Diethylcarbamazine: A potential treatment drug for pulmonary hypertension?

Diethylcarbamazine: A potential treatment drug for pulmonary hypertension?

Accepted Manuscript Diethylcarbamazine: A potential treatment drug for pulmonary hypertension? Edlene Lima Ribeiro, Ingrid Tavares Fragoso, Fabiana O...

1MB Sizes 3 Downloads 77 Views

Accepted Manuscript Diethylcarbamazine: A potential treatment drug for pulmonary hypertension?

Edlene Lima Ribeiro, Ingrid Tavares Fragoso, Fabiana Oliveira dos Santos Gomes, Amanda Costa Oliveira, Amanda Karoline Soares e Silva, Patrícia Martins e Silva, Bianca Torres Ciambarella, Isalira Peroba Rezende Ramos, Christina Alves Peixoto PII: DOI: Reference:

S0041-008X(17)30354-X doi: 10.1016/j.taap.2017.08.015 YTAAP 14034

To appear in:

Toxicology and Applied Pharmacology

Received date: Revised date: Accepted date:

5 June 2017 9 August 2017 25 August 2017

Please cite this article as: Edlene Lima Ribeiro, Ingrid Tavares Fragoso, Fabiana Oliveira dos Santos Gomes, Amanda Costa Oliveira, Amanda Karoline Soares e Silva, Patrícia Martins e Silva, Bianca Torres Ciambarella, Isalira Peroba Rezende Ramos, Christina Alves Peixoto , Diethylcarbamazine: A potential treatment drug for pulmonary hypertension?. The address for the corresponding author was captured as affiliation for all authors. Please check if appropriate. Ytaap(2017), doi: 10.1016/j.taap.2017.08.015

This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

ACCEPTED MANUSCRIPT 1 Diethylcarbamazine: a potential treatment drug for pulmonary hypertension?

Edlene Lima Ribeiroa,b, Ingrid Tavares Fragosoa,b, Fabiana Oliveira dos Santos Gomesa,b, Amanda Costa Oliveira a,b, Amanda Karoline Soares e Silva a,b, Patrícia Martins e Silva c , Bianca Torres Ciambarella c , Isalira Peroba Rezende

SC RI P

a

T

Ramosd, Christina Alves Peixoto a*

Laboratory of Ultrastructure, Aggeu Magalhães Research Center – CPqAM,

Pernambuco, Brazil. Federal University of Pernambuco, Brazil.

c

Laboratory of Inflammation – FIOCRUZ, Rio de Janeiro, Brazil. National Center Structural Biology and Bio-imaging, Carlos Chagas Filho

Biophysics Institute and

MA

d

NU

b

Department of Radiology - University Hospital

ED

Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.

PT

*Corresponding author: Dr Christina Alves Peixoto. aLaboratory of

CE

Ultrastructure, Aggeu Magalhães Research Center, Avenida Moraes Rego s/n, Cidade Universitária, 50670-420, Recife, PE, Brazil. Fax: 55-81-21012500,

AC

Phone: 55-81-21012557.

ACCEPTED MANUSCRIPT 2

ABSTRACT

The present study demonstrated the potential effects of diethylcarbamazine

DEC for 28 days.

50 mg/kg body weight of

SC RI P

(600mg/kg) was administered once per week, and

T

(DEC) on monocrotaline (MCT)-induced pulmonary hypertension. MCT solution

Three C57Bl/6 male mice groups (n=10) were studied:

Control; MCT28, and MCT28/DEC. Echocardiography analysis was performed

NU

and lung tissues were collected for light microscopy (haematoxylin-eosin and Masson's trichrome staining), immunohistochemistry (αSMA, FADD, caspase 8,

MA

caspase 3, BAX, BCL2, cytochrome C and caspase 9) western blot (FADD, caspase 8, caspase 3, BAX, BCL2, cytochrome C and caspase 9) and qRt-PCR

ED

(COL-1α and αSMA). Echocardiography analysis demonstrated an increase in the pulmonary arterial blood flow gradient and velocity in the systole and RV

PT

area in the MCT28 group, while treatment with DEC resulted in a significant

CE

reduction in these parameters. Deposition of collagen fibers and αSMA staining around the pulmonary arteries was evident in the MCT28 group, while treatment

AC

with DEC reduced both. Western blot analysis revealed a decrease in BMPR2 in the MCT28 group, in contrast DEC treatment resulted in a significant increase in the level of BMPR2. DEC also significantly reduced the level of VEGF compared to the MCT28 group. Apoptosis extrinsic and intrinsic pathway markers were reduced in the MCT28 group. After treatment with DEC these levels returned to baseline. The results of this study indicate that DEC

ACCEPTED MANUSCRIPT 3 attenuates PH in an experimental monocrotaline-induced model by inhibiting a series of markers involved in cell proliferation/death.

Key

words:

Diethylcarbamazine,

monocrotaline,

hypertension,

AC

CE

PT

ED

MA

NU

SC RI P

T

apoptosis.

pulmonary

ACCEPTED MANUSCRIPT 4

INTRODUCTION Pulmonary hypertension (PH) is a life-threatening progressive disorder associated with abnormally elevated pulmonary pressure and right heart failure.

T

It is a disease of a complex etiology and pathobiology that results from

SC RI P

interactions between the genetic make-up of an individual and the surrounding environment [1, 2].

The initial pathological events of the pulmonary artery dysregulation

NU

involve the proliferation of the smooth muscle cells. Several lines of evidence suggested that increased proliferation and decreased apoptosis of the

MA

pulmonary arterial smooth muscle cells can mediate thickening of the pulmonary vasculature, which would subsequently lead to a reduced inner

ED

diameter and increased pulmonary vascular resistance [3]. Endothelial cell (EC) apoptosis and apoptosis resistance seems to play

PT

crucial roles in the development of plexiform lesions that feature in the

CE

pathogenesis of PH. Subsequently, EC injury associated with smooth muscle cell (SMC) proliferation facilitates vascular remodeling and eventually leads to

AC

narrowed vascular lumen, increased pulmonary vascular resistance, increased pulmonary arterial pressure, and right heart failure [4]. The imbalance between cell death and proliferation occurs in every stage of pulmonary vascular remodeling and the pathogenesis of PH, and involves every cell type in the vasculature including, but not limited to ECs, SMCs, and fibroblasts [5]. Intriguingly, PH pathogenesis involves both inappropriate apoptosis and over-proliferation. Apoptosis in ECs, after initial environmental insults, has been

ACCEPTED MANUSCRIPT 5 recognized as one of the crucial events that trigger pulmonary vascular remodeling in PH [6]. Despite extensive studies, a detailed understanding of the cellular and molecular mechanisms involved in the transition from initial apoptosis to apoptosis resistant proliferation of ECs and SMCs has yet to be established [4].

T

The MCT model is considered by some to be a toxic model, as it is

SC RI P

suggested that MCT rats die from hepatic veno-occlusive disease with liver failure, instead of right ventricle failure [7]. MCT is known to cause pulmonary endothelial injury and pulmonary hypertension in humans and rats [8, 9], but

NU

has little effect on mice [10].

The drug diethylcarbamazine (DEC) is used throughout the world against

MA

lymphatic filariasis. However, in recent years many studies have described other pharmacological activities of DEC. It has been established that DEC

eicosanoid

production

ED

interferes with the cyclooxygenase and lipoxygenase pathways, reducing and

acting

as

an

anti-inflammatory

drug

[11].

PT

Furthermore, DEC inhibits the activation of NF-kB, suppressing target genes

CE

involved in pulmonary inflammatory response [12]. DEC has also been shown to be effective in different models of lung inflammation, such as tropical eosinophilia,

AC

pulmonary

pulmonary

hypertension,

eosinophilic

pulmonary

inflammation and asthma [13, 14, 15, 16]. Queto et al. (2010) [15] reported that DEC suppressed the pulmonary and bone marrow eosinophilia by CD95L/CD95 signaling. Since the CD95L (FasL) is a ligant for the apoptosis inducing receptor CD95 (Fas), these results suggest that DEC can possibly act as an apoptosis inducer. The aim of the present study was to evaluate the cell death markers and action of DEC on a monocrotaline-induced pulmonary hypertension model.

ACCEPTED MANUSCRIPT 6

MATERIALS AND METHODS Animals Thirty male C57BL/6 mice, weighing 26-30 g (11 weeks), were used in all

T

experiments. The mice were examined to determine their health status and

SC RI P

acclimated to a laboratory environment of 23-24°C. They were kept in a 12/12 h day/night cycle photoperiod, housed in metal cages and fed a standard diet with water ad libitum [17, 18]. All experimental procedures were approved by the Committee

for

Animal

Experimentation

(Prot.

63/2014

NU

Ethics

Drugs and experimental Design

MA

FIOCRUZ/CPqAM).

ED

Diethylcarbamazine citrate (DEC) was obtained from Sigma (St. Louis, MO, D8765) and dissolved in distilled water. The lymphatic filariasis therapeutic

PT

dose regimens recommended by the World Health Organization of 6 mg/kg

CE

were used. Considering that the total metabolism rate of a mouse is approximately seven times that of a human, the present study used 50 mg/kg

AC

body weight of DEC, adjusted according to the body weight of mice, administered through drinking water for 28 days [12]. The mice received an intraperitoneal (i.p) injection of MCT (600mg/kg SigmaAldrich, St. Louis, MO, USA) dissolved in saline solution and administered once per week (0, 7, 14 and 21 th days) as described elsewhere [19, 20, 21]. Mice were randomly allocated into three groups of ten (N = 10) animals each:

ACCEPTED MANUSCRIPT

SC RI P

T

7

Determination of right ventricular hypertrophy (RVH), pulmonary artery and left ventricular function

Canada)

on

day

28.

During

the

procedure,

isofluorane/O2

MA

Toronto,

NU

Echocardiography analysis was performed using a VisualSonic Vevo770,

administration was administered using a facemask to keep the mice lightly

ED

anesthetized, with heart rates in the range of 300-350 bpm. The right ventricle was visualized from the right parasternal long axis view with a 704 RMV

PT

scanhead. The right ventricular wall thickness was measured from images produced in M-mode, using the depth interval (mm) generic measurement tool

CE

(Vevo770 v3.0 software, VisualSonics). Doppler flow images were recorded

AC

from the left parasternal long axis view with a 707 B scanhead pointing slightly towards the left shoulder to visualize the pulmonary artery. Volume was measured at the level of the pulmonary valve, and several indices of pulmonary artery blood flow (velocity time integral, peak and mean pressure gradient and peak and mean velocity) were assessed using the pulmonary valve protocol measurement tool [19, 22].

Histological examination

ACCEPTED MANUSCRIPT 8 The lung fragments were washed twice in PBS pH 7.2 and fixed in Bouin solution for 8 hours (1% saturated picric acid, formaldehyde and 40% glacial acetic acid), before being dehydrated in an increasing ethanol series, cleared in xylene, embedded and included in purified paraffin (VETEC, São Paulo, SP, Brazil). Tissue sections of 5 μm were cut using a microtome (Leica RM

SC RI P

hematoxylin/eosin and studied using light microscopy [23].

T

2125RT) and deparaffinized with xylene. They were then stained with

Immunohistochemical Localization

NU

The paraffin sections of lung tissue were mounted onto slides. After being deparaffinized, the tissues were incubated overnight at 4°C with primary

MA

antibody anti- α-SMA (1:100 cat. ab 5694) anti-FADD (1:50 cat. Sc 6036), anticaspase 8 (1:50 cat. Sc5263), anti-caspase 3 (1:100 cat. Ab4011), anti-BAX

ED

(1:50 cat. Ab7977), anti-BCL2 (1:100 cat. Ab7973), anti-cytochrome C (1:50 cat. Sc13156), anti-caspase 9 (1:50 cat. sc56076). The antigen-antibody reaction

PT

was visualized with avidin-biotin peroxidase (Dako Universal LSAB + Kit,

CE

Peroxidase), using 3,3-diaminobenzidine as a chromogen. Imaging was performed by light microscopy. Five pictures at the same magnification were

AC

quantitatively analyzed using the Gimp 2.8 software program (GNU Image Manipulation Program, UNIX platforms) [24]. RNA extraction and quantitative real-time polymerase chain reaction (q RTPCR) Total RNA from mouse tissues was isolated using Trizol reagent (Invitrogen, Carlsbad, CA, USA). The forward and reverse primers used for each

gene

were

as

follows:

Collagen

Type



(COL-1α):

5’-

ACCEPTED MANUSCRIPT 9 GAACGGTCCACGATTGCATG-3’ and 5’-GGCATGTTGCTAGGCACGAAG-3’, αSMA:

5’-ATCTGGCACCACTCTTTCTA-3’

GTACGTCCAGAGGCATAGAG-3’,

GAPDH

and

(endogenous

control):

5’5′-

AGGTCGGTGTGAACGGATTTG-3′ and 5′-TGTAGACCATGTAGTTGAGGTCA3′. All reactions were performed in triplicate and included the following: 1 μL of

T

cDNA; 5 μM of each primer; 2x SYBR Green PCR Master Mix (Applied

SC RI P

Biosystems); and water added to give a final volume of 25 µL. The relative amount of mRNA was determined using the comparative threshold (Ct) method by normalizing target cDNA Ct values to those of GAPDH. Fold increase ratios

NU

were calculated relative to control (basal conditions) for each group using the

MA

formula 2e-ΔΔCt [24].

Western Blot Analysis

ED

The lungs were submerged in liquid nitrogen and the total proteins were

PT

extracted using an extraction cocktail (10 mM ethylenediaminetetraacetic acid (EDTA), 2 mM phenylmethylsulfonyl fluoride (PMSF), 100 mM sodium fluoride

CE

(NaF), 10 mM sodium pyrophosphate, 10 mM sodium orthovanadate (NaVO4),

AC

10 mg of aprotinin, and 100 mM Tris(hydroxymethyl)aminomethane, pH 7.4). Western blotting and the subsequent quantification of each blot were performed as previously described [25]. The primary antibodies for anti-BAX, anti-Bcl2 and anti-caspase 3 were obtained from Abcam (CA, USA), while anti-caspase 8, anti-FADD, anti-caspase 9 and anti-cytochrome c were obtained from Santa Cruz Biotechnologies (Santa Cruz, CA). Secondary antibodies and β-actin were acquired from Sigma-Aldrich (USA).

ACCEPTED MANUSCRIPT 10 Data Analysis GraphPad Prism software (version 6) was used for statistical analysis. Data was expressed as mean ± standard deviation. Differences between the control and treatment groups were analyzed using analysis of variance (ANOVA), prior to Tukey’s post hoc test or the Student's t-test being carried out.

RESULTS

NU

Effect of DEC on pulmonary hypertension

SC RI P

T

Probability values less than 0.05 were considered significant.

In humans, transthoracic echocardiography is an excellent noninvasive

MA

screening test for patients with symptoms or risk factors for PH by providing direct and/or indirect signs of elevated pulmonary artery pressure (PAP).

ED

Doppler analysis at the pulmonary valve level, recorded by ultrasonography in lightly anesthetized mice (heart rate 300-350 bpm) demonstrated an increased

PT

in the pulmonary arterial blood flow gradient and velocity in the systole and RV

CE

area in the MCT28 group, while treatment with DEC resulted in a significant reduction in these parameters (Table 1).

AC

Tissue lesions of pulmonary hypertension are characterized by changes in all components of the pulmonary arterial walls. The MCT28 group exhibited remarkable changes in the pulmonary arteries, arterioles and in the pulmonary parenchyma, as well as congestion and atelectasis. Treatment with DEC reduced the lung damage (Figure 1A).

Effect of DEC on pulmonary arteriole muscularization

ACCEPTED MANUSCRIPT 11 Vascular remodeling and fibrosis are among the key pathological features in PAH. One of the main features of vascular remodeling seen in PAH is collagen deposition in the remodeled pulmonary vessels. Masson’s trichrome staining revealed a significant increase in collagen deposition in the pulmonary interstitium, around the arteries, vessels and bronchioles. In contrast, after

T

treatment with DEC there was an evident reduction of collagen (Figure 1B), a

SC RI P

finding also observed in the gene expression of COL-1α mRNA (Figure 1E). αSMA is a marker of the expression of a smooth-muscle phenotype, expressed by the PSMCs of existing vessel walls in both the normal and

NU

hypertensive lung. However, an increase in αSMA expression can lead to a thickening of the middle layer of the pulmonary arteries. Immunolabeling of the

MA

lung sections with αSMA revealed a significant increase in the MCT28 group, in comparison with the control group, mainly around the arteries and vessels,

ED

demonstrating the muscularization process. In contrast, the DEC group decreased the expression of αSMA (Figures 1C,D). Those same results were

CE

PT

observed in the gene expression of αSMA (Figure 1F).

Effect of DEC on growth factors

AC

Studies have identified the bone morphogenetic protein (BMP) pathway, a member of the TGF-superfamily of receptors, as having particular importance in PAH pathogenesis, suggesting that this pathway might be important in the pathogenesis of a variety of common clinical situations in which pulmonary hypertension is a feature. Western blot analysis revealed a decrease in BMPR2 in the MCT28 group, compared to the control group. In contrast, treatment with

ACCEPTED MANUSCRIPT 12 DEC increased the BMPR2 level significantly, returning to levels observed in the control group (Figures 2A,B). Models of pulmonary hypertension have been shown to be associated with increased levels of vascular endothelial growth factor (VEGF) transcripts. Dysregulation can cause increased vascular permeability and stimulate

T

neovascularization in the physiological and pathological processes. Western

SC RI P

blot analysis revealed an increase in VEGF in the MCT28 group, compared to the control group, whereas treatment with DEC significantly reduced the VEGF

NU

expression (Figures 2A,B).

Effect of DEC on expression of Apoptotic Pathways proteins

MA

Apoptosis is triggered and modulated by two pathways. The intrinsic pathway involves the mitochondria in response to stress, such as reactive

ED

oxygen species, nutrient deprivation or DNA damage, whereas the extrinsic pathway is induced by receptor binding to pro-apoptotic death ligands such as

PT

tumor necrosis factor–α (TNF-α) and Fas.

CE

The extrinsic pathway markers FADD, caspase 8 and caspase 3 were analyzed by immunohistochemistry and western blotting, through which a

AC

significant reduction of these markers was observed in the MCT group. In contrast, treatment with DEC significantly increased levels of FADD, caspase 8 and caspase 3 (Figures 3 A-E and G-H). The intrinsic pathway markers BAX, cytochrome C and caspase 9 displayed reduced levels of expression, other than Bcl2 which did not show a significant change. Treatment with DEC increased the levels of expression of BAX, cytochrome C and caspase 9, demonstrating that this drug exerts an

ACCEPTED MANUSCRIPT 13 effect on both the intrinsic and the extrinsic pathways of apoptosis (Figure 4 A-H and I-J).

DISCUSSION In the present study, we presented evidence that MCT promotes

T

inactivation of the apoptosis pathway, as well as interfering with growth factors.

SC RI P

The pathobiological mechanisms of PH have been extensively studied. The PH “phenotype” is characterized by endothelial dysfunction, a decreased ratio of apoptosis/proliferation in PASMCs, and a thickened, disordered adventitia in

NU

which there is excessive activation of adventitial metalloproteases. Like cancer and atherosclerosis, PAH does not have a single origin, but a variety of

MA

underlying causes [26].

ED

While recent advances have led to greater recognition and new therapies, relatively few therapies are still used for PH. The MCT model

PT

continues to be a frequently investigated model of PH, as it offers technical simplicity, reproducibility, and low cost compared with other models of PH.

CE

Elucidating the pathobiology of PH continues to be critical for the design of new

AC

and effective therapeutic strategies, and animal models are fundamental to achieving this objective [27]. The C57BL/6J strain has displayed the advantage of presenting a knockout series for several genes, which allows the functional analyze of different cytokines and growth factors in PH development. DEC is a drug that is used against lymphatic filariasis all over the world, however, in recent years many studies have described other pharmacological activities of DEC [11]. Ribeiro et al. (2014) [23] showed that DEC had an antiinflammatory effect in acute lung injury, and Fragoso et al. (2017) [24] found

ACCEPTED MANUSCRIPT 14 that DEC prevented inflammatory cells accumulation and accelerated the inflammation resolution by stimulating apoptosis. Interestingly, a 1985 study tested the hypothesis that monocrotaline would activate arachidonic acid metabolism in rats. These authors described that the arachidonate metabolism was activated before pulmonary hypertension developed, and that inflammatory

T

cell infiltration in the alveolus followed the hypertensive process. Furthermore,

SC RI P

DEC treatment attenuated both monocrotaline-induced inflammatory response and the pulmonary hypertension [28].

Experimental models are important tools for the study of the pathogenic

NU

mechanisms of PH, and for the development of new therapeutic strategies. Established models of PH include chronic exposure to hypoxia, monocrotaline

MA

(MCT), overexpression or knockout mice (IL-6 overexpression, BMPR2 model). Perivascular inflammation is common in the remodeling of blood vessels, both

ED

in animal models and in human PH. The monocrotaline acute toxic model is by

peripheral

accumulation

of

mononuclear

PT

characterized

vascular

damage

inflammatory

caused

cells,

by

without

a

massive

formation

of

CE

obstructive intimal lesions [29]. Our studies expanded on monocrotalineinduced PH previous results by demonstrating that DEC also had an action on

AC

growth and apoptotic factors. Pulmonary arterial hypertension (PAH) is a particularly fatal form of PH that is characterized by increased pulmonary vascular pressure, caused by pathological remodeling [30]. The pathology is associated with abnormal connective tissue deposition and characterized by structural and functional changes in the pulmonary vasculature, including vascular smooth muscle cell

ACCEPTED MANUSCRIPT 15 proliferation hypertrophy and excess collagen formation and remodeling pulmonary vessels [29]. Circulating levels of N-terminal propeptide of type III procollagen (PIIINP), Carboxyterminal telopeptide

of type

I collagen (CITP), matrix

metalloproteinase 9 (MMP-9) and tissue inhibitor of metalloproteinase 1 (TIMP)

T

are elevated in PAH patients, with the results suggesting that the elevated

SC RI P

levels were markers of the state of the disease rather than of the etiology of PAH. Furthermore, circulating markers of new collagen formation, type 1 collagen degradation, elastase (MMP9) activity, and inhibition of matrix

NU

metalloproteinase by a ubiquitous MMP inhibitor (TIMP1) may be indicative of active vascular remodeling and be clinically relevant [31].

MA

The sequence of morphologic changes that lead to neomuscularization of the microvessels has also been seen, and there is a series of studies that

ED

characterize the evolving phenotype of smooth-muscle cells (PSMCs). We studied α-SMA expression, a recognized first marker of developing smooth-

PT

muscle cells during microvessel wall remodeling in both an experimental and

CE

human model [32, 33]. We described that DEC promoted a reduction in the αSMA expression, inhibiting the development of morphological changes in the

AC

pulmonary arteries.

A number of studies have examined the effect of BMPR2 and VEGF in inducing pulmonary artery smooth muscle cell (PASMC) apoptosis in human and experimental models [34, 35, 36]. Bone Morphogenetic Proteins (BMPs) are synthesized and secreted from a variety of cell types including pulmonary vascular smooth muscle and endothelial cells and play an important role in regulating cell proliferation, apoptosis and differentiation [5, 34, 37]. Mutations

ACCEPTED MANUSCRIPT 16 in the gene encoding a bone morphogenetic protein type II receptor (BMPRII) are the most frequent cause of PH. Emerging studies suggest that modulation of BMPRII signaling is a promising alternative that could prevent and reverse pulmonary vascular remodeling [38]. In monocrotaline-induced PH, there was a decrease in levels of BMPR2 expression, consistent with PH development. After

SC RI P

T

treatment with DEC significant increased levels of BMPR2 were evident. Reduced levels of vascular endothelial growth factor (VEGF) in both hypoxia‑ and MCT‑ induced PH models have been described in literature [39,40]. VEGF has been known to confer a potent protective effect on ECs from

NU

apoptosis through the extrinsic pathway [41]. In a variety of experimental PH

MA

models, EC apoptosis has been shown to be associated with reduced levels of VEGF [39,40]. The MCT28 group exhibited high levels of VEGF in protein analysis, demonstrating an increase in these proteins. After DEC treatment a

ED

significant reduction of VEGF levels was observed. Another interesting fact was

PT

imbalance of the extrinsic apoptosis pathway markers, where it was observed that levels of FADD, active caspase-8 and caspase-3 decreased in the MCT28

CE

group in comparison with the control group, whereas after treatment with DEC

AC

these apoptotic proteins had a similar expression to the control group. Similar results were observed in the intrinsic pathway. Some hypotheses have been discussed in relation to apoptosis and lung cells. The lung is the most vascular organ in the body and the massive pulmonary endothelial surface area is almost directly exposed to the environment through the air we breathe [42]. Thus, it is very likely that even in healthy individuals, there are episodes of endothelial cell (EC) injury induced by environmental triggers resulting in waves of apoptosis, but the normal

ACCEPTED MANUSCRIPT 17 reparative mechanisms involving the proliferation and migration of neighboring ECs and possibly the homing of circulating endothelial progenitor cells are sufficient to restore vascular continuity and maintain the integrity of pulmonary circulation [43]. Endothelial cells show an apoptosis-resistant phenotype in idiopathic

T

pulmonary hypertension [4]. Recently, a study using a thromboembolic

SC RI P

pulmonary hypertension rodent model confirmed the imbalance between proapoptotic and anti-apoptotic proteins. The thromboembolism led to the upregulation of Bad and the down-regulation of Bcl-2, associated to decreased

NU

mRNA and protein levels of FoxO1, a member of the FoxO family that plays important role in cell cycle, proliferation, apoptosis, and tumorigenesis [44].

MA

It has been demonstrated that increased PASMC proliferation and/or inhibited PASMCS apoptosis both contribute to the inducing of pulmonary

ED

vascular medial hypertrophy. However, the precise mechanisms involved in the regulation of PASMC proliferation and apoptosis in PH are still incompletely

PT

understood. Another important factor is that the BMPR2 pathway prevents EC

CE

apoptosis and maintains the integrity of the lung microvascular cells, and thus inactivating BMPR2 mutations play a crucial role in triggering pathological

AC

vascular remodeling [5].

The present work has some limitations, although a monocrotalineinduced pulmonary hypertension has been effectively established in C57Bl-6 mice in this study, ECS and PASMCS were difficult to isolate and in vitro assays have not been performed. Therefore, functional assays to characterize the mechanism of DEC on apoptosis/proliferation and pulmonary remodeling have been left to be investigated further.

ACCEPTED MANUSCRIPT 18 In conclusion, the results of the present study indicate that DEC attenuates PH in an experimental induced-monocrotaline model by inhibiting a series of markers involved in apoptosis resistance that plays an important role in pulmonary vascular remodeling.

T

ACKNOWLEDGEMENTS: The present study was supported by the following

SC RI P

Brazilian fostering agencies: Fundação de Amparo à Ciência e Tecnologia do Estado de Pernambuco (FACEPE), Centro de Pesquisas Aggeu Magalhães

NU

(CPqAM/FIOCRUZ) and National Center Structural Biology and Bio-imaging.

1-

MA

REFERENCES

McLaughlin VV, Archer SL, Badesch DB, Barst RJ, Farber HW, Lindner

ED

JR, Mathier MA, McGoon MD, Park MH, Rosenson RS, Rubin LJ, Tapson VF,

PT

Varga J; American College of Cardiology Foundation Task Force on Expert Consensus Documents; American Heart Association; American College of

CE

Chest Physicians; American Thoracic Society, Inc; Pulmonary Hypertension Association. ACCF/AHA 2009 expert consensus document on pulmonary

AC

hypertension a report of the American College of Cardiology Foundation Task Force on Expert Consensus Documents and the American Heart Association developed in collaboration with the American College of Chest Physicians; American Thoracic Society, Inc.; and the Pulmonary Hypertension Association. J Am Coll Cardiol. 2009 Apr 28;53(17):1573-619.

ACCEPTED MANUSCRIPT 19 2-

Rosenkranz

S.

Pulmonary

hypertension 2015: current definitions,

terminology, and novel treatment options. Clin Res Cardiol. 2015, 104(3):197– 207. 3 - Maron BA, Loscalzo J. Pulmonary hypertension: pathophysiology and

T

signaling pathways. Handb Exp Pharmacol. 2013, 218:31–58.

SC RI P

4 - Jin Y, Choi AM. Cross talk between autophagy and apoptosis in pulmonary hypertension. Pulm Circ. 2012 Oct;2(4):407-14.

5 - Guignabert C, Dorfmuller P. Pathology and pathobiology of pulmonary

NU

hypertension. Semin Respir Crit Care Med. 2013 Oct;34(5):551-9.

MA

6- Rajagopalan N, Simon MA, Suffoletto MS, Shah H, Edelman K, Mathier MA, López-Candales A. Noninvasive estimation of pulmonary vascular resistance in

ED

pulmonary hypertension. Echocardiography. 2009 May;26(5):489-94. 7- Ruiter G, de Man FS, Schalij I, Sairras S, Grünberg K, Westerhof N, van der

PT

Laarse WJ, Vonk-Noordegraaf A. Reversibility of the monocrotaline pulmonary

Price LC, Wort SJ, Perros F, Dorfmüller P, Huertas A, Montani D, Cohen-

AC

8-

CE

hypertension rat model. Eur Respir J. 2013 Aug;42(2):553-6.

Kaminsky S, Humbert M. Inflammation in pulmonary arterial hypertension. Chest. 2012 Jan;141(1):210-221. 9-

Schultze

AE,

Roth

RA,.

Chronic

pulmonary

hypertension–the

monocrotalina model and involvement of the hemostatic system. J Toxicol Environ Health B Crit Rev.1998, 1: 271–346.

ACCEPTED MANUSCRIPT 20 10-

Molteni

A,

Ward

WF,

Ts'ao

CH,

Solliday

NH.

Monocrotaline

pneumotoxicity in mice. Virchows Arch B Cell Pathol Incl Mol Pathol. 1989;57(3):149-55. 11-

Peixoto CA, Silva BS. Anti-inflammatory effects of diethylcarbamazine: a

Santos LA, Ribeiro EL, Barbosa KP, Fragoso IT, Gomes FO, Donato MA,

SC RI P

12-

T

review. Eur J Pharmacol. 2014 Jul 5;734:35-41.

Silva BS, Silva AK, Rocha SW, França ME, Rodrigues GB, Silva TG, Peixoto CA. Diethylcarbamazine inhibits NF-κB activation in acute lung injury induced

13-

NU

by carrageenan in mice. Int Immunopharmacol. 2014 Nov;23(1):153-62. Boggild AK, Keystone JS, Kain KC. Tropical pulmonary eosinophilia: a

MA

case series in a setting of nonendemicity. Clin Infect Dis. 2004 Oct

14-

ED

15;39(8):1123-8.

Morganroth ML, Stenmark KR, Morris KG, Murphy RC, Mathias M,

pulmonary

hypertension

in

awake

rats.

Am

Rev

Respir

Dis.

1985

CE

Apr;131(4):488-92.

Queto T, Xavier-Elsas P, Gardel MA, de Luca B, Barradas M, Masid D, E

AC

15-

PT

Reeves JT, Voelkel NF. Diethylcarbamazine inhibits acute and chronic hypoxic

Silva PM, Peixoto CA, Vasconcelos ZM, Dias EP, Gaspar-Elsas MI. Inducible nitric oxide synthase/CD95L-dependent suppression of pulmonary and bone marrow eosinophilia by diethylcarbamazine. Am J Respir Crit Care Med. 2010 Mar 1;181(5):429-37.

ACCEPTED MANUSCRIPT 21 16-

Zuo L, Christofi FL, Wright VP, Bao S, Clanton TL. Lipoxygenase-

dependent superoxide release in skeletal muscle. J Appl Physiol (1985). 2004 Aug;97(2):661-8. Epub 2004 Apr 23. 17-

Kumar

S, Wei

C, Thomas

CM, Kim IK, Seqqat R, Kumar R, Baker

T

KM, Jones WK, Gupta S. Cardiac-specific genetic inhibition of nuclear factor-κB

SC RI P

prevents right ventricular hypertrophy induced by monocrotaline. Am J Physiol Heart Circ Physiol. 2012 Apr 15;302(8):H1655-66.

18- Li L, Wei C, Kim IK, Janssen-Heininger Y, Gupta S. Inhibition of nuclear

NU

factor-κB in the lungs prevents monocrotaline-induced pulmonary hypertension in mice. Hypertension. 2014 Jun;63(6):1260-9.

increased

in

MA

19- Seta F, Rahmani M, Turner PV, Funk CD. Pulmonary oxidative stress is cyclooxygenase-2

knockdown

mice

with

mild

pulmonary

ED

hypertension induced by monocrotaline. PLoS One. 2011;6(8):e23439.

PT

20- Dumitrascu R, Koebrich S, Dony E, Weissmann N, Savai R, Pullamsetti SS, Ghofrani HA, Samidurai A, Traupe H, Seeger W, Grimminger F, Schermuly RT.

CE

Characterization of a murine model of monocrotaline pyrrole-induced acute lung

AC

injury. BMC Pulm Med. 2008 Dec 17;8:25. 21- Susumu Hosokawa, Go Haraguchi, Akihito Sasaki, Hirokuni Arai, Susumu Muto, Akiko Itai, Shozaburo Doi, Shuki Mizutani, and Mitsuaki Isobe. Pathophysiological roles of nuclear factor kappaB (NF-kB) in pulmonary arterial hypertension:

effects

of

synthetic

selective

0354.Cardiovascular Research 2013 99, 35–43.

NF-kB

inhibitor

IMD-

ACCEPTED MANUSCRIPT 22 22 - Bossone E, D'Andrea A, D'Alto M, Citro R, Argiento P, Ferrara F, Cittadini A, Rubenfire M, Naeije R. Echocardiography in pulmonary arterial hypertension: from diagnosis to prognosis. J Am Soc Echocardiogr. 2013 Jan;26(1):1-14. 23-

Ribeiro EL, Barbosa KP, Fragoso IT, Donato MA, Gomes FO, da Silva

T

BS, Soares e Silva AK, Rocha SW, da Silva Junior VA, Peixoto CA.

SC RI P

Diethylcarbamazine Attenuates the Development of Carrageenan-Induced Lung Injury in Mice. Mediators Inflammation. 2014; 2014: 105120. 24-

Fragoso IT, Ribeiro EL, Gomes FO, Donato MA, Silva AK, Oliveira AC,

NU

Araújo SM, Barbosa KP, Santos LA, Peixoto CA. Diethylcarbamazine attenuates LPS-induced acute lung injury in mice by apoptosis of inflammatory

MA

cells. Pharmacol Rep. 2017 Feb;69(1):81-89. 25-

Liu Y, Wu H, Nie YC, Chen JL, Su WW, Li PB. Naringin attenuates acute

lung

injury

LPS-treated

mice

ED

in

by

inhibiting

NF-κB

pathway.

Int

26-

PT

Immunopharmacol. 2011 Oct;11(10):1606-12. Farber HW, Loscalzo J Pulmonary arterial hypertension. N Engl J Med.

Gomez-Arroyo JG, Farkas L, Alhussaini AA, Farkas D, Kraskauskas D,

AC

27-

CE

2004, 351(16):1655–1665.

Voelkel NF, Bogaard HJ. The monocrotaline model of pulmonary hypertension in perspective. Am J Physiol Lung Cell Mol Physiol. 2012 302(4):L363-9. 28-

Stenmark KR, Morganroth ML, Remigio LK, Voelkel NF, Murphy RC,

Henson PM, Mathias MM, Reeves JT. Alveolar inflammation and arachidonate metabolism in monocrotaline-induced pulmonary hypertension. Am J Physiol. 1985 Jun;248(6 Pt 2):H859-66.

ACCEPTED MANUSCRIPT 23 29- Stenmark KR, Meyrick B, Galie N, Mooi WJ, McMurtry IF. Animal models of pulmonary arterial hypertension: the hope for etiological discovery and pharmacological

cure.

Am

J

Physiol

Lung

Cell

Mol

Physiol.

2009

Dec;297(6):L1013-32.

pulmonary

arterial hypertension. Nat Rev Cardiol. 2011

21;8(8):443-55. 31-

Jun

SC RI P

disease:

T

30- Schermuly RT, Ghofrani HA, Wilkins MR, Grimminger F. Mechanisms of

Safdar Z, Tamez E, Chan W, Arya B, Ge Y, Deswal A, Bozkurt B, Frost A

NU

Entman M4. Circulating collagen biomarkers as indicators of disease severity in pulmonary arterial hypertension. JACC Heart Fail. 2014 Aug;2(4):412-21. Jones R, Jacobson M, Steudel W. alpha-smooth-muscle actin and

MA

32-

microvascular precursor smooth-muscle cells in pulmonary hypertension. Am J

Kawai-Kowase K, Owens GK. Multiple repressor pathways contribute to

PT

33-

ED

Respir Cell Mol Biol. 1999 Apr;20(4):582-94.

phenotypic switching of vascular smooth muscle cells. Am J Physiol Cell

Zhang S, Fantozzi I, Tigno DD, Yi ES, Platoshyn O, Thistlethwaite PA,

AC

34-

CE

Physiol. 2007 Jan;292(1):C59-69. Epub 2006 Sep 6.

Kriett JM, Yung G, Rubin LJ, Yuan JX. Bone morphogenetic proteins induce apoptosis in human pulmonary vascular smooth muscle cells. Am J Physiol Lung Cell Mol Physiol. 2003 Sep;285(3):L740-54. 35-

Morty RE, Nejman B, Kwapiszewska G, Hecker M, Zakrzewicz A, Kouri

FM, Peters DM, Dumitrascu R, Seeger W, Knaus P, Schermuly RT, Eickelberg O. Dysregulated bone morphogenetic protein signaling in monocrotaline-

ACCEPTED MANUSCRIPT 24 induced pulmonary arterial hypertension. Arterioscler Thromb Vasc Biol. 2007 May;27(5):1072-8. 36-

Takahashi H, Goto N, Kojima Y, Tsuda Y, Morio Y, Muramatsu M,

Fukuchi Y. Downregulation of type II bone morphogenetic protein receptor in

T

hypoxic pulmonary hypertension. Am J Physiol Lung Cell Mol Physiol. 2006

37-

SC RI P

Mar;290(3):L450-8.

Song Y, Coleman L, Shi J, Beppu H, Sato K, Walsh K, Loscalzo J, Zhang

YY. Inflammation, endothelial injury, and persistent pulmonary hypertension in

NU

heterozygous BMPR2-mutant mice. Am J Physiol Heart Circ Physiol. 2008 Aug;295(2):H677-90.

MA

38 - Orriols M, Gomez-Puerto MC, Ten Dijke P., 2017. BMP type II receptor as

39-

ED

a therapeutic target in pulmonary arterial hypertension. Cell Mol Life Sci. 26. Partovian C, Adnot S, Eddahibi S, Teiger E, Levame M, Dreyfus P,

PT

Raffestin B, Frelin C. Heart and lung VEGF mRNA expression in rats with monocrotaline- or hypoxia-induced pulmonary hypertension. Am J Physiol. 1998

Arcot SS, Lipke DW, Gillespie MN, Olson JW. Alterations of growth

AC

40-

CE

Dec;275(6 Pt 2):H1948-56.

factor transcripts in rat lungs during development of monocrotaline-induced pulmonary hypertension. Biochem Pharmacol. 1993 Sep 14;46(6):1086-91. 41 -

Chen YH, Wu HL, Chen CK, Huang YH, Yang BC, Wu LW. Angiostatin

antagonizes the action of VEGF‑A in human endothelial cells via two distinct pathways. Biochem Biophys Res Commun. 2003, 310:804‑10.

ACCEPTED MANUSCRIPT 25 42-

Henson PM, Tuder RM. Apoptosis in the lung: induction, clearance and

detection. Am J Physiol Lung Cell Mol Physiol. 2008 Apr;294(4):L601-11. 43 -

Jurasz P, Courtman D, Babaie S, Stewart DJ. Role of apoptosis in

pulmonary hypertension: from experimental models to clinical trials. Pharmacol

T

Ther. 2010 Apr;126(1):1-8.

SC RI P

44 - Deng C, Zhong Z, Wu D, Chen Y, Lian N, Ding H, Zhang Q, Lin Q, Wu S. Role of FoxO1 and apoptosis in pulmonary vascular remolding in a rat model of chronic

thromboembolic

pulmonary

hypertension.

Rep.

2017

May

MA

NU

23;7(1):2270.

Sci

Legends

ED

Table 1: Pulmonary artery blood velocity after DEC treatment: Velocity-time integral (VTI, cm) mean and peak gradient (mmHg) and mean and peak velocity

PT

(mm/s) of blood flow in the pulmonary artery and area of the right ventricle (mm2) were measured from Doppler waveforms acquired by ultrasound

AC

CE

imaging. N=5. Mean±SD; p<0.05 vs SHAM, #p<0.05 vs MCT28

Fig. 1: Effect of DEC treatment on histological alterations in lung after MCT-induced pulmonary hypertension. A: Representative images of H&E staining demonstrating, interstitial edema with thickening of the septum alveolar, infiltrates of inflammatory cells with the presence of activated macrophages and plexiform lesions of pulmonary arteries and emphysema in MCT28 groups. Administration of DEC significantly attenuated the lung damage. Histological analysis of the control group did not reveal any morphological changes. B:

ACCEPTED MANUSCRIPT 26 Representative

images

of

Masson’s

trichrome

staining

demonstrating

significantly increased collagen deposition and MCT28 group. Administration of DEC significantly reduced collagen. C: Immunohistochemical localization of αSMA labeling around the arteries, vessels and bronchioles and MCT28 group. After treatment with DEC there was a reduction of immunostaining for α-SMA.

T

D: Quantitative densitometry analysis (GIMP2 analyzed) α-SMA. Data is

SC RI P

expressed as mean ± S.D. from n = 5 mice for each group. E e F: Relative expression of mRNA COL-1α and αSMA showing a significant increase in the MCT28 group. After treatment with DEC there was a reduction of expression

NU

COL-1α and αSMA respectively. Data is expressed as mean ± S.D. from n = 5 mice for each group. The letter at the top of the columns represents the groups

MA

where there was a significant difference with the cited group: a - CONT; b –

ED

MCT28; c- MCT28/DEC (p < 0.05).

Fig. 2: Effect of DEC treatment on expression of BMPR2 and VEGF: A:

PT

Representative western blotting protein expression for BMPR2 and VEGF

CE

respectively. Data is expressed as mean ± S.D from n = 3 mice for each group. B: Quantitative densitometry analysis (ImageJ analyzed). The letter on the top

AC

of the columns represents the groups where there was a significant difference with the cited group: a - CONT; b – MCT28; c- MCT28/DEC (p < 0.05).

Fig. 3: Effect of DEC treatment on the extrinsic pathway of apoptosis: Immunohistochemical localization for FADD, C8 and C3 (A, B and C respectively). D-F: Quantitative densitometry analysis (GIMP2 analyzed). Data is expressed as mean ± S.D. from n = 5 mice for each group. G: Representative

ACCEPTED MANUSCRIPT 27 Western blotting showing protein expression for FADD, C8 and C3 in the CONT, MCT28 and MCT28/DEC groups. β-actin was used as an internal loading control. Data is expressed as mean ± S.D. from n = 3 mice for each group. H: Quantitative densitometry analysis (ImageJ analyzed). The letter on the top of the columns represents the groups where there was a significant

T

difference with the cited group: a - CONT; b – MCT28; c- MCT28/DEC (p <

SC RI P

0.05).

Fig. 4: Effect of DEC treatment on the intrinsic pathway of apoptosis:

NU

Immunohistochemical localization for BAX, BCL2, C9 and Cytocrome C (A, B, C and D respectively). E-H: Quantitative densitometry analysis (GIMP2

MA

analyzed). Data is expressed as mean ± S.D. from n = 5 mice for each group. ND: not detected I: Representative Western blotting showing protein expression

ED

for BAX, BCL2, C9 and Cytocrome C in the CONT, MCT28 and MCT28/DEC groups. β-actin was used as an internal loading control. Data is expressed as

PT

mean ± S.D. from n = 3 mice for each group. H: Quantitative densitometry

CE

analysis (ImageJ analyzed). The letter on the top of the columns represents the groups where there was a significant difference with the cited group: a - CONT;

AC

b – MCT28; c- MCT28/DEC (p < 0.05).

ACCEPTED MANUSCRIPT

MA

NU

SC RI P

T

28

AC

CE

PT

ED

Figure 1

ACCEPTED MANUSCRIPT

SC RI P

T

29

AC

CE

PT

ED

MA

NU

Figure 2

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA

NU

SC RI P

T

30

ACCEPTED MANUSCRIPT

ED

MA

NU

SC RI P

T

31

AC

CE

PT

Figure 3

ACCEPTED MANUSCRIPT

AC

CE

PT

ED

MA

NU

SC RI P

T

32

ACCEPTED MANUSCRIPT

ED

MA

NU

SC RI P

T

33

AC

CE

PT

Figure 4

ACCEPTED MANUSCRIPT 34

VTI, cm

2.27 ± 0.4 4.28 ± 0.35* 3.15 ± 0.24#

Peak Gradient, mmHg

Mean Velocity, mm/s

Peak velocity, mm/s

Area RV (mm2 )

0.84 ± 0.22 3.40 ± 0.69* 2.42 ± 0.45#

268.0 ±49.49 565.5± 42.39* 461.8± 40.48#

473.4± 86.49 959.2± 67.87* 775.1± 75.80#

11.40 ± 0.52 17.57 ± 0.97* 12.78 ± 1.63#

CE

PT

ED

MA

NU

SC RI P

T

Table 1

AC

SHAM MCT28 MCT28/DEC

Mean Gradient, mmHg 0.29 ± 0.1 1.19 ± 0.22* 0.84 ± 0.15#

ACCEPTED MANUSCRIPT 35 Highlights

CE

PT

ED

MA

NU

SC RI P

T

First study on the therapeutic effects of diethylcarbamazine in lung hypertension. Lung hypertension exhibited reductions apoptosis extrinsic and intrinsic pathways Diethylcarbamazine inhibited markers involved in cell proliferation/death.

AC

  