Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
bioRxiv ; 2024 Mar 17.
Article in English | MEDLINE | ID: mdl-38496652

ABSTRACT

Classical G protein-coupled receptor (GPCR) signaling takes place in response to extracellular stimuli and involves receptors and heterotrimeric G proteins located at the plasma membrane. It has recently been established that GPCR signaling can also take place from intracellular membrane compartments, including endosomes that contain internalized receptors and ligands. While the mechanisms of GPCR endocytosis are well understood, it is not clear how internalized receptors are supplied with G proteins. To address this gap we use gene editing, confocal microscopy, and bioluminescence resonance energy transfer to study the distribution and trafficking of endogenous G proteins. We show here that constitutive endocytosis is sufficient to supply newly internalized endocytic vesicles with 20-30% of the G protein density found at the plasma membrane. We find that G proteins are present on early, late, and recycling endosomes, are abundant on lysosomes, but are virtually undetectable on the endoplasmic reticulum, mitochondria, and the medial Golgi apparatus. Receptor activation does not change heterotrimer abundance on endosomes. Our results provide a detailed subcellular map of endogenous G protein distribution, suggest that G proteins may be partially excluded from nascent endocytic vesicles, and are likely to have implications for GPCR signaling from endosomes and other intracellular compartments.

2.
Chem Commun (Camb) ; 59(100): 14823-14826, 2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38009240

ABSTRACT

By leveraging natural saturated fatty acids with distinct melting points and swift reversible phase transitions, we correlated external thermal cues to monomer and excimer emissions of difluoroboron ß-diketonate fluorophores. This integration yielded a ratiometric fluorescent thermometer showcasing unparalleled sensitivity and thermochromism in the physiological temperature range.

3.
iScience ; 26(12): 108308, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38025772

ABSTRACT

Low response rates and immune-related adverse events limit the remarkable impact of cancer immunotherapy. To improve clinical outcomes, preclinical studies have shown that combining immunotherapies with N-terminal Hsp90 inhibitors resulted in improved efficacy, even though induction of an extensive heat shock response (HSR) and less than optimal dosing of these inhibitors limited their clinical efficacy as monotherapies. We discovered that the natural product Enniatin A (EnnA) targets Hsp90 and destabilizes its client oncoproteins without inducing an HSR. EnnA triggers immunogenic cell death in triple-negative breast cancer (TNBC) syngeneic mouse models and exhibits superior antitumor activity compared to Hsp90 N-terminal inhibitors. EnnA reprograms the tumor microenvironment (TME) to promote CD8+ T cell-dependent antitumor immunity by reducing PD-L1 levels and activating the chemokine receptor CX3CR1 pathway. These findings provide strong evidence for transforming the immunosuppressive TME into a more tumor-hostile milieu by engaging Hsp90 with therapeutic agents involving novel mechanisms of action.

4.
Nat Chem Biol ; 19(6): 687-694, 2023 06.
Article in English | MEDLINE | ID: mdl-36646958

ABSTRACT

G protein-coupled receptors (GPCRs) selectively activate at least one of the four families of heterotrimeric G proteins, but the mechanism of coupling selectivity remains unclear. Structural studies emphasize structural complementarity of GPCRs and nucleotide-free G proteins, but selectivity is likely to be determined by transient intermediate-state complexes that exist before nucleotide release. Here we study coupling to nucleotide-decoupled G protein variants that can adopt conformations similar to receptor-bound G proteins without releasing nucleotide, and are therefore able to bypass intermediate-state complexes. We find that selectivity is degraded when nucleotide release is not required for GPCR-G protein complex formation, to the extent that most GPCRs interact with most nucleotide-decoupled G proteins. These findings demonstrate the absence of absolute structural incompatibility between noncognate receptor-G protein pairs, and are consistent with the hypothesis that transient intermediate states are partly responsible for coupling selectivity.


Subject(s)
Heterotrimeric GTP-Binding Proteins , Receptors, G-Protein-Coupled , Receptors, G-Protein-Coupled/metabolism , Protein Conformation , Heterotrimeric GTP-Binding Proteins/chemistry , Heterotrimeric GTP-Binding Proteins/metabolism
5.
Mol Psychiatry ; 28(2): 588-600, 2023 02.
Article in English | MEDLINE | ID: mdl-36357671

ABSTRACT

Posttraumatic stress disorder (PTSD) after the pandemic has emerged as a major neuropsychiatric component of post-acute COVID-19 syndrome, yet the current pharmacotherapy for PTSD is limited. The use of adrenergic drugs to treat PTSD has been suggested; however, it is hindered by conflicting clinical results and a lack of mechanistic understanding of drug actions. Our studies, using both genetically modified mice and human induced pluripotent stem cell-derived neurons, reveal a novel α2A adrenergic receptor (α2AAR)-spinophilin-cofilin axis in the hippocampus that is critical for regulation of contextual fear memory reconsolidation. In addition, we have found that two α2 ligands, clonidine and guanfacine, exhibit differential abilities in activating this signaling axis to disrupt fear memory reconsolidation. Stimulation of α2AAR with clonidine, but not guanfacine, promotes the interaction of the actin binding protein cofilin with the receptor and with the dendritic spine scaffolding protein spinophilin to induce cofilin activation at the synapse. Spinophilin-dependent regulation of cofilin is required for clonidine-induced disruption of contextual fear memory reconsolidation. Our results inform the interpretation of differential clinical observations of these two drugs on PTSD and suggest that clonidine could provide immediate treatment for PTSD symptoms related to the current pandemic. Furthermore, our study indicates that modulation of dendritic spine morphology may represent an effective strategy for the development of new pharmacotherapies for PTSD.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Animals , Humans , Mice , Actin Depolymerizing Factors/pharmacology , Adrenergic Agents/pharmacology , Clonidine/pharmacology , Fear/physiology , Induced Pluripotent Stem Cells/metabolism , Microfilament Proteins/metabolism , Receptors, Adrenergic, alpha-2/metabolism
6.
PLoS One ; 16(4): e0247743, 2021.
Article in English | MEDLINE | ID: mdl-33886554

ABSTRACT

A large number of GPCRs are potentially valuable drug targets but remain understudied. Many of these lack well-validated activating ligands and are considered "orphan" receptors, and G protein coupling profiles have not been defined for many orphan GPCRs. Here we asked if constitutive receptor activity can be used to determine G protein coupling profiles of orphan GPCRs. We monitored nucleotide-sensitive interactions between 48 understudied orphan GPCRs and five G proteins (240 combinations) using bioluminescence resonance energy transfer (BRET). No receptor ligands were used, but GDP was used as a common G protein ligand to disrupt receptor-G protein complexes. Constitutive BRET between the same receptors and ß-arrestins was also measured. We found sufficient GDP-sensitive BRET to generate G protein coupling profiles for 22 of the 48 receptors we studied. Altogether we identified 48 coupled receptor-G protein pairs, many of which have not been described previously. We conclude that receptor-G protein complexes that form spontaneously in the absence of guanine nucleotides can be used to profile G protein coupling of constitutively-active GPCRs. This approach may prove useful for studying G protein coupling of other GPCRs for which activating ligands are not available.


Subject(s)
Protein Interaction Maps , Receptors, G-Protein-Coupled/metabolism , Arrestin/metabolism , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Luminescent Measurements , Protein Interaction Mapping
7.
Cell Death Dis ; 11(10): 884, 2020 10 20.
Article in English | MEDLINE | ID: mdl-33082322

ABSTRACT

Resistance to molecular therapies frequently occur due to genetic changes affecting the targeted pathway. In myeloid and lymphoid leukemias/lymphomas resulting from constitutive activation of FGFR1 kinases, resistance has been shown to be due either to mutations in FGFR1 or deletions of PTEN. RNA-Seq analysis of the resistant clones demonstrates expression changes in cell death pathways centering on the p53 upregulated modulator of apoptosis (Puma) protein. Treatment with different tyrosine kinase inhibitors (TKIs) revealed that, in both FGFR1 mutation and Pten deletion-mediated resistance, sustained Akt activation in resistant cells leads to compromised Puma activation, resulting in suppression of TKI-induced apoptosis. This suppression of Puma is achieved as a result of sequestration of inactivated p-Foxo3a in the cytoplasm. CRISPR/Cas9 mediated knockout of Puma in leukemic cells led to an increased drug resistance in the knockout cells demonstrating a direct role in TKI resistance. Since Puma promotes cell death by targeting Bcl2, TKI-resistant cells showed high Bcl2 levels and targeting Bcl2 with Venetoclax (ABT199) led to increased apoptosis in these cells. In vivo treatment of mice xenografted with resistant cells using ABT199 suppressed leukemogenesis and led to prolonged survival. This in-depth survey of the underlying genetic mechanisms of resistance has identified a potential means of treating FGFR1-driven malignancies that are resistant to FGFR1 inhibitors.


Subject(s)
Apoptosis Regulatory Proteins/drug effects , Down-Regulation/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins/drug effects , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Animals , Apoptosis Regulatory Proteins/metabolism , Drug Resistance, Neoplasm/drug effects , Humans , Leukemia/pathology , Lymphoma/genetics , Mice , Signal Transduction/drug effects
8.
Proc Natl Acad Sci U S A ; 117(35): 21723-21730, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32817560

ABSTRACT

G proteins are activated when they associate with G protein-coupled receptors (GPCRs), often in response to agonist-mediated receptor activation. It is generally thought that agonist-induced receptor-G protein association necessarily promotes G protein activation and, conversely, that activated GPCRs do not interact with G proteins that they do not activate. Here we show that GPCRs can form agonist-dependent complexes with G proteins that they do not activate. Using cell-based bioluminescence resonance energy transfer (BRET) and luminescence assays we find that vasopressin V2 receptors (V2R) associate with both Gs and G12 heterotrimers when stimulated with the agonist arginine vasopressin (AVP). However, unlike V2R-Gs complexes, V2R-G12 complexes are not destabilized by guanine nucleotides and do not promote G12 activation. Activating V2R does not lead to signaling responses downstream of G12 activation, but instead inhibits basal G12-mediated signaling, presumably by sequestering G12 heterotrimers. Overexpressing G12 inhibits G protein receptor kinase (GRK) and arrestin recruitment to V2R and receptor internalization. Formyl peptide (FPR1 and FPR2) and Smoothened (Smo) receptors also form complexes with G12 that are insensitive to nucleotides, suggesting that unproductive GPCR-G12 complexes are not unique to V2R. These results indicate that agonist-dependent receptor-G protein association does not always lead to G protein activation and may in fact inhibit G protein activation.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/metabolism , Bioluminescence Resonance Energy Transfer Techniques/methods , Cyclic AMP/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/physiology , GTP-Binding Protein alpha Subunits, Gs/metabolism , GTP-Binding Protein alpha Subunits, Gs/physiology , GTP-Binding Proteins/metabolism , HEK293 Cells , Humans , Ligands , Protein Binding/physiology , Receptors, Vasopressin/metabolism , Signal Transduction/physiology , Vasopressins/metabolism , beta-Arrestins/metabolism
9.
Int J Cancer ; 146(8): 2243-2254, 2020 04 15.
Article in English | MEDLINE | ID: mdl-31525277

ABSTRACT

Constitutive activation of FGFR1, as a result of diverse chromosome translocations, is the hallmark of stem cell leukemia/lymphoma syndrome. The BCR-FGFR1 variant is unique in that the BCR component contributes a serine-threonine kinase (STK) to the N-terminal end of the chimeric FGFR1 kinase. We have deleted the STK domain and mutated the critical Y177 residue and demonstrate that the transforming activity of these mutated genes is reduced compared to the BCR-FGFR1 parental kinase. In addition, we demonstrate that deletion of the FGFR1 tyrosine kinase domain abrogates transforming ability, which is not compensated for by BCR STK activity. Unbiased screening for proteins that are inactivated as a result of loss of the BCR STK identified activated S6 kinase and SHP2 kinase. Genetic and pharmacological inhibition of SHP2 function in SCLL cells expressing BCR-FGFR1 in vitro leads to reduced viability and increased apoptosis. In vivo treatment of SCLL in mice with SHP099 leads to suppression of leukemogenesis, supporting an important role for SHP2 in FGFR1-driven leukemogenesis. In combination with the BGJ398 FGFR1 inhibitor, cell viability in vitro is further suppressed and acts synergistically with SHP099 in vivo suggesting a potential combined targeted therapy option in this subtype of SCLL disease.


Subject(s)
Leukemia/metabolism , Lymphoma/metabolism , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins c-bcr/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Transformation, Neoplastic , Drug Synergism , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukemia/drug therapy , Leukemia/genetics , Leukemia/pathology , Lymphoma/drug therapy , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Inbred BALB C , Oncogene Proteins, Fusion/genetics , Phenylurea Compounds/administration & dosage , Phenylurea Compounds/pharmacology , Piperidines/administration & dosage , Piperidines/pharmacology , Protein Domains , Protein Tyrosine Phosphatase, Non-Receptor Type 11/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Proto-Oncogene Proteins c-bcr/biosynthesis , Proto-Oncogene Proteins c-bcr/genetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Receptor, Fibroblast Growth Factor, Type 1/genetics
11.
J Biol Chem ; 294(48): 18122-18130, 2019 11 29.
Article in English | MEDLINE | ID: mdl-31628193

ABSTRACT

MicroRNA-150-5p (miR-150-5p) plays a complex role in normal early hematopoietic development and is also implicated in the development of various different leukemias. We have reported previously that, in myeloid and lymphoid malignancies associated with dysregulated fibroblast growth factor receptor 1 (FGFR1) activities, miR-150-5p is down-regulated compared with healthy cells. Here, using murine cells, we found that this down-regulation is accompanied by CpG methylation of the miR-150-5p promoter region. Of note, analysis of human acute lymphoblastic leukemia (ALL) cohorts also revealed an inverse relationship between miR-150-5p expression and disease progression. We also found that the DNA methyltransferase 1 (DNMT1) enzyme is highly up-regulated in FGFR1-driven leukemias and lymphomas and that FGFR1 inhibition reduces DNMT1 expression. DNMT1 knockdown in stem cell leukemia/lymphoma (SCLL) cells increased miR-150-5p levels and reduced levels of the MYB proto-oncogene transcription factor, a key regulator of leukemogenesis. FGFR1 directly activates the MYC proto-oncogene basic helix-loop-helix transcription factor, which, as we show here, binds and activates the DNMT1 promoter. MYC knockdown decreased DNMT1 expression, which, in turn, increased miR-150-5p expression. One of the known targets of miR-150-5p is MYB, and treatment of leukemic cells with the MYB inhibitor mebendazole dose-dependently increased apoptosis and reduced cell viability. Moreover, mebendazole treatment of murine xenografts models of FGFR1-driven leukemias enhanced survival. These findings provide evidence that MYC activates MYB by up-regulating DNMT1, which silences miR-150-5p and promotes SCLL progression. We propose that inclusion of mebendazole in a combination therapy with FGFR1 inhibitors may be a valuable option to manage SCLL.


Subject(s)
Carcinogenesis/metabolism , CpG Islands , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , DNA, Neoplasm/metabolism , Leukemia/metabolism , MicroRNAs/biosynthesis , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , RNA, Neoplasm/biosynthesis , Receptor, Fibroblast Growth Factor, Type 1/biosynthesis , Carcinogenesis/genetics , Carcinogenesis/pathology , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA, Neoplasm/genetics , Humans , Leukemia/genetics , Leukemia/pathology , MicroRNAs/genetics , Neoplasm Proteins/genetics , Proto-Oncogene Mas , RNA, Neoplasm/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics
13.
Am J Pathol ; 189(12): 2450-2458, 2019 12.
Article in English | MEDLINE | ID: mdl-31542393

ABSTRACT

The WASF3 gene has been implicated in cancer cell movement, invasion, and metastasis by regulating genetic pathways important in these processes. Invasion and metastasis assays, however, are largely centered on xenograft models in immune-compromised mice. To facilitate analysis of the role of WASF3 in the spontaneous development of cancer cell metastasis, we generated a Wasf3 null strain by deleting exons 4 and 5, which encode essential motifs for Wasf3 function. On exposure to cre-recombinase a stop codon is generated immediately downstream in exon 6. Using a cytomegalovirus (CMV)-cre strain, Wasf3 constitutively was inactivated, which led to viable mice with no visible morphologic or behavioral abnormalities. There was no abnormal development or function of the mouse mammary gland in the Wasf3 null mice and brain development was normal. In the mouse mammary tumor virus (MMTV)-driven polyoma middle-T oncogene strain, which shows early onset breast cancer development and metastasis, Wiskott-Aldrich syndrome protein family member 3 (Wasf3) is up-regulated in metastatic lesions. When this oncogene was introduced onto the Wasf3-null background, although metastasis was observed in these mice, there was a reduction in the number and size of metastatic lesions in the lungs. These data provide evidence for a role in WASF3 in the development of metastasis in a spontaneous model of breast cancer.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Lung Neoplasms/secondary , Wiskott-Aldrich Syndrome Protein Family/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Movement , Cohort Studies , Disease Models, Animal , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Invasiveness , Signal Transduction , Tumor Cells, Cultured , Wiskott-Aldrich Syndrome Protein Family/genetics
14.
Nat Immunol ; 20(9): 1231-1243, 2019 09.
Article in English | MEDLINE | ID: mdl-31358999

ABSTRACT

Understanding resistance to antibody to programmed cell death protein 1 (PD-1; anti-PD-1) is crucial for the development of reversal strategies. In anti-PD-1-resistant models, simultaneous anti-PD-1 and vaccine therapy reversed resistance, while PD-1 blockade before antigen priming abolished therapeutic outcomes. This was due to induction of dysfunctional PD-1+CD38hi CD8+ cells by PD-1 blockade in suboptimally primed CD8 cell conditions induced by tumors. This results in erroneous T cell receptor signaling and unresponsiveness to antigenic restimulation. On the other hand, PD-1 blockade of optimally primed CD8 cells prevented the induction of dysfunctional CD8 cells, reversing resistance. Depleting PD-1+CD38hi CD8+ cells enhanced therapeutic outcomes. Furthermore, non-responding patients showed more PD-1+CD38+CD8+ cells in tumor and blood than responders. In conclusion, the status of CD8+ T cell priming is a major contributor to anti-PD-1 therapeutic resistance. PD-1 blockade in unprimed or suboptimally primed CD8 cells induces resistance through the induction of PD-1+CD38hi CD8+ cells that is reversed by optimal priming. PD-1+CD38hi CD8+ cells serve as a predictive and therapeutic biomarker for anti-PD-1 treatment. Sequencing of anti-PD-1 and vaccine is crucial for successful therapy.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , CD8-Positive T-Lymphocytes/immunology , Drug Resistance, Neoplasm/immunology , Membrane Glycoproteins/metabolism , Neoplasms/immunology , Programmed Cell Death 1 Receptor/immunology , ADP-ribosyl Cyclase 1/genetics , Animals , Antibodies/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/immunology , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Humans , Immunotherapy/methods , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/immunology
15.
Biochim Biophys Acta Mol Basis Dis ; 1865(3): 688-695, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30625381

ABSTRACT

Inflammation and cellular energetics play critical roles in organ dysfunction following hemorrhagic shock. Recent studies suggest a putative role for sirtuin 1 (SIRT1) in potentiating mitochondrial function and improving organ function following hemorrhagic shock in animal models. SIRT1 is an NAD+ dependent protein deacetylase and increased availability of NAD+ has been shown to augment SIRT1 activity. As niacin is a precursor of NAD+, in this study, we tested whether niacin can improve survival following hemorrhagic shock. However niacin also mediates its biological action by binding to its receptor, hydroxyl-carboxylic acid receptor 2 (HCA2 or Gpr109a); so we examined whether the effect of niacin is mediated by binding to Gpr109a or by increasing NAD+ availability. We found that niacin administered intravenously to rats subjected to hemorrhagic injury (HI) in the absence of fluid resuscitation resulted in a significantly prolonged duration of survival. However, treatment of rats with similar doses of nicotinamide mononucleotide (NMN), a precursor to NAD+ that does not bind Gpr109a, did not extend survival following HI. The duration of survival due to niacin treatment was significantly reduced in Gpr109a-/- mice subjected to HI. These experiments demonstrated that the Gpr109a receptor-mediated pathway contributed significantly to niacin mediated salutary effect. Further studies showed improvement in markers of cellular energetics and attenuation of inflammatory response with niacin treatment. In conclusion, we report that Gpr109a-dependent signalling is important in restoring cellular energetics and immunometabolism following hemorrhagic shock.


Subject(s)
Niacin/therapeutic use , Receptors, G-Protein-Coupled/genetics , Shock, Hemorrhagic/drug therapy , Animals , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardium/metabolism , Myocardium/pathology , NAD/metabolism , NADP/metabolism , Niacin/metabolism , Permeability/drug effects , Receptors, G-Protein-Coupled/metabolism , Shock, Hemorrhagic/genetics , Shock, Hemorrhagic/mortality , Shock, Hemorrhagic/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Survival Analysis
16.
RSC Adv ; 9(33): 19086-19094, 2019 Jun 14.
Article in English | MEDLINE | ID: mdl-35516882

ABSTRACT

Nanoslits composed of different layered nanomaterials attract great attention in the theoretical and experimental investigations of nanofluidic devices due to their geometric simplicity and unique surface properties. Although many efforts have witnessed simulations of water molecules inside slit-like nanochannels formed by graphenes, the thermodynamic properties and transport behavior of water inside nanoslits formed by different two-dimensional materials are seldom investigated. In this paper, we choose nanoslits formed by graphene, boron nitride (hBN), and molybdenum disulfide (MoS2) as models, and study the water properties inside these nanoslits using traditional molecular dynamics simulations at different pressures. It is shown that water molecules can form a planar square at high pressure (10 kbar) in all three types of nanoslit. The nanoslits affect diffusion coefficient, orientation of water molecules, number of hydrogen bonds and life-time of hydrogen bonding significantly. The self-diffusion coefficients of water molecules in different nanoslits are all lower than that of bulk water. The diffusion coefficients are significantly affected by the special ordered structure of water, which is caused by the unique surface structure of the nanoslit. The results of the present work will be helpful to understand the unique behavior of confined water in nanoslits composed of different nanomaterials and provide theoretical guidance for many applications, such as desalination and nano-energy conversion.

17.
Cancer Res ; 79(1): 114-124, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30413411

ABSTRACT

Transformation of hematopoietic stem cells by the BCR-FGFR1 fusion kinase found in a variant of stem cell leukemia/lymphoma (SCLL) syndrome leads to development of B-lymphomas in syngeneic mice and humans. In this study, we show that the relatively rapid onset of this leukemia is potentially related to oncogenic domains within the BCR component. BCR recruited a guanidine nucleotide exchange factor (GEF) domain to the fusion kinase to facilitate activation of small GTPases such as the Ras homology gene family, member A (RHOA). Deletion of this GEF domain increased leukemogenesis, enhanced cell survival and proliferation, and promoted stem cell expansion and lymph node metastasis. This suggests that, in an SCLL context, the presence of the endogenous GEF motif leads to reduced leukemogenesis. Indeed, loss of the GEF domain suppressed activation of RHOA and PTEN, leading to increased activation of AKT. Loss of the GEF domain enhanced cell proliferation and invasion potential, which was also observed in cells in which RHOA is knocked down, supported by the observation that overexpression of RHOA leads to reduced viability and invasion. In vivo depletion of RHOA in SCLL cells significantly increased disease progression and shortened latency. Collectively, these data show that the BCR GEF domain affects phenotypes associated with progression of SCLL through suppression of RHOA signaling. SIGNIFICANCE: RHOA activation is a critical event in the progression of BCR-FGFR1-driven leukemogenesis in stem cell leukemia and lymphoma syndrome and is regulated by the BCR GEF domain.


Subject(s)
Guanine Nucleotide Exchange Factors/metabolism , Leukemia, Experimental/pathology , Lymphoma/pathology , Precursor Cells, B-Lymphoid/pathology , Proto-Oncogene Proteins c-bcr/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , rho GTP-Binding Proteins/metabolism , Animals , Cell Movement , Cell Proliferation , Cells, Cultured , Guanine Nucleotide Exchange Factors/genetics , Leukemia, Experimental/genetics , Leukemia, Experimental/metabolism , Lymphoma/genetics , Lymphoma/metabolism , Mice , Mice, Inbred BALB C , Precursor Cells, B-Lymphoid/metabolism , Protein Domains , Proto-Oncogene Proteins c-bcr/genetics , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction , rho GTP-Binding Proteins/genetics , rhoA GTP-Binding Protein
18.
Cancer Res ; 78(13): 3522-3531, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29735550

ABSTRACT

The development of myeloid and lymphoid neoplasms related to overexpression of FGFR1 kinases as a result of chromosome translocations depends on the promotion of a stem cell phenotype, suppression of terminal differentiation, and resistance to apoptosis. These phenotypes are related to the stem cell leukemia/lymphoma syndrome (SCLL), which arises through the effects of the activated FGFR1 kinase on gene transcription, which includes miRNA dysregulation. In a screen for miRNAs that are directly regulated by FGFR1, and which stimulate cell proliferation and survival, we identified miR-339-5p, which is highly upregulated in cells carrying various different chimeric kinases. Overexpression of miR-339-5p in SCLL cell types enhances cell survival and inhibition of its function leads to reduced cell viability. miR-339-5p overexpression protects cells from the consequences of FGFR1 inactivation, promoting cell-cycle progression and reduced apoptosis. Transient luciferase reporter assays and qRT-PCR detection of endogenous miR-339-5p expression in stably transduced cell lines demonstrated that BCR-FGFR1 can directly regulate miR-339-5p expression. This correlation between miR-339-5p and FGFR1 expression is also seen in primary human B-cell precursor acute lymphoblastic leukemia. In a screen to identify targets of miR-339-5p, we identified and verified the BCL2L11 and BAX genes, which can promote apoptosis. In vivo, SCLL cells forced to overexpress miR-339-5p show a more rapid onset of disease and poorer survival compared with parental cells expressing endogenous levels of miR-339-5p. Analysis of human primary B-cell precursor ALL shows a significant higher expression of miR339-5p compared with the two cohorts of CLL patient samples, suggesting direct roles in disease progression and supporting the evidence generated in mouse models of SCLL.Significance: Proapoptiotic genes that are direct targets of miR-339-5p significantly influence promotion and aggressive development of leukemia/lymphomas associated with FGFR1 overexpression. Cancer Res; 78(13); 3522-31. ©2018 AACR.


Subject(s)
Bcl-2-Like Protein 11/genetics , Leukemia/genetics , Lymphoma/genetics , MicroRNAs/metabolism , bcl-2-Associated X Protein/genetics , Animals , Bcl-2-Like Protein 11/metabolism , Cell Line, Tumor/transplantation , Cell Survival/genetics , Chromosomes, Human, Pair 8/genetics , Disease Models, Animal , Down-Regulation , Female , HEK293 Cells , Hematopoietic Stem Cells/pathology , Humans , Leukemia/pathology , Lymphoma/pathology , Mice , Mice, Inbred BALB C , NIH 3T3 Cells , Oncogene Proteins, Fusion/metabolism , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Syndrome , Translocation, Genetic , bcl-2-Associated X Protein/metabolism
19.
Epigenetics Chromatin ; 10(1): 54, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29126443

ABSTRACT

BACKGROUND: Ubiquitin C-terminal hydrolase isozyme L1 (UCHL1) is primarily expressed in neuronal cells and neuroendocrine cells and has been associated with various diseases, including many cancers. It is a multifunctional protein involved in deubiquitination, ubiquitination and ubiquitin homeostasis, but its specific roles are disputed and still generally undetermined. RESULTS: Herein, we demonstrate that UCHL1 is associated with genomic DNA in certain prostate cancer cell lines, including DU 145 cells derived from a brain metastatic site, and in HEK293T embryonic kidney cells with a neuronal lineage. Chromatin immunoprecipitation and sequencing revealed that UCHL1 localizes to TTAGGG repeats at telomeres and interstitial telomeric sequences, as do TRF1 and TRF2, components of the shelterin complex. A weak or transient interaction between UCHL1 and the shelterin complex was confirmed by immunoprecipitation and proximity ligation assays. UCHL1 and RAP1, also known as TERF2IP and a component of the shelterin complex, were bound to the nuclear scaffold. CONCLUSIONS: We demonstrated a novel feature of UCHL1 in binding telomeres and interstitial telomeric sites.


Subject(s)
Telomere-Binding Proteins/metabolism , Telomere/metabolism , Ubiquitin Thiolesterase/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Protein Binding , Shelterin Complex , Telomeric Repeat Binding Protein 1/metabolism , Telomeric Repeat Binding Protein 2/metabolism
20.
Sci Rep ; 7(1): 2671, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28572638

ABSTRACT

Hemorrhagic shock is a leading cause of death in people under the age of 45 and accounts for almost half of trauma-related deaths. In order to develop a treatment strategy based on potentiating mitochondrial function, we investigated the effect of the orphan drug dichloroacetate (DCA) on survival in an animal model of hemorrhagic shock in the absence of fluid resuscitation. Hemorrhagic shock was induced in rats by withdrawing 60% of the blood volume and maintaining a hypotensive state. The studies demonstrated prolonged survival of rats subjected to hemorrhagic injury (HI) when treated with DCA. In separate experiments, using a fluid resuscitation model we studied mitochondrial functional alterations and changes in metabolic networks connected to mitochondria following HI and treatment with DCA. DCA treatment restored cardiac mitochondrial membrane potential and tissue ATP in the rats following HI. Treatment with DCA resulted in normalization of several metabolic and molecular parameters including plasma lactate and p-AMPK/AMPK, as well as Ach-mediated vascular relaxation. In conclusion we demonstrate that DCA can be successfully used in the treatment of hemorrhagic shock in the absence of fluid resuscitation; therefore DCA may be a good candidate in prolonged field care following severe blood loss.


Subject(s)
Chloroacetates/administration & dosage , Mitochondria/drug effects , Mitochondria/metabolism , Shock, Hemorrhagic/metabolism , Animals , Dose-Response Relationship, Drug , Kaplan-Meier Estimate , Male , Membrane Potential, Mitochondrial , Orphan Drug Production , Rats, Sprague-Dawley , Shock, Hemorrhagic/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...