bims-pimaco Biomed News
on PI3K and MAPK signalling in colorectal cancer
Issue of 2022‒05‒15
thirteen papers selected by
Lucas B. Zeiger
Beatson Institute for Cancer Research


  1. FASEB J. 2022 May;36 Suppl 1
      PIK3CA, the gene for the lipid kinase p110α is one of the most frequently mutated oncogenes across all types of cancer. p110α is an enzyme that catalyzes the formation of phosphatidylinositol 3, 4, 5 Triphosphate (PIP3 ). PIP3 recruits effector proteins which regulate growth, proliferation and motility. Due to this role as a master cell regulator, the activity of p110α is maintained in an inactive confirmation and is only activated downstream of Receptor Tyrosine Kinases (RTKs) and RAS family of GTPases. p110α is maintained in an inactive confirmation through interactions with its regulatory subunit as well as inhibitory contacts with the C-terminus. However, oncogenic mutations in p110α breaks these inhibitory interactions and drives hyperactivity without the need for activation signals leading to uncontrolled cell growth. We provide molecular insights into the regulation of oncogenic mutants of p110α using Hydrogen-Deuterium Exchange Mass Spectrometry (HDX-MS). HDX-MS reveals the dynamic changes between the natural cytosolic state and fully-active membrane bound states of the enzyme. We find unique molecular mechanisms regulating how mutants at the c-terminus activate lipid kinase activity (H1047R, M1043L, G1049R and N1068KLKR). Using extensive biophysical tools, biochemical assays, and MD simulations, we have tested the oncogenic potential of these mutations. Our results elucidates a unifying theory towards the regulation of PI3Kα and how oncogenic mutations drive hyperactivity. This will aid in understanding the regulation of PI3Kα and in developing isoform specific inhibitors.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.L7656
  2. FASEB J. 2022 May;36 Suppl 1
      The conserved kinase mTOR (mechanistic target of rapamycin) regulates cell metabolism and promotes cell growth, proliferation, and survival in response to diverse environmental cues (e.g., nutrients; growth factors; hormones). mTOR forms the catalytic core of two multiprotein complexes, mTORC1 and mTORC2, which possess unique downstream targets and cellular functions. While mTORC1 and mTORC2 often respond to distinct upstream cues, they share a requirement for PI3K in their activation by growth factors. While many studies agree that amino acids activate mTORC1 but not mTORC2, several studies reported paradoxical activation of mTORC2 by amino acids. We noted that stimulating amino acid starved cells with a commercial mixture of amino acids increased mTORC2-dependent Akt S473 phosphorylation rapidly while re-feeding cells with complete DMEM containing amino acids failed to do so. Interestingly, we found the pH of the commercial amino acid mixture to be ~ pH 10. Upon controlling for pH, stimulating starved cells with amino acids at pH 10 but not 7.4 increased mTORC2 signaling. Moreover, DMEM at alkaline pH was sufficient to increase mTORC2 catalytic activity and signaling. Using a fluorescent pH-sensitive dye (cSNARF-1-AM) coupled to ratio-metric live cell imaging, we confirmed that alkaline extracellular pH (pHe) translated into a rapid increase in intracellular pH (pHi). Moreover, blunting this increase with a pharmacological inhibitor of an H+ transporter attenuated the increase in mTORC2 signaling by pHe. Alkaline pHi also activated AMPK, a canonical sensor of energetic stress that promotes mTORC2 signaling, as reported previously by us. Functionally, we found that alkaline pHi attenuated apoptosis caused by growth factor withdrawal through activation of AMPK-mTORC2 signaling. These results indicate that alkaline pHi augments mTORC2 signaling to promote cell survival, in part through AMPK. In the course of this work, we noted that pHi increased phosphorylation of several downstream targets of PI3K (e.g., Akt P-T308 and P-S473; S6K1 P-T389 and P-T229; PRAS40 P-T246; Tsc2 P-S939), suggesting that PI3K itself responds to changes in pHi. Indeed, alkaline pHi increased PI-3',4',5'-P3 levels in a manner sensitive to the PI3K inhibitor BYL-719. Thus, alkaline pHi elevates PI3K activity, which increases both mTORC1 and mTORC2 signaling. Mechanistically, we found that activation of PI3K by alkaline pHi induced dissociation of Tsc2 from lysosomal membranes, thereby relieving TSC-mediated suppression of Rheb, a mTORC1-activating GTPase. Functionally, we found that activation of PI3K by alkaline pHi increased mTORC1-mediated 4EBP1 phosphorylation, which initiates cap-dependent translation by eIF4E. Alkaline pHi also increased mTORC1-driven protein synthesis. Taken together, these findings reveal alkaline pHi as a previously unrecognized activator of PI3K-mTORC1/2 signaling that promotes protein synthesis and cell survival. As elevated pHi represents an under-appreciated hallmark of cancer cells, these findings suggest that by alkaline pHi sensing by the PI3K-mTOR axis and AMPK-mTORC2 axes may contribute to tumorigenesis.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.L7803
  3. FASEB J. 2022 May;36 Suppl 1
      Ras proteins are among the most widely studied proto-oncogene products. Ras proteins are membrane-anchored GTPases that participate in multiple signaling cascades regulating crucial cellular processes including cell survival, proliferation, and differentiation. Ras mutations and/or deregulated Ras activities frequently lead to inflammation and malignant transformation. Recent studies have also shown that Ras mutations are associated with immuno-resistance by positive regulation of PD-L1 expression. Indoleamine 2,3-dioxygenases 1 (IDO1) is a heme-containing enzyme that catalyzes the rate limiting step of conversion of tryptophan into kynurenine (Kyn). Extensive research in the past has shown that IDO1 and its catalytic product Kyn mediate immune tolerance, thus promoting tumorigenesis in vivo. IDO1 expression is activated by immune cytokines including interferon-γ (IFN-γ)and interleukin-6 (IL-6). However, whether there is a mechanistic link between oncogenic KRas and IDO1 expression and how IDO1 expression contributes to immune invasion of transformed cells with KRas mutations remain unclear. Here we report that oncogenic KRas significantly enhanced IFN-γ-induced IDO1 expression. In H358 lung carcinoma cells, IDO1 expression induced by IFN-γ was at least in part dependent on ERK activation, and treatment with ARS-1620, a covalent KRasG12C inhibitor, suppressed IDO1 expression induced by IFN-γαμμα in a concentration-dependent manner. IDO1 expression was also induced by IFN-γ in HCT116 colon carcinoma cells that harbored a KRasG12D mutant allele, but not in HCT116 cells with wild-type KRas, suggesting that IFN-γ induced IDO1 expression requires Ras activation. In addition, KRas downregulation by specific siRNAs decreased IFN-γ-induced IDO1 expression in A549 lung cancer cells. Taken together, our study strongly suggests that Ras/ERK signaling pathway plays an important role in promoting IDO1 expression induced by IFN-γ.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.L7486
  4. FASEB J. 2022 May;36 Suppl 1
      The mechanistic target of rapamycin complex 1 (mTORC1) senses diverse signals to regulate cell growth and metabolism. The complex is present at the plasma membrane, nucleus, lysosomes, and the outer mitochondrial membrane. Such spatial compartmentation has been suggested to enhance signaling efficiency and specificity. For instance, we recently discovered nuclear mTORC1 activity, which is distinctly regulated from the canonical lysosomal mTORC1 (Zhou et al., 2020). Previous studies have shown that mTOR is present at the outer mitochondrial membrane (OMM), but it is not clear whether mTORC1 is active at this location and what the functional consequences are. To investigate this, we targeted our FRET-based mTORC1 activity reporter, TORCAR (Zhou et al., 2015), to the OMM and probed the subcellular activity of mTORC1. We found that platelet-derived growth factor (PDGF) stimulation increases mTORC1 activity at the OMM in addition to at the lysosome and in the nucleus, whereas insulin specifically stimulates mTORC1 activity at the OMM without affecting the lysosomal and nuclear activities. We further dissected the regulation of mitochondrial mTORC1 activity and applied a novel approach of identifying new mTORC1 substrates. Elucidating the signaling events that lead to subcellular mTORC1 activity at mitochondria and its downstream functions will increase our understanding of the roles that mTORC1 may play in diseases associated with altered metabolism or mitochondrial dysfunction, such as diabetes and cancer.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R4944
  5. Cancer Res. 2022 May 10. pii: canres.0804.2022. [Epub ahead of print]
      Mutations in RAS isoforms (KRAS, NRAS, and HRAS) are among the most frequent oncogenic alterations in many cancers, making these proteins high priority therapeutic targets. Effectively targeting RAS isoforms requires an exact understanding of their active, inactive, and druggable conformations. However, there is no structural catalog of RAS conformations to guide therapeutic targeting or examining the structural impact of RAS mutations. Here we present an expanded classification of RAS conformations based on analyses of the catalytic switch 1 (SW1) and switch 2 (SW2) loops. From 721 human KRAS, NRAS, and HRAS structures available in the Protein Data Bank (206 RAS-protein co-complexes, 190 inhibitor-bound, and 325 unbound, including 204 WT and 517 mutated structures), we created a broad conformational classification based on the spatial positions of Y32 in SW1 and Y71 in SW2. Clustering all well-modeled SW1 and SW2 loops using a density-based machine learning algorithm defined additional conformational subsets, some previously undescribed. Three SW1 conformations and nine SW2 conformations were identified, each associated with different nucleotide states (GTP-bound, nucleotide-free, and GDP-bound) and specific bound proteins or inhibitor sites. The GTP-bound SW1 conformation could be further subdivided based on the hydrogen bond type made between Y32 and the GTP γ-phosphate. Further analysis clarified the catalytic impact of G12D and G12V mutations and the inhibitor chemistries that bind to each druggable RAS conformation. Overall, this study has expanded our understanding of RAS structural biology, which could facilitate future RAS drug discovery.
    DOI:  https://doi.org/10.1158/0008-5472.CAN-22-0804
  6. Am Soc Clin Oncol Educ Book. 2022 Apr;42 1-13
      Despite the discovery of RAS oncogenes in human tumor DNA 40 years ago, the development of effective targeted therapies directed against RAS has lagged behind those more successful advancements in the field of therapeutic tyrosine kinase inhibitors targeting other oncogenes such as EGFR, ALK, and ROS1. The discoveries that (1) malignant RAS oncogenes differ from their wild-type counterparts by only a single amino acid change and (2) covalent inhibition of the cysteine residue at codon 12 of KRASG12C in its inactive GDP-bound state resulted in effective inhibition of oncogenic RAS signaling and have catalyzed a dramatic shift in mindset toward KRAS-driven cancers. Although the development of allele-selective KRASG12C inhibitors has changed a treatment paradigm, the clinical activity of these agents is more modest than tyrosine kinase inhibitors targeting other oncogene-driven cancers. Heterogeneous resistance mechanisms generally result in the restoration of RAS/mitogen-activated protein kinase pathway signaling. Many approaches are being evaluated to overcome this resistance, with many combinatorial clinical trials ongoing. Furthermore, because KRASG12D and KRASG12V are more prevalent than KRASG12C, there remains an unmet need for additional therapeutic strategies for these patients. Thus, our current translational standing could be described as "the end of the beginning," with additional discovery and research innovation needed to address the enormous disease burden imposed by RAS-mutant cancers. Here, we describe the development of KRASG12C inhibitors, the challenges of resistance to these inhibitors, strategies to mitigate that resistance, and new approaches being taken to address other RAS-mutant cancers.
    DOI:  https://doi.org/10.1200/EDBK_351333
  7. FASEB J. 2022 May;36 Suppl 1
      mTOR, which is part of mTOR complex 1 (mTORC1) and mTORC2, controls cellular metabolism in response to levels of nutrients and other growth signals. A hallmark of mTORC2 activation is the phosphorylation of Akt, which becomes upregulated in cancer. How mTORC2 modulates Akt phosphorylation remains poorly understood. Here, we found that the RNA binding protein, AUF1 (ARE/poly(U)-binding/degradation factor 1), modulates mTORC2/Akt signaling. AUF1 is required for Akt phosphorylation. It also mediates phosphorylation of the mTORC2-modulated metabolic enzyme GFAT1 at Ser243. Reciprocally, mTORC2 could also modulate AUF1. Conditions that enhance mTORC2 signaling, such as serum restimulation or acute glutamine withdrawal augments AUF1 phosphorylation while mTOR inhibition abolishes AUF1 phosphorylation. Our findings unravel a role for AUF1 in mTORC2/Akt signaling. Targeting AUF1 could serve as a novel treatment strategy for cancers with upregulated mTORC2/Akt signaling.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.0R396
  8. FASEB J. 2022 May;36 Suppl 1
      BRAF is one member of the RAF serine/threonine kinase family and a key component of the RAS-RAF-MEK-ERK (MAPK) signaling pathway, controlling cell growth, proliferation, differentiation, migration and survival. BRAF is the most frequently mutated kinase in human cancers, with over 200 mutations discovered, among which V600E and G469A are the most common BRAF mutations. Current therapies with ATP-competitive inhibitors are potent against BRAFV600E , yet they display "paradoxical activation" and drug resistance toward BRAFG469A andother non-V600 BRAF mutants. Thus, novel BRAF therapies towards non-V600 variants are urgently needed. However, the biochemical properties of non-V600 BRAF mutants and the cause of drug response are unclear due to the lack of structures of BRAF mutants. In this study, we applied molecular dynamics (MD) simulations to investigate the structure, dynamics and functions of oncogenic BRAFG469A , in comparison with BRAFWT and BRAFV600E . For each BRAF type (WT or mutant), MD simulations are carried out for its monomer and dimer with or without 1 or 2 bound vemurafenib in explicit aqueous solution. Our work provides atomic information on mechanism of aberrant activation by G469A mutation, "paradoxical activation" and structural dynamics of the nucleotide pocket, activation loop, and dimer and inhibitor/BRAF interfaces. G469A mutation makes hydrophobic contact between methyl of A469 with alkyl chain of K483 in the binding pocket. This stabilizes the K483-E501 salt-bridge, which functions to bring αC-helix towards the active site. Thus, G469A stabilizes the active conformation by positioning αC-helix close to the P-loop through increasingly stabilized salt-bridge relative to WT and V600E. Consequently, our simulations illustrate inability of vemurafenib to disrupt active conformation of BRAFG469A by separating αC-helix from the nucleotide binding pocket. This decreased structural fluctuation upon inhibitor binding to one protomer explains the ability of "paradoxical activation" of BRAF dimer through allosteric activation.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R3086
  9. J Cancer Res Clin Oncol. 2022 May 11.
      INTRODUCTION: KRAS mutations (KRASmut), PIK3CAmut, BRAFmut, and mismatch repair deficiency (dMMR) have been associated with the Warburg-effect. We previously observed differential associations between energy balance-related factors (BMI, clothing-size, physical activity) and colorectal cancer (CRC) subtypes based on the Warburg-effect. We now investigated whether associations between energy balance-related factors and risk of CRC differ between subgroups based on mutation and MMR status.METHODS: Information on molecular features was available for 2349 incident CRC cases within the Netherlands Cohort Study (NLCS), with complete covariate data available for 1934 cases and 3911 subcohort members. Multivariable-adjusted Cox-regression was used to estimate associations of energy balance-related factors with risk of CRC based on individual molecular features (KRASmut; PIK3CAmut; BRAFmut; dMMR) and combinations thereof (all-wild-type + MMR-proficient (pMMR); any-mutation/dMMR).
    RESULTS: In men, BMI and clothing-size were positively associated with risk of colon, but not rectal cancer, regardless of molecular features subgroups; the strongest associations were observed for PIK3CAmut colon cancer. In women, however, BMI and clothing-size were only associated with risk of KRASmut colon cancer (p-heterogeneityKRASmut versus all-wild-type+pMMR = 0.008). Inverse associations of non-occupational physical activity with risk of colon cancer were strongest for any-mutation/dMMR tumors in men and women, and specifically for PIK3CAmut tumors in women. Occupational physical activity was inversely associated with both combination subgroups of colon cancer in men.
    CONCLUSION: In men, associations did not vary according to molecular features. In women, a role of KRAS mutations in the etiological pathway between adiposity and colon cancer is suggested, and of PIK3CA mutations between physical activity and colon cancer.
    Keywords:  Colorectal cancer; Energy balance; Etiological heterogeneity; Mismatch repair/microsatellite instability; Mutations; Prospective cohort study
    DOI:  https://doi.org/10.1007/s00432-022-04019-9
  10. FASEB J. 2022 May;36 Suppl 1
      The PI 3-kinase (PI3K) and AKT signaling pathway plays a critical role in regulating all aspects of normal cellular physiology, and is also frequently deregulated in human pathophysiologies, most evidently in cancer and diabetes. Growth factors and hormones stimulate PI3K leading to the biosynthesis of the lipid-derived second messenger PIP3. In turn, PIP3 elicits the membrane recruitment of the protein kinase AKT, originally discovered in 1987 by Staal and colleagues as v-Akt, a transforming oncogene. In the early 1990s, three independent groups cloned and described the cellular homolog c-AKT, a serine/threonine protein kinase with a high degree of homology to other AGC family protein kinases. In the ensuing three decades, the mechanisms by which AKT transduces signals to cell growth, proliferation, motility and metabolism were uncovered. Three AKT isoforms exist in humans encoded by distinct genes (AKT1, AKT2, AKT3), and although originally thought to function redundantly, many studies have shown that AKT isoforms have non-overlapping and unique roles in both normal physiology and disease. Similarly, genetic lesions in the PI3K and AKT oncogenes have been described, and many of the genes that contribute to PI3K/AKT pathway activation and also signal termination have been found to be altered in human cancers. Numerous drugs that inhibit PI3K as well AKT have been developed for therapeutic use in patients, and many of these are being evaluated in late-stage clinical trials. During the lecture, I will highlight the major advances in PI3K and AKT field over the past 30 years, with a focus on mechanistic insight into this ubiquitous lipid signaling pathway. Genetic lesions in the PI3K/AKT pathway in human cancers will also be discussed, as well as efforts to target this pathway therapeutically. The second part of the lecture will focus on recent efforts in our laboratory to uncover novel mechanisms of AKT signaling and biology, with an emphasis on breast cancer and with a focus on metabolic reprogramming mediated by AKT. I will also present recent efforts aimed at targeting AKT with novel therapies, including degrader technologies and how these have illuminated novel aspects of AKT biology. I will conclude with some personal thoughts and future perspectives as to where the field is going, gaps in knowledge and what studying AKT for 30 years has taught me.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.0I219
  11. Sci Rep. 2022 May 13. 12(1): 7965
      KRAS mutation is responsible for 40-50% of colorectal cancers (CRCs). RNA-seq data and bioinformatics methods were used to analyze the transcriptional profiles of KRAS mutant (mtKRAS) in comparison with the wild-type (wtKRAS) cell lines, followed by in-silico and quantitative real-time PCR (qPCR) validations. Gene set enrichment analysis showed overrepresentation of KRAS signaling as an oncogenic signature in mtKRAS. Gene ontology and pathway analyses on 600 differentially-expressed genes (DEGs) indicated their major involvement in the cancer-associated signal transduction pathways. Significant hub genes were identified through analyzing PPI network, with the highest node degree for PTPRC. The evaluation of the interaction between co-expressed DEGs and lncRNAs revealed 12 differentially-expressed lncRNAs which potentially regulate the genes majorly enriched in Rap1 and RAS signaling pathways. The results of the qPCR showed the overexpression of PPARG and PTGS2, and downregulation of PTPRC in mtKRAS cells compared to the wtKRAS one, which confirming the outputs of RNA-seq analysis. Further, significant upregualtion of miR-23b was observed in wtKRAS cells. The comparison between the expression level of hub genes and TFs with expression data of CRC tissue samples deposited in TCGA databank confirmed them as distinct biomarkers for the discrimination of normal and tumor patient samples. Survival analysis revealed the significant prognostic value for some of the hub genes, TFs, and lncRNAs. The results of the present study can extend the vision on the molecular mechanisms involved in KRAS-driven CRC pathogenesis.
    DOI:  https://doi.org/10.1038/s41598-022-11697-5
  12. FASEB J. 2022 May;36 Suppl 1
      The blood-brain barrier (BBB) is a specialized microvasculature integral for brain tissue-fluid homeostasis that is comprised of brain endothelial cells (BECs) and pericytes. Disruption of the BBB and subsequent vascular leakage of protein-rich fluids is toxic to surrounding neurons and is an underlying risk factor in neurodegenerative disorders. BECs and pericytes form adhesions through the transmembrane protein Neural (N)-cadherin. Although BEC-pericyte interactions are critical for maintaining the BBB, the role of N-cadherin adhesions in regulating the BBB remain unclear. Our previous work demonstrated that mutant mice lacking Cdh2 (N-cadherin) in ECs or pericytes exhibited a size-dependent increase in BBB permeability without affecting vessel or pericyte coverage. These findings raise the possibility that N-cadherin junctions activate outside-in signaling to strengthen the BBB. Analysis of tight junction (TJ) proteins occludin and claudins 1 and 5 in microvascular BECs of the cortex demonstrated a significant reduction in the accumulation of occludin, but not claudins, at TJs of KO mice. To delineate the signaling mechanism by which N-cadherin adhesion-mediated signaling regulates occludin TJs, we utilize biomimetic surfaces (Ncdh-BioS) bearing covalently linked N-cadherin extracellular domain to induce N-cadherin adhesion in BEC monolayers in vitro. Consistent with our observations in mice, assembly of N-cadherin junctions induced the accumulation of occludin and ZO1 at TJs in BEC monolayers. Depletion of N-cadherin reversed these events, suggesting that N-cadherin adhesion-induced signaling assembles or stabilizes occludin TJs. Analysis of occludin-Dendra 2 kinetics revealed decreased internalization rates of occludin from TJs in BECs grown on Ncdh-BioS as compared to collagen, suggesting that N-cadherin signaling stabilized occludin TJs. Furthermore, formation of N-cadherin adhesion complexes led to activation of phosphoinositide 3-kinase (PI3K) signaling as evidenced by the spatial redistribution of Akt to the plasma membrane as well as Akt phosphorylation. Pharmacological inhibition of class I PI3K with Copanlisib or depletion of PI3K p110β, but not of other PI3K catalytic isoforms, abolished Akt activation and increased the rate of occludin internalization from TJs in BECs grown on Ncdh-BioS. Cumulatively, these data demonstrates that N-cadherin outside-in signaling strengthens the BBB by stabilizing occludin TJs through PI3K p110β signaling.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.R5589
  13. FASEB J. 2022 May;36 Suppl 1
      Directed cell migration is an important biological process that is necessary for the embryonic development, maintenance of multicellular organisms and immune response to external stimuli. Dysregulation of cell migration is implicated in the onset and progression of diseases including cardiovascular diseases and cancer metastasis. Many studies have identified genes and proteins that are important for directed cell migration, but the process remains incompletely understood. The mechanistic Target of Rapamycin Complex 2 (mTORC2) is a complex of proteins that have been identified as important players in cell migration. mTORC2 is known to have a role in regulating F-actin organization and actomyosin (MyoII). Moreover, there is increasing evidence that shows a correlation between mTORC2 and cancer cell migration and invasion. Despite this knowledge, the role and regulation of mTORC2 is not fully understood. Recently, we found that Rap1, a member of the Ras superfamily of small GTPases with conserved roles in cytoskeleton remodeling and cell adhesion, is a conserved binding partner of mTORC2 component RIP3/SIN1. We previously showed that Rap1 positively regulates mTORC2 activation in response to the chemoattractant cAMP in the model organism Dictyostelium discoideum. Moreover, we recently found that expression of a constitutively active Rap1 mutant (Rap1CA) potentiates the insulin-induced activation of mTORC2 in human HEK293 cells. We then undertook a study to investigate if Rap1 regulation of mTORC2 is part of a mechanism involved in regulating human cell migration. We observed that basal mTORC2 activity, as well as that induced by several potential promigratory signals, including insulin, epidermal growth factor (EGF), insulin-like growth factor-1 (IGF-1), and lysophosphatidic acid (LPA), was increased by the overexpression of wild-type Rap1 or Rap1CA. Therefore, these observations suggest that Rap1 plays a role in positively regulating mTORC2 activation in HEK293 cells in response to stimulation of promigratory signals, similar to our previous observations made in Dictyostelium. Ongoing studies now aim at determining how Rap1 regulates the function of mTORC2 in cell migration.
    DOI:  https://doi.org/10.1096/fasebj.2022.36.S1.0R360