Dr Agnieszka Konopacka

Group Leader: Induced Proximity Therapeutics

OrcID: 0000-0003-2192-1928

Email: [email protected]

Location: Sutton

Headshot of Agnieszka Konopacka in front of an ICR building

OrcID: 0000-0003-2192-1928

Email: [email protected]

Location: Sutton

Dr Agnieszka Konopacka has received a PhD in Medical Sciences in 2008 from the Mossakowski Medical Research Centre in Warsaw, Poland. Following postdoctoral training at Bristol University, where she studied molecular mechanisms in the brain that regulate cardiovascular homeostasis, in 2014 she moved to pharma industry to work on various neuroscience drug discovery programs at Pfizer Neusentis, and on regulatory oncology assay development at Horizon Discovery.

In 2017 she joined the GSK Protein Degradation Group pioneering innovative targeted protein degradation technologies, where she led multiple drug discovery initiatives in the oncology, immuno-oncology and immunology portfolio projects. She was elected a GSK Fellow to foster innovation in drug discovery and she was involved in multiple collaborations between pharma industry and academia. She supervised and mentored multiple industrial placement and PhD students. For her contributions to drug discovery, she was honoured with several industry awards including two prestigious ‘GSK Innovation, Performance, Trust’ Awards.

In 2024 she joined the ICR to lead biology at the newly opened Centre for Protein Degradation. Her research focuses on discovery of novel molecular glues and PROTAC degraders and enabling novel E3 ligases for cancer therapy, and provides comprehensive drug discovery platforms for target validation, cell-based screening, mechanistic profiling and in vitro efficacy studies.

She is a committee member of the Society for Medicines Research and the Women in TPD group, engaged in organising scientific events and mentoring.

Types of Publications

Journal articles

Röth, S., Kocaturk, N.M., Sathyamurthi, P.S., Carton, B., Watt, M., Macartney, T.J., Chan, K.-.H., Isidro-Llobet, A., Konopacka, A., Queisser, M.A., Sapkota, G.P. (2023). Identification of KLHDC2 as an efficient proximity-induced degrader of K-RAS, STK33, β-catenin, and FoxP3. Cell Chemical Biology, Vol.30(10), p. 1261. show abstract

Targeted protein degradation (TPD), induced by enforcing target proximity to an E3 ubiquitin ligase using small molecules has become an important drug discovery approach for targeting previously undruggable disease-causing proteins. However, out of over 600 E3 ligases encoded by the human genome, just over 10 E3 ligases are currently utilized for TPD. Here, using the affinity-directed protein missile (AdPROM) system, in which an anti-GFP nanobody was linked to an E3 ligase, we screened over 30 E3 ligases for their ability to degrade 4 target proteins, K-RAS, STK33, β-catenin, and FoxP3, which were endogenously GFP-tagged. Several new E3 ligases, including CUL2 diGly receptor KLHDC2, emerged as effective degraders, suggesting that these E3 ligases can be taken forward for the development of small-molecule degraders, such as proteolysis targeting chimeras (PROTACs). As a proof of concept, we demonstrate that a KLHDC2-recruiting peptide-based PROTAC connected to chloroalkane is capable of degrading HALO-GFP protein in cells.

Bárez-López, S., Konopacka, A., Cross, S.J., Greenwood, M., Skarveli, M., Murphy, D., Greenwood, M.P. (2022). Transcriptional and Post-Transcriptional Regulation of Oxytocin and Vasopressin Gene Expression by CREB3L1 and CAPRIN2. Neuroendocrinology, Vol.112(11), pp. 1058-1077. show abstract

<h4>Introduction</h4>Water homoeostasis is achieved by secretion of the peptide hormones arginine vasopressin (AVP) and oxytocin (OXT) that are synthesized by separate populations of magnocellular neurones (MCNs) in the supraoptic and paraventricular (PVN) nuclei of the hypothalamus. To further understand the molecular mechanisms that facilitate biosynthesis of AVP and OXT by MCNs, we have explored the spatiotemporal dynamic, both mRNA and protein expression, of two genes identified by our group as being important components of the osmotic defence response: Caprin2 and Creb3l1.<h4>Methods</h4>By RNA in situ hybridization and immunohistochemistry, we have characterized the expression of Caprin2 and Creb3l1 in MCNs in the basal state, in response to dehydration, and during rehydration in the rat.<h4>Results</h4>We found that Caprin2 and Creb3l1 are expressed in AVP and OXT MCNs and in response to dehydration expression increases in both MCN populations. Protein levels mirror the increase in transcript levels for both CREB3L1 and CAPRIN2. In view of increased CREB3L1 and CAPRIN2 expression in OXT neurones by dehydration, we explored OXT-specific functions for these genes. By luciferase assays, we demonstrate that CREB3L1 may be a transcription factor regulating Oxt gene expression. By RNA immunoprecipitation assays and Northern blot analysis of Oxt mRNA poly(A) tails, we have found that CAPRIN2 binds to Oxt mRNA and regulates its poly(A) tail length. Moreover, in response to dehydration, Caprin2 mRNA is subjected to nuclear retention, possibly to regulate Caprin2 mRNA availability in the cytoplasm.<h4>Conclusion</h4>The exploration of the spatiotemporal dynamics of Creb3l1- and Caprin2-encoded mRNAs and proteins has provided novel insights beyond the AVP-ergic system, revealing novel OXT-ergic system roles of these genes in the osmotic defence response.

Stacey, P., Lithgow, H., Lewell, X., Konopacka, A., Besley, S., Green, G., Whatling, R., Law, R., Röth, S., Sapkota, G.P., Smith, I.E.D., Burley, G.A., Harling, J., Benowitz, A.B., Queisser, M.A., Muelbaier, M. (2021). A Phenotypic Approach for the Identification of New Molecules for Targeted Protein Degradation Applications. SLAS Discovery, Vol.26(7), pp. 885-895. show abstract

Targeted protein degradation is an emerging new strategy for the modulation of intracellular protein levels with applications in chemical biology and drug discovery. One approach to enable this strategy is to redirect the ubiquitin-proteasome system to mark and degrade target proteins of interest (POIs) through the use of proteolysis targeting chimeras (PROTACs). Although great progress has been made in enabling PROTACs as a platform, there are still a limited number of E3 ligases that have been employed for PROTAC design. Herein we report a novel phenotypic screening approach for the identification of E3 ligase binders. The key concept underlying this approach is the high-throughput modification of screening compounds with a chloroalkane moiety to generate HaloPROTACs in situ, which were then evaluated for their ability to degrade a GFP-HaloTag fusion protein in a cellular context. As proof of concept, we demonstrated that we could generate and detect functional HaloPROTACs in situ, using a validated Von Hippel-Lindau (VHL) binder that successfully degraded the GFP-HaloTag fusion protein in living cells. We then used this method to prepare and screen a library of approximately 2000 prospective E3 ligase-recruiting molecules.

Chung, C.-.W., Dai, H., Fernandez, E., Tinworth, C.P., Churcher, I., Cryan, J., Denyer, J., Harling, J.D., Konopacka, A., Queisser, M.A., Tame, C.J., Watt, G., Jiang, F., Qian, D., Benowitz, A.B. (2020). Structural Insights into PROTAC-Mediated Degradation of Bcl-xL. ACS Chemical Biology, Vol.15(9), pp. 2316-2323.
Roth, S., Macartney, T.J., Konopacka, A., Chan, K.-.H., Zhou, H., Queisser, M.A., Sapkota, G.P. (2020). Targeting Endogenous K-RAS for Degradation through the Affinity-Directed Protein Missile System. Cell Chemical Biology, Vol.27(9), p. 1151.
Loh, S.-.Y., Jahans-Price, T., Greenwood, M.P., Greenwood, M., Hoe, S.-.Z., Konopacka, A., Campbell, C., Murphy, D., Hindmarch, C.C.T. (2017). Unsupervised Network Analysis of the Plastic Supraoptic Nucleus Transcriptome Predicts Caprin2 Regulatory Interactions. eNeuro, Vol.4(6). show abstract

The supraoptic nucleus (SON) is a group of neurons in the hypothalamus responsible for the synthesis and secretion of the peptide hormones vasopressin and oxytocin. Following physiological cues, such as dehydration, salt-loading and lactation, the SON undergoes a function related plasticity that we have previously described in the rat at the transcriptome level. Using the unsupervised graphical lasso (Glasso) algorithm, we reconstructed a putative network from 500 plastic SON genes in which genes are the nodes and the edges are the inferred interactions. The most active nodal gene identified within the network was <i>Caprin2</i>. <i>Caprin2</i> encodes an RNA-binding protein that we have previously shown to be vital for the functioning of osmoregulatory neuroendocrine neurons in the SON of the rat hypothalamus. To test the validity of the Glasso network, we either overexpressed or knocked down <i>Caprin2</i> transcripts in differentiated rat pheochromocytoma PC12 cells and showed that these manipulations had significant opposite effects on the levels of putative target mRNAs. These studies suggest that the predicative power of the Glasso algorithm within an <i>in vivo</i> system is accurate, and identifies biological targets that may be important to the functional plasticity of the SON.

Konopacka, A., Greenwood, M., Loh, S.-.Y., Paton, J., Murphy, D. (2015). RNA binding protein Caprin-2 is a pivotal regulator of the central osmotic defense response. eLife, Vol.4, p. e09656. show abstract

In response to an osmotic challenge, the synthesis of the antidiuretic hormone arginine vasopressin (AVP) increases in the hypothalamus, and this is accompanied by extension of the 3' poly(A) tail of the AVP mRNA, and the up-regulation of the expression of RNA binding protein Caprin-2. Here we show that Caprin-2 binds to AVP mRNAs, and that lentiviral mediated shRNA knockdown of Caprin-2 in the osmotically stimulated hypothalamus shortens the AVP mRNA poly(A) tail at the same time as reducing transcript abundance. In a recapitulated in vitro system, we confirm that Caprin-2 over-expression enhances AVP mRNA abundance and poly(A) tail length. Importantly, we show that Caprin-2 knockdown in the hypothalamus decreases urine output and fluid intake, and increases urine osmolality, urine sodium concentration, and plasma AVP levels. Thus Caprin-2 controls physiological mechanisms that are essential for the body's response to osmotic stress.

Konopacka, A., Qiu, J., Yao, S.T., Greenwood, M.P., Greenwood, M., Lancaster, T., Inoue, W., Mecawi, A.D.S., Vechiato, F.M.V., de Lima, J.B.M., Coletti, R., Hoe, S.Z., Martin, A., Lee, J., Joseph, M., Hindmarch, C., Paton, J., Antunes-Rodrigues, J., Bains, J., Murphy, D. (2015). Osmoregulation requires brain expression of the renal Na-K-2Cl cotransporter NKCC2. Journal of Neuroscience, Vol.35(13), pp. 5144-5155. show abstract

The Na-K-2Cl cotransporter 2 (NKCC2) was thought to be kidney specific. Here we show expression in the brain hypothalamo-neurohypophyseal system (HNS), wherein upregulation follows osmotic stress. The HNS controls osmotic stability through the synthesis and release of the neuropeptide hormone, arginine vasopressin (AVP). AVP travels through the bloodstream to the kidney, where it promotes water conservation. Knockdown of HNS NKCC2 elicited profound effects on fluid balance following ingestion of a high-salt solution-rats produced significantly more urine, concomitant with increases in fluid intake and plasma osmolality. Since NKCC2 is the molecular target of the loop diuretics bumetanide and furosemide, we asked about their effects on HNS function following disturbed water balance. Dehydration-evoked GABA-mediated excitation of AVP neurons was reversed by bumetanide, and furosemide blocked AVP release, both in vivo and in hypothalamic explants. Thus, NKCC2-dependent brain mechanisms that regulate osmotic stability are disrupted by loop diuretics in rats.

Murphy, D., Konopacka, A., Hindmarch, C., Paton, J.F.R., Sweedler, J.V., Gillette, M.U., Ueta, Y., Grinevich, V., Lozic, M., Japundzic-Zigon, N. (2012). The hypothalamic-neurohypophyseal system: from genome to physiology. Journal of Neuroendocrinology, Vol.24(4), pp. 539-553. show abstract

The elucidation of the genomes of a large number of mammalian species has produced a huge amount of data on which to base physiological studies. These endeavours have also produced surprises, not least of which has been the revelation that the number of protein coding genes needed to make a mammal is only 22 333 (give or take). However, this small number belies an unanticipated complexity that has only recently been revealed as a result of genomic studies. This complexity is evident at a number of levels: (i) cis-regulatory sequences; (ii) noncoding and antisense mRNAs, most of which have no known function; (iii) alternative splicing that results in the generation of multiple, subtly different mature mRNAs from the precursor transcript encoded by a single gene; and (iv) post-translational processing and modification. In this review, we examine the steps being taken to decipher genome complexity in the context of gene expression, regulation and function in the hypothalamic-neurohypophyseal system (HNS). Five unique stories explain: (i) the use of transcriptomics to identify genes involved in the response to physiological (dehydration) and pathological (hypertension) cues; (ii) the use of mass spectrometry for single-cell level identification of biological active peptides in the HNS, and to measure in vitro release; (iii) the use of transgenic lines that express fusion transgenes enabling (by cross-breeding) the generation of double transgenic lines that can be used to study vasopressin (AVP) and oxytocin (OXT) neurones in the HNS, as well as their neuroanatomy, electrophysiology and activation upon exposure to any given stimulus; (iv) the use of viral vectors to demonstrate that somato-dendritically released AVP plays an important role in cardiovascular homeostasis by binding to V1a receptors on local somata and dendrites; and (v) the use of virally-mediated optogenetics to dissect the role of OXT and AVP in the modulation of a wide variety of behaviours.

Konopacka, A., Konopacki, F.A., Albrecht, J. (2009). Protein kinase G is involved in ammonia-induced swelling of astrocytes. Journal of Neurochemistry, Vol.109, pp. 246-251.
Konopacka, A., Zielinska, M., Albrecht, J. (2008). Ammonia inhibits the C-type natriuretic peptide-dependent cyclic GMP synthesis and calcium accumulation in a rat brain endothelial cell line. Neurochemistry International, Vol.52(6), pp. 1160-1166.
Zielinska, M., Fresko, I., Konopacka, A., Felipo, V., Albrecht, J. (2007). Hyperammonemia inhibits the natriuretic peptide receptor 2 (NPR-2)-mediated cyclic GMP synthesis in the astrocytic compartment of rat cerebral cortex slices. NeuroToxicology, Vol.28(6), pp. 1260-1263.
Konopacka, A., Fresko, I., Piaskowski, S., Albrecht, J., Zielinska, M. Ammonia affects the activity and expression of soluble and particulate GC in cultured rat astrocytes. Neurochemistry International, Vol.48(6-7), pp. 553-558.
Röth, S., Macartney, T.J., Konopacka, A., Queisser, M.A., Sapkota, G. Targeting Endogenous K-RAS for Degradation Through the Affinity-Directed Protein Missile System. SSRN Electronic Journal.