9 research outputs found

    Вплив антигіпертензивної терапії на рівень сечової кислоти у хворих на артеріальну гіпертензію з гіперурикемією

    Get PDF
    Артеріальна гіпертензія (АГ) є найбільш поширеним хронічним захворюванням у світі і значною мірою визначає високу смертність та інвалідність від серцево-судинних і церебро-васкулярних захворювань. При цитуванні документа, використовуйте посилання http://essuir.sumdu.edu.ua/handle/123456789/2723

    Untargeted Metabolomic Analysis of Sjögren–Larsson Syndrome Reveals a Distinctive Pattern of Multiple Disrupted Biochemical Pathways

    Get PDF
    Sjögren–Larsson syndrome (SLS) is a rare inherited neurocutaneous disease characterized by ichthyosis, spastic diplegia or tetraplegia, intellectual disability and a distinctive retinopathy. SLS is caused by bi-allelic mutations in ALDH3A2, which codes for fatty aldehyde dehydrogenase (FALDH) and results in abnormal lipid metabolism. The biochemical abnormalities in SLS are not completely known, and the pathogenic mechanisms leading to symptoms are still unclear. To search for pathways that are perturbed in SLS, we performed untargeted metabolomic screening in 20 SLS subjects along with age- and sex-matched controls. Of 823 identified metabolites in plasma, 121 (14.7%) quantitatively differed in the overall SLS cohort from controls; 77 metabolites were decreased and 44 increased. Pathway analysis pointed to disrupted metabolism of sphingolipids, sterols, bile acids, glycogen, purines and certain amino acids such as tryptophan, aspartate and phenylalanine. Random forest analysis identified a unique metabolomic profile that had a predictive accuracy of 100% for discriminating SLS from controls. These results provide new insight into the abnormal biochemical pathways that likely contribute to disease in SLS and may constitute a biomarker panel for diagnosis and future therapeutic studies

    Metabolic and endocrine aspects of coronary disease

    Get PDF
    The early history, the nature and the incidence of coronary atherosclerosis and ischaemic heart disease have been reviewed. Evidence has been presented to suggest that there are aetiological factors which favour the development of ischaemic heart disease without influencing the incidence of coronary atherosclerosis. The close relationship of cholesterol metabolism to coronary atherosclerosis and to ischaemic heart disease has been emphasised. The circulating lipids and lipoproteins have been studied in health in relation to age and sex. They were abnormal in the majority of patients with ischaemic heart disease, particularly in those under the age of 50. Analysis of the circulating lipids and lipoprotein could aid the solution of an equivocal diagnosis in young subjects suspected of having ischaemic heart disease. The effects of endogenous hormones and the action of administered hormones on the circulating lipids and lipoproteins have been described in detail. Hormones can also influence the fluid state of the blood and the tonicity and metabolism of arteries. It has been postulated that the homeostasis of cholesterol metabolism, of the fluid state of the blood and of arterial metabolism could be disturbed by alteration of the physiological endocrine balance. The thesis has been proposed that an endocrine imbalance could contribute to the development of coronary atherosclerosis and of ischaemic heart disease. The relationship of this thesis to existing theories of the aetiology of ischaemic heart disease has been considered in detail. Finally, some therapeutic implications of these metabolic and endocrine aspects of coronary disease have been considered

    Coronary thrombosis: a study of 128 cases with special reference to its relationship to angina pectoris

    Get PDF
    It is observed from this study that angina pectoris and coronary thrombosis are the different phases of the same process being caused by anoxaemia of heart muscle. The underlying factor in both the conditions 154. excluding the precipitating factors is the same i.e. atherosclerosis of coronary arteries Atheromatous changes in angina pectoris however are more or less of a uniform nature, resulting usually in narrowing of the lumen of these vessels, whereas in coronary thrombosis the changes are generally of a patchy type with or without much narrowing of the caliber of coronary arteries. The precipitating factors no doubt are different, but while coronary thrombosis is caused in a person with Sclerotic coronary arteries at rest, Angina pectoris results in the same individual in excitement and with exertion

    Uric acid, metabolism, neuro-endocrine-immune complex, 258 s.

    Get PDF
    LITERATURE 1. Avtandylov H.G. Medical morphometry. Moscow. Medicine; 1990: 384 p. 2. Baevsky R.M., Kirillov O.I., Kletskin S.Z. Mathematical analysis of heart rate changes during stress. Moscow. Science; 1984: 221 p. 3. Baevsky R.M., Ivanov H.G. Variability of the heart rhythm: theoretical aspects and opportunities for clinical application. Ultrasound and functional diagnostics. 2001; 3: 106-127. 4. Bazarnova M.A. Cytological examination of punctate spleen. Guide to practical classes in clinical laboratory diagnostics. K.: Higher school; 1988: 263-264. 5. Balanovsky V.P., Popovych I.L., Karpynets SV. About the ambivalent-equilibrator nature of the influence of Naftusya medicinal water on the human body. ANU reports. Mat., prop., tech. science 1993; 3: 154-158. 6. Belousova OI, Fedotova MY. Comparative data on changes in the number of lymphocytes of the spleen, goiter and bone marrow in the early stages after irradiation in a wide range of doses. Radiobiology-radiotherapy. 1968; 9(3): 309-313. 7. Bilas V.R., Popovych I.L. The role of microflora and organic substances of Naftusya water in its modulating effect on the neuroendocrine-immune complex and metabolism. Medical hydrology and rehabilitation. 2009; 7(1): 68-102. 8. Biloshitsky P.V. Temperature, information, water, anabiosis, immortality. Health and longevity. Kyiv; 2007: 46-47. 9. Harkavy L.H., Kvakina E.B., Kuzmenko T.S. Antistress reactions and activating therapy. Moscow. Imedis; 1998: 654 p. 10. Harkavy L.H., Kvakina E.B., Ukolova M.A. Adaptation reactions and resistance of the body. Rostov-on-Don. Rostov University Publishing House, 3rd ed.; 1990: 224 p. 11. Harkavy L.H., Romasyuk S.I., Barantsev F.G., Kuzmenko T.S., Otkydach S.A., Tattsov O.V., Barantseva L.P. Activation therapy in the sanatorium-resort stage of rehabilitation of patients with internal organ diseases. Sochi; 2000: 94 p. 12. Gozhenko A.I. Theory of disease. Odesa. Phoenix; 2018: 236 p. 13. Gozhenko A.I. Dysregulation as the basis of the pathophysiology of homeostasis. Clinical and experimental pathology. 2004; 3 (2): 191-193. 14. Gozhenko A.I. Functional-metabolic continuum. The National Academy of Sciences of Ukraine. 2016; 22(1): 3-8. 15. Gozhenko A.I., Korda M.M., Popadynets O.O., Popovych I.L. Entropy, harmony, synchronization and their neuro-endocrine-immune correlates. Odesa. Phoenix; 2021: 232 p. 16. Horyachkovsky A.M. Clinical biochemistry. Odesa: Astroprint; 1998: 608 p. 17. Datsko O.R., Bubnyak A.B., Ivasivka S.V. The organic component of Naftusya mineral water. Development of an idea about its composition and origin. Medical hydrology and rehabilitation. 2008; 6(1): 168-174. 18. Ivasivka S.V., Popovych I.L., Aksentiychuk B.I., Flunt I.S. Physiological activity of uric acid and its role in the mechanism of action of Naftusya water. K. Computer Press; 2004: 163 p. 19. Lapovets L.Y., Lutsik B.D. Laboratory immunology. Kyiv. 2004. 173 p. 20. Perederiy V.G., Zemskov A.M., Bychkova N.G., Zemskov V.M. Immune status, principles of its evaluation and correction of immune disorders. K. Health; 1995: 211 p. 21. Popovych I.L. Informational effects of Naftusya bioactive water in rats: modulation of entropy, reversal of desynchronizing and limitation of disharmonizing effect of water-immersion stress on informational components of the neuro-endocrine-immune system and metabolism, which correlates with a gastroprotective effect. Medical hydrology and rehabilitation. 2007; 5(3): 50-70. 22. I.L. Popovych Concept of neuro-endocrine-immune complex. Medical hydrology and rehabilitation. 2009; 7(2): 9-18. 23. I.L. Popovych Stress-limiting adaptogenic mechanism of biological and therapeutic activity of Naftusya water. Kyiv. Computer press; 2011: 300 c. 24. Popovich I.L., Flunt I.S., Alekseev O.I. etc. Sanogenetic principles of rehabilitation at the Truskavets resort of urological patients of the Chernobyl contingent. Kyiv. Computer press; 2003: 192 p. 25. Popovich I.L., Flunt I.S., Nischeta I.V., Loboda M.V., Aksentiychuk B.I., Pryima B.G., Tserkovnyuk R.G. General adaptation reactions and resistance of the organism of liquidators of the Chernobyl accident. Kyiv. Computer press; 2000: 117 p. 26. Portnychenko A.G. Ukrainian balneology: scientific trends of the last decade (scientometric analysis). Medical hydrology and rehabilitation. 2015; 13(4): 41-52. 27. Smagliy V.S., Gozhenko A.I., Badyuk N.S., Popovych I.L. Variants of uric acid metabolism and their immune and microbial accompaniments in patients with complex neuro-endocrine-immune dysfunction. In: VIII National Congress of Pathophysiologists of Ukraine "Pathological Physiology - Health Care of Ukraine" (Odesa, May 13-15, 2020). Odesa; 2020: 314-315. 28. Khaitov R.M., Pinegin B.V., Istamov K.I. Ecological immunology. Moscow. INWARD; 1995: 219 p. 29. Khmelevsky Yu.V., Usatenko O.K. Basic biochemical constants of a person in normal and pathological conditions. Kyiv. Health; 1987: 160 p. 30. Efroimson V.P. Some biological factors of mental activity. VIET. 1987; 4: 74-84. 31. Yushkovska O.H. The use of information theory to study the adaptive reactions of athletes' bodies. Medical rehabilitation Spa therapy Physiotherapy. 2001; 1 (25): 40-43. 32. Abdel Aziz N., Tallima H., Hafez E.A., El Ridi R. Papain-based vaccination modulates Schistosoma mansoni infection-induced cytokine signals. Scand J Immunol. 2016;83(2):128–138. 33. Ahbap E., Sakaci T., Kara E., Sahutoglu T., Koc Y., Basturk T. Serum uric acid levels and inflammatory markers with respect to dipping status: a retrospective analysis of hypertensive patients with or without chronic kidney disease . Clin Exp Hypertens. 2016;38(6):555–563. 34. Akbar S.R., Long D.M., Hussain K., Alhajhusain A., Ahmed U.S., Iqbal H.I. Hyperuricemia: an early marker for severity of illness in sepsis. Int J Nephrol. 2015; 301021. 35. Alberti K.G., Eckel R.H., Grundy S.M., Zimmet P.Z., Cleeman J.I., Donato K.A. Harmonizing the metabolic syndrome: a joint interim statement of the international diabetes federation task force on epidemiology and prevention; national heart, lung, and blood institute; american heart association; world heart federation; international atherosclerosis society; and international association for the study of obesity. Circulation. 2009;120(16):1640–1645. 36. Aldenderfer M.S., Blashfield R.K. Cluster analysis (Second printing, 1985) [transl. from English in Russian]. In: Factor, Discriminant and Cluster Analysis. Moscow. Finance and Statistics; 1989: 139-214. 37. Alvarez-Lario B., Macarrón-Vicente J. Is there anything good in uric acid? QJM. 2011;104(12):1015–1024.] 38. Alvarez-Lario B., Macarron-Vicente J. Uric acid and evolution. Rheumatology. 2010;49(11):2010–2015. 39. Amaral F.A., Costa V.V., Tavares L.D., Sachs D., Coelho F.M., Fagundes C.T. NLRP3 inflammasome-mediated neutrophil recruitment and hypernociception depend on leukotriene B(4) in a murine model of gout. Arthritis Rheum. 2012;64(2):474–484. 40. Amaral K.B., Silva T.P., Malta K.K., Carmo L.A.S., Dias F.F., Almeida M.R. Natural Schistosoma mansoni infection in the wild reservoir Nectomys squamipes leads to excessive lipid droplet accumulation in hepatocytes in the absence of liver functional impairment. Plos One. 2016;11(11):e0166979. 41. Amaral L.M., Cunningham M.W., Jr, Cornelius D.C., LaMarca B. Preeclampsia: long-term consequences for vascular health. Vasc Health Risk Manage. 2015; 11: 403–415. 42. Ames B.N., Cathcart R., Schwiers E., Hochstein P. Uric acid provides an antioxidant defense in humans against oxidant- and radical-caused aging and cancer: a hypothesis. Proc Natl Acad Sci U S A. 1981;78(11):6858–6862. 43. Amsellem V., Abid S., Poupel L., Parpaleix A., Rodero M., Gary-Bobo G., Latiri M., Dubois-Rande J.L., Lipskaia L., Combadiere C., et al. Roles for the CX3CL1/CX3CR1 and CCL2/CCR2 Chemokine Systems in Hypoxic Pulmonary Hypertension. Am. J. Respir. Cell. Mol. Biol. 2017; 56:597–608. 44. Ando K., Takahashi H., Watanabe T., Daidoji H., Otaki Y., Nishiyama S. Impact of serum uric acid levels on coronary plaque stability evaluated using integrated backscatter intravascular ultrasound in patients with coronary artery disease. J Atheroscler Thromb. 2016;23(8):932–939. 45. Andreadou E., Nikolaou C., Gournaras F., Rentzos M., Boufidou F., Tsoutsou A. Serum uric acid levels in patients with Parkinson's disease: their relationship to treatment and disease duration. Clin Neurol Neurosurg. 2009;111(9):724–728. 46. Annanmaki T., Muuronen A., Murros K. Low plasma uric acid level in Parkinson's disease. Mov Disord. 2007;22(8):1133–1137. 47. Anthony R.M., Rutitzky L.I., Urban J.F., Jr., Stadecker M.J., Gause W.C. Protective immune mechanisms in helminth infection. Nat Rev Immunol. 2007;7(12):975–987. Review. 48. Anzai N., Ichida K., Jutabha P., Kimura T., Babu E., Jin C.J. Plasma urate level is directly regulated by a voltage-driven urate efflux transporter URATv1 (SLC2A9) in humans. J Biol Chem. 2008;283(40):26834–26838. 49. Apasov S, Chen JF, Smith P, Sitkovsky M. A2A receptor dependent and A2A receptor independent effects of extracellular adenosine on murine thymocytes in the condition of adenosine deaminase deficiency. Blood. 2000; 95(12): 3859-3867. 50. Araya J., Rodrigo R., Videla L.A., Thielemann L., Orellana M., Pettinelli P. Increase in long-chain polyunsaturated fatty acid n - 6/n - 3 ratio in relation to hepatic steatosis in patients with non- alcoholic fatty liver disease. Clin Sci (Lond) 2004;106(6):635–643. 51. Aribas A., Kayrak M., Ulucan S., Keser A., Demir K., Alibasic H. The relationship between uric acid and erectile dysfunction in hypertensive subjects. Blood Press. 2014;23: 370–376. 52. Arvola L, Bertelsen G, Hassaf D, Ytrehus K. Positive inotropic and sustained anti-beta-adrenergic effect of diadenosine pentaphosphate in human and guinea pig hearts. Role of dinucleotide receptors and adenosine receptors. Acta Physiol Scand. 2004;182(3):277-285. 53. Arnold I.C., Mathisen S., Schulthess J., Danne C., Hegazy A.N., Powrie F. CD11c(+) monocyte/macrophages promote chronic Helicobacter hepaticus-induced intestinal inflammation through the production of IL-23. Mucosal Immunol. 2016; 9:352–363. 54. Auerbach A. Dose-ResponseAnalysis When There Is a Correlation between Affinity and Efficacy. Mol. Pharmacol. 2016; 89:297–302. 55. Babio N., Martínez-González M.A., Estruch R., Wärnberg J., Recondo J., Ortega-Calvo M. Associations between serum uric acid concentrations and metabolic syndrome and its components in the PREDIMED study. Nutr Metab Cardiovasc Dis. 2015;25(2):173–180. 56. Bakhtiari S., Toosi P., Samadi S., Bakhshi M. Assessment of uric acid level in the saliva of patients with oral lichen planus. Med Princ Pract. 2017;26(1):57–60. 57. Barabé F, Gilbert C, Liao N, Bourgoin SG, Naccache PH. Crystal-induced neutrophil activationVI. Involvement of FcgammaRIIIB (CD16) and CD11b in response to inflammatory microcrystals. FASEB J. 1998;12(2):209-220. doi:10.1096/fasebj.12.2.209 58. Barakat R., Abou El-Ela N.E., Sharaf S., El Sagheer O., Selim S., Tallima H. Efficacy and safety of arachidonic acid for treatment of school-age children in Schistosoma mansoni high-endemic regions. Am J Trop Med Hyg. 2015;92(4):797–804. 59. Bardin T., Richette P. Definition of hyperuricemia and gouty conditions. Curr. Opin. Rheumatol. 2014;26: 186–191. 60. Barikbin B., Yousefi M., Rahimi H., Hedayati M., Razavi S.M., Lotfi S. Antioxidant status in patients with lichen planus. Clin Exp Dermatol. 2011;36(8):851–854. 61. Bartáková V., Kuricová K., Pácal L., Nová Z., Dvořáková V., Švrčková M. Hyperuricemia contributes to the faster progression of diabetic kidney disease in type 2 diabetes mellitus. J Diabetes Complications. 2016;30(7):1300–1307. 62. Barylyak L.G., Malyuchkova R.V., Tolstanov O.B., Tymochko O.B., Hryvnak R.F., Uhryn M.R. Comparative estimation of informativeness of leukocytary index of adaptation by Garkavi and by Popovych. Medical Hydrology and Rehabilitation. 2013; 11(1): 5-20. 63. Basseville A., Bates S. Gout, genetics and ABC transporters. F1000 Biology Reports. 2011;3: 23. 64. Beck L.H. Requiem for gouty nephropathy. Kidney Int. 1986;30(2):280–287. 65. Becker B.F. Towards the physiological function of uric acid. Free Radical Biol Med. 1993;14(6):615–631. 66. Bellomo G., Venanzi S., Verdura C., Saronio P., Esposito A., Timio M. Association of uric acid with changes in kidney function in healthy normotensive individuals. Am J Kidney Dis. 2010;56(2):264–272. 67. Berntson GG, Bigger JT jr, Eckberg DL, Grossman P, Kaufman PG, Malik M, Nagaraja HN, Porges SW, Saul JP, Stone PH, Van der Molen MW. Heart Rate Variability: Origins, methods, and interpretive caveats. Psychophysiology. 1997; 34: 623-648. 68. Bianco C. Population of lymphocytes bearing a membrane receptor for antigen-antibody complex. J Exp Med. 1970; 134(4): 702-720. 69. Bjorkander S., Heidari-Hamedani G., Bremme K., Gunnarsson I., Holmlund U. Peripheral monocyte expression of the chemokine receptors CCR2, CCR5 and CXCR3 is altered at parturition in healthy women and in women with systemic lupus erythematosus. Scand. J. Immunol. 2013;77: 200–212. 70. Birch RE, Rosenthal AK, Polmer SH. Pharmacological modification of immunoregulatory T lymphocytes. II. Modulation of T lymphocyte cell surface characteristics. Clin Exp Immunol. 1982; 48(1): 231-238. 71. Bjornstad P., Lanaspa M.A., Ishimoto T., Kosugi T., Kume S., Jalal D. Fructose and uric acid in diabetic nephropathy. Diabetology. 2015;58(9):1993–2002. 72. Bjornstad P., Maahs D.M., Johnson R.J., Rewers M., Snell-Bergeon J.K. Estimated insulin sensitivity predicts regression of albuminuria in Type 1 diabetes. Diabetes Med. 2015;32(2):257–261. 73. Bjornstad P., Roncal C., Milagres T., Pyle L., Lanaspa M.A., Bishop F.K. Hyperfiltration and uricosuria in adolescents with type 1 diabetes. Pediatr Nephrol. 2016;(5):787–793. 74. Bjornstad P., Snell-Bergeon J.K., McFann K., Wadwa R.P., Rewers M., Rivard C.J. Serum uric acid and insulin sensitivity in adolescents and adults with and without type 1 diabetes. J Diabetes Complications. 2014;28(3):298–304. 75. Bobulescu I.A., Moe O.W. Renal transport of uric acid: evolving concepts and uncertainties. Adv Chronic Kidney Dis. 2012;19(6):358–371. 76. Bocarsly M.E., Powell E.S., Avena N.M., Hoebel B.G. High-fructose corn syrup causes characteristics of obesity in rats: increased body weight, body fat and triglyceride levels. Pharmacol Biochem Behav. 2010;97: 101–106. 77. Broz P., Dixit V.M. Inflammasomes: mechanism of assembly, regulation and signaling. Nat Rev Immunol. 2016;16(7):407–420. 78. Bruno C.M., Pricoco G., Cantone D., Elisa Marino E., Bruno F. Tubular handling of uric acid and factors influencing its renal excretion: a short review. EMJ Nephrol. 2016;4(1):92–97. 79. Busso N., Ea H.K. The mechanisms of inflammation in gout and pseudogout (CPP-induced arthritis) Rheumatismo. 2012;63(4):230–237. 80. Busso N., So A. Mechanisms of inflammation in gout. Arthritis Res Ther. 2010;12(2):206. 81. Busso N., So A. Microcrystals as DAMPs and their role in joint inflammation. Rheumatology (Oxford) 2012;51(7):1154–1160. 82. Cammalleri L., Malaguarnera M. Rasburicase represents a new tool for hyperuricemia in tumor lysis syndrome and in gout. International Journal of Medical Sciences. 2007;4(2):83–93. doi: 10.7150/ijms.4.83. 83. Campion E.W., Glynn R.J., DeLabry L.O. Asymptomatic hyperuricemia. Risks and consequences in the Normative Aging Study. Am J Med. 1987; 82:421. 84. Carito V., Ceccanti M., Tarani L., Ferraguti G., Chaldakov G. N., Fiore M. Neurotrophins' modulation by olive polyphenols. Current Medicinal Chemistry. 2016;23(28):3189–3197. 85. Carvalho LAC, Lopes JPPB, Kaihami GH, Silva RP, Bruni-Cordoso A, Baldini RL, Meotti FC. Uric acid disrupts hypochlorous acid production and bactericidal activity of HL-60 cells. Redox Biology. 2018; 16: 179-188. 86. Chakraborti G., Biswas R., Chakraborti S., Sen P.K. Altered serum uric acid level in lichen planus patients. Indian J Dermatol. 2014;59(6):558–561. 87. Chang B.S. Ancient insights into uric acid metabolism in primates. Proc Natl Acad Sci USA. 2014;111(10):3657–3658. 88. Chaudhary K., Malhotra K., Sowers J., Aroor A. Uric Acid - key ingredient in the recipe for cardiorenal metabolic syndrome. Cardiorenal Med. 2013;3(3):208–220. 89. Chen CJ, Shi Y, Hearn A, et al. MyD88-dependent IL-1 receptor signaling is essential for gouty inflammation stimulated by monosodium urate crystals. J Clin Invest. 2006;116(8):2262-2271. 90. Chen H., Cao G., Chen D. Q., et al. Metabolomics insights into activated redox signaling and lipid metabolism dysfunction in chronic kidney disease progression. Redox Biology. 2016; 10:168–178. 91. Chen Y., Xu B., Sun W., Sun J., Wang T., Xu Y. Impact of the serum uric acid level on subclinical atherosclerosis in middle-aged and elderly Chinese. J Atheroscler Thromb. 2015;22(8):823–832. 92. Chen-Xu M., Yokose C., Rai S.K., Pillinger M.H., Choi H.K. Contemporary Prevalence of Gout and Hyperuricemia in the United States and Decadal Trends: The National Health and Nutrition Examination Survey, 2007–2016. Arthritis Rheumatol. 2019; 71:991–999. 93. Cheung K.J., Tzameli I., Pissios P., Rovira I., Gavrilova O., Ohtsubo T. Xanthine oxidoreductase is a regulator of adipogenesis and PPARγ activity. Cell Metab. 2007; 5:115–128. 94. Cheungpasitporn W., Thongprayoon C., Harrison A.M., Erickson S.B. Admission hyperuricemia increases the risk of acute kidney injury in hospitalized patients. Clin Kidney J. 2016;9(1):51–56. 95. Chiquete E., Ruiz-Sandoval J.L., Murillo-Bonilla L.M., Arauz A., Orozco-Valera D.R., Ochoa-Guzmán A. Serum uric acid and outcome after acute ischemic stroke: PREMIER study. Cerebrovasc Dis. 2013;35(2):168–174. 96. Cho J., Kim C., Kang D.R., Park J. Hyperuricemia and uncontrolled hypertension in treated hypertensive patients: K-MetS study. Medicine (Baltimore) 2016;95(28): e4177. 97. Choi H.K., Curhan G. Independent impact of gout on mortality and risk for coronary heart disease. Circulation. 2007;116(8):894–900. 98. Choi H.K., Ford E.S. Prevalence of the metabolic syndrome in individuals with hyperuricemia. Am J Med. 2007;120(5):442–447. 99.Choi Y.J., Shin H.S., Choi H.S., Park J.W., Jo I., Oh E.S. Uric acid induces fat accumulation via generation of endoplasmic reticulum stress and SREBP-1c activation in hepatocytes. Lab Invest. 2014;94(10):1114–1125. 100.Chou Y.C., Kuan J.C., Yang T., Chou W.Y., Hsieh P.C., Bai C.H. Elevated uric acid level as a significant predictor of chronic kidney disease: a cohort study with repeated measurements. J Nephrol. 2015;28(4):457–462. 101. Cicerchi C., Li N., Kratzer J., Garcia G., Roncal-Jimenez C.A., Tanabe K. Uric acid-dependent inhibition of AMP kinase induces hepatic glucose production in diabetes and starvation: evolutionary implications of the uricase loss in hominids FASEB J. 2014;(8):3339–3350. 102. Cicero A., Rosticci M., Tartagni E., Parini A., Grandi E., D'Addato S. Serum uric acid level, but not renal function or arterial stiffness, is associated to worse blood pressure control in general practice : data from the brisighella heart study. J Hypertens. 2015;33(Suppl 1):e22. 103. Cicero A.F., Rosticci M., Fogacci F., Grandi E., D'Addato S., Borghi C. High serum uric acid is associated with poorly controlled blood pressure and higher arterial stiffness in hypertensive subjects. Eur J Intern Med. 2017;37: 38–42. 104. Cinel I, Gür S. Direct inotropic effects of propofol and adenosine on rat atrial muscle: possible mechanisms. Pharmacol Res. 2000;42(2):123-128. 105. Clarson L.E., Chandratre P., Hider S.L., Belcher J., Heneghan C., Roddy E. Increased cardiovascular mortality associated with gout: a systematic review and meta-analysis. Eur J Prev Cardiol. 2015;22(3):335–343. 106. Clémençon B., Lüscher B.P., Fine M., Baumann M.U., Surbek D.V., Bonny O. Expression, purification, and structural insights for the human uric acid transporter, GLUT9, using the Xenopus laevis oocytes system. PLoS ONE. 2014;9(10):e108852. 107. Cleophas M. C., Joosten L. A., Stamp L. K., Dalbeth N., Woodward O. M., Merriman T. R. ABCG2 polymorphisms in gout: insights into disease susceptibility and treatment approaches. Ph. DArmacogenomics and Personalized Medicine. 2017; 10:129–142. 108. Conen D, Wietlisbach V, Bovet P, Shamlaye C, Riesen W, Paccaud F, et al. Prevalence of hyperuricemia and relationship of serum uric acid with cardiovascular risk factors in a developing country. BMC public health. 2004; 4:9. 109. Convento M.S., Pessoa E., Dalboni M.A., Borges F.T., Schor N. Pro-inflammatory and oxidative effects of noncrystalline uric acid in human mesangial cells: contribution to hyperuricemic glomerular damage. Urol Res. 2011;39(1):21–27. 110.Cristóbal-García M., García-Arroyo F.E., Tapia E., Osorio H., Arellano-Buendía A.S., Madero M. Renal oxidative stress induced by long-term hyperuricemia alters mitochondrial function and maintains systemic hypertension. Oxid Med Cell Longev. 2015; 535686. 111. Culleton B.F., Larson M.G., Kannel W.B., Levy D. Serum uric acid and risk for cardiovascular disease and death: the Framingham Heart Study. Ann Intern Med. 1999;131(1):7–13. 112. Dalbeth N., Merriman T. Crystal ball gazing: new therapeutic targets for hyperuricemia and gout. Rheumatology (Oxford). 2009;48(3):222–226. 113. De Cosmo S., Viazzi F., Pacilli A., Giorda C., Ceriello A., Gentile S. Serum uric acid and risk of CKD in Type 2 diabetes. Clin J Am Soc Nephrol. 2015;10(11):1921–1929. 114. De Duve C, Wattiaux R. Functions of lysosomes. Annu Rev Physiol. 1966; 28:435–492. 115. De Vera M., Rahman M.M., Rankin J., Kopec J., Gao X., Choi H. Gout and the risk of Parkinson's disease: a cohort study. Art

    A qualitative study exploring the experiences of access and pathways to health care among BME community groups residing in Ayrshire

    Get PDF
    A review of the literature showed that Black and minority ethnic (BME) communities carry a disproportionately higher burden of illness than the general population and experience greater inequalities in health and health care provision. A growing body of research confirms that BME groups are under-represented in research. The main objective of the present study was to gain insights into the perspectives of BME community groups' experiences of accessing general and sexual health care services in Ayrshire. Semi-structured interviews were conducted with 11 participants, n = 5 men (age range: 32-65; mean age = 52.4 years), and n = 6 women (age range: 27-60; mean age = 47.67 years). Data were analysed employing Interpretative Phenomenological Analysis (IPA). The analysis is illustrated through the use of four super-ordinate themes: ‘It’s a Trust Thing’, ‘Minding the Gap(s)’, ‘Sexual Health: a Culture of Silence’, and ‘Personal Perception of Risk’. The findings both support and add to the existing trust literature by presenting a heuristic model of trust, and by showing that participants trust is dynamic in character, has a role, and serves various roles and functions that impact decisions about accessing health services. Significant gaps in knowledge about available health services and unique communication challenges that prevent full access to health care and health promotion information were found, resulting in unmet needs. Analysis charted culturally driven factors that prohibit discussions about sexual health concerns and found complex cognitions involved in the personal perception of risk that was meaningfully understood by participants that direct towards understanding risk in terms of a heuristic model. The implications for clinical practice, health promotion, health service development, and the direction of future research will be discussed.awd_pdtunpub2619_ethesesunpu

    Gout and Metabolic Disease: Investigation of Potential Relationship in the New Zealand Population

    Get PDF
    Hyperuricaemia, pathologically defined as the presence of higher levels of serum urate, results from a compromise in the delicate balance between the production and excretion of urate primarily in the liver and the kidneys, respectively. Hyperuricaemia is a prerequisite for gout, a painful inflammatory arthritis. The symptoms of gout arise from the body’s immune response to monosodium urate crystals that accumulate in the synovial fluid of the joints. Hyperuricaemia and gout are complex traits. A number of genetic loci confer risk to develop hyperuricaemia. Genome-wide association studies (GWAS), an indispensable tool in population genetics, has identified at least twenty eight genomic loci that contain variants affecting serum urate concentration. Gene-environment interactions also play a significant role in this context. Exogenous factors such as the intake of purine-rich foods increase the frequency of gout flares. Population-specific genetic effects on gout are as evident, if not more, as for other complex phenotypes. The prevalence of gout is much higher in the New Zealand Polynesian population compared to other populations. Approximately 7% of New Zealand Māori and Pacific Island people and 3% of New Zealand Europeans are affected by gout. The coexistence of metabolic conditions with gout, usually called gout-comorbidities, adds another level of complexity. However, not many studies have attempted to address the causal relationship between these traits. In fact, my research project was instigated as an attempt to study the causal associations between gout and its comorbidities and fill in some gap in the scientific literature. The research was, however, limited to three metabolic conditions/comorbidities of gout – imbalanced iron homeostasis, metabolic syndrome and disrupted lipid metabolism. My study shows an association between increased serum ferritin and the risk of gout and seeded an idea that the consumption of iron-rich diet may play a role in increasing the frequency and severity of gout flares. Genetic association analysis using two variants in the HFE gene was done to confirm the association between ferritin and urate, which showed positive association in a smaller dataset and provoked the idea to investigate the causality, if it exists, between gout and iron metabolism. Using the robust ‘Two-sample Mendelian randomisation’ approach and exploiting summary statistics data from two large GWA studies, I was able to find an evidence of a causal effect of iron on urate metabolism, but not urate on iron metabolism. In the context of the metabolic syndrome, the role of variants within/near the ADRB3, MC3R, MC4R and ADTRP genes were investigated. The positive effects identified for these variants supported the possible involvement of obesity and insulin resistance-related genes in gout pathophysiology. With the help of gene sequencing-based rare variant analyses, several novel population-specific association signals were found within the coding regions of two lipid-related genes, LRP2 and A1CF. Polynesian-specific novel genetic effects were identified to be predictive for gout for common variants within the LRP2 gene. Rare variants within the LRP2 gene were also identified and a higher prevalence of non-synonymous polymorphisms that can increase the risk of hyperuricaemia was observed in European individuals compared to Polynesians. These results indicated LRP2 to contribute to the difference in gout prevalence between Māori and Pacific Island individuals compared to the New Zealand European population. Collectively, my study reports a causal role of iron and ferritin in increasing serum urate concentration and the involvement of imbalanced iron homeostasis in hyperuricaemia. Also, positive genetic associations indicated that genes contributing to metabolic syndrome and lipid metabolism can increase the risk of gout, and also have population-specific effects for the Polynesian and European ancestral groups in New Zealand
    corecore