• Sonuç bulunamadı

SONUÇ VE ÖNERİLER

Belgede ŞEKİLLER DİZİNİ (sayfa 66-79)

Sonuçlar;

Sıçanlarda bakteriyel enfeksiyonların prenatal-postnatal etkileşimini inceleyen mevcut tez çalışmasında aşağıdaki sonuçlara varılmıştır:

o Prenatal bakteriyel enfeksiyon, yavruların canlı ağırlıklarını düşürmüş, nötrofillerin % oranını yükseltmiş ve lenfositlerin % oranını düşürmüştür ve bu nedenle prenatal bir programlamanın oluşmuştuğu düşünülmüştür;

o Postnatal bakteriyel enfeksiyon, karaciğer ağırlığını, karaciğer indeksini ve TNF-alfa ile IL-1beta düzeylerini yükseltmiştir;

o Prenatal dönemde bakteriyel enfeksiyon geçiren sıçan yavrularının postnatal dönemde tekrar enfeksiyona maruz kalması kortikosteron düzeyini etkilemezken kalp dokusunda SOD düzeyini düşürmüştür.

o Bir diğer ifadeyle bakteriyel enfeksiyonlar prenatal dönemde oluşurlarsa postnatal 60. günde immünolojik savunma hücrelerini etkilerken, sitokinler ve HPG aksında değişikliğe yol açmamıştır. Postnatal bakteriyel enfeksiyonlar ise kısa vadede (4 saat içinde) karaciğer ağırlığı ile sitokinleri artırmıştır.

Öneriler;

Mevcut çalışma, prenatal bakteriyel enfeksiyonların uzun vadede postnatal hayati parametreleri etkileyebileceğini (canlı ağırlık, savunma hücreleri) ve prenatal dönemdeki bakteriyel enfeksiyonların postnatal dönemde oluşan bakteriyel enfeksiyonlarla yanıtı modifiye edebileceğini gösterdiğinden,

o Prenatal ve postnatal dönemlerde ve farklı günlerde yapılması, o Farklı dozları içeren yeni çalışmalar yapılması,

o Canlı ağırlık değişimlerinin detaylı (haftalık) incelenmesi, önerilebilir.

52 KAYNAKLAR

1- Aly HA, Lightfoot DA, El-Shemy HA. Bacterial lipopolysaccharide-induced oxidative stress in adult rat Sertoli cells in vitro. Toxicology in Vitro 2010, 24:

1266–72.

2- Mohamadina AM, Elberry AA, Mohamed A, Elkablawyc HS, Gawad A, Abbasi FAA. Montelukast, a leukotriene receptor antagonist abrogates lipopolysaccharide-induced toxicity and oxidative stress in rat liver.

Pathophysiology 2011, 18: 235–42.

3- Geyik S, Kumcu EK, Büyüknaca HS, Arıdogan A, Göçmen C, Önder S. Effects of vitamin E and sodium selenate on impaired contractile activity by bacterial lipopolysaccharide in the rat vas deferens. Naunyn-Schmied Arch Pharmacol Naunyn-Schmied Arch Pharmacol 2009, 380: 1–9.

4- Yang CC, Ma MC, Chien CT, Wu MS, Sun WK, Chen CF. Hypoxic preconditioning attenuates lipopolysaccharideinduced oxidative stress in rat kidneys. J Physiol 2007, 582: 407–19.

5- Dong HP, Chunag IJ, Wang DC, Huang LJ, Lee CI, Tsai JH, Yang RC.

Lipopolysaccharide-stimulated leukocytes contribute to platelet aggregative dysfunction, which is attenuated by catalase in rats. Kaohsiung J Med Sci 2010, 26(11): 584-92.

6- Remacle C, Bieswal F, Bol V, Reusens B. Developmental programming of adult obesity and cardiovascular disease in rodents by maternal nutrition. Am J Clin Nutr 2011, 4: 1-7.

7- Reusens B, Theys N, Dumortier O, Goosse K, Remacle C. Maternal malnutrition programs the endocrine pancreas in progeny. Am J Clin Nutr 2011, 94: 1824–9.

8- Mohn CE, Solari JF, Laurentiis AD, Bornstein SR, Bornstein ME, Rettori V.

Adrenal gland responses to lipopolysaccharide after stress and ethanol administration in male rats. Stress 2011, 14(2): 216-26.

9- Raetz CRH, Whitfield C. Lipopolysaccharide Endotoxins. Annu Rev Biochem 2002, 71: 635–700.

10- Caroff M, Karibian D. Structure of bacterial lipopolysaccharides. Carbohydr Res 2003, 338(23): 2431–47.

11- Topçu WT, Söyletir G, Doğanay M. İnfeksiyon Hastalıklar ve Mikrobiyolojisi, 1.

Baskı. İstanbul, Nobel tıp kitapevi 2002: 110.

53 12- İskit AB. Sepsiste Deneysel Modeller, Hacettepe Universitesi Tıp Fakultesi,

Farmakoloji Anabilim Dalı, Yoğun Bakım Dergisi 2005, 5(2): 133-6.

13- Brandtzaeg P. Significance and pathogenesis of septic shock. Curr Top Microbiol Immunol 1996, 216: 15-37.

14- Endotoxin lipopolysaccharide schematic.

(http://hermes.mbl.edu/marine_org/images/animals/Limulus/blood/lps.html) 8 Temmuz 2015.

15- Wang H, Yang LL, Hu YF, Wang BW, Huang YY. Maternal LPS Exposure during Pregnancy Impairs Testicular Development, Steroidogenesis and Spermatogenesis in Male Offspring. Plos One 2014, 9(9): e106786.

16- Hurley JC. Concordance of endotoxemia with gram-negative bacteremia in patients with gram-negative sepsis: a meta-analysis. J Clin Microbiol 1994, 32:

2120–7.

17- Aisemberg J, Vercelli C, Billi S, Ribeiro ML, Ogando D. Nitric oxide mediates prostaglandins’ deleterious effect on lipopolysaccharide-triggered murine fetal resorption. Proc Natl Acad Sci U S A 2007, 104: 7534–39.

18- Ogando DG, Paz D, Cella M, Franchi AM. The fundamental role of increased production of nitric oxide in lipopolysaccharide-induced embryonic resorption in mice. Reproduction 2003, 125: 95–110.

19- Chen YH, Zhao M, Chen X, Zhang Y, Wang H. Zinc supplementation during pregnancy protects against lipopolysaccharide-induced fetal growth restriction and demise through its anti-inflammatory effect. J Immunol 2012, 189: 454–63.

20- Bernardi MM, Kirsten TB, Matsuoka SM, Teodorov E, Habr SF, Penteado SH, Palermo-Neto J. Prenatal Lipopolysaccharide Exposure Affects Maternal Behavior and Male Offspring Sexual Behavior in Adulthood.

Neuroimmunomodulation 2010, 17(1): 47-55.

21- French SS, Chester EM, Demas GE. Maternal immune activation affects litter success, size and neuroendocrine responses related to behavior in adult offspring.

Physiol Behav 2013, 2(119): 175-84.

22- Oskvig DB, Elkahloun AG, Johnson KR, Phillips TM, Herkenham M. Maternal immune activation by LPS selectively alters specific gene expression profiles of interneuron migration and oxidative stress in the fetus without triggering a fetal immune response. Brain, Behavior and Immunity 2012, 26: 623–34.

54 23- Holekamp KE, Dloniak SM. Maternal effects in mammals. Chicago: University

of Chicago Press; 2009: 227-5.

24- Meyer U, Nyffeler M, Engler A, Urwyler A, Schedlowski M, Knuesel I. The time of prenatal immune challenge determines the specificity of inflammation-mediated brain and behavioral pathology. J Neurosci 2006, 26: 4752–62.

25- Brown AS, Begg MD, Gravenstein S, Schaefer CA, Wyatt RJ, Bresnahan M.

Serologic evidence of prenatal influenza in the etiology of schizophrenia. Arch GenPsychiatry 2004, 61(8): 774-80.

26- Solati J, Hajikhani R, Rashidieh B, Fatipour Jalilian M. Effects of prenatal lipopolysaccharide exposure on reproductive activities and serum concentrations of pituitary-gonadal hormones in mice offspring. Int J Fertil Steril 2012, 6(1): 51-8.

27- Hodyl NA. Innate immune dysfunction in the neonatal rat following prenatal endotoxin exposure. J Neuroimmunol 2008, 204: 126–30.

28- Williams CL, Teeling JL, Perry VH and Fleming TP. Williams et al. Mouse maternal systemic inflammation at the zygote stage causes blunted cytokine responsiveness in lipopolysaccharide-challenged adult offspring. BMC Biology 2011, 9: 49-51.

29- Dammann O, Kuban KCK, Leviton A. Perinatal infection, fetal inflammatory response, white matter damage and cognitive limitation in children born preterm.

Ment Retard Dev Disabil 2002, 8: 46-50.

30- Saliba E, Henrot A. Inflammatory mediators and neonatal brain damage. Biol.

Neonate 2001, 79: 224-7.

31- Golan HM, Lev V, Hallak M, Sorokin Y, Huleihel M. Specific neurodevelopmental damage in mice offspring following maternal inflammation during pregnancy. Neuropharmacology 2005, 48: 903-17.

32- Dinarello CA. Proinflammatory cytokines. Chest 2000, 118: 503-8.

33- Bilbo SD, Schwarz JM. Early-life programming of later-life brain and behavior:

a critical role for the immune system. Front Behav Neurosci 2009, 3(14): 1-14.

34- Atladottir HO, Thorsen P, Ostergaard L, Schendel DE, Lemcke S, Abdallah, M, Parner ET. Maternal infection requiring hospitalization during pregnancy and autism spectrum disorders. J Autism Dev Disord 2010, 40: 1423– 30.

55 35- Elovitz MA, Brown AG, Breen K, Anton L, Maubert M, Burd I. Intrauterine inflammation, insufficient to induce parturition, still evokes fetal and neonatal brain injury. Int. J. Devl Neuroscience 2011, 29: 663–71.

36- Enayatia M, Solati J, Hosseinic MH, Shahia HR, Sakid G, Salari AA. Maternal infection during late pregnancy increases anxiety- and depression-like behaviors with increasing age in male offspring. Brain Research Bulletin 2012, 87: 295–

302.

37- Lowe GC, Luheshi GN, Williams S. Maternal infection and fever during late gestation are associated with altered synaptic transmission in the hippocampus of juvenile offspring rats. Am J Physiol Regul Integr Comp Physiol 2008, 295: 1563–

71.

38- Getahun D, Ananth CV, Oyelese Y, Peltier MR, Smulian JC, Vintzileos AM.

Acute and chronic respiratory diseases in pregnancy: associations with spontaneous premature rupture of membranes. J Maternal Fetal Neonatal Medicine 2007, 20: 669–75.

39- Velten M, Britt JRD, Heyob KM, Welty SE, Eiberger B, Tipple TE, Rogers LK.

Prenatal inflammation exacerbates hyperoxia induced functional and structural changes in adult mice. Am J Physiol Regul Integr Comp Physiol 2012, 303: 279–

90.

40- Kirsten TB, Oliveira BPS, Oliveira APL, Kieling K, Lima WT, Neto JP, Bernardi MM. Single early prenatal lipopolysaccharide exposure prevents subsequent airway inflammation response in an experimental model of asthma. Life Sciences 2011, 89: 15–9.

41- Wei YL, Li XH, Zhou JZ. Prenatal exposure to lipopolysaccharide results in increases in blood pressure and body weight in rats. Acta Pharmacol Sin 2007, 28: 651–6.

42- Zhao S, Zhang H, Cao D, Liu Y, Li X. Lipopolysaccharide Exposure during Pregnancy Leads to Aortic Dysfunction in Offspring Rats. PLoS ONE 2014, 9(7):

e102273.

43- Hao1 XQ, Zhang HG, Yuan ZB, Yang DL, Hao LY, Li XH. Prenatal exposure to lipopolysaccharide alters the intrarenal renin–angiotensin system and renal damage in offspring rats. Hypertension Research 2010, 33: 76–82.

44- Haoa XQ, Konga T, Zhangc SY, Zhaoa ZS. Alteration of embryonic AT2-R and inflammatory cytokines gene expression induced by prenatal exposure to

56 lipopolysaccharide affects renal development. Experimental and Toxicologic Pathology 2013, 65: 433– 9.

45- Gao M, Zhang X, Chen X, Mi C, Tang Y. Prenatal Exposure to Lipopolysaccharide Results in Local RAS Activation in the Adipose Tissue of Rat Offspring. PLoS ONE 2014, 9(10): 1371-6.

46- Pararas MV, Skevaki CL, Kafetzis DA. Preterm birth due to maternal infection:

causative pathogens and modes of prevention. Eur J Clin Microbiol Infect Dis 2006, 25: 562–9.

47- Kramer BW, Ikegami M, Moss TJ, Nitsos I, Newnham JP, Jobe AH. Endotoxin induced chorioamnionitis modulates innate immunity of monocytes in preterm sheep. Am J Respir Crit Care Med 2005, 171: 73-7.

48- Surriga O, Ortega A, Jadeja V, Bellafronte A, Lasala N, Zhou H. Altered hepatic inflammatory response in the offspring following prenatal LPS exposure.

Immunology Letters 2009, 123: 88–95.

49- Gerhold K, Avagyan A, Seib C, Frei R, Steinle J, Ahrens B, Dittrich AM, Blumchen K, Lauener R, Hamelmann E. Prenatal initiation of endotoxin airway exposure prevents subsequent allergen-induced sensitization and airway inflammation in mice. J Allergy Clin Immunol 2006, 118: 666-73.

50- Lasala N, Zhou H. Effects of maternal exposure to LPS on the inflammatory response in the offspring. Journal of Neuroimmunology 2007, 189: 95–101.

51- Hodyl NA, Walker FR, Krivanek KM, Clifton VL, Hodgson DM. Prenatal endotoxin exposure alters behavioural pain responses to lipopolysaccharide in adult offspring. Physiology & Behavior 2010, 100: 143–7.

52- Isolauri E, Huurre A, Salminen S, Impivaara O. The allergy epidemic extends beyond the past few decades. Clin Exp Allergy 2004, 34: 1007-10.

53- Dujmovic I, Mangano K, Pekmezovic T, Quattrocchi C, Mesaros S, Stojsavljevic N. The analysis of IL-1 beta and its naturally occurring inhibitors in multiple sclerosis: the elevation of IL-1 receptor antagonist and IL-1 receptor type II after steroid therapy. Journal Neuroimmunol 2009, 207: 101–06.

54- Contassot E, Beer HD, French LE. Interleukin-1, inflammasomes, autoinflammation and the skin. Swiss Med Wkly 2012, 142: 13590.

55- Camargo JF, Correa PA, Castiblanco J, Anaya JM. Interleukin-1beta polymorphisms in Colombian patients with autoimmune rheumatic diseases.

Genes Immun 2004, 5(8): 609–14.

57 56- Aggelakis K, Zacharaki F, Dardiotis E, Xiromerisiou G, Tsimourtou V, Ralli S, et al. Interleukin-1B and interleukin-1 receptor antagonist gene polymorphisms in Greek multiple sclerosis (MS) patients with bout-onset MS. Neurol Sci 2010, death and inflammation to therapeutic giants – past, present and future. Cytokine

& Growth Factor Reviews 2014, 25: 453–72.

59- Michalska AG, Tadeusz J, Rachwalska P, Bugajski J. Cytokines, prostaglandins and nitric oxide in the regulation of stress-response systems. Pharmacological Reports 2013, 65: 1655-62.

60- Deng Y, Ren X, Yang L, Lin Y, Wu X. A JNK-dependent pathway is required for TNFa-induced apoptosis. Cell 2003, 115: 61–70.

61- Stamou P, Kontoyiannis DL. Posttranscriptional regulation of TNF mRNA: a paradigm of signal-dependent mRNA utilization and its relevance to pathology.

Curr Dir Autoimmun 2010, 11: 61–79.

62- Catalani A, Alemà GS, Cinque C, Zuena AR, Casolini P. Maternal corticosterone effects on hypothalamus–pituitary–adrenal axis regulation and behavior of the offspring in rodents, Neuroscience and Biobehavioral Reviews 2013, 35: 1502–

17.

63- Harbuz M. Neuroendocrine function and chronic inflammatory stress.

Experimental Physiology 2002, 87: 519–25.

64- Vreugdenhil E, Kloet ER, Schaaf1 M, Datson NA. Genetic dissection of corticosterone receptor function in the rat hippocampus. European Neuropsychopharmacology 2001, 11: 423–30.

65- Maccari S, Morley FS.. Effects of prenatal restraint stress on the hypothalamus–

pituitary–adrenal axis and related behavioural and neurobiological alterations.

Psychoneuroendocrinology 2007, 32: 10–15.

66- Schoenfeld TJ, Gould E. Stress, stress hormones, and adult neurogenesis.

Experimental Neurology 2012, 233: 12–21.

58 67- Buonocore G, Perrone S, Tataranno ML. Oxygen toxicity: Chemistry and biology of reactive oxy- gen species. Seminars in Fetal and Neonatal Medicine 2010, 15:

186-90.

68- Auten RL, Davis JM. Oxygen toxicity and reactive oxygen species: The devil is in the details. Pedi- atric Research 2009, 66: 121-27.

69- Young IS, Woodside JV. Antioxidants in health and disease. J Clin Pathol 2001, 54: 176-86.

70- Perrona S, Tatarano MS, Stazzoni G, Buonocore G. Oxidative stress anad free radicals related diseases of the newborn. Advances in Bioscience and Biotechnology 2012, 3: 1043- 50.

71- Toro J, Rodrigo R. Oxidative stress: Basic owerview. Nova Science Puplishers, Inc 2009 Ed Rodrigo R. Chapter 1 ISBN:978-1-60741-554-1

72- Sabuncuoğlu S, Özgüneş H. Kemoterapi, serbest radikaller ve oksidatif stres.

Hacettepe Üniversitesi Eczacılık Fakültesi Dergisi 2011, 31: 137-50.

73- Lushchak VI Free radicals, reactive oxygen species, oxidative stress and its classification. Chem Biol Interact 2014, 28: 164-75.

74- Lushchak VI, Bagnyukova TV, Husak VV, Luzhna LL, Lushchak OV, Storey KB. Hyperoxia results in transient oxidative stress and an adaptive response by antioxidant enzymes in goldfish tissues, Int. J. Biochem. Cell Biol 2005, 37:

1670–80.

75- Birben E, Sahiner UM, Sackesen C, Erzurum S, Katayci O. Oxidative stress and antioxidant defense. World Allergy Organ J 2012, 5(1): 9–19.

76- Demir T, Aydemir A, Güler S, Serdaroğlu E, Kurutepe M, Donma O, Yıldırım N.

Akut ve stabil KOAH olgularında oksidatif stres. Solunum 1999, 1: 43-47.

77- Sun Y, Oberley LW, Li YA. A simple method for clinical assay of superoxide dismutase. Cli Chem 1988, 34: 497-500.

78- Durak I, Yurtaslani Z, Canbolat O, Akyol O. A methodological approach to superoxide dismutase activity assay based on inhibition of nitroblue tetrazolium (NBT) reduction. Clin Chem Acta 1993, 214: 103-4.

79- Esterbauer H, Cheeseman KH. Determination of aldehydic lipid peroxidation product: malonaldehyde and 4-hydroxynonenal In Packer L. Methods Enzymol 1990, 16: 407-21.

80- Aebi H. Catalase. In: Bergmeyer HU (ed). Methods of enzymatic analysis.

Academic pres: New York and London 1974, 673-7.

59 81- Lowry OH, Rosenbrough NJ, Farr AL, Randall RJ. Protein measurment with the

Folin phenol reagent. J Biol Chem 1951, 193: 265-75.

82- Hodyl NA, Krivanek KM, Lawrence E, Clifton VL, Hodgson DM. Prenatal exposure to a pro-inflammatory stimulus causes delays in the development of the innate immune response to LPS in the offspring. Journal of Neuroimmunology 2007, 190: 61–71.

83- Chen K, Geng S, Yuan R, Diao N, Upchurch Z, Li L. Super-low dose endotoxin pre-conditioning exacerbates sepsis mortality. Ebio Med 2015, 2(4): 324-33.

84- Wei YL, Li XH, Zhou JZ. Prenatal exposure to lipopolysaccharide results in increases in blood pressure and body weight in rats. Acta Pharmacol Sin 2007, 28 (5): 651–6.

85- Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, Fava F, Tuohy KM, Chabo C. Metabolic Endotoxemia Initiates Obesity and Insulin Resistance. Diabetes 2007, 56: 1761–72.

86- Zhao R, Zhau H, Su BS. A critical role for interleukin-1B in the progression of autoimmune diseases. International immunopharmacology 2013, 17: 658-69.

87- Jepson MM, Pell JM, Bates PC, Mıllward DJ. The effects of endotoxaemia on protein metabolism in skeletal muscle and liver of fed and fasted rats. Biochem.

J. 1986, 235: 329-36.

88- Chen Y, Du Y, Li Y, Wang X, Gao P, Yang G.Panaxadiol Saponin and Dexamethasone Improve Renal Function in Lipopolysaccharide-Induced Mouse Model of Acute Kidney Injury. PLoS ONE 2015, 10(7): e0134653.

89- Khodir AE, Ghoneim HA, Rahim MA, Suddek GM. Montelukast attenuates lipopolysaccharide-induced cardiac injury in rats. Hum Exp Toxicol 2015 doi 10.1177.

90- Kao SJ, Wang D, Lin HI, Chen HI. N-Acetylcysteıne Abrogates Acute Lung Injury Induced By Endotoxın. Clinical And Experimental Pharmacology And Physiology 2006, 33: 33–40.

91- Doursout MF, Schurdell MS, Young LM, Osuagwu U, Hook DM, Poindexter BJ, Schiess MC, Bick DLM, Bick RJ. Inflammatory Cells and Cytokines in the Olfactory Bulb of a Rat Model of Neuroinflammation; Insights into Neurodegeneration. Journal Of Interferon & Cytokıne Research 2013, 33: 376-83.

60 92- Zager A, Pinheiro ML, Paula VF, Ribeiro A, Neto JP. Increased cell-mediated immunity in male mice offspring exposed to maternal immune activation during late gestation. International Immunopharmacology 2013, 17: 633–7.

93- Von Dadelszen P, Watson RWG, Noorwali F. Maternal neutrophil apoptosis in normal pregnancy, preeclampsia and normotensive intrauterine growth restriction. Am J Obstet Gynecol 1999, 181: 408–14.

94- Luo D, Schowengerdt KO, Stegner JJ. Decreased functional caspase-3 expression in umbilical cord blood neutrophils is linked to delayed apoptosis. Pediatr Res 2003, 53: 859–64.

95- Triantafilou M, Triantafilou K. The dynamics of LPS recognition: complex orchestration of multiple receptors. J Endotoxin Res 2005, 11: 5–11.

96- Kramer BW, Kallapur SG, Moss TJM, Nitsos I, Polglase GP, Newnham JP, Jobe AH. Modulation of fetal inflammatory response upon exposure to LPS by chorioamnion, lung or gut in sheep. Am J Obstet Gynecol 2010, 202: 771–9.

97- Bernardi MM, Teixeira LP, Oliveira APL, Lima WT, Neto JP, Kirsten TB.

Neonatal lipopolysaccharide exposure induces sexually dimorphic sickness behavior in adult rats. Psychology & Neuroscience 2014, 7: 113 – 23.

98- Solati J, Kleehaupt E, Kratz O, Moll GH, Goluba Y. Inverse effects of lipopolysaccharides on anxiety in pregnant mice and their offspring. Physiology

& Behavior 2015, 139: 369–74.

99- Tamion F, Bauer F, Richard V, Laude K, Renet S, Slama M, Thuillez C.

Myocardial Dysfunction in Early State of Endotoxemia Role of Heme-Oxygenase-1. Journal of Surgical Research 2010, 158: 94–103.

100-Sebai H, Sani M, Aouani E, Boughanmi NG. Cardioprotective effect of .resveratrol on lipopolysaccharideinduced oxidative stress in rat. Drug and .Chemical Toxicology 2011, 34: 146-50.

101-Shaul VB, Lomnitski L, Nyska A, Zurovsky Y, Bergman M, Grossman S. The .effect of natural antioxidants, NAO and apocynin, on oxidative stress in the rat .heart following LPS challenge. Toxicology Letters 2001, 123: 1–10.

102-Recknagel P, Gonnert FA, Halilbasic E, Gajda M, Jbeily N, Lupp A, Rubio I, .Claus RA, Kortgen A, Trauner M, Singer M, Bauer M. Mechanisms and

61 .functional consequences of liver failure substantially differ between endotoxaemia and faecal peritonitis in rats. Liver Int 2013, 33: 283–93.

103-Sebai H, Sani M, Boughanmi NG, Aouani E. Prevention of lipopolysaccharide-.induced mouse lethality by resveratrol. Food and Chemical Toxicology 2010, 48:

.1543–9.

104-Resveratrol, a red wine polyphenol, attenuates lipopolysaccharide-induced .oxidative stress in rat liver. Sebai H, Sani M, Yacoubi MT, Aouani E, Boughanmi NG, Attia MB. Ecotoxicology and Environmental Safety 2010, 73: 1078–83.

105-Ajuwon OR, Oguntibeju OO, Marnewick JL. Amelioration of lipopolysaccharide-induced liver injury by aqueous rooibos (Aspalathus linearis) extract via inhibition of pro-inflammatory cytokines and oxidative stress. BMC Complementary and Alternative Medicine 2014, 14: 392–404.

62

EKLER

EK.1 ÖZGEÇMİŞ

1986 yılında Malatya’da doğdu. Ortaöğretim ve lise eğitimini Malatya’da tamamladı. 2009 yılında Fırat Üniversitesi Sağlık Yüksekokulu Sağlık Memurluğu bölümünden mezun oldu. 2009-2011 yıllarında Hemşirelik yaptıktan sonra 2012 yılında Atatürk Üniversitesinde Araştırma Görevlisi olarak çalıştı, 2013 yılında İnönü Üniversitesi Sağlık Bilimleri Fakültesinde Araştırma Görevlisi olarak atandı, Halk Sağlığı Hemşireliği Anabilim Dalında Yüksek Lisans eğitimini bitirip, 2015 yılında Doktoro eğitimine başladı. 2010 yılında başlamış olduğu Fizyoloji Anabilim Dalında Yüksek Lisansı devam etmektedir. Evli, bir çocuk sahibi ve orta derecede İngilizce bilmektedir.

63

EK.2. ETİK ONAY FORMU

64

Belgede ŞEKİLLER DİZİNİ (sayfa 66-79)

Benzer Belgeler