrial ROS
participate in the activation of NLRP3 inflammasome, we proposed to study the possible participation of NCLX in this
process.

Materials and Methods: To this end, we have used the compound ITH12575, a synthetic derivative of CGP37157, and
we have studied its effect in glial primary cultures of mouse and in the murine macrophage cell line J774 A.1 exposed to
NLRP3-activation conditions.

Results: Stimulation of glial cultures with lipopolysaccharide (LPS) 1 μg/ml during 3’5 hours, followed by ATP 5 mM 30
min induced NLRP3 inflammasome activation and IL-1β release. Inhibition of NCLX by ITH12575 reduced the release of
this pro-inflammatory cytokine in a concentration-dependent manner (1, 3 and 10 μM). Oxidative stress parameters
(ROS and RNS) induced by LPS treatment of glial cultures were also reduced by ITH12575. LPS treatment of macrophages
during 24 hours induced the stabilization of the hypoxia-inducible factor 1-alpha (HIF-1α) and pro-IL-1β expression, an
effect that was notably potentiated under hypoxic conditions (1% O2). In these conditions, pharmacological inhibition of
NCLX by ITH12575 reduced both HIF-1α stabilization and pro-IL-1β protein levels, which suggests a possible mechanism
by which mitochondria can be participating in the activation of the inflammasome.

Conclusion: From these results we can conclude that (i) inhibition of mitochondrial NCLX by ITH12575 significantly
reduces IL-1β release in glial cultures stimulated with LPS+ATP and (ii) NCLX inhibition by ITH12575 reduces oxidative
stress parameters induced by LPS in glial cultures.

e00118
Mitochondria function and morphology alterations precede
neurosecretion impairment in chromaffin cells of the SOD1G93A
mouse model of amyotrophic lateral sclerosis.

Iago Méndez-lópez1, Carmen Martínez-Ramírez1, Antonio G. García1, Fernando Padín Nogueira1,2.

*Corresponding author:
Iago Méndez-López, Instituto Teófilo Hernando and Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de
Madrid, Madrid, Spain. E-mail: iagomendez@hotmail.es

Details of affiliation

1Instituto Teófilo Hernando and Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain.
2Departamento de Ciencias Médicas, Facultad de Medina, Universidad de Castilla La Mancha, Ciudad Real, Spain.

Funding

SAF 2013-44108-P grant to AGG; FPI BES-2014-069005 grant to I.M-L., MINECO, Spain. Continuous support or Fundación Teófilo
Hernando is also acknowledged.

Competing Interests:

Authors have no conflict of interest to the presented work.

Keywords: ALS, SOD1G93A, chromaffin cell, mitochondrial ultrastructure, fusion pore, exocytosis.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00118

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Iago Méndez-lópez, Carmen Martínez-Ramírez, Antonio G. García, Fernando Padín Nogueira. Mitochondria function and morphology alterations precede neurosecretion impairment in chromaffin cells of the SOD1G93A mouse model of amyotrophic lateral sclerosis. IBJ Plus 2018 (S2):e00118 doi: 10.24217/2531-0151.18v1s2.00118.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Amyotrophic lateral sclerosis (ALS) is characterized with a selective loss of motor neurons that cause
paralysis and respiratory failure. Hyperexcitability and Ca2+-dependent glutamate excitotoxicity has been hypothesized
to be involved in ALS pathogenesis. Exploring the chromaffin cell (CC) of SOD1G93A mouse model of familiar ALS, we
found that the fusion pore kinetics of exocytosis is slowed but with higher catecholamine quantal size when the disease
is already established (Calvo-Gallardo et al., Am J Physiol Cell Physiol 2015;308:C1-C19). To go further in the study of
these neurosecretion alterations, we investigate the exocytosis before the disease onset (30 days postnatal), and we
focus in the study of mitochondrial ultrastructure and function as a crucial organelle involved in the process.

Material and Methods: Mitochondrial ultrastructure was explored by transmission electron microscopy (TEM) and
analyzed with ImageJ software. Luminometer was used to measure ATP levels in CC cultures with the commercial
CellTiter-Glo® kit. Reactive oxygen species (ROS) production was monitored 30 minutes with the fluorescent dye
H2DCFDA. Fusion pore kinetic was studied by amperometry, eliciting exocytosis by 1 minute acetylcholine stimulus.

Results: TEM showed that mitochondria from SOD1G93A CCs have the following alterations with respect to wildtype
CCs: i) more number and small sized; ii) increased mitochondrial intermembrane space; iii) lower number and swollen
cristae. These ultrastructural changes suggesting mitochondrial fission and ultrastructure damage were accompanied
by lower ATP production and a higher rate of ROS generation. However, we fail in observe such significant differences in
the fusion pore kinetics.

Conclusion: The described mitochondrial alterations shown an interesting non-motor neuron degeneration in this ALS
model at presymptomatic stages. However, the kinetic of the exocytotic fusion pore have not been affected, contrary to
the slowed secretion observed once the paralysis is already established. These results evidence that this mitochondrial
alterations precede the functional changes linked to neurotransmitter release. In spite of having lower clinical relevance
than in later stages, it could generate some clues about the initiation and progression of the disease. Our data consolidate
the mitochondria as a potential target and the sympathetic-adrenal system affectation as an interesting new approach
for ALS diagnosis.

e00119
Vascular damage in obesity associated to pge2 derived mpges-1 through aldosterone/mineralocorticoid-receptor route.

María González-Amor1*, Luis M. Beltrán2, Ana B. García-Redondo1,3, Raquel Rodrígues-Díez1, Constanza Ballesteros1, Mercedes Salaices1,3, Ana M.
Briones1,3.

*Corresponding author:
maria.gonzalezamor@uam.es

Details of affiliation

1Dpt. Farmacología, Facultad de Medicina, UAM. IdiPAZ. Madrid, Spain.
2Servicio de Medicina Interna, Hospital Universitario La Paz, UAM. IdiPAZ. Madrid. Spain.
3Ciber de enfermedades cardiovasculares (CiberCV), Spain.

Funding

ISCIII-Fondo Europeo de Desarrollo Regional: PI13/01488; MINECO: SAF 2016-80305-P; Roche-IDIPAZ; CIBERCV.

Competing Interests:

No competing interests.

Keywords: vascular damage, obesity, mPGES-1.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00119

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: María González-Amor, Luis M. Beltrán, Ana B. García-Redondo, Raquel Rodrígues-Díez, Constanza Ballesteros, Mercedes
Salaices, Ana M. Briones. Vascular damage in obesity associated to pge2 derived mpges-1 through aldosterone/mineralocorticoidreceptor
route. IBJ Plus 2018 (S2):e00119 doi: 10.24217/2531-0151.18v1s2.00119.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Adipocytes are more than fat storage cells since they release a number of factors contributing to energy homeostasis
and vascular tone and structure. We demonstrated that adipocytes are a source of aldosterone in response to Ang II
and that this is facilitated by mPGES-1-derived ProstaglandinE2 (PGE2) (unpublished). However, whether this pathway
is activated in obesity is unknown. Mineralocorticoid-Receptor (MR), is involved in the stiffness and the endothelial
dysfunction observed in obesity. We determined if mPGES-1 participates in aldosterone production from adipocytes in
obesity and whether this is involved in endothelial dysfunction and vascular stiffness observed in this pathology.

Epididimal fat from DBA mPGES-1+/+ and mPGES-1-/- mice fed with normal or high fat diet (HFD) was analyzed. CYP11B2
and MR expression were studied by qRT-PCR. Changes in vascular function and stiffness were studied using wire and
pressure myographs. 3T3-L1 adipocytes were stimulated with 16,16-DimethylProstaglandinE2 (DPGE2). Furthermore,
visceral fat was obtained from patients and gene data were correlated with parameters of vascular stiffness.

We found that CYP11B2 mRNA expression is augmented in the adipose tissue of the mPGES-1+/+ HFD mice, but not in
mPGES-1-/-. However, we did not find differences in the MR. Moreover, DPGE2 increases CYP11B2 mRNA expression in
3T3-L1 adipocytes. HFD provokes endothelial dysfunction in both genotypes, which is prevented by eplerenone. HFD
induces vascular stiffness in mPGES-1+/+ but not in mPGES-1-/-mice. Preliminary data in patients show positive correlations
between mPGES-1, CYP11B2 and MR gene expression. Moreover, there is a positive correlation between pulse wave
velocity and CYP11B2 gene expression.

Our study suggest that mPGES-1-derived PGE2 is involved in the excessive aldosterone synthase expression observed in
adipose tissue in obesity and this might have a role in the vascular damage observed in this pathology.

Abstracts PhD Programme in Clinical and Health Psychology:

e00120
Assessing individual change without knowing the test properties: Item bootstrapping.

Juan Botella1, Desirée Blázquez1, Javier Revuelta1, Manuel Suero1.

*Corresponding author:
Desirée Blázquez, Department of Social Psychology and Methodology (UAM), Madrid, Spain. E-mail: desiree.blazquez@yahoo.com

Details of affiliation

1Department of Social Psychology and Methodology, Universidad Autónoma de Madrid, Madrid, Spain.

Funding

The research was carried out without funding support.

Competing Interests:

The research was conducted in the absence of any commercial or financial relationships that could be construed
as a potential conflict of competing interests.

Keywords: Bootstrap, Individual change, Reliable change, Significant change, Psychometric properties.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00120

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Juan Botella, Desirée Blázquez, Javier Revuelta, Manuel Suero. Assessing individual change without knowing the test properties: Item bootstrapping. IBJ Plus 2018 (S2):e00120 doi: 10.24217/2531-0151.18v1s2.00120.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Botella et al. (2018) proposed to use the ‘non parametric bootstrap’ method (Efron & Tibshirani, 1994)
to the responses given by an individual to the items of a test in order to create confidence intervals for an individual’s
true test score for situations in which classical procedures cannot be used. In six databases containing the responses to
several psychological scales, two procedures were applied to create the confidence intervals; a classical one, Estimating
the True Score (ETS; Gulliksen, 1950), and the Bootstrap of items (BSI). When there was an expected change in the
criterion of interest after an intervention and when there was not, the rates of significant change obtained with both
procedures were very similar. These results suggested that BSI was a promising solution when other methods could not
be applied. However, evidence is needed from different research contexts to assess the performance of BSI.

Material and Method: On the basis of the Partial Credit Model (Masters, 1982), a IRT model for polytomous response
data, two simulation studies were programmed in R to examine the performance of the BSI technique in comparison to
the classical ETS method. In study 1, focused on no change scenarios, we examine the influence of several test features
on the width of the BSI confidence interval and its coverage rates of the true score. The factors assessed are the subjects’
trait level, skewness of the trait distribution, number of items’ categories, number of items, and internal consistency
reliability (Cronbach’s alpha). Study 2 was carried out to analyze the significant change rates of BSI and ETS given a
change in the subjects’ trait level.

Results: Study 1 shows that the BSI confidence interval is narrower as the subject’s trait level gets more extreme and
the test internal consistency is higher. The BSI coverage rates of the true score reach appropriate values when the test is
made up of at least 20-25 items. Results from study 2 reveal that BSI has lesser statistical power to detect a significant
change than ETS as the change in the subject’s trait level is bigger.

Conclusion: The classical procedure ETS seems to be yet the best option. Nevertheless, the classical procedures for
elaborating confidence intervals involve knowing several properties of the test given a fixed sample and, sometimes,
these properties are unknown or are not trustworthy. Given the differences in the performance of the methods
examined, BSI is a good option to create confidence intervals for an individual’s true test score for situations in which
classical procedures cannot be used.

e00121
Physical Activity as a Potential Protective Factor in People at
Increased Genetic Risk for Alzheimer’s Disease.

Jaisalmer de Frutos-Lucas1, 2, Fernando Maestú Unturbe2, 3, Juan Manuel Serrano1, Simon Laws4

*Corresponding author:
Jaisalmer de Frutos Lucas: jaisalmer.dfl@gmail.com

Details of affiliation

1Biological and Health Psychology, Universidad Autónoma de Madrid
2Center for Biomedical Technology
3Psicología Experimental, Procesos Cognitivos y Logopedia, Universidad Complutense de Madrid
4School of Medical and Health Sciences, Edith Cowan University

Funding

La Caixa Foundation PhD Scholarship to Jaisalmer de Frutos

Competing Interests:

No Conflict of interests.

Keywords: Physical Activity, Genetics, Alzheimer’s Disease, healthy Aging

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00121

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Jaisalmer de Frutos-Lucas, Fernando Maestú Unturbe, Juan Manuel Serrano, Simon Laws. Physical Activity as a Potential
Protective Factor in People at Increased Genetic Risk for Alzheimer’s Disease. IBJ Plus 2018 (S2):e00121 doi: 10.24217/2531-
0151.18v1s2.00121.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Alzheimer’s disease (AD) is the most common cause of dementia. Unfortunately, no cure has been found
to prevent the advance of this devastating neurodegenerative disease. The AD neuropathological process begins up
to 20 years before we are able to detect the first clinical manifestations of the disease. Thus, it remains possible that
pharmacological and non-pharmacological interventions are more effective in preclinical stages of AD. On the other
hand, physical activity (PA) has been thought to improve health and cognition since ancient times. However, it has
not been until recently that we have started to understand the mechanisms that underlie this relationship. Therefore,
the aim of my studies is to investigate how PA influences different biomarkers of AD (grey matter volumes, functional
connectivity, blood markers of neuroinflammation, brain amyloid deposition, etc.) in different samples of healthy elders
who are at increased risk for AD, either because they are carriers of different genetic risk factors or because they are
relatives in first degree of AD patients.

Methods: For these studies I have been able to have access to two main sources of data. These are the Australian imaging,
biomarkers & lifestyle flag study of ageing (aibl) dataset and that of the longitudinal studies in healthy and pathological
aging carried out at the Laboratory of Cognitive and Computational Neuroscience (Center for Biomedical Technology).
The set of assays that compose these protocols include: magnetoencephalography, magnetic resonance imaging,
genotyping, neuroinflammation assays, nutritional assessment, physical activity measurement, neuropsychological
assessment, ophthalmological assessment among others.

Results: Preliminary results suggest that although healthy older adults do in general benefit from PA, such benefit is
larger or exclusive to those who are not at increased genetic risk for AD. Genetic risk for poor dopamine transmission
has also been identified as a risk factor for low PA engagement and cognitive decline.

Conclusion: Higher levels of PA are associated to lower levels of different early markers of AD. However, this effect tends
to be more moderate or non-existent carriers of certain genetic risk factors. Nevertheless, physical inactivity increases
the risk of AD even further in those who are already at increased risk for AD.

e00122
The impact of subjective well-being on mortality.

Natalia Martín-María MS1,2,3, Marta Miret PhD1,2,3*, Francisco Félix Caballero PhD1,2,3, Josep Maria Haro MD2,4, José Luis Ayuso-Mateos PhD-MD1,2,3

*Corresponding author:
Natalia Martín-María. Department of Psychiatry, Universidad Autónoma de Madrid, Spain. E-mail: natalia.martinm@uam.es

Details of affiliation

1Department of Psychiatry, Universidad Autónoma de Madrid, Spain.
2Instituto de Salud Carlos III, Centro de Investigación Biomédica en Red de Salud Mental. CIBERSAM, Spain.
3Department of Psychiatry, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Madrid, Spain.
4Parc Sanitari Sant Joan de Déu, Universitat de Barcelona, Sant Boi de Llobregat, Barcelona, Spain

Funding

The research leading to these results has received funding from the European Union Horizon 2020 Framework Programme
for Research and Innovation under grant agreement 635316 (ATHLOS Project), from the European Community’s Seventh Framework
Programme (FP7/2007-2013) under grant agreement number 223071 (COURAGE in Europe), from the Spanish Ministry of Science
and Innovation ACI- Promociona (ACI2009-1010), from the Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM)
Mental Health and Disability Instruments Library Platform, and from the Instituto de Salud Carlos III-FIS research grants PS09/00295,
PS09/01845, PI12/01490, and PI13/00059. Projects PI12/01490 and PI13/00059 have been co-funded by the European Union European
Regional Development Fund (ERDF) “A Way to Build Europe.” The study was supported by the Instituto de Salud Carlos III Centro de
Investigación Biomédica en Red de Salud Mental (CIBERSAM). NMM is supported by the programme “Contratos predoctorales para
Formación de Personal Investigador, FPI-UAM,” Universidad Autónoma de Madrid, Spain.

Competing Interests:

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Keywords: Subjective well-being longitudinal study, meta-analysis, mortality, depression.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00122

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Natalia Martín-María MS, Marta Miret, Francisco Félix Caballero, Josep Maria Haro, José Luis Ayuso-Mateos. The impact of
subjective well-being on mortality. IBJ Plus 2018 (S2):e00122 doi: 10.24217/2531-0151.18v1s2.00122.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Subjective well-being has been recognized as an important global health issue. The aims of the present
study are: 1) to review the existing evidence on whether subjective well-being is a protective factor for mortality in
the general population, analyzing the differential impact of evaluative, experienced, and eudaimonic well-being, and
2) to disentangle the differential influence that positive affect, negative affect, and evaluative well-being might have
on mortality, after adjusting for health and other lifestyle factors and to analyze whether this association is different in
people with and without depression.

Methods: A systematic review of longitudinal studies on the general population was carried out in the PsycINFO, Web of
Science, and PubMed databases. Data on the studies’ characteristics, quality, and the effects of variables were extracted.
A meta-analysis was conducted on the studies included in the systematic review. In the empirical part of the study, a
nationally representative sample of 4,753 people from the general population in Spain was followed up after 3 years.
Analyses were performed with Cox regression models among the total sample and separately in people with and without
depression.

Results: A total of 62 articles that investigated mortality in general populations, involving 1,259,949 participants, were
found. The meta-analysis showed that subjective well-being was a protective factor for mortality [pooled HR= 0.920;
95% CI = (0.905, 0.934)]. The impact of subjective well-being on survival was significant in both men and women. The
three aspects of subjective well-being were significant protective factors for mortality. The same results were found in
the longitudinal study, in which all three well-being variables showed separately a statistically significant association
with mortality, after adjusting for age, sex, and years of education. However, after adjusting for health status and the
other well-being components, only positive affect remained as marginally associated with a decreased risk of mortality
in the overall sample [HR = 0.87; 95% CI = 0.73–1.03], and in particular among individuals without depression [HR = 0.82;
95% CI = 0.68–0.99].
Conclusion: Our results from the meta-analysis suggest that evaluative, experienced, and eudaimonic well-being are
associated with a decreased risk of mortality. However, our analysis adjusting for health status and the other well-being
components, showed that only positive affect is inversely associated with mortality in individuals without depression.
Future research should focus on assessing interventions associated with a higher level of positive affect in order to
produce longevity gains.

Abstracts Other PhD Programmes:

e00123
PPRV effects on the differentiation of sheep monocyte-derived dendritic cells.

Rodríguez-Martín D1, Rojas JM1, Martín V1, Sevilla N1*.

*Corresponding author:
sevilla@inia.es

Details of affiliation

1Centro de Investigación en Sanidad Animal (CISA-INIA), Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Ctra. Algete a El Casar,
Valdeolmos, 28130, Madrid, Spain.

Funding

DRM is supported by a “Garantía Juvenil” Contract Ref.56640 co-financed by the European Social Fund and the Youth
Employment Initiative. This work was funded by Grants AGL2015-64290R and ADENONET-Redes de Excelencia from Spanish Ministerio
de Economía y Competitividad; Grant S2013/ABI-2906-PLATESA from Comunidad de Madrid and the European Union (FEDER funds);
and European Grant 731914-VetBionetH2020.

Competing Interests:

The authors declare that they have no competing interests.

Keywords: PPRV, sheep, monocyte-derived dendritic cells, differentiation, cell markers, phagocytosis

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00123

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Rodríguez-Martín D, Rojas JM, Martín V, Sevilla N. PPRV effects on the differentiation of sheep monocyte-derived dendritic
cells. IBJ Plus 2018 (S2):e00123 doi: 10.24217/2531-0151.18v1s2.00123.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Peste des Petits Ruminants virus (PPRV) is the causative agent of an economically important disease,
which affects small ruminants, limiting the productivity of the livestock. PPRV produces immunosuppression which can
derive in opportunistic infections that increase mortality in infected animals. PPRV is a Morbillivirus, Paramyxoviridae
family, closely related to measles and rinderpest virus. PPRV infects immune cells through the signalling lymphocyte
activating molecule (SLAM) and this tropism could contribute to its immunosuppressive properties. Since dendritic cells
(DC) are professional antigen presentation cells essential to the development of an effective adaptive immune response,
we evaluated PPRV effects on DC differentiation and activity.

Material and Methods: CD14+ cells (monocytes) were isolated from ovine Peripheral Blood Mononuclear Cells (PBMC)
using anti-CD14 antibody magnetic sorting, and differentiated into immature monocyte-derived DC (iMoDC) with GMCSF
and IL-4 for 72h. To determine PPRV effects on DC differentiation, CD14+ cells were infected with the virulent ICV-89
PPRV strain prior to differentiation.

Results: After 72h with GM-CSF and IL-4, ovine monocytes presented classic DC characteristics such as changed
morphology (increased size, multiple dendrites), increased DC cell markers expression levels (CD11b, CD11c, DC-SIGN,
MHC-II, MHC-I, CD1, CD1w2, DC80 and CD86) and increased phagocytic activity. Thus CD14+ monocytes were successfully
differentiated into iMoDC after 72h. Viral production was detected in iMoDC culture supernatants differentiated from
PPRV-infected monocytes. iMoDC infection was further confirmed by anti-PPRV-N antibody staining. A slight increase in
cell death was observed in PPRV-infected iMoDC. At 72h, PPRV-infected iMoDC decreased CD11b, CD11c, MHC-II, CD1
and CD1w2 expression, while CD80 and CD86 co-stimulation markers levels remain unchanged. PPRV infection reduced
the phagocytic activity of iMoDC as detected by flow cytometry and confocal microscopy. Furthermore, in allogeneic
MLR assays, the antigen-presenting capacity of infected iMoDC was reduced.

Conclusions: We were able to successfully differentiate functional iMoDC from sheep CD14+ monocytes. PPRV
infection in monocytes disrupted their differentiation to iMoDC by downregulating a number of iMoDC surface
markers and reducing their phagocytic and antigen-presenting ability. PPRV disruption of professional antigen
presenting cell function could therefore contribute to the immunosuppressive effects of the virus.

e00124
Palbociclib radiosensitizes colon and lung cancer cell lines in a p53-dependent manner.

D.M. Fernández-Aroca1, O. Roche1, M. Ortega-Muelas1, R. Pascual-Serra1, S. Sabater2, R. Olivares-Martin1, MJ. Ruiz-Hidalgo1,3, R. Sánchez-Prieto4

*Corresponding author:
Diego M. Fernández Aroca, Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, UCLM, Unidad
Asociada de Biomedicina CSIC-UCLM, Albacete, España E-mail: DiegoManuel.Fernandez@alu.uclm.es

Details of affiliation

¹Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, UCLM, Unidad Asociada de Biomedicina
CSIC-UCLM, Albacete, España.
²Servicio de Oncología Radioterápica, Complejo Hospitalario Universitario de Albacete (CHUA), Albacete, España
³Área de Bioquímica y Biología Molecular, Dpto. de Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina de Albacete, UCLM
⁴Departamento de Biología del cáncer, Instituto de Investigaciones Biomédicas “Alberto Sols”. CSIC-UAM, Madrid, España

Funding

Study supported by Fundación Leticia Castillejo Castillo

Competing Interests:

The authors declare that they have no conflicting interests.

Keywords: Palbociclib, radiosentitizing, p53, radiation.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00124

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: D.M. Fernández-Aroca, O. Roche, M. Ortega-Muelas, R. Pascual-Serra, S. Sabater, R. Olivares-Martin, MJ. Ruiz-Hidalgo, R.
Sánchez-Prieto. Palbociclib radiosensitizes colon and lung cancer cell lines in a p53-dependent manner. IBJ Plus 2018 (S2):e00124 doi:
10.24217/2531-0151.18v1s2.00124.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Cell cycle regulatory proteins are one of the most used targets in cancer-targeted therapy. One example
is palbociclib, a CDK4/6 inhibitor that suppresses their enzymatic activity by the inhibition of their phosphorylating
capacity making them unable to phosphorylate their substrate: the retinoblastoma protein (pRB), which results in G1-
phase cell cycle arrest.
On the other hand, radiotherapy is one of the main tools in cancer therapy, in fact, it has been estimated that
approximately 50% of patients need their use at some point during the course of the disease. In this sense, the use of
radiosensitizing drugs becomes very important, because allows greater treatment effectivity.
Recent studies points to the potential of palbociclib as a radiosensitizer agent, nevertheless, it has not been established
its molecular mechanism of action.

Material and methods: The aim of this study was to check the radiosensitizing effect of palbociclib, as well as establishing
its molecular mechanism of action in colorectal (HCT116 y HT-29) and lung (A549 y H1299) cancer cell lines. We evaluated
the palbociclib toxicity by MTT. We measured the functionality of palbociclib in two ways, by Western Blot (measuring
pRB phosphorylation) and by flow cytometry (analyzing cell-cycle arrest). Furthermore, to analyze the needed of p53 for
the radiosensitizing effect, we use the HCT116 isogenic model p53+/+ and p53 -/-; and we knockdown p53 by shRNA in
A549 cells.

Results: Palbociclib shows the same toxicity in all the employed cell lines, and moreover induces the same G1-phase cell
cycle arrest and inhibits the phosphorylation in pRB. Palbociclib induces radiosensitivity if the cell line presents a fully
functional p53 protein, on the contrary, in those lines lacking p53 or expressing a non-functional p53 protein we do not
observe this radiosensitizing effect.

Conclusion: Our results demonstrate the radiosensitizing effect of palbociclib and the needed of a functional p53 protein
for this effect, providing a new clinical use for this inhibitor. These results could allow doctors to predict the effectiveness
of the treatment in a patient using the p53 protein as a biomarker.

e00125
Erk5 pathway is a new indirect target of sorafenib.

M. Ortega-Muelas1, L. Muñoz-Martinez-Blanco3, R. Pascual-Serra1, O. Roche1, D.M. Fernández-Aroca1, R.Olivares-Martin1, M.J. Ruiz-Hidalgo1,2, B.
Belandia3, R. Sánchez-Prieto1,3.

*Corresponding author:
Marta Ortega Muelas, Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, UCLM, Unidad
Asociada de Biomedicina CSIC-UCLM, Albacete, España. E-mail: mortega@iib.uam.es

Details of affiliation

1Laboratorio de Oncología, Unidad de Medicina Molecular, Centro Regional de Investigaciones Biomédicas, UCLM, Unidad Asociada de Biomedicina
CSIC-UCLM, Albacete, España.
2Área de Bioquímica y Biología Molecular, Dpto. de Química Inorgánica, Orgánica y Bioquímica, Facultad de Medicina de Albacete, UCLM.
3Departamento de Biología del Cáncer, Instituto de Investigaciones Biomédicas “Alberto Sols”. CSIC-UAM, Madrid, España.

Funding

MINECO (SAF2015-64215-R). Fundación Leticia Castillejo.

Competing Interests:

All the authors declare that there is no competing financial interest.

Keywords: Sorafenib, ERK5, MAPK.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00125

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: M. Ortega-Muelas, L. Muñoz-Martinez-Blanco, R. Pascual-Serra, O. Roche, D.M. Fernández-Aroca, R.Olivares-Martin, M.J.
Ruiz-Hidalgo, B. Belandia, R. Sánchez-Prieto. Erk5 pathway is a new indirect target of sorafenib. IBJ Plus 2018 (S2):e00125 doi:
10.24217/2531-0151.18v1s2.00125.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Sorafenib is a tyrosine kinase inhibitor that was developed as a B-RAF inhibitor. Afterwards it was discovered
that Sorafenib could also inhibit other kinases like VEGF-R and PDEGF-R. The fundamental therapeutic mechanisms of
Sorafenib are anti-angiogenesis and anti-cell proliferation. Its use has been approved in Renal Cell Carcinoma (RCC),
Gastrointestinal Stromal Tumor (GIST), Thyroid Cancer and Hepatocellular Carcinoma.

Material and Methods: An experimental model of human epithelioid cervix cancer cells (Hela) was used to analyse
how Sorafenib impacts on MEK5 function assessing the inhibition of the phosphorylation of its immediate downstream
MAPK, ERK5. Moreover, cell viability by MTT assay, and migration of cancer cells by wound healing assay were also
studied.

Results and Conclusion: In this study, we demonstrated that MEK5/ERK5 signaling pathway is a new target of Sorafenib.
Thus, we showed that Sorafenib provokes a partial inhibition of MEK5 activity upon treatment with EGF or by expression
of a constitutively active MEK5 mutant form. Furthermore, we have confirmed our observations by using specific shRNA
against ERK5.
Therefore, our data indicate that part of the therapeutic effect of Sorafenib could be mediated through the inhibitory
effect exerted onto ERK5 signalling pathway, discarding other MAPK as ERK1/2 or p38MAPK pathways. In sum, our data
demonstrate that ERK5 pathway is an indirect target of Sorafenib, providing new therapeutic opportunities in the use of
this novel tyrosine kinase inhibitor.

e00126
Mitochondrial activity plays a critical role in multiple myeloma resistance.

Ortiz-Ruiz Alejandra1,2, Ruiz-Heredia Yanira1, Morales María Luz1,2, Bárcenas Carmen3, García-Martin Rosa María3, Garrido Vanesa1, Baquero Irene1, Alonso Rafael1, Martínez-López Joaquín1,2, Linares María1,2*, Gallardo Miguel1,2*.

*Corresponding autor: María Linares, Haematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain and Haematology Neoplasms group, CNIO, Madrid, Spain; marialinares@vet.ucm.es. Miguel Gallardo, Haematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain and Haematology Neoplasms group, CNIO, Madrid, Spain; miguelgallardodelgado@gmail.com.

Details of affiliation

1Haematology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.
²Haematology Neoplasms group, CNIO, Madrid, Spain.
3Pathology Department, Hospital Universitario 12 de Octubre, Madrid, Spain.

Funding

This study was supported by the Subdirección General de Investigación Sanitaria grants PI13/02387 and PI16/01530, and
the CRIS against Cancer foundation grant 2014/0120

Competing Interests:

The authors declare no conflict of interest

Keywords: Multiple myeloma, resistance, mitochondria.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00126

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Ortiz-Ruiz Alejandra, Ruiz-Heredia Yanira, Morales María Luz, Bárcenas Carmen, García-Martin Rosa María, Garrido Vanesa,
Baquero Irene, Alonso Rafael, Martínez-López Joaquín, Linares María, Gallardo Miguel. Mitochondrial activity plays a critical role in
multiple myeloma resistance. IBJ Plus 2018 (S2):e00126 doi: 10.24217/2531-0151.18v1s2.00126.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: Mitochondria control crucial biological pathways such as proliferation, apoptosis and cell growth. The
implication of its activity in the pathogenesis of Multiple Myeloma (MM) stills remains unknown.
We have studied the impact of mitochondrial genes, protein expression, and activity in MM progression and treatment
resistance. Furthermore, we have studied the potential exploitation of mitochondrial activity as a functional target in
the MM therapy.

Methods: We have performed gene expression studies by RT-PCR of known factors that regulate and are involved in
the mitochondrial function (c-Myc, TFAM, EF-Tu, NRF1 and hnRNPK) in a total of 34 sample patients at different MM
stages. To validate gene expression results, we developed an immunohistochemistry (IHC) assay of COX II, representative
protein of mitochondrial burden in a total sample of 49 patients. We analyzed the mitochondrial activity with the study
of COX2 histoenzymatic reaction in 15 patients. Finally, we have tested the effect in plasma cells of Metformin and
Tigecycline, in monotherapy and in combination with Bortezomib over four MM cell lines (JJN3, L363, NCI-H929 and
NCI-H929 R20). Besides, we analyze the mitochondrial DNA (mtDNA) burden in these cell lines. Our further studies
include in vivo validation in NSG mice models of results obtained in vitro.

Results: We have observed a significant overexpression of genes C-Myc, TFAM, EF-Tu, and a higher expression trend
of hnRNPK in MM relapsed patients compared with MGUS and newly diagnose MM groups (p-value *< 0.05; p-value
** < 0.001) (Fig. 1A). Moreover, IHC reveals overexpression of mitochondrial COXII protein in newly diagnose MM and
relapsed groups compared with MGUS (Fig. 1B). By a functional assay we have demonstrated that gene and protein
overexpression drives to an increase of activity, comparing MGUS and MM at diagnosis versus MM at relapse (p-value
***< 0.0001) (Fig. 1C). We confirmed the correlation between higher mitochondrial burden and resistance to bortezomib
in JJN3 and L363, and NCI-H929 and its resistant, NCI-H929 R20 (p-value*<0.05) (Fig. D). In vitro drug assays showed a
synergistic effect of tigecycline with bortezomib, suggesting
that could be used as a potential therapy in combination for
MM patients.

Conclusions: Mitochondrial machinery plays a critical role
in the development, progression and resistance of MM
patients. Mitochondrial protein components that generates
the activity could be prospective targets for MM treatment.
Tigecycline demonstrates synergistic effect with Bortezomib
suggesting potential use as novel drug combination therapy
in MM patients.

e00127
From stem cells to unique neurons: Specification of the Drosophila melanogaster Orcokinin A neurons.

Irene Rubio-Ferrera1, Luis Clarembaux-Badell1, M Ángel Berrocal-Rubio1, Pablo Baladrón1, Núria Niell1, Hugo Gabilondo2, Jonathan Benito-Sipos1*.

*Corresponding author:
Jonathan Benito-Sipos. 1Universidad Autónoma de Madrid, Madrid (Spain). E-mail: jonathan.benito@uam.es

Details of affiliation

1Universidad Autónoma de Madrid, Departamento de Biología, Facultad de Ciencias, E 28049 Madrid, Spain.
2Centro de Biología Molecular Severo Ochoa (CBMSO), Departamento de Desarrollo y Regeneración, E 28049 Madrid, Spain.

Funding

This work was supported by a grant from the Spanish Ministerio de Economía y Competitividad (BFU2013-43858-P).

Competing Interests:

The authors have declared that no competing interests exist.

Keywords: neuron, stem cells, development, cell fate specification.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00127

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: Irene Rubio-Ferrera, Luis Clarembaux-Badell, M Ángel Berrocal-Rubio, Pablo Baladrón, Núria Niell, Hugo Gabilondo, Jonathan
Benito-Sipos. From stem cells to unique neurons: Specification of the Drosophila melanogaster Orcokinin A neurons. IBJ Plus 2018
(S2):e00127 doi: 10.24217/2531-0151.18v1s2.00127.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

One of the major challenges in the field of Developmental Neurobiology and Regenerative Medicine based on Stem
Cells (SCs) is to understand the basic mechanisms of cell specification. Discovering how SCs produce a progeny that
differentiates into diverse cell fates is essential to understand the normal development of an organism, but also to
understand some pathological situations and take advantage of the therapeutic potential of this system. This is
particularly interesting in tissue repair and regeneration after any damage or pathology.
Regarding the Central Nervous System (CNS), it is critical to understand how each neural subtype is generated under
physiological conditions. Thus, we could acquire the capacity to produce specific neurons from neural SCs to replace
those that have been lost or damaged.

Nowadays, improving in vivo studies of neural SCs manipulation becomes essential. However, the high conservation of the basic processes that regulate the neural progenitor differentiation supports the study of simpler model organisms.
Therefore, the main objective of this project is to advance in a logical and efficient way in our understanding about generation and maintenance of neuronal diversity. For that purpose, we study the specification of the abdominal Orcokinin A neuropeptidergic
neurons in Drosophila melanogaster.
First, we have found that these cells are generated at late stage of lineage progression in the Neuroblast 5-3 (NB 5-3) during the castor-grainyhead temporal window.

Second, we have found that their correct specification depends on Hox genes input. In particular, Ubx and abd-A appear to be involved in the establishment of the Orcokinin A terminal fate, while Antp and Abd-B seems to trigger a different fate in the segments they govern.
We have found that although the Dpp pathway is active in these neurons, the pathway does not seem to determine the Orcokinin A fate. Additionally, the Notch pathway is inactive in these neurons, which suggest that they are the Notch-OFF counterpart in the case they were sibling neurons born from a GMC cell. 

Finally, we have identified the cis-regulatory modules (CRM) that direct the expression of Orcokinin A to address the molecular mechanisms involved in the specification of these neurons. We have begun dissecting their organization by extensive in vivo studies. These involve mutant and misexpression analysis and transcription factor binding site mutagenesis. In addition, the CRISPR/Cas9 system will be use to delete this specific CRM to validate its in vivo necessity.
With this set of experiments, we attempt to resolve key questions that are hampering progress in the knowledge of Developmental Neurobiology and Stem Cells Biology.

e00088
A comprehensive study of Trichomonas vaginalis infection: a step forward to understand the pathobiology of isolates from Madrid, Spain.

C. Bolumburu1*, V. Zamora2, J.J. Nogal-Ruiz1, A. Gómez-Barrio1, M. Muñoz-Algarra2, J.A. Escario1, A. Ibáñez-Escribano1.

*Corresponding author:
C. Bolumburu. Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n,
28040, Madrid, Spain. E-mail: celiabol@ucm.es

Details of affiliation

1Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid,
Spain.
2Servicio de Microbiología, Hospital Universitario Puerta de Hierro-Majadahonda, Majadahonda, Madrid, España.

Funding

The study was funded by the Complutense University of Madrid research group “Antiparasitic Therapy” ref. 911120.

Competing Interests:

None declared.

Keywords: Trichomonosis, resistance, epidemiology, sexually transmitted infection, Trichomonas vaginalis virus, Mycoplasma,
genotype, microsatellite.

Published May 18, 2018.

DOI: 10.24217/2531-0151.18v1s2.00088

Copyright: © 2018 Author. This is an open-access article distributed under the terms of the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Cite as: C. Bolumburu, V. Zamora, J.J. Nogal-Ruiz, A. Gómez-Barrio, M. Muñoz-Algarra, J.A. Escario, A. Ibáñez-Escribano. A comprehensive
study of Trichomonas vaginalis infection: a step forward to understand the pathobiology of isolates from Madrid, Spain. IBJ Plus 2018
(S2):e00088 doi: 10.24217/2531-0151.18v1s2.00088.

Edited: Madrid, España.

Editor: Alberto M. Borobia Pérez.

Abstract

Introduction: The protozoan parasite Trichomonas vaginalis (TV) is the causative agent of the most common non-viral
sexually transmitted infection (STI) in the world. Trichomonosis is a very complex disease characterized by a broad
range of symptoms, as TV isolates vary in their virulence, pathogenicity and drug resistance, and a remarkable ability to
evade the host immune system using a wide variety of mechanisms. The last WHO report estimates an incidence of 276
million new cases every year.TV infection has been associated with an increased risk of several diseases as cervix and
prostate cancer, adverse pregnancy outcomes, infections caused by HIV, Chlamydia trachomatis, Neisseria gonorroheae,
Treponema pallidum, and human papilloma virus (HPV), especially the high-risk HPV type 16. Despite the fact that
trichomonosis exceeds the incidence of chlamydia, gonorrhea and syphilis together, nowadays this parasitic infection is
not a reportable disease.

Procedure: Our study is phased in three stages: (1) Epidemiological study in a STI clinic and a hospital of Madrid (Spain)
to study the trichomonosis in this region, (2) biological characterization of TV isolates to identify relevant phenotypic
characteristics as metronidazole resistance, presence of endosymbionts in the parasite (TVV and Mycoplasma), for the
correct parasite’s classification between type 1 or 2, and (3) molecular characterization of TV isolates to identify length
polymorphism microsatellite as genetic markers which support biologic characterization for intraespecific differentiation.

Results: As to date, 35 TV isolates have been received, 16 of which have been completely characterized, and 19 are being
analyzed. The epidemiological study showed that 8 of each 1000 vaginal swabs analyzed in the Hospital, was positive
to T. vaginalis in 2017, and comparing with the previous year, the incidence of trichomonosis in women attending this
hospital during 2017 increased a 118 percent. In addition, 2017 was the first year which broke the falling trend in the
incidence of trichomonosis in the hospital since 2013.

Discussion: Our findings demonstrated a high prevalence of T. vaginalis isolates infected with TVVs or M. hominis. The
MIC to metronidazole in M. hominis-infected T. vaginalis isolates tend to be higher than the respective non-infected
strains. The opposite result was found in TVV- infected T. vaginalis isolates, whose MIC tend to be lower than the
respective non-infected ones. This is the first study evaluating incident T. vaginalis in women attending a Hospital of
Madrid since the 90’s. Further investigation should assess the benefits of routinely screening women in STD clinics and
hospitals for T. vaginalis.