Preconception Exposure to Fine Particulate Matter Leads to Cardiac Dysfunction in Adult Male Offspring

Vineeta Tanwar, Jeremy M Adelstein, Jacob A Grimmer, Dane J Youtz, Aashish Katapadi, Benjamin P Sugar, Michael J Falvo, Lisa A Baer, Kristin I Stanford, Loren E Wold, Vineeta Tanwar, Jeremy M Adelstein, Jacob A Grimmer, Dane J Youtz, Aashish Katapadi, Benjamin P Sugar, Michael J Falvo, Lisa A Baer, Kristin I Stanford, Loren E Wold

Abstract

Background Particulate matter (particles < 2.5 μm [ PM 2.5]) exposure during the in utero and postnatal developmental periods causes cardiac dysfunction during adulthood. Here, we investigated the potential priming effects of preconception exposure of PM 2.5 on cardiac function in adult offspring. Methods and Results Male and female friend leukemia virus b (FVB) mice were exposed to either filtered air ( FA ) or PM 2.5 at an average concentration of 38.58 μg/m3 for 6 hours/day, 5 days/week for 3 months. Mice were then crossbred into 2 groups: (1) FA male× FA female (both parents were exposed to FA preconception) and, (2) PM 2.5male× PM 2.5female (both parents were exposed to PM 2.5 preconception). Male offspring were divided: (1) preconception FA (offspring born to FA exposed parents) and, (2) preconception PM 2.5 (offspring born to PM 2.5 exposed parents) and analyzed at 3 months of age. Echocardiography identified increased left ventricular end systolic volume and reduced posterior wall thickness, reduced %fractional shortening and %ejection fraction in preconception PM 2.5 offspring. Cardiomyocytes isolated from preconception PM 2.5 offspring showed reduced %peak shortening, -dL/dT, TPS 90 and slower calcium reuptake (tau). Gene and protein expression revealed modifications in markers of inflammation ( IL -6, IL -15, TNF α, NF қB, CRP , CD 26E, CD 26P, intercellular adhesion molecule 1, and monocyte chemoattractant protein-1) profibrosis (collagen type III alpha 1 chain), oxidative stress ( NOS 2), antioxidants (Nrf2, SOD , catalase), Ca2+ regulatory proteins ( SERCA 2a, p- PLN , NCX ), and epigenetic regulators (Dnmt1, Dnmt3a, Dnmt3b, Sirt1, and Sirt2) in preconception PM 2.5 offspring. Conclusions Preconception exposure to PM 2.5 results in global cardiac dysfunction in adult offspring, suggesting that abnormalities during development are not limited to the prenatal or postnatal periods but can also be determined before conception.

Keywords: calcium; cardiovascular function; inflammation; myocyte; particulate matter; preconception.

Figures

Figure 1
Figure 1
Body weight parameters from mice born to preconception FA or PM 2.5‐exposed parents. A, Birth weight (n=42–49 pups/group), (B) body weight (n=12–13 mice/group), (C) body surface area (BSA) (n=12–13 mice/group), (D) heart weight (n=12–13 mice/group), (E) heart weight/body weight (HW/BW) (n=12–13 mice/group), (F) heart weight/tibial length (HW/TL) (n=12–13). Data are expressed as ±SEM. *P<0.05, ***P<0.001 vs preconception FA controls. A, Represents birth weight of all offspring (irrespective of sex). B through F, Represent parameters that were analyzed in 3‐month‐old male offspring only. PC FA indicates preconception filtered air; PC PM2.5, preconception particulate matter (<2.5 μm in diameter).
Figure 2
Figure 2
Transthoracic echocardiography to assess cardiac function of 3‐month‐old mice that were born to preconception FA or PM 2.5 exposed parents. A, LVESd, (B) PWTs, (C) %FS, (D) %EF. Data were collected from 10 to 12 mice in each group. Five beat cycles were captured and 3 loops averaged per assessment. Data are expressed as ±SEM. %EF indicates %ejection fraction; %FS, %fractional shortening; LVESd, left ventricular end‐diastolic dimension; PC FA, preconception filtered air; PC PM2.5, preconception particulate matter (<2.5 μm in diameter); PWTs, posterior wall thickness during systole. *P<0.05, **P<0.01 vs PC FA controls.
Figure 3
Figure 3
In vitro cardiomyocyte functional and calcium signaling parameters obtained from 3‐month‐old mice that were born to preconception filtered air or particulate matter (A) %peak shortening (%PS), (B) negative velocity (‐dL/dT), (C) time‐to‐peak shortening (TPS90), (D) fluorescence decay rate (τ), (E) calcium transient amplitude (Δ340/380). Data were collected from 70 to 80 cardiomyocytes isolated from 9 to 10 mice per group taken from individual litters. Data are expressed as mean±SEM. %PS indicates %peak shortening; ‐dL/dT, negative velocity; PC FA indicates preconception filtered air; PC PM2.5, preconception particulate matter (<2.5 μm in diameter); TPS90, time‐to‐peak shortening. *P<0.05, **P<0.01 vs PC FA controls.
Figure 4
Figure 4
Western blot analysis showing expression Ca2+ handling proteins (A) SERCA‐2A, (B) p‐PLN; (C) NCX. Representative Western blot shown above and quantification below. Data were obtained from 3‐month‐old mouse hearts (n=6 mice/group) that were born to preconception filtered air or particulate matter (<2.5 μm in diameter) exposed parents and expressed as mean±SEM. NCX indicates Na+/Ca2+ exchanger; PC FA indicates preconception filtered air; PC PM2.5, preconception particulate matter (<2.5 μm in diameter); p‐PLN, phosphorylated phospholamban; SERCA‐2A, sarco/endoplasmic reticulum Ca2+‐ATPase. **P<0.01, ***P<0.001 vs PC FA controls.
Figure 5
Figure 5
A, Western blot analysis showing expression of NOS‐2. Representative Western blot is shown on the top. Quantitative polymerase chain reaction analysis showing expression of (B) Nrf2, (C) SOD, (D) CAT. Data were obtained from 3‐month‐old mouse hearts (n=6 mice/group) that were born to preconception FA or PM 2.5 exposed parents and expressed as mean±SEM. CAT indicates catalase; NOS‐2, nitric oxide synthase 2; Nrf2, nuclear factor (erythroid‐derived 2)‐like 2; SOD, superoxide dismutase; PC FA, preconception filtered air; PC PM2.5, preconception particulate matter (<2.5 μm in diameter). *P<0.05, **P<0.01 vs preconception FA controls.
Figure 6
Figure 6
Quantitative polymerase chain reaction expression of (A) IL‐6, (B) IL‐15, (C) TNF‐α, (D) NF‐κB, (E) CRP, (F) E‐selectin (CD26E), (G) P‐selectin (CD26P), (H) ICAM‐1. I, Western blot analysis showing expression of MCP‐1. Representative Western blot was shown on the right and quantification on the left. Data were obtained from 3‐month‐old mouse hearts (n=6 mice/group) that were born to preconception FA or PM 2.5 exposed parents and expressed as mean±SEM. CRP indicates C‐reactive protein; ICAM1, intercellular adhesion molecule 1; IL‐6, interleukin 6; IL‐15, interleukin 15; MCP‐1, monocyte chemoattractant protein‐1; NF‐κB, nuclear factor kappa‐light‐chain‐enhancer of activated B cells; PC FA, preconception filtered air; PC PM2.5, preconception particulate matter (<2.5 μm in diameter); TNF‐α, tumor necrosis factor‐alpha. **P<0.01, ***P<0.001 vs PC FA controls.
Figure 7
Figure 7
A, Quantitative polymerase chain reaction expression of collagen3a1 (Col3a1) and (B) protein expression of Col3a1 via Western blot analysis. Representative Western blot is shown above and quantification below. Data were obtained from 3‐month‐old mouse hearts (n=6 mice/group) that were born to preconception FA or PM 2.5‐exposed parents and expressed as mean±SEM. PC FA indicates preconception filtered air; PC PM2.5, preconception particulate matter (<2.5 μm in diameter). *P<0.05, **P<0.01 vs PC FA controls.
Figure 8
Figure 8
Western blot analysis showing expression of (A) DNA methyltransferases‐1 (Dnmt1) and (B) DNA methyltransferases‐3a (Dnmt3a). Representative Western blots are shown on the left and quantification on the right. Quantitative polymerase chain reaction analysis showing expression of (C) Sirt1 and (D) Sirt2. Data were obtained from 3‐month‐old mouse hearts (n=6 mice/group) that were born to preconception FA or PM 2.5‐exposed parents and expressed as mean±SEM. Dnmt1 indicates DNA methyltransferases‐1; Dnmt3a, DNA methyltransferases; PC FA, preconception filtered air; PC PM2.5, preconception particulate matter (<2.5 μm in diameter); Sirt1, sirtuin 1; Sirt2, sirtuin 2. *P<0.05, **P<0.01 vs PC FA controls.

References

    1. Thurston G, Lippmann M. Ambient particulate matter air pollution and cardiopulmonary diseases. Semin Respir Crit Care Med. 2015;36:422–432.
    1. Franklin BA, Brook R, Arden Pope C III. Air pollution and cardiovascular disease. Curr Probl Cardiol. 2015;40:207–238.
    1. Wang T, Lang GD, Moreno‐Vinasco L, Huang Y, Goonewardena SN, Peng YJ, Svensson EC, Natarajan V, Lang RM, Linares JD, Breysse PN, Geyh AS, Samet JM, Lussier YA, Dudley S, Prabhakar NR, Garcia JG. Particulate matter induces cardiac arrhythmias via dysregulation of carotid body sensitivity and cardiac sodium channels. Am J Respir Cell Mol Biol. 2012;46:524–531.
    1. Byrd JB, Morishita M, Bard RL, Das R, Wang L, Sun Z, Spino C, Harkema J, Dvonch JT, Rajagopalan S, Brook RD. Acute increase in blood pressure during inhalation of coarse particulate matter air pollution from an urban location. J Am Soc Hypertens. 2016;10:133–139.e4.
    1. Argacha JF, Collart P, Wauters A, Kayaert P, Lochy S, Schoors D, Sonck J, de Vos T, Forton M, Brasseur O, Beauloye C, Gevaert S, Evrard P, Coppieters Y, Sinnaeve P, Claeys MJ. Air pollution and ST‐elevation myocardial infarction: a case‐crossover study of the Belgian STEMI registry 2009–2013. Int J Cardiol. 2016;223:300–305.
    1. Tanwar V, Adelstein JM, Grimmer JA, Youtz DJ, Sugar BP, Wold LE. PM2.5 exposure in utero contributes to neonatal cardiac dysfunction in mice. Environ Pollut. 2017;230:116–124.
    1. Tanwar V, Gorr MW, Velten M, Eichenseer CM, Long VP III, Bonilla IM, Shettigar V, Ziolo MT, Davis JP, Baine SH, Carnes CA, Wold LE. In utero particulate matter exposure produces heart failure, electrical remodeling, and epigenetic changes at adulthood. J Am Heart Assoc. 2017;6:e005796 DOI: 10.1161/JAHA.117.005796.
    1. Guerrero‐Bosagna C, Settles M, Lucker B, Skinner MK. Epigenetic transgenerational actions of vinclozolin on promoter regions of the sperm epigenome. PLoS One. 2010;5:e13100.
    1. Zama AM, Uzumcu M. Fetal and neonatal exposure to the endocrine disruptor methoxychlor causes epigenetic alterations in adult ovarian genes. Endocrinology. 2009;150:4681–4691.
    1. Shi L, Wu J. Epigenetic regulation in mammalian preimplantation embryo development. Reprod Biol Endocrinol. 2009;7:59.
    1. Robledo CA, Yeung E, Mendola P, Sundaram R, Maisog J, Sweeney AM, Barr DB, Louis GM. Preconception maternal and paternal exposure to persistent organic pollutants and birth size: the LIFE study. Environ Health Perspect. 2015;123:88–94.
    1. Rappazzo KM, Daniels JL, Messer LC, Poole C, Lobdell DT. Exposure to fine particulate matter during pregnancy and risk of preterm birth among women in New Jersey, Ohio, and Pennsylvania, 2000–2005. Environ Health Perspect. 2014;122:992–997.
    1. Dadvand P, Parker J, Bell ML, Bonzini M, Brauer M, Darrow LA, Gehring U, Glinianaia SV, Gouveia N, Ha EH, Leem JH, van den Hooven EH, Jalaludin B, Jesdale BM, Lepeule J, Morello‐Frosch R, Morgan GG, Pesatori AC, Pierik FH, Pless‐Mulloli T, Rich DQ, Sathyanarayana S, Seo J, Slama R, Strickland M, Tamburic L, Wartenberg D, Nieuwenhuijsen MJ, Woodruff TJ. Maternal exposure to particulate air pollution and term birth weight: a multi‐country evaluation of effect and heterogeneity. Environ Health Perspect. 2013;121:267–373.
    1. Cheung MC, Spalding PB, Gutierrez JC, Balkan W, Namias N, Koniaris LG, Zimmers TA. Body surface area prediction in normal, hypermuscular, and obese mice. J Surg Res. 2009;153:326–331.
    1. Wold LE, Ying Z, Hutchinson KR, Velten M, Gorr MW, Velten C, Youtz DJ, Wang A, Lucchesi PA, Sun Q, Rajagopalan S. Cardiovascular remodeling in response to long‐term exposure to fine particulate matter air pollution. Circ Heart Fail. 2012;5:452–461.
    1. Watkins AJ, Lucas ES, Wilkins A, Cagampang FR, Fleming TP. Maternal periconceptional and gestational low protein diet affects mouse offspring growth, cardiovascular and adipose phenotype at 1 year of age. PLoS One. 2011;6:e28745.
    1. Gardebjer EM, Cuffe JS, Pantaleon M, Wlodek ME, Moritz KM. Periconceptional alcohol consumption causes fetal growth restriction and increases glycogen accumulation in the late gestation rat placenta. Placenta. 2014;35:50–57.
    1. Witt WP, Mandell KC, Wisk LE, Cheng ER, Chatterjee D, Wakeel F, Park H, Zarak D. Infant birthweight in the US: the role of preconception stressful life events and substance use. Arch Womens Ment Health. 2016;19:529–542.
    1. Bell ML, Belanger K, Ebisu K, Gent JF, Lee HJ, Koutrakis P, Leaderer BP. Prenatal exposure to fine particulate matter and birth weight: variations by particulate constituents and sources. Epidemiology. 2010;21:884–891.
    1. Kjoller‐Hansen L, Steffensen R, Grande P. Beneficial effects of ramipril on left ventricular end‐diastolic and end‐systolic volume indexes after uncomplicated invasive revascularization are associated with a reduction in cardiac events in patients with moderately impaired left ventricular function and no clinical heart failure. J Am Coll Cardiol. 2001;37:1214–1220.
    1. Migrino RQ, Young JB, Ellis SG, White HD, Lundergan CF, Miller DP, Granger CB, Ross AM, Califf RM, Topol EJ. End‐systolic volume index at 90 to 180 minutes into reperfusion therapy for acute myocardial infarction is a strong predictor of early and late mortality. The Global Utilization of Streptokinase and t‐PA for Occluded Coronary Arteries (GUSTO)‐I Angiographic Investigators. Circulation. 1997;96:116–121.
    1. Velten M, Gorr MW, Youtz DJ, Velten C, Rogers LK, Wold LE. Adverse perinatal environment contributes to altered cardiac development and function. Am J Physiol Heart Circ Physiol. 2014;306:H1334–H1340.
    1. Yu X, Kennedy RH, Liu SJ. JAK2/STAT3, not ERK1/2, mediates interleukin‐6‐induced activation of inducible nitric‐oxide synthase and decrease in contractility of adult ventricular myocytes. J Biol Chem. 2003;278:16304–16309.
    1. Feng Q, Lu X, Jones DL, Shen J, Arnold JM. Increased inducible nitric oxide synthase expression contributes to myocardial dysfunction and higher mortality after myocardial infarction in mice. Circulation. 2001;104:700–704.
    1. Umar S, van der Laarse A. Nitric oxide and nitric oxide synthase isoforms in the normal, hypertrophic, and failing heart. Mol Cell Biochem. 2010;333:191–201.
    1. Zimmet JM, Hare JM. Nitroso‐redox interactions in the cardiovascular system. Circulation. 2006;114:1531–1544.
    1. Xu X, Liu C, Xu Z, Tzan K, Zhong M, Wang A, Lippmann M, Chen LC, Rajagopalan S, Sun Q. Long‐term exposure to ambient fine particulate pollution induces insulin resistance and mitochondrial alteration in adipose tissue. Toxicol Sci. 2011;124:88–98.
    1. Glorieux C, Zamocky M, Sandoval JM, Verrax J, Calderon PB. Regulation of catalase expression in healthy and cancerous cells. Free Radic Biol Med. 2015;87:84–97.
    1. Yano S, Yano N. Regulation of catalase enzyme activity by cell signaling molecules. Mol Cell Biochem. 2002;240:119–130.
    1. Slezak J, Tribulova N, Pristacova J, Uhrik B, Thomas T, Khaper N, Kaul N, Singal PK. Hydrogen peroxide changes in ischemic and reperfused heart. Cytochemistry and biochemical and X‐ray microanalysis. Am J Pathol. 1995;147:772–781.
    1. Peretz A, Peck EC, Bammler TK, Beyer RP, Sullivan JH, Trenga CA, Srinouanprachnah S, Farin FM, Kaufman JD. Diesel exhaust inhalation and assessment of peripheral blood mononuclear cell gene transcription effects: an exploratory study of healthy human volunteers. Inhal Toxicol. 2007;19:1107–1119.
    1. Tsai DH, Amyai N, Marques‐Vidal P, Wang JL, Riediker M, Mooser V, Paccaud F, Waeber G, Vollenweider P, Bochud M. Effects of particulate matter on inflammatory markers in the general adult population. Part Fibre Toxicol. 2012;9:24.
    1. Wollert KC, Taga T, Saito M, Narazaki M, Kishimoto T, Glembotski CC, Vernallis AB, Heath JK, Pennica D, Wood WI, Chien KR. Cardiotrophin‐1 activates a distinct form of cardiac muscle cell hypertrophy. Assembly of sarcomeric units in series VIA gp130/leukemia inhibitory factor receptor‐dependent pathways. J Biol Chem. 1996;271:9535–9545.
    1. Terrell AM, Crisostomo PR, Wairiuko GM, Wang M, Morrell ED, Meldrum DR. Jak/STAT/SOCS signaling circuits and associated cytokine‐mediated inflammation and hypertrophy in the heart. Shock. 2006;26:226–234.
    1. Prabhu SD. Cytokine‐induced modulation of cardiac function. Circ Res. 2004;95:1140–1153.
    1. Yang YY, Liu H, Nam SW, Kunos G, Lee SS. Mechanisms of TNFalpha‐induced cardiac dysfunction in cholestatic bile duct‐ligated mice: interaction between TNFalpha and endocannabinoids. J Hepatol. 2010;53:298–306.
    1. Sun M, Chen M, Dawood F, Zurawska U, Li JY, Parker T, Kassiri Z, Kirshenbaum LA, Arnold M, Khokha R, Liu PP. Tumor necrosis factor‐alpha mediates cardiac remodeling and ventricular dysfunction after pressure overload state. Circulation. 2007;115:1398–1407.
    1. Moe KT, Khairunnisa K, Yin NO, Chin‐Dusting J, Wong P, Wong MC. Tumor necrosis factor‐alpha‐induced nuclear factor‐kappaB activation in human cardiomyocytes is mediated by NADPH oxidase. J Physiol Biochem. 2014;70:769–779.
    1. Liu H, Lee SS. Nuclear factor‐kappaB inhibition improves myocardial contractility in rats with cirrhotic cardiomyopathy. Liver Int. 2008;28:640–648.
    1. Gupta S, Young D, Sen S. Inhibition of NF‐kappaB induces regression of cardiac hypertrophy, independent of blood pressure control, in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol. 2005;289:H20–H29.
    1. Kawamura N, Kubota T, Kawano S, Monden Y, Feldman AM, Tsutsui H, Takeshita A, Sunagawa K. Blockade of NF‐kappaB improves cardiac function and survival without affecting inflammation in TNF‐alpha‐induced cardiomyopathy. Cardiovasc Res. 2005;66:520–529.
    1. Vogel CF, Sciullo E, Wong P, Kuzmicky P, Kado N, Matsumura F. Induction of proinflammatory cytokines and C‐reactive protein in human macrophage cell line U937 exposed to air pollution particulates. Environ Health Perspect. 2005;113:1536–1541.
    1. Niwa Y, Hiura Y, Sawamura H, Iwai N. Inhalation exposure to carbon black induces inflammatory response in rats. Circ J. 2008;72:144–149.
    1. Munkhaugen J, Otterstad JE, Dammen T, Gjertsen E, Moum T, Husebye E, Gullestad L. The prevalence and predictors of elevated C‐reactive protein after a coronary heart disease event. Eur J Prev Cardiol. 2018;25:923–931. DOI: 10.1177/2047487318768940.
    1. Pradhan AD, Rifai N, Ridker PM. Soluble intercellular adhesion molecule‐1, soluble vascular adhesion molecule‐1, and the development of symptomatic peripheral arterial disease in men. Circulation. 2002;106:820–825.
    1. Rana JS, Arsenault BJ, Despres JP, Cote M, Talmud PJ, Ninio E, Wouter Jukema J, Wareham NJ, Kastelein JJ, Khaw KT, Boekholdt SM. Inflammatory biomarkers, physical activity, waist circumference, and risk of future coronary heart disease in healthy men and women. Eur Heart J. 2011;32:336–344.
    1. Dawson J, Miltz W, Mir AK, Wiessner C. Targeting monocyte chemoattractant protein‐1 signalling in disease. Expert Opin Ther Targets. 2003;7:35–48.
    1. Sheikine Y, Hansson GK. Chemokines and atherosclerosis. Ann Med. 2004;36:98–118.
    1. Becker LC. Yin and yang of MCP‐1. Circ Res. 2005;96:812–814.
    1. Pope CA III, Bhatnagar A, McCracken JP, Abplanalp W, Conklin DJ, O'Toole T. Exposure to fine particulate air pollution is associated with endothelial injury and systemic inflammation. Circ Res. 2016;119:1204–1214.
    1. Kampfrath T, Maiseyeu A, Ying Z, Shah Z, Deiuliis JA, Xu X, Kherada N, Brook RD, Reddy KM, Padture NP, Parthasarathy S, Chen LC, Moffatt‐Bruce S, Sun Q, Morawietz H, Rajagopalan S. Chronic fine particulate matter exposure induces systemic vascular dysfunction via NADPH oxidase and TLR4 pathways. Circ Res. 2011;108:716–726.
    1. Gorr MW, Youtz DJ, Eichenseer CM, Smith KE, Nelin TD, Cormet‐Boyaka E, Wold LE. In vitro particulate matter exposure causes direct and lung‐mediated indirect effects on cardiomyocyte function. Am J Physiol Heart Circ Physiol. 2015;309:H53–H62.
    1. Mack M. Inflammation and fibrosis. Matrix Biol. 2018;68–69:106–121.
    1. Day J, Savani S, Krempley BD, Nguyen M, Kitlinska JB. Influence of paternal preconception exposures on their offspring: through epigenetics to phenotype. Am J Stem Cells. 2016;5:11–18.
    1. Baccarelli A, Rienstra M, Benjamin EJ. Cardiovascular epigenetics: basic concepts and results from animal and human studies. Circ Cardiovasc Genet. 2010;3:567–573.
    1. Turunen MP, Aavik E, Yla‐Herttuala S. Epigenetics and atherosclerosis. Biochim Biophys Acta. 2009;1790:886–891.
    1. Cantone L, Iodice S, Tarantini L, Albetti B, Restelli I, Vigna L, Bonzini M, Pesatori AC, Bollati V. Particulate matter exposure is associated with inflammatory gene methylation in obese subjects. Environ Res. 2017;152:478–484.
    1. Bosch‐Presegue L, Vaquero A. Sirtuin‐dependent epigenetic regulation in the maintenance of genome integrity. FEBS J. 2015;282:1745–1767.
    1. Peng L, Yuan Z, Ling H, Fukasawa K, Robertson K, Olashaw N, Koomen J, Chen J, Lane WS, Seto E. SIRT1 deacetylates the DNA methyltransferase 1 (DNMT1) protein and alters its activities. Mol Cell Biol. 2011;31:4720–4734.
    1. Heo J, Lim J, Lee S, Jeong J, Kang H, Kim Y, Kang JW, Yu HY, Jeong EM, Kim K, Kucia M, Waigel SJ, Zacharias W, Chen Y, Kim IG, Ratajczak MZ, Shin DM. Sirt1 regulates DNA methylation and differentiation potential of embryonic stem cells by antagonizing Dnmt3 l. Cell Rep. 2017;18:1930–1945.

Source: PubMed

3
Subscribe