• No results found

Impact of SETD2 mutation in the

N/A
N/A
Protected

Academic year: 2021

Share "Impact of SETD2 mutation in the "

Copied!
16
0
0

Loading.... (view fulltext now)

Full text

(1)

Impact of SETD2 mutation in the

regulation of Autophagy in Clear Cell Renal Cell Carcinoma Cells

Hanzhao Zhang

__________________________________________

Master Degree Project in Medical Research, 30 credits. Spring 2018

Department: Department of Environmental Medicine, Karolinska Institutet Supervisor: Bertrand Joseph, Patricia González-Rodríguez

(2)

Abstract

Background: Autophagy is an important cellular pathway in cell growth and maintenance, during which unnecessary organelles and materials in cells can be degraded. Regulation of autophagy is related to tumorigenesis. SETD2 is a histone methyltransferase and mutation of SETD2 has been identified in different human cancers, especially in clear cell renal cell carcinoma. Previous results of the laboratory showed evidence of SETD2 involvement in autophagy regulation. Therefore, the focus of this report is on autophagy regulation of SETD2 in clear cell renal cell carcinoma.

Aim: To investigate the impact of SETD2 mutation in autophagy pathway in clear cell renal cell carcinoma cells

Methods: Cells were cultured in standard procedure in optimal medium. Immunoblotting was used to examine protein expression level. RT-qPCR was used to determine mRNA expression level of certain gene. Localization of protein expression was detected by immunofluorescence.

Results: SETD2 deficient cell lines showed less accumulation of p62 and LC3B when blocking the fusion of autophagosome and lysosome. In SETD2 deficient cell lines, an extra band and higher signal of free ATG12 were found when blotted with ATG12-ATG5. Expression of alternative splicing marker was also lower when SETD2 was mutated.

Conclusion: SETD2 might be related to a lower autophagic flux and might be involved in autophagosome formation by affecting formation of ATG12-ATG5-ATG16L complex. SETD2 deficiency might also decrease alternative splicing. Further investigations are required to give solid statement.

Key words: Autophagy, SETD2, ccRCC, ATG12-ATG5 complex

(3)

Introduction

Autophagy is a cellular pathway involved in cell growth and development, which helps maintain the balance between synthesis and degradation of proteins and organelles. Autophagy also plays a role in embryogenesis, cell differentiation, cellular death and lifespan extension (Tsukamoto et al., 2008;

Yorimitsu and Klionsky, 2005). Previous research has also reported the function of autophagy in many diseases. It is possible that autophagy can be protective in the progression of cancer and neurodegenerative disease such as Alzheimer’s disease (Cuervo, 2004; Yorimitsu and Klionsky, 2005).

Moreover, autophagy is upregulated under multiple stress conditions such as hypoxia, starvation, pathogen infection, etc. to help cells adapt and survive those unfavorable conditions (Tsukada and Ohsumi, 1993). During autophagy, cells degrade unnecessary proteins and organelles and recycle the components (Klionsky and Emr, 2000). There are three major forms of autophagy: macroautophagy, chaperone-mediated autophagy and microautophagy. Macroautophagy is the most prevalent form and most studied. During macroautophagy, autophagosome, a double-membrane structure will form from the sequestration of a region of cytoplasm and will wrap cytosolic components such as protein aggregates, damage organelles and intracellular bacteria. The autophagosome will fuse with a lysosome, which introduces acid hydrolases to the autophagosome that forms a mature structure called autolysosome (Dunn, 1990a). The degraded cargo in the autolysosome will then be recycled and reused for different cellular purposes (Dunn, 1990b).

In studies about autophagy, two widely used autophagy markers are LC3B and p62/SQSTM1 (p62 hereafter). LC3 is a light chain subunit of microtubule-associated proteins (MAPs) (Mann and Hammarback, 1994). Due to different post-transcriptional modification, two forms of LC3B are included in autophagosome formation, LC3-I and LC3-II. The cytosolic, LC3-I, binds to phosphatidylethanolamine (PE) to form LC3-II through lipidation, which is located on the autophagosome membrane The process includes two ubiquitin-like reactions, under catalysis of the E1- like enzyme, ATG7 and then E2-like enzyme, ATG3. LC3-II then is recruited on autophagosome membrane and later degraded when autophagosomes fuse with lysosomes (Tanida et al., 2008).

Therefore, LC3B has been used as an autophagy marker and LC3BII has been used to detect autophagosome formation. p62 is a polyubiquitin-binding protein which is involved in autophagy pathway via LC3. A direct interaction of p62 and LC3B has been found on autophagosomal membrane (Bjørkøy et al., 2005) and a depletion of p62 results in less recruitment of LC3B on autophagosomal membrane (Pankiv et al., 2007). Since p62 bodies are degraded by autophagy via interaction with LC3,

(4)

analysis of both are used to detect autophagic flux (Bjørkøy et al., 2005) and the co-localization of LC3B and p62 is a reliable measure to study autophagosome formation (Jiang and Mizushima, 2015).

In autophagy pathway, autophagy-related (ATG) proteins play an important role in the regulation in different steps, from the autophagosome formation to the fusion with lysosome(He and Klionsky, 2009).

For instance, one essential conjugation system of ATG proteins in autophagosome formation is the conjugation of ATG12-ATG5-ATG16L, which is a ubiquitin-like conjugation system (Mizushima et al., 1998). In this system, the C-terminal carboxyl group of ATG12 is activated by ATG7, the E1 enzyme and then transferred to ATG10, the E2 enzyme. After that, activated ATG12 bind to the lysine residue of ATG5 via an isopeptide bond. Later on, the complex also binds to ATG16L in mammalian cells (Nakatogawa, 2013). ATG12-ATG5 is also found functional in LC3 lipidation on autophagosomal membrane (Otomo et al., 2013). In fact, previous research showed failure in ATG12-ATG5-ATG16L complex and autophagosome formation when cells are mutant in ATG7 and ATG10 (Mizushima et al., 1998).

Although ATG proteins are known essential for autophagy, further knowledge is required in order to understand in the transcriptional control of ATG genes. Previous studies of our laboratory were focused on searching for transcriptional regulators in autophagy. By screening of over 150 mutants in single transcription factors and analysis on some ATG proteins expression level, a histone demethylase protein, Rph1/KDM4 was found to be an autophagy repressor (Bernard et al., 2015). Based on this finding, the research was expanded to the impact of SETD2, a histone methyltransferase, on the autophagy pathway.

SETD2 is the H3 lysine 36 methyltransferase, which uses dimethylated H3K36 (H3K36me2) as substrate to synthesize trimethylated H3K36 (H3K36me3) (de Almeida et al., 2011; Sun et al., 2005). To note, SETD2 is the exclusive enzyme for H3K36 trimethylation. SETD2 mutations are identified in different human cancers according to the analysis of cancer genomics, which suggests the tumor suppression function of SETD2 (Gao et al., 2013). The first reported SETD2 mutation is in cases of clear cell renal cell carcinoma (ccRCC), which is the most common and aggressive subtype of kidney tumors (Dalgliesh et al., 2010). SETD2 mutation was also reported in ccRCC cell lines (Duns et al., 2012). SETD2 mutation was also reported in ccRCC cell lines (Morris and Latif, 2017). Therefore, SETD2 mutation in ccRCC can affect genome stability and the expression of many functional genes. To note, the most common mutation in ccRCC is inactivation of Von Hippel Lindau (VHL) tumor suppressor, which is related to chromosome 3p loss and found in ~80% sporadic cases of ccRCC (Gnarra et al., 1994; Zbar et al., 1987). Inactivation of VHL leads to activation of hypoxia inducible factors (Shen and Kaelin), resulting in hypoxia response that affects cell metabolism(Shen and Kaelin, 2013). Therefore, impact of SETD2 on autophagy in ccRCC is studied with or without VHL mutation in this paper.

(5)

Due to the important role of SETD2 in histone modification, studies have been done on the relation between SETD2 and alternative splicing. A genome-wide analysis of human cells lines showed cells with intron truncation gave lower signal of H3K36me3 marking (de Almeida et al., 2011). Impact on alternative splicing is usually a global transcriptional regulator. Recent studies have revealed that splicing can affect essential genes in autophagy (Paronetto et al., 2016).

Based on the possible impact of SETD2 on autophagy and the link between SETD2 and ccRCC, the research in this report is focused on the impact of SETD2 mutation on regulation of autophagy in ccRCC.

Aim

Ÿ Investigate the impact of SETD2 on autophagy regulation in ccRCC

Ÿ Investigate the relation of SETD2 and autophagosome formation

Ÿ Study the role of SETD2 in alternative splicing

Materials and Methods Cell culture and treatments

A panel of ccRCC cell lines were used: ACHN, Caki-1 (carrying a SETD2 mutation), Caki-2 (carrying a VHL mutation), A498 (carrying SETD2 and VHL mutations). All cells were cultured at 37°C, 5% CO2.

ACHN and A498 were cultured in Dulbecco's Modified Eagle Medium supplemented with 10% fetal bovine serum, 1% L-Glutamine and 1% penicillin/streptomycin. Caki-1 and Caki-2 were cultured in RPMI 1640 medium supplemented with 10% fetal bovine serum, 1% L-Glutamine and 1%

penicillin/streptomycin. Bafilomycin A1 (40µM) was used to treat cells.

Immunoblotting

250,000 ~ 300,000 cells were seeded in each well on 6 well plates and harvested with Laemmli buffer (5x) at the indicated timepoint. Cells were sonicated and boiled at 99oC for ~10 minutes. The whole cell lysates were subjected to SDS-polyacrylamide gel electrophoresis (PAGE) and blotted with specific primary antibodies diluted in 5% bovine serum albumin with 0.1% sodium azide. Secondary antibodies with fluorescence were incubated and developed by Odyssey® CLx Imaging Syetem. Quantification was done using Image J. Primary antibodies used in the research were: anti-SETD2 (rabbit, Abcam, ab69836), anti-H3K36me3 (rabbit, Cell Signaling, 9763), anti-LC3B (rabbit, Sigma, L7543), anti-p62 (rabbit, Cell Signaling, 5114), anti-ATG7 (rabbit, GeneTex, GTX61647), anti-ATG12 (rabbit, Abcam, ab155589), anti-SF3B3 (rabbit, Abcam, ab86992), anti-Actin (mouse, Sigma, A3853). Secondary antibodies used in the research were: IRDye® 680RD Goat anti-Rabbit IgG, IRDye® 800CW Goat anti-Mouse IgG.

(6)

RT-qPCR

RNA was extracted using the RNeasy Kit (Qiagen). After determining RNA concentration with Nanodrop, in total 1µg RNA was used for first strand synthesis with SuperScript IV Reverse Transcriptase (Invitrogen) to synthesize cDNA. QPCR was performed on StepOnePlus™ Real-Time PCR System (Applied Biosystems).

Immunofluorescence

For immunofluorescence, cells were cultured on coverslips. Cells were fixed in cold methanol or 4%

paraformaldehyde for 15min at room temperature. After blocking with Duolink® Blocking Solution, primary antibodies were incubated at 4oC overnight. The day after, secondary antibodies were incubated in room temperature for 1 hour. The slides were washed 3 times with PBS. Mounting media with DAPI (Duolink) was used to mount the slides. The slides were analyzed under confocal microscope. Primary antibodies used: p62 (mouse, Abcam, ab56416) and LC3B (rabbit, Sigma, L7543). Secondary antibodies used: Alexa Fluor 594 (goat anti mouse, Molecular Probes, A11005) and Alexa Fluor 488 (goat anti rabbit, Molecular Probes, A11008).

Results

Characterization of renal clear carcinoma cell lines

As mentioned previously, cell lines used in this study (ACHN, Caki-1, Caki-2 and A498) were all ccRCC cell lines derived from human, either from tumor or metastatic site. In order to characterize the mentioned ccRCC cell lines, we examined SETD2, VHL and H3K36me3 expression level either by immunoblotting or RT-qPCR (Fig.1).

As expected, Caki-1 did not express SETD2 at protein level compared to ACHN and A498 (Fig.1a).

Since SETD2 is the sole protein responsible for H3K36 trimethylation, SETD2 deficiency can result in loss of the histone marker, H3K36me3. Therefore, H3K36me3 protein level was also examined to investigate SETD2 function. As shown in Fig.1b, a complete loss was found in Caki-1 and A498 cell lines, compared to ACHN. Although SETD2 remained intact in A498, loss of H3K36me3 could still suggest the non-functional SETD2. Non-functional SETD2 was also found in previous research when characterizing A498 ccRCC cell lines (Pfister et al., 2015). RT-qPCR was used to examine VHL gene expression level (Fig.1c). Compared to ACHN, VHL expression was much lower in Caki-2 and A498.

Therefore, all cell lines used in the research were carrying corresponding mutations.

(7)

Baseline autophagy in ccRCC cell lines with SETD2 mutation

After characterization of mutations in cell lines, baseline level of autophagy was studied by the protein level of LC3 and p62. As mentioned previously, LC3 is involved in autophagosome formation. Cytosolic LC3-I is conjugated to PE to form LC3-II, which is located on autophagosome membrane (Tanida et al., 2008). Since intra-autophagosomal LC3-II is degraded during the fusion of autophagosome and lysosome, LC3-II expression level can be a marker to reveal the dynamic of autophagy (Tanida et al., 2005). In other words, LC3-II is a marker for autophagic activity. Therefore, we analyzed LC3 expression level in ccRCC cell lines which carries mutations in SETD2 and VHL. By immunoblotting, both LC3-I and LC3-II can be detected using antibodies for LC3.

p62 is found to directly interact with LC3 on autophagosome membrane and degraded via LC3 pathway(Bjørkøy et al., 2005). Although the role of p62 in autophagy is not clear, the interaction of p62 and LC3 are widely used as a marker for autophagy flux, which is more related to degradation rate after fusion of autophagosome and lysosome (Bjørkøy et al., 2005). Moreover, co-localization of p62 and LC3 works as a marker for autophagosome formation since the interaction locates the formation of autophagosome membrane (Jiang and Mizushima, 2015).

Fig.1

a-b. SETD2 and H3K36me3 protein level were examined by immunobloting in ACHN, Caki-1 and A498 cell lines.

c. VHL gene expression level was examined by RT-qPCR.

(ACHN: ccRCC cells; Caki-1: SETD2-/- ccRCC cells; Caki-2: VHL-/- ccRCC cells; A498: SETD2-/- VHL-/- ccRCC cells)

(8)

During autophagy process, the degradation and recycle start after the fusion of autophagosome and lysosome. The level of autophagy depends on the amount of autophagosomes and the degradation level of autolysosome, which have to be taken into consideration when studying autophagy. Bafilomycin A1 (Baf A1) is a specific inhibitor of vacuolar H+ ATPase and is found to block the fusion of autophagosome and lysosome (Yamamoto et al., 1998). By treatment with Baf A1, the total amount of autophagosome can be detected.

As shown in Fig.2a, the overall LC3 expression level was lower in Caki-1 and A498, compared to ACHN (Ratio LC3/Actin). However, the expression of LC3-II, which was more relevant to autophagosome formation, was the opposite. Caki-1 and A498 showed higher expression in LC3-II (Ratio LC3-II/Actin). In order to investigate whether the increase in LC3-II revealed higher autophagic activity, Baf A1 treatment showed the total amount of autophagosome. From Ratio LC3-II accumulation, although Caki-1 and A498 had higher expression in LC3BII, the amount of total accumulation of LC3BII was not higher compared to ACHN. This might indicate that it was the degradation rate instead of total autophagic activity that resulted in the change of LC3-II expression level in Caki-1 and A498.

In p62 blotting (Fig.2b), We found that at baseline levels the expression of p62 in Caki-1 was similar to that in ACHN cells, while only ccRCC cell lines that carry VHL mutations showed a clear increase in p62 level in Caki-2 and A498 (Ratio p62/Actin). When treated with Baf A1, there was accumulation of p62 in all cell lines but in Caki-2, the accumulation was not as significant as the other three. Caki-1 and A498 also showed less accumulation than ACHN (Ratio p62 Baf A1), which meant the degraded p62 that was restored by Baf A1 treatment was less. It might indicate a lower autophagic flux.

Preliminary data in the laboratory showed higher expression level of LC3B in Caki-1 by RT-qPCR (Fig.2c), which is different from immunoblotting results. In order to examine whether the difference was due to different level of autophagic activity, a more reliable detection of autophagosome formation, the co-localization of p62 and LC3, was performed by immunofluorescence (Fig.2d). There was difference in the pattern of co-localization in Caki-1, Caki-2 and A498, compared to ACHN. For instance, although higher expression of LC3-II was detected in Caki-1 by immunoblotting, the co-localization was not significantly higher, indicating that LC3-II alone might not well reveal the autophagic activity. However, without further investigation, no more information can be obtained.

(9)

0 0.2 0.4 0.6 0.8 1 1.2 1.4

ACHN CAKI1 A498

MAP1LC3B

a

b

c

(10)

Impact of SETD2 on ATG12-ATG5-ATG16L system

ATG12-ATG5-ATG16L conjugation system is a ubiquitin-like system which plays an important role in autophagosome formation. It is found to be involved in lipidation of LC3-I (Otomo et al., 2013). In order to investigate the impact of SETD2 on ATG12-ATG5-ATG16L system, the expression level of ATG12- ATG5 complex and free ATG12 was detected (Fig.3a). ATG12-ATG5 complex is very stable, making it possible to be detected by immunoblotting. The expression level of ATG12-ATG5 complex differed little among four cell lines. However, there was another heavier band appearing in ACHN, Caki-1 and A498 but not in Caki-2 (black arrow). Moreover, the signal of this band was significantly higher in Caki-1 and

ACHN

Caki-1

A498 Caki- 2

Fig.2

a. Immunoblotting and quantification of LC3 protein level.

b. Immunoblotting and quantification of p62 protein level.

c. Preliminary RT-qPCR data of LC3B

d. Immunofluorescence results demonstrated the co-localization of LC3B and p62.

(ACHN: ccRCC cells; Caki-1: SETD2-/- ccRCC cells; Caki-2: VHL-/- ccRCC cells; A498: SETD2-/- VHL-/- ccRCC cells)

(11)

A498, which are SETD2 deficient cell lines, than ACHN. Also, signal of free ATG12 was only detected in SETD2 deficient cell lines, Caki-1 and A498, not in ACHN or Caki-2. When treated with Baf A1, the expression level of ATG12-ATG5 and free ATG12 changed little, which indicated the accumulation was very low or even no accumulation. Then the E1 enzyme in this conjugation system, ATG7, was examined by immunoblotting (Fig.3b). However, no significant difference of ATG7 expression level was found.

Discussion

During the research, we found that Caki-2 (VHL-/-) cells did not react to Baf A1 as Caki-1 (SETD2-/-) and A498 (VHL-/-, SETD2-/-). When treated with Baf A1, no accumulation or very little accumulation could be found (Fig.2a, Fig.2b). This indicates that it is possible that in Caki-2, the level of

Fig.3

a. The amount of ATG12-ATG5 protein complex was similar among four cell lines. However, an extra band appeared in ACHN, Caki-1 and A498 (black arrow). Signal of this band was higher in Caki-1 and A498, the SETD2 deficient cell lines.

Free ATG12 was detected in Caki-1 and A498, not in ACHN or Caki-2. The signal was too low compared to that in Caki-1 and A498. When treated with Baf A1, neither ATG12-ATG5 nor free ATG12 showed accumulation.

b. The expression of ATG7 was detected by immunoblotting. However, the change in ATG7 among different cell lines was little.

(ACHN: ccRCC cells; Caki-1: SETD2-/- ccRCC cells; Caki-2: VHL-/- ccRCC cells; A498: SETD2-/- VHL-/- ccRCC cells) a

b

(12)

autophagosome formation has reached the limit under normal condition, making it unable to accumulate more when blocking the fusion. It has been reported previously that loss of VHL affects regulation of the entire network and promotes LC3-dependent oncogenic autophagy. This is mainly due to loss of a tumor suppressing microRNA, miR-204 (Cost and Czyzyk-Krzeska, 2015). Although Caki-2 was found to have lower expression of LC3BII than Caki-1 and A498 by immunoblotting (Fig.2b), the protein level alone was not potent enough to reveal autophagosome formation. It is possible that only a small portion of LC3BII in Caki-1 and A498 are actually involved in autophagosome. To validate this, further investigation of co-localization of p62 and LC3B by immunofluorescence is needed.

Although the co-localization of p62 and LC3B gives more reliable information of autophagosome formation, the protein expression level can give us a picture of autophagy level and autophagic flux.

LC3BII had higher protein expression in Caki-1 and A498, SETD2 deficient cell lines, compared to ACHN. However, when blocking with Baf A1, the accumulation of LC3BII was not higher in Caki-1 and A498 (Fig.2b). Possible explanation is that it can be either not all LC3BII detected in immunoblotting were used in autophagosome formation or the degradation rate after fusion was lower in Caki-1 and A498. It is also possible to be the combination of two explanations. p62 is used to detect autophagic flux since it is degraded via interaction with LC3B after fusion of autophagosome and lysosome. However, it has been reported previously that p62 targeting on autophagosomal membrane depends not on LC3 binding but self-oligomerization instead. It is also reported p62 co-localized with other proteins like NBR1 on autophagosome formation site in cytoplasm in very early state of autophagosome formation, while LC3 is more important in later closure of autophagosome (Itakura and Mizushima, 2011).

Therefore, p62 protein expression level by immunoblotting may not well reveal the degradation via LC3 interaction. When treated with Baf A1, however, the accumulation level can give some information of autophagic flux. The accumulation can indicate proteins that are degraded through fusion of autophagosome and lysosome. Less accumulation of p62 in Caki-1 (Fig.2b Ratio p62 accumulation) compared to ACHN indicates fewer proteins degraded through the fusion, which might imply a lower rate in degradation. Therefore, loss of SETD2 can be related in a lower rate in autophagic flux.

Analysis of co-localization of p62 and LC3B by immunofluorescence gives information about autophagosome formation. Some difference in expression pattern can be observed in Fig.2d. For instance, the co-localization (orange dots) seems more in Caki-2. In Caki-1, although LC3B expression seems higher, the co-localization is less compared to ACHN. However, before further investigation of results, no solid conclusion can be drawn.

Here an essential conjugation system in autophagosome formation was studied, ATG12-ATG5-ATG16L.

As mentioned above, formation of ATG12-ATG5-ATG16L requires two ubiquitin-like reactions,

(13)

catalyzed by the E1-like enzyme ATG7 and the E2-like enzyme ATG10. Protein expression level of ATG12-ATG5 complex, free ATG12 and ATG7 was examined (Fig.3). No accumulation was detected with Baf A1 treatment. This is more likely due to errors in operation or the reagent was not working well.

When comparing baseline level of ATG12-ATG5 in each cell line, no significant difference was found.

Since ATG12-ATG5 is not the only ATG complex involved in autophagosome formation, whether the detected expression of ATG12-ATG5 can be linked to autophagosome formation requires further studies into complex like ATG8-PE. ATG8-PE is another ubiquitin-like conjugation system involved in autophagosome formation and is closely related to ATG12-ATG5-ATG16L. ATG8 shares the E1-like enzyme ATG7 with ATG12 and then is transferred to the amino group of PE with activation by the E-2 like enzyme ATG3. Moreover, ATG12-ATG5 works as E3-like enzyme for ATG8-PE conjugation system, which increases ATG3 activity and helps localize ATG8-PE to form autophagosome membrane (Nakatogawa, 2013). Since the expression of ATG7 also showed little change among cell lines, expression of other involved ATG proteins will be helpful to better interpret the results.

An interesting finding was the extra band (Fig.3a black arrow) and significantly higher expression of free ATG12 in Caki-1 and A498. Although the extra band also appeared in ACHN, the signal was much higher in Caki-1 and A498. This might indicate that the extra band and free ATG12 expression are SETD2 dependent. Little explanation can be found in previous publications. It has been reported that SETD7, a histone methyltransferase that monomethylates lysine 4 of Histone 3, methylates lysine 151 on ATG16L and impairs the binding to ATG12-ATG5 to inhibit hypoxia/reoxygenation-induced autophagy in cardiomyocytes (Song et al., 2018). Since ATG16L interacts directly with ATG12-ATG5, it is worthwhile to examine ATG16L expression level for further experiments. Another possible explanation is that, as mentioned above, since previous publications showed results implicating SETD2 is related to alternative splicing, different isoforms of ATG12 or ATG5 can be formed due to change in splicing.

However, different isoforms might not be able to form ATG12-ATG5 complex or the complex they form is not functional. Higher signal of free ATG12 expression in SETD2 deficient cell lines can also be explained by different dysfunctional isoforms of ATG12. To conclude, the reason is still unclear and the hypotheses require further investigation in involved protein such as ATG16L and ATG5.

SETD2 is proposed to be related to alternative splicing as mentioned before. In this paper, SF3B3 (also named SAP130), a component of the splicing factor SF3B complex, was examined and a decrease was found in Caki-1 and A498, the SETD2 deficient cell lines. This results validates previous results that recruitment of methyltransferase SETD2 to H3K36me2 depends on alternative splicing (de Almeida et al., 2011). However, studies in ccRCC model systems gave opposite results in SETD2 and alternative splicing. By depletion of SETD2, no difference was found in exon usage or intron truncation, which were

(14)

SETD2-dependent alternative splicing events observed previously (Kanu et al., 2015). This contradiction might be due to the use of ccRCC model system. More studies of splicing markers are required to validate either view of SETD2 and alternative splicing.

To conclude, loss of SETD2 function might be related to a lower autophagic flux. SETD2 might also be involved in formation of ATG12-ATG5-ATG16L complex during autophagosome formation. Moreover, SETD2 is possible to affect alternative splicing as a global transcriptional regulator. However, results and discussion described in this report still require much further investigation. For further experiment, co- localization of LC3B and p62 will be studied. Other involved ATG proteins and complexes such as ATG5, ATG8-PE will be examined. Alternative splicing mechanism will also be further studied. Gain or loss of function of SETD2 will be studied to validate the function of SETD2. Protein expression level will be validated by RT-qPCR. Moreover, cells that induced autophagy will be used in future experiments.

Acknowledgements

I would like to give my thanks to my supervisor Bertrand Joseph and Patricia González-Rodríguez for the help and guidance in the work. Preliminary data from the laboratory was provided by Patricia González-Rodríguez.

References

Bernard, A., Jin, M., González-Rodríguez, P., Füllgrabe, J., Delorme-Axford, E., Backues, S.K., Joseph, B., and Klionsky, D.J. (2015). Rph1/KDM4 mediates nutrient-limitation signaling that leads to the transcriptional induction of autophagy. Curr Biol 25, 546-555.

Bjørkøy, G., Lamark, T., Brech, A., Outzen, H., Perander, M., Overvatn, A., Stenmark, H., and Johansen, T. (2005). p62/SQSTM1 forms protein aggregates degraded by autophagy and has a protective effect on huntingtin-induced cell death. J Cell Biol 171, 603-614.

Cost, N.G., and Czyzyk-Krzeska, M.F. (2015). Regulation of autophagy by two products of one gene:

TRPM3 and miR-204. Mol Cell Oncol 2, e1002712.

Dalgliesh, G.L., Furge, K., Greenman, C., Chen, L., Bignell, G., Butler, A., Davies, H., Edkins, S., Hardy, C., Latimer, C., et al. (2010). Systematic sequencing of renal carcinoma reveals inactivation of histone modifying genes. Nature 463, 360-363.

de Almeida, S.F., Grosso, A.R., Koch, F., Fenouil, R., Carvalho, S., Andrade, J., Levezinho, H., Gut, M., Eick, D., Gut, I., et al. (2011). Splicing enhances recruitment of methyltransferase HYPB/Setd2 and methylation of histone H3 Lys36. Nat Struct Mol Biol 18, 977-983.

(15)

Dunn, W.A. (1990a). Studies on the mechanisms of autophagy: formation of the autophagic vacuole. J Cell Biol 110, 1923-1933.

Dunn, W.A. (1990b). Studies on the mechanisms of autophagy: maturation of the autophagic vacuole. J Cell Biol 110, 1935-1945.

Duns, G., Hofstra, R.M., Sietzema, J.G., Hollema, H., van Duivenbode, I., Kuik, A., Giezen, C., Jan, O., Bergsma, J.J., Bijnen, H., et al. (2012). Targeted exome sequencing in clear cell renal cell carcinoma tumors suggests aberrant chromatin regulation as a crucial step in ccRCC development. Hum Mutat 33, 1059-1062.

Gao, J., Aksoy, B.A., Dogrusoz, U., Dresdner, G., Gross, B., Sumer, S.O., Sun, Y., Jacobsen, A., Sinha, R., Larsson, E., et al. (2013). Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 6, pl1.

Gnarra, J.R., Tory, K., Weng, Y., Schmidt, L., Wei, M.H., Li, H., Latif, F., Liu, S., Chen, F., and Duh, F.M. (1994). Mutations of the VHL tumour suppressor gene in renal carcinoma. Nat Genet 7, 85-90.

He, C., and Klionsky, D.J. (2009). Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 43, 67-93.

Itakura, E., and Mizushima, N. (2011). p62 Targeting to the autophagosome formation site requires self- oligomerization but not LC3 binding. J Cell Biol 192, 17-27.

Jiang, P., and Mizushima, N. (2015). LC3- and p62-based biochemical methods for the analysis of autophagy progression in mammalian cells. Methods 75, 13-18.

Kanu, N., Grönroos, E., Martinez, P., Burrell, R.A., Yi Goh, X., Bartkova, J., Maya-Mendoza, A., Mistrík, M., Rowan, A.J., Patel, H., et al. (2015). SETD2 loss-of-function promotes renal cancer branched evolution through replication stress and impaired DNA repair. Oncogene 34, 5699-5708.

Mann, S.S., and Hammarback, J.A. (1994). Molecular characterization of light chain 3. A microtubule binding subunit of MAP1A and MAP1B. J Biol Chem 269, 11492-11497.

Mizushima, N., Noda, T., Yoshimori, T., Tanaka, Y., Ishii, T., George, M.D., Klionsky, D.J., Ohsumi, M., and Ohsumi, Y. (1998). A protein conjugation system essential for autophagy. Nature 395, 395-398.

Morris, M.R., and Latif, F. (2017). The epigenetic landscape of renal cancer. Nat Rev Nephrol 13, 47-60.

Nakatogawa, H. (2013). Two ubiquitin-like conjugation systems that mediate membrane formation during autophagy. Essays Biochem 55, 39-50.

Otomo, C., Metlagel, Z., Takaesu, G., and Otomo, T. (2013). Structure of the human ATG12~ATG5 conjugate required for LC3 lipidation in autophagy. Nat Struct Mol Biol 20, 59-66.

(16)

Pankiv, S., Clausen, T.H., Lamark, T., Brech, A., Bruun, J.A., Outzen, H., Øvervatn, A., Bjørkøy, G., and Johansen, T. (2007). p62/SQSTM1 binds directly to ATG8/LC3 to facilitate degradation of ubiquitinated protein aggregates by autophagy. J Biol Chem 282, 24131-24145.

Paronetto, M.P., Passacantilli, I., and Sette, C. (2016). Alternative splicing and cell survival: from tissue homeostasis to disease. Cell Death Differ 23, 1919-1929.

Pfister, S.X., Markkanen, E., Jiang, Y., Sarkar, S., Woodcock, M., Orlando, G., Mavrommati, I., Pai, C.C., Zalmas, L.P., Drobnitzky, N., et al. (2015). Inhibiting WEE1 Selectively Kills Histone H3K36me3- Deficient Cancers by dNTP Starvation. Cancer Cell 28, 557-568.

Shen, C., and Kaelin, W.G. (2013). The VHL/HIF axis in clear cell renal carcinoma. Semin Cancer Biol 23, 18-25.

Song, H., Feng, X., Zhang, M., Jin, X., Xu, X., Wang, L., Ding, X., Luo, Y., Lin, F., Wu, Q., et al.

(2018). Crosstalk between lysine methylation and phosphorylation of ATG16L1 dictates the apoptosis of hypoxia/reoxygenation-induced cardiomyocytes. Autophagy, 1-20.

Sun, X.J., Wei, J., Wu, X.Y., Hu, M., Wang, L., Wang, H.H., Zhang, Q.H., Chen, S.J., Huang, Q.H., and Chen, Z. (2005). Identification and characterization of a novel human histone H3 lysine 36-specific methyltransferase. J Biol Chem 280, 35261-35271.

Tanida, I., Minematsu-Ikeguchi, N., Ueno, T., and Kominami, E. (2005). Lysosomal turnover, but not a cellular level, of endogenous LC3 is a marker for autophagy. Autophagy 1, 84-91.

Tanida, I., Ueno, T., and Kominami, E. (2008). LC3 and Autophagy. Methods Mol Biol 445, 77-88.

Tsukada, M., and Ohsumi, Y. (1993). Isolation and characterization of autophagy-defective mutants of Saccharomyces cerevisiae. FEBS Lett 333, 169-174.

Yamamoto, A., Tagawa, Y., Yoshimori, T., Moriyama, Y., Masaki, R., and Tashiro, Y. (1998).

Bafilomycin A1 prevents maturation of autophagic vacuoles by inhibiting fusion between

autophagosomes and lysosomes in rat hepatoma cell line, H-4-II-E cells. Cell Struct Funct 23, 33-42.

Zbar, B., Brauch, H., Talmadge, C., and Linehan, M. (1987). Loss of alleles of loci on the short arm of chromosome 3 in renal cell carcinoma. Nature 327, 721-724.

References

Related documents

According to Jonsson (2005), companies that apply a carrying charge in inventory management ought to investigate how it is determined in order achieve a reasonable size. Closely

In: Redaktionskommitté Anders Brändström, Sören Edvinsson, Tom Ericsson och Peter Sköld (ed.), Befolkningshistoriska perspektiv: festskrift till Lars-Göran Tedebrand

The big difference is the latter are sport inspired slick fashion design, whilst the Fraction collection is expressive sport wear design.. The collection has developed quite

Paper III Single molecule analysis of the hypermutable tetranucleotide locus D21S1245 through sperm genotyping: a heterogeneous pattern of mutation but no clear male age effect In

I en utredning som lades fram 1983 fastställdes det att staten därför har: ”förpliktelser att visa respekt för samisk kultur och samiska näringar och ge utrymme

In the spinal cord from rabbits heterozygous for the sauteur allele (+/s am ), the number of neurons expressing RORB was approximately 25% lower than in the wild-type animals (Fig

This book is the first to bring together a broad range of policy-relevant per- spectives on negative emissions and, in particular, on bioenergy with carbon capture and storage:

Married women in Uganda is a group that is stroked both by the crisis in social reproduction and the unfavorable conditions on the labor market, and this study will contribute