Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 124
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 84(4): 776-790.e5, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38211588

ABSTRACT

TANK-binding kinase 1 (TBK1) is a potential therapeutic target in multiple cancers, including clear cell renal cell carcinoma (ccRCC). However, targeting TBK1 in clinical practice is challenging. One approach to overcome this challenge would be to identify an upstream TBK1 regulator that could be targeted therapeutically in cancer specifically. In this study, we perform a kinome-wide small interfering RNA (siRNA) screen and identify doublecortin-like kinase 2 (DCLK2) as a TBK1 regulator in ccRCC. DCLK2 binds to and directly phosphorylates TBK1 on Ser172. Depletion of DCLK2 inhibits anchorage-independent colony growth and kidney tumorigenesis in orthotopic xenograft models. Conversely, overexpression of DCLK2203, a short isoform that predominates in ccRCC, promotes ccRCC cell growth and tumorigenesis in vivo. Mechanistically, DCLK2203 elicits its oncogenic signaling via TBK1 phosphorylation and activation. Taken together, these results suggest that DCLK2 is a TBK1 activator and potential therapeutic target for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinogenesis/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Doublecortin-Like Kinases , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
2.
Mol Cell ; 77(6): 1294-1306.e5, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32023483

ABSTRACT

von Hippel-Lindau (VHL) is a critical tumor suppressor in clear cell renal cell carcinomas (ccRCCs). It is important to identify additional therapeutic targets in ccRCC downstream of VHL loss besides hypoxia-inducible factor 2α (HIF2α). By performing a genome-wide screen, we identified Scm-like with four malignant brain tumor domains 1 (SFMBT1) as a candidate pVHL target. SFMBT1 was considered to be a transcriptional repressor but its role in cancer remains unclear. ccRCC patients with VHL loss-of-function mutations displayed elevated SFMBT1 protein levels. SFMBT1 hydroxylation on Proline residue 651 by EglN1 mediated its ubiquitination and degradation governed by pVHL. Depletion of SFMBT1 abolished ccRCC cell proliferation in vitro and inhibited orthotopic tumor growth in vivo. Integrated analyses of ChIP-seq, RNA-seq, and patient prognosis identified sphingosine kinase 1 (SPHK1) as a key SFMBT1 target gene contributing to its oncogenic phenotype. Therefore, the pVHL-SFMBT1-SPHK1 axis serves as a potential therapeutic avenue for ccRCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/pathology , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Repressor Proteins/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Animals , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/metabolism , Cell Cycle , Cell Movement , Cell Proliferation , Genome-Wide Association Study , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Phosphotransferases (Alcohol Group Acceptor)/genetics , Prognosis , Prolyl Hydroxylases/genetics , Prolyl Hydroxylases/metabolism , Repressor Proteins/genetics , Tumor Cells, Cultured , Ubiquitination , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Xenograft Model Antitumor Assays
3.
Proc Natl Acad Sci U S A ; 120(32): e2216532120, 2023 08 08.
Article in English | MEDLINE | ID: mdl-37523561

ABSTRACT

We analyzed transcriptional data from 104 HPV+ (Human papillomavirus) HNSCC (head and neck squamous cell carcinoma) tumors together with two publicly available sources to identify highly robust transcriptional programs (modules) which could be detected consistently despite heterogeneous sequencing and quantification methodologies. Among 22 modules identified, we found a single module that naturally subclassifies HPV+ HNSCC tumors based on a bimodal pattern of gene expression, clusters all atypical features of HPV+ HNSCC biology into a single subclass, and predicts patient outcome in four independent cohorts. The subclass-defining gene set was strongly correlated with Nuclear factor kappa B (NF-κB) target expression. Tumors with high expression of this NF-κB module were rarely associated with activating PIK3CA alterations or viral integration, and also expressed higher levels of HPHPV E2 and had decreased APOBEC mutagenesis. Alternatively, they harbored inactivating alterations of key regulators of NF-κB, TNF receptor associated factor 3 (TRAF3), and cylindromatosis (CYLD), as well as retinoblastoma protein (RB1). HPV+ HNSCC cells in culture with experimental depletion of TRAF3 or CYLD displayed increased expression of the subclass-defining genes, as well as robust radio-sensitization, thus recapitulating both the tumor transcriptional state and improved treatment response observed in patient data. Across all gene sets investigated, methylation to expression correlations were the strongest for the subclass-defining, NF-κB-related genes. Increased tumor-infiltrating CD4+ T cells and increased Estrogen receptors alpha (ERα) expression were identified in NF-κB active tumors. Based on the relatively high rates of cure in HPV+ HNSCC, deintensification of therapy to reduce treatment-related morbidity is being studied at many institutions. Tumor subclassification based on oncogenic subtypes may help guide the selection of therapeutic intensity or modality for patients with HPV+ HNSCC.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Papillomavirus Infections , Humans , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/radiotherapy , NF-kappa B/genetics , NF-kappa B/metabolism , TNF Receptor-Associated Factor 3/genetics , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/metabolism , Papillomavirus Infections/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/radiotherapy , Human Papillomavirus Viruses , Carcinogenesis , Papillomaviridae/genetics , Papillomaviridae/metabolism
4.
Proc Natl Acad Sci U S A ; 119(36): e2119854119, 2022 09 06.
Article in English | MEDLINE | ID: mdl-36037364

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is characterized by the loss of tumor suppressor Von Hippel Lindau (VHL) function. VHL is the component of an E3 ligase complex that promotes the ubiquitination and degradation of hypoxia inducible factor α (HIF-α) (including HIF1α and HIF2α) and Zinc Fingers And Homeoboxes 2 (ZHX2). Our recent research showed that ZHX2 contributed to ccRCC tumorigenesis in a HIF-independent manner. However, it is still unknown whether ZHX2 could be modified through deubiquitination even in the absence of pVHL. Here, we performed a deubiquitinase (DUB) complementary DNA (cDNA) library binding screen and identified USP13 as a DUB that bound ZHX2 and promoted ZHX2 deubiquitination. As a result, USP13 promoted ZHX2 protein stability in an enzymatically dependent manner, and depletion of USP13 led to ZHX2 down-regulation in ccRCC. Functionally, USP13 depletion led to decreased cell proliferation measured by two-dimensional (2D) colony formation and three-dimensional (3D) anchorage-independent growth. Furthermore, USP13 was essential for ccRCC tumor growth in vivo, and the effect was partially mediated by its regulation on ZHX2. Our findings support that USP13 may be a key effector in ccRCC tumorigenesis.


Subject(s)
Carcinoma, Renal Cell , Homeodomain Proteins , Kidney Neoplasms , Transcription Factors , Ubiquitin-Specific Proteases , Carcinogenesis/genetics , Carcinoma, Renal Cell/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Kidney Neoplasms/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Ubiquitin-Specific Proteases/genetics , Ubiquitin-Specific Proteases/metabolism , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Von Hippel-Lindau Tumor Suppressor Protein/metabolism
5.
J Am Chem Soc ; 145(3): 1512-1517, 2023 01 25.
Article in English | MEDLINE | ID: mdl-36630539

ABSTRACT

mRNA display is revolutionizing peptide drug discovery through its ability to quickly identify potent peptide binders of therapeutic protein targets. Methods to expand the chemical diversity of display libraries are continually needed to increase the likelihood of identifying clinically relevant peptide ligands. Orthogonal aminoacyl-tRNA synthetases (ORSs) have proven utility in cellular genetic code expansion, but are relatively underexplored for in vitro translation (IVT) and mRNA display. Herein, we demonstrate that the promiscuous ORS p-CNF-RS can incorporate noncanonical amino acids at amber codons in IVT, including the novel substrate p-cyanopyridylalanine (p-CNpyrA), to enable a pyridine-thiazoline (pyr-thn) macrocyclization in mRNA display. Pyr-thn-based selections against the deubiquitinase USP15 yielded a potent macrocyclic binder that exhibits good selectivity for USP15 and close homologues over other ubiquitin-specific proteases (USPs). Overall, this work exemplifies how promiscuous ORSs can both expand side chain diversity and provide structural novelty in mRNA display libraries through a heterocycle forming macrocyclization.


Subject(s)
Amino Acyl-tRNA Synthetases , RNA, Messenger/genetics , RNA, Messenger/metabolism , Amino Acyl-tRNA Synthetases/metabolism , Genetic Code , Amino Acids/chemistry , Peptides/genetics , RNA, Transfer/metabolism
6.
J Virol ; 96(17): e0082622, 2022 09 14.
Article in English | MEDLINE | ID: mdl-35938868

ABSTRACT

Viruses evolve mechanisms to exploit cellular pathways that increase viral fitness, e.g., enhance viral replication or evade the host cell immune response. The ubiquitin-proteosome system, a fundamental pathway-regulating protein fate in eukaryotes, is hijacked by all seven classes of viruses. Members of the Cullin-RING family of ubiquitin (Ub) ligases are frequently co-opted by divergent viruses because they can target a broad array of substrates by forming multisubunit assemblies comprised of a variety of adapters and substrate receptors. For example, the linker subunit DDB1 in the cullin 4-RING (CRL4)-DDB1 Ub ligase (CRL4DDB1) interacts with an H-box motif found in several unrelated viral proteins, including the V protein of simian virus 5 (SV5-V), the HBx protein of hepatitis B virus (HBV), and the recently identified pUL145 protein of human cytomegalovirus (HCMV). In HCMV-infected cells, pUL145 repurposes CRL4DDB1 to target STAT2, a protein vital to the antiviral immune response. However, the details of how these divergent viral sequences hijack DDB1 is not well understood. Here, we use a combination of binding assays, X-ray crystallography, alanine scanning, cell-based assays, and computational analysis to reveal that viral H-box motifs appear to bind to DDB1 with a higher affinity than the H-box motifs from host proteins DCAF1 and DDB2. This analysis reveals that viruses maintain native hot-spot residues in the H-box motif of host DCAFs and also acquire favorable interactions at neighboring residues within the H-box. Overall, these studies reveal how viruses evolve strategies to produce high-affinity binding and quality interactions with DDB1 to repurpose its Ub ligase machinery. IMPORTANCE Many different viruses modulate the protein machinery required for ubiquitination to enhance viral fitness. Specifically, several viruses hijack the cullin-RING ligase CRL4DDB1 to degrade host resistance factors. Human cytomegalovirus (HCMV) encodes pUL145 that redirects CRL4DDB1 to evade the immune system through the targeted degradation of the antiviral immune response protein STAT2. However, it is unclear why several viruses bind specific surfaces on ubiquitin ligases to repurpose their activity. We demonstrate that viruses have optimized H-box motifs that bind DDB1 with higher affinity than the H-box of native binders. For viral H-boxes, native interactions are maintained, but additional interactions that are absent in host cell H-boxes are formed, indicating that rewiring CRL4DDB1 creates a selective advantage for the virus. The DDB1-pUL145 peptide structure reveals that water-mediated interactions are critical to the higher affinity. Together, our data present an interesting example of how viral evolution can exploit a weakness in the ubiquitination machinery.


Subject(s)
Cullin Proteins , Cytomegalovirus Infections , DNA-Binding Proteins , Viral Proteins , Cullin Proteins/metabolism , Cytomegalovirus Infections/immunology , DNA-Binding Proteins/metabolism , Humans , Protein Binding , Protein Conformation , STAT2 Transcription Factor/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Viral Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 117(23): 13023-13032, 2020 06 09.
Article in English | MEDLINE | ID: mdl-32461361

ABSTRACT

Clear cell renal cell carcinoma (ccRCC) is characterized by loss of tumor suppressor Von Hippel Lindau (VHL) function, which leads to accumulation of hypoxia inducible factor α (including HIF1α and HIF2α). HIF2α was previously reported to be one of the major oncogenic drivers in ccRCC, however, its therapeutic targets remain challenging. Here we performed a deubiquitinase (DUB) complementary DNA (cDNA) library binding screen and discovered that ubiquitin-specific peptidase 37 (USP37) is a DUB that binds HIF2α and promotes HIF2α deubiquitination. As a result, USP37 promotes HIF2α protein stability in an enzymatically dependent manner, and depletion of USP37 leads to HIF2α down-regulation in ccRCC. Functionally, USP37 depletion causes decreased cell proliferation measured by MTS, two-dimensional (2D) colony formation as well as three-dimensional (3D) anchorage- independent growth. USP37 is also essential for maintaining kidney tumorigenesis in an orthotopic xenograft model and its depletion leads to both decreased primary kidney tumorigenesis and spontaneous lung metastasis. Our results suggest that USP37 is a potential therapeutic target in ccRCC.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Carcinoma, Renal Cell/pathology , Endopeptidases/metabolism , Kidney Neoplasms/pathology , Animals , Carcinogenesis , Carcinoma, Renal Cell/genetics , Cell Line, Tumor , Chromatin Immunoprecipitation Sequencing , Down-Regulation , Endopeptidases/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Kidney Neoplasms/genetics , Mice , Protein Stability , RNA, Small Interfering/metabolism , RNA-Seq , Ubiquitination , Xenograft Model Antitumor Assays
8.
Mol Cell ; 45(6): 719-30, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22342344

ABSTRACT

The IκB kinase (IKK) pathway is an essential mediator of inflammatory, oncogenic, and cell stress pathways. Recently IKK was shown to be essential for autophagy induction in mammalian cells independent of its ability to regulate NF-κB, but the mechanism by which this occurs is unclear. Here we demonstrate that the p85 regulatory subunit of PI3K is an IKK substrate, phosphorylated at S690 in vitro and in vivo in response to cellular starvation. Cells expressing p85 S690A or inhibited for IKK activity exhibit increased Akt activity following cell starvation, demonstrating that p85 phosphorylation is required for starvation-induced PI3K feedback inhibition. S690 is in a conserved region of the p85 cSH2 domain, and IKK-mediated phosphorylation of this site results in decreased affinity for tyrosine-phosphorylated proteins and decreased PI3K membrane localization. Finally, leucine deprivation is shown to be necessary and sufficient for starvation-induced, IKK-mediated p85 phosphorylation and PI3K feedback inhibition.


Subject(s)
I-kappa B Kinase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Starvation/metabolism , Amino Acid Sequence , Amino Acids/metabolism , Animals , Cell Line , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Conserved Sequence , Feedback, Physiological , Fibroblasts/metabolism , Humans , I-kappa B Kinase/genetics , Leucine/metabolism , Mice , Mice, Inbred C57BL , Phosphorylation , Phosphotyrosine/metabolism , src Homology Domains
9.
J Biol Chem ; 293(41): 15977-15990, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30131338

ABSTRACT

Thioridazine is an antipsychotic that has been shown to induce cell death and inhibit self-renewal in a broad spectrum of cancer cells. The mechanisms by which these effects are mediated are currently unknown but are presumed to result from the inhibition of dopamine receptor 2 (DRD2). Here we show that the self-renewal of several, but not all, triple-negative breast cancer cell lines is inhibited by thioridazine. The inhibition of self-renewal by thioridazine in these cells is mediated by DRD2 inhibition. Further, we demonstrate that DRD2 promotes self-renewal in these cells via a STAT3- and IL-6-dependent mechanism. We also show that thioridazine induces a G1 arrest and a loss in cell viability in all tested cell lines. However, the reduction in proliferation and cell viability is independent of DRD2 and STAT3. Our results indicate that although there are cell types in which DRD2 inhibition results in inhibition of STAT3 and self-renewal, the dramatic block in cancer cell proliferation across many cell lines caused by thioridazine treatment is independent of DRD2 inhibition.


Subject(s)
Breast Neoplasms/drug therapy , Cell Self Renewal/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Receptors, Dopamine D2/metabolism , STAT3 Transcription Factor/antagonists & inhibitors , Thioridazine/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chlorpromazine/pharmacology , Dopamine D2 Receptor Antagonists/pharmacology , Humans , STAT3 Transcription Factor/metabolism
10.
Immunity ; 33(1): 35-47, 2010 Jul 23.
Article in English | MEDLINE | ID: mdl-20643337

ABSTRACT

Distal cis-regulatory elements play essential roles in the T lineage-specific expression of cytokine genes. We have mapped interactions of three trans-acting factors-NF-kappaB, STAT4, and T-bet-with cis elements in the Ifng locus. We find that RelA is critical for optimal Ifng expression and is differentially recruited to multiple elements contingent upon T cell receptor (TCR) or interleukin-12 (IL-12) plus IL-18 signaling. RelA recruitment to at least four elements is dependent on T-bet-dependent remodeling of the Ifng locus and corecruitment of STAT4. STAT4 and NF-kappaB therefore cooperate at multiple cis elements to enable NF-kappaB-dependent enhancement of Ifng expression. RelA recruitment to distal elements was similar in T helper 1 (Th1) and effector CD8(+) T (Tc1) cells, although T-bet was dispensable in CD8 effectors. These results support a model of Ifng regulation in which distal cis-regulatory elements differentially recruit key transcription factors in a modular fashion to initiate gene transcription induced by distinct activation signals.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , STAT4 Transcription Factor/metabolism , T-Box Domain Proteins/metabolism , Th1 Cells/metabolism , Transcription Factor RelA/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Differentiation , Cells, Cultured , Chromatin Assembly and Disassembly/genetics , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-12/immunology , Interleukin-12/metabolism , Interleukin-18/immunology , Interleukin-18/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , NF-kappa B/genetics , NF-kappa B/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Regulatory Elements, Transcriptional/genetics , STAT4 Transcription Factor/genetics , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Th1 Cells/immunology , Th1 Cells/pathology , Transcription Factor RelA/genetics , Transcription Factor RelA/immunology , Transcriptional Activation
11.
J Biol Chem ; 292(13): 5405-5417, 2017 03 31.
Article in English | MEDLINE | ID: mdl-28188292

ABSTRACT

Cytosolic DNA can elicit beneficial as well as undesirable immune responses. For example, viral or microbial DNA triggers cell-intrinsic immune responses to defend against infections, whereas aberrant cytosolic accumulation of self-DNA results in pathological conditions, such as autoimmunity. Given the importance of these DNA-provoked responses, a better understanding of their molecular mechanisms is needed. Cytosolic DNA engages stimulator of interferon genes (STING) to activate TANK-binding kinase 1 (TBK1), which subsequently phosphorylates the transcription factor interferon regulatory factor 3 (IRF3) to promote interferon expression. Recent studies have reported that additional transcription factors, including nuclear factor κB (NF-κB) and signal transducer and activator of transcription 6 (STAT6), are also activated by cytosolic DNA, suggesting that cytosolic DNA-induced gene expression is orchestrated by multiple factors. Here we show that cytosolic DNA activates STAT3, another member of the STAT family, via an autocrine mechanism involving interferon ß (IFNß) and IL-6. Additionally, we observed a novel cytosolic DNA-induced phosphorylation at serine 754 in the transactivation domain of STAT3. Upon cytosolic DNA stimulation, Ser754 is directly phosphorylated by TBK1 in a STING-dependent manner. Moreover, Ser754 phosphorylation inhibits cytosolic DNA-induced STAT3 transcriptional activity and selectively reduces STAT3 target genes that are up-regulated in response to cytosolic DNA. Taken together, our results suggest that cytosolic DNA-induced STAT3 activation via IFNß and IL-6 is restrained by Ser754 phosphorylation of STAT3. Our findings reveal a new signaling axis downstream of the cytosolic DNA pathway and suggest potential interactions between innate immune responses and STAT3-driven oncogenic pathways.


Subject(s)
DNA/immunology , Protein Serine-Threonine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Autocrine Communication , Cell Line , DNA/physiology , Gene Expression Regulation/immunology , Humans , Immunity, Innate , Interferon-beta , Interleukin-6 , Membrane Proteins/metabolism , Phosphorylation
12.
Biol Blood Marrow Transplant ; 23(4): 569-580, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28161607

ABSTRACT

Idiopathic pneumonia syndrome (IPS) is a noninfectious inflammatory disorder of the lungs that occurs most often after fully myeloablative allogeneic hematopoietic stem cell transplantation (HSCT). IPS can be severe and is associated with high 1-year mortality rates despite existing therapies. The canonical nuclear factor-(NF) κB signaling pathway has previously been linked to several inflammatory disorders of the lung, including asthma and lung allograft rejection. It has never been specifically targeted as a novel IPS treatment approach, however. Here, we report that the IκB kinase 2 (IKK2) antagonist BAY 65-5811 or "compound A," a highly potent and specific inhibitor of the NF-κB pathway, was able to improve median survival times and recipient oxygenation in a well-described mouse model of IPS. Compound A impaired the production of the proinflammatory chemokines CCL2 and CCL5 within the host lung after transplantation. This resulted in significantly lower numbers of donor lung infiltrating CD4+ and CD8+ T cells and reduced pulmonary inflammatory cytokine production after allograft. Compound A's beneficial effects appeared to be specific for limiting pulmonary injury, as the drug was unable to improve outcomes in a B6 into B6D2 haplotype-matched murine HSCT model in which recipient mice succumb to lethal acute graft-versus-host disease of the gastrointestinal tract. Collectively, our data suggest that the targeting of the canonical NF-κB pathway with a small molecule IKK2 antagonist may represent an effective and novel therapy for the specific management of acute lung injury that can occur after allogeneic HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation/adverse effects , I-kappa B Kinase/antagonists & inhibitors , Lung Injury/drug therapy , Molecular Targeted Therapy/methods , NF-kappa B/metabolism , Pneumonia/drug therapy , Animals , Lung Injury/etiology , Mice , Treatment Outcome
13.
Immunol Rev ; 246(1): 327-45, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22435564

ABSTRACT

The cellular response to survive or to undergo death is fundamental to the benefit of the organism, and errors in this process can lead to autoimmunity and cancer. The transcription factor nuclear factor κB (NF-κB) functions to block cell death through transcriptional induction of genes encoding anti-apoptotic and antioxidant proteins. This is essential for survival of activated cells of the immune system and for cells undergoing a DNA damage response. In Ras-transformed cells and tumors as well as other cancers, NF-κB functions to suppress apoptosis--a hallmark of cancer. Critical prosurvival roles for inhibitor of NF-κB kinase (IKK) family members, including IKKε and TBK1, have been reported, which are both NF-κB-dependent and -independent. While the roles of NF-κB in promoting cell survival in lymphocytes and in cancers is relatively clear, evidence has been presented that NF-κB can promote cell death in particular contexts. Recently, IKK was shown to play a critical role in the induction of autophagy, a metabolic response typically associated with cell survival but which can lead to cell death. This review provides an historical perspective, along with new findings, regarding the roles of the IKK and NF-κB pathways in regulating cell survival.


Subject(s)
I-kappa B Kinase/metabolism , Immunity/physiology , NF-kappa B/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Animals , B-Lymphocytes/metabolism , Cell Death , Cell Survival , DNA Damage , Drug Resistance, Neoplasm/genetics , Humans , I-kappa B Kinase/genetics , NF-kappa B/genetics , Neoplasms/genetics , Signal Transduction , T-Lymphocytes/metabolism , Tumor Suppressor Protein p53/metabolism
14.
J Biol Chem ; 289(36): 25227-40, 2014 Sep 05.
Article in English | MEDLINE | ID: mdl-24990947

ABSTRACT

The serine/threonine protein kinase Akt promotes cell survival, growth, and proliferation through phosphorylation of different downstream substrates. A key effector of Akt is the mammalian target of rapamycin (mTOR). Akt is known to stimulate mTORC1 activity through phosphorylation of tuberous sclerosis complex 2 (TSC2) and PRAS40, both negative regulators of mTOR activity. We previously reported that IκB kinase α (IKKα), a component of the kinase complex that leads to NF-κB activation, plays an important role in promoting mTORC1 activity downstream of activated Akt. Here, we demonstrate IKKα-dependent regulation of mTORC1 using multiple PTEN null cancer cell lines and an animal model with deletion of IKKα. Importantly, IKKα is shown to phosphorylate mTOR at serine 1415 in a manner dependent on Akt to promote mTORC1 activity. These results demonstrate that IKKα is an effector of Akt in promoting mTORC1 activity.


Subject(s)
I-kappa B Kinase/metabolism , Multiprotein Complexes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Enzyme Activation , Female , HEK293 Cells , HeLa Cells , Humans , I-kappa B Kinase/genetics , Immunoblotting , Male , Mechanistic Target of Rapamycin Complex 1 , Mice, Knockout , Multiprotein Complexes/genetics , Mutation , Phosphorylation , Protein Binding , Proto-Oncogene Proteins c-akt/genetics , RNA Interference , Regulatory-Associated Protein of mTOR , Serine/genetics , Serine/metabolism , TOR Serine-Threonine Kinases/genetics , Tuberous Sclerosis Complex 2 Protein , Tumor Suppressor Proteins/metabolism
15.
Eur J Immunol ; 44(6): 1662-71, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24752751

ABSTRACT

Acute graft-versus-host disease (aGvHD) is a major limitation to the use of allogeneic stem cell transplantation for the treatment of patients with relapsed malignant disease. Previous work using animals lacking secondary lymphoid tissue (SLT) suggested that activation of donor T cells in SLT is critically important for the pathogenesis of aGvHD. However, these studies did not determine if impaired migration into, and more importantly, out of SLT, would ameliorate aGvHD. Here, we show that T cells from mice lacking Coronin 1A (Coro 1A(-/-)), an actin-associated protein shown to be important for thymocyte egress, do not mediate acute GvHD. The attenuation of aGvHD was associated with decreased expression of the critical trafficking proteins C-C chemokines receptor type 7 (CCR7) and sphingosine 1 phosphate receptor on donor T cells. This was mediated in part by impaired activation of the canonical NF-κB pathway in the absence of Coro 1A. As a result of these alterations, donor T cells from Coro 1A(-/-) mice were not able to initially traffic to SLT or exit SLT after BM transplantation. However, this alteration did not abrogate the graft-versus-leukemia response. Our data suggest that blocking T-cell migration into and out of SLT is a valid approach to prevent aGvHD.


Subject(s)
Bone Marrow Transplantation , Cell Movement/immunology , Graft vs Host Disease/immunology , Microfilament Proteins/immunology , T-Lymphocytes/immunology , Acute Disease , Allografts , Animals , Cell Movement/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Mice , Mice, Inbred BALB C , Mice, Knockout , Microfilament Proteins/isolation & purification , NF-kappa B/genetics , NF-kappa B/immunology , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/immunology , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/pathology
16.
Blood ; 121(25): 5015-24, 2013 Jun 20.
Article in English | MEDLINE | ID: mdl-23670180

ABSTRACT

Hematopoiesis is a tightly regulated process resulting in the production of blood cells. Self-renewal and differentiation of hematopoietic stem cells (HSCs) are key processes in hematopoietic development. Disruption of these steps can lead to altered cell distribution and disease. To investigate the role of the nuclear factor-κB subunit RelA/p65 in the regulation of HSCs in vivo, we generated mice lacking RelA/p65 in the hematopoietic compartment. Using this model system, we show that loss of p65 severely impairs HSC function and occurs in conjunction with increased hematopoietic stem and progenitor cell cycling, extramedullary hematopoiesis, and differentiation defects. Gene array studies of phenotypic HSCs indicate the up-regulation of genes normally expressed in lineage restricted cells, as well as the down-regulation of genes involved in HSC maintenance and homeostasis. We hypothesize that changes in gene expression in p65-deficient cells lead to decreased self-renewal and differentiation efficiency of hematopoietic stem and progenitor cells. These studies demonstrate that p65 is an important regulator of hematopoiesis through the transcription of genes involved in HSC fate.


Subject(s)
Gene Expression Regulation/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Transcription Factor RelA/metabolism , Animals , Cell Differentiation/physiology , Female , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Transcription Factor RelA/genetics
17.
Cancer Cell ; 12(3): 239-51, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17785205

ABSTRACT

LZAP has been reported to inhibit cellular proliferation and clonogenic growth. Here, we report that decreased LZAP expression promoted cellular transformation, xenograft tumor growth, and xenograft tumor vascularity. Loss of LZAP also increased cellular invasion, and MMP-9 expression dependent on NF-kappaB. LZAP directly bound to RelA, impaired serine 536 phosphorylation of RelA, increased HDAC association with RelA, inhibited basal and stimulated NF-kappaB transcriptional activity, and was found at the promoter of selective NF-kappaB-responsive genes. LZAP protein levels were markedly decreased in 32% of primary HNSCCs (n = 28) and decreased LZAP levels in primary HNSCC correlated with increased expression of the NF-kappaB-regulated genes IL-8 and IkappaBalpha. In aggregate, these data support a role of LZAP in NF-kappaB regulation and tumor suppression.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , NF-kappa B/antagonists & inhibitors , Nerve Tissue Proteins/physiology , Tumor Suppressor Proteins/physiology , Animals , Apoptosis , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Cycle Proteins , Cell Transformation, Neoplastic/metabolism , Gene Expression Regulation , HeLa Cells , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/pathology , Histone Deacetylases/metabolism , Humans , I-kappa B Proteins/metabolism , Interleukin-8/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , Neoplasm Invasiveness , Transcription Factor RelA/metabolism , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/physiology
18.
Blood ; 120(12): 2529-36, 2012 Sep 20.
Article in English | MEDLINE | ID: mdl-22896003

ABSTRACT

Recent data reveal an important role for B cells in the pathogenesis of chronic GVHD (cGVHD). Patients with cGVHD have delayed B-cell reconstitution and elevated BAFF to B-cell ratios compared to patients without cGVHD. The mechanisms promoting and sustaining B-cell activation in this disease, however, remain unknown. As BAFF increases murine B-cell metabolism and survival and maintains autoreactive B-cell clones, we performed ex vivo analyses of peripheral B cells from 51 patients who either had or did not have active cGVHD and were greater than 1 year from the time of allogeneic hematopoietic stem cell transplantation. We found that B cells from patients with active cGVHD were in a heightened metabolic state and were resistant to apoptosis. Exogenous BAFF treatment amplified cell size and survival in B cells from these patients. We found significantly increased signaling through ERK and AKT that associated with decreased levels of proapoptotic Bim, suggesting a mechanistic link between elevated BAFF levels and aberrant B-cell survival. Thus, we identify a role for BAFF in the pathogenesis of cGVHD and define B-cell activation and survival pathways suitable for novel therapeutic development in cGVHD.


Subject(s)
Apoptosis , B-Cell Activating Factor/metabolism , B-Lymphocytes/immunology , Graft vs Host Disease/etiology , Hematologic Neoplasms/complications , Hematopoietic Stem Cell Transplantation/adverse effects , Signal Transduction , Adult , Aged , B-Cell Activating Factor/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Size , Cells, Cultured , Chronic Disease , Female , Flow Cytometry , Follow-Up Studies , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Hematologic Neoplasms/pathology , Hematologic Neoplasms/therapy , Humans , Immunoblotting , Lymphocyte Activation , Male , Middle Aged , Transplantation, Homologous , Young Adult
19.
J Immunol ; 189(9): 4417-25, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22993204

ABSTRACT

Dihydroartemisinin (DHA) is an important derivative of the herb medicine Artemisia annua L., used in ancient China. DHA is currently used worldwide to treat malaria by killing malaria-causing parasites. In addition to this prominent effect, DHA is thought to regulate cellular functions, such as angiogenesis, tumor cell growth, and immunity. Nonetheless, how DHA affects T cell function remains poorly understood. We found that DHA potently suppressed Th cell differentiation in vitro. Unexpectedly, however, DHA greatly promoted regulatory T cell (Treg) generation in a manner dependent on the TGF-ßR:Smad signal. In addition, DHA treatment effectively reduced onset of experimental autoimmune encephalomyelitis (EAE) and ameliorated ongoing EAE in mice. Administration of DHA significantly decreased Th but increased Tregs in EAE-inflicted mice, without apparent global immune suppression. Moreover, DHA modulated the mammalian target of rapamycin (mTOR) pathway, because mTOR signal was attenuated in T cells upon DHA treatment. Importantly, enhanced Akt activity neutralized DHA-mediated effects on T cells in an mTOR-dependent fashion. This study therefore reveals a novel immune regulatory function of DHA in reciprocally regulating Th and Treg cell generation through the modulating mTOR pathway. It addresses how DHA regulates immune function and suggests a new type of drug for treating diseases in which mTOR activity is to be tempered.


Subject(s)
Artemisinins/pharmacology , Inflammation/prevention & control , Signal Transduction/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Signal Transduction/drug effects , T-Lymphocytes, Regulatory/pathology , TOR Serine-Threonine Kinases/physiology
20.
Nat Commun ; 15(1): 3805, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714664

ABSTRACT

Genomic alterations that activate Fibroblast Growth Factor Receptor 2 (FGFR2) are common in intrahepatic cholangiocarcinoma (ICC) and confer sensitivity to FGFR inhibition. However, the depth and duration of response is often limited. Here, we conduct integrative transcriptomics, metabolomics, and phosphoproteomics analysis of patient-derived models to define pathways downstream of oncogenic FGFR2 signaling that fuel ICC growth and to uncover compensatory mechanisms associated with pathway inhibition. We find that FGFR2-mediated activation of Nuclear factor-κB (NF-κB) maintains a highly glycolytic phenotype. Conversely, FGFR inhibition blocks glucose uptake and glycolysis while inciting adaptive changes, including switching fuel source utilization favoring fatty acid oxidation and increasing mitochondrial fusion and autophagy. Accordingly, FGFR inhibitor efficacy is potentiated by combined mitochondrial targeting, an effect enhanced in xenograft models by intermittent fasting. Thus, we show that oncogenic FGFR2 signaling drives NF-κB-dependent glycolysis in ICC and that metabolic reprogramming in response to FGFR inhibition confers new targetable vulnerabilities.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Glucose , Glycolysis , NF-kappa B , Receptor, Fibroblast Growth Factor, Type 2 , Signal Transduction , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Humans , NF-kappa B/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Receptor, Fibroblast Growth Factor, Type 2/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 2/genetics , Animals , Glycolysis/drug effects , Glucose/metabolism , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/drug therapy , Mice , Cell Line, Tumor , Signal Transduction/drug effects , Xenograft Model Antitumor Assays , Mitochondria/metabolism , Mitochondria/drug effects , Pyrimidines/pharmacology , Autophagy/drug effects , Gene Expression Regulation, Neoplastic/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL