@article{YangHeydarianKozjakPavlovicetal.2020, author = {Yang, Tao and Heydarian, Motaharehsadat and Kozjak-Pavlovic, Vera and Urban, Manuela and Harbottle, Richard P. and Rudel, Thomas}, title = {Folliculin Controls the Intracellular Survival and Trans-Epithelial Passage of Neisseria gonorrhoeae}, series = {Frontiers in Cellular and Infection Microbiology}, volume = {10}, journal = {Frontiers in Cellular and Infection Microbiology}, number = {422}, issn = {2235-2988}, doi = {10.3389/fcimb.2020.00422}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-211372}, year = {2020}, abstract = {Neisseria gonorrhoeae, a Gram-negative obligate human pathogenic bacterium, infects human epithelial cells and causes sexually transmitted diseases. Emerging multi-antibiotic resistant gonococci and increasing numbers of infections complicate the treatment of infected patients. Here, we used an shRNA library screen and next-generation sequencing to identify factors involved in epithelial cell infection. Folliculin (FLCN), a 64 kDa protein with a tumor repressor function was identified as a novel host factor important for N. gonorrhoeae survival after uptake. We further determined that FLCN did not affect N. gonorrhoeae adherence and invasion but was essential for its survival in the cells by modulating autophagy. In addition, FLCN was also required to maintain cell to cell contacts in the epithelial layer. In an infection model with polarized cells, FLCN inhibited the polarized localization of E-cadherin and the transcytosis of gonococci across polarized epithelial cells. In conclusion, we demonstrate here the connection between FLCN and bacterial infection and in particular the role of FLCN in the intracellular survival and transcytosis of gonococci across polarized epithelial cell layers.}, language = {en} } @phdthesis{Yang2021, author = {Yang, Tao}, title = {Functional insights into the role of a bacterial virulence factor and a host factor in Neisseria gonorrhoeae infection}, doi = {10.25972/OPUS-20895}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-208959}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2021}, abstract = {Neisseria gonorrhoeae (GC) is a human specific pathogenic bacterium. Currently, N. gonorrhoeae developed resistance to virtually all the available antibiotics used for treatment. N. gonorrhoeae starts infection by colonizing the cell surface, followed by invasion of the host cell, intracellular persistence, transcytosis and exit into the subepithelial space. Subepithelial bacteria can reach the bloodstream and disseminate to other tissues causing systemic infections, which leads to serious conditions such as arthritis and pneumonia. A number of studies have well established the host-pathogen interactions during the initial adherence and invasion steps. However, the mechanism of intracellular survival and traversal is poorly understood so far. Hence, identification of novel bacterial virulence factors and host factors involved in the host-pathogen interaction is a crucial step in understanding disease development and uncovering novel therapeutic approaches. Besides, most of the previous studies about N. gonorrhoeae were performed in the conventional cell culture. Although they have provided insights into host-pathogen interactions, much information about the native infection microenvironment, such as cell polarization and barrier function, is still missing. This work focused on determining the function of novel bacterial virulence factor NGFG_01605 and host factor (FLCN) in gonococcal infection. NGFG_01605 was identified by Tn5 transposon library screening. It is a putative U32 protease. Unlike other proteins in this family, it is not secreted and has no ex vivo protease activity. NGFG_01605 knockout decreases gonococcal survival in the epithelial cell. 3D models based on T84 cell was developed for the bacterial transmigration assay. NGFG_01605 knockout does not affect gonococcal transmigration. The novel host factor FLCN was identified by shRNA library screening in search for factors that affected gonococcal adherence and/or internalization. We discovered that FLCN did not affect N. gonorrhoeae adherence and invasion but was essential for bacterial survival. Since programmed cell death is a host defence mechanism against intracellular pathogens, we further explored apoptosis and autophagy upon gonococcal infection and determined that FLCN did not affect apoptosis but inhibited autophagy. Moreover, we found that FLCN inhibited the expression of E-cadherin. Knockdown of E- cadherin decreased the autophagy flux and supported N. gonorrhoeae survival. Both non-polarized and polarized cells are present in the cervix, and additionally, E-cadherin represents different polarization properties on these different cells. Therefore, we established 3-D models to better understand the functions of FLCN. We discovered that FLCN was critical for N. gonorrhoeae survival in the 3-D environment as well, but not through inhibiting autophagy. Furthermore, FLCN inhibits the E-cadherin expression and disturbs its polarization in the 3-D models. Since N. gonorrhoeae can cross the epithelial cell barriers through both cell-cell junctions and transcellular migration, we further explored the roles FLCN and E-cadherin played in transmigration. FLCN delayed N. gonorrhoeae transmigration, whereas the knockdown of E-cadherin increased N. gonorrhoeae transmigration. In summary, we revealed roles of the NGFG_01605 and FLCN-E-cadherin axis play in N. gonorrhoeae infection, particularly in relation to intracellular survival and transmigration. This is also the first study that connects FLCN and human-specific pathogen infection.}, language = {en} } @article{TrivanovicVolkmannStoeckletal.2023, author = {Trivanovic, Drenka and Volkmann, Noah and Stoeckl, Magdalena and Tertel, Tobias and Rudert, Maximilian and Giebel, Bernd and Herrmann, Marietta}, title = {Enhancement of immunosuppressive activity of mesenchymal stromal cells by platelet-derived factors is accompanied by apoptotic priming}, series = {Stem Cell Reviews and Reports}, volume = {19}, journal = {Stem Cell Reviews and Reports}, number = {3}, doi = {10.1007/s12015-022-10471-4}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-324669}, pages = {713-733}, year = {2023}, abstract = {The pro-inflammatory phase of bone healing, initiated by platelet activation and eventually hematoma formation, impacts bone marrow mesenchymal stromal cells (MSCs) in unknown ways. Here, we created platelet-rich plasma (PRP) hydrogels to study how platelet-derived factors modulate functional properties of encapsulated MSCs in comparison to a non-inflammatory fibrin (FBR) hydrogel environment. MSCs were isolated from human bone marrow, while PRP was collected from pooled apheresis thrombocyte concentrates and used for hydrogel preparation. After their encapsulation in hydrogels for 72 h, retrieved MSCs were analyzed for immunomodulatory activities, apoptosis, stem cell properties, senescence, CD9\(^+\), CD63\(^+\) and CD81\(^+\) extracellular vesicle (EV) release, and metabolism-related changes. PRP-hydrogels stimulated immunosuppressive functions of MSCs, along with their upregulated susceptibility to cell death in communication with PBMCs and augmented caspase 3/7 activity. We found impaired clonal growth and cell cycle progression, and more pronounced β-galactosidase activity as well as accumulation of LC3-II-positive vacuoles in PRP-MSCs. Stimuli derived from PRP-hydrogels upregulated AKT and reduced mTOR phosphorylation in MSCs, which suggests an initiation of survival-related processes. Our results showed that PRP-hydrogels might represent a metabolically stressful environment, inducing acidification of MSCs, reducing polarization of the mitochondrial membrane and increasing lipid accumulation. These features were not detected in FBR-MSCs, which showed reduced CD63\(^+\) and CD81\(^+\) EV production and maintained clonogenicity. Our data revealed that PRP-derived hematoma components cause metabolic adaptation of MSCs followed by increased immune regulatory functions. For the first time, we showed that PRP stimuli represent a survival challenge and "apoptotic priming" that are detrimental for stem cell-like growth of MSCs and important for their therapeutic consideration.}, language = {en} } @article{TolayBuchberger2021, author = {Tolay, Nazife and Buchberger, Alexander}, title = {Comparative profiling of stress granule clearance reveals differential contributions of the ubiquitin system}, series = {Life Science Alliance}, volume = {4}, journal = {Life Science Alliance}, number = {5}, doi = {10.26508/lsa.202000927}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-259810}, pages = {e202000927}, year = {2021}, abstract = {Stress granules (SGs) are cytoplasmic condensates containing untranslated mRNP complexes. They are induced by various proteotoxic conditions such as heat, oxidative, and osmotic stress. SGs are believed to protect mRNPs from degradation and to enable cells to rapidly resume translation when stress conditions subside. SG dynamics are controlled by various posttranslationalmodifications, but the role of the ubiquitin system has remained controversial. Here, we present a comparative analysis addressing the involvement of the ubiquitin system in SG clearance. Using high-resolution immuno-fluorescence microscopy, we found that ubiquitin associated to varying extent with SGs induced by heat, arsenite, H2O2, sorbitol, or combined puromycin and Hsp70 inhibitor treatment. SG-associated ubiquitin species included K48- and K63-linked conjugates, whereas free ubiquitin was not significantly enriched. Inhibition of the ubiquitin activating enzyme, deubiquitylating enzymes, the 26S proteasome and p97/VCP impaired the clearance of arsenite- and heat-induced SGs, whereas SGs induced by other stress conditions were little affected. Our data underline the differential involvement of the ubiquitin system in SG clearance, a process important to prevent the formation of disease-linked aberrant SGs.}, language = {en} } @article{StojanovićFuchsFiedleretal.2020, author = {Stojanović, Stevan D. and Fuchs, Maximilian and Fiedler, Jan and Xiao, Ke and Meinecke, Anna and Just, Annette and Pich, Andreas and Thum, Thomas and Kunz, Meik}, title = {Comprehensive bioinformatics identifies key microRNA players in ATG7-deficient lung fibroblasts}, series = {International Journal of Molecular Sciences}, volume = {21}, journal = {International Journal of Molecular Sciences}, number = {11}, issn = {1422-0067}, doi = {10.3390/ijms21114126}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-285181}, year = {2020}, abstract = {Background: Deficient autophagy has been recently implicated as a driver of pulmonary fibrosis, yet bioinformatics approaches to study this cellular process are lacking. Autophagy-related 5 and 7 (ATG5/ATG7) are critical elements of macro-autophagy. However, an alternative ATG5/ATG7-independent macro-autophagy pathway was recently discovered, its regulation being unknown. Using a bioinformatics proteome profiling analysis of ATG7-deficient human fibroblasts, we aimed to identify key microRNA (miR) regulators in autophagy. Method: We have generated ATG7-knockout MRC-5 fibroblasts and performed mass spectrometry to generate a large-scale proteomics dataset. We further quantified the interactions between various proteins combining bioinformatics molecular network reconstruction and functional enrichment analysis. The predicted key regulatory miRs were validated via quantitative polymerase chain reaction. Results: The functional enrichment analysis of the 26 deregulated proteins showed decreased cellular trafficking, increased mitophagy and senescence as the major overarching processes in ATG7-deficient lung fibroblasts. The 26 proteins reconstitute a protein interactome of 46 nodes and miR-regulated interactome of 834 nodes. The miR network shows three functional cluster modules around miR-16-5p, miR-17-5p and let-7a-5p related to multiple deregulated proteins. Confirming these results in a biological setting, serially passaged wild-type and autophagy-deficient fibroblasts displayed senescence-dependent expression profiles of miR-16-5p and miR-17-5p. Conclusions: We have developed a bioinformatics proteome profiling approach that successfully identifies biologically relevant miR regulators from a proteomics dataset of the ATG-7-deficient milieu in lung fibroblasts, and thus may be used to elucidate key molecular players in complex fibrotic pathological processes. The approach is not limited to a specific cell-type and disease, thus highlighting its high relevance in proteome and non-coding RNA research.}, language = {en} } @article{SiegmundWagnerWajant2022, author = {Siegmund, Daniela and Wagner, Jennifer and Wajant, Harald}, title = {TNF receptor associated factor 2 (TRAF2) signaling in cancer}, series = {Cancers}, volume = {14}, journal = {Cancers}, number = {16}, issn = {2072-6694}, doi = {10.3390/cancers14164055}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-286073}, year = {2022}, abstract = {Tumor necrosis factor (TNF) receptor associated factor-2 (TRAF2) has been originally identified as a protein interacting with TNF receptor 2 (TNFR2) but also binds to several other receptors of the TNF receptor superfamily (TNFRSF). TRAF2, often in concert with other members of the TRAF protein family, is involved in the activation of the classical NFκB pathway and the stimulation of various mitogen-activated protein (MAP) kinase cascades by TNFRSF receptors (TNFRs), but is also required to inhibit the alternative NFκB pathway. TRAF2 has also been implicated in endoplasmic reticulum (ER) stress signaling, the regulation of autophagy, and the control of cell death programs. TRAF2 fulfills its functions by acting as a scaffold, bringing together the E3 ligase cellular inhibitor of apoptosis-1 (cIAP1) and cIAP2 with their substrates and various regulatory proteins, e.g., deubiquitinases. Furthermore, TRAF2 can act as an E3 ligase by help of its N-terminal really interesting new gene (RING) domain. The finding that TRAF2 (but also several other members of the TRAF family) interacts with the latent membrane protein 1 (LMP1) oncogene of the Epstein-Barr virus (EBV) indicated early on that TRAF2 could play a role in the oncogenesis of B-cell malignancies and EBV-associated non-keratinizing nasopharyngeal carcinoma (NPC). TRAF2 can also act as an oncogene in solid tumors, e.g., in colon cancer by promoting Wnt/β-catenin signaling. Moreover, tumor cell-expressed TRAF2 has been identified as a major factor-limiting cancer cell killing by cytotoxic T-cells after immune checkpoint blockade. However, TRAF2 can also be context-dependent as a tumor suppressor, presumably by virtue of its inhibitory effect on the alternative NFκB pathway. For example, inactivating mutations of TRAF2 have been associated with tumor development, e.g., in multiple myeloma and mantle cell lymphoma. In this review, we summarize the various TRAF2-related signaling pathways and their relevance for the oncogenic and tumor suppressive activities of TRAF2. Particularly, we discuss currently emerging concepts to target TRAF2 for therapeutic purposes.}, language = {en} } @article{ScherzadMeyerKleinsasseretal.2017, author = {Scherzad, Agmal and Meyer, Till and Kleinsasser, Norbert and Hackenberg, Stephan}, title = {Molecular Mechanisms of Zinc Oxide Nanoparticle-Induced Genotoxicity Short Running Title: Genotoxicity of ZnO NPs}, series = {Materials}, volume = {10}, journal = {Materials}, number = {12}, doi = {10.3390/ma10121427}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-169948}, pages = {1427}, year = {2017}, abstract = {Background: Zinc oxide nanoparticles (ZnO NPs) are among the most frequently applied nanomaterials in consumer products. Evidence exists regarding the cytotoxic effects of ZnO NPs in mammalian cells; however, knowledge about the potential genotoxicity of ZnO NPs is rare, and results presented in the current literature are inconsistent. Objectives: The aim of this review is to summarize the existing data regarding the DNA damage that ZnO NPs induce, and focus on the possible molecular mechanisms underlying genotoxic events. Methods: Electronic literature databases were systematically searched for studies that report on the genotoxicity of ZnO NPs. Results: Several methods and different endpoints demonstrate the genotoxic potential of ZnO NPs. Most publications describe in vitro assessments of the oxidative DNA damage triggered by dissoluted Zn2+ ions. Most genotoxicological investigations of ZnO NPs address acute exposure situations. Conclusion: Existing evidence indicates that ZnO NPs possibly have the potential to damage DNA. However, there is a lack of long-term exposure experiments that clarify the intracellular bioaccumulation of ZnO NPs and the possible mechanisms of DNA repair and cell survival.}, language = {en} } @phdthesis{Riedel2007, author = {Riedel, Alexander}, title = {Untersuchungen zur endogenen MHC-Klasse-II-restringierten Pr{\"a}sentation nukle{\"a}rer Antigene}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-25183}, school = {Universit{\"a}t W{\"u}rzburg}, year = {2007}, abstract = {Die endogene Pr{\"a}sentation von intrazellul{\"a}ren Antigenen auf Major-Histokompatibilit{\"a}tskomplex Klasse-II (MHC-II) -Molek{\"u}len ist von entscheidender Bedeutung f{\"u}r eine Reihe von immunologischen Prozessen. Die mechanistischen Grundlagen dieses Pr{\"a}sentationsweges sind aber noch weitgehend unverstanden. Ziel dieser Arbeit war es, einen Beitrag zum molekularen Verst{\"a}ndnis der Abl{\"a}ufe zu leisten, die an der endogenen Pr{\"a}sentation nukle{\"a}rer Antigene auf MHC-II-Molek{\"u}len beteiligt sind. Dazu sollte am Beispiel des nukle{\"a}r lokalisierten Modellantigens Neomycin-Phosphotransferase II (NucNeoR) sowie des viralen Kernantigens Epstein-Barr-virus nuclear antigen 3C (EBNA3C) und entsprechender antigenspezifischer MHC-II-restringierter CD4+ T-Zellen die verantwortlichen Pr{\"a}sentationswege in professionell und nicht-professionell antigenpr{\"a}sentierenden Zellen untersucht werden. In beiden Zellsystemen wurde NucNeoR {\"u}ber einen endogenen Pr{\"a}sentationsweg und nicht {\"u}ber die Freisetzung und Wiederaufnahme als exogenes Protein auf MHC-II-Molek{\"u}len pr{\"a}sentiert. Durch die Verwendung chemischer Inhibitoren konnte eine Beteiligung der Autophagie an der endogenen Antigenpr{\"a}sentation nachgewiesen werden. Da Autophagie ausschließlich im Zytoplasma stattfindet, wurde nach m{\"o}glichen Eintrittspforten f{\"u}r nukle{\"a}re Proteine in diesen Abbauweg gesucht. F{\"u}r die Autophagie-abh{\"a}ngige Pr{\"a}sentation von NucNeoR war weder ein CRM1-vermittelter aktiver Export des Antigens aus dem Kern ins Zytoplasma, noch eine Aufl{\"o}sung der Kernmembran im Rahmen der Zellteilung und der dadurch bedingten Durchmischung nukle{\"a}rer und zytoplasmatischer Bestandteile notwendig. Mit Hilfe eines konditionalen Antigenexpressionsystems und der Auftrennung antigenexprimierender Zellen nach Zellzyklusphasen konnte eine verst{\"a}rkte Antigenpr{\"a}sentation in der G1/0-Phase nachgewiesen werden, die mit fortschreitendem Zellzyklus immer mehr abnahm. Die Antigenpr{\"a}sentation korrelierte dabei mit der ebenfalls im Laufe des Zellzyklus abnehmenden Transkriptions- bzw. Translationsrate des Antigens, aber nicht mit der absoluten Menge an Antigen in den Zellen. Bei abgeschalteter Antigentranskription dagegen korrelierte die Antigenpr{\"a}sentation mit der MHC-II-Oberfl{\"a}chenexpression, die von der G1/0- bis hin zur G2/M-Phase kontinuierlich zunahm. Eine {\"a}hnliche Korrelation von Antigentranskription/ Antigentranslation und Autophagie-abh{\"a}ngiger Antigenpr{\"a}sentation wurde auch f{\"u}r EBNA3C und die zytoplasmatisch lokalisierte NeoR-Variante beobachtet. Diese Ergebnisse identifizieren die Autophagie-abh{\"a}ngige Pr{\"a}sentation neusynthetisierter Proteine als den verantwortlichen molekularen Mechanismus f{\"u}r die endogene Pr{\"a}sentation der untersuchten nukle{\"a}ren Antigene auf MHC-II-Molek{\"u}len. Durch die Kopplung von Translation und autophagischem Abbau erlangen Proteine unabh{\"a}ngig von ihrer subzellul{\"a}ren Lokalisation Zugang zu diesem Pr{\"a}sentationsweg und erweitern so das Spektrum der intrazellul{\"a}ren Antigene, die einer CD4+ T-Zell{\"u}berwachung unterliegen.}, subject = {Autophagie}, language = {de} } @article{OttoHahlbrockEichetal.2016, author = {Otto, Christoph and Hahlbrock, Theresa and Eich, Kilian and Karaaslan, Ferdi and J{\"u}rgens, Constantin and Germer, Christoph-Thomas and Wiegering, Armin and K{\"a}mmerer, Ulrike}, title = {Antiproliferative and antimetabolic effects behind the anticancer property of fermented wheat germ extract}, series = {BMC Complementary and Alternative Medicine}, volume = {16}, journal = {BMC Complementary and Alternative Medicine}, number = {160}, doi = {10.1186/s12906-016-1138-5}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-146013}, year = {2016}, abstract = {Background Fermented wheat germ extract (FWGE) sold under the trade name Avemar exhibits anticancer activity in vitro and in vivo. Its mechanisms of action are divided into antiproliferative and antimetabolic effects. Its influcence on cancer cell metabolism needs further investigation. One objective of this study, therefore, was to further elucidate the antimetabolic action of FWGE. The anticancer compound 2,6-dimethoxy-1,4-benzoquinone (DMBQ) is the major bioactive compound in FWGE and is probably responsible for its anticancer activity. The second objective of this study was to compare the antiproliferative properties in vitro of FWGE and the DMBQ compound. Methods The IC\(_{50}\) values of FWGE were determined for nine human cancer cell lines after 24 h of culture. The DMBQ compound was used at a concentration of 24 μmol/l, which is equal to the molar concentration of DMBQ in FWGE. Cell viability, cell cycle, cellular redox state, glucose consumption, lactic acid production, cellular ATP levels, and the NADH/NAD\(^+\) ratio were measured. Results The mean IC\(_{50}\) value of FWGE for the nine human cancer cell lines tested was 10 mg/ml. Both FWGE (10 mg/ml) and the DMBQ compound (24 μmol/l) induced massive cell damage within 24 h after starting treatment, with changes in the cellular redox state secondary to formation of intracellular reactive oxygen species. Unlike the DMBQ compound, which was only cytotoxic, FWGE exhibited cytostatic and growth delay effects in addition to cytotoxicity. Both cytostatic and growth delay effects were linked to impaired glucose utilization which influenced the cell cycle, cellular ATP levels, and the NADH/NAD\(^+\) ratio. The growth delay effect in response to FWGE treatment led to induction of autophagy. Conclusions FWGE and the DMBQ compound both induced oxidative stress-promoted cytotoxicity. In addition, FWGE exhibited cytostatic and growth delay effects associated with impaired glucose utilization which led to autophagy, a possible previously unknown mechanism behind the influence of FWGE on cancer cell metabolism.}, language = {en} } @article{OelschlaegelWeissSadanSalpeteretal.2020, author = {Oelschlaegel, Diana and Weiss Sadan, Tommy and Salpeter, Seth and Krug, Sebastian and Blum, Galia and Schmitz, Werner and Schulze, Almut and Michl, Patrick}, title = {Cathepsin inhibition modulates metabolism and polarization of tumor-associated macrophages}, series = {Cancers}, volume = {12}, journal = {Cancers}, number = {9}, issn = {2072-6694}, doi = {10.3390/cancers12092579}, url = {http://nbn-resolving.de/urn:nbn:de:bvb:20-opus-213040}, year = {2020}, abstract = {Stroma-infiltrating immune cells, such as tumor-associated macrophages (TAM), play an important role in regulating tumor progression and chemoresistance. These effects are mostly conveyed by secreted mediators, among them several cathepsin proteases. In addition, increasing evidence suggests that stroma-infiltrating immune cells are able to induce profound metabolic changes within the tumor microenvironment. In this study, we aimed to characterize the impact of cathepsins in maintaining the TAM phenotype in more detail. For this purpose, we investigated the molecular effects of pharmacological cathepsin inhibition on the viability and polarization of human primary macrophages as well as its metabolic consequences. Pharmacological inhibition of cathepsins B, L, and S using a novel inhibitor, GB111-NH\(_2\), led to changes in cellular recycling processes characterized by an increased expression of autophagy- and lysosome-associated marker genes and reduced adenosine triphosphate (ATP) content. Decreased cathepsin activity in primary macrophages further led to distinct changes in fatty acid metabolites associated with increased expression of key modulators of fatty acid metabolism, such as fatty acid synthase (FASN) and acid ceramidase (ASAH1). The altered fatty acid profile was associated with an increased synthesis of the pro-inflammatory prostaglandin PGE\(_2\), which correlated with the upregulation of numerous NF\(_k\)B-dependent pro-inflammatory mediators, including interleukin-1 (IL-1), interleukin-6 (IL-6), C-C motif chemokine ligand 2 (CCL2), and tumor necrosis factor-alpha (TNFα). Our data indicate a novel link between cathepsin activity and metabolic reprogramming in macrophages, demonstrated by a profound impact on autophagy and fatty acid metabolism, which facilitates a pro-inflammatory micromilieu generally associated with enhanced tumor elimination. These results provide a strong rationale for therapeutic cathepsin inhibition to overcome the tumor-promoting effects of the immune-evasive tumor micromilieu.}, language = {en} }