bims-lymeca Biomed News
on Lysosome metabolism in cancer
Issue of 2022‒02‒06
eleven papers selected by
Charilaos Filippakis
Harvard University


  1. Proc Natl Acad Sci U S A. 2022 Feb 08. pii: e2114912119. [Epub ahead of print]119(6):
      Cells acquire essential nutrients from the environment and utilize adaptive mechanisms to survive when nutrients are scarce. How nutrients are trafficked and compartmentalized within cells and whether they are stored in response to stress remain poorly understood. Here, we investigate amino acid trafficking and uncover evidence for the lysosomal transit of numerous essential amino acids. We find that starvation induces the lysosomal retention of leucine in a manner requiring RAG-GTPases and the lysosomal protein complex Ragulator, but that this process occurs independently of mechanistic target of rapamycin complex 1 activity. We further find that stored leucine is utilized in protein synthesis and that inhibition of protein synthesis releases lysosomal stores. These findings identify a regulated starvation response that involves the lysosomal storage of leucine.
    Keywords:  leucine; lysosome; mTOR
    DOI:  https://doi.org/10.1073/pnas.2114912119
  2. Cancer Sci. 2022 Feb 02.
      Cancer cells secrete large amounts of extracellular vesicles (EVs) originating from multivesicular bodies (MVBs). Mature MVBs fuse either with the plasma membrane for release as EVs often referred as to exosomes or with lysosomes for degradation. However, the mechanisms regulating MVB fate remain unknown. Here, we investigated the regulators of MVB fate by analyzing the effects of signaling inhibitors on EV secretion from cancer cells engineered to secrete luciferase-labeled EVs. Inhibition of the oncogenic MEK/ERK pathway suppressed EV release and activated lysosome formation. MEK/ERK-mediated lysosomal inactivation impaired MVB degradation, resulting in increased EV secretion from cancer cells. Moreover, MEK/ERK inhibition prevented c-MYC expression and induced the nuclear translocation of MiT/TFE transcription factors, thereby promoting the activation of lysosome-related genes, including the gene encoding a subunit of vacuolar-type H+ -ATPase, which is responsible for lysosomal acidification and function. Furthermore, c-MYC upregulation was associated with lysosomal genes downregulation in MEK/ERK-activated renal cancer cells/tissues. These findings suggest that the MEK/ERK/c-MYC pathway controls MVB fate and promotes EV production in human cancers by inactivating lysosomal function.
    Keywords:  MEK/ERK; c-MYC; extracellular vesicles; lysosome; renal cancer
    DOI:  https://doi.org/10.1111/cas.15288
  3. Mol Biol Cell. 2022 Feb 02. mbcE21100526
      Lysosomal degradative compartments hydrolyze macromolecules to generate basic building blocks that fuel metabolic pathways in our cells. They also remove misfolded proteins and control size, function and number of cytoplasmic organelles via constitutive and regulated autophagy. These catabolic processes attract interest because their defective functioning is linked to human disease and their molecular components are promising pharmacologic targets. The capacity to quantitatively assess them is highly sought for. Here, we present a tandem-fluorescent reporter consisting of a HaloTag-GFP chimera appended at the C- or at the N-terminus of select polypeptides to monitor protein and organelle delivery to the lysosomal compartment. The Halo-GFP changes color upon fluorescent pulse with cell-permeable HaloTag ligands and, again, upon delivery to acidic, degradative lysosomal compartments, where the fluorescent ligand-associated HaloTag is relatively stable, whereas the GFP portion is not, as testified by loss of the green fluorescence and generation of a protease-resistant, fluorescent HaloTag fragment. The Halo-GFP tandem fluorescent reporter presented in our study allows quantitative and, crucially, time-resolved analyses of protein and organelle transport to the lysosomal compartment by high resolution confocal laser scanning microscopy, antibody-free electrophoretic techniques and flow cytometry.
    DOI:  https://doi.org/10.1091/mbc.E21-10-0526
  4. Transl Cancer Res. 2021 Mar;10(3): 1313-1323
      Background: Extracellular and cell-surface molecules remain the most common druggable cancer targets. However, intracellular therapeutic modalities are gaining momentum. The overexpression of stress-induced phosphoprotein 1 (STIP1), an adaptor protein that coordinates the functions of different chaperones in protein folding, has been reported in several solid malignancies. Here, we investigated the effects of intracellular STIP1 inhibition, attained either through the HEPES-mediated cytosolic delivery of anti-STIP1 antibodies or the use of a cell-penetrating signal-tagged peptide 520, in different human cancer cell lines and luciferase-expressing murine ovarian cancer cells (MOSEC/Luc) tumor-bearing C57BL/6 mice.Methods: The effects of STIP1 in different human cell lines were determined by cell viability, cell cytotoxicity and cell apoptosis assays. Immunoblotting was used to assess the relevant proteins found in this study and tumor xenograft mice models were also employed.
    Results: Intracellular targeting of STIP1 inhibited cancer cell line growth and promoted caspase 3-dependent apoptotic cell death. Moreover, the intracellular delivery of anti-STIP1 antibodies facilitated the degradation of STIP1 and two of its client proteins, lysine-specific demethylase 1 and Janus kinase 2. In vivo studies demonstrated that survival of mice bearing experimental tumors was improved by administration of anti-STIP1 antibodies.
    Conclusions: Our findings demonstrate that the cytosolic inhibition of STIP1 in tumor cells is feasible and provides a solid basis for further investigation of STIP1 as an intracellular cancer target. Our findings demonstrate that cytosolic inhibition of STIP1 in tumor cells is feasible and provide a solid basis for further exploration of STIP1 as an intracellular cancer target.
    Keywords:  Intracellular targeting; cancer therapy; cytosol; intracellular protein delivery; stress-induced phosphoprotein 1 (STIP1)
    DOI:  https://doi.org/10.21037/tcr-20-3333
  5. Biochem J. 2022 Feb 11. 479(3): 273-288
      Membrane traffic in eukaryotic cells is mediated by transport vesicles that bud from a precursor compartment and are transported to their destination compartment where they dock and fuse. To reach their intracellular destination, transport vesicles contain targeting signals such as Rab GTPases and polyphosphoinositides that are recognized by tethering factors in the cytoplasm and that connect the vesicles with their respective destination compartment. The final step, membrane fusion, is mediated by SNARE proteins. SNAREs are connected to targeting signals and tethering factors by multiple interactions. However, it is still debated whether SNAREs only function downstream of targeting and tethering or whether they also participate in regulating targeting specificity. Here, we review the evidence and discuss recent data supporting a role of SNARE proteins as targeting signals in vesicle traffic.
    Keywords:  membrane fusion; snare proteins; trafficking
    DOI:  https://doi.org/10.1042/BCJ20210719
  6. Histol Histopathol. 2022 Feb 02. 18426
      Microphthalmia-associated transcription factor (MITF/MiT) family is a group of basic helix-loop-helix leucine zipper (bHLH-LZ) transcription factors including TFE3 (TFEA), TFEB, TFEC and MITF. The first renal neoplasms involving MITF family translocation were renal cell carcinomas with chromosome translocations involving ASPL-TFE3/t(X;17)(p11.23;q25) or MALAT1-TFEB/t(6;11)(p21.1;q12), and now it is known as MiT family translocation RCC in 2016 WHO classification. Translocations involving MITF family genes also are found in other tumor types, such as perivascular epithelioid cell neoplasm (PEComa), Alveolar soft part sarcoma (ASPS), epithelioid hemangioendothelioma, ossifying fibromyxoid tumor (OFMT), and clear cell tumor with melanocytic differentiation and ACTIN-MITF translocation. In this review, we summarize the features of different types of neoplasms with MITF family translocations.
    DOI:  https://doi.org/10.14670/HH-18-426
  7. Front Oncol. 2021 ;11 787953
      Treatment of patients with castration-resistant prostate cancer (CRPC) remains a major clinical challenge. We previously showed that estrogenic effects contribute to CRPC progression and are primarily caused by the increased endogenous estradiol produced via highly expressed aromatase. However, the mechanism of aromatase upregulation and its role in CRPC are poorly described. In this study, we report that HeyL is aberrantly upregulated in CRPC tissues, and its expression is positively correlated with aromatase levels. HeyL overexpression increased endogenous estradiol levels and estrogen receptor-α (ERα) transcriptional activity by upregulating CYP19A1 expression, which encodes aromatase, enhancing prostate cancer stem cell (PCSC) properties in PC3 cells. Mechanistically, HeyL bound to the CYP19A1 promoter and activated its transcription. HeyL overexpression significantly promoted bicalutamide resistance in LNCaP cells, which was reversed by the aromatase inhibitor letrozole. In PC3 cells, the HeyL-aromatase axis promoted the PCSC phenotype by upregulating autophagy-related genes, while the autophagy inhibitor chloroquine (CQ) suppressed the aromatase-induced PCSC phenotype. The activated HeyL-aromatase axis promoted PCSC autophagy via ERα-mediated estrogenic effects. Taken together, our results indicated that the HeyL-aromatase axis could increase endogenous estradiol levels and activate ERα to suppress PCSC apoptosis by promoting autophagy, which enhances the understanding of how endogenous estrogenic effects influence CRPC development.
    Keywords:  CRPC; HeyL; aromatase; autophagy; prostate cancer stem cell
    DOI:  https://doi.org/10.3389/fonc.2021.787953
  8. Nat Cancer. 2021 Mar;2(3): 245-246
      
    DOI:  https://doi.org/10.1038/s43018-021-00192-x
  9. Transl Cancer Res. 2020 Mar;9(3): 1926-1930
      Background: Arsenic trioxide (ATO) has been proved useful for the treatment of acute promyelocytic leukemia (APL). Apoptosis is the result of the cytotoxic effect of ATO, apoptotic mediated cell death confirmed by DNA fragmentation and Annexin V staining. Although signaling associated with ATO-induced apoptosis has been well defined, it is still unknown whether other forms of cell death are involved in ATO-induced cell death.Methods: Western blotting, cytotoxicity assay, transmission electron microscopy were used to evaluate other forms of cell death in U251 cells.
    Results: We found that pyroptotic mediated cell death was observed in U251 cells after ATO treatment, which was confirmed by observing the increased gasdermin E (GSDME) cleavage, lactate dehydrogenase (LDH) release and transmission electron microscopy imaging. Consistent with previous results, caspase-3 was activated by ATO, which was also important for GSDME cleavage and subsequent pyroptosis.
    Conclusions: We reported that GSDME mediated pyroptosis involved in ATO induced cell death in astroglioma cells.
    Keywords:  Arsenic trioxide (ATO); gasdermin E (GSDME); pyroptosis
    DOI:  https://doi.org/10.21037/tcr.2020.02.17