Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Proteins ; 90(4): 919-935, 2022 04.
Article in English | MEDLINE | ID: mdl-34773424

ABSTRACT

Detailed description of the mechanism of action of the therapeutic antibodies is essential for the functional characterization and future optimization of potential clinical agents. We recently developed KD035, a fully human antibody targeting vascular endothelial growth factor receptor 2 (VEGFR2). KD035 blocked VEGF-A, and VEGF-C-mediated VEGFR2 activation, as demonstrated by the in vitro binding and competition assays and functional cellular assays. Here, we report a computational model of the complex between the variable fragment of KD035 (KD035(Fv)) and the domains 2 and 3 of the extracellular portion of VEGFR2 (VEGFR2(D2-3)). Our modeling was guided by a priori experimental information including the X-ray structures of KD035 and related antibodies, binding assays, target domain mapping and comparison of KD035 affinity for VEGFR2 from different species. The accuracy of the model was assessed by molecular dynamics simulations, and subsequently validated by mutagenesis and binding analysis. Importantly, the steps followed during the generation of this model can set a precedent for future in silico efforts aimed at the accurate description of the antibody-antigen and more broadly protein-protein complexes.


Subject(s)
Antibodies , Vascular Endothelial Growth Factor A , Humans , Molecular Dynamics Simulation , Vascular Endothelial Growth Factor A/metabolism
2.
Cell Chem Biol ; 29(3): 423-435.e10, 2022 03 17.
Article in English | MEDLINE | ID: mdl-34715056

ABSTRACT

Efforts to target glucose metabolism in cancer have been limited by the poor potency and specificity of existing anti-glycolytic agents and a poor understanding of the glucose dependence of cancer subtypes in vivo. Here, we present an extensively characterized series of potent, orally bioavailable inhibitors of the class I glucose transporters (GLUTs). The representative compound KL-11743 specifically blocks glucose metabolism, triggering an acute collapse in NADH pools and a striking accumulation of aspartate, indicating a dramatic shift toward oxidative phosphorylation in the mitochondria. Disrupting mitochondrial metabolism via chemical inhibition of electron transport, deletion of the malate-aspartate shuttle component GOT1, or endogenous mutations in tricarboxylic acid cycle enzymes, causes synthetic lethality with KL-11743. Patient-derived xenograft models of succinate dehydrogenase A (SDHA)-deficient cancers are specifically sensitive to KL-11743, providing direct evidence that TCA cycle-mutant tumors are vulnerable to GLUT inhibitors in vivo.


Subject(s)
Citric Acid Cycle , Neoplasms , Aspartic Acid/metabolism , Glucose/metabolism , Humans , Mitochondria/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism
3.
J Med Chem ; 63(10): 5201-5211, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32282207

ABSTRACT

Aerobic glycolysis, originally identified by Warburg as a hallmark of cancer, has recently been implicated in immune cell activation and growth. Glucose, the starting material for glycolysis, is transported through the cellular membrane by a family of glucose transporters (GLUTs). Therefore, targeting glucose transporters to regulate aerobic glycolysis is an attractive approach to identify potential therapeutic agents for cancers and autoimmune diseases. Herein, we describe the discovery and optimization of a class of potent, orally bioavailable inhibitors of glucose transporters, targeting both GLUT1 and GLUT3.


Subject(s)
Drug Discovery/methods , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transport Proteins, Facilitative/metabolism , Glucose/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Caco-2 Cells , Dose-Response Relationship, Drug , Drug Discovery/trends , Glycolysis/drug effects , Glycolysis/physiology , Humans , Mice , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats
4.
AIDS Res Hum Retroviruses ; 20(10): 1063-71, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15585097

ABSTRACT

CXCR4, the specific receptor for the chemokine SDF-1 alpha that also binds CXCR4-using HIV gp120s, affects survival of different cell types, including neurons. However, current data show that the outcome of CXCR4 activation on neuronal survival may vary depending on the ligand and/or the cellular conditions. In this study, we have systematically compared the effects of SDF-1 alpha and gp120(IIIB) (with or without CD4) on several intracellular pathways involved in cell survival, including MAP kinases and Akt-dependent pathways. Our data show that gp120(IIIB) and SDF-1 alpha are both potent activators of MAP kinases in neuronal and non-neuronal cells, though the kinetic of these responses is slightly different. Furthermore, unlike SDF-1 alpha, and independently of CD4, gp120(IIIB) is unable to stimulate Akt and some of its antiapoptotic targets (NF-kappa B and MDM2)--despite its ability to activate other signaling pathways in the same conditions. Finally, the viral protein is more efficient in recruiting some effectors (e.g., JNK) than others in comparison with SDF-1 alpha (EC(50) = 0.1 vs. 0.6 nM). We conclude that the intrinsic efficacy of the two ligands is significantly different and is pathway dependent. These findings have important implications for our understanding of CXCR4-mediated responses in the CNS, as well as the role of this coreceptor in HIV neuropathogenesis.


Subject(s)
Apoptosis , HIV Envelope Protein gp120/metabolism , HIV-1/pathogenicity , Neurons/physiology , Receptors, CXCR4/metabolism , Animals , Astrocytes/physiology , Cell Line, Tumor , Cell Survival , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/metabolism , Enzyme Activation , Gene Expression Regulation , HIV Envelope Protein gp120/genetics , Humans , Mitogen-Activated Protein Kinases/metabolism , Rats , Receptors, CXCR4/genetics , Signal Transduction
5.
J Neurovirol ; 12(3): 211-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16877302

ABSTRACT

The chemokine receptor CXCR4 functions as human immunodeficiency virus (HIV)-1 coreceptor and is involved in acquired immunodeficiency virus (AIDS) neuropathogenesis. CXCR4 is expressed by most cell types in the brain, including microglia, astrocytes, and neurons. Studies have shown that the HIV envelope protein gp120 binds to neuronal CXCR4 and activates signal transduction pathways leading to apoptosis. However, the natural CXCR4 ligand (CXCL12) has been referred to induce both neuronal survival and death. Here the authors used flow cytometry to determine whether gp120 and CXCL12 differ in their ability to induce CXCR4 internalization in the human neuroblastoma cells SH-SY5Y, which constitutively express CXCR4. As expected, increasing concentration of CXCL12 reduced surface expression of CXCR4 in a time-and concentration-dependent manner. Conversely, gp120IIIB (monomeric or oligomeric, in presence or absence of soluble CD4) did not change CXCR4 membrane levels. Similar results were obtained in a murine lymphocyte cell line (300-19) stably expressing human CXCR4. Nevertheless, gp120IIIB was still able to activate intracellular signaling and proapoptotic pathways, via CXCR4. These results show that gp120IIIB toxicity and signaling do not require CXCR4 internalization in SH-SY5Y cells, and suggest that the viral protein may alter normal CXCR4 trafficking thus, interfering with activation of prosurvival pathways.


Subject(s)
Apoptosis/physiology , HIV Envelope Protein gp120/metabolism , Neurons/pathology , Neurons/virology , Receptors, CXCR4/metabolism , Apoptosis/drug effects , Blotting, Western , Cell Line, Tumor , Chemokine CXCL12 , Chemokines, CXC/pharmacology , Endocytosis/physiology , Flow Cytometry , HIV Envelope Protein gp120/pharmacology , Humans , Neuroblastoma , Signal Transduction/drug effects , Signal Transduction/physiology
6.
J Neurovirol ; 12(6): 492-500, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17162664

ABSTRACT

The chemokine receptor CXCR4 regulates neuronal survival and differentiation and is involved in a number of pathologies, including cancer and human immunodeficiency virus (HIV). Recent data suggest that chemokines act in concert with neurotransmitters and neuropeptides, such as opioids. This study aimed to determine whether mu-opioid agonists alter the effect of CXCL12 (the specific CXCR4 ligand) on central neurons. Neuronal expression of CXCR4 and micro-opioid receptors (MORs) was analyzed by Western blot, immunostaining, and flow cytometry. Single-cell studies showed that all CXCR4-positive neurons coexpress MORs. Treatment of neuronal cultures with the selective MOR agonist DAMGO or the endogenous peptide endomorphin-1 inhibited intracellular signaling pathways (ERK1/2 and Akt) activated by CXCL12. Furthermore, DAMGO abolished the neuroprotective effect of CXCL12 in N-methyl-d-aspartate (NMDA) neurotoxicity studies. The effects of DAMGO and endomorphin-1 were inhibited by a general or a micro-specific opioid receptor antagonist, and not caused by changes in neuronal CXCR4 levels. DAMGO did not affect CXCL12-induced internalization of CXCR4. The authors propose that interactions between MOR and CXCR4 signaling can modulate the action of CXCL12 on neuronal survival-which may have important implications to neuroAIDS as well as other neuroinflammatory disorders.


Subject(s)
Analgesics, Opioid/pharmacology , Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology , Neurons/drug effects , Receptors, CXCR4/metabolism , Receptors, Opioid, mu/metabolism , Animals , Blotting, Western , Cell Survival , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Flow Cytometry , Humans , Immunohistochemistry , Microscopy, Confocal , Narcotic Antagonists/pharmacology , Neurons/metabolism , Oligopeptides/metabolism , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Receptor Cross-Talk/drug effects , Receptor Cross-Talk/physiology , Receptors, CXCR4/drug effects , Receptors, Opioid, mu/drug effects
7.
Mol Cell Neurosci ; 30(1): 58-66, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16005638

ABSTRACT

Abnormal activation of CXCR 4 during inflammatory/infectious states may lead to neuronal dysfunction or damage. The major goal of this study was to determine the coupling of CXCR 4 to p53-dependent survival pathways in primary neurons. Neurons were stimulated with the HIV envelope protein gp120(IIIB) or the endogenous CXCR 4 agonist, SDF-1 alpha. We found that gp120 stimulates p53 activity and induces expression of the p53 pro-apoptotic target Apaf-1 in cultured neurons. Inhibition of CXCR 4 by AMD 3100 abrogates the effect of gp120 on both p53 and Apaf-1. Moreover, gp120 neurotoxicity is markedly reduced by the p53-inhibitor, pifithrin-alpha. The viral protein also regulates p53 phosphorylation and expression of other p53-responsive genes, such as MDM 2 and p21. Conversely, SDF-1 alpha, which can promote neuronal survival, increases p53 acetylation and p21 expression in neurons. Thus, the stimulation of different p53 targets could be instrumental in determining the outcome of CXCR 4 activation on neuronal survival in neuro-inflammatory disorders.


Subject(s)
Neurons/cytology , Neurons/metabolism , Receptors, CXCR4/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptotic Protease-Activating Factor 1 , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/pharmacology , HIV Envelope Protein gp120/toxicity , Phosphorylation/drug effects , Proteins/metabolism , Rats , Transcription, Genetic , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL