Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Glia ; 67(8): 1558-1570, 2019 08.
Article in English | MEDLINE | ID: mdl-31058365

ABSTRACT

GPR124 is involved in embryonic development and remains expressed by select organs. The importance of GPR124 during development suggests that its aberrant expression might participate in tumor growth. Here we show that both increases and decreases in GPR124 expression in glioblastoma cells reduce cell proliferation by differentially altering the duration mitotic progression. Using mass spectrometry-based proteomics, we discovered that GPR124 interacts with ch-TOG, a known regulator of both microtubule (MT)-plus-end assembly and mitotic progression. Accordingly, changes in GPR124 expression and ch-TOG similarly affect MT assembly measured by real-time microscopy in cells. Our study describes a novel molecular interaction involving GPR124 and ch-TOG at the plasma membrane that controls glioblastoma cell proliferation by modifying MT assembly rates and controlling the progression of distinct phases of mitosis.


Subject(s)
Brain Neoplasms/metabolism , Cell Proliferation/physiology , Glioblastoma/metabolism , Microtubules/metabolism , Mitosis/physiology , Receptors, G-Protein-Coupled/metabolism , Adult , Brain/metabolism , Cell Membrane/metabolism , Cells, Cultured , Female , Gene Expression , Humans , Male , Microtubule-Associated Proteins/metabolism , Middle Aged
2.
Mol Cancer Ther ; 15(9): 2018-29, 2016 09.
Article in English | MEDLINE | ID: mdl-27325686

ABSTRACT

Glioblastoma multiforme is a devastating and intractable type of cancer. Current antineoplastic drugs do not improve the median survival of patients diagnosed with glioblastoma multiforme beyond 14 to 15 months, in part because the blood-brain barrier is generally impermeable to many therapeutic agents. Drugs that target microtubules (MT) have shown remarkable efficacy in a variety of cancers, yet their use as glioblastoma multiforme treatments has also been hindered by the scarcity of brain-penetrant MT-targeting compounds. We have discovered a new alkylindole compound, ST-11, that acts directly on MTs and rapidly attenuates their rate of assembly. Accordingly, ST-11 arrests glioblastoma multiforme cells in prometaphase and triggers apoptosis. In vivo analyses reveal that unlike current antitubulin agents, ST-11 readily crosses the blood-brain barrier. Further investigation in a syngeneic orthotopic mouse model of glioblastoma multiforme shows that ST-11 activates caspase-3 in tumors to reduce tumor volume without overt toxicity. Thus, ST-11 represents the first member of a new class of brain-penetrant antitubulin therapeutic agents. Mol Cancer Ther; 15(9); 2018-29. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Microtubules/metabolism , Tubulin Modulators/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Caspase 3/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Mice , Nanoparticles , Pilot Projects , Solubility , Tubulin Modulators/administration & dosage , Tubulin Modulators/pharmacokinetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
3.
Glia ; 63(10): 1797-808, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25914169

ABSTRACT

Ligands targeting G protein-coupled receptors (GPCR) expressed by microglia have been shown to regulate distinct components of their activation process, including cell proliferation, migration and differentiation into M1 or M2 phenotypes. Cannabinoids, including the active component of the Cannabis plant, tetrahydrocannabinol (THC), and the synthetic alkylindole (AI) compound, WIN55212-2 (WIN-2), activate two molecularly identified GPCRs: CB1 and CB2 . Previous studies reported that WIN-2 activates an additional unknown GPCR that is not activated by plant-derived cannabinoids, and evidence indicates that microglia express these receptors. Detailed studies on the role of AI-sensitive receptors in microglial cell activation were difficult as no selective pharmacological tools were available. Here, three newly-developed AI analogues allowed us to determine if microglia express AI-sensitive receptors and if so, study how they regulate the microglial cell activation process. We found that mouse microglia in primary culture express functional AI-sensitive receptors as measured by radioligand binding and changes in intracellular cAMP levels, and that these receptors control both basal and ATP-stimulated migration. AI analogues inhibit cell proliferation stimulated by macrophage-colony stimulating factor (M-CSF) without affecting basal cell proliferation. Remarkably, AI analogues do not control the expression of effector proteins characteristic of M1 or M2 phenotypes; yet activating microglia with M1 and M2 cytokines reduces the microglial response to AI analogues. Our results suggest that microglia express functional AI-sensitive receptors that control select components of their activation process. Agonists of these novel targets might represent a novel class of therapeutics to influence the microglial cell activation process.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , Microglia/physiology , Receptors, G-Protein-Coupled/metabolism , Adenosine Triphosphate/pharmacology , Animals , Animals, Newborn , Benzoxazines/pharmacology , Brain/cytology , Calcium Channel Blockers/pharmacology , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokine CXCL10/metabolism , Cyclic AMP/metabolism , Cytokines/pharmacology , Dronabinol/pharmacology , Mice , Mice, Inbred C57BL , Microglia/drug effects , Morpholines/pharmacology , Naphthalenes/pharmacology , Nitric Oxide/metabolism
4.
Neuron ; 83(2): 361-371, 2014 Jul 16.
Article in English | MEDLINE | ID: mdl-25033180

ABSTRACT

The serine hydrolase α/ß-hydrolase domain 6 (ABHD6) hydrolyzes the most abundant endocannabinoid (eCB) in the brain, 2-arachidonoylglycerol (2-AG), and controls its availability at cannabinoid receptors. We show that ABHD6 inhibition decreases pentylenetetrazole (PTZ)-induced generalized tonic-clonic and myoclonic seizure incidence and severity. This effect is retained in Cnr1(-/-) or Cnr2(-/-) mice, but blocked by addition of a subconvulsive dose of picrotoxin, suggesting the involvement of GABAA receptors. ABHD6 inhibition also blocked spontaneous seizures in R6/2 mice, a genetic model of juvenile Huntington's disease known to exhibit dysregulated eCB signaling. ABHD6 blockade retained its antiepileptic activity over chronic dosing and was not associated with psychomotor or cognitive effects. While the etiology of seizures in R6/2 mice remains unsolved, involvement of the hippocampus is suggested by interictal epileptic discharges, increased expression of vGLUT1 but not vGAT, and reduced Neuropeptide Y (NPY) expression. We conclude that ABHD6 inhibition may represent a novel antiepileptic strategy.


Subject(s)
Anticonvulsants/therapeutic use , Brain/drug effects , Carbamates/therapeutic use , Monoacylglycerol Lipases/antagonists & inhibitors , Seizures/drug therapy , Animals , Anticonvulsants/pharmacology , Behavior, Animal/drug effects , Brain/physiopathology , Carbamates/pharmacology , Male , Mice , Mice, Knockout , Pentylenetetrazole , Receptors, Cannabinoid/genetics , Seizures/chemically induced , Seizures/physiopathology
5.
Eur J Neurosci ; 37(3): 429-40, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23167744

ABSTRACT

Cannabinoid receptor 1 (CB(1) receptor) controls several neuronal functions, including neurotransmitter release, synaptic plasticity, gene expression and neuronal viability. Downregulation of CB(1) expression in the basal ganglia of patients with Huntington's disease (HD) and animal models represents one of the earliest molecular events induced by mutant huntingtin (mHtt). This early disruption of neuronal CB(1) signaling is thought to contribute to HD symptoms and neurodegeneration. Here we determined whether CB(1) downregulation measured in patients with HD and mouse models was ubiquitous or restricted to specific striatal neuronal subpopulations. Using unbiased semi-quantitative immunohistochemistry, we confirmed previous studies showing that CB(1) expression is downregulated in medium spiny neurons of the indirect pathway, and found that CB(1) is also downregulated in neuropeptide Y (NPY)/neuronal nitric oxide synthase (nNOS)-expressing interneurons while remaining unchanged in parvalbumin- and calretinin-expressing interneurons. CB(1) downregulation in striatal NPY/nNOS-expressing interneurons occurs in R6/2 mice, Hdh(Q150/Q150) mice and the caudate nucleus of patients with HD. In R6/2 mice, CB(1) downregulation in NPY/nNOS-expressing interneurons correlates with diffuse expression of mHtt in the soma. This downregulation also occludes the ability of cannabinoid agonists to activate the pro-survival signaling molecule cAMP response element-binding protein in NPY/nNOS-expressing interneurons. Loss of CB(1) signaling in NPY/nNOS-expressing interneurons could contribute to the impairment of basal ganglia functions linked to HD.


Subject(s)
Basal Ganglia/metabolism , Down-Regulation , Huntington Disease/metabolism , Interneurons/metabolism , Neuropeptide Y/metabolism , Receptor, Cannabinoid, CB1/metabolism , Adult , Aged , Animals , Basal Ganglia/cytology , Calbindin 2 , Cannabinoid Receptor Agonists/pharmacology , Case-Control Studies , Cyclic AMP/metabolism , Disease Models, Animal , Female , Gene Expression , Humans , Huntingtin Protein , Interneurons/classification , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Nerve Tissue Proteins/genetics , Neuropeptide Y/genetics , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase/metabolism , Nuclear Proteins/genetics , Parvalbumins/genetics , Parvalbumins/metabolism , Receptor, Cannabinoid, CB1/agonists , Receptor, Cannabinoid, CB1/genetics , S100 Calcium Binding Protein G/genetics , S100 Calcium Binding Protein G/metabolism , Serotonin Plasma Membrane Transport Proteins/genetics , Serotonin Plasma Membrane Transport Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...