Skip to main content

Plasma membrane and nuclear phosphatidylinositol 4,5-bisphosphate signalling in cancer

Abstract

The development of metastasis is a leading cause of cancer-related death that involves specific changes in the plasma membrane (PM) and nucleus of cancer cells. Elevated levels of membrane lipids, including sphingomyelin, cholesterol, and phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2), in the PM, contribute to changes in membrane rigidity, lipid raft formation, and actin polymerisation dynamics, processes that drive cell invasion. This review discusses the relationship between well-studied cytoplasmic phosphoinositides and their lesser-known nuclear counterparts, highlighting their functional role in metastatic progression. Nuclear phosphoinositides, particularly PI(4,5)P2, are essential for regulating transcription factors and chromatin organisation, thereby shaping gene expression patterns. We also explore the role of PI(4,5)P2 and its metabolism in cancer cell invasiveness and metastasis, proposing a model in which the dysregulation of cytosolic and/or nuclear PI(4,5)P2 pool triggers malignant transformation. Understanding the PI(4,5)P2-related mechanisms underlying metastasis may provide insights into potential therapeutic targets, paving the way for more effective therapies and improved patient outcomes.

Graphical Abstract

Introduction

Cancer is a leading cause of death worldwide [1]. In most cases, the increased invasiveness of cancer cells is responsible for their lethal potential. Metastasis is defined as the spread of cancer cells from their primary site to neighbouring tissues and other organs via blood vessels and the lymphatic system. This process can be divided into five sequential stages. Invasion, a crucial step in the progression of metastasis, followed by intravasation, survival in circulation, extravasation, and colonisation [2]. A deeper understanding of the molecular mechanisms underlying this metastatic cascade will lead to the development of effective therapies and improved patient prognosis.

Metastatic progression is associated with altered composition and dynamics of the plasma membrane (PM) and cell nucleus. Given the complex nature of the PM, consisting of a lipid bilayer and various proteins, these alterations can occur as changes in membrane rigidity, increased lipid raft formation, or differences in PM signalling [3], as well as alterations in actin polymerisation and depolymerisation, which contribute to enhanced cellular invasion [4]. Lipid rafts are highly ordered microdomains enriched in sphingomyelin and cholesterol [5], serving as key platforms for physiological processes, including signalling, migration, trafficking, cellcell communication, and cellmatrix interaction [6]. Although the enrichment of sphingomyelin and cholesterol mainly characterises lipid rafts, significant amounts of phosphoinositides (PIPs) are also present [7, 8]. Among these, phosphatidylinositol 4,5bisphosphate (PI(4,5)P2), is a crucial regulator of Factin dynamics, significantly influencing cell dynamics and invasive potential [9]. Disruption of lipid rafts within the PM, responsible for organising specific proteins and regulating lipid microdomains, can modulate membraneassociated signalling pathways, such as the phosphoinositide 3kinase/protein kinase B (PI3K/Akt) pathway [3, 10]. This, in turn, affects the migratory and invasive behaviour of cancer cells by altering actin remodelling, promoting invadopodia formation, and enhancing their capacity to degrade the extracellular matrix (ECM). Ultimately, these changes increase the metastatic potential of cancer cells [4].

In addition, nuclear dynamics affect cancer metastasis by altering the shape and size of the nucleus and the structure of the nuclear envelope [11, 12]. Studies have demonstrated that nuclear morphological changes are prevalent in cancer cells and may influence their invasiveness. For instance, cancer cells frequently exhibit enlarged and irregularly shaped nuclei, which have been linked to an elevated capacity for metastasis in a multitude of distinct cancer types [11, 12]. Moreover, dysregulation of nuclear signalling pathways, including those mediated by transcription factors, chromatin modifiers, and DNA damage response proteins, can significantly affect gene expression patterns involved in cancer metastasis. Transcription factors play a critical role in the regulation of gene expression associated with cell motility, invasion, and metastasis [2]. Dysregulated expression or activity of these transcription factors can promote the expression of prometastatic genes while inhibiting the expression of genes involved in tumour suppression and cell adhesion [2]. Furthermore, alterations in chromatin structure and epigenetic modifications can also contribute to cancer metastasis by affecting gene expression patterns [13]. Epigenetic modifications, such as DNA methylation or histone acetylation, have been demonstrated to regulate the expression of genes involved in metastatic progression. For instance, aberrant DNA methylation patterns at the promoter regions of tumour suppressor genes can result in their silencing, thereby promoting metastasis [14].

Nuclear phosphoinositides have emerged as essential regulators of processes involved in metastasis and invasiveness of cancer cells [15,16,17]. These lipid molecules not only participate in signalling cascades that control gene expression and cellular responses to extracellular signals but also contribute to the regulation of nuclear envelope dynamics and chromatin organisation [18,19,20]. Recent studies have underscored the importance of nuclear phosphoinositides in modulating the activities of transcription factors and chromatin modifiers involved in metastatic progression [21, 22].

In this review, we focus on PI(4,5)P2, the most abundant phosphoinositide and a key regulator of cell invasion. We highlight the potential implications of the interplay between cytoplasmic and nuclear PI(4,5)P2 pathways in driving cancer invasiveness.

The role of plasma membrane phosphoinositide-associated structures and molecules in cancer progression and metastasis

Lipids, which serve as the backbone of cell membranes, are essential for maintaining the cellular structure. Membrane composition comprises a mix of three main lipid groups: glycerophospholipids (e.g., phosphatidylcholine, phosphatidylinositol, phosphatidylethanolamine, phosphatidylserine), sphingolipids (e.g., sphingomyelin), and sterols (e.g., cholesterol) [23, 24]. In addition, membrane dynamics, together with lipid heterogeneity, leads to the formation of specialised domains that selectively recruit or exclude specific proteins. These dynamic sphingomyelincholesterolrich microdomains, known as lipid rafts, play a crucial role in membrane compartmentalisation [5, 25,26,27], and are involved in crucial processes associated with cancer progression and metastasis [28]. Lipid raft marker proteins, such as caveolin-1 and flotillin-1 and − 2, have been shown to regulate key cancer processes. In metastatic prostate cancer, phosphorylated caveolin-1 facilitates autophosphorylation of VEGFR2 and its colocalisation at focal adhesion complexes. This, in turn, stimulates angiogenic signalling, which is crucial for new blood vessel formation and tumour growth [29]. The upregulation of flotillin 2 in gastric cancer cells is required for TGFβinduced epithelialtomesenchymal transition (EMT), which increases cancer cell motility and invasiveness [30]. Moreover, overexpression of both flotillin 2 mRNA and protein in breast cancer is associated with poor prognosis, irrespective of disease stage [31]. Since lipid rafts are not the primary focus of this review, we do not discuss this topic in further detail. Researchers interested in a comprehensive overview of lipid rafts in cancer are referred to a recent review [3].

Apart from receptors, phosphoinositides (PIPs) are among the most important molecules in the plasma membrane that are involved in cell signalling [15, 32]. They enable plasma membrane receptors to transmit and amplify signal received from signalling molecules, such as growth factors or hormones. The most common interacting domains found in PIP binding proteins include the pleckstrin homology (PH) domain, Fab-1, YGL023, Vps27, and EEA1 (FYVE), Phox homology (PX), 4.1 protein, ezrin, radixin, moesin (FERM) domain, and plant homeodomain (PHD) finger domains, as reviewed in detail elsewhere [33,34,35]. PIPs are distributed asymmetrically between the two plasma membrane leaflets. In the outer leaflet, phosphoinositides are associated with GPIanchored proteins in the lipid rafts. PI(4,5)P2 and other phosphorylated isoforms predominantly localize in the inner plasma membrane leaflet because of their critical roles in numerous signalling cascades [42] and regulatory processes [36, 37]. The localisation of PI(4,5)P2 in the inner membrane is significantly affected by the surrounding lipid environment. Lipid domains comprising sphingomyelin with shorter acyl chains provide a stable environment for PI(4,5)P2, promoting its accumulation. Conversely, when the membrane domains are enriched in sphingomyelin with long acyl chains, PI(4,5)P2 is expelled from these domains and instead localises to the less ordered membrane regions [7].

Impact of phosphoinositide imbalance on cancer progression

Two classes of enzymes, kinases and phosphatases, are involved in maintaining the phosphoinositide balance. The class I kinases phosphatidylinositol-4phosphate 5-kinases (PIP5K, PI4P 5-kinase) and class II kinases phosphatidylinositol 5phosphate 4kinase (PIP4K, PI5P 4-kinase) catalyse the phosphorylation of PI4P and PI5P, respectively, to PI(4,5)P2 [32, 37, 38]. PI(4,5)P2 is also a substrate of secondary messengers diacylglycerol (DAG) and inositol trisphosphate (IP3), which are formed during PI(4,5)P2 hydrolysis by phospholipase C (PLC). The hydrolysis of PI(4,5)P2 to PI4P is catalysed by phosphoinositide 5phosphatases (e.g. Synaptojanin-1, Synaptojanin-2, inositol polyphosphate-5phosphatase B/E/J/K, OCRL inositol polyphosphate-5phosphatase, SHIP2) [32, 37]. These phosphatases, particularly Synaptojanin-2 and SHIP1/2, also catalyse the hydrolysis of PI(3,4,5)P3 into PI(3,4)P2.The formation of PI(3,4,5)P3 from PI(4,5)P2 is catalysed by class I PI3K kinases [17, 32, 39], while the antagonistic reaction is mediated by the phosphatase and tensin homolog deleted on chromosome 10 (PTEN) [28, 40]. PTEN is a tumour suppressor protein and lipid phosphatase. Depending on its posttranslational modifications, PTEN is targeted to the nucleus or the plasma membrane [41]. Dysfunction of either kinases or phosphatases causes an imbalance between phosphoinositide levels in the cell and is frequently observed in many diseases, including obesity, type 2 diabetes, endometriosis, neurodegenerative diseases, and cancer [42]. Cancer cells frequently exhibit an imbalance in the levels of phosphoinositides, with a notable increase in the total cellular levels of PI(3,4,5)P3 and a decrease in the total cellular levels of PI(4,5)P2. This imbalance is largely attributed to the loss or inactivation of PTEN or activating mutations in the gene PIK3CA, both of which result in elevated levels of PI(3,4,5)P3 [42, 43]. It is still unknown whether these changes affect the cytoplasmic and nuclear PI(4,5)P2 pools in the same way or if they differ from each other. Research suggests that both the cytoplasm and nucleus maintain distinct pools of PIP enzymes, yet some precursors may translocate between these two compartments [44].

PIK3CA encodes the PI3Kα catalytic subunit p110α and is one of the most frequently mutated oncogenes in various types of cancer [44]. The regulatory subunit of PI3K, p85, is encoded by PIK3R1 (isoforms p85α, p55α, and p50α), PIK3R2 (isoform p85β), and PIK3R3 (isoform p55γ). The PI3K signalling pathway is regulated by Src kinase through two mechanisms. Firstly, by direct phosphorylation of the p85 subunit, which recruits PI3K to the plasma membrane, where it can interact with PI(4,5)P2 [45, 46]. Secondly, Src inhibits phosphatase activity and reduces PTEN stability, affecting its ability to hydrolyse PI(3,4,5)P3 [47]. Src, a member of the nonreceptor proteintyrosine kinase family, controls a variety of cellular processes, such as differentiation, proliferation, and adhesion, through a complex regulatory mechanism [48]. During this process, increased Src affinity to the plasma membrane regulates cell motility [49]. Moreover, elevated Src activity has been detected in numerous cancer types, such as skin, ovarian, lung, and head and neck cancers [48, 50]. In squamous cell carcinomas (SCC), including those originating from the head and neck region (HNSCC), lung, vulva and uterine cervical mucosa, an increase in the copy number of chromosome 3q is frequently observed, leading to the amplification of PIK3CA [51,52,53]. The Cancer Genome Atlas for HNSCC revealed that PIK3CA is the most frequently altered gene [54]. However, a significant association between PIK3CA mutation status and overall survival or diseasefree survival was not observed. Therefore, the clinicopathological and prognostic significance of mutated PIK3CA in SCC tissues remains under debate [55]. In smokingassociated HNSCC, universal lossoffunction TP53 mutations and CDKN2A inactivation have been detected, whereas, in HPVassociated HNSCC, mutations in the helical domain of the oncogene PIK3CA, loss of TRAF3, and amplification of the cell cycle gene E2F1 are frequently observed [54]. Importantly, in HPVpositive patients, wildtype PIK3CA was associated with a 4.64fold increase in the risk of recurrence or death. This implies that stratifying patients according to their HPV status followed by their PIK3CA status can help predict prognosis and identify patients who may benefit from deintensified treatment combined with targeted therapies against the affected signalling pathway. In the future, it might be worth considering not only to investigate the mutation per se but also to determine the cellular effects, such as altered cytoplasmic and/or nuclear quantity and localisation of PI(3,4,5)P3, PI(4,5)P2 and other phosphoinositides [56].

Fig. 1
figure 1

The dynamic regulation of the cytosolic PI(4,5)P2/PI(3,4,5)P3 levels plays a crucial role in many key cell processes, including cell growth, proliferation or organisation of the actin cytoskeleton. The figure illustrates how the disruption of PI(4,5)P2 homeostasis modulates PI3K/Akt downstream signalling and actin cytoskeleton dynamics. (A) In normal cells, the two actin forms, monomeric actin (Gactin) and filamentous actin (F-actin) are under constant dynamic conversion driven by PI(4,5)P2 via actin regulatory proteins. (B) In cancer cells, the imbalanced PI(4,5)P2/PI(3,4,5)P2 levels influence the hyperactivation of Akt. This, in turn, leads to alterations in actin remodelling dynamics, which can be observed as increased actin polymerisation, invadopodia formation and, in consequence, enhanced cell motility, invasivity and metastasis

Phosphoinositide-mediated actin remodelling in cancer progression

High levels of PI(3,4,5)P3 activate PH domain effector proteins (e.g. Akt, Cdc42, Rac) and downstream signalling involved in processes, such as cell growth, proliferation, survival, and metabolism, which can lead to increased invasiveness, metastasis, and resistance to therapy in cancer cells. Metabolic dysregulation caused by the increased hydrolysis of PI(4,5)P2 by PLC leads to increased production of inositol trisphosphate (IP3) and diacylglycerol (DAG). This process triggers the release of calcium ions (Ca2+) from intracellular stores, which subsequently activates protein kinase C (PKC). As a result, various signalling pathways, including RAS/RAF/MAPK/ERK and JAK/STAT, are initiated, playing key roles in the regulation of angiogenesis, cell proliferation, survival, and autophagy [2, 39]. Furthermore, the organisation of the actin cytoskeleton is influenced by the PI(4,5)P2 and PI(3,4,5)P3 metabolism. The actin cytoskeleton is a dynamic structure of actin filaments responsible for maintaining cell shape, vesicle trafficking, cell division, signalling, and invasiveness (Fig. 1). PI(4,5)P2, which transiently regulates actinbinding proteins (ABPs), is responsible for actin cytoskeletal dynamics by regulating the formation of actin filaments (Factin) from monomeric actin molecules (Gactin) [9]. The neuronal WiskottAldrich syndrome protein (NWASP) is recruited to the cell membrane by PI(4,5)P2. It changes its conformation and activates the actinrelated protein 2/3 (ARP2/3) complex, which exhibits nucleation and branching activities. Binding to PI(4,5)P2 inhibits profilin activity, which delivers monomeric Gactin to the barbed ends. Similarly, gelsolin and cofilin severing activities were blocked by PI(4,5)P2. Active gelsolin has an additional capping ability that blocks the free ends of actin. In contrast, once dissociated from the membrane, active cofilin accelerates the dynamics of actin turnover through severing and depolymerisation activity [37]. Cofilin activity is dependent on phosphorylation, a process that is tightly regulated by RhoGTPase, LIM kinase (LIMK), and phosphoinositide binding. The Rho, Rac, and Cdc42 members of the Rho family of small GTPases play pivotal roles in orchestrating actin reorganisation via their diverse downstream effectors [57]. Rho, Rac, and Cdc42 are known regulators of podosomes. Podosomes are actinbased structures that mediate cellmatrix contacts [4]. They consist of more than 300 proteins, including ABPs, such as NWASP, Arp2/3, cofilin, and cortactin. They can be found in different cell types like monocytic cells, osteoclasts, and endothelial cells. They participate in various physiological processes such as immune surveillance, bone degradation, and angiogenic sprouting. These processes facilitate extracellular matrix (ECM) degradation and invasion of the surrounding tissues. In podosomes, the PI3K/Akt signalling pathway and Src kinase promote actin polymerisation and matrix metalloproteinase (MMP) secretion [4, 28]. Similar actinbased and lipid raftenriched structures, with the addition of Diaphanousrelated formins (DRFs), have been identified in highly invasive cancer cells [4]. In this context, the protrusive structures with ECM degradation activity that drive proteolytic invasion are known as invadopodia. They play a crucial role in increased cellular migration, angiogenesis, and metastasis of cancer cells [28, 58]. Similar to podosomes, invadopodia activity and formation are regulated by PI(4,5)P2/PI(3,4)P2 levels, PTEN inactivation, and actin turnover. Moreover, invadopodia formation has also been shown to be lipid raftdependent. In breast cancer cells, colocalisation of the matrix metalloproteinase MT1MMP with caveolin-1 promotes the recruitment of invadopodia to lipid rafts. At the same time, CAV1 silencing and lipid raft disruption by mβCD were found to inhibit MT1MMP activation and abolish invadopodia formation [59]. In addition, another study demonstrated that caveolin-1 activation of MT1MMP occurs through the PI3K/AKT/mTOR pathway [60].

The role of nuclear phosphoinositides in chromatin regulation, transcription, and cancer progression

The role of actin in the cytoplasm is well known, but its presence in the cell nucleus has been debated for many years. Recently, the development of advanced imaging techniques has made it possible to visualise and study nuclear actin in detail [61, 62]. Actin lacks a nuclear localisation signal (NLS); thus, its nuclear localisation depends on active transport. This shuttle transport is enabled by cofilin and importin 9, which are responsible for importing actin into the nucleus, and profilin and exportin 6, which facilitate actin export [63]. The main limitations of this process are the availability of Gactin in the cytoplasm and the level of unphosphorylated cofilin. For a long time, the inability to visualise filamentous nuclear actin led to the assumption that actin did not form filaments within the nucleus. However, there is now evidence that nuclear actin filaments can be observed during specific physiological processes or pathological conditions, although they are mainly present as monomers [61, 64]. One of these processes is the regulation of chromatin regulatory complexes. Actin interacts in a PI(4,5)P2dependent manner with the mammalian ATPdependent chromatinremodelling complex (SWI/SNFlike BAF complex), which is involved in a variety of cellular processes ranging from gene activation and repression to cell development and differentiation. [65, 66]. In homologydirected DNA damage repair (DDR), the N-WASP/Arp2/3 complex mediates nuclear actin polymerisation, which is required for the clustering of DNA breaks [63]. In addition, actin has been reported to be involved in the initiation and regulation of transcription mediated by all three RNA polymerases [67,68,69]. Additionally, nuclear Gactin plays a role in Pol II pause release through its interaction with PTEFb [70]. Interestingly, upon serum stimulation, Arp2/3 and N-WASP promote actin polymerisation, which plays a crucial role in enhancing Pol II clustering and significantly changing the dynamics of transcription condensates. The larger and more stable clusters transcribe genes associated with survival, proliferation, angiogenesis, migration, EMT, and drug resistance [71].

In comparison to actin, the presence of phosphoinositides in the cell nucleus is less controversial and more widely accepted [72, 73]. Phosphoinositides are located in nucleoplasmic membraneless nuclear structures such as nuclear speckles or nucleoli [74,75,76]. Recently, researchers have started to explore the potential role of phosphoinositides in regulating nuclear processes. The most abundant phosphoinositide detected in these structures was PI(4,5)P2, which is thought to play a role in the spatial and functional organisation of the nucleus (Fig. 2) [75]. The cell nucleus possesses its own pool of phosphoinositidemetabolizing enzymes, which is distinct from the cytoplasmic pool and thus enables them to control their involvement in nuclear processes specifically [77,78,79].

Fig. 2
figure 2

Schematic illustration of the nuclear PI(4,5)P2 role in normal cells (A) and cancer cells (B). Figure (A) highlights the mechanism of the transcriptional regulation of Pol II in normal cells. The myosin phosphatase Rho-interacting protein (MPRIP) mediates the interaction between Pol II initiation condensate (Tyr1P-CTD) and PI(4,5)P2-containing structures which enables the release of RNAPII from the promoter-proximal pausing. Figure (B) highlights the implications of the altered nuclear PI(4,5)P2 levels in carcinogenesis

The expanding role of PI(4,5)P2 in nuclear architecture and organization

Previously, the dysregulation of the nuclear PI(4,5)P2 pool and its metabolism were overlooked, as the observed changes were more aligned with the total cellular PI(4,5)P2 levels. However, recent exploration of the nuclear PI(4,5)P2 has uncovered its role in the crucial nuclear processes, whose dysregulation is associated with various pathological conditions, including cancer [56, 80, 81]. These include the chromatin remodelling process mentioned above, as well as the acylation and deacylation of histones, RNA processing, DDR, regulation of Pol II transcript activity, premRNA splicing, and polyadenylation [19, 74, 82, 83]. For these processes to function properly, it is essential that appropriate proteins and nucleic acids are concentrated in a specific location. This can be achieved through the formation of biomolecular condensates that concentrate molecules and separate them from their surroundings [84]. The liquid-liquid phase separation (LLPS) phenomenon plays an essential role in this organisational process [85, 86]. The resulting condensates are usually spherical, dynamically coalesce, and undergo rapid fluorescence recovery after photobleaching, demonstrating a high degree of internal dynamics [87]. The hallmark of the LLPS process is multivalent weak inter and intramolecular interactions, where proteins undergoing LLPS are characterised by distinctive features, such as multiple structured modular domains as well as intrinsically disordered regions (IDRs) or oligomerisation domains [84]. Recently, PIPs have been suggested to be novel LLPS regulatory molecules [22, 74, 88]. The PI(4,5)P2 effector protein and Factin regulator, myosin phosphatase Rhointeracting protein (MPRIP), was identified as a component of the Pol II initiation condensate and as a transcription modulator with condensation capacity [89]. MPRIP recruits myosin 1 C (NM1)/Pol II complex via Pol II Tyr1PCTD to PI(4,5)P2containing structures [21]. Notably, a recent study by Dumilie et al. showed that PI(4,5)P2 is a key component of natural condensates and is enriched in mediator (MED1) condensates [22]. Together with BDR4, MED1 is a transcriptional cofactor found in superenhancers that can compartmentalise the Pol II transcriptional apparatus [90]. The BRD4MED1 SuperEnhancer condensates play crucial roles in boosting the transcription of protooncogenes, which cancer cells use to regulate cell proliferation and survival [91]. Thus, the abnormal LLPS process of PI(4,5)P2containing condensates may lead to the formation of pathological structures with abnormal physicochemical properties and trigger a malignant transcriptional program leading to disease, including cancer progression and metastasis. In addition, the crucial DDR pathway, which is critical for genome stability, may also be affected by abnormal LLPS and contribute to its dysregulation and, ultimately, to carcinogenesis. During the DDR response, the interaction of PI5P, a precursor of PI(4,5)P2, with an inhibitor of growth protein 2 (ING2) leads to the acylation of the tumour suppressor protein p53, thereby transactivating the p53dependent apoptotic pathway [16, 18].

A previously published study showed an increase in PI(4,5)P2 levels in HPV8induced skin tumour samples as well as in HPV16positive samples from lowgrade cervical intraepithelial neoplasia (CIN I) to invasive cervical cancer [92]. HPV E6 protein is known to induce ubiquitination and degradation of the p53 protein [93, 94], while PI(4,5)P2 is known to stabilise p53 [16]. An unknown epigenetic modification may be present in these cancers that protects p53 from degradation and simultaneously ensures cancer cell survival. The elevated nuclear PI(4,5)P2 levels observed in betaHPVpositive skin and HPV16positive cervical tumours highlight the clinical significance of these findings in HPVmediated tumorigenesis. These results established a novel link between HPV infection and nuclear PI(4,5)P2 metabolism. Given that elevated nuclear PI(4,5)P2 is detected as early as in CIN I lesions, HPVinfected cells represent a promising model for studying the role of PI(4,5)P2 in tumour progression and metastasis. The absence of elevated PI(4,5)P2 signals in other skin cancers, such as malignant melanoma and Merkel cell carcinoma, strongly indicates a specific effect on HPVassociated skin tumours [95]. The specificity of these signals warrants further investigation to determine whether similar molecular patterns exist in other cancers or are unique to HPVassociated cancers. The methodology developed by Hoboth et al. can be used for this purpose on paraffinembedded section patient material obtained from clinicians [96]. Several vital questions must be addressed in future research. First, it is important to investigate whether elevated nuclear PI(4,5)P2 levels are also associated with HPV16positive oropharyngeal squamous cell carcinoma (OPSCC) compared to nonHPVrelated OPSCC to determine whether the cancer origin and tumour grade affect the elevated nuclear PI(4,5)P2 levels. It also needs to be clarified whether an increase in nuclear PI(4,5)P2 levels is specific to oncogenic skin and mucosainfecting HPV types or if it is a characteristic of HPVinduced tumours or tumours in general. As mentioned above, PI(4,5)P2 is known to localise to nuclear speckles, nuclear lipid islets, and nucleoli [7]. The compartmentalisation of the nucleus and, thus, the existence of different pools of PI(4,5)P2 might allow local finetuning of specific nuclear processes [12]. Using superresolution threedimensional structured illumination microscopy, PI(4,5)P2 was found to localise to nuclear speckles in HPVpositive associated cancers, and the precise link between nuclear speckle function and nuclear PI(4,5)P2 remain elusive and needs to be determined. Furthermore, it should be investigated whether only nuclear PI(4,5)P2 levels are dysregulated in HPVinfected tissues or if other phosphoinositide species are also differentially regulated in HPVrelated tumours. An increase in PI(4,5)P2 may alter PI(3,4,5)P3 levels. However, it remains unclear which nuclear kinases or phosphatases are dysregulated in the presence of the virus. Further research is needed to clarify the mechanisms behind these potential changes in phosphoinositide levels and to understand their implications for the pathogenesis and progression of HPVrelated cancers. Additionally, exploring the interplay between these phosphoinositides may provide insight into early diagnosis, as nuclear PI(4,5)P2 levels changes are observed as early as CIN I or novel therapeutic targets for managing HPVassociated malignancies.

Concluding remarks and prospective directions

Phosphoinositides are fundamental lipids involved in various cellular processes, including the regulation of membrane dynamics and signalling cascades, as well as in nuclear processes such as chromatin organisation. Dysregulation of phosphoinositides, whether in the cytosolic or nuclear pools, can contribute to the progression of several diseases, particularly cancers. Dysregulation of the PI3K signalling pathway has been observed in various types of cancer, underscoring the potential of PI3K as a target for therapeutic intervention. While research involving therapeutic targeting of the PI3K pathway has progressed, the therapeutic efficacy of PI3K inhibitors continues to be challenged by their toxicity, acquired drug resistance, and offtarget activity. Currently, five small molecule PI3K inhibitors have been clinically approved by the U.S. Food and Drug Administration and the European Medicines Agency [97]. Nonetheless, advancement of techniques, such as highthroughput screening, have facilitated the design of novel smallmolecule inhibitors with higher specificity, selectivity, and potency. As a result, more than 60 novel smallmolecule PI3K inhibitors are currently undergoing clinical trials [97].

Recent data suggest that nuclear PI(4,5)P2 plays a crucial role in organising nuclear processes, including RNA processing, DDR, and regulation of Pol II transcript activity [21, 22, 83, 88, 98, 99]. Moreover, elevated nuclear PI(4,5)P2 levels appear to be linked to cancer progression [92]. Studies have also suggested that PI(4,5)P2 is a key component of LLPS biocondensates [22, 88]. However, given that the nucleoplasm is a membraneless environment, a significant question arises regarding the structural form of PI(4,5)P2 condensates. One possibility is that it forms a lipid structure, such as micelles, to hide its hydrophobic acyl chains from the unfavourable environment. Alternatively, the acyl groups of the PIP may integrate into protein condensate structures, forming hydrophobic cores. Another possible scenario is that PI(4,5)P2 may be covalently linked to certain proteins, as shown by Marx et al. [92]. Such conjugation is thought to induce a conformational change in the protein, resulting in the acyl chains being shielded from an aqueous environment. This type of conjugation would represent the novel posttranslational modification of unknown metabolic pathway. Finally, similar to SF-1 protein, certain nuclear proteins might possess hydrophobic binding pockets where they accommodate PI(4,5)P2 acyl chains, leaving the head exposed to the external environment [100]. Therefore, a deeper understanding of the metabolic pathways, structure and functions of nuclear PI(4,5)P2 may provide valuable insights into the mechanisms underlying cancer invasiveness and metastasis.

Table 1 Impact of four potential PI(4,5)P2 deregulation patterns in cancer cells on Cancer Progression and Metastasis

Signals originating from cytosolic PIPs may also influence nuclear events and vice versa. Some PIPs enzyme precursors are known to shuttle between the nucleus and cytoplasm, allowing for a degree of interconnected regulation [72]. In summary, there are four different patterns in which deregulation of the PIP pathway and its levels can influence cancer progression. The table lists the three with the most likely metastatic ability (high cytoplasmlow nucleus, low cytoplasmhigh nucleus, and high cytoplasmhigh nucleus) and the one with the least invasive ability (low cytoplasmlow nucleus), as it is most likely to contribute to cancer cell death (Table 1). Deciphering the detailed mechanisms of this complex and precise system will not only enhance our understanding of PIPs metabolism in cancers but may also uncover novel therapeutic targets for malignancies characterised by aberrant nuclear lipid signalling.

Data availability

No datasets were generated or analysed during the current study.

Abbreviations

ABD:

Adaptor-binding domain

CIN:

Cervical intraepithelial neoplasia

DDR:

DNA damage repair

ECM:

Extracellular matrix

HNSCC:

Head and neck squamous cell carcinomas

LLPS:

Liquidliquid phase separation

OPSCC:

Oropharyngeal squamous cell carcinoma

PI(4,5)P2:

Phosphatidylinositol 4,5-bisphosphate

PI3K:

Phosphoinositide 3-kinase

PIP4K:

Phosphatidylinositol 5-phosphate 4-kinase

PIP5K:

Phosphatidylinositol-4-phosphate 5-kinase

PIPs:

Phosphoinositides

PM:

Plasma membrane

PTEN:

Phosphatase and tensin homolog deleted on chromosome 10

RBD:

Ras-binding domain

SCC:

Squamous cell carcinomas

References

  1. WHO. Cancer. World Health Organization. [Internet]. www.who.int. 2022 [cited WH2024 Oct 16]. Available from: https://www.who.int/news-room/fact-sheets/detail/cancer, accessed: 16.05.2024.

  2. Fares J, Fares MY, Khachfe HH, Salhab HA, Fares Y. Molecular principles of metastasis: a hallmark of cancer revisited. Signal Transduct Target Ther. Springer Nature; 2020.

  3. Greenlee JD, Subramanian T, Liu K, King MR. Rafting Down the Metastatic Cascade: The Role of Lipid Rafts in Cancer Metastasis, Cell Death, and Clinical Outcomes. Cancer Res [Internet]. 2021;81:5–17. Available from: https://aacrjournals.publicaciones.saludcastillayleon.es/cancerres/article/81/1/5/649260/Rafting-Down-the-Metastatic-Cascade-The-Role-of

  4. Linder S, Cervero P, Eddy R, Condeelis J. Mechanisms and roles of podosomes and invadopodia. Nat Rev Mol Cell Biol [Internet]. 2023;24:86–106. Available from: http://www.ncbi.nlm.nih.gov/pubmed/36104625

  5. Levental I, Levental KR, Heberle FA. Lipid Rafts: Controversies Resolved, Mysteries Remain. Trends Cell Biol [Internet]. 2020;30:341–53. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0962892420300313

  6. Sezgin E, Levental I, Mayor S, Eggeling C. The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat Rev Mol Cell Biol [Internet]. 2017;18:361–74. Available from: https://www.nature.com/articles/nrm.2017.16

  7. Li S, Huang F, Xia T, Shi Y, Yue T. Phosphatidylinositol 4,5-Bisphosphate sensing lipid raft via Inter-leaflet Coupling regulated by acyl chain length of Sphingomyelin. Langmuir. 2023;39:5995–6005.

    Article  CAS  PubMed  Google Scholar 

  8. Myeong J, Park C-G, Suh B-C, Hille B. Compartmentalization of phosphatidylinositol 4,5-bisphosphate metabolism into plasma membrane liquid-ordered/raft domains. Proceedings of the National Academy of Sciences [Internet]. 2021;118. Available from: https://doiorg.publicaciones.saludcastillayleon.es/10.1073/pnas.2025343118

  9. Mandal K. Review of PIP2 in cellular signaling, functions and diseases. Int J Mol Sci MDPI AG; 2020. pp. 1–20.

  10. Mollinedo F, Gajate C. Lipid rafts as signaling hubs in cancer cell survival/death and invasion: Implications in tumor progression and therapy. J Lipid Res [Internet]. 2020;61:611–35. Available from: https://doiorg.publicaciones.saludcastillayleon.es/10.1194/jlr.TR119000439

  11. Singh I, Lele TP. Nuclear Morphological abnormalities in Cancer: a search for unifying mechanisms. Results Probl Cell Differ. Springer Science and Business Media Deutschland GmbH; 2022. pp. 443–67.

  12. Fischer EG, Karger AG. 2020. pp. 511–9.

  13. Chatterjee A, Rodger EJ, Eccles MR. Epigenetic drivers of tumourigenesis and cancer metastasis. Semin Cancer Biol. Academic Press; 2018. pp. 149–59.

  14. Geissler F, Nesic K, Kondrashova O, Dobrovic A, Swisher EM, Scott CL et al. The role of aberrant DNA methylation in cancer initiation and clinical impacts. Ther Adv Med Oncol. 2024;16.

  15. Hammond GRV, Burke JE. Novel roles of phosphoinositides in signaling, lipid transport, and disease. Curr Opin Cell Biol. Elsevier Ltd; 2020. pp. 57–67.

  16. Wang YH, Sheetz MP. When PIP2 meets p53: Nuclear Phosphoinositide Signaling in the DNA damage response. Front Cell Dev Biol. Frontiers Media S.A.; 2022.

  17. Deng S, Leong HC, Datta A, Gopal V, Kumar AP, Yap CT. PI3K/AKT Signaling Tips the Balance of Cytoskeletal forces for Cancer Progression. Cancers (Basel). MDPI; 2022.

  18. Hamann BL, Blind RD. Nuclear phosphoinositide regulation of chromatin. J Cell Physiol [Internet]. 2018;233:107–23. Available from: https://onlinelibrary.wiley.com/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1002/jcp.25886

  19. Castano E, Yildirim S, Fáberová V, Krausová A, Uličná L, Paprčková D, et al. Nuclear phosphoinositides—versatile regulators of genome functions. Cells. MDPI; 2019.

  20. Sztacho M, Sobol M, Balaban C, Escudeiro Lopes SE, Hozák P. Nuclear phosphoinositides and phase separation: Important players in nuclear compartmentalization. Adv Biol Regul [Internet]. 2019;71:111–7. Available from: https://doiorg.publicaciones.saludcastillayleon.es/10.1016/j.jbior.2018.09.009

  21. Balaban C, Sztacho M, Antiga L, Miladinović A, Harata M, Hozák P. PIP2-Effector protein MPRIP regulates RNA polymerase II condensation and transcription. Biomolecules. 2023;13.

  22. Dumelie JG, Chen Q, Miller D, Attarwala N, Gross SS, Jaffrey SR. Biomolecular condensates create phospholipid-enriched microenvironments. Nat Chem Biol. 2024;20:302–13.

    Article  CAS  PubMed  Google Scholar 

  23. Coskun Ü, Simons K. Cell Membranes: The Lipid Perspective. Structure [Internet]. 2011 [cited 2023 Jan 28];19:1543–8. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0969212611003649

  24. Holthuis JCM, Menon AK. Lipid landscapes and pipelines in membrane homeostasis. Nature. 2014;510:48–57.

    Article  CAS  PubMed  Google Scholar 

  25. Varshney P, Yadav V, Saini N. Lipid rafts in immune signalling: current progress and future perspective. Immunology. 2016;149:13–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Raghunathan K, Kenworthy AK. Dynamic pattern generation in cell membranes: current insights into membrane organization. Biochim Biophys Acta Biomembr. 2018;1860:2018–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Biernatowska A, Wójtowicz K, Trombik T, Sikorski AF, Czogalla A. MPP1 determines the mobility of Flotillins and controls the confinement of raft-Associated molecules. Cells. 2022;11:1–13.

    Article  Google Scholar 

  28. Yamaguchi H, Oikawa T. Membrane lipids in invadopodia and podosomes: key structures for cancer invasion and metastasis. Oncotarget [Internet]. 2010;1:320–8. Available from: http://www.ncbi.nlm.nih.gov/pubmed/21307399

  29. Tahir SA, Park S, Thompson TC. Caveolin-1 regulates VEGF-stimulated angiogenic activities in prostate cancer and endothelial cells. Cancer Biol Ther [Internet]. 2009;8:2284–94. Available from: http://www.tandfonline.com/doi/abs/https://doiorg.publicaciones.saludcastillayleon.es/10.4161/cbt.8.23.10138

  30. Li Q, Peng J, Li X, Leng A, Liu T. miR-449a targets Flot2 and inhibits gastric cancer invasion by inhibiting TGF-β-mediated EMT. Diagn Pathol. 2015;10.

  31. Wang X, Zhang Y, Zhao Y, Liang Y, Xiang C, Zhou H et al. CD24 promoted cancer cell angiogenesis via Hsp90-mediated STAT3/VEGF signaling pathway in colorectal cancer. Oncotarget [Internet]. 2016;7:55663–76. Available from: https://www.oncotarget.com/lookup/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.18632/oncotarget.10971

  32. Dickson EJ, Hille B. Understanding phosphoinositides: rare, dynamic, and essential membrane phospholipids. Biochemical Journal. Portland Press Ltd; 2019. pp. 1–23.

  33. Singh N, Reyes-Ordoñez A, Compagnone MA, Moreno JF, Leslie BJ, Ha T et al. Redefining the specificity of phosphoinositide-binding by human PH domain-containing proteins. Nat Commun. 2021;12.

  34. Pemberton JG, Balla T. Polyphosphoinositide-Binding Domains: Insights from Peripheral Membrane and Lipid-Transfer Proteins. Adv Exp Med Biol. Springer; 2019. pp. 77–137.

  35. Viaud J, Mansour R, Antkowiak A, Mujalli A, Valet C, Chicanne G, et al. Phosphoinositides: important lipids in the coordination of cell dynamics. Biochimie. Elsevier B.V.; 2016. pp. 250–8.

  36. Janmey PA, Bucki R, Radhakrishnan R. Regulation of actin assembly by PI(4,5)P2 and other inositol phospholipids: An update on possible mechanisms. Biochem Biophys Res Commun. Elsevier B.V.; 2018. pp. 307–14.

  37. Wills RC, Hammond GRV. PI(4,5)P2: signaling the plasma membrane. Biochemical Journal. Portland Press Ltd; 2022. pp. 2311–25.

  38. Mansat M, Kpotor AO, Chicanne G, Picot M, Mazars A, Flores-Flores R et al. MTM1-mediated production of phosphatidylinositol 5-phosphate fuels the formation of podosome-like protrusions regulating myoblast fusion. Proc Natl Acad Sci U S A. 2024;121.

  39. Obeng EO, Rusciano I, Marvi MV, Fazio A, Ratti S, Follo MY et al. Phosphoinositide-dependent signaling in cancer: a focus on phospholipase C isozymes. Int J Mol Sci. 2020;21.

  40. Posor Y, Jang W, Haucke V. Phosphoinositides as membrane organizers. Nat Rev Mol Cell Biol Nat Res; 2022. pp. 797–816.

  41. González-García A, Garrido A, Carrera AC. Targeting PTEN Regulation by Post translational modifications. Cancers (Basel). MDPI; 2022.

  42. Bryant JM, Blind RD. Signaling through non-membrane nuclear phosphoinositide binding proteins in human health and disease. J Lipid Res. American Society for Biochemistry and Molecular Biology Inc.; 2019. pp. 299–311.

  43. Luongo F, Colonna F, Calapà F, Vitale S, Fiori ME, De Maria R, Cancers. (Basel). MDPI AG; 2019.

  44. Lawrence MS, Stojanov P, Mermel CH, Robinson JT, Garraway LA, Golub TR, et al. Discovery and saturation analysis of cancer genes across 21 tumour types. Nature. 2014;505:495–501.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Luo J, Field SJ, Lee JY, Engelman JA, Cantley LC. The p85 regulatory subunit of phosphoinositide 3-kinase down-regulates IRS-1 signaling via the formation of a sequestration complex. J Cell Biol. 2005;170:455–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Cuevas BD, Lu Y, Mao M, Zhang J, LaPushin R, Siminovitch K, et al. Tyrosine phosphorylation of p85 relieves its inhibitory activity on Phosphatidylinositol 3-Kinase. J Biol Chem. 2001;276:27455–61.

    Article  CAS  PubMed  Google Scholar 

  47. Lu Y, Yu Q, Liu JH, Zhang J, Wang H, Koul D, et al. Src family protein-tyrosine kinases alter the function of PTEN to regulate phosphatidylinositol 3-kinase/AKT cascades. J Biol Chem. 2003;278:40057–66.

    Article  CAS  PubMed  Google Scholar 

  48. Pelaz SG, Tabernero A. Src: coordinating metabolism in cancer. Oncogene. Springer Nature; 2022. pp. 4917–28.

  49. Koudelková L, Pelantová M, Brůhová Z, Sztacho M, Pavlík V, Pánek D et al. Phosphorylation of tyrosine 90 in SH3 domain is a new regulatory switch controlling src kinase. Elife. 2023;12.

  50. Irby RB, Yeatman TJ. Role of Src expression and activation in human cancer. Oncogene [Internet]. 2000;19:5636–42. Available from: http://www.ncbi.nlm.nih.gov/pubmed/11114744

  51. Glaviano A, Foo ASC, Lam HY, Yap KCH, Jacot W, Jones RH, et al. PI3K/AKT/mTOR signaling transduction pathway and targeted therapies in cancer. Mol Cancer. BioMed Central Ltd; 2023.

  52. Klussmann JP, Mooren JJ, Lehnen M, Claessen SMH, Stenner M, Huebbers CU, et al. Genetic signatures of HPV-related and unrelated oropharyngeal carcinoma and their prognostic implications. Clin Cancer Res. 2009;15:1779–86.

    Article  CAS  PubMed  Google Scholar 

  53. Leemans CR, Snijders PJF, Brakenhoff RH. The molecular landscape of head and neck cancer. Nat Rev Cancer Nat Publishing Group; 2018. pp. 269–82.

  54. Lawrence MS, Sougnez C, Lichtenstein L, Cibulskis K, Lander E, Gabriel SB, et al. Comprehensive genomic characterization of head and neck squamous cell carcinomas. Nature. 2015;517:576–82.

    Article  CAS  Google Scholar 

  55. Cochicho D, Esteves S, Rito M, Silva F, Martins L, Montalvão P et al. PIK3CA gene mutations in HNSCC: systematic review and correlations with HPV Status and Patient Survival. Cancers (Basel). 2022;14.

  56. Davis WJ, Lehmann PZ, Li W. Nuclear PI3K signaling in cell growth and tumorigenesis. Front Cell Dev Biol. Frontiers Media S.A.; 2015.

  57. Xu J, Huang Y, Zhao J, Wu L, Qi Q, Liu Y, et al. Cofilin: a promising protein implicated in Cancer Metastasis and apoptosis. Front Cell Dev Biol. Frontiers Media S.A.; 2021.

  58. Augoff K, Hryniewicz-Jankowska A, Tabola R. Invadopodia: clearing the way for cancer cell invasion. Ann Transl Med. 2020;8:902–902.

    Article  PubMed  PubMed Central  Google Scholar 

  59. Yamaguchi H, Takeo Y, Yoshida S, Kouchi Z, Nakamura Y, Fukami K. Lipid rafts and caveolin-1 are required for invadopodia formation and extracellular matrix degradation by human breast cancer cells. Cancer Res. 2009;69:8594–602.

    Article  CAS  PubMed  Google Scholar 

  60. Yang H, Guan L, Li S, Jiang Y, Xiong N, Li L et al. Mechanosensitive caveolin-1 activation-induced PI3K/Akt/mTOR signaling pathway promotes breast cancer motility, invadopodia formation and metastasis in vivo [Internet]. Oncotarget. 2016. Available from: www.impactjournals.com/oncotarget/

  61. Huang Y, Zhang S, Park JI. Nuclear actin dynamics in Gene expression, DNA repair, and Cancer. Results Probl Cell Differ. Springer Science and Business Media Deutschland GmbH; 2022. pp. 625–63.

  62. Plessner M, Melak M, Chinchilla P, Baarlink C, Grosse R. Nuclear F-actin formation and reorganization upon cell spreading. J Biol Chem. 2015;290:11209–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Ulferts S, Prajapati B, Grosse R, Vartiainen MK. Emerging properties and functions of actin and actin filaments inside the nucleus. Cold Spring Harb Perspect Biol. 2021;13:1–16.

    Article  Google Scholar 

  64. Serebryannyy LA, Parilla M, Annibale P, Cruz CM, Laster K, Gratton E, et al. Persistent nuclear actin filaments inhibit transcription by RNA polymerase II. J Cell Sci. 2016;129:3412–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Rando OJ, Zhao K, Janmey P, Crabtree GR. Phosphatidylinositol-dependent actin filament binding by the SWISNF-like BAF chromatin remodeling complex [Internet]. 2001. Available from: https://www.pnas.org

  66. Alfert A, Moreno N, Kerl K. The BAF complex in development and disease. Epigenetics Chromatin [Internet]. 2019;12:19. Available from: http://www.ncbi.nlm.nih.gov/pubmed/30898143

  67. Philimonenko VV, Zhao J, Iben S, Dingová H, Kyselá K, Kahle M et al. Nuclear actin and myosin I are required for RNA polymerase I transcription. Nat Cell Biol [Internet]. 2004;6:1165–72. Available from: http://www.ncbi.nlm.nih.gov/pubmed/15558034

  68. Hofmann WA, Stojiljkovic L, Fuchsova B, Vargas GM, Mavrommatis E, Philimonenko V, et al. Actin is part of pre-initiation complexes and is necessary for transcription by RNA polymerase II. Nat Cell Biol. 2004;6:1094–101.

    Article  CAS  PubMed  Google Scholar 

  69. Hu P, Wu S, Hernandez N. A role for β-actin in RNA polymerase III transcription. Genes Dev. 2004;18:3010–5.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Qi T, Tang W, Wang L, Zhai L, Guo L, Zeng X. G-actin participates in RNA polymerase II-dependent transcription elongation by recruiting positive transcription elongation factor b (P-TEFb). J Biol Chem. 2011;286:15171–81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Wei M, Fan X, Ding M, Li R, Shao S, Hou Y et al. Nuclear actin regulates inducible transcription by enhancing RNA polymerase II clustering. Sci Adv [Internet]. 2020;6:6515. Available from: http://www.ncbi.nlm.nih.gov/pubmed/32494599

  72. Schramp M, Hedman A, Li W, Tan X, Anderson R. PIP kinases from the cell membrane to the nucleus. Subcell Biochem. 2015;58:25–59.

    Article  Google Scholar 

  73. Jungmichel S, Sylvestersen KB, Choudhary C, Nguyen S, Mann M, Nielsen ML. Specificity and commonality of the phosphoinositide-binding proteome analyzed by quantitative Mass Spectrometry. Cell Rep. 2014;6:578–91.

    Article  CAS  PubMed  Google Scholar 

  74. Sztacho M, Sobol M, Balaban C, Escudeiro Lopes SE, Hozák P. Nuclear phosphoinositides and phase separation: important players in nuclear compartmentalization. Adv Biol Regul. Elsevier Ltd; 2019. pp. 111–7.

  75. Vidalle MC, Sheth B, Fazio A, Marvi MV, Leto S, Koufi FD, et al. Nuclear Phosphoinositides as Key determinants of Nuclear functions. Biomolecules. Multidisciplinary Digital Publishing Institute (MDPI); 2023.

  76. Sztacho M, Šalovská B, Červenka J, Balaban C, Hoboth P, Hozák P. Limited proteolysis-coupled mass spectrometry identifies phosphatidylinositol 4,5-bisphosphate effectors in human nuclear proteome. Cells. 2021;10:1–16.

    Article  Google Scholar 

  77. Cocco L, Martelli AM, Barnabei O, Manzoli FA. Nuclear inositol lipid signaling. Adv Enzyme Regul [Internet]. 2001;41:361–84. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0065257100000170

  78. Irvine RF. Nuclear lipid signalling. Nat Rev Mol Cell Biol. 2003;4:349–60.

    Article  CAS  PubMed  Google Scholar 

  79. Ratti S, Ramazzotti G, Faenza I, Fiume R, Mongiorgi S, Billi AM et al. Nuclear inositide signaling and cell cycle. Adv Biol Regul [Internet]. 2018;67:1–6. Available from: https://linkinghub.elsevier.com/retrieve/pii/S2212492617301732

  80. Casalin I, Ceneri E, Ratti S, Manzoli L, Cocco L, Follo MY. Nuclear Phospholipids and Signaling: An Update of the Story. Cells [Internet]. 2024;13:713. Available from: https://www.mdpi.com/2073-4409/13/8/713

  81. Fiume R, Stijf-Bultsma Y, Shah ZH, Keune WJ, Jones DR, Jude JG, et al. PIP4K and the role of nuclear phosphoinositides in tumour suppression. Biochim Biophys Acta Mol Cell Biol Lipids. Elsevier B.V.; 2015. pp. 898–910.

  82. Shah ZH, Jones DR, Sommer L, Foulger R, Bultsma Y, D’Santos C et al. Nuclear phosphoinositides and their impact on nuclear functions. FEBS J. 2013. pp. 6295–310.

  83. Hoboth P, Sztacho M, Hozák P. Nuclear patterns of phosphatidylinositol 4,5- and 3,4‐bisphosphate revealed by super‐resolution microscopy differ between the consecutive stages of RNA polymerase II transcription. FEBS J [Internet]. 2024; Available from: https://febs.onlinelibrary.wiley.com/doi/https://doiorg.publicaciones.saludcastillayleon.es/10.1111/febs.17136

  84. Sabari BR, Dall’Agnese A, Young RA. Biomolecular Condensates in the Nucleus. Trends Biochem Sci [Internet]. 2020;45:961–77. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0968000420301675

  85. Povarova OI, Antifeeva IA, Fonin AV, Turoverov KK, Kuznetsova IM. The role of liquid–liquid phase separation in actin polymerization. Int J Mol Sci MDPI; 2023.

  86. Weirich KL, Banerjee S, Dasbiswas K, Witten TA, Vaikuntanathan S, Gardel ML. Liquid behavior of cross-linked actin bundles. Proc Natl Acad Sci U S A. 2017;114:2131–6.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Alberti S, Saha S, Woodruff JB, Franzmann TM, Wang J, Hyman AA. A user’s guide for phase separation assays with purified proteins. J Mol Biol. 2018;430:4806–20.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Sztacho M, Červenka J, Šalovská B, Antiga L, Hoboth P, Hozák P. The RNA-dependent association of phosphatidylinositol 4,5-bisphosphate with intrinsically disordered proteins contribute to nuclear compartmentalization. Papenfort K, editor. PLoS Genet [Internet]. 2024;20:e1011462. Available from: https://doiorg.publicaciones.saludcastillayleon.es/10.1371/journal.pgen.1011462

  89. Balaban C, Sztacho M, Blažíková M, Hozák P. The F-Actin-binding MPRIP forms phase-separated condensates and associates with PI(4,5)P2 and active RNA polymerase II in the cell nucleus. Cells. 2021;10.

  90. Sabari BR, Dall’Agnese A, Boija A, Klein IA, Coffey EL, Shrinivas K et al. Coactivator condensation at super-enhancers links phase separation and gene control. Science (1979). 2018;361.

  91. Qian H, Zhu M, Tan X, Zhang Y, Liu X, Yang L. Super-enhancers and the super-enhancer reader BRD4: tumorigenic factors and therapeutic targets. Cell Death Discov. 2023;9.

  92. Marx B, Hufbauer M, Zigrino P, Majewski S, Markiefka B, Sachsenheimer T et al. Phospholipidation of nuclear proteins by the human papillomavirus E6 oncoprotein: implication in carcinogenesis [Internet]. Oncotarget. 2018. Available from: www.oncotarget.com

  93. Akgül B, Karle P, Adam M, Fuchs PG, Pfister HJ. Dual role of tumor suppressor p53 in regulation of DNA replication and oncogene E6-promoter activity of Epidermodysplasia Verruciformis-associated human papillomavirus type 8. Virology. 2003;308:279–90.

    Article  PubMed  Google Scholar 

  94. Lazić D, Hufbauer M, Zigrino P, Buchholz S, Kazem S, Feltkamp MCW, et al. Human papillomavirus type 8 E6 oncoprotein inhibits transcription of the PDZ protein Syntenin-2. J Virol. 2012;86:7943–52.

    Article  PubMed  PubMed Central  Google Scholar 

  95. Hoboth P, Sztacho M, Quaas A, Akgül B, Hozák P. Quantitative super-resolution microscopy reveals the differences in the nanoscale distribution of nuclear phosphatidylinositol 4,5-bisphosphate in human healthy skin and skin warts. Front Cell Dev Biol. 2023;11.

  96. Hoboth P, Šebesta O, Sztacho M, Castano E, Hozák P. Dual-color dSTORM imaging and ThunderSTORM image reconstruction and analysis to study the spatial organization of the nuclear phosphatidylinositol phosphates. MethodsX. 2021;8.

  97. Li H, Wen X, Ren Y, Fan Z, Zhang J, He G, et al. Targeting PI3K family with small-molecule inhibitors in cancer therapy: current clinical status and future directions. Mol Cancer. BioMed Central Ltd; 2024.

  98. Miladinović A, Antiga L, Venit T, Bayona-Hernandez A, Červenka J, Labala RK et al. The perinucleolar compartment and the oncogenic super-enhancers are part of the same phase-separated structure filled with phosphatidylinositol 4,5bisphosphate and long noncoding RNA HANR. Adv Biol Regul. 2024.

  99. Wang YH, Hariharan A, Bastianello G, Toyama Y, Shivashankar GV, Foiani M et al. DNA damage causes rapid accumulation of phosphoinositides for ATR signaling. Nat Commun. 2017;8.

  100. Blind RD, Sablin EP, Kuchenbecker KM, Chiu HJ, Deacon AM, Das D, et al. The signaling phospholipid PIP3 creates a new interaction surface on the nuclear receptor SF-1. Proc Natl Acad Sci U S A. 2014;111:15054–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Institute for Cancer Research (NPO EXCELES, ID Project No. LX22NPO5102), Charles University Cooperatio Program: research area “Oncology and Haematology”, Fond F Program EXCELENCE1–223011/1LF and the German Research Foundation (AK 42/10 − 1).

Author information

Authors and Affiliations

Authors

Contributions

AC and MS conceptualized the review manuscript. AC prepared the figures and drafted the manuscript. AC, SR, BA and MS wrote and reviewed the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Martin Sztacho.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chytła, A., Rattay, S., Akgül, B. et al. Plasma membrane and nuclear phosphatidylinositol 4,5-bisphosphate signalling in cancer. Lipids Health Dis 24, 39 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12944-025-02452-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12944-025-02452-6

Keywords