Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Volkan I. Sayin is active.

Publication


Featured researches published by Volkan I. Sayin.


Science Translational Medicine | 2014

Antioxidants Accelerate Lung Cancer Progression in Mice

Volkan I. Sayin; Mohamed X. Ibrahim; Erik Larsson; Jonas A. Nilsson; Per Lindahl; Martin O. Bergo

The antioxidants acetylcysteine and vitamin E accelerate tumor progression and reduce survival in mouse models of lung cancer by disrupting the ROS-p53 axis. The Dark Side of Antioxidants Antioxidants, or chemical compounds that prevent oxidation of other molecules, are widely marketed as dietary supplements with a variety of health claims. One particular characteristic that was frequently attributed to antioxidants is an ability to decrease the risk of cancer. However, a number of studies have shed doubt on this claim in recent years, as emerging evidence has suggested that antioxidants may actually increase the risk of some forms of cancer. Now, a study by Sayin and coauthors sheds new light on this issue and suggests that antioxidants may have a particularly detrimental effect in lung cancer development. When mice carrying mutations that increase their risk of lung cancer were treated with antioxidants, their early precancerous lesions progressed more quickly, and the mice developed more tumors and at more advanced stages. The antioxidants did reduce oxidative stress and DNA damage as expected, but at the same time, they also reduced the expression of p53, a key tumor suppressor protein. This work was done in cells and in mice, but the authors took care to make it as relevant to humans as possible. Thus, the mice were treated with types and doses of antioxidants (vitamin E and acetylcysteine) that healthy humans use, and the results were confirmed in human lung cancer cell lines. Although the current study does not show what would happen to wild-type mice or healthy people using antioxidants, it provides evidence for a procarcinogenic role of antioxidants in people who are already at a higher risk of cancer, such as smokers. Antioxidants are widely used to protect cells from damage induced by reactive oxygen species (ROS). The concept that antioxidants can help fight cancer is deeply rooted in the general population, promoted by the food supplement industry, and supported by some scientific studies. However, clinical trials have reported inconsistent results. We show that supplementing the diet with the antioxidants N-acetylcysteine (NAC) and vitamin E markedly increases tumor progression and reduces survival in mouse models of B-RAF– and K-RAS–induced lung cancer. RNA sequencing revealed that NAC and vitamin E, which are structurally unrelated, produce highly coordinated changes in tumor transcriptome profiles, dominated by reduced expression of endogenous antioxidant genes. NAC and vitamin E increase tumor cell proliferation by reducing ROS, DNA damage, and p53 expression in mouse and human lung tumor cells. Inactivation of p53 increases tumor growth to a similar degree as antioxidants and abolishes the antioxidant effect. Thus, antioxidants accelerate tumor growth by disrupting the ROS-p53 axis. Because somatic mutations in p53 occur late in tumor progression, antioxidants may accelerate the growth of early tumors or precancerous lesions in high-risk populations such as smokers and patients with chronic obstructive pulmonary disease who receive NAC to relieve mucus production.


Science Translational Medicine | 2015

Antioxidants can increase melanoma metastasis in mice

Le Gal K; Mohamed X. Ibrahim; Wiel C; Volkan I. Sayin; Murali K. Akula; Christin Karlsson; Martin Dalin; Levent M. Akyürek; Per Lindahl; Jonas A. Nilsson; Martin O. Bergo

Antioxidants increase migration and invasion of human melanoma cells and accelerate metastasis in an endogenous mouse model of malignant melanoma. Another strike against antioxidants Antioxidants are found in a variety of foods and dietary supplements and are frequently used with the goal of preventing cancer, but mounting evidence suggests that they may not be as beneficial as once thought. Clinical studies have shown mixed or no benefits, and other works demonstrated that antioxidants may accelerate the progression of lung cancer. Now, Le Gal et al. discovered that some common antioxidants increase the rate of melanoma cell migration and invasion and increase metastasis in a mouse model. These are early findings, and additional work will be required to confirm the generalizability of this observation. Nevertheless, the results suggest a need for caution in the use of antioxidants, especially for patients with existing cancer. Antioxidants in the diet and supplements are widely used to protect against cancer, but clinical trials with antioxidants do not support this concept. Some trials show that antioxidants actually increase cancer risk and a study in mice showed that antioxidants accelerate the progression of primary lung tumors. However, little is known about the impact of antioxidant supplementation on the progression of other types of cancer, including malignant melanoma. We show that administration of N-acetylcysteine (NAC) increases lymph node metastases in an endogenous mouse model of malignant melanoma but has no impact on the number and size of primary tumors. Similarly, NAC and the soluble vitamin E analog Trolox markedly increased the migration and invasive properties of human malignant melanoma cells but did not affect their proliferation. Both antioxidants increased the ratio between reduced and oxidized glutathione in melanoma cells and in lymph node metastases, and the increased migration depended on new glutathione synthesis. Furthermore, both NAC and Trolox increased the activation of the small guanosine triphosphatase (GTPase) RHOA, and blocking downstream RHOA signaling abolished antioxidant-induced migration. These results demonstrate that antioxidants and the glutathione system play a previously unappreciated role in malignant melanoma progression.


Science | 2013

Targeting Isoprenylcysteine Methylation Ameliorates Disease in a Mouse Model of Progeria

Mohamed X. Ibrahim; Volkan I. Sayin; Murali K. Akula; Meng Liu; Loren G. Fong; Stephen G. Young; Martin O. Bergo

Methylation and Methuselah? Hutchinson-Gilford progeria syndrome (HGPS) and other prelamin A–associated progeroid disorders arise when farnesylated and methylated forms of prelamin A accumulate at the nuclear envelope. Ibrahim et al. (p. 1330, published online 16 May; see the Perspective by Johnson) show that reducing the activity of the isoprenylcysteine carboxyl methyltransferase (ICMT) mislocalizes prelamin A, triggers prelamin A–dependent AKT-mTOR signaling, and eliminates disease phenotypes in 30-week-old progeria model mice. Reduced ICMT expression increased the proliferation and delayed the premature senescence of progeria model mouse fibroblasts and cells from children with HGPS. Reduced protein methyltransferase activity improves progeria-like disease phenotypes. [Also see Perspective by Johnson] Several progeroid disorders, including Hutchinson-Gilford progeria syndrome (HGPS) and restrictive dermopathy (ZMPSTE24 deficiency), arise when a farnesylated and methylated form of prelamin A accumulates at the nuclear envelope. Here, we found that a hypomorphic allele of isoprenylcysteine carboxyl methyltransferase (ICMT) increased body weight, normalized grip strength, and prevented bone fractures and death in Zmpste24-deficient mice. The reduced ICMT activity caused prelamin A mislocalization within the nucleus and triggered prelamin A–dependent activation of AKT-mammalian target of rapamycin (mTOR) signaling, which abolished the premature senescence of Zmpste24-deficient fibroblasts. ICMT inhibition increased AKT-mTOR signaling and proliferation and delayed senescence in human HGPS fibroblasts but did not reduce the levels of misshapen nuclei in mouse and human cells. Thus, targeting ICMT might be useful for treating prelamin A–associated progeroid disorders.


Nature Medicine | 2017

Keap1 loss promotes Kras -driven lung cancer and results in dependence on glutaminolysis

Rodrigo Romero; Volkan I. Sayin; Shawn M. Davidson; Matthew R. Bauer; Simranjit X. Singh; Sarah LeBoeuf; Triantafyllia R. Karakousi; Donald Christian Ellis; Arjun Bhutkar; Francisco J. Sánchez-Rivera; Lakshmipriya Subbaraj; Britney Martinez; Roderick T. Bronson; Justin R. Prigge; Edward E. Schmidt; Craig J. Thomas; Chandra Goparaju; Angela M. Davies; Igor Dolgalev; Adriana Heguy; Viola Allaj; John T. Poirier; Andre L. Moreira; Charles M. Rudin; Harvey I. Pass; Matthew G. Vander Heiden; Tyler Jacks; Thales Papagiannakopoulos

Treating KRAS-mutant lung adenocarcinoma (LUAD) remains a major challenge in cancer treatment given the difficulties associated with directly inhibiting the KRAS oncoprotein. One approach to addressing this challenge is to define mutations that frequently co-occur with those in KRAS, which themselves may lead to therapeutic vulnerabilities in tumors. Approximately 20% of KRAS-mutant LUAD tumors carry loss-of-function mutations in the KEAP1 gene encoding Kelch-like ECH-associated protein 1 (refs. 2, 3, 4), a negative regulator of nuclear factor erythroid 2-like 2 (NFE2L2; hereafter NRF2), which is the master transcriptional regulator of the endogenous antioxidant response. The high frequency of mutations in KEAP1 suggests an important role for the oxidative stress response in lung tumorigenesis. Using a CRISPR–Cas9-based approach in a mouse model of KRAS-driven LUAD, we examined the effects of Keap1 loss in lung cancer progression. We show that loss of Keap1 hyperactivates NRF2 and promotes KRAS-driven LUAD in mice. Through a combination of CRISPR–Cas9-based genetic screening and metabolomic analyses, we show that Keap1- or Nrf2-mutant cancers are dependent on increased glutaminolysis, and this property can be therapeutically exploited through the pharmacological inhibition of glutaminase. Finally, we provide a rationale for stratification of human patients with lung cancer harboring KRAS/KEAP1- or KRAS/NRF2-mutant lung tumors as likely to respond to glutaminase inhibition.


Oncogene | 2016

Oncogene-induced senescence underlies the mutual exclusive nature of oncogenic KRAS and BRAF

J Cisowski; Volkan I. Sayin; Meng Liu; Christin Karlsson; Martin O. Bergo

KRAS and BRAF are among the most commonly mutated oncogenes in human cancer that contribute to tumorigenesis in both distinct and overlapping tissues. However, KRAS and BRAF mutations are mutually exclusive; they never occur in the same tumor cell. The reason for the mutual exclusivity is unknown, but there are several possibilities. The two mutations could be functionally redundant and not create a selective advantage to tumor cells. Alternatively, they could be deleterious for the tumor cell and induce apoptosis or senescence. To distinguish between these possibilities, we activated the expression of BRAFV600E and KRASG12D from their endogenous promoters in mouse lungs. Although the tumor-forming ability of BRAFV600E was higher than KRASG12D, KRASG12D tumors were larger and more advanced. Coactivation of BRAFV600E and KRASG12D markedly reduced lung tumor numbers and overall tumor burden compared with activation of BRAFV600E alone. Moreover, several tumors expressed only one oncogene, suggesting negative selection against expression of both. Similarly, expression of both oncogenes in mouse embryonic fibroblasts essentially stopped proliferation. The expression of both oncogenes hyperactivated the MEK-ERK-cyclin D pathway but reduced proliferation by increasing the production of p15, p16 and p19 proteins encoded by the Ink4/Arf locus and thereby increased senescence-associated β-galactosidase-positive cells. The data suggest that coexpression of BRAFV600E and KRASG12D in early tumorigenesis leads to negative selection due to oncogene-induced senescence.


Circulation Research | 2014

Loss of One Copy of Zfp148 Reduces Lesional Macrophage Proliferation and Atherosclerosis in Mice by Activating p53

Volkan I. Sayin; Omar M. Khan; Lara E. Pehlivanoglu; Anna Staffas; Mohamed X. Ibrahim; Annika Asplund; Pia Ågren; Anna Nilton; Göran Bergström; Martin O. Bergo; Jan Borén; Per Lindahl

Rationale: Cell proliferation and cell cycle control mechanisms are thought to play central roles in the pathogenesis of atherosclerosis. The transcription factor Zinc finger protein 148 (Zfp148) was shown recently to maintain cell proliferation under oxidative conditions by suppressing p53, a checkpoint protein that arrests proliferation in response to various stressors. It is established that inactivation of p53 accelerates atherosclerosis, but whether increased p53 activation confers protection against the disease remains to be determined. Objective: We aimed to test the hypothesis that Zfp148 deficiency reduces atherosclerosis by unleashing p53 activity. Methods and Results: Mice harboring a gene-trap mutation in the Zfp148 locus (Zfp148gt/+) were bred onto the apolipoprotein E (Apoe)–/– genetic background and fed a high-fat or chow diet. Loss of 1 copy of Zfp148 markedly reduced atherosclerosis without affecting lipid metabolism. Bone marrow transplantation experiments revealed that the effector cell is of hematopoietic origin. Peritoneal macrophages and atherosclerotic lesions from Zfp148gt/+Apoe–/– mice showed increased levels of phosphorylated p53 compared with controls, and atherosclerotic lesions contained fewer proliferating macrophages. Zfp148gt/+Apoe–/– mice were further crossed with p53-null mice (Trp53–/– [the gene encoding p53]). There was no difference in atherosclerosis between Zfp148gt/+Apoe–/– mice and controls on a Trp53+/– genetic background, and there was no difference in levels of phosphorylated p53 or cell proliferation. Conclusions: Zfp148 deficiency increases p53 activity and protects against atherosclerosis by causing proliferation arrest of lesional macrophages, suggesting that drugs targeting macrophage proliferation may be useful in the treatment of atherosclerosis.


Nature Communications | 2016

Pan-cancer transcriptomic analysis associates long non-coding RNAs with key mutational driver events

Arghavan Ashouri; Volkan I. Sayin; Jimmy Van den Eynden; Simranjit X. Singh; Thales Papagiannakopoulos; Erik Larsson

Thousands of long non-coding RNAs (lncRNAs) lie interspersed with coding genes across the genome, and a small subset has been implicated as downstream effectors in oncogenic pathways. Here we make use of transcriptome and exome sequencing data from thousands of tumours across 19 cancer types, to identify lncRNAs that are induced or repressed in relation to somatic mutations in key oncogenic driver genes. Our screen confirms known coding and non-coding effectors and also associates many new lncRNAs to relevant pathways. The associations are often highly reproducible across cancer types, and while many lncRNAs are co-expressed with their protein-coding hosts or neighbours, some are intergenic and independent. We highlight lncRNAs with possible functions downstream of the tumour suppressor TP53 and the master antioxidant transcription factor NFE2L2. Our study provides a comprehensive overview of lncRNA transcriptional alterations in relation to key driver mutational events in human cancers.


PLOS ONE | 2013

Zfp148 Deficiency Causes Lung Maturation Defects and Lethality in Newborn Mice That Are Rescued by Deletion of p53 or Antioxidant Treatment

Volkan I. Sayin; Anna Nilton; Mohamed X. Ibrahim; Pia Ågren; Erik Larsson; Marleen M. Petit; Lillemor Mattsson Hultén; Marcus Ståhlman; Bengt R. Johansson; Martin O. Bergo; Per Lindahl

The transcription factor Zfp148 (Zbp-89, BFCOL, BERF1, htβ) interacts physically with the tumor suppressor p53 and is implicated in cell cycle control, but the physiological role of Zfp148 remains unknown. Here we show that Zfp148 deficiency leads to respiratory distress and lethality in newborn mice. Zfp148 deficiency prevented structural maturation of the prenatal lung without affecting type II cell differentiation or surfactant production. BrdU analyses revealed that Zfp148 deficiency caused proliferation arrest of pulmonary cells at E18.5–19.5. Similarly, Zfp148-deficient fibroblasts exhibited proliferative arrest that was dependent on p53, raising the possibility that cell stress is part of the underlying mechanism. Indeed, Zfp148 deficiency lowered the threshold for activation of p53 under oxidative conditions. Moreover, both in vivo and cellular phenotypes were rescued on Trp53 +/− or Trp53 −/− backgrounds and by antioxidant treatment. Thus, Zfp148 prevents respiratory distress and lethality in newborn mice by attenuating oxidative stress–dependent p53-activity during the saccular stage of lung development. Our results establish Zfp148 as a novel player in mammalian lung maturation and demonstrate that Zfp148 is critical for cell cycle progression in vivo.


Oncotarget | 2016

Targeting Zfp148 activates p53 and reduces tumor initiation in the gut.

Anna Nilton; Volkan I. Sayin; Zhiyuan V. Zou; Sama I. Sayin; Cecilia Bondjers; Nadia Gul; Pia Ågren; Per Fogelstrand; Ola Nilsson; Martin O. Bergo; Per Lindahl

The transcription factor Zinc finger protein 148 (Zfp148, ZBP-89, BFCOL, BERF1, htβ) interacts physically with the tumor suppressor p53, but the significance of this interaction is not known. We recently showed that knockout of Zfp148 in mice leads to ectopic activation of p53 in some tissues and cultured fibroblasts, suggesting that Zfp148 represses p53 activity. Here we hypothesize that targeting Zfp148 would unleash p53 activity and protect against cancer development, and test this idea in the APCMin/+ mouse model of intestinal adenomas. Loss of one copy of Zfp148 markedly reduced tumor numbers and tumor-associated intestinal bleedings, and improved survival. Furthermore, after activation of β-catenin-the initiating event in colorectal cancer-Zfp148 deficiency activated p53 and induced apoptosis in intestinal explants of APCMin/+ mice. The anti-tumor effect of targeting Zfp148 depended on p53, as Zfp148 deficiency did not affect tumor numbers in APCMin/+ mice lacking one or both copies of Trp53. The results suggest that Zfp148 controls the fate of newly transformed intestinal tumor cells by repressing p53 and that targeting Zfp148 might be useful in the treatment of colorectal cancer.


PLOS ONE | 2013

Zinc finger protein 148 is dispensable for primitive and definitive hematopoiesis in mice.

Anna Nilton; Volkan I. Sayin; Anna Staffas; Erik Larsson; Julia Rolf; Marleen M. Petit; Lars Palmqvist; Birgitta Swolin; Susanna Cardell; Per Lindahl

Hematopoiesis is regulated by transcription factors that induce cell fate and differentiation in hematopoietic stem cells into fully differentiated hematopoietic cell types. The transcription factor zinc finger protein 148 (Zfp148) interacts with the hematopoietic transcription factor Gata1 and has been implicated to play an important role in primitive and definitive hematopoiesis in zebra fish and mouse chimeras. We have recently created a gene-trap knockout mouse model deficient for Zfp148, opening up for analyses of hematopoiesis in a conventional loss-of-function model in vivo. Here, we show that Zfp148-deficient neonatal and adult mice have normal or slightly increased levels of hemoglobin, hematocrit, platelets and white blood cells, compared to wild type controls. Hematopoietic lineages in bone marrow, thymus and spleen from Zfp148 gt/gt mice were further investigated by flow cytometry. There were no differences in T-cells (CD4 and CD8 single positive cells, CD4 and CD8 double negative/positive cells) in either organ. However, the fraction of CD69- and B220-positive cells among lymphocytes in spleen was slightly lower at postnatal day 14 in Zfp148 gt/gt mice compared to wild type mice. Our results demonstrate that Zfp148-deficient mice generate normal mature hematopoietic populations thus challenging earlier studies indicating that Zfp148 plays a critical role during hematopoietic development.

Collaboration


Dive into the Volkan I. Sayin's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Per Lindahl

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Nilton

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar

Erik Larsson

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pia Ågren

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Staffas

University of Gothenburg

View shared research outputs
Researchain Logo
Decentralizing Knowledge