Psikopatolojilerde gen-çevre etkileşimi: Stresle ilgili genetik ve epigenetik süreçler

Stresli yaşam olaylarının birçok psikolojik problemle ilişki içinde olduğu uzun zamandır bilinmektedir. Stres ve psikiyatrik rahatsızlıklar arasındaki ilişki, biyolojik ve çevresel faktörlerin etkileşimlerini inceleyen çalışmalar sonucunda daha anlaşılır hale gelmektedir. Bu makalede, insan vücudundaki genetik ve epigenetik süreçleri içeren biyolojik mekanizmalar aracılığıyla psikopatolojiler üzerinde etkili olan stresle ilişkili çevresel faktörlere değinilmektedir. Otonom sinir sistemi ve hipotalamus- hipofiz-adrenal aksı gibi zihin sağlığı ve stres yönetimi açısından önemli yapıların işleyişinde rol alan genler üzerindeki tek nükleotid polimorfizmleri ve DNA metilasyonları, psikopatolojilerdeki gen-çevre etkileşimi mekanizmalarına ışık tutmaktadır. Erken ve yakın dönem olumsuz çevresel yaşantıların biyolojik süreçler ile bağlantılı bir biçimde psikiyatrik rahatsızlıklarla olan ilişkisine yönelik bulgular dikkat çekicidir. Özellikle, CRHR1, FKBP5, CRHBP, SLC6A4, NR3C1, OXTR ve BDNF gibi stresle ilgili genlerin depresyon, intihar, bipolar bozukluk, kaygı bozukluğu, çeşitli kişilik bozuklukları ve travma sonrası stres bozukluğu gibi çok sayıda psikiyatrik problemle ilişkisi çevresel yaşantılarla etkileşim içerisinde rapor edilmiştir. Söz konusu ilişkilerin bu problemlere yönelik tedavi yaklaşımlarının etkinliğinde rol oynadığını gösteren çalışmalar hızla artmaktadır. Bulgular yorumlanırken göz önünde bulundurulması gereken olası kısıtlılıklar ve gelecek çalışmalar için öneriler tartışılmıştır.

Gene-environment interaction in psychopathologies: Stress-related genetic and epigenetic influences

It has long been known that stressful life events are related to various psychological problems. The relationship between stress and psychiatric disorders has been further recognized due to reports of studies investigating interactions between biological and environmental factors. This paper is about the effect of stress-related environmental factors on psychopathologies through relevant biological mechanisms, such as genetic and epigenetic processes, in human body. Single-nucleotide polymorphisms and DNA methylation profiles of the genes playing role in the autonomic nervous system and the hypothalamic-pituitary-adrenal axis, which are essential for mental health and stress regulation, shed light upon gene-environment interactions in psychopathologies. Research results showing an association between early/late adversities and psychiatric problems through biological mechanisms deserve note. Relationships between genes related to stress regulation, such as CRHR1, FKBP5, CRHBP, SLC6A4, NR3C1, OXTR and BDNF, and various psychiatric conditions (e.g., depression, suicide, anxiety, bipolar disorder, personality disorders and post-traumatic stress disorder) have been suggested in an interaction with environmental factors. The number of studies reporting similar associations for therapeutic approaches to these disorders is also on the increase. Possible limitations when interpreting findings and suggestions for future research have also been discussed.

___

  • Babenko, O., Kovalchuk, I. ve Metz, G. A. (2015). Stressinduced perinatal and transgenerational epigenetic programming of brain development and mental health. Neuroscience & Biobehavioral Reviews, 48, 70-91.
  • Bakulski, K. M., Halladay, A., Hu, V. W., Mill, J. ve Fallin, M. D. (2016). Epigenetic research in neuropsychiatric disorders: The “tissue issue”. Current Behavioral Neuroscience Reports, 3(3), 264-274.
  • Bakusic, J., Schaufeli, W., Claes, S. ve Godderis, L. (2017). Stress, burnout and depression: A systematic review on DNA methylation mechanisms. Journal of Psychosomatic Research, 92, 34-44.
  • Barker, E. D., Walton, E. ve Cecil, C. A. (2018). Annual research review: DNA methylation as a mediator in the association between risk exposure and child and adolescent psychopathology. Journal of Child Psychology and Psychiatry, 59(4), 303-322.
  • Beach, S. R., Brody, G. H., Lei, M. K., Gibbons, F. X., Gerrard, M., Simons, R. L., Cutrona, C. E. ve Philibert, R. A. (2013). Impact of child sex abuse on adult psychopathology: A genetically and epigenetically informed investigation. Journal of Family Psychology, 27(1), 3-11.
  • Beach, S. R., Brody, G. H., Todorov, A. A., Gunter, T. D. ve Philibert, R. A. (2011). Methylation at 5HTT mediates the impact of child sex abuse on women's antisocial behavior: An examination of the Iowa adoptee sample. Psychosomatic Medicine, 73(1), 83-87.
  • Binder, E. B., Bradley, R. G., Liu, W., Epstein, M. P., Deveau, T. C., Mercer, K. B., Tang, Y., Gillespie, C. F., Heim, C. M., Nemeroff, C. B., Schwartz, A. C., Cubells, J. F. ve Ressler, K. J. (2008). Association of FKBP5 polymorphisms and childhood abuse with risk of posttraumatic stress disorder symptoms in adults. Jama, 299(11), 1291-1305.
  • Booij, L., Szyf, M., Carballedo, A., Frey, E. M., Morris, D., Dymov, S., Vaisheva, F., Ly, V., Fahey, C., Meaney, J., Gill, M. ve Frodl, T. (2015). DNA methylation of the serotonin transporter gene in peripheral cells and stress-related changes in hippocampal volume: A study in depressed patients and healthy controls. PloS One, 10(3), e0119061.
  • Bradley, R. G., Binder, E. B., Epstein, M. P., Tang, Y., Nair, H. P., Liu, W., Gillespie, C. F., Berg, T., Evces, M., Newport, D. J., Stowe, Z. N., Heim, C. M., Nemeroff, C. B., Schwartz, A., Cubells, J. F. ve Ressler, K. J. (2008).
  • Influence of child abuse on adult depression: Moderation by the corticotropin-releasing hormone receptor gene. Archives of General Psychiatry, 65(2), 190-200.
  • Carlberg, L., Scheibelreiter, J., Hassler, M. R., Schloegelhofer, M., Schmoeger, M., Ludwig, B., Kasper, S., Aschauer, H., Egger, G. ve Schosser, A. (2014). Brain-derived neurotrophic factor (BDNF)–epigenetic regulation in unipolar and bipolar affective disorder. Journal of Affective Disorders, 168, 399-406.
  • Chen, Y. ve Baram, T. Z. (2016). Toward understanding how early-life stress reprograms cognitive and emotional brain networks. Neuropsychopharmacology, 41(1), 197- 206.
  • D'Addario, C., Dell'Osso, B., Galimberti, D., Palazzo, M. C., Benatti, B., Di Francesco, A., Scarpini, E., Altamura, A. C. ve Maccarrone, M. (2013). Epigenetic modulation of BDNF gene in patients with major depressive disorder. Biological Psychiatry, 73(2), e6-e7. D'Addario, C., Dell'Osso, B., Palazzo, M. C., Benatti, B., Lietti, L., Cattaneo, E., Galimberti, D., Fenoglio, C., Cortini, F., Scarpini, E., Arosio, B., Di Francesco, A., Di Benedetto, M., Romualdi, P., Candeletti, S., Mari, D., Bergamaschini, L., Bresolin, N., Maccarrone, M. ve Altamura, A. C. (2012). Selective DNA methylation of BDNF promoter in bipolar disorder: Differences among patients with BDI and BDII. Neuropsychopharmacology, 37(7), 1647-1655.
  • Dempster, E. L., Pidsley, R., Schalkwyk, L. C., Owens, S., Georgiades, A., Kane, F., Kalidindi, S., Picchioni, M., Kravariti, E., Toulopoulou, T., Murray, R. M. ve Mill, J. (2011). Disease-associated epigenetic changes in monozygotic twins discordant for schizophrenia and bipolar disorder. Human Molecular Genetics, 20(24), 4786-4796.
  • Docherty, S. ve Mill, J. (2008). Epigenetic mechanisms as mediators of environmental risks for psychiatric disorders. Psychiatry, 7(12), 500-506.
  • Duman, E. A. ve Canli, T. (2015). Influence of life stress, 5- HTTLPR genotype, and SLC6A4 methylation on gene expression and stress response in healthy Caucasian males. Biology of Mood & Anxiety Disorders, 5(1), 2.
  • Essex, M. J., Thomas Boyce, W., Hertzman, C., Lam, L. L., Armstrong, J. M., Neumann, S. M. ve Kobor, M. S. (2013). Epigenetic vestiges of early developmental adversity: Childhood stress exposure and DNA methylation in adolescence. Child Development, 84(1), 58-75.
  • Gatt, J. M., Nemeroff, C. B., Dobson-Stone, C., Paul, R. H., Bryant, R. A., Schofield, P. R., Gordon, E., Kemp, A. H. ve Williams, L. M. (2009). Interactions between BDNF Val66Met polymorphism and early life stress predict brain and arousal pathways to syndromal depression and anxiety. Molecular Psychiatry, 14(7), 681-695. Gee, D. G. ve Casey, B. J. (2015). The impact of developmental timing for stress and recovery. Neurobiology of Stress, 1, 184-194.
  • Glover, V., O'Donnell, K. J., O'Connor, T. G. ve Fisher, J. (2018). Prenatal maternal stress, fetal programming, and mechanisms underlying later psychopathology-a global perspective. Development and Psychopathology, 30(3), 843-854.
  • Gunnar, M. ve Quevedo, K. (2007). The neurobiology of stress and development. Annual Review of Psychology, 58(1), 145-173.
  • Hackman, D. A., Farah, M. J. ve Meaney, M. J. (2010). Socioeconomic status and the brain: Mechanistic insights from human and an
  • Hannon, E., Lunnon, K., Schalkwyk, L. ve Mill, J. (2015). Interindividual methylomic variation across blood, cortex, and cerebellum: Implications for epigenetic studies Heim, C. ve Binder, E. B. (2012). Current research trends in early life stress and depression: Review of human studies on sensitive periods, gene-environment interactions, and epigenetics. Experimental Neurology, 233(1), 102-111.
  • Hompes, T., Izzi, B., Gellens, E., Morreels, M., Fieuws, S., Pexsters, A., Schops, G., Dom, M., Van Bree, R., Freson, K., Verhaeghe, J., Spitz, B., Demyttenaere, K., Glover, V., Van den Bergh, B., Allegaert, K. ve Claes, S. (2013). Investigating the influence of maternal cortisol and emotional state during pregnancy on the DNA methylation status of the glucocorticoid receptor gene (NR3C1) promoter region in cord blood. Journal of Psychiatric Research, 47(7), 880-891.
  • Ising, M., Depping, A. M., Siebertz, A., Lucae, S., Unschuld, P. G., Kloiber, S., Horstmann, S., Uhr, M., Müller-Myhsok, B. ve Holsboer, F. (2008). Polymorphisms in the FKBP5 gene region modulate recovery from psychosocial stress in healthy controls. European Journal of Neuroscience, 28(2), 389-398.
  • Januar, V., Saffery, R. ve Ryan, J. (2015). Epigenetics and depressive disorders: A review of current progress and future directions. International Journal of Epidemiology, 44(4), 1364-1387.
  • Jin, M. J., Jeon, H., Hyun, M. H. ve Lee, S. H. (2019). Influence of childhood trauma and brain-derived neurotrophic factor Val66Met polymorphism on posttraumatic stress symptoms and cortical thickness. Scientific Reports, 9(1), 1-12.
  • Kato, T., Iwamoto, K., Kakiuchi, C., Kuratomi, G. ve Okazaki, Y. (2005). Genetic or epigenetic difference causing discordance between monozygotic twins as a clue to molecular basis of mental disorders. Molecular Psychiatry, 10(7), 622-630.
  • Klengel, T., Pape, J., Binder, E. B. ve Mehta, D. (2014). The role of DNA methylation in stress-related psychiatric disorders. Neuropharmacology, 80, 115-132.
  • Koenen, K. C., Uddin, M., Chang, S. C., Aiello, A. E., Wildman, D. E., Goldmann, E. ve Galea, S. (2011). SLC6A4 methylation modifies the effect of the number of traumatic events on risk for posttraumatic strss disorder. Depression and Anxiety, 28(8), 639-647.
  • Kumsta, R. (2019). The role of epigenetics for understanding mental health difficulties and its implications for psychotherapy research. Psychology and Psychotherapy: Theory, Research and Practice, 92(2), 190-207.
  • Labonte, B., Yerko, V., Gross, J., Mechawar, N., Meaney, M. J., Szyf, M. ve Turecki, G. (2012). Differential glucocorticoid receptor exon 1B, 1C, and 1H expression and methylation in suicide completers with a history of childhood abuse. Biological Psychiatry, 72(1), 41-48.
  • Lenroot, R. K. ve Giedd, J. N. (2006). Brain development in children and adolescents: Insights from anatomical magnetic resonance imaging. Neuroscience & Biobehavioral Reviews, 30(6), 718-729.
  • Liu, Z., Zhu, F., Wang, G., Xiao, Z., Tang, J., Liu, W., Wang, H., Liu, H., Wang, X., Wu, Y., Cao, Z. ve Li, W. (2007). Association study of corticotropin-releasing hormone receptor1 gene polymorphisms and antidepressant response in major depressive disorders. Neuroscience Letters, 414(2), 155-158.
  • Machin, G. (2009). Non-identical monozygotic twins, intermediate twin types, zygosity testing, and the nonrandom nature of monozygotic twinning: A review. American Journal of Medical Genetics Part C: Seminars in Medical Genetics, 151(2), 110-127.
  • Mann, J. J. ve Currier, D. M. (2010). Stress, genetics and epigenetic effects on the neurobiology of suicidal behavior and depression. European Psychiatry, 25(5), 268- 271.
  • Maud, C., Ryan, J., McIntosh, J. E. ve Olsson, C. A. (2018). The role of oxytocin receptor gene (OXTR) DNA methylation (DNAm) in human social and emotional functioning: A systematic narrative review. BMC Psychiatry, 18(1), 154.
  • McGowan, P. O., Sasaki, A., D'alessio, A. C., Dymov, S., Labonté, B., Szyf, M., Turecki, G. ve Meaney, M. J. (2009). Epigenetic regulation of the glucocorticoid receptor in human brain associates with childhood abuse. Nature Neuroscience, 12(3), 342-348.
  • Meaney, M. J. (2010). Epigenetics and the biological definition of gene×environment interactions. Child Development, 81(1), 41-79.
  • Meehl, P. E. (1962). Schizotaxia, schizotypy, schizophrenia. American Psychologist, 17(12), 827-838.
  • Mill, J., Dempster, E., Caspi, A., Williams, B., Moffitt, T. ve Craig I. (2006). Evidence for monozygotic twin (MZ) discordance in methylation level at two CpG sites in the promoter region of the catechol-O-methyltransferase (COMT) gene. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics, 141(4), 421-425.
  • Moore, S. R. (2017). Commentary: What is the case for candidate gene approaches in the era of high‐throughput genomics? A response to Border and Keller (2017). Journal of Child Psychology and Psychiatry, 58(3), 331-334.
  • Nguyen, A., Rauch, T. A., Pfeifer, G. P. ve Hu, V. W. (2010). Global methylation profiling of lymphoblastoid cell lines reveals epigenetic contributions to autism spectrum disorders and a novel autism candidate gene, RORA, whose protein product is reduced in au tistic brain. The FASEB Journal, 24(8), 3036-3051.
  • Oberlander, T. F., Weinberg, J., Papsdorf, M., Grunau, R., Misri, S. ve Devlin, A. M. (2008). Prenatal exposure to maternal depression, neonatal methylation of human glucocorticoid receptor gene (NR3C1) and infant cortisol stress responses. Epigenetics, 3(2), 97-106.
  • Palma-Gudiel, H., Córdova-Palomera, A., Leza, J. C. ve Fañanás, L. (2015). Glucocorticoid receptor gene (NR3C1) methylation processes as mediators of early adversity in stress-related disorders causality: A critical review. Neuroscience & Biobehavioral Reviews, 55, 520-535.
  • Pechtel, P. ve Pizzagalli, D. A. (2011). Effects of early life stress on cognitive and affective function: An integrated review of human literature. Psychopharmacology, 214(1), 55-70.
  • Perroud, N., Paoloni-Giacobino, A., Prada, P., Olié, E., Salzmann, A., Nicastro, R., Guillaume, S., Mouthon, D.,Stouder, C., Dieben, K., Huguelet, P., Courtet, P. ve Malafosse,
  • A. (2011). Increased methylation of glucocorticoid receptor gene (NR3C1) in adults with a history of childhood maltreatment: A link with the severity and type of trauma. Translational Psychiatry, 1(12), e59-e59.
  • Perroud, N., Rutembesa, E., Paoloni-Giacobino, A., Mutaba ruka, J., Mutesa, L., Stenz, L., Malafosse, A. ve Karege, F. (2014). The Tutsi genocide and transgenerational transmission of maternal stress: Epigenetics and biology of the HPA axis. The World Journal of Biological Psychiatry, 15(4), 334-345.
  • Perroud, N., Salzmann, A., Prada, P., Nicastro, R., Hoeppli, M. E., Furrer, S., Ardu, S., Krejci, I., Karege, F. ve Malafosse, A. (2013). Response to psychotherapy in borderline personality disorder and methylation status of the BDNF gene. Translational Psychiatry, 3(1), e207-e207.
  • Plomin, R., DeFries, J. C., Knopik, V. S. ve Neiderhiser, J. M. (2013). Behavioral genetics (6. baskı). Worth. Polanczyk, G., Caspi, A., Williams, B., Price, T. S., Danese, A., Sugden, K., Uher, R., Poulton, R. veMoffitt, T. E. (2009). Protective effect of CRHR1 gene variants on the development of adult depression following childhood maltreatment: Replication and extension. Archives of General Psychiatry, 66(9), 978-985.
  • Radtke, K. M., Ruf, M., Gunter, H. M., Dohrmann, K., Schauer, M., Meyer, A. ve Elbert, T. (2011). Transgenerational impact of intimate partner violence on methylation in the promoter of the glucocorticoid receptor. Translational Psychiatry, 1(7), e21-e21.
  • Radtke, K. M., Schauer, M., Gunter, H. M., Ruf-Leuschner, M., Sill, J., Meyer, A. ve Elbert, T. (2015). Epigenetic modifications of the glucocorticoid receptor gene are associated with the vulnerability to psychopathology in childhood maltreatment. Translational Psychiatry, 5(5), e571-e571.
  • Roberts, S., Keers, R., Lester, K. J., Coleman, J. R. I., Breen, G., Arendt, K., Blatter-Meunier, J., Cooper, P., Creswell, C., Fjermestad, K., Havik, O. E., Herren, C., Hogendoorn, S. M., Hudson, J. L., Krause, K., Lyneham, H. J., Morris, T., Nauta, M., Rapee, R. M., Rey, Y. … Wong, C. C. Y. (2015). HPA axis related genes and response to psychological therapies: Genetics and epigenetics. Depression and Anxiety, 32(12), 861-870.
  • Roberts, S., Lester, K. J., Hudson, J. L., Rapee, R. M., Creswell, C., Cooper, P. J., Thirlwall, K. J., Coleman, J. R. I., Breen, G., Wong, C. C. Y. ve Eley, T. C. (2014). Serotonin tranporter methylation and response to cognitive behaviour therapy in children with anxiety disorders. Translational Psychiatry, 4(9), e444.
  • Rosa, A., Picchioni, M. M., Kalidindi, S., Loat, C. S., Knight, J., Toulopoulou, T., Vonk, R., van der Schot, A. C., Nolen, W., Kahn, R. S., McGuffin, P., Murray, R. M. ve Craig, I. W. (2007). Differential methylation of the Xchromosome is a possible source of discordance for bipolar disorder female monozygotic twins. American Journal of Medical Genetics Part B: Neuropsychiatric Genetics, 147(4), 459-462.
  • Roy, A., Hodgkinson, C. A., DeLuca, V., Goldman, D. ve Enoch, M. A. (2012). Two HPA axis genes, CRHBP and FKBP5, interact with childhood trauma to increase the risk for suicidal behavior. Journal of Psychiatric Research, 46(1), 72-79.
  • Smoller, J. W., Andreassen, O. A., Edenberg, H. J., Faraone, S. V., Glatt, S. J. ve Kendler, K. S. (2019). Psychiatric genetics and the structure of psychopathology. Molecular Psychiatry, 24(3), 409-420.
  • Stricker, S. H., Köferle, A. ve Beck, S. (2017). From profiles to function in epigenomics. Nature Reviews Genetics, 18(1), 51-66.
  • Turecki, G., Ernst, C., Jollant, F., Labonté, B. ve Mechawar, N. (2012). The neurodevelopmental origins of suicidal behavior. Trends in Neurosciences, 35(1), 14-23.
  • Tyrka, A. R., Price, L. H., Marsit, C., Walters, O. C. ve Carpenter, L. L. (2012). Childhood adversity and epigenetic modulation of the leukocyte glucocorticoid receptor: Preliminary findings in healthy adults. PloS One, 7(1), e30148.
  • Unternaehrer, E., Luers, P., Mill, J., Dempster, E., Meyer, A. H., Staehli, S., Lieb, R., Hellhammer, D. H. veMeinlschmidt, G. (2012). Dynamic changes in DNA methylation of stress-associated genes (OXTR, BDNF) after acute psychosocial stress. Translational Psychiatry, 2(8), e150- e150.
  • Wang, Q., Shelton, R. C. ve Dwivedi, Y. (2018). Interaction between early-life stress and FKBP5 gene variants in major depressive disorder and post-traumatic stress disorder: A systematic review and meta-analysis. Journal of Affective Disorders, 225, 422-428.
  • Yehuda, R., Daskalakis, N. P., Desarnaud, F., Makotkine, I., Lehrner, A. L., Koch, E., Flory, J. D., Buxbaum, J. D.,
  • Meaney, M. J. ve Bierer, L. M. (2013). Epigenetic biomarkers as predictors and correlates of symptom improvement following psychotherapy in combat veterans with PTSD. Frontiers in Psychiatry, 4, 118.
  • Yehuda, R., Flory, J. D., Bierer, L. M., Henn-Haase, C., Lehrner, A., Desarnaud, F., Makotkine, I., Daskalakis, N. P., Marmar, C. R. ve Meaney, M. J. (2015). Lower
  • methylation of glucocorticoid receptor gene promoter 1F in peripheral blood of veterans with posttraumatic stress disorder. Biological Psychiatry, 77(4), 356-364.
  • Yehuda, R., Golier, J. A., Yang, R. K. ve Tischler, L. (2004). Enhanced sensitivity to glucocorticoids in peripheral mononuclear leukocytes in posttraumatic stress disorder. Biological Psychiatry, 55(11), 1110-1116.
  • Zannas, A. S., Wiechmann, T., Gassen, N. C. ve Binder, E. B. (2016). Gene–stress–epigenetic regulation of FKBP5: Clinical and translational implications. Neuropsychopharmacology, 41(1), 261-274.
Klinik Psikoloji Dergisi-Cover
  • ISSN: 2602-4438
  • Yayın Aralığı: Yılda 3 Sayı
  • Başlangıç: 2017
  • Yayıncı: Klinik Psikoloji Araştırmaları Derneği
Sayıdaki Diğer Makaleler

Yale-Brown Obsesyon Kompulsiyon Ölçeği-Türkçe Öz Bildirim Formu: Güvenirlik ve geçerlik çalışması

Filiz KOÇOĞLU, Başak BAHTİYAR

Psikoterapideki tabu: Olumsuz etkilere dair sistematik bir gözden geçirme

Esra ALTIN GÖK, Orçun YORULMAZ

The investigation of the mediating role of impostor phenomenon in the relationship between maladaptive perfectionism and depression among residents

Elif Elvan MİREL, Hale ÖGEL-BALABAN

Investigating alexithymia, emotional expression, childhood trauma, and attachment in self-reported disordered eating behaviour

İlayda ÖZDEMİR, Şermin TÜKEL

Çevrimiçi psikoterapi yöntemlerinin farklı gruplar (yetişkin, çocuk, ergen ve aile) temelinde incelenmesi

Seda OKTAY, Ezgi Didem MERDAN YILDIZ, Pelin KARACA DİNÇ, Gülsen ERDEN

Psikopatolojilerde gen-çevre etkileşimi: Stresle ilgili genetik ve epigenetik süreçler

Esra ZIVRALI YARAR

Çocuk kaybı sonrası ebeveynlerin ilişkileri

Serkan ÖZGÜN, Belgin DERYALAR, İlke Sine EĞECİ, Bülent AYKUTOĞLU, Asuman BÜYÜKCAN TETİK, Gökçen BULUT

COVID-19 pandemisi sürecinde sigara ve alkol kullanan bireylerde travma sonrası stres bozukluğu semptomlarının incelenmesi

Sevda ACAR, Hadiye Bostancı DEMİRCİ, Derya ŞAŞMAN KAYLI, Görkem YARARBAŞ

Investigating the symptoms of post-traumatic stress disorder in individuals who use alcohol and cigarette during the COVID-19 pandemic

Sevda ACAR, Hadiye Bostancı DEMİRCİ, Derya ŞAŞMAN KAYLI, Görkem YARARBAŞ