Startseite Class IA PI3K isoforms lead to differential signalling downstream of PKB/Akt
Artikel Open Access

Class IA PI3K isoforms lead to differential signalling downstream of PKB/Akt

  • Hazal B. Catalak Yilmaz ORCID logo , Mahnoor Sulaiman ORCID logo , Ozlem Aybuke Isik ORCID logo und Onur Cizmecioglu ORCID logo EMAIL logo
Veröffentlicht/Copyright: 20. Dezember 2023

Abstract

Objectives

The catalytic subunits of Class IA PI3K, p110α, p110β, and p110δ, phosphorylates phosphatidylinositol 4,5-bisphosphate (PIP2) into phosphatidylinositol 3,4,5-trisphosphate (PIP3) on the plasma membrane. In cancer, these catalytic subunits are usually found to be altered or amplified. Because pan-PI3K inhibition results in systemic toxicities, finding specific targets for the ubiquitous PI3K isoforms offers considerable potential for enhancing the effectiveness of PI3K-targeted therapy.

Methods

We aim to delineate the isoform-specific druggable targets of the PI3K by deleting PIK3CA (encoding p110α) and PIK3CB (encoding p110β) by Cre mediated excision and ectopically expressing p110α, p110β, or p110δ with or without myristoylation (Myr) tag in mouse embryonic fibroblasts (MEFs). Myr is a lipidation signal that translocates proteins to plasma membrane permanently. This translocation renders p110s constitutively activated as they remain in close proximity to PIP2 on the membrane.

Results

Unique and redundant Akt targets are identified downstream of different PI3K isoforms. mTORC1, one of the targets of fully-activated Akt, has been observed to be differentially regulated in MEFs upon expression of p110α or p110β. The varying dependencies on mTORC1 and Rac1 led us to analyse a potential scaffolding function of p110β with Rac1 to mediate phosphorylation and activation of mTOR using platforms for the modeling of biomolecular complexes. We also documented that p110α and p110β support cell cycle kinetics differentially.

Conclusions

This study suggests differential regulation of protein translation, metabolism, cell cycle, and survival signaling downstream of unique p110 targets, underlying the importance of cancer treatment according to the deregulated p110 isoform.

Introduction

Class I Phosphoinositide 3-Kinases (PI3Ks) are lipid kinases which catalyze the phosphorylation of PIP2 (phosphatidylinositol 3,4-bisphosphate) into PIP3 (phosphatidylinositol 3,4,5-trisphosphate) on the plasma membrane. p110α, β, and δ are the catalytic subunits of Class IA PI3Ks, and they form obligatory couples with regulatory subunits of the same class that stabilize and inhibit catalytic subunits. Although p110α and p110β are expressed ubiquitously, p110δ is specific to the hematopoietic cells [1]. PTEN (Phosphatase and Tensin Homolog) is the lipid phosphatase removing 3′-phosphate of PIP3, and it is considered a tumor suppressor whose deletion is a common occurrence in cancer [2, 3].

p110 isoforms differ by their selection of membrane microdomain localization, interaction partners, and different modes of interaction [4, 5]. The plasma membrane is a laterally compartmentalized platform composed of different domains based on their lipid and protein constituents, where cellular signals are initiated and regulated [4, 6]. p110α has generally been restricted to nonraft regions, although p110β may also be detected in lipid rafts. p110α is primarily active downstream of receptor tyrosine kinases (RTKs), p110β is activated downstream of G-protein coupled receptors (GPCRs) [4, 6]. Of note, a number of RTKs and GPCRs localize selectively to raft or nonraft membrane areas. Different interaction partners facilitate the localization of p110s to different membrane domains. p110α is localized to nonraft regions through its interaction with Ras; on the other hand, p110β interacts with Rac1 or Cdc42 to be localized downstream of GPCRs. p110δ also interacts with Ras, but as it has been reported to be activated downstream of both RTKs and GPCRs, it is portrayed as the intermediate isoform [4, 7].

Upon receptor activation, PI3Ks are translocated to the plasma membrane and they catalyze PIP3 phosphorylation. This leads to the activation of AKT [8]. The main AKT isoform AKT1, is vital for survival, migration, and polarization, while AKT2 functions to regulate glucose homeostasis. Different AKT isoforms have been observed to be upregulated in varying cancer subtypes [9].

Upon activation, AKT phosphorylates mTORC1 component PRAS40, leading to the release of PRAS40’s inhibition on mTORC1, thus activating mTOR. mTOR mainly exists in two complexes, designated as mTORC1 and mTORC2. They are distinguished by their short-term sensitivity and insensitivity to Rapamycin, respectively. mTORC2 is activated upon PI3K signaling, however mTORC1 can be activated upon amino acids, low energy levels, hypoxia, and growth factor signals [10]. Following activation of these primary PI3K signal transducers, downstream effectors such as 4E-BP1, S6K, SGK, and ULK1 are activated, which play important roles in survival, ribosome synthesis, translation, autophagy and metabolism [10, 11].

In order to delineate the isoform-specific actions of class IA p110 isoforms, we utilized the myristoylation (Myr) tag to constitutively localize and activate them at the plasma membrane [3]. Our results indicate that different p110 isoforms promote varying levels of AKT and mTOR phosphorylations. Moreover, we identified a possible scaffolding function for p110β on mTOR autophosphorylation and modelled this possibility by docking simulations. In addition, different p110 isoforms promote varying levels of Src and Stat3 phosphorylation. We show that p110β expressing cells have higher tyrosine-phosphorylated Src, depicting elevated growth signaling [12, 13]. To further investigate the varying phosphorylation patterns among growth inducing effectors, we analyzed cell cycle kinetics in overexpression models of p110α and p110β. We observed that p110β overexpressing cells are more sensitive to genotoxic stress and Src inhibition. Overall, this work sheds light on the various functions provided by the p110 isoforms as well as the consequences of their activation.

Materials and methods

Maintenance and generation of cell lines

Generation of p110α/p110β floxed MEFs and retroviral transductions were performed as described before [4]. MEFs are transduced with retroviral pBABE constructs with puromycin, neomycin and blasticidin resistance. Transduced cells were treated with adenoviruses carrying Cre-recombinase to generate PIK3CA and PIK3CB knock-outs. MEFs depicted as wild-type (WT MEFs) are untreated p110α/p110β floxed cells. All of the MEFs were grown in 8 % FBS 1 % Pen/Strep high glucose DMEM.

2D growth assay

A total of 4,000 cells were seeded to each well of 12-well plates in triplicates. When DMSO control wells are at least 80 % confluent, the plates were fixed with 10 % acetic acid 10 % ethanol in dH2O. The wells are stained with crystal violet stain. The staining is quantified after destaining them with 100 mL acetic acid, 900 mL dH2O and transferred to a 96-well plate for measurements. The quantification is performed with a microplate reader at 592 nm.

Inhibitors BYL719, KIN193, CAL101, Rapamycin, Dasatinib and EHT1864 were all from Selleckchem. Cisplatin and doxorubicin were purchased from Koçak Farma and Deva Holding.

Western blot

The cells were scraped on ice, and pellets were lysed with RIPA buffer treated with phosphatase and protease inhibitor cocktails. Nitrocellulose membranes (GE Healthcare Life Sciences) were used. To obtain total protein levels, membranes previously probed with phosphoprotein antibodies were stripped with Millipore Membrane Stripping Solution, and reprobed with total protein antibodies. All antibodies used in the study were purchased from CST.

Cell cycle analysis with PI staining

Tyrpsinized cells were fixed with 70 % ethanol in dH2O on ice for 3 h. Ethanol was removed with subsequent ice-cold PBS washing steps. The cells were resuspended in 50 µL 100 μg/mL RNaseA (Sigma-Aldrich), and incubated at 37 °C for 15 min. The cells were resuspended in 200 µL 50 μg/mL propidium iodide (Sigma-Aldrich), and incubated in dark for 1 h on ice. The cell clumps were removed by using a strainer and the cells were counted and gated with CytoFlex (Beckman Coulter).

Docking experiments with HADDOCK

The following protein models were used for docking simulations obtained from Protein Data Bank (PDB):

Model ID Used molecule part Organism
2Y3A p110β and p85α icSH2 Mouse
4OVU p110α and p85α niSH2 Human
2RMK Rac1 Human
5H64 mTORC1 Human

The unknown residues were filled with FASTA sequences from UniProt. Default HADDOCK parameters were used. Active residues were selected using existing literature [1314].

For mTORC1 docking experiments, the models were aligned to the crystal structure (6BCU) from reference [15] on PyMol and possible residues that may partake in mTORC1-Rac1-p110α and p110β complexes were identified.

Statistical analysis and band intensity quantifications

The experiments were performed as triplicates and the graphs show mean±standard deviation. The Student’s t-test was used to analyze the significance, and p value less than 0.05 was considered significant. Quantification of band intensities was performed with ImageJ (NIH, Bethesda, MD).

Results

MEFs expressing unique p110 isoforms display distinct levels of Akt1 S473 phosphorylation

To investigate isoform-specific roles of class IA p110 catalytic subunits, we used MEFs whose first exons of p110α and p110β were floxed [4]. We ectopically expressed myristoylation (Myr)-tagged p110α, p110β and p110δ in these MEFs, and using an adenoviral Cre-recombinase; we deleted the endogenous PIK3CA and PIK3CB so that these MEFs would be expressing only the activated exogenous constructs. Wild-type p110α/β floxed MEFs were used as controls. p110α/β flox background is depicted as α, β +/+ throughout the manuscript.

First we checked the levels of the exogenously expressed p110 proteins. p110α, p110β and p110δ (a minor PI3K isoform in MEFs) were all expressed at comparable levels (Figure 1A). To demonstrate the validity of our cellular models, we determined the relative sensitivity of our cell lines to isoform-specific PI3K inhibitors in 2D-growth assays. BYL719 specifically targets p110α, whereas KIN193 and CAL101 inhibit p110β, and p110δ respectively [16, 17]. BYL719 almost completely blocks the growth of myr-p110α MEFs, and KIN193 inhibits myr-p110β MEFs significantly (Figure 1B). In line with previous findings [18], BYL719 at the doses used can also block the growth of myr-p110δ MEFs as well as CAL101. To investigate whether activated p110s coordinate isoform-specific roles in MEFs, we examined the downstream consequences of activated PI3K signalling. PI3K activity leads to Akt phosphorylation on Thr308, and Ser473, leading to its full activation [8]. Thus, Akt Ser473 phosphorylation indicates the activated Akt. We employed WT and myr-p110 MEFs and used a phospho-specific antibody for Akt1 Ser473 to highlight the activation profile of the major AKT isoform in MEFs. We observed different Akt1 Ser473 phosphorylation levels among these cell lines, the highest among them being myr-p110β MEFs (Figure 1C). Interestingly, the phospho-Akt1 level of myr-p110δ MEFs was significantly low, comparable to the WT controls. Then, we asked whether phosphorylated Akt1 effectively relays the signal to the downstream components in these MEFs with antibodies detecting phosphorylated Akt substrates (Figure 1D). Of note, the most abundant phosphorylations were observed on proteins around 250 kDa, 75 kDa, and 37–50 kDa sizes in myr-p110β MEFs. As expected, the phospho-AKT substrate signatures of myr-p110δ and myr-p110α MEFs were not as prominent as their myr-p110β counterparts. Overall, MEFs that express various activated p110s exhibit differential activation of Akt1 and Akt substrate phosphorylation, suggestive of isoform-specific functions.

Figure 1: 
Ectopic expression of activated p110β leads to a higher level of Akt activation in comparison to other Class IA PI3Ks in MEFs. (A) Relative protein levels of myristoylated p110s and WT cell lines. Each well contains 40 µg of protein lysate. Samples were probed with the indicated antibodies in an immunoblot. GAPDH is used as loading control. (B) Crystal violet stained plates were read at OD 592 nm to infer cellular growth (n=3). The DMSO controls of each cell line is normalized to 100 %, and the remaining reads adjusted to that. Averages were shown with SEM. Statistical significance was checked with Student’s t-test. *: p≤0.05, **: p≤0.01. (C) Proteins were harvested from cells grown to subconfluency. Each well of SDS – PAGE contains 30 µg protein lysate. Indicated antibodies were used in an immunoblot, GAPDH serves as loading control. Band intensity quantifications of the GAPDH normalized signals are indicated. (D) Proteins were harvested from exponentially growing cells. Each well contains 30 µg protein lysate. The approximated molecular weight values are noted at the right hand side of the blot. In the upper panel, phospho-AKT substrate antibody was used to assess level of AKT activity and GAPDH serves as loading control.
Figure 1:

Ectopic expression of activated p110β leads to a higher level of Akt activation in comparison to other Class IA PI3Ks in MEFs. (A) Relative protein levels of myristoylated p110s and WT cell lines. Each well contains 40 µg of protein lysate. Samples were probed with the indicated antibodies in an immunoblot. GAPDH is used as loading control. (B) Crystal violet stained plates were read at OD 592 nm to infer cellular growth (n=3). The DMSO controls of each cell line is normalized to 100 %, and the remaining reads adjusted to that. Averages were shown with SEM. Statistical significance was checked with Student’s t-test. *: p≤0.05, **: p≤0.01. (C) Proteins were harvested from cells grown to subconfluency. Each well of SDS – PAGE contains 30 µg protein lysate. Indicated antibodies were used in an immunoblot, GAPDH serves as loading control. Band intensity quantifications of the GAPDH normalized signals are indicated. (D) Proteins were harvested from exponentially growing cells. Each well contains 30 µg protein lysate. The approximated molecular weight values are noted at the right hand side of the blot. In the upper panel, phospho-AKT substrate antibody was used to assess level of AKT activity and GAPDH serves as loading control.

Activated p110δ relays phospho-tyrosine signals through JAK/STAT pathway

As myr-p110δ MEFs did not activate the Akt arm of the signaling as much as their p110β counterparts, we hypothesized that p110δ may be transmitting the signals via the tyrosine phosphorylation cascades [19, 20]. We analyzed the tyrosine phosphorylation signatures of myr-p110 MEFs. We observed two distinctive increments in tyrosine phosphorylation: at 130 kDa and at 60 kDa, in the myr-p110δ and the myr-p110β lanes, respectively (Figure 2A). As p110δ is functional mostly in immunological cells and JAK1/2/3 are around 130 kDa, we suspected that pattern might represent activated JAK/STAT signaling [6, 21]. Thus we investigated the phosphorylation of an essential downstream regulator of JAK/STAT signaling, STAT3 [22] (Figure 2B). STAT3 phosphorylation was highly detectable in myr-p110δ MEFs. Previously, it has been stated that Src becomes phosphorylated on a scaffold in the presence of p110β [13]. We wanted to see if the 60 kDa phosphoprotein could indeed be Src because it is prominent in fibroblasts. To corroborate this, we analyzed phospho-Src amongst control, myr-p110α, and myr-p110β expressing MEFs (Figure 2C). Phospho-Src levels were found to be high in p110β but not in p110α-expressing cells. Since the phosphorylated Src is expected to be activated, we investigated whether these results translated to varying levels of Src dependency in myr-p110α and myr-p110β MEFs in 2D-growth assays with a selective Src inhibitor, Dasatinib (Figure 2D). Almost in all inhibitor concentrations tested, we determined that myr-p110β MEFs are significantly more dependent on Src in comparison to their p110α counterparts. In short, our data thus far supports the notion that active p110β and p110δ induce differential activation of Src and STAT3 respectively.

Figure 2: 
p110β and p110δ activate differential tyrosine phosphorylation cascades. (A) Phospho-tyrosine blot of myristoylation constructs. The approximated molecular weight values are noted at the right hand side of the blot. GAPDH is used as loading control. (B) Myr-PI3K MEFs were exponentially grown, 30 µg lysate was used per cell line. Immunoblots were performed with phospho-STAT3, STAT3 and ACTIN antibodies. Band intensity quantifications of p-STAT3 were indicated for each sample. (C) Myr-p110α and myr-p110β MEFs were grown exponentially, 30 µg lysate was used per cell line. Indicated antibodies were used in an immunoblot, GAPDH serves as loading control. (D) 10,000 cells were seeded to six well plate. Indicated doses of Dasatinib treatment were conducted for 5 days. Cells were then fixed and crystal violet stainings were quantified in comparison to DMSO controls (n=3). Percents of growth inhibition were displayed with standard deviation, statistical significance was checked with Student’s t-test. Ns: p>0.05, *: p≤0.05, ***: p≤0.001.
Figure 2:

p110β and p110δ activate differential tyrosine phosphorylation cascades. (A) Phospho-tyrosine blot of myristoylation constructs. The approximated molecular weight values are noted at the right hand side of the blot. GAPDH is used as loading control. (B) Myr-PI3K MEFs were exponentially grown, 30 µg lysate was used per cell line. Immunoblots were performed with phospho-STAT3, STAT3 and ACTIN antibodies. Band intensity quantifications of p-STAT3 were indicated for each sample. (C) Myr-p110α and myr-p110β MEFs were grown exponentially, 30 µg lysate was used per cell line. Indicated antibodies were used in an immunoblot, GAPDH serves as loading control. (D) 10,000 cells were seeded to six well plate. Indicated doses of Dasatinib treatment were conducted for 5 days. Cells were then fixed and crystal violet stainings were quantified in comparison to DMSO controls (n=3). Percents of growth inhibition were displayed with standard deviation, statistical significance was checked with Student’s t-test. Ns: p>0.05, *: p≤0.05, ***: p≤0.001.

Activated p110β has a scaffold function that involves mTORC1

As the greatest levels of Akt1 Ser473 phosphorylation in MEFs was found in myr-p110β MEFs, we decided to study its potential causes and consequences. To do that, we used myr-p110α and myr-p110β MEFs and inhibited p110s by treating them with their respective inhibitors. Although in both cell lines phospho-S6 is totally eliminated upon inhibitor treatment, Pras40 phosphorylation was more persistently decreased in myr-p110α MEFs upon BYL719 treatment. On the other hand, myr-p110β MEFs treated with KIN193 exhibit sustained mTOR auto-phosphorylation despite the catalytic inhibition (Figure 3A) [23]. These results indicate that activated p110β might induce a differential signaling module that partially spares mTORC1 activity. To investigate this possibility further, we used short term Rapamycin treatment to target mTORC1, and EHT1864 to catalytically inhibit Rac1 (Figure 3B), as Rac1 is involved in activation of p110β [4]. We showed that catalytic inhibition of Rac1 decreases mTOR levels as well as its auto-phosphorylation, and these effects are further exacerbated when EHT1864 is combined with Rapamycin in myr-p110β MEFs. Interestingly, myr-p110α MEFs did not exhibit a similar response under identical circumstances. These results support the notion that inhibition of Rac1 catalytic activity inhibits phosphorylation of mTORC1 in myr-p110β MEFs possibly through a previously unidentified, enzyme independent function of p110β.

Figure 3: 
Activated p110α and p110β produce distinct marks for mTOR auto-phosphorylation. (A) Indicated cells were grown to subconfluency and treated with indicated inhibitors at 2 μM concentrations for 2 h before the harvest. Each well contains 40 µg of protein lysate. Immunoblots were performed with the indicated antibodies, GAPDH serves as loading control. (B) Indicated cells were grown to subconfluency and treated with indicated inhibitors for 2 h before the harvest. 100 nM Rapamycin and 5 μM EHT1844 were used either alone or in combination. Each well contains 40 µg of protein lysate. Immunoblots were performed with the indicated antibodies, GAPDH serves as loading control.
Figure 3:

Activated p110α and p110β produce distinct marks for mTOR auto-phosphorylation. (A) Indicated cells were grown to subconfluency and treated with indicated inhibitors at 2 μM concentrations for 2 h before the harvest. Each well contains 40 µg of protein lysate. Immunoblots were performed with the indicated antibodies, GAPDH serves as loading control. (B) Indicated cells were grown to subconfluency and treated with indicated inhibitors for 2 h before the harvest. 100 nM Rapamycin and 5 μM EHT1844 were used either alone or in combination. Each well contains 40 µg of protein lysate. Immunoblots were performed with the indicated antibodies, GAPDH serves as loading control.

Previously, it has been shown that mTORC1 becomes activated by RHEB [15]. As we observed a similar pattern where Rac1 catalytic activity somehow activated mTORC1 where p110β is present, we wanted to investigate the possibility of mTORC1, Rac1, p110β, and p85α icSH2 to form a complex in which mTORC1 becomes activated. Since Rheb and Rac1 are both GTPases, we modeled the interaction based on the crystal structure of mTORC1, whose PDB ID is 6BCU [15, 24]. We discovered putative contact sites by aligning Rac1 to Rheb on 6BCU in PyMOL and utilized them as input for HADDOCK, an online data-driven docking program. The catalytic sites of mTORC1 were inserted as passive residues to prevent proteins from docking to those areas [25, 26]. mTORC1, p110β, and Rac1 formed the best cluster (Figure 4A). We explored if p110α could form a comparable complex. HADDOCK simulations failed to produce an mTORC1–p110α complex (Figure 4B). We conducted a competitive docking experiment where p110α and p110β were instructed to engage with mTORC1 at the same residues to validate our claim. Intriguingly, p110α was not docked to mTORC1, but p110β was successfully shown to interact with mTORC1, where Rac1 plays a bridging role between mTOR and p110β (Figure 4C). The scores of the best clusters of the docking experiments could be found in Table 1. The HADDOCK score is calculated as sum of various molecular interactions, where the negative values indicate the high probability of a given cluster of models [27]. The Z-Score indicates the deviation of the cluster from the average, and better results have more negative Z-Scores. Overall, our findings suggest that Rac1 may also allosterically activate mTORC1 similar to RHEB, in a p110β-containing complex [15].

Figure 4: 
p110β but not p110α emerges as an interactor of mTOR in molecular docking simulations. (A) The best docked model generated by HADDOCK. p110β crystalized with p85 α IC SH2 domains, Rac1, and mTORC1 (mTOR, mLST8, RAPTOR) were used as input models for HADDOCK. Blue: mTORC1, green: p110β with p85α, magenta: Rac1. (B) The best docked model generated by HADDOCK. p110α crystalized with p85 α NI SH2 domains, Rac1, and mTORC1 (mTOR, mLST8, RAPTOR) were used as input models for HADDOCK. Blue: mTORC1, green: p110α with p85α, magenta: Rac1. (C) The best docked model generated by HADDOCK. p110β crystalized with p85 α IC SH2 domains, Rac1, mTORC1 (mTOR, mLST8, RAPTOR), and p110α with p85α were used as input models for HADDOCK. Blue: mTORC1, green: p110β with p85 α IC SH2, magenta: Rac1, yellow: p110α with p85α.
Figure 4:

p110β but not p110α emerges as an interactor of mTOR in molecular docking simulations. (A) The best docked model generated by HADDOCK. p110β crystalized with p85 α IC SH2 domains, Rac1, and mTORC1 (mTOR, mLST8, RAPTOR) were used as input models for HADDOCK. Blue: mTORC1, green: p110β with p85α, magenta: Rac1. (B) The best docked model generated by HADDOCK. p110α crystalized with p85 α NI SH2 domains, Rac1, and mTORC1 (mTOR, mLST8, RAPTOR) were used as input models for HADDOCK. Blue: mTORC1, green: p110α with p85α, magenta: Rac1. (C) The best docked model generated by HADDOCK. p110β crystalized with p85 α IC SH2 domains, Rac1, mTORC1 (mTOR, mLST8, RAPTOR), and p110α with p85α were used as input models for HADDOCK. Blue: mTORC1, green: p110β with p85 α IC SH2, magenta: Rac1, yellow: p110α with p85α.

Table 1:

Various HADDOCK parameters for p110α/p110β-mTOR docking simulations.

Parameters and results 4A: mTORC1, Rac1, p110β, p85α icSH2 4B: mTORC1, Rac1, p110α, p85α niSH2 4C: mTORC1, Rac1, p110α with p85α niSH2, p110β with p85α icSH2

All models successfully docked? Yes No Although p110β with Rac1 and p85α were successfully docked to mTORC1, p110α was not
HADDOCK score −200.2±4.7 −91.3±2.8 −176.5±10.2
Cluster size 3 45 2
RMSD from the overall lowest-energy structure 21.4±2.4 54.8±21.8 93.6±51.3
van der Waals energy −107.3±15.5 −50.9±4.9 −107.2±1.2
Electrostatic energy −436.0±62.0 −145.0±31.4 −494.8±71.2
Desolvation energy −25.6±6.5 −13.5±2.0 −18.8±12.3
Restraints violation energy 199.2±12.4 21.2±16.6 485.6±133.2
Buried surface area 4,554.0±354.3 1,584.3±92.0 4,123.0±467.9
Z-score −2.7 −1.5 −1.0

p110β expressing MEFs exhibit higher G2/M activity than p110α MEFs

We have shown that myr-p110β expressing MEFs have higher levels of phosphorylated and activated Akt1 (Figure 1C and D). As one of the roles of Akt is to promote cell cycle [28], we wanted to investigate whether cell cycle kinetics differ between p110α and p110β expressing MEFs via FACS analysis. For these sets of experiments, we used α, β +/+ Cre MEFs with p110α and p110β ectopic expression without the myr tag, to investigate the basal cell cycle kinetics. We observed that the p110α and p110β expressing MEFs have distinct cell cycle distributions (Figure 5A and B), with more p110β MEFs collected at the G2/M phase than the p110α MEFs (%42.19 vs. %21.47, respectively). Furthermore, fewer p110β MEFs are found in G1 and S phase, than p110α MEFs (%37.22 and %12.84 vs. %53.97 and %19.68, respectively, Figure 5C). These results indicate that p110β expression promotes a higher mitotic index.

Figure 5: 
Cell cycle analyses of double knock-out addback p110α and p110β MEFs. Cells were grown and harvested at 50 % confluency. The same gates were used for both of the cell lines. (A) FACS analysis of cycling DKO addback p110α MEFs. (B) FACS analysis of cycling DKO addback p110β MEFs. (C) Schematic representation of the cell cycle distribution of addback cell lines with mean and SEM (n=2).
Figure 5:

Cell cycle analyses of double knock-out addback p110α and p110β MEFs. Cells were grown and harvested at 50 % confluency. The same gates were used for both of the cell lines. (A) FACS analysis of cycling DKO addback p110α MEFs. (B) FACS analysis of cycling DKO addback p110β MEFs. (C) Schematic representation of the cell cycle distribution of addback cell lines with mean and SEM (n=2).

Activated p110β expressing MEFs are more sensitive to genotoxic stress

We observed that Akt1 Ser473 and Akt substrate phosphorylation (Figure 1C and D) levels were the highest in myr-p110β MEFs. We wished to study the consequences of the differential in Akt activation between p110 isoforms. As Akt has many effectors responsible for activating the cell cycle [29], we reasoned that myr-p110α and β MEFs may have different growth rates and sensitivities for cytotoxic agents as was suggested by our FACS results (Figure 5A and B). We performed 2D growth assays to understand whether growth rates differ among MEFs expressing activated p110s in comparison to controls (Figure 6A). According to our results, MEFs that express myr-p110β exhibit the fastest proliferation amongst the other lines. Cancer is characterized by uncontrolled cell proliferation as a result of cell cycle dysregulation. In response to genotoxic stress inducers, fast-cycling cells tend to accumulate more damage. We therefore used increasing doses of doxorubicin and cisplatin to examine the sensitivity of activated p110 MEFs in response to genotoxic stress. Myr-p110β MEFs are found to be the most sensitive cell line for genotoxic stress induced by doxorubicin and cisplatin (Figure 6B and C), having the lowest IC50 value among the other activated p110 MEF lines. Due to the fact that myr-p110β MEFs exhibited the highest phosphorylation level of Akt1 Ser473 among the activated p110 MEFs, we hypothesized that in these MEFs, Akt effectors that regulate cell cycle are upregulated, and as these cells cycle more frequently, they are more susceptible to genotoxic stress.

Figure 6: 
Myr-p110β expression sensitizes MEF more to genotoxic stress inducers in comparison to the other Class IA PI3Ks. (A) Indicated cells were seeded (104) on 6-well plates, cells were fixed after 3 days and crystal violet stainings were performed. Absolute quantifications of stained cells are displayed (n=3) with standard deviation. Statistical significance was checked with Student’s t-test. Ns: p>0.05, *: p≤0.05. (B) Depiction of IC50 values of doxorubicin of each depicted cell line, calculated from dose – response curves (0–200 μM) in three independent biological replicates. WT MEFs are used as baseline control. (C) Depiction of IC50 values of Cisplatin of each depicted cell line, calculated from dose – response curves (0–5 μM) in three independent biological replicates. WT MEFs are used as baseline control. Circles represent mean IC50 values for WT MEFs, whereas square, triangles and reverse triangles display the IC50 values for myr-p110α, myr-p110β and myr-p110δ MEFs respectively.
Figure 6:

Myr-p110β expression sensitizes MEF more to genotoxic stress inducers in comparison to the other Class IA PI3Ks. (A) Indicated cells were seeded (104) on 6-well plates, cells were fixed after 3 days and crystal violet stainings were performed. Absolute quantifications of stained cells are displayed (n=3) with standard deviation. Statistical significance was checked with Student’s t-test. Ns: p>0.05, *: p≤0.05. (B) Depiction of IC50 values of doxorubicin of each depicted cell line, calculated from dose – response curves (0–200 μM) in three independent biological replicates. WT MEFs are used as baseline control. (C) Depiction of IC50 values of Cisplatin of each depicted cell line, calculated from dose – response curves (0–5 μM) in three independent biological replicates. WT MEFs are used as baseline control. Circles represent mean IC50 values for WT MEFs, whereas square, triangles and reverse triangles display the IC50 values for myr-p110α, myr-p110β and myr-p110δ MEFs respectively.

Discussion

Previously, p110β was demonstrated to signal downstream of GPCRs [4]. Here, we propose two additional roles for p110β. We identified a possible alternative PI3K/mTOR signalling axis where p110β interacts with mTORC1 via Rac1. Secondly, we observed an increase in phosphorylation of Src upon activation of p110β.

It is expected to observe increased Akt Thr308 and Ser473 phosphorylations in myristoylated p110 MEFs compared to WT MEFs as Akt becomes activated downstream of PI3K [30]. These residues may not always reflect the activation status of Akt, rather, they may be read as phosphorylation of different downstream substrates of Akt, as in the case of Thr308 being enough in lipid and muscle cells and requirement for Ser473 phosphorylation in other cell lines for glucose intake [30, 31]. The different levels of Akt phosphorylation on Ser473, as well as their receptor selectivity, suggest that p110α and p110β play distinct but complementary functions in cellular physiology [32].

MEFs expressing p110β exhibited increased Src phosphorylation. It is not clear whether only the scaffolding or the catalytic action of p110β is causing the increase in phospho-Src. This increment in Src phosphorylation as a result of scaffolding role of p110β could be interpreted as GPCR internalization followed by autophagy [13, 33, 34]. Tyr350 on the β2-adrenergic receptor is phosphorylated by Src in response to insulin binding to its receptor [4, 5]. p110β is one of the potential actors in Src autophosphorylation through its scaffolding function, leading to receptor internalization [13]. This phosphorylation has been portrayed as an anchor for SH2 domain-containing proteins, such as c-Src and p85 [33]. It might be a different strategy to maintain glucose levels while favouring anabolism over catabolism up until the internalized receptor is broken down during autophagy. Additionally, under that setting, it is anticipated that mTORC1 will be activated to favour anabolic processes that result in the synthesis of cellular nutrition and to permit autophagy, which may be aided by p110β-mediated Src phsophorylation [11]. Another possible explanation for the increment in Src phosphorylation in the presence of activated p110β, is that the FAK-Src complex becomes activated and phosphorylates p130Cas, creating docking points for CRKL to recruit p110β and induce RAC activity. Increased RAC activity leads to an elevation in p110β activity, thus creating a positive feedback loop where Src, p110β, and Rac1 become activated [13].

In myr-p110δ MEFs, the phospho-Akt Ser473 and its substrate phosphorylation levels were the lowest, which indicated that p110δ signalling may be using an alternative route. From size-descriptive phospho-tyrosine blots, we suspected that JAK proteins would be phosphorylated the most in myr-p110δ MEFs. Then, we showed that cells with activated p110δ have increased Stat3 tyrosine phosphorylation. Previously, cells transformed by oncogenic H1047R p110α mutants were demonstrated to be dependent on Stat3 to induce oncogenicity [35]. It is highly possible that myr-p110δ MEFs also use the same network to induce oncogenicity depending on Stat3. Whether the same mechanism is valid for both activated p110α and p110δ needs to be studied further.

The mTOR autophosphorylation on Ser2481 enabled us to differentiate whether the outcome of p110β activity leads to an increase in its catalytic potential [36, 37]. Since Ser2481 phosphorylation is affected by Rac1 catalytic inhibition solely in myr-p110β MEFs, Rac1 and p110β interaction seems to be maintaining mTOR catalytic activity through the scaffolding role of p110β, unaffected by its catalytic activity. A recent report described a Rac1 dependent mechanism regulating the stability of mTOR [38]. An E3 ubiquitin ligase mediated process explains why we see a decrease in mTOR levels upon combined inhibition with Rapamycin and EHT1864. To investigate whether a complex consisting of mTORC1, Rac, p110β, and p85α icSH2, we utilized the crystal structures and docked them with HADDOCK. We used a previously established active mTORC1 and RHEB crystal structure to test if our complex might activate like RHEB. By replacing p110β with p110α, we showed that p110β was a critical component of the complex. Overall, it seems plausible to assume that Rac1 and p110β interact with mTOR in the same complex.

Furthermore, we demonstrated that MEFs which depend on only p110α or p110β have distinct cell cycle kinetics. It was previously presented that p110α has a critical role in re-entry into cell cycle, and we observed p110α-dependent MEFs accumulate more at G1 phase in comparison to wild-type counterparts [39]. The increased accumulation of p110β-dependent MEFs in G2/M may be due to the earlier conclusion of the S phase [14]. The fact that p110β-dependent MEFs end up in G2/M phase in comparison to their p110α counterparts explains why p110β MEFs proliferate faster than others. It also clarifies why p110β-dependent MEFs are the most sensitive cell line for genotoxic insult among other cell lines.

In conclusion, we show isoform-specific and redundant roles of p110s in signal transduction. Our findings provide important clarifications about isoform-specific roles of p110s in the normal cellular setting.


Corresponding author: Onur Cizmecioglu, Department of Molecular Biology and Genetics, Ihsan Dogramaci Bilkent University, Ankara, Türkiye, Phone: +90 312 2902138, E-mail:

Acknowledgments

We thank Jean J. Zhao, Shaozhen Xie, Jing Ni, and Thomas M. Roberts for the reagents.

  1. Research ethics: Not applicable.

  2. Informed consent: Not applicable.

  3. Author contributions: Research concept and design: H.B.C.Y., O.C. Collection and/or assembly of data: H.B.C.Y., M.S., O.A.I. Data analysis and interpretation: H.B.C.Y., M.S., O.A.I., O.C. writing the article: H.B.C.Y., O.C. Critical revision of the article H.B.C.Y., M.S., O.A.I., O.C. final approval of article: O.C. All authors have accepted responsibility for the entire content of this manuscript and approved its submission.

  4. Competing interests: The authors state no conflict of interests.

  5. Research funding: The project was funded by The Scientific and Technological Research Council of Turkey (TÜBİTAK), project number 119Z068.

  6. Data availability: The raw data can be obtained on request from the corresponding author.

References

1. Rommel, C, Vanhaesebroeck, B, Vogt, PK. Phosphoinositide 3-kinase in health and disease. London: Springer; 2010:267–78 pp.10.1007/978-3-642-14816-3Suche in Google Scholar

2. Wozniak, DJ, Kajdacsy-Balla, A, Macias, V, Ball-Kell, S, Zenner, ML, Bie, W, et al.. PTEN is a protein phosphatase that targets active PTK6 and inhibits PTK6 oncogenic signaling in prostate cancer. Nat Commun 2017;8, https://doi.org/10.1038/s41467-017-01574-5,Suche in Google Scholar PubMed PubMed Central

3. Sueta, A, Yamamoto, Y, Yamamoto-Ibusuki, M, Hayashi, M, Takeshita, T, Yamamoto, S, et al.. An integrative analysis of PIK3CA mutation, PTEN, and INPP4B expression in terms of trastuzumab efficacy in HER2-positive breast cancer. PLoS One 2014;9:1–17, https://doi.org/10.1371/journal.pone.0116054.Suche in Google Scholar PubMed PubMed Central

4. Cizmecioglu, O, Ni, J, Xie, S, Zhao, JJ, Roberts, TM. Rac1-mediated membrane raft localization of PI3K/p110β is required for its activation by GPCRs or PTEN loss. Elife 2016;5:1–21, https://doi.org/10.7554/elife.17635.Suche in Google Scholar

5. Burke, JE, Williams, RL. Dynamic steps in receptor tyrosine kinase mediated activation of class IA phosphoinositide 3-kinases (PI3K) captured by H/D exchange (HDX-MS). Adv Biol Regul [Internet] 2013;53:97–110, https://doi.org/10.1016/j.jbior.2012.09.005.Suche in Google Scholar PubMed PubMed Central

6. Okkenhaug, K, Ali, K, Vanhaesebroeck, B. Antigen receptor signalling: a distinctive role for the p110δ isoform of PI3K. Trends Immunol 2007;28:80–7, https://doi.org/10.1016/j.it.2006.12.007.Suche in Google Scholar PubMed PubMed Central

7. Fritsch, R, De Krijger, I, Fritsch, K, George, R, Reason, B, Kumar, MS, et al.. RAS and RHO families of GTPases directly regulate distinct phosphoinositide 3-kinase isoforms. Cell 2013;153:1050, https://doi.org/10.1016/j.cell.2013.04.031.Suche in Google Scholar PubMed PubMed Central

8. Liu, P, Cheng, H, Roberts, TM, Zhao, JJ. Targeting the phosphoinositide 3-kinase pathway in cancer. Nat Rev Drug Discov 2009;8:627–44, https://doi.org/10.1038/nrd2926.Suche in Google Scholar PubMed PubMed Central

9. Sahlberg, SH, Mortensen, AC, Haglöf, J, Engskog, MKR, Arvidsson, T, Pettersson, C, et al.. Different functions of AKT1 and AKT2 in molecular pathways, cell migration and metabolism in colon cancer cells. Int J Oncol 2017;50:5–14, https://doi.org/10.3892/ijo.2016.3771.Suche in Google Scholar PubMed PubMed Central

10. Saxton, RA, Sabatini, DM. mTOR signaling in growth, metabolism, and disease. Cell 2017;168:960–76, https://doi.org/10.1016/j.cell.2017.02.004.Suche in Google Scholar PubMed PubMed Central

11. Liu, GY, Sabatini, DM. mTOR at the nexus of nutrition, growth, ageing and disease. Nat Rev Mol Cell Biol 2020;21:183–203, https://doi.org/10.1038/s41580-019-0199-y.Suche in Google Scholar PubMed PubMed Central

12. Hunter, T. Tyrosine phosphorylation: thirty years and counting. Curr Opin Cell Biol 2009;21:140–6, https://doi.org/10.1016/j.ceb.2009.01.028.Suche in Google Scholar PubMed PubMed Central

13. Zhang, J, Gao, X, Schmit, F, Adelmant, G, Eck, MJ, Marto, JA, et al.. CRKL mediates p110β-dependent PI3K signaling in PTEN-deficient cancer cells. Cell Rep 2017;20:549–57, https://doi.org/10.1016/j.celrep.2017.06.054.Suche in Google Scholar PubMed PubMed Central

14. Marqués, M, Kumar, A, Poveda, AM, Zuluaga, S, Hernández, C, Jackson, S, et al.. Specific function of phosphoinositide 3-kinase beta in the control of DNA replication. Proc Natl Acad Sci U S A 2009;106:7525–30, https://doi.org/10.1073/pnas.0812000106.Suche in Google Scholar PubMed PubMed Central

15. Yang, H, Jiang, X, Li, B, Yang, HJ, Miller, M, Yang, A, et al.. Mechanisms of mTORC1 activation by RHEB and inhibition by PRAS40. Nature 2017;552:368–73, https://doi.org/10.1038/nature25023.Suche in Google Scholar PubMed PubMed Central

16. Schmit, F, Utermark, T, Zhang, S, Wang, Q, Von, T, Roberts, TM, et al.. PI3K isoform dependence of PTEN-deficient tumors can be altered by the genetic context. Proc Natl Acad Sci U S A 2014;111:6395–400, https://doi.org/10.1073/pnas.1323004111.Suche in Google Scholar PubMed PubMed Central

17. Vanhaesebroeck, B, Stephens, L, Hawkins, P. PI3K signalling: the path to discovery and understanding. Nat Rev Mol Cell Biol 2012;13:195–203, https://doi.org/10.1038/nrm3290.Suche in Google Scholar PubMed

18. Furet, P, Guagnano, V, Fairhurst, RA, Imbach-Weese, P, Bruce, I, Knapp, M, et al.. Discovery of NVP-BYL719 a potent and selective phosphatidylinositol-3 kinase alpha inhibitor selected for clinical evaluation. Bioorganic Med Chem Lett 2013;23:3741–8, https://doi.org/10.1016/j.bmcl.2013.05.007.Suche in Google Scholar PubMed

19. Sugiyama, MG, Fairn, GD, Antonescu, CN. Akt-ing up just about everywhere: compartment-specific Akt activation and function in receptor tyrosine kinase signaling. Front Cell Dev Biol 2019;7:1–24, https://doi.org/10.3389/fcell.2019.00070.Suche in Google Scholar PubMed PubMed Central

20. Bretz, NP, Salnikov, AV, Perne, C, Keller, S, Wang, X, Mierke, CT, et al.. CD24 controls Src/STAT3 activity in human tumors. Cell Mol Life Sci 2012;69:3863–79, https://doi.org/10.1007/s00018-012-1055-9.Suche in Google Scholar PubMed

21. Johnston, JA, Bacon, CM, Finbloom, DS, Rees, RC, Kaplan, D, Shibuya, K, et al.. Tyrosine phosphorylation and activation of STAT5, STAT3, and Janus kinases by interleukins 2 and 15. Proc Natl Acad Sci U S A 1995;92:8705–9, https://doi.org/10.1073/pnas.92.19.8705.Suche in Google Scholar PubMed PubMed Central

22. Demir, M, Cizmecioglu, O. ZAP70 activation compensates for loss of class IA PI3K isoforms through activation of the JAK–STAT3 pathway. Cancer Diagnosis Progn 2022;2:391–404, https://doi.org/10.21873/cdp.10122.Suche in Google Scholar PubMed PubMed Central

23. Wang, L, Harris, TE, Roth, RA, Lawrence, JC. PRAS40 regulates mTORC1 kinase activity by functioning as a direct inhibitor of substrate binding. J Biol Chem 2007;282:20036–44, https://doi.org/10.1074/jbc.m702376200.Suche in Google Scholar PubMed

24. Schöneborn, H, Raudzus, F, Coppey, M, Neumann, S, Heumann, R. Perspectives of RAS and RHEB GTPase signaling pathways in regenerating brain neurons. Int J Mol Sci 2018;19:1–37, https://doi.org/10.3390/ijms19124052.Suche in Google Scholar PubMed PubMed Central

25. Honorato, RV, Koukos, PI, Jiménez-García, B, Tsaregorodtsev, A, Verlato, M, Giachetti, A, et al.. Structural biology in the clouds: the WeNMR-EOSC ecosystem. Front Mol Biosci 2021;8:1–7, https://doi.org/10.3389/fmolb.2021.729513.Suche in Google Scholar PubMed PubMed Central

26. Van Zundert, GCP, Rodrigues, JPGLM, Trellet, M, Schmitz, C, Kastritis, PL, Karaca, E, et al.. The HADDOCK2.2 web server: user-friendly integrative modeling of biomolecular complexes. J Mol Biol 2016;428:720–5, https://doi.org/10.1016/j.jmb.2015.09.014.Suche in Google Scholar PubMed

27. Chakravarty, D, Guharoy, M, Robert, CH, Chakrabarti, P, Janin, J. Reassessing buried surface areas in protein-protein complexes. Protein Sci 2013;22:1453–7, https://doi.org/10.1002/pro.2330.Suche in Google Scholar PubMed PubMed Central

28. Vadlakonda, L, Pasupuleti, M, Pallu, R. Role of PI3K-AKT-mTOR and Wnt signaling pathways in transition of G1-S phase of cell cycle in cancer cells. Front Oncol 2013:1–7. https://doi.org/10.3389/fonc.2013.00085.Suche in Google Scholar PubMed PubMed Central

29. Manning, BD, Cantley, LC. AKT/PKB signaling: navigating downstream. Cell 2007;129:1261–74, https://doi.org/10.1016/j.cell.2007.06.009.Suche in Google Scholar PubMed PubMed Central

30. Manning, BD, Toker, A. AKT/PKB signaling: navigating the network. Cell 2017;169:381–405. https://doi.org/10.1016/j.cell.2017.04.001.Suche in Google Scholar PubMed PubMed Central

31. Yudushkin, I. Getting the akt together: guiding intracellular akt activity by pi3k. Biomolecules 2019;9:1–14. https://doi.org/10.3390/biom9020067.Suche in Google Scholar PubMed PubMed Central

32. Guillermet-Guibert, J, Bjorklof, K, Salpekar, A, Gonella, C, Ramadani, F, Bilancio, A, et al.. The p110β isoform of phosphoinositide 3-kinase signals downstream of G protein-coupled receptors and is functionally redundant with p110γ. Proc Natl Acad Sci U S A 2008;105:8292–7, https://doi.org/10.1073/pnas.0707761105.Suche in Google Scholar PubMed PubMed Central

33. Tobin, AB. G-protein-coupled receptor phosphorylation: where, when and by whom. Br J Pharmacol 2008;153:167–76, https://doi.org/10.1038/sj.bjp.0707662.Suche in Google Scholar PubMed PubMed Central

34. Chen, C-C, Jeon, S-M, Bhaskar, PT, Nogueira, V, Sundararajan, D, Tonic, I, et al.. FoxOs inhibit mTORC1 and activate Akt by inducing the expression of Sestrin3 and Rictor. Dev Cell 2010;18:592–604, https://doi.org/10.1016/j.devcel.2010.03.008.Suche in Google Scholar PubMed PubMed Central

35. Hart, JR, Liao, L, Yates, JR, Vogt, PK. Essential role of stat3 in pi3k-induced oncogenic transformation. Proc Natl Acad Sci U S A 2011;108:13247–52, https://doi.org/10.1073/pnas.1110486108.Suche in Google Scholar PubMed PubMed Central

36. Chiang, GG, Abraham, RT. Phosphorylation of mammalian target of rapamycin (mTOR) at Ser-2448 is mediated by p70S6 kinase. J Biol Chem 2005;280:25485–90, https://doi.org/10.1074/jbc.m501707200.Suche in Google Scholar

37. Soliman, GA, Acosta-Jaquez, HA, Dunlop, EA, Ekim, B, Maj, NE, Tee, AR, et al.. mTOR Ser-2481 autophosphorylation monitors mTORC-specific catalytic activity and clarifies rapamycin mechanism of action. J Biol Chem 2010;285:7866–79, https://doi.org/10.1074/jbc.m109.096222.Suche in Google Scholar

38. Turgu, B, El-Naggar, A, Kogler, M, Tortola, L, Zhang, HF, Hassan, M, et al.. The HACE1 E3 ligase mediates RAC1-dependent control of mTOR signaling complexes. EMBO Rep 2023;24:e56815. https://doi.org/10.15252/embr.202356815.Suche in Google Scholar PubMed PubMed Central

39. Cizmecioglu, O. p110α activity at the M-to-G1 transition is critical for cellular proliferation and reentry into the cell cycle. Turkish J Biol 2022;46:207–15, https://doi.org/10.55730/1300-0152.2609.Suche in Google Scholar PubMed PubMed Central

Received: 2023-07-02
Accepted: 2023-11-21
Published Online: 2023-12-20

© 2023 the author(s), published by De Gruyter, Berlin/Boston

This work is licensed under the Creative Commons Attribution 4.0 International License.

Artikel in diesem Heft

  1. Frontmatter
  2. Review
  3. Quercetin induces cytotoxicity and apoptosis, reduces metastasis and drug resistance in oral cancer cells
  4. Research Articles
  5. Comparison of results of two hematological analyzer systems: Dirui BF-7200 and Sysmex XN-1000
  6. User verification of Abbott Alinity HQ hematology analyzer
  7. Erythrocyte labile iron pool indicating concealed iron overload in non-transfusion-dependent β-thalassemia
  8. Flow cytometric analysis of lymphocyte subsets, monocytes, and HLA-DR expressions on these cells in patients with COVID-19
  9. Comparative vascular effects of levetiracetam and valproate with hyperhomocysteinemia in rat models
  10. Evaluation of systemic inflammatory and fibrosis indices in Saprochaete capitata infections: a retrospective case-control study
  11. Class IA PI3K isoforms lead to differential signalling downstream of PKB/Akt
  12. Enzymatic comparison and expression pattern of pig B4GALNT2 and B4GALNT2-like proteins
  13. Does COVID-19 affect thyroid more than non-COVID-19 infections? A retrospective study
  14. Desmin’s conformational modulation by hydrophobicity
  15. Assessment of myogenic potency in patient-derived fibroblasts with c.1289-2A>G Desmin mutation
  16. GSH-related enzyme activity and tumor relation: glutathione peroxidase and glutathione reductase status under hypoxia in HepG2 cells
  17. Can triglyceride related indices be reliable markers in the assessment of polycystic ovarian syndrome?
  18. Vascular endothelial growth factor (VEGF)-C and its receptors, soluble VEGFR-2 and VEGFR-3, in polycystic ovary syndrome
  19. Platelet activating factor acetylhydrolase is associated with cardiac valvular calcification in dialysis patients
  20. Reticulated platelets and coronary slow flow: a study in stable coronary artery disease
  21. Identification of the role of TG2 on the expression of TGF-β, TIMP-1 and TIMP-2 in aged skin
  22. Unveiling the link: Helicobacter pylori infection and impact on ischemia modified albumin, thiol, and disulfide levels
Heruntergeladen am 6.10.2025 von https://www.degruyterbrill.com/document/doi/10.1515/tjb-2023-0146/html?lang=de
Button zum nach oben scrollen