Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Cell Biochem ; 125(3): e30526, 2024 03.
Artigo em Inglês | MEDLINE | ID: mdl-38229533

RESUMO

Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are devastating neurodegenerative diseases with no effective cure. GGGGCC repeat expansion in C9orf72 is the most common genetic cause of both ALS and FTD. A key pathological feature of C9orf72 related ALS/FTD is the presence of abnormal dipeptide repeat proteins translated from GGGGCC repeat expansion, including poly Glycine-Arginine (GR). In this study, we observed that (GR)50 conferred significant mitochondria damage and cytotoxicity. Metformin, the most widely used clinical drug, successfully relieved (GR)50 induced mitochondrial damage and inhibited (GR)50 related cytotoxicity. Further research revealed metformin effectively restored mitochondrial function by upregulating AKT phosphorylation in (GR)50 expressed cells. Taken together, our results indicated restoring mitochondrial function with metformin may be a rational therapeutic strategy to reduce poly(GR) toxicity in C9orf72 ALS/FTD patients.


Assuntos
Esclerose Lateral Amiotrófica , Demência Frontotemporal , Humanos , Demência Frontotemporal/tratamento farmacológico , Demência Frontotemporal/genética , Demência Frontotemporal/metabolismo , Esclerose Lateral Amiotrófica/tratamento farmacológico , Esclerose Lateral Amiotrófica/genética , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteína C9orf72/genética , Proteína C9orf72/metabolismo , Fosforilação , Dipeptídeos
3.
Mol Cell ; 54(1): 119-132, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24657168

RESUMO

The apoptotic actions of p53 require its phosphorylation by a family of phosphoinositide-3-kinase-related-kinases (PIKKs), which include DNA-PKcs and ATM. These kinases are stabilized by the TTT (Tel2, Tti1, Tti2) cochaperone family, whose actions are mediated by CK2 phosphorylation. The inositol pyrophosphates, such as 5-diphosphoinositol pentakisphosphate (IP7), are generated by a family of inositol hexakisphosphate kinases (IP6Ks), of which IP6K2 has been implicated in p53-associated cell death. In the present study we report an apoptotic signaling cascade linking CK2, TTT, the PIKKs, and p53. We demonstrate that IP7, formed by IP6K2, binds CK2 to enhance its phosphorylation of the TTT complex, thereby stabilizing DNA-PKcs and ATM. This process stimulates p53 phosphorylation at serine 15 to activate the cell death program in human cancer cells and in murine B cells.


Assuntos
Apoptose , Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Proteínas de Transporte/metabolismo , Caseína Quinase II/metabolismo , Proteína Quinase Ativada por DNA/metabolismo , Proteínas de Ligação a DNA/metabolismo , Fosfatos de Inositol/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas de Ligação a Telômeros/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Proteínas Mutadas de Ataxia Telangiectasia/genética , Linfócitos B/enzimologia , Linfócitos B/patologia , Sítios de Ligação , Proteínas de Transporte/genética , Caseína Quinase II/genética , Proteína Quinase Ativada por DNA/genética , Proteínas de Ligação a DNA/genética , Estabilidade Enzimática , Células HCT116 , Células HEK293 , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos Knockout , Neoplasias/enzimologia , Neoplasias/genética , Neoplasias/patologia , Proteínas Nucleares/genética , Fosforilação , Fosfotransferases (Aceptor do Grupo Fosfato)/deficiência , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Proteínas Proto-Oncogênicas c-ets/genética , Interferência de RNA , Serina , Transdução de Sinais , Proteínas de Ligação a Telômeros/genética , Transfecção , Proteína Supressora de Tumor p53/genética
4.
Proc Natl Acad Sci U S A ; 116(8): 3278-3287, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30718399

RESUMO

Cells express a family of three inositol hexakisphosphate kinases (IP6Ks). Although sharing the same enzymatic activity, individual IP6Ks mediate different cellular processes. Here we report that IP6K3 is enriched at the leading edge of migrating cells where it associates with dynein intermediate chain 2 (DIC2). Using immunofluorescence microscopy and total internal reflection fluorescence microscopy, we found that DIC2 and IP6K3 are recruited interdependently to the leading edge of migrating cells, where they function coordinately to enhance the turnover of focal adhesions. Deletion of IP6K3 causes defects in cell motility and neuronal dendritic growth, eventually leading to brain malformations. Our results reveal a mechanism whereby IP6K3 functions in coordination with DIC2 in a confined intracellular microenvironment to promote focal adhesion turnover.


Assuntos
Dineínas do Citoplasma/genética , Dendritos/genética , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Encéfalo/metabolismo , Encéfalo/patologia , Adesão Celular/genética , Movimento Celular/genética , Microambiente Celular/genética , Adesões Focais/genética , Células HEK293 , Humanos , Neurônios/metabolismo
5.
Circ Res ; 122(3): 457-472, 2018 02 02.
Artigo em Inglês | MEDLINE | ID: mdl-29279301

RESUMO

RATIONALE: Inositol polyphosphate multikinase (IPMK) and its major product inositol pentakisphosphate (IP5) regulate a variety of cellular functions, but their role in vascular biology remains unexplored. OBJECTIVE: We have investigated the role of IPMK in regulating angiogenesis. METHODS AND RESULTS: Deletion of IPMK in fibroblasts induces angiogenesis in both in vitro and in vivo models. IPMK deletion elicits a substantial increase of VEGF (vascular endothelial growth factor), which mediates the regulation of angiogenesis by IPMK. The regulation of VEGF by IPMK requires its catalytic activity. IPMK is predominantly nuclear and regulates gene transcription. However, IPMK does not apparently serve as a transcription factor for VEGF. HIF (hypoxia-inducible factor)-1α is a major determinant of angiogenesis and induces VEGF transcription. IPMK deletion elicits a major enrichment of HIF-1α protein and thus VEGF. HIF-1α is constitutively ubiquitinated by pVHL (von Hippel-Lindau protein) followed by proteasomal degradation under normal conditions. However, HIF-1α is not recognized and ubiquitinated by pVHL in IPMK KO (knockout) cells. IP5 reinstates the interaction of HIF-1α and pVHL. HIF-1α prolyl hydroxylation, which is prerequisite for pVHL recognition, is interrupted in IPMK-deleted cells. IP5 promotes HIF-1α prolyl hydroxylation and thus pVHL-dependent degradation of HIF-1α. Deletion of IPMK in mouse brain increases HIF-1α/VEGF levels and vascularization. The increased VEGF in IPMK KO disrupts blood-brain barrier and enhances brain blood vessel permeability. CONCLUSIONS: IPMK, via its product IP5, negatively regulates angiogenesis by inhibiting VEGF expression. IP5 acts by enhancing HIF-1α hydroxylation and thus pVHL-dependent degradation of HIF-1α.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosfatos de Inositol/metabolismo , Neovascularização Fisiológica/fisiologia , Fosfotransferases (Aceptor do Grupo Álcool)/fisiologia , Animais , Barreira Hematoencefálica , Células Cultivadas , Técnicas de Cocultura , Meios de Cultivo Condicionados/farmacologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Técnicas de Inativação de Genes , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Proteólise , RNA Interferente Pequeno/genética , Organismos Livres de Patógenos Específicos , Fator A de Crescimento do Endotélio Vascular/biossíntese , Fator A de Crescimento do Endotélio Vascular/genética , Proteína Supressora de Tumor Von Hippel-Lindau/metabolismo
6.
Proc Natl Acad Sci U S A ; 114(8): 2036-2041, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28154132

RESUMO

Inositol hexakisphosphate kinase 1 (IP6K1), which generates 5-diphosphoinositol pentakisphosphate (5-IP7), physiologically mediates numerous functions. We report that IP6K1 deletion leads to brain malformation and abnormalities of neuronal migration. IP6K1 physiologically associates with α-actinin and localizes to focal adhesions. IP6K1 deletion disrupts α-actinin's intracellular localization and function. The IP6K1 deleted cells display substantial decreases of stress fiber formation and impaired cell migration and spreading. Regulation of α-actinin by IP6K1 requires its kinase activity. Deletion of IP6K1 abolishes α-actinin tyrosine phosphorylation, which is known to be regulated by focal adhesion kinase (FAK). FAK phosphorylation is substantially decreased in IP6K1 deleted cells. 5-IP7, a product of IP6K1, promotes FAK autophosphorylation. Pharmacologic inhibition of IP6K by TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl)purine] recapitulates the phenotype of IP6K1 deletion. These findings establish that IP6K1 physiologically regulates neuronal migration by binding to α-actinin and influencing phosphorylation of both FAK and α-actinin through its product 5-IP7.


Assuntos
Actinina/metabolismo , Movimento Celular/fisiologia , Quinase 1 de Adesão Focal/metabolismo , Neurônios/fisiologia , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Animais , Encéfalo/anormalidades , Encéfalo/enzimologia , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Proteína-Tirosina Quinases de Adesão Focal , Humanos , Fosfatos de Inositol/metabolismo , Camundongos , Camundongos Knockout , Fosforilação , Fosfotransferases (Aceptor do Grupo Fosfato)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Interferência de RNA , RNA Interferente Pequeno/metabolismo
7.
J Neurosci ; 38(34): 7409-7419, 2018 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-30006360

RESUMO

Inositol hexakisphosphate kinases (IP6Ks) regulate various biological processes. Among pyrophosphates generated by IP6Ks, diphosphoinositol pentakisphosphate (IP7), and bis-diphosphoinositol tetrakisphosphate have been extensively characterized. IP7 is produced in mammals by a family of inositol hexakisphosphate kinases, IP6K1, IP6K2, and IP6K3, which have distinct biological functions. We report that IP6K2 binds protein 4.1.N with high affinity and specificity. Nuclear translocation of 4.1N, which is required for its principal functions, is dependent on IP6K2. Both of these proteins are highly expressed in granule cells of the cerebellum where their interaction regulates Purkinje cell morphology and cerebellar synapses. The deletion of IP6K2 in male/female mice elicits substantial defects in synaptic influences of granule cells upon Purkinje cells as well as notable impairment of locomotor function. Moreover, the disruption of IP6K2-4.1N interactions impairs cell viability. Thus, IP6K2 and its interaction with 4.1N appear to be major determinants of cerebellar disposition and psychomotor behavior.SIGNIFICANCE STATEMENT Inositol phosphates are produced by a family of inositol hexakisphosphate kinases (IP6Ks)-IP6K1, IP6K2, and IP6K3. Of these, the physiological roles of IP6K2 in the brain have been least characterized. In the present study, we report that IP6K2 binds selectively to the neuronal protein 4.1N. Both of these proteins are highly expressed in granule cells of the cerebellum. Using IP6K2 knock-out (KO) mice, we establish that IP6K2-4.1N interactions in granule cells regulate Purkinje cell morphology, the viability of cerebellar neurons, and psychomotor behavior.


Assuntos
Cerebelo/fisiologia , Proteínas do Citoesqueleto/fisiologia , Proteínas de Membrana/fisiologia , Atividade Motora/fisiologia , Proteínas do Tecido Nervoso/fisiologia , Neurônios/fisiologia , Neuropeptídeos/fisiologia , Fosfotransferases (Aceptor do Grupo Fosfato)/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular , Sobrevivência Celular , Cerebelo/citologia , Cerebelo/enzimologia , Comportamento Exploratório , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/deficiência , Neurônios/enzimologia , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Ligação Proteica , Desempenho Psicomotor/fisiologia , Células de Purkinje/enzimologia , Células de Purkinje/fisiologia , Teste de Desempenho do Rota-Rod , Sinapses/fisiologia
8.
Proc Natl Acad Sci U S A ; 112(6): 1773-8, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25617365

RESUMO

The inositol pyrophosphates, molecular messengers containing an energetic pyrophosphate bond, impact a wide range of biologic processes. They are generated primarily by a family of three inositol hexakisphosphate kinases (IP6Ks), the principal product of which is diphosphoinositol pentakisphosphate (IP7). We report that IP6K2, via IP7 synthesis, is a major mediator of cancer cell migration and tumor metastasis in cell culture and in intact mice. IP6K2 acts by enhancing cell-matrix adhesion and decreasing cell-cell adhesion. This action is mediated by IP7-elicited nuclear sequestration and inactivation of the tumor suppressor liver kinase B1 (LKB1). Accordingly, inhibitors of IP6K2 offer promise in cancer therapy.


Assuntos
Adesão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Fosfatos de Inositol/metabolismo , Metástase Neoplásica/fisiopatologia , Fosfotransferases (Aceptor do Grupo Fosfato)/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Quinases Ativadas por AMP , Animais , Western Blotting , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Matriz Extracelular/metabolismo , Humanos , Imunoprecipitação , Fosfatos de Inositol/biossíntese , Camundongos , Camundongos Nus , Microscopia de Fluorescência , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo
9.
J Neurosci ; 35(31): 11056-67, 2015 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-26245967

RESUMO

The inositol hexakisphosphate kinases (IP6Ks) are the principal enzymes that generate inositol pyrophosphates. There are three IP6Ks (IP6K1, 2, and 3). Functions of IP6K1 and IP6K2 have been substantially delineated, but little is known of IP6K3's role in normal physiology, especially in the brain. To elucidate functions of IP6K3, we generated mice with targeted deletion of IP6K3. We demonstrate that IP6K3 is highly concentrated in the brain in cerebellar Purkinje cells. IP6K3 physiologically binds to the cytoskeletal proteins adducin and spectrin, whose mutual interactions are perturbed in IP6K3-null mutants. Consequently, IP6K3 knock-out cerebella manifest abnormalities in Purkinje cell structure and synapse number, and the mutant mice display deficits in motor learning and coordination. Thus, IP6K3 is a major determinant of cytoskeletal disposition and function of cerebellar Purkinje cells. SIGNIFICANCE STATEMENT: We identified and cloned a family of three inositol hexakisphosphate kinases (IP6Ks) that generate the inositol pyrophosphates, most notably 5-diphosphoinositol pentakisphosphate (IP7). Of these, IP6K3 has been least characterized. In the present study we generated IP6K3 knock-out mice and show that IP6K3 is highly expressed in cerebellar Purkinje cells. IP6K3-deleted mice display defects of motor learning and coordination. IP6K3-null mice manifest aberrations of Purkinje cells with a diminished number of synapses. IP6K3 interacts with the cytoskeletal proteins spectrin and adducin whose altered disposition in IP6K3 knock-out mice may mediate phenotypic features of the mutant mice. These findings afford molecular/cytoskeletal mechanisms by which the inositol polyphosphate system impacts brain function.


Assuntos
Proteínas de Ligação a Calmodulina/metabolismo , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Células de Purkinje/metabolismo , Espectrina/metabolismo , Sinapses/metabolismo , Citoesqueleto de Actina/metabolismo , Animais , Forma Celular/fisiologia , Camundongos , Camundongos Knockout , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Células de Purkinje/citologia
10.
J Biol Chem ; 289(43): 29631-41, 2014 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-25164819

RESUMO

D-Serine, an endogenous co-agonist for the glycine site of the synaptic NMDA glutamate receptor, regulates synaptic plasticity and is implicated in schizophrenia. Serine racemase (SR) is the enzyme that converts L-serine to D-serine. In this study, we demonstrate that SR interacts with the synaptic proteins, postsynaptic density protein 95 (PSD-95) and stargazin, forming a ternary complex. SR binds to the PDZ3 domain of PSD-95 through the PDZ domain ligand at its C terminus. SR also binds to the C terminus of stargazin, which facilitates the cell membrane localization of SR and inhibits its activity. AMPA receptor activation internalizes SR and disrupts its interaction with stargazin, therefore derepressing SR activity, leading to more D-serine production and potentially facilitating NMDA receptor activation. These interactions regulate the enzymatic activity as well as the intracellular localization of SR, potentially coupling the activities of NMDA and AMPA receptors. This shuttling of a neurotransmitter synthesizing enzyme between two receptors appears to be a novel mode of synaptic regulation.


Assuntos
Canais de Cálcio/metabolismo , Ácido Glutâmico/metabolismo , Guanilato Quinases/metabolismo , Proteínas de Membrana/metabolismo , N-Metilaspartato/metabolismo , Racemases e Epimerases/metabolismo , Transmissão Sináptica , Ácido alfa-Amino-3-hidroxi-5-metil-4-isoxazol Propiônico/metabolismo , Animais , Biocatálise , Membrana Celular/metabolismo , Proteína 4 Homóloga a Disks-Large , Feminino , Células HEK293 , Humanos , Masculino , Camundongos , Modelos Biológicos , Ligação Proteica , Ratos , Receptores de AMPA/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo
11.
J Cardiovasc Transl Res ; 17(1): 197-215, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37615888

RESUMO

Water-soluble myo-inositol phosphates have long been characterized as second messengers. The signaling properties of these compounds are determined by the number and arrangement of phosphate groups on the myo-inositol backbone. Recently, higher inositol phosphates with pyrophosphate groups were recognized as signaling molecules. 5-Diphosphoinositol 1,2,3,4,6-pentakisphosphate (5PP-InsP5) is the most abundant isoform, constituting more than 90% of intracellular inositol pyrophosphates. 5PP-InsP5 can be further phosphorylated to 1,5-bisdiphosphoinositol 2,3,4,6-tetrakisphosphate (InsP8). These two molecules, 5PP-InsP5 and InsP8, are present in various subcellular compartments, where they participate in regulating diverse cellular processes such as cell death, energy homeostasis, and cytoskeletal dynamics. The synthesis and metabolism of inositol pyrophosphates are subjected to tight regulation, allowing for their highly specific functions. Blocking the 5PP-InsP5/InsP8 signaling pathway by inhibiting the biosynthesis of 5PP-InsP5 demonstrates therapeutic benefits in preclinical studies, and thus holds promise as a therapeutic approach for certain diseases treatment, such as metabolic disorders.


Assuntos
Difosfatos , Fosfatos de Inositol , Animais , Fosfatos de Inositol/metabolismo , Transdução de Sinais , Mamíferos/metabolismo
12.
Mol Brain ; 17(1): 8, 2024 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-38350944

RESUMO

Inositol pyrophosphates are key signaling molecules that regulate diverse neurobiological processes. We previously reported that the inositol pyrophosphate 5-InsP7, generated by inositol hexakisphosphate kinase 1 (IP6K1), governs the degradation of Na+/K+-ATPase (NKA) via an autoinhibitory domain of PI3K p85α. NKA is required for maintaining electrochemical gradients for proper neuronal firing. Here we characterized the electrophysiology of IP6K1 knockout (KO) neurons to further expand upon the functions of IP6K1-regulated control of NKA stability. We found that IP6K1 KO neurons have a lower frequency of action potentials and a specific deepening of the afterhyperpolarization phase. Our results demonstrate that deleting IP6K1 suppresses neuronal excitability, which is consistent with hyperpolarization due to an enrichment of NKA. Given that impaired NKA function contributes to the pathophysiology of various neurological diseases, including hyperexcitability in epilepsy, our findings may have therapeutic implications.


Assuntos
Inositol , ATPase Trocadora de Sódio-Potássio , Transdução de Sinais , Transporte Proteico , Neurônios/fisiologia
13.
Cardiovasc Res ; 120(8): 954-970, 2024 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-38252884

RESUMO

AIMS: Adiponectin is an adipocyte-derived circulating protein that exerts cardiovascular and metabolic protection. Due to the futile degradation of endogenous adiponectin and the challenges of exogenous administration, regulatory mechanisms of adiponectin biosynthesis are of significant pharmacological interest. METHODS AND RESULTS: Here, we report that 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7) generated by inositol hexakisphosphate kinase 1 (IP6K1) governed circulating adiponectin levels via thiol-mediated protein quality control in the secretory pathway. IP6K1 bound to adiponectin and DsbA-L and generated 5-InsP7 to stabilize adiponectin/ERp44 and DsbA-L/Ero1-Lα interactions, driving adiponectin intracellular degradation. Depleting 5-InsP7 by either IP6K1 deletion or pharmacological inhibition blocked intracellular adiponectin degradation. Whole-body and adipocyte-specific deletion of IP6K1 boosted plasma adiponectin levels, especially its high molecular weight forms, and activated AMPK-mediated protection against myocardial ischaemia-reperfusion injury. Pharmacological inhibition of 5-InsP7 biosynthesis in wild-type but not adiponectin knockout mice attenuated myocardial ischaemia-reperfusion injury. CONCLUSION: Our findings revealed that 5-InsP7 is a physiological regulator of adiponectin biosynthesis that is amenable to pharmacological intervention for cardioprotection.


Assuntos
Adiponectina , Camundongos Endogâmicos C57BL , Camundongos Knockout , Traumatismo por Reperfusão Miocárdica , Animais , Adiponectina/metabolismo , Adiponectina/genética , Adiponectina/sangue , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/genética , Traumatismo por Reperfusão Miocárdica/sangue , Traumatismo por Reperfusão Miocárdica/enzimologia , Fosfotransferases (Aceptor do Grupo Fosfato)/metabolismo , Fosfotransferases (Aceptor do Grupo Fosfato)/genética , Fosfatos de Inositol/metabolismo , Adipócitos/metabolismo , Adipócitos/enzimologia , Adipócitos/efeitos dos fármacos , Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Masculino , Camundongos , Modelos Animais de Doenças , Transdução de Sinais , Proteólise , Humanos
14.
Mol Pharmacol ; 83(5): 1099-108, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23470287

RESUMO

Inhibition of oxidative stress and inflammation in vascular endothelial cells (ECs) may represent a new therapeutic strategy against endothelial activation. Sinapic acid (SA), a phenylpropanoid compound, is found in natural herbs and high-bran cereals and has moderate antioxidant activity. We aimed to develop new SA agents with the properties of antioxidation and blocking EC activation for possible therapy of cardiovascular disease. We designed and synthesized 10 SA derivatives according to their chemical structures. Preliminary screening of the compounds involved scavenging hydroxyl radicals and 2,2-diphenyl-1-picrylhydrazyl (DPPH(⋅)), croton oil-induced ear edema in mice, and analysis of the mRNA expression of adhesion molecules in ECs. 1-Acetyl-sinapic acyl-4-(3'-chlorine-)benzylpiperazine (SA9) had the strongest antioxidant and anti-inflammatory activities both in vitro and in vivo. Thus, the effect of SA9 was further studied. SA9 inhibited tumor necrosis factor α-induced upregulation of adhesion molecules in ECs at both mRNA and protein levels, as well as the consequent monocyte adhesion to ECs. In vivo, result of face-to-face immunostaining showed that SA9 reduced lipopolysaccharide-induced expression of intercellular adhesion molecule-1 in mouse aortic intima. To study the molecular mechanism, results from luciferase assay, nuclear translocation of NF-κB, and Western blot indicated that the mechanism of the anti-inflammatory effects of SA9 might be suppression of intracellular generation of ROS and inhibition of NF-κB activation in ECs. SA9 is a prototype of a novel class of antioxidant with anti-inflammatory effects in ECs. It may represent a new therapeutic approach for preventing endothelial activation in cardiovascular disorders.


Assuntos
Ácidos Cumáricos/farmacologia , Células Endoteliais/efeitos dos fármacos , Endotélio Vascular/efeitos dos fármacos , Animais , Anti-Inflamatórios/síntese química , Anti-Inflamatórios/farmacologia , Antioxidantes/síntese química , Antioxidantes/farmacologia , Aorta/efeitos dos fármacos , Aorta/metabolismo , Compostos de Bifenilo/farmacologia , Adesão Celular/efeitos dos fármacos , Ácidos Cumáricos/síntese química , Óleo de Cróton/farmacologia , Orelha , Edema/induzido quimicamente , Edema/tratamento farmacológico , Edema/metabolismo , Células Endoteliais/metabolismo , Endotélio Vascular/metabolismo , Sequestradores de Radicais Livres/síntese química , Sequestradores de Radicais Livres/farmacologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Inflamação/induzido quimicamente , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Molécula 1 de Adesão Intercelular/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/efeitos dos fármacos , Monócitos/metabolismo , NF-kappa B/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Picratos/farmacologia , Fator de Necrose Tumoral alfa/metabolismo
15.
Biochem Biophys Res Commun ; 438(2): 364-9, 2013 Aug 23.
Artigo em Inglês | MEDLINE | ID: mdl-23892038

RESUMO

In cancer patients, the development of resistance to anti-angiogenic agents targeting the VEGF pathway is common. Increased pericyte coverage of the tumor vasculature undergoing VEGF targeted therapy has been suggested to play an important role in resistance. Therefore, reducing the pericytes coverage of the tumor vasculature has been suggested to be a therapeutic approach in breaking the resistance to and increasing the efficacy of anti-angiogenic therapies. To screen compound libraries, a simple in vitro assay of blood vessel maturation demonstrating endothelial cells and pericytes association while forming lumenized vascular structures is needed. Unfortunately, previously described 3-dimensional, matrix based assays are laborious and challenging from an image and data acquisition perspective. For these reasons they generally lack the scalability needed to perform in a high-throughput environment. With this work, we have developed a novel in vitro blood vessel maturation assay, in which lumenized, vascular structures form in one optical plane and mesenchymal progenitor cells (10T1/2) differentiate into pericyte-like cells, which associate with the endothelial vessels (HUVECs). The differentiation of the 10T1/2 cells into pericyte-like cells is visualized using a GFP reporter controlled by the alpha smooth muscle actin promoter (SMP-8). The organization of these vascular structures and their recruited mural cells in one optical plane allows for automated data capture and subsequent image analysis. The ability of this assay to screen for inhibitors of pericytes recruitment was validated. In summary, this novel assay of in vitro blood vessel maturation provides a valuable tool to screen for new agents with therapeutic potential.


Assuntos
Inibidores da Angiogênese/farmacologia , Vasos Sanguíneos/efeitos dos fármacos , Vasos Sanguíneos/patologia , Avaliação Pré-Clínica de Medicamentos/métodos , Actinas/metabolismo , Animais , Benzamidas/farmacologia , Linhagem Celular , Técnicas de Cocultura , Células Endoteliais/citologia , Fibroblastos/citologia , Proteínas de Fluorescência Verde/metabolismo , Células HEK293 , Células Endoteliais da Veia Umbilical Humana , Humanos , Mesilato de Imatinib , Indóis/farmacologia , Lentivirus/metabolismo , Camundongos , Músculo Liso/metabolismo , Neovascularização Fisiológica , Pericitos/citologia , Piperazinas/farmacologia , Regiões Promotoras Genéticas , Pirimidinas/farmacologia , Pirróis/farmacologia , Células-Tronco/citologia , Sunitinibe
16.
Circ Res ; 108(4): 410-7, 2011 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-21193741

RESUMO

RATIONALE: Endothelial cells (ECs) have distinct mechanotransduction mechanisms responding to laminar versus disturbed flow patterns. Endothelial dysfunction, affected by imposed flow, is one of the earliest events leading to atherogenesis. The involvement of γ/δ T lymphocytes in endothelial dysfunction under flow is largely unknown. OBJECTIVE: To investigate whether shear stress regulates membrane translocation of ATP synthase ß chain (ATPSß) in ECs, leading to the increased γ/δ T-lymphocyte adhesion and the related functions. METHOD AND RESULTS: We applied different flow patterns to cultured ECs. Laminar flow decreased the level of membrane-bound ATPSß (ecto-ATPSß) and depleted membrane cholesterol, whereas oscillatory flow increased the level of ecto-ATPSß and membrane cholesterol. Incubating ECs with cholesterol or depleting cellular cholesterol with ß-cyclodextrin mimicked the effect of oscillatory or laminar flow, respectively. Knockdown caveolin-1 by small interfering RNA prevented ATPSß translocation in response to laminar flow. Importantly, oscillatory flow or cholesterol treatment elevated the number of γ/δ T cells binding to ECs, which was blocked by anti-ATPSß antibody. Furthermore, the incubation of γ/δ T cells with ECs increased tumor necrosis fact α and interferon-γ secretion from T cells and vascular cell adhesion molecule-1 expression in ECs. In vivo, γ/δ T-cell adhesion and ATPSß membrane translocation was elevated in the aortic inner curvature and disturbed flow areas in partially ligated carotid arteries of ApoE(-/-) mice fed a high-fat diet. CONCLUSIONS: This study provides evidence that disturbed flow and hypercholesterolemia synergistically promote γ/δ T-lymphocyte activation by the membrane translocation of ATPSß in ECs and in vivo in mice, which is a novel mechanism of endothelial activation.


Assuntos
Comunicação Celular/fisiologia , Endotélio Vascular/citologia , ATPases Mitocondriais Próton-Translocadoras/fisiologia , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Estresse Mecânico , Linfócitos T/citologia , Linfócitos T/metabolismo , Adenosina Trifosfatases/fisiologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Transporte Biológico/fisiologia , Caveolina 1/fisiologia , Adesão Celular/fisiologia , Membrana Celular/fisiologia , Células Cultivadas , Colesterol/metabolismo , Endotélio Vascular/fisiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais
17.
Biomed Pharmacother ; 161: 114449, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36857911

RESUMO

The antifungal drug itraconazole has been repurposed to anti-angiogenic agent, but the mechanisms of action have been elusive. Here we report that itraconazole disrupts focal adhesion dynamics and cytoskeletal remodeling, which requires 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7). We find that inositol hexakisphosphate kinase 1 (IP6K1) binds Arp2 and generates 5-InsP7 to recruit coronin, a negative regulator of the Arp2/3 complex. IP6K1 also produces focal adhesion-enriched 5-InsP7, which binds focal adhesion kinase (FAK) at the FERM domain to promote its dimerization and phosphorylation. Itraconazole treatment elicits displacement of IP6K1/5-InsP7, thus augments 5-InsP7-mediated inhibition of Arp2/3 complex and reduces 5-InsP7-mediated FAK dimerization. Itraconazole-treated cells display reduced focal adhesion dynamics and actin cytoskeleton remodeling. Accordingly, itraconazole severely disrupts cell motility, an essential component of angiogenesis. These results demonstrate critical roles of IP6K1-generated 5-InsP7 in regulating focal adhesion dynamics and actin cytoskeleton remodeling and reveal functional mechanisms by which itraconazole inhibits cell motility.


Assuntos
Fosfatos de Inositol , Itraconazol , Itraconazol/farmacologia , Fosfatos de Inositol/metabolismo , Adesões Focais , Difosfatos/metabolismo , Movimento Celular , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Fosforilação , Células Endoteliais/metabolismo , Adesão Celular
18.
Microvasc Res ; 84(3): 270-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22835519

RESUMO

BACKGROUND: Endothelial progenitor cells (EPCs) participate in post-natal vasculogenesis. Maintaining the preliminary progenitor phenotype and good proliferation capacity of EPCs is key to their use in treating cardiovascular ischemic diseases. However, transcriptional regulation in EPCs remains largely unknown. We investigated the effect of leukemia inhibitory factor (LIF) combined with vascular endothelial growth factor (VEGF) on EPCs and the potential roles of Krüppel-like transcription factors (KLFs). METHODS AND RESULTS: Co-treatment with LIF and VEGF (100 ng/ml each) (V+L) could increase EPC colony-forming units and CD34 expression, which reflects the EPC progenitor phenotype and alleviated differentiation of EPCs. The effect was associated with Akt activation and increased expression of KLF4. Upregulation of KLF4 induced by V+L could be inhibited by transfection with dominant-negative Akt adenovirus. Furthermore, overexpression of KLF4 in EPCs enhanced the expression of CD34 and alleviated cell differentiation but did not increase the phosphorylation of Akt. CONCLUSIONS: LIF combined with VEGF can maintain the preliminary, progenitor phenotype of EPCs and alleviate cell differentiation by upregulating KLF4, which may provide new insights into transcriptional regulation in EPCs.


Assuntos
Células Endoteliais/citologia , Regulação da Expressão Gênica , Fatores de Transcrição Kruppel-Like/metabolismo , Fator Inibidor de Leucemia/metabolismo , Células-Tronco/citologia , Antígenos CD34/biossíntese , Diferenciação Celular , Proliferação de Células , Sangue Fetal/citologia , Citometria de Fluxo/métodos , Humanos , Fator 4 Semelhante a Kruppel , Microcirculação , Fenótipo , Fosforilação , Fator A de Crescimento do Endotélio Vascular/metabolismo
19.
Biochim Biophys Acta ; 1801(7): 702-10, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20382259

RESUMO

Caveolae and its structural protein caveolin-1 (Cav-1) are abundant in vascular endothelial cells (ECs). We examined whether caveolae are involved in monocyte adhesion to ECs responding to a synergy of hypercholesterolemia and inflammation. Treating human umbilical vein ECs with cholesterol enhanced endotoxin lipopolysaccharide (LPS)-induced monocyte adhesion. Use of isolated caveolae-enriched membranes revealed that cell adhesion molecules (CAMs), including intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1), co-localized with Cav-1 in caveolae. LPS upregulated CAMs expression and increased the co-localization. Cholesterol exposure decreased the level of CAMs in the caveolae. Co-immunoprecipitation and confocal microscopy revealed that ICAM-1 interacted with Cav-1. Electron microscopy showed that ICAM-1 was mainly located in caveolae. Cholesterol exposure decreased this interaction and drove ICAM-1 out of caveolae. Knockdown of Cav-1 reduced the synergistic effects of cholesterol and inflammation. In vivo, ICAM-1 and Cav-1 co-localization was lower in the aortic endothelium of ApoE(-)(/)(-) mice than in that of wild-type controls. Cav-1 negatively regulates monocyte adhesion by the co-localization of CAMs in caveolae, which is disturbed by cholesterol. Thus, our study suggests a molecular basis underlying the synergistic effects of hypercholesterolemia and inflammation in atherogenesis.


Assuntos
Cavéolas/metabolismo , Colesterol/metabolismo , Endotélio Vascular/metabolismo , Hipercolesterolemia/metabolismo , Monócitos/metabolismo , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Bovinos , Cavéolas/patologia , Caveolina 1/genética , Caveolina 1/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/genética , Endotélio Vascular/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Humanos , Hipercolesterolemia/genética , Hipercolesterolemia/patologia , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Molécula 1 de Adesão Intercelular/genética , Molécula 1 de Adesão Intercelular/metabolismo , Lipopolissacarídeos/farmacologia , Camundongos , Camundongos Knockout , Monócitos/patologia , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
20.
Sci Adv ; 6(44)2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-33115740

RESUMO

Sodium/potassium-transporting adenosine triphosphatase (Na+/K+-ATPase) is one of the most abundant cell membrane proteins and is essential for eukaryotes. Endogenous negative regulators have long been postulated to play an important role in regulating the activity and stability of Na+/K+-ATPase, but characterization of these regulators has been elusive. Mechanisms of regulating Na+/K+-ATPase homeostatic turnover are unknown. Here, we report that 5-diphosphoinositol 1,2,3,4,6-pentakisphosphate (5-InsP7), generated by inositol hexakisphosphate kinase 1 (IP6K1), promotes physiological endocytosis and downstream degradation of Na+/K+-ATPase-α1. Deletion of IP6K1 elicits a twofold enrichment of Na+/K+-ATPase-α1 in plasma membranes of multiple tissues and cell types. Using a suite of synthetic chemical biology tools, we found that 5-InsP7 binds the RhoGAP domain of phosphatidylinositol 3-kinase (PI3K) p85α to disinhibit its interaction with Na+/K+-ATPase-α1. This recruits adaptor protein 2 (AP2) and triggers the clathrin-mediated endocytosis of Na+/K+-ATPase-α1. Our study identifies 5-InsP7 as an endogenous negative regulator of Na+/K+-ATPase-α1.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA