Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38.947
Filter
Add more filters

Publication year range
1.
Cell ; 187(14): 3726-3740.e43, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38861993

ABSTRACT

Many growth factors and cytokines signal by binding to the extracellular domains of their receptors and driving association and transphosphorylation of the receptor intracellular tyrosine kinase domains, initiating downstream signaling cascades. To enable systematic exploration of how receptor valency and geometry affect signaling outcomes, we designed cyclic homo-oligomers with up to 8 subunits using repeat protein building blocks that can be modularly extended. By incorporating a de novo-designed fibroblast growth factor receptor (FGFR)-binding module into these scaffolds, we generated a series of synthetic signaling ligands that exhibit potent valency- and geometry-dependent Ca2+ release and mitogen-activated protein kinase (MAPK) pathway activation. The high specificity of the designed agonists reveals distinct roles for two FGFR splice variants in driving arterial endothelium and perivascular cell fates during early vascular development. Our designed modular assemblies should be broadly useful for unraveling the complexities of signaling in key developmental transitions and for developing future therapeutic applications.


Subject(s)
Cell Differentiation , Fibroblast Growth Factors , Receptors, Fibroblast Growth Factor , Signal Transduction , Animals , Humans , Receptors, Fibroblast Growth Factor/metabolism , Fibroblast Growth Factors/metabolism , Mice , Ligands , Calcium/metabolism , MAP Kinase Signaling System
2.
Cell ; 186(17): 3606-3618.e16, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37480850

ABSTRACT

Injury induces systemic responses, but their functions remain elusive. Mechanisms that can rapidly synchronize wound responses through long distances are also mostly unknown. Using planarian flatworms capable of whole-body regeneration, we report that injury induces extracellular signal-regulated kinase (Erk) activity waves to travel at a speed 10-100 times faster than those in other multicellular tissues. This ultrafast propagation requires longitudinal body-wall muscles, elongated cells forming dense parallel tracks running the length of the organism. The morphological properties of muscles allow them to act as superhighways for propagating and disseminating wound signals. Inhibiting Erk propagation prevents tissues distant to the wound from responding and blocks regeneration, which can be rescued by a second injury to distal tissues shortly after the first injury. Our findings provide a mechanism for long-range signal propagation in large, complex tissues to coordinate responses across cell types and highlight the function of feedback between spatially separated tissues during whole-body regeneration.


Subject(s)
Planarians , Regeneration , Animals , MAP Kinase Signaling System , Muscles , Phosphorylation , Planarians/physiology , Protein Processing, Post-Translational
3.
Cell ; 185(4): 585-602.e29, 2022 02 17.
Article in English | MEDLINE | ID: mdl-35051368

ABSTRACT

The relevance of extracellular magnesium in cellular immunity remains largely unknown. Here, we show that the co-stimulatory cell-surface molecule LFA-1 requires magnesium to adopt its active conformation on CD8+ T cells, thereby augmenting calcium flux, signal transduction, metabolic reprogramming, immune synapse formation, and, as a consequence, specific cytotoxicity. Accordingly, magnesium-sufficiency sensed via LFA-1 translated to the superior performance of pathogen- and tumor-specific T cells, enhanced effectiveness of bi-specific T cell engaging antibodies, and improved CAR T cell function. Clinically, low serum magnesium levels were associated with more rapid disease progression and shorter overall survival in CAR T cell and immune checkpoint antibody-treated patients. LFA-1 thus directly incorporates information on the composition of the microenvironment as a determinant of outside-in signaling activity. These findings conceptually link co-stimulation and nutrient sensing and point to the magnesium-LFA-1 axis as a therapeutically amenable biologic system.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Magnesium/metabolism , Animals , Bacterial Infections/immunology , Caloric Restriction , Cell Line, Tumor , Cytotoxicity, Immunologic , HEK293 Cells , Humans , Immunologic Memory , Immunological Synapses/metabolism , Immunotherapy , Lymphocyte Activation/immunology , MAP Kinase Signaling System , Magnesium/administration & dosage , Male , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Phosphorylation , Proto-Oncogene Proteins c-jun/metabolism
4.
Cell ; 183(2): 490-502.e18, 2020 10 15.
Article in English | MEDLINE | ID: mdl-33002410

ABSTRACT

The non-receptor protein tyrosine phosphatase (PTP) SHP2, encoded by PTPN11, plays an essential role in RAS-mitogen-activated protein kinase (MAPK) signaling during normal development. It has been perplexing as to why both enzymatically activating and inactivating mutations in PTPN11 result in human developmental disorders with overlapping clinical manifestations. Here, we uncover a common liquid-liquid phase separation (LLPS) behavior shared by these disease-associated SHP2 mutants. SHP2 LLPS is mediated by the conserved well-folded PTP domain through multivalent electrostatic interactions and regulated by an intrinsic autoinhibitory mechanism through conformational changes. SHP2 allosteric inhibitors can attenuate LLPS of SHP2 mutants, which boosts SHP2 PTP activity. Moreover, disease-associated SHP2 mutants can recruit and activate wild-type (WT) SHP2 in LLPS to promote MAPK activation. These results not only suggest that LLPS serves as a gain-of-function mechanism involved in the pathogenesis of SHP2-associated human diseases but also provide evidence that PTP may be regulated by LLPS that can be therapeutically targeted.


Subject(s)
Mitogen-Activated Protein Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , A549 Cells , Animals , Child , Child, Preschool , Female , Gain of Function Mutation/genetics , HEK293 Cells , Human Umbilical Vein Endothelial Cells , Humans , MAP Kinase Signaling System/physiology , Male , Mice , Mouse Embryonic Stem Cells , Mutation/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Signal Transduction , src Homology Domains/genetics
5.
Cell ; 181(2): 424-441.e21, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32234521

ABSTRACT

KRAS mutant pancreatic ductal adenocarcinoma (PDAC) is characterized by a desmoplastic response that promotes hypovascularity, immunosuppression, and resistance to chemo- and immunotherapies. We show that a combination of MEK and CDK4/6 inhibitors that target KRAS-directed oncogenic signaling can suppress PDAC proliferation through induction of retinoblastoma (RB) protein-mediated senescence. In preclinical mouse models of PDAC, this senescence-inducing therapy produces a senescence-associated secretory phenotype (SASP) that includes pro-angiogenic factors that promote tumor vascularization, which in turn enhances drug delivery and efficacy of cytotoxic gemcitabine chemotherapy. In addition, SASP-mediated endothelial cell activation stimulates the accumulation of CD8+ T cells into otherwise immunologically "cold" tumors, sensitizing tumors to PD-1 checkpoint blockade. Therefore, in PDAC models, therapy-induced senescence can establish emergent susceptibilities to otherwise ineffective chemo- and immunotherapies through SASP-dependent effects on the tumor vasculature and immune system.


Subject(s)
Aging/physiology , Carcinoma, Pancreatic Ductal/pathology , Vascular Remodeling/physiology , Animals , CD8-Positive T-Lymphocytes/immunology , Carcinoma, Pancreatic Ductal/microbiology , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, ras/genetics , Humans , Immunotherapy/methods , MAP Kinase Signaling System/physiology , Mice , Pancreatic Neoplasms/pathology , Retinoblastoma Protein/immunology , Signal Transduction/genetics , Tumor Microenvironment , Vascular Remodeling/genetics
6.
Cell ; 173(6): 1413-1425.e14, 2018 05 31.
Article in English | MEDLINE | ID: mdl-29754815

ABSTRACT

BRAF(V600E) mutant melanomas treated with inhibitors of the BRAF and MEK kinases almost invariably develop resistance that is frequently caused by reactivation of the mitogen activated protein kinase (MAPK) pathway. To identify novel treatment options for such patients, we searched for acquired vulnerabilities of MAPK inhibitor-resistant melanomas. We find that resistance to BRAF+MEK inhibitors is associated with increased levels of reactive oxygen species (ROS). Subsequent treatment with the histone deacetylase inhibitor vorinostat suppresses SLC7A11, leading to a lethal increase in the already-elevated levels of ROS in drug-resistant cells. This causes selective apoptotic death of only the drug-resistant tumor cells. Consistently, treatment of BRAF inhibitor-resistant melanoma with vorinostat in mice results in dramatic tumor regression. In a study in patients with advanced BRAF+MEK inhibitor-resistant melanoma, we find that vorinostat can selectively ablate drug-resistant tumor cells, providing clinical proof of concept for the novel therapy identified here.


Subject(s)
Drug Resistance, Neoplasm , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Amino Acid Transport System y+/metabolism , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Humans , MAP Kinase Kinase 1/metabolism , MAP Kinase Signaling System , Melanoma/genetics , Mice , Mutation , Neoplasm Transplantation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Reactive Oxygen Species/metabolism , Skin Neoplasms/genetics , Treatment Outcome , Vorinostat/pharmacology
7.
Cell ; 175(1): 186-199.e19, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30220457

ABSTRACT

Mutations or aberrant upregulation of EZH2 occur frequently in human cancers, yet clinical benefits of EZH2 inhibitor (EZH2i) remain unsatisfactory and limited to certain hematological malignancies. We profile global posttranslational histone modification changes across a large panel of cancer cell lines with various sensitivities to EZH2i. We report here oncogenic transcriptional reprogramming mediated by MLL1's interaction with the p300/CBP complex, which directs H3K27me loss to reciprocal H3K27ac gain and restricts EZH2i response. Concurrent inhibition of H3K27me and H3K27ac results in transcriptional repression and MAPK pathway dependency in cancer subsets. In preclinical models encompassing a broad spectrum of EZH2-aberrant solid tumors, a combination of EZH2 and BRD4 inhibitors, or a triple-combination including MAPK inhibition display robust efficacy with very tolerable toxicity. Our results suggest an attractive precision treatment strategy for EZH2-aberrant tumors on the basis of tumor-intrinsic MLL1 expression and concurrent inhibition of epigenetic crosstalk and feedback MAPK activation.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/genetics , Histone-Lysine N-Methyltransferase/physiology , Myeloid-Lymphoid Leukemia Protein/physiology , Animals , Carcinogenesis/genetics , Cell Cycle Proteins , Cell Line, Tumor , Epigenesis, Genetic/genetics , Epigenomics/methods , Female , Gene Expression Regulation, Neoplastic/genetics , Histone Code/drug effects , Histone Code/genetics , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/physiology , Humans , MAP Kinase Signaling System , Male , Mice , Mice, Inbred BALB C , Mice, SCID , Mutation , Myeloid-Lymphoid Leukemia Protein/genetics , Nuclear Proteins/metabolism , Nuclear Proteins/physiology , Polycomb Repressive Complex 2/metabolism , Transcription Factors/metabolism , Transcription Factors/physiology , Transcriptional Activation , Xenograft Model Antitumor Assays/methods , p300-CBP Transcription Factors/physiology
8.
Cell ; 168(3): 427-441.e21, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111074

ABSTRACT

Human apolipoprotein E (ApoE) apolipoprotein is primarily expressed in three isoforms (ApoE2, ApoE3, and ApoE4) that differ only by two residues. ApoE4 constitutes the most important genetic risk factor for Alzheimer's disease (AD), ApoE3 is neutral, and ApoE2 is protective. How ApoE isoforms influence AD pathogenesis, however, remains unclear. Using ES-cell-derived human neurons, we show that ApoE secreted by glia stimulates neuronal Aß production with an ApoE4 > ApoE3 > ApoE2 potency rank order. We demonstrate that ApoE binding to ApoE receptors activates dual leucine-zipper kinase (DLK), a MAP-kinase kinase kinase that then activates MKK7 and ERK1/2 MAP kinases. Activated ERK1/2 induces cFos phosphorylation, stimulating the transcription factor AP-1, which in turn enhances transcription of amyloid-ß precursor protein (APP) and thereby increases amyloid-ß levels. This molecular mechanism also regulates APP transcription in mice in vivo. Our data describe a novel signal transduction pathway in neurons whereby ApoE activates a non-canonical MAP kinase cascade that enhances APP transcription and amyloid-ß synthesis.


Subject(s)
Amyloid beta-Protein Precursor/genetics , Apolipoproteins E/metabolism , MAP Kinase Signaling System , Alzheimer Disease/metabolism , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Humans , Mice , Neurons/metabolism , Protein Isoforms/metabolism
9.
Cell ; 168(5): 890-903.e15, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28162770

ABSTRACT

The genetic dependencies of human cancers widely vary. Here, we catalog this heterogeneity and use it to identify functional gene interactions and genotype-dependent liabilities in cancer. By using genome-wide CRISPR-based screens, we generate a gene essentiality dataset across 14 human acute myeloid leukemia (AML) cell lines. Sets of genes with correlated patterns of essentiality across the lines reveal new gene relationships, the essential substrates of enzymes, and the molecular functions of uncharacterized proteins. Comparisons of differentially essential genes between Ras-dependent and -independent lines uncover synthetic lethal partners of oncogenic Ras. Screens in both human AML and engineered mouse pro-B cells converge on a surprisingly small number of genes in the Ras processing and MAPK pathways and pinpoint PREX1 as an AML-specific activator of MAPK signaling. Our findings suggest general strategies for defining mammalian gene networks and synthetic lethal interactions by exploiting the natural genetic and epigenetic diversity of human cancer cells.


Subject(s)
Gene Regulatory Networks , Leukemia, Myeloid, Acute/genetics , Animals , Carrier Proteins , Cell Line, Tumor , Clustered Regularly Interspaced Short Palindromic Repeats , Epigenesis, Genetic , Genes, Essential , Humans , MAP Kinase Signaling System , Mice , Mitochondrial Proteins , Protein Processing, Post-Translational , ras Proteins/genetics
10.
Cell ; 168(5): 817-829.e15, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28215705

ABSTRACT

Investigating therapeutic "outliers" that show exceptional responses to anti-cancer treatment can uncover biomarkers of drug sensitivity. We performed preclinical trials investigating primary murine acute myeloid leukemias (AMLs) generated by retroviral insertional mutagenesis in KrasG12D "knockin" mice with the MEK inhibitor PD0325901 (PD901). One outlier AML responded and exhibited intrinsic drug resistance at relapse. Loss of wild-type (WT) Kras enhanced the fitness of the dominant clone and rendered it sensitive to MEK inhibition. Similarly, human colorectal cancer cell lines with increased KRAS mutant allele frequency were more sensitive to MAP kinase inhibition, and CRISPR-Cas9-mediated replacement of WT KRAS with a mutant allele sensitized heterozygous mutant HCT116 cells to treatment. In a prospectively characterized cohort of patients with advanced cancer, 642 of 1,168 (55%) with KRAS mutations exhibited allelic imbalance. These studies demonstrate that serial genetic changes at the Kras/KRAS locus are frequent in cancer and modulate competitive fitness and MEK dependency.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzamides/therapeutic use , Colorectal Neoplasms/genetics , Diphenylamine/analogs & derivatives , MAP Kinase Signaling System , Proto-Oncogene Proteins p21(ras)/genetics , Animals , Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Cell Line, Tumor , Clonal Evolution , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Diphenylamine/pharmacology , Diphenylamine/therapeutic use , Drug Resistance, Neoplasm , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , MAP Kinase Signaling System/drug effects , Mice , Mutation , Retroviridae
11.
Mol Cell ; 84(1): 142-155, 2024 Jan 04.
Article in English | MEDLINE | ID: mdl-38118452

ABSTRACT

Cellular homeostasis is continuously challenged by environmental cues and cellular stress conditions. In their defense, cells need to mount appropriate stress responses that, dependent on the cellular context, signaling intensity, and duration, may have diverse outcomes. The stress- and mitogen-activated protein kinase (SAPK/MAPK) system consists of well-characterized signaling cascades that sense and transduce an array of different stress stimuli into biological responses. However, the physical and chemical nature of stress signals and how these are sensed by individual upstream MAP kinase kinase kinases (MAP3Ks) remain largely ambiguous. Here, we review the existing knowledge of how individual members of the large and diverse group of MAP3Ks sense specific stress signals through largely non-redundant mechanisms. We emphasize the large knowledge gaps in assigning function and stress signals for individual MAP3K family members and touch on the potential of targeting this class of proteins for clinical benefit.


Subject(s)
JNK Mitogen-Activated Protein Kinases , MAP Kinase Kinase Kinases , Animals , MAP Kinase Kinase Kinases/genetics , MAP Kinase Kinase Kinases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Signal Transduction , Phosphorylation , p38 Mitogen-Activated Protein Kinases/metabolism , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Mammals/metabolism
12.
Nat Immunol ; 20(6): 677-686, 2019 06.
Article in English | MEDLINE | ID: mdl-31110312

ABSTRACT

Consumption of a high-energy Western diet triggers mild adaptive ß cell proliferation to compensate for peripheral insulin resistance; however, the underlying molecular mechanism remains unclear. In the present study we show that the toll-like receptors TLR2 and TLR4 inhibited the diet-induced replication of ß cells in mice and humans. The combined, but not the individual, loss of TLR2 and TLR4 increased the replication of ß cells, but not that of α cells, leading to enlarged ß cell area and hyperinsulinemia in diet-induced obesity. Loss of TLR2 and TLR4 increased the nuclear abundance of the cell cycle regulators cyclin D2 and Cdk4 in a manner dependent on the signaling mediator Erk. These data reveal a regulatory mechanism controlling the proliferation of ß cells in diet-induced obesity and suggest that selective targeting of the TLR2/TLR4 pathways may reverse ß cell failure in patients with diabetes.


Subject(s)
Insulin-Secreting Cells/metabolism , Obesity/etiology , Obesity/metabolism , Toll-Like Receptor 2/genetics , Toll-Like Receptor 4/genetics , Animals , Cell Proliferation , Cyclin D2/metabolism , Cyclin-Dependent Kinase 4/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Female , Humans , Insulin/blood , Insulin/metabolism , Insulin-Secreting Cells/ultrastructure , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , MAP Kinase Signaling System , Male , Mice , Mice, Knockout , Multiprotein Complexes/metabolism , Obesity/drug therapy , Parabiosis , Protein Binding , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
13.
Cell ; 165(7): 1658-1671, 2016 Jun 16.
Article in English | MEDLINE | ID: mdl-27212238

ABSTRACT

Macrophages are multifunctional cells that perform diverse roles in health and disease. Emerging evidence has suggested that these innate immune cells might also be capable of developing immunological memory, a trait previously associated with the adaptive system alone. While recent studies have focused on the dramatic macrophage reprogramming that follows infection and protects against secondary microbial attack, can macrophages also develop memory in response to other cues? Here, we show that apoptotic corpse engulfment by Drosophila macrophages is an essential primer for their inflammatory response to tissue damage and infection in vivo. Priming is triggered via calcium-induced JNK signaling, which leads to upregulation of the damage receptor Draper, thus providing a molecular memory that allows the cell to rapidly respond to subsequent injury or infection. This remarkable plasticity and capacity for memory places macrophages as key therapeutic targets for treatment of inflammatory disorders.


Subject(s)
Drosophila/immunology , Immunologic Memory , Macrophages/immunology , Animals , Apoptosis , Drosophila/cytology , Drosophila Proteins/metabolism , Escherichia coli , Immunity, Innate , MAP Kinase Signaling System , Membrane Proteins/metabolism , Phagocytosis
14.
Cell ; 165(1): 35-44, 2016 Mar 24.
Article in English | MEDLINE | ID: mdl-26997480

ABSTRACT

PD-1 immune checkpoint blockade provides significant clinical benefits for melanoma patients. We analyzed the somatic mutanomes and transcriptomes of pretreatment melanoma biopsies to identify factors that may influence innate sensitivity or resistance to anti-PD-1 therapy. We find that overall high mutational loads associate with improved survival, and tumors from responding patients are enriched for mutations in the DNA repair gene BRCA2. Innately resistant tumors display a transcriptional signature (referred to as the IPRES, or innate anti-PD-1 resistance), indicating concurrent up-expression of genes involved in the regulation of mesenchymal transition, cell adhesion, extracellular matrix remodeling, angiogenesis, and wound healing. Notably, mitogen-activated protein kinase (MAPK)-targeted therapy (MAPK inhibitor) induces similar signatures in melanoma, suggesting that a non-genomic form of MAPK inhibitor resistance mediates cross-resistance to anti-PD-1 therapy. Validation of the IPRES in other independent tumor cohorts defines a transcriptomic subset across distinct types of advanced cancer. These findings suggest that attenuating the biological processes that underlie IPRES may improve anti-PD-1 response in melanoma and other cancer types.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Resistance, Neoplasm , Melanoma/drug therapy , Neoplasm Metastasis/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents/adverse effects , BRCA2 Protein/genetics , Humans , MAP Kinase Signaling System/drug effects , Melanoma/genetics , Neoplasm Metastasis/genetics , Nivolumab , Transcriptome
15.
Cell ; 165(3): 643-55, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27104980

ABSTRACT

Oncogenic activation of RAS genes via point mutations occurs in 20%-30% of human cancers. The development of effective RAS inhibitors has been challenging, necessitating new approaches to inhibit this oncogenic protein. Functional studies have shown that the switch region of RAS interacts with a large number of effector proteins containing a common RAS-binding domain (RBD). Because RBD-mediated interactions are essential for RAS signaling, blocking RBD association with small molecules constitutes an attractive therapeutic approach. Here, we present evidence that rigosertib, a styryl-benzyl sulfone, acts as a RAS-mimetic and interacts with the RBDs of RAF kinases, resulting in their inability to bind to RAS, disruption of RAF activation, and inhibition of the RAS-RAF-MEK pathway. We also find that ribosertib binds to the RBDs of Ral-GDS and PI3Ks. These results suggest that targeting of RBDs across multiple signaling pathways by rigosertib may represent an effective strategy for inactivation of RAS signaling.


Subject(s)
Glycine/analogs & derivatives , RNA-Binding Proteins/chemistry , Signal Transduction/drug effects , Sulfones/pharmacology , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/drug effects , Crystallography, X-Ray , Dimerization , Glycine/administration & dosage , Glycine/chemistry , Glycine/pharmacology , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Pancreatic Neoplasms/drug therapy , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , RNA-Binding Proteins/metabolism , Sequence Alignment , Sulfones/administration & dosage , Sulfones/chemistry , ras Proteins/metabolism , Polo-Like Kinase 1
16.
Immunity ; 54(12): 2740-2755.e6, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34644536

ABSTRACT

T follicular helper (Tfh) cells play essential roles in regulating humoral immunity, especially germinal center reactions. However, how CD4+ T cells integrate the antigenic and costimulatory signals in Tfh cell development is still poorly understood. Here, we found that phorbol 12-myristate 13-acetate (PMA) + ionomycin (P+I) stimulation, together with interleukin-6 (IL-6), potently induce Tfh cell-like transcriptomic programs in vitro. The ERK kinase pathway was attenuated under P+I stimulation; ERK2 inhibition enhanced Tfh cell development in vitro and in vivo. We observed that inducible T cell costimulator (ICOS), but not CD28, lacked the ability to activate ERK, which was important in sustaining Tfh cell development. The transcription factor Zfp831, whose expression was repressed by ERK, promoted Tfh cell differentiation by directly upregulating the expression of the transcription factors Bcl6 and Tcf7. We have hence identified an ERK-Zfp831 axis, regulated by costimulation signaling, in critical regulation of Tfh cell development.


Subject(s)
DNA-Binding Proteins/metabolism , Germinal Center/immunology , Inducible T-Cell Co-Stimulator Protein/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , T Follicular Helper Cells/immunology , Animals , Cell Differentiation , Hepatocyte Nuclear Factor 1-alpha/metabolism , Immunity, Humoral , Interleukin-6/metabolism , Lymphocyte Activation , MAP Kinase Signaling System , Mice , Mice, Knockout , Transcriptome
17.
Cell ; 162(1): 72-83, 2015 Jul 02.
Article in English | MEDLINE | ID: mdl-26119340

ABSTRACT

Identifying the molecular mechanisms that underlie aging and their pharmacological manipulation are key aims for improving lifelong human health. Here, we identify a critical role for Ras-Erk-ETS signaling in aging in Drosophila. We show that inhibition of Ras is sufficient for lifespan extension downstream of reduced insulin/IGF-1 (IIS) signaling. Moreover, direct reduction of Ras or Erk activity leads to increased lifespan. We identify the E-twenty six (ETS) transcriptional repressor, Anterior open (Aop), as central to lifespan extension caused by reduced IIS or Ras attenuation. Importantly, we demonstrate that adult-onset administration of the drug trametinib, a highly specific inhibitor of Ras-Erk-ETS signaling, can extend lifespan. This discovery of the Ras-Erk-ETS pathway as a pharmacological target for animal aging, together with the high degree of evolutionary conservation of the pathway, suggests that inhibition of Ras-Erk-ETS signaling may provide an effective target for anti-aging interventions in mammals.


Subject(s)
Drosophila melanogaster/metabolism , Longevity , MAP Kinase Signaling System , Aging , Animals , Drosophila Proteins/metabolism , Eye Proteins/metabolism , Insulin Receptor Substrate Proteins/metabolism , MAP Kinase Signaling System/drug effects , Models, Animal , Protein Kinase Inhibitors/pharmacology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Repressor Proteins/metabolism
18.
Cell ; 161(5): 967-970, 2015 May 21.
Article in English | MEDLINE | ID: mdl-26000477

ABSTRACT

RAF links RAS, one of the most potent human oncogenes, to its effector ERK and to proliferation. This role is evolutionarily conserved, but while simpler multicellular organisms express one RAF, mammals have three. This Minireview highlights common and divergent features of RAF paralogs, their signaling outputs, and roles in tumorigenesis.


Subject(s)
Carcinogenesis , MAP Kinase Signaling System , Proto-Oncogene Proteins c-raf/metabolism , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Neoplasms/metabolism , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/genetics , Signal Transduction
19.
Cell ; 161(3): 595-609, 2015 Apr 23.
Article in English | MEDLINE | ID: mdl-25892225

ABSTRACT

Organisms must be able to respond to low oxygen in a number of homeostatic and pathological contexts. Regulation of hypoxic responses via the hypoxia-inducible factor (HIF) is well established, but evidence indicates that other, HIF-independent mechanisms are also involved. Here, we report a hypoxic response that depends on the accumulation of lactate, a metabolite whose production increases in hypoxic conditions. We find that the NDRG3 protein is degraded in a PHD2/VHL-dependent manner in normoxia but is protected from destruction by binding to lactate that accumulates under hypoxia. The stabilized NDRG3 protein binds c-Raf to mediate hypoxia-induced activation of Raf-ERK pathway, promoting angiogenesis and cell growth. Inhibiting cellular lactate production abolishes the NDRG3-mediated hypoxia responses. Our study, therefore, elucidates the molecular basis for lactate-induced hypoxia signaling, which can be exploited for the development of therapies targeting hypoxia-induced diseases.


Subject(s)
Hypoxia/metabolism , Lactic Acid/metabolism , Cell Hypoxia , Cell Line , Gene Expression Regulation , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/metabolism , Intracellular Signaling Peptides and Proteins , MAP Kinase Signaling System , Neovascularization, Pathologic/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Oxygen/metabolism , Protein Binding , raf Kinases/metabolism
20.
Cell ; 160(1-2): 161-76, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25594179

ABSTRACT

Axonal death disrupts functional connectivity of neural circuits and is a critical feature of many neurodegenerative disorders. Pathological axon degeneration often occurs independently of known programmed death pathways, but the underlying molecular mechanisms remain largely unknown. Using traumatic injury as a model, we systematically investigate mitogen-activated protein kinase (MAPK) families and delineate a MAPK cascade that represents the early degenerative response to axonal injury. The adaptor protein Sarm1 is required for activation of this MAPK cascade, and this Sarm1-MAPK pathway disrupts axonal energy homeostasis, leading to ATP depletion before physical breakdown of damaged axons. The protective cytoNmnat1/Wld(s) protein inhibits activation of this MAPK cascade. Further, MKK4, a key component in the Sarm1-MAPK pathway, is antagonized by AKT signaling, which modulates the degenerative response by limiting activation of downstream JNK signaling. Our results reveal a regulatory mechanism that integrates distinct signals to instruct pathological axon degeneration.


Subject(s)
Axons/pathology , MAP Kinase Signaling System , Adenosine Triphosphate/metabolism , Animals , Armadillo Domain Proteins/genetics , Armadillo Domain Proteins/metabolism , Cell Death , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , MAP Kinase Kinase 4/metabolism , Mice , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Proto-Oncogene Proteins c-akt/metabolism , Retinal Ganglion Cells/metabolism , Retinal Ganglion Cells/pathology
SELECTION OF CITATIONS
SEARCH DETAIL