Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
Cancer Res Commun ; 3(5): 943-951, 2023 05.
Article En | MEDLINE | ID: mdl-37377894

Interferons (IFNs) are cytokines with potent antineoplastic and antiviral properties. IFNα has significant clinical activity in the treatment of myeloproliferative neoplasms (MPN), but the precise mechanisms by which it acts are not well understood. Here, we demonstrate that chromatin assembly factor 1 subunit B (CHAF1B), an Unc-51-like kinase 1 (ULK1)-interactive protein in the nuclear compartment of malignant cells, is overexpressed in patients with MPN. Remarkably, targeted silencing of CHAF1B enhances transcription of IFNα-stimulated genes and promotes IFNα-dependent antineoplastic responses in primary MPN progenitor cells. Taken together, our findings indicate that CHAF1B is a promising newly identified therapeutic target in MPN and that CHAF1B inhibition in combination with IFNα therapy might offer a novel strategy for treating patients with MPN. Significance: Our findings raise the potential for clinical development of drugs targeting CHAF1B to enhance IFN antitumor responses in the treatment of patients with MPN and should have important clinical translational implications for the treatment of MPN and possibly in other malignancies.


Bone Marrow Neoplasms , Myeloproliferative Disorders , Neoplasms , Humans , Myeloproliferative Disorders/drug therapy , Interferon-alpha/pharmacology , Chromatin Assembly Factor-1/genetics
2.
Bioessays ; 45(3): e2200203, 2023 03.
Article En | MEDLINE | ID: mdl-36642848

Interferons (IFNs) are a diverse group of cytokines whose potent antitumor effects have piqued the interest of scientists for decades. Some of the most sustained clinical accomplishments have been in the field of myeloproliferative neoplasms (MPNs). Here, we discuss how both historical and novel breakthroughs in our understanding of IFN function may lead to more effective therapies for MPNs. The particular relevance and importance of modulating the novel IFN-regulated ULK1 pathway to optimize IFN responses is highlighted.


Hematologic Neoplasms , Interferons , Humans , Interferons/therapeutic use , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/pathology , Hematologic Neoplasms/drug therapy
3.
Mol Cancer Res ; 21(4): 332-344, 2023 04 01.
Article En | MEDLINE | ID: mdl-36573964

Immune checkpoint inhibitors (ICI) have transformed the treatment of melanoma. However, the majority of patients have primary or acquired resistance to ICIs, limiting durable responses and patient survival. IFNγ signaling and the expression of IFNγ-stimulated genes correlate with either response or resistance to ICIs, in a context-dependent manner. While IFNγ-inducible immunostimulatory genes are required for response to ICIs, chronic IFNγ signaling induces the expression of immunosuppressive genes, promoting resistance to these therapies. Here, we show that high levels of Unc-51 like kinase 1 (ULK1) correlate with poor survival in patients with melanoma and overexpression of ULK1 in melanoma cells enhances IFNγ-induced expression of immunosuppressive genes, with minimal effects on the expression of immunostimulatory genes. In contrast, genetic or pharmacologic inhibition of ULK1 reduces expression of IFNγ-induced immunosuppressive genes. ULK1 binds IRF1 in the nuclear compartment of melanoma cells, controlling its binding to the programmed death-ligand 1 promoter region. In addition, pharmacologic inhibition of ULK1 in combination with anti-programmed cell death protein 1 therapy further reduces melanoma tumor growth in vivo. Our data suggest that targeting ULK1 represses IFNγ-dependent immunosuppression. These findings support the combination of ULK1 drug-targeted inhibition with ICIs for the treatment of patients with melanoma to improve response rates and patient outcomes. IMPLICATIONS: This study identifies ULK1, activated downstream of IFNγ signaling, as a druggable target to overcome resistance mechanisms to ICI therapy in metastatic melanoma.


Immune Checkpoint Inhibitors , Melanoma , Humans , Immune Checkpoint Inhibitors/therapeutic use , Melanoma/drug therapy , Melanoma/genetics , Interferon-gamma/pharmacology , Immunosuppression Therapy , Immune Tolerance , Autophagy-Related Protein-1 Homolog/genetics , Intracellular Signaling Peptides and Proteins/genetics
4.
Mol Immunol ; 147: 1-9, 2022 07.
Article En | MEDLINE | ID: mdl-35489289

The novel coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been shown to hijack angiotensin converting enzyme 2 (ACE2) for entry into mammalian cells. A short isoform of ACE2, termed deltaACE2 (dACE2), has recently been identified. In contrast to ACE2, the short dACE2 isoform lacks the ability to bind the spike protein of SARS-CoV-2. Several studies have proposed that expression of ACE2 and/or dACE2 is induced by interferons (IFNs). Here, we report that drug-targeted inhibition or silencing of Unc51-like kinase 1 (ULK1) results in repression of type I IFN-induced expression of the dACE2 isoform. Notably, dACE2 is expressed in various squamous tumors. In efforts to identify pharmacological agents that target this pathway, we found that fisetin, a natural flavonoid, is an ULK1 inhibitor that decreases type I IFN-induced dACE2 expression. Taken together, our results establish a requirement for ULK1 in the regulation of type I IFN-induced transcription of dACE2 and raise the possibility of clinical translational applications of fisetin as a novel ULK1 inhibitor.


Angiotensin-Converting Enzyme 2 , COVID-19 , Animals , Interferon-alpha , Mammals , Protein Isoforms/genetics , Protein Isoforms/metabolism , SARS-CoV-2
5.
Nat Commun ; 13(1): 1750, 2022 04 01.
Article En | MEDLINE | ID: mdl-35365653

Interferons (IFNs) are key initiators and effectors of the immune response against malignant cells and also directly inhibit tumor growth. IFNα is highly effective in the treatment of myeloproliferative neoplasms (MPNs), but the mechanisms of action are unclear and it remains unknown why some patients respond to IFNα and others do not. Here, we identify and characterize a pathway involving PKCδ-dependent phosphorylation of ULK1 on serine residues 341 and 495, required for subsequent activation of p38 MAPK. We show that this pathway is essential for IFN-suppressive effects on primary malignant erythroid precursors from MPN patients, and that increased levels of ULK1 and p38 MAPK correlate with clinical response to IFNα therapy in these patients. We also demonstrate that IFNα treatment induces cleavage/activation of the ULK1-interacting ROCK1/2 proteins in vitro and in vivo, triggering a negative feedback loop that suppresses IFN responses. Overexpression of ROCK1/2 is seen in MPN patients and their genetic or pharmacological inhibition enhances IFN-anti-neoplastic responses in malignant erythroid precursors from MPN patients. These findings suggest the clinical potential of pharmacological inhibition of ROCK1/2 in combination with IFN-therapy for the treatment of MPNs.


Myeloproliferative Disorders , Neoplasms , Antiviral Agents/therapeutic use , Feedback , Humans , Interferon-alpha/pharmacology , Interferon-alpha/therapeutic use , Myeloproliferative Disorders/drug therapy , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/metabolism , Neoplasms/drug therapy , Signal Transduction , rho-Associated Kinases/metabolism
6.
Oncotarget ; 12(10): 955-966, 2021 May 11.
Article En | MEDLINE | ID: mdl-34012509

The MAPK-interacting kinases 1 and 2 (MNK1/2) have generated increasing interest as therapeutic targets for acute myeloid leukemia (AML). We evaluated the therapeutic potential of the highly-selective MNK1/2 inhibitor Tomivosertib on AML cells. Tomivosertib was highly effective at blocking eIF4E phosphorylation on serine 209 in AML cells. Such inhibitory effects correlated with dose-dependent suppression of cellular viability and leukemic progenitor colony formation. Moreover, combination of Tomivosertib and Venetoclax resulted in synergistic anti-leukemic responses in AML cell lines. Mass spectrometry studies identified novel putative MNK1/2 interactors, while in parallel studies we demonstrated that MNK2 - RAPTOR - mTOR complexes are not disrupted by Tomivosertib. Overall, these findings demonstrate that Tomivosertib exhibits potent anti-leukemic properties on AML cells and support the development of clinical translational efforts involving the use of this drug, alone or in combination with other therapies for the treatment of AML.

7.
Oncogene ; 40(18): 3273-3286, 2021 05.
Article En | MEDLINE | ID: mdl-33846574

We provide evidence that a member of the human Schlafen (SLFN) family of proteins, SLFN5, is overexpressed in human pancreatic ductal adenocarcinoma (PDAC). Targeted deletion of SLFN5 results in decreased PDAC cell proliferation and suppresses PDAC tumorigenesis in in vivo PDAC models. Importantly, high expression levels of SLFN5 correlate with worse outcomes in PDAC patients, implicating SLFN5 in the pathophysiology of PDAC that leads to poor outcomes. Our studies establish novel regulatory effects of SLFN5 on cell cycle progression through binding/blocking of the transcriptional repressor E2F7, promoting transcription of key genes that stimulate S phase progression. Together, our studies suggest an essential role for SLFN5 in PDAC and support the potential for developing new therapeutic approaches for the treatment of pancreatic cancer through SLFN5 targeting.


Pancreatic Neoplasms , Carcinogenesis , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Pancreatic Neoplasms
8.
Cancers (Basel) ; 13(5)2021 Mar 02.
Article En | MEDLINE | ID: mdl-33801234

The interferons (IFNs) are essential components of the immune response against infections and malignancies. IFNs are potent promoters of the anti-tumor response, but there is also evidence that feedback mechanisms regulated by IFNs negatively control immune responses to avoid hyper-activation and limit inflammation. This balance of responses plays an important role in cancer surveillance, immunoediting and response to anticancer therapeutic approaches. Here we review the roles of both type I and type II IFNs on the control of the immune response against malignancies in the context of effects on both malignant cells and cells of the immune system in the tumor microenvironment.

9.
Cancers (Basel) ; 13(4)2021 Feb 16.
Article En | MEDLINE | ID: mdl-33669271

Cytokines are key molecules within the tumor microenvironment (TME) that can be used as biomarkers to predict the magnitude of anti-tumor immune responses. During immune monitoring, it has been customary to predict outcomes based on the abundance of a single cytokine, in particular IFN-γ or TGF-ß, as a readout of ongoing anti-cancer immunity. However, individual cytokines within the TME can exhibit dual opposing roles. For example, both IFN-γ and TGF-ß have been associated with pro- and anti-tumor functions. Moreover, cytokines originating from different cellular sources influence the crosstalk between CD4+ and CD8+ T cells, while the array of cytokines expressed by T cells is also instrumental in defining the mechanisms of action and efficacy of treatments. Thus, it becomes increasingly clear that a reliable readout of ongoing immunity within the TME will have to include more than the measurement of a single cytokine. This review focuses on defining a panel of cytokines that could help to reliably predict and analyze the outcomes of T cell-based anti-tumor therapies.

10.
Semin Immunol ; 43: 101299, 2019 06.
Article En | MEDLINE | ID: mdl-31771762

The interferons (IFNs) are cytokines with important antineoplastic and immune modulatory effects. These cytokines have been conserved through evolution as important elements of the immune surveillance against cancer. Despite this, defining their precise and specific roles in the generation of antitumor responses remains challenging. Emerging evidence suggests the existence of previously unknown roles for IFNs in the control of the immune response against cancer that may redefine our understanding on how these cytokines function. Beyond the engagement of classical JAK-STAT signaling pathways that promote transcription and expression of gene products, the IFNs engage multiple other signaling cascades to generate products that mediate biological responses and outcomes. There is recent emerging evidence indicating that IFNs control the expression of both traditional immune checkpoints like the PD-L1/PD1 axis, but also less well understood "intracellular" immune checkpoints whose targeting may define new approaches for the treatment of malignancies.


Immunotherapy/trends , Interferons/metabolism , Neoplasms/immunology , Animals , B7-H1 Antigen/metabolism , Humans , Immunity , Immunologic Surveillance , Programmed Cell Death 1 Receptor/metabolism , Signal Transduction
11.
Oncotarget ; 10(67): 7112-7121, 2019 Dec 24.
Article En | MEDLINE | ID: mdl-31903169

MAPK interacting kinase (MNK), a downstream effector of mitogen-activated protein kinase (MAPK) pathways, activates eukaryotic translation initiation factor 4E (eIF4E) and plays a key role in the mRNA translation of mitogenic and antiapoptotic genes in acute myeloid leukemia (AML) cells. We examined the antileukemic properties of a novel MNK inhibitor, SEL201. Our studies provide evidence that SEL201 suppresses eIF4E phosphorylation on Ser209 in AML cell lines and in primary patient-derived AML cells. Such effects lead to growth inhibitory effects and leukemic cell apoptosis, as well as suppression of leukemic progenitor colony formation. Combination of SEL201 with 5'-azacytidine or rapamycin results in synergistic inhibition of AML cell growth. Collectively, these results suggest that SEL201 has significant antileukemic activity and further underscore the relevance of the MNK pathway in leukemogenesis.

12.
J Biol Chem ; 294(3): 827-837, 2019 01 18.
Article En | MEDLINE | ID: mdl-30487288

Type I interferons (IFNs) induce expression of multiple genes that control innate immune responses to invoke both antiviral and antineoplastic activities. Transcription of these interferon-stimulated genes (ISGs) occurs upon activation of the canonical Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathways. Phosphorylation and acetylation are both events crucial to tightly regulate expression of ISGs. Here, using mouse embryonic fibroblasts and an array of biochemical methods including immunoblotting and kinase assays, we show that sirtuin 2 (SIRT2), a member of the NAD-dependent protein deacetylase family, is involved in type I IFN signaling. We found that SIRT2 deacetylates cyclin-dependent kinase 9 (CDK9) in a type I IFN-dependent manner and that the CDK9 deacetylation is essential for STAT1 phosphorylation at Ser-727. We also found that SIRT2 is subsequently required for the transcription of ISGs and for IFN-driven antiproliferative responses in both normal and malignant cells. These findings establish the existence of a previously unreported signaling pathway whose function is essential for the control of JAK-STAT signaling and the regulation of IFN responses. Our findings suggest that targeting sirtuin activities may offer an avenue in the development of therapies for managing immune-related diseases and cancer.


Cyclin-Dependent Kinase 9/metabolism , Interferon Type I/metabolism , STAT1 Transcription Factor/metabolism , Signal Transduction , Sirtuin 2/metabolism , Acetylation , Animals , Cyclin-Dependent Kinase 9/genetics , Humans , Interferon Type I/genetics , Mice , Mice, Knockout , Phosphorylation , STAT1 Transcription Factor/genetics , Sirtuin 2/genetics , Transcription, Genetic , U937 Cells
13.
Sci Signal ; 11(557)2018 11 20.
Article En | MEDLINE | ID: mdl-30459284

It is well established that activation of the transcription factor signal transducer and activator of transcription 1 (STAT1) is required for the interferon-γ (IFN-γ)-mediated antiviral response. Here, we found that IFN-γ receptor stimulation also activated Unc-51-like kinase 1 (ULK1), an initiator of Beclin-1-mediated autophagy. Furthermore, the interaction between ULK1 and the mitogen-activated protein kinase kinase kinase MLK3 (mixed lineage kinase 3) was necessary for MLK3 phosphorylation and downstream activation of the kinase ERK5. This autophagy-independent activity of ULK1 promoted the transcription of key antiviral IFN-stimulated genes (ISGs) and was essential for IFN-γ-dependent antiviral effects. These findings define a previously unknown IFN-γ pathway that appears to be a key element of the antiviral response.


Autophagy-Related Protein-1 Homolog/metabolism , Interferon-gamma/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , MAP Kinase Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase 7/metabolism , Animals , Autophagy , Beclin-1/metabolism , Cell Line, Tumor , Class III Phosphatidylinositol 3-Kinases/metabolism , Cytokines/metabolism , Gene Expression Profiling , Humans , Immunity, Innate , Mice , Multigene Family , Phosphorylation , Protein Binding , Receptors, Interferon/metabolism , Recombinant Proteins/metabolism , Signal Transduction , Transcription, Genetic , U937 Cells , Virus Diseases/metabolism , Interferon gamma Receptor , Mitogen-Activated Protein Kinase Kinase Kinase 11
14.
Mol Cell Biol ; 38(16)2018 08 15.
Article En | MEDLINE | ID: mdl-29866656

Although members of the Slfn family have been implicated in the regulation of type I interferon (IFN) responses, the mechanisms by which they mediate their effects remain unknown. In the present study, we provide evidence that targeted disruption of the Slfn2 gene leads to increased transcription of IFN-stimulated genes (ISGs) and enhanced type I IFN-mediated antiviral responses. We demonstrate that Slfn2 interacts with protein phosphatase 6 regulatory subunit 1 (PPP6R1), leading to reduced type I IFN-induced activation of nuclear factor kappa B (NF-κB) signaling, resulting in reduced expression of ISGs. Altogether, these data suggest a novel mechanism by which Slfn2 controls ISG expression and provide evidence for a critical role for Slfn2 in the regulation of IFN-mediated biological responses.


Cell Cycle Proteins/metabolism , Interferon Type I/metabolism , NF-kappa B/metabolism , Animals , Binding Sites/genetics , Cell Cycle Proteins/deficiency , Cell Cycle Proteins/genetics , Cells, Cultured , Gene Expression , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , NIH 3T3 Cells , Phosphoprotein Phosphatases/metabolism , Promoter Regions, Genetic , Signal Transduction
15.
J Biol Chem ; 292(11): 4743-4752, 2017 03 17.
Article En | MEDLINE | ID: mdl-28174303

The precise signaling mechanisms by which type II IFN receptors control expression of unique genes to induce biological responses remain to be established. We provide evidence that Sin1, a known element of the mammalian target of rapamycin complex 2 (mTORC2), is required for IFNγ-induced phosphorylation and activation of AKT and that such activation mediates downstream regulation of mTORC1 and its effectors. These events play important roles in the assembly of the eukaryotic translation initiation factor 4F (eIF4F) and mRNA translation of IFN-stimulated genes. Interestingly, IFNγ-induced tyrosine phosphorylation of STAT1 is reduced in cells with targeted disruption of Sin1, leading to decreased transcription of several IFNγ-inducible genes in an mTORC2-independent manner. Additionally, our studies establish that Sin1 is essential for generation of type II IFN-dependent antiviral effects and antiproliferative responses in normal and malignant hematopoiesis. Together, our findings establish an important role for Sin1 in both transcription and translation of IFN-stimulated genes and type II IFN-mediated biological responses, involving both mTORC2-dependent and -independent functions.


Adaptor Proteins, Signal Transducing/immunology , Carrier Proteins/immunology , Interferon-gamma/immunology , Animals , Cell Line , Humans , Immunity, Innate , Mice , Phosphorylation , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/immunology , STAT1 Transcription Factor/chemistry , STAT1 Transcription Factor/immunology , Signal Transduction
16.
Cytokine ; 89: 116-121, 2017 01.
Article En | MEDLINE | ID: mdl-27094611

Dysregulation of mRNA translation leads to aberrant activation of cellular pathways that promote expansion and survival of leukemic clones. A key element of the initiation translation complex is eIF4E (eukaryotic translation initiation factor 4E). The mitogen-activated protein kinase (MAPK) and mammalian target of rapamycin (mTOR) pathways play important roles in the regulation of eIF4E expression and downstream functional outcomes. Mitogen-activated protein kinase interacting protein kinases (Mnks) control translation by phosphorylation of eIF4E, whereas the mTOR kinase phosphorylates/de-activates the eIF4E inhibitor, 4E-BP1, to release translational repression. Both pathways are often abnormally activated in leukemia cells and promote cell survival events by controlling expression of oncogenic proteins. Targeting these pathways may provide approaches to avoid aberrant proliferation and neoplastic transformation.


Eukaryotic Initiation Factor-4E/metabolism , Leukemia, Myeloid, Acute/metabolism , Neoplasm Proteins/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Eukaryotic Initiation Factor-4E/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Protein Kinase Inhibitors/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/genetics
17.
Blood ; 128(3): 410-4, 2016 07 21.
Article En | MEDLINE | ID: mdl-27307295

Mitogen-activated protein kinase interacting protein kinases (Mnks) play important roles in the development and progression of acute myeloid leukemia (AML) by regulating eukaryotic translation initiation factor 4E (eIF4E) activation. Inhibiting Mnk1/2-induced phosphorylation of eIF4E may represent a unique approach for the treatment of AML. We provide evidence for antileukemic effects of merestinib, an orally bioavailable multikinase inhibitor with suppressive effects on Mnk activity. Our studies show that merestinib effectively blocks eIF4E phosphorylation in AML cells and suppresses primitive leukemic progenitors from AML patients in vitro and in an AML xenograft model in vivo. Our findings provide evidence for potent preclinical antileukemic properties of merestinib and support its clinical development for the treatment of patients with AML.


Adenosine Triphosphatases/antagonists & inhibitors , Cation Transport Proteins/antagonists & inhibitors , Enzyme Inhibitors/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Neoplasm Proteins/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Animals , Cation Transport Proteins/metabolism , Cell Line, Tumor , Copper-Transporting ATPases , Eukaryotic Initiation Factor-4E/metabolism , Humans , Leukemia, Myeloid, Acute/enzymology , Mice , Neoplasm Proteins/metabolism , Xenograft Model Antitumor Assays
18.
Cytokine Growth Factor Rev ; 29: 17-22, 2016 06.
Article En | MEDLINE | ID: mdl-27068414

The human serine/threonine kinase ULK1 is the human homolog of the Caenorhabditis elegans Unc-51 kinase and of the Saccharomyces cerevisiae autophagy-related protein kinase Atg1. As Unc-51 and Atg1, ULK1 regulates both axon growth and autophagy, respectively, in mammalian cells. However, a novel immunoregulatory role of ULK1 has been recently described. This kinase was shown to be required for regulation of both type I interferon (IFN) production and induction of type I IFN signaling. Optimal regulation of IFN production is crucial for generation of effective IFN-immune responses, and defects in such networks can be detrimental for the host leading to uncontrolled pathogen infection, tumor growth, or autoimmune diseases. Thus, ULK1 plays a central role in IFN-dependent immunity. Here we review the diverse roles of ULK1, with special focus on its importance to type I IFN signaling, and highlight important future study questions.


Autophagy-Related Protein-1 Homolog/immunology , Autophagy/immunology , Interferon Type I/immunology , Intracellular Signaling Peptides and Proteins/immunology , Signal Transduction/immunology , Animals , Autoimmune Diseases/immunology , Autophagy-Related Proteins/immunology , Caenorhabditis elegans/immunology , Caenorhabditis elegans Proteins/immunology , Humans , Infections/immunology , Neoplasms/immunology , Protein Kinases/immunology , Protein Serine-Threonine Kinases/immunology , Saccharomyces cerevisiae/immunology , Saccharomyces cerevisiae Proteins/immunology
19.
J Biol Chem ; 291(5): 2389-96, 2016 Jan 29.
Article En | MEDLINE | ID: mdl-26645692

We provide evidence for a unique pathway engaged by the type II IFN receptor, involving mTORC2/AKT-mediated downstream regulation of mTORC1 and effectors. These events are required for formation of the eukaryotic translation initiation factor 4F complex (eIF4F) and initiation of mRNA translation of type II interferon-stimulated genes. Our studies establish that Rictor is essential for the generation of type II IFN-dependent antiviral and antiproliferative responses and that it controls the generation of type II IFN-suppressive effects on normal and malignant hematopoiesis. Together, our findings establish a central role for mTORC2 in IFNγ signaling and type II IFN responses.


Carrier Proteins/metabolism , Eukaryotic Initiation Factor-4F/metabolism , Interferon-gamma/metabolism , Multiprotein Complexes/metabolism , Receptors, Interferon/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Chemokine CXCL10/metabolism , Fibroblasts/metabolism , Gene Expression Regulation , Hematopoiesis , Hematopoietic Stem Cells/cytology , Humans , Interferon-Stimulated Gene Factor 3, gamma Subunit/metabolism , Mechanistic Target of Rapamycin Complex 1 , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Knockout , Phosphorylation , Polyribosomes/metabolism , Protein Biosynthesis , Rapamycin-Insensitive Companion of mTOR Protein , U937 Cells
...