Network


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

Hotspot


Dive into the research topics where Sanna Kaikkonen is active.

Publication


Featured researches published by Sanna Kaikkonen.


Cellular and Molecular Life Sciences | 2009

PIAS proteins: pleiotropic interactors associated with SUMO.

Miia M. Rytinki; Sanna Kaikkonen; Petri Pehkonen; Tiina Jääskeläinen; Jorma J. Palvimo

The interactions and functions of protein inhibitors of activated STAT (PIAS) proteins are not restricted to the signal transducers and activators of transcription (STATs), but PIAS1, -2, -3 and -4 interact with and regulate a variety of distinct proteins, especially transcription factors. Although the majority of PIAS-interacting proteins are prone to modification by small ubiquitin-related modifier (SUMO) proteins and the PIAS proteins have the capacity to promote the modification as RING-type SUMO ligases, they do not function solely as SUMO E3 ligases. Instead, their effects are often independent of their Siz/PIAS (SP)-RING finger, but dependent on their capability to noncovalently interact with SUMOs or DNA through their SUMO-interacting motif and scaffold attachment factor-A/B, acinus and PIAS domain, respectively. Here, we present an overview of the cellular regulation by PIAS proteins and propose that many of their functions are due to their capability to mediate and facilitate SUMO-linked protein assemblies.


Molecular Endocrinology | 2009

SUMO-Specific Protease 1 (SENP1) Reverses the Hormone-Augmented SUMOylation of Androgen Receptor and Modulates Gene Responses in Prostate Cancer Cells

Sanna Kaikkonen; Tiina Jääskeläinen; Ulla Karvonen; Miia M. Rytinki; Harri Makkonen; Daniel Gioeli; Bryce M. Paschal; Jorma J. Palvimo

The acceptor sites for small ubiquitin-like modifier (SUMO) are conserved in the N-terminal domains of several nuclear receptors. Here, we show that androgens induce rapid and dynamic conjugation of SUMO-1 to androgen receptor (AR). Nuclear import of AR is not sufficient for SUMOylation, because constitutively nuclear apo-ARs or antagonist-bound ARs are only very weakly modified by SUMO-1 in comparison with agonist-bound ARs. Of the SUMO-specific proteases (SENP)-1, -2, -3, -5, and -6, only SENP1 and SENP2 are efficient in cleaving AR-SUMO-1 conjugates in intact cells and in vitro. Both SENP1 and -2 are nuclear and found at sites proximal to AR. Their expression promotes AR-dependent transcription, but in a promoter-selective fashion. SENP1 and -2 stimulated the activity of holo-AR on compound androgen response element-containing promoters. The effects of SENP1 and -2 on AR-dependent transcription were dependent on catalytic activity and required intact SUMO acceptor sites in AR, indicating that their coactivating effects are mainly due to their direct isopeptidase activity on holo-AR. In prostate cancer cells, ectopic expression of SENP1, but not that of SENP2, increased the transcription activity of endogenous AR. Silencing of SENP1 attenuated the expression of several AR target genes and blunted androgen-stimulated growth of LNCaP cells. Our results indicate that SENP1 reverses the ligand-induced SUMOylation of AR and helps fine tune the cellular responses to androgens in a target promoter-selective manner.


Nucleic Acids Research | 2014

SUMOylation regulates the chromatin occupancy and anti-proliferative gene programs of glucocorticoid receptor

Ville Paakinaho; Sanna Kaikkonen; Harri Makkonen; Vladimir Benes; Jorma J. Palvimo

In addition to the glucocorticoids, the glucocorticoid receptor (GR) is regulated by post-translational modifications, including SUMOylation. We have analyzed how SUMOylation influences the activity of endogenous GR target genes and the receptor chromatin binding by using isogenic HEK293 cells expressing wild-type GR (wtGR) or SUMOylation-defective GR (GR3KR). Gene expression profiling revealed that both dexamethasone up- and downregulated genes are affected by the GR SUMOylation and that the affected genes are significantly associated with pathways of cellular proliferation and survival. The GR3KR-expressing cells proliferated more rapidly, and their anti-proliferative response to dexamethasone was less pronounced than in the wtGR-expressing cells. ChIP-seq analyses indicated that the SUMOylation modulates the chromatin occupancy of GR on several loci associated with cellular growth in a fashion that parallels with their differential dexamethasone-regulated expression between the two cell lines. Moreover, chromatin SUMO-2/3 marks, which were associated with active GR-binding sites, showed markedly higher overlap with the wtGR cistrome than with the GR3KR cistrome. In sum, our results indicate that the SUMOylation does not simply repress the GR activity, but regulates the activity of the receptor in a target locus selective fashion, playing an important role in controlling the GR activity on genes influencing cell growth.


Molecular and Cellular Biology | 2012

Dynamic SUMOylation Is Linked to the Activity Cycles of Androgen Receptor in the Cell Nucleus

Miia Rytinki; Sanna Kaikkonen; Päivi Sutinen; Ville Paakinaho; Vesa Rahkama; Jorma J. Palvimo

ABSTRACT Despite of the progress in the molecular etiology of prostate cancer, the androgen receptor (AR) remains the major druggable target for the advanced disease. In addition to hormonal ligands, AR activity is regulated by posttranslational modifications. Here, we show that androgen induces SUMO-2 and SUMO-3 (SUMO-2/3) modification (SUMOylation) of the endogenous AR in prostate cancer cells, which is also reflected in the chromatin-bound receptor. Although only a small percentage of AR is SUMOylated at the steady state, AR SUMOylation sites have an impact on the receptors stability, intranuclear mobility, and chromatin interactions and on expression of its target genes. Interestingly, short-term proteotoxic and cell stress, such as hyperthermia, that detaches the AR from the chromatin triggers accumulation of the SUMO-2/3-modified AR pool which concentrates into the nuclear matrix compartment. Alleviation of the stress allows rapid reversal of the SUMO-2/3 modifications and the AR to return to the chromatin. In sum, these results suggest that the androgen-induced SUMOylation is linked to the activity cycles of the holo-AR in the nucleus and chromatin binding, whereas the stress-induced SUMO-2/3 modifications sustain the solubility of the AR and protect it from proteotoxic insults in the nucleus.


Journal of Molecular Biology | 2008

ZNF451 is a novel PML body- and SUMO-associated transcriptional coregulator.

Ulla Karvonen; Tiina Jääskeläinen; Miia M. Rytinki; Sanna Kaikkonen; Jorma J. Palvimo

Covalent modification by small ubiquitin-related modifiers (SUMOs) is an important means to regulate dynamic residency of transcription factors within nuclear compartments. Here, we identify a multi-C(2)H(2)-type zinc finger protein (ZNF), ZNF451, as a novel nuclear protein that can be associated with promyelocytic leukemia bodies. In keeping with its interaction with SUMO E2 conjugase Ubc9 and SUMOs, ZNF451 is covalently modified by SUMOs (sumoylated) at several, albeit nonconsensus, sites. Interestingly, noncovalent SUMO-binding activity of ZNF451 (SUMO-interacting motif) is also important for its sumoylation. SUMO modifications regulate the nuclear compartmentalization of ZNF451, since coexpression of ZNF451 with SUMO-specific proteases SENP1 or SENP2, both capable of desumoylating the protein, redistributes ZNF451 from nuclear domains to speckles and nucleoplasm. Interaction of ZNF451 with PIAS1 (protein inhibitor of activated STAT 1) is not manifested as PIAS1s E3 SUMO ligase activity towards ZNF451 but results in disintegration of ZNF451 nuclear domains and recruitment of ZNF451 to androgen receptor (AR) speckles. ZNF451 interacts weakly, but in a SUMO-1-enhanced fashion, with AR. ZNF451 does not harbor an intrinsic transcription activation function, but interestingly, ablation of endogenous ZNF451 in prostate cancer cells significantly decreases expression of several AR target genes. Thus, we suggest that ZNF451 exerts its effects via SUMO modification machinery and trafficking of transcription regulators between promyelocytic leukemia bodies and nucleoplasm.


Nucleic Acids Research | 2015

SUMO ligase PIAS1 functions as a target gene selective androgen receptor coregulator on prostate cancer cell chromatin

Sari Toropainen; Marjo Malinen; Sanna Kaikkonen; Miia Rytinki; Tiina Jääskeläinen; Biswajyoti Sahu; Olli A. Jänne; Jorma J. Palvimo

Androgen receptor (AR) is a ligand-activated transcription factor that plays a central role in the development and growth of prostate carcinoma. PIAS1 is an AR- and SUMO-interacting protein and a putative transcriptional coregulator overexpressed in prostate cancer. To study the importance of PIAS1 for the androgen-regulated transcriptome of VCaP prostate cancer cells, we silenced its expression by RNAi. Transcriptome analyses revealed that a subset of the AR-regulated genes is significantly influenced, either activated or repressed, by PIAS1 depletion. Interestingly, PIAS1 depletion also exposed a new set of genes to androgen regulation, suggesting that PIAS1 can mask distinct genomic loci from AR access. In keeping with gene expression data, silencing of PIAS1 attenuated VCaP cell proliferation. ChIP-seq analyses showed that PIAS1 interacts with AR at chromatin sites harboring also SUMO2/3 and surrounded by H3K4me2; androgen exposure increased the number of PIAS1-occupying sites, resulting in nearly complete overlap with AR chromatin binding events. PIAS1 interacted also with the pioneer factor FOXA1. Of note, PIAS1 depletion affected AR chromatin occupancy at binding sites enriched for HOXD13 and GATA motifs. Taken together, PIAS1 is a genuine chromatin-bound AR coregulator that functions in a target gene selective fashion to regulate prostate cancer cell growth.


Molecular Endocrinology | 2013

Prostaglandin 15d-PGJ2 Inhibits Androgen Receptor Signaling in Prostate Cancer Cells

Sanna Kaikkonen; Ville Paakinaho; Päivi Sutinen; Anna-Liisa Levonen; Jorma J. Palvimo

Androgen signaling, in particular overexpression of the androgen receptor (AR), is critical for the growth and progression of prostate cancer. Because the AR is amenable to targeting by small-molecule inhibitors, it remains the major druggable target for the advanced disease. Inflammation has also been implicated in the cancerous growth in the prostate. Here we show that 15-deoxy-Δ(12,14)-prostaglandin J(2) (15d-PGJ(2)), an endogenously produced antiinflammatory prostaglandin, targets the AR and acts as a potent AR inhibitor, rapidly repressing AR target genes, such as FKBP51 and TMPRSS2 in prostate cancer cells. However, exposure of prostate cancer cells to 15d-PGJ(2) does not simply evoke a general inhibition of nuclear receptor activity or transcription because under the same conditions, peroxisome proliferator-activated receptor-γ is activated by 15d-PGJ(2). Moreover, 15d-PGJ(2) rapidly triggers modifications of AR by small ubiquitin-related modifier-2/3 (SUMO-2/3), which may modulate the repressing effect of 15d-PGJ(2) on AR-dependent transcription. Chromatin immunoprecipitation assays indicate that the inhibitory effect of 15d-PGJ(2) on FKBP51 and TMPRSS2 expression occurs in parallel with the inhibition of the AR binding to the regulatory regions of these genes. However, the DNA-binding activity is not the only AR function targeted by 15d-PGJ(2) because the prostaglandin also blunted the androgen-dependent interaction between the AR amino and carboxy termini. In conclusion, our results identify 15d-PGJ(2) as a potent and direct inhibitor of androgen signaling, suggesting novel possibilities in restricting the AR activity in prostate cancer cells.


Biochimica et Biophysica Acta | 2010

SUMOylation can regulate the activity of ETS-like transcription factor 4.

Sanna Kaikkonen; Harri Makkonen; Miia Rytinki; Jorma J. Palvimo

ETS-like transcription factor 4 (ELK4) (a.k.a. serum response factor accessory protein 1) belongs to the ternary complex factor (TCF) subfamily of E twenty-six (ETS) domain transcription factors. Compared to the other TCF subfamily members, ELK1 and ELK3 (NET), there is limited information of the mechanisms regulating the ELK4 activity. Here, we show that the ELK4 can be covalently modified (SUMOylated) by small ubiquitin-related modifier (SUMO) 1 protein, an important regulator of signaling and transcription. SUMOylation of ELK4 was reversed by SUMO-specific proteases (SENP) 1 and 2 and stimulated by SUMO E3 ligase PIAS3. Conserved lysine residue 167 that is located in the NET inhibitory domain of ELK4 was identified as the main site of SUMO-1 conjugation. Interestingly, mutation of the K167 disrupting the SUMOylation markedly enhanced the transcriptional activity of the ELK4, but weakened its repressive function on c-fos promoter. In conclusion, our results suggest that covalent modification by SUMO-1 can regulate the activity of ELK4, contributing to the transcriptional repression by the ELK4.


Molecular and Cellular Endocrinology | 2014

Preclinical pharmacology of FL442, a novel nonsteroidal androgen receptor modulator

Pekka K. Poutiainen; Tuulia Huhtala; Tiina Jääskeläinen; Aleksanteri Petsalo; Jenni Küblbeck; Sanna Kaikkonen; Jorma J. Palvimo; Hannu Raunio; Ale Närvänen; Mikael Peräkylä; Risto O. Juvonen; Paavo Honkakoski; Reino Laatikainen; Juha T. Pulkkinen

The preclinical profiles of two most potent compounds of our recently published cycloalkane[d]isoxazole pharmacophore-based androgen receptor (AR) modulators, FL442 (4-(3a,4,5,6,7,7a-hexahydro-benzo[d]isoxazol-3-yl)-2-(trifluoromethyl)benzonitrile) and its nitro analog FL425 (3-(4-nitro-3-(trifluoromethyl)phenyl)-3a,4,5,6,7,7a-hexahydrobenzo[d]isoxazole), were explored to evaluate their druggability for the treatment of AR dependent prostate cancer. The studies revealed that both compounds are selective to AR over other closely related steroid hormone receptors and that FL442 exhibits equal inhibition efficiency towards the androgen-responsive LNCaP prostate cancer cell line as the most widely used antiandrogen bicalutamide and the more recently discovered enzalutamide. Notably, FL442 maintains antiandrogenic activity with enzalutamide-activated AR mutant F876L. In contrast to bicalutamide, FL442 does not stimulate the VCaP prostate cancer cells which express elevated levels of the AR. Distribution analyses showed that [(14)CN]FL442 accumulates strongly in the mouse prostate. In spite of its low plasma concentration obtained by intraperitoneal administration, FL442 significantly inhibited LNCaP xenograft tumor growth. These findings provide a preclinical proof for FL442 as a promising AR targeted candidate for a further optimization.


Molecular and Cellular Biology | 2014

Electrophilic lipid mediator 15-deoxy-Δ12,14-prostaglandin j2 modifies glucocorticoid signaling via receptor SUMOylation.

Ville Paakinaho; Sanna Kaikkonen; Anna-Liisa Levonen; Jorma J. Palvimo

ABSTRACT Cortisol, the central stress hormone in humans, activates the glucocorticoid receptor (GR). Anti-inflammatory effects are the most important pharmaceutical effects mediated by the GR. Inasmuch as electrophilic cyclopentenone prostaglandin 15-deoxy-Δ12,14-prostaglandin J2 (15d-PGJ2) has potent anti-inflammatory properties and activates the SUMOylation pathway, we have investigated the effect of 15d-PGJ2 on glucocorticoid signaling and receptor SUMOylation. To this end, we studied isogenic HEK293 cells expressing either wild-type GR or SUMOylation-defective GR. Interestingly, 15d-PGJ2 triggered SUMO-2 and -3 (SUMO-2/3) modification in the primary SUMOylation sites of the GR. Gene expression profiling and pathway analyses indicate that 15d-PGJ2 inhibits GR signaling in a genome-wide fashion that is significantly dependent on the GR SUMOylation sites. Chromatin immunoprecipitation assays showed that the repressive effect of 15d-PGJ2 on GR target gene expression occurs in parallel with the inhibition of receptor binding to the target gene chromatin. Furthermore, depletion of UBC9, the sole SUMO E2 conjugase, from HEK293 cells confirmed the involvement of active SUMOylation in the regulatory process. Taken together, our data indicate that GR SUMOylation modulates the glucocorticoid signaling during acute cell stress. Our data also suggest that GR SUMOylation modulates cross talk of the glucocorticoid signaling with other transcription factors that are responsive to cell stress.

Collaboration


Dive into the Sanna Kaikkonen's collaboration.

Top Co-Authors

Avatar

Jorma J. Palvimo

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Tiina Jääskeläinen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Ville Paakinaho

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Miia Rytinki

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Päivi Sutinen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Harri Makkonen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar

Anna-Liisa Levonen

University of Eastern Finland

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Vesa Rahkama

University of Eastern Finland

View shared research outputs
Researchain Logo
Decentralizing Knowledge