Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
J Allergy Clin Immunol ; 141(2): 608-619.e7, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28807414

RESUMO

BACKGROUND: Chronic itch is a highly debilitating symptom that underlies many medical disorders with no universally effective treatments. Although unique neuronal signaling cascades in the sensory ganglia and spinal cord have been shown to critically promote the pathogenesis of chronic itch, the role of skin-associated cells remains poorly understood. OBJECTIVE: We sought to examine the cutaneous mechanisms underlying transient receptor potential vanilloid 4 (TRPV4)-mediated allergic and nonallergic chronic itch. METHODS: Expression of TRPV4 in chronic itch and healthy control skin preparations was examined by using real-time RT-PCR. Trpv4eGFP mice were used to study the expression and function of TRPV4 in the skin by means of immunofluorescence staining, flow cytometry, calcium imaging, and patch-clamp recordings. Genetic and pharmacologic approaches were used to examine the role and underlying mechanisms of TRPV4 in mouse models of dry skin-associated chronic itch and spontaneous scratching associated with squaric acid dibutylester-induced allergic contact dermatitis. RESULTS: TRPV4 is selectively expressed by dermal macrophages and epidermal keratinocytes in mice. Lineage-specific deletion of TRPV4 in macrophages and keratinocytes reduces allergic and nonallergic chronic itch in mice, respectively. Importantly, TRPV4 expression is significantly increased in skin biopsy specimens from patients with chronic idiopathic pruritus in comparison with skin from healthy control subjects. Moreover, TRPV4-dependent chronic itch requires 5-hydroxytryptamine (5-HT) signaling secondary to activation of distinct 5-HT receptors in mice with allergic and those with nonallergic chronic itch conditions. CONCLUSION: Our study reveals previously unrecognized mechanisms by which TRPV4-expressing epithelial and immune cells in the skin critically and dynamically mediate chronic itch and unravels novel targets for therapeutics in the setting of chronic itch.


Assuntos
Dermatite Alérgica de Contato/imunologia , Derme/imunologia , Regulação da Expressão Gênica/imunologia , Queratinócitos/imunologia , Macrófagos/imunologia , Prurido/imunologia , Canais de Cátion TRPV/imunologia , Animais , Doença Crônica , Dermatite Alérgica de Contato/genética , Dermatite Alérgica de Contato/patologia , Derme/patologia , Feminino , Regulação da Expressão Gênica/genética , Humanos , Queratinócitos/patologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout , Prurido/genética , Prurido/patologia , Canais de Cátion TRPV/genética
2.
Am J Physiol Renal Physiol ; 315(6): F1626-F1636, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30207167

RESUMO

Transient receptor potential cation channel subfamily V member 4 (TRPV4)-mediated Ca2+ signaling induces early activation of small/intermediate Ca2+-activated K+ channels, SK3 (KCNN3) and IK1 (KCNN4), which leads to membrane hyperpolarization and enhanced Ca2+ influx, which is critical for subsequent activation of the large conductance Ca2+-activated K+ channel BK (KCNMA1) and K+ secretion in kidney cortical collecting duct (CCD) cells. The focus of the present study was to determine if such coordinated hierarchical/sequential activation of these channels in CCD was orchestrated within caveolae, a known microcompartment underlying selective Ca2+-signaling events in other cells. In K+-secreting mouse principal cell (PC) line, mCCDcl1 cells, knockdown of caveolae caveolin-1 (CAV-1) depressed TRPV4-mediated Ca2+ signaling and activation of SK3, intermediate conductance channel (IK1), and BK. Immunofluorescence colocalization analysis and coimmunoprecipitation assays demonstrated direct coupling of TRPV4 with each of the KCa channels in both mCCDcl1 and whole mouse kidney homogenates. Likewise, extending this analysis to CAV-1 demonstrates colocalization and direct coupling of CAV-1 with TRPV4, SK3, IK1, and BK, providing strong support for coupling of the channels in caveolae microdomains. Furthermore, differential expression of CAV-1 along the CCD was apparent where CAV-1 was strongly expressed within and along the cell borders of kidney PCs and intercalated cells (ICs), although significantly less in ICs. It is concluded that caveolae provide a key microdomain in PCs and ICs for coupling of TRPV4 with SK3, IK1, and BK that directly contributes to TRPV4-mediated Ca2+ signaling in these domains leading to rapid and sequential coupling of TRPV4-SK3/IK1-BK that may play a central role in mediating Ca2+-dependent regulation of BK and K+ secretion.


Assuntos
Sinalização do Cálcio , Cavéolas/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Túbulos Renais Coletores/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Caveolina 1/genética , Caveolina 1/metabolismo , Linhagem Celular , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/genética , Túbulos Renais Coletores/citologia , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/genética , Potenciais da Membrana , Camundongos Endogâmicos C57BL , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética , Canais de Cátion TRPV/genética
3.
Am J Physiol Renal Physiol ; 312(6): F1081-F1089, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28274924

RESUMO

The large-conductance Ca2+-activated K+ channel, BK (KCNMA1), is expressed along the connecting tubule (CNT) and cortical collecting duct (CCD) where it underlies flow- and Ca2+-dependent K+ secretion. Its activity is partially under the control of the mechanosensitive transient receptor potential vanilloid type 4 (TRPV4) Ca2+-permeable channel. Recently, we identified three small-/intermediate-conductance Ca2+-activated K+ channels, SK1 (KCNN1), SK3 (KCNN3), and IK1 (KCNN4), with notably high Ca2+-binding affinities, that are expressed in CNT/CCD and may be regulated by TRPV4-mediated Ca2+ influx. The K+-secreting CCD mCCDcl1 cells, which express these channels, were used to determine whether SK1/3 and IK1 are activated on TRPV4 stimulation and whether they contribute to Ca2+ influx and activation of BK. Activation of TRPV4 (GSK1016790A) modestly depolarized the membrane potential and robustly increased intracellular Ca2+, [Ca2+]i Inhibition of both SK1/3 and IK1 by application of apamin and 1-[(2-chlorophenyl)diphenylmethyl]-1H-pyrazole (TRAM-34), respectively, further depolarized the membrane potential and markedly suppressed the TRPV4-mediated rise in [Ca2+]i Application of BK inhibitor iberiotoxin after activation of TRPV4 without apamin/TRAM-34 also reduced [Ca2+]i and further intensified membrane depolarization, demonstrating BK involvement. However, the BK-dependent effects on [Ca2+]i and membrane potential were largely abolished by pretreatment with apamin and TRAM-34, identical to that observed by separately suppressing TRPV4-mediated Ca2+ influx, demonstrating that SK1/3-IK1 channels potently contribute to TRPV4-mediated BK activation. Our data indicate a direct correlation between TRPV4-mediated Ca2+ signal and BK activation but where early activation of SK1/3 and IK1 channels are critical to sufficiently enhanced Ca2+ entry and [Ca2+]i levels required for activation of BK.


Assuntos
Cálcio/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/metabolismo , Túbulos Renais Coletores/metabolismo , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Potássio/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Agonistas dos Canais de Cálcio/farmacologia , Sinalização do Cálcio , Células Cultivadas , Canais de Potássio Ativados por Cálcio de Condutância Intermediária/antagonistas & inibidores , Túbulos Renais Coletores/efeitos dos fármacos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/antagonistas & inibidores , Potenciais da Membrana , Camundongos , Bloqueadores dos Canais de Potássio/farmacologia , Canais de Potássio Ativados por Cálcio de Condutância Baixa/antagonistas & inibidores , Canais de Cátion TRPV/agonistas
4.
Kidney Int ; 91(6): 1398-1409, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28187982

RESUMO

To maintain potassium homeostasis, kidneys exert flow-dependent potassium secretion to facilitate kaliuresis in response to elevated dietary potassium intake. This process involves stimulation of calcium-activated large conductance maxi-K (BK) channels in the distal nephron, namely the connecting tubule and the collecting duct. Recent evidence suggests that the TRPV4 channel is a critical determinant of flow-dependent intracellular calcium elevations in these segments of the renal tubule. Here, we demonstrate that elevated dietary potassium intake (five percent potassium) increases renal TRPV4 mRNA and protein levels in an aldosterone-dependent manner and causes redistribution of the channel to the apical plasma membrane in native collecting duct cells. This, in turn, leads to augmented TRPV4-mediated flow-dependent calcium ion responses in freshly isolated split-opened collecting ducts from mice fed the high potassium diet. Genetic TRPV4 ablation greatly diminished BK channel activity in collecting duct cells pointing to a reduced capacity to excrete potassium. Consistently, elevated potassium intake induced hyperkalemia in TRPV4 knockout mice due to deficient renal potassium excretion. Thus, regulation of TRPV4 activity in the distal nephron by dietary potassium is an indispensable component of whole body potassium balance.


Assuntos
Hiperpotassemia/metabolismo , Túbulos Renais/metabolismo , Potássio na Dieta/metabolismo , Eliminação Renal , Canais de Cátion TRPV/metabolismo , Adaptação Fisiológica , Animais , Cálcio/metabolismo , Predisposição Genética para Doença , Homeostase , Hiperpotassemia/genética , Hiperpotassemia/fisiopatologia , Túbulos Renais/fisiopatologia , Canais de Potássio Ativados por Cálcio de Condutância Alta/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Potássio na Dieta/administração & dosagem , Receptores de Mineralocorticoides/metabolismo , Canais de Cátion TRPV/deficiência , Canais de Cátion TRPV/genética
5.
J Biol Chem ; 288(28): 20306-14, 2013 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-23709216

RESUMO

We have recently documented that the Ca(2+)-permeable TRPV4 channel, which is abundantly expressed in distal nephron cells, mediates cellular Ca(2+) responses to elevated luminal flow. In this study, we combined Fura-2-based [Ca(2+)]i imaging with immunofluorescence microscopy in isolated split-opened distal nephrons of C57BL/6 mice to probe the molecular determinants of TRPV4 activity and subcellular distribution. We found that activation of the PKC pathway with phorbol 12-myristate 13-acetate significantly increased [Ca(2+)]i responses to flow without affecting the subcellular distribution of TRPV4. Inhibition of PKC with bisindolylmaleimide I diminished cellular responses to elevated flow. In contrast, activation of the PKA pathway with forskolin did not affect TRPV4-mediated [Ca(2+)]i responses to flow but markedly shifted the subcellular distribution of the channel toward the apical membrane. These actions were blocked with the specific PKA inhibitor H-89. Concomitant activation of the PKA and PKC cascades additively enhanced the amplitude of flow-induced [Ca(2+)]i responses and greatly increased basal [Ca(2+)]i levels, indicating constitutive TRPV4 activation. This effect was precluded by the selective TRPV4 antagonist HC-067047. Therefore, the functional status of the TRPV4 channel in the distal nephron is regulated by two distinct signaling pathways. Although the PKA-dependent cascade promotes TRPV4 trafficking and translocation to the apical membrane, the PKC-dependent pathway increases the activity of the channel on the plasma membrane.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Néfrons/metabolismo , Proteína Quinase C/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Colforsina/farmacologia , Proteínas Quinases Dependentes de AMP Cíclico/antagonistas & inibidores , Ativação Enzimática/efeitos dos fármacos , Fura-2/química , Fura-2/metabolismo , Técnicas In Vitro , Indóis/farmacologia , Isoquinolinas/farmacologia , Túbulos Renais Coletores/metabolismo , Maleimidas/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência , Morfolinas/farmacologia , Néfrons/efeitos dos fármacos , Perfusão/métodos , Ésteres de Forbol/farmacologia , Proteína Quinase C/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Transporte Proteico/efeitos dos fármacos , Pirróis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Sulfonamidas/farmacologia , Canais de Cátion TRPV/antagonistas & inibidores
6.
J Am Soc Nephrol ; 24(4): 604-16, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23411787

RESUMO

The molecular mechanism of cyst formation and expansion in autosomal recessive polycystic kidney disease (ARPKD) is poorly understood, but impaired mechanosensitivity to tubular flow and dysfunctional calcium signaling are important contributors. The activity of the mechanosensitive Ca(2+)-permeable TRPV4 channel underlies flow-dependent Ca(2+) signaling in murine collecting duct (CD) cells, suggesting that this channel may contribute to cystogenesis in ARPKD. Here, we developed a method to isolate CD-derived cysts and studied TRPV4 function in these cysts laid open as monolayers and in nondilated split-open CDs in a rat model of ARPKD. In freshly isolated CD-derived cyst monolayers, we observed markedly impaired TRPV4 activity, abnormal subcellular localization of the channel, disrupted TRPV4 glycosylation, decreased basal [Ca(2+)]i, and loss of flow-mediated [Ca(2+)]i signaling. In contrast, nondilated CDs of these rats exhibited functional TRPV4 with largely preserved mechanosensitive properties. Long-term systemic augmentation of TRPV4 activity with a selective TRPV4 activator significantly attenuated the renal manifestations of ARPKD in a time-dependent manner. At the cellular level, selective activation of TRPV4 restored mechanosensitive Ca(2+) signaling as well as the function and subcellular distribution of TRPV4. In conclusion, the functional status of TRPV4, which underlies mechanosensitive Ca(2+) signaling in CD cells, inversely correlates with renal cystogenesis in ARPKD. Augmenting TRPV4 activity may have therapeutic potential in ARPKD.


Assuntos
Cálcio/metabolismo , Cistos/metabolismo , Túbulos Renais Coletores/patologia , Rim Policístico Autossômico Recessivo/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cistos/fisiopatologia , Túbulos Renais Coletores/metabolismo , Túbulos Renais Coletores/fisiopatologia , Rim Policístico Autossômico Recessivo/fisiopatologia , Ratos , Ratos Sprague-Dawley , Transdução de Sinais
7.
J Biol Chem ; 287(12): 8782-91, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22298783

RESUMO

The TRPV4 Ca(2+)-permeable channel is sensitive to mechanical stimuli. In the current study we have employed immunocytochemical staining in kidney slices and functional assessments (Ca(2+) imaging) in isolated, split-opened, tubule segments to define TRPV4 sites of expression and flow-dependent function in the collecting duct system. Staining patterns revealed strong expression of TRPV4 along the entire collecting duct system with highest levels at the apical (luminal)/subapical region of the principal cells (PCs), the dominant cell type, with more diffuse staining in intercalated cells (ICs). Using fluorescence Ca(2+) imaging and the selective TRPV4 agonist, GSK1016790A, we demonstrated functional TRPV4 channels in PCs and ICs of split-opened cortical collecting ducts and connecting tubules. The agonist was ineffective in inducing a rise in [Ca(2+)](i) in the absence of extracellular Ca(2+) or in tubules from TRPV4-deficient animals. Most importantly, a 10-fold elevation in luminal (apical) fluid flow induced a rapid and sustained influx of Ca(2+) that was abolished by the TRPV channel inhibitor, ruthenium red, or in tubules isolated from TRPV4 deficient animals. We concluded that TRPV4 is highly expressed along the entire collecting duct system where it appears to function as a sensor/transducer of flow-induce mechanical stresses.


Assuntos
Túbulos Renais/metabolismo , Mecanotransdução Celular , Canais de Cátion TRPV/metabolismo , Animais , Camundongos , Estresse Mecânico , Canais de Cátion TRPV/genética
8.
Biochim Biophys Acta ; 1821(7): 994-1002, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22521764

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) are one of the most widely consumed pharmaceuticals, yet both the mechanisms involved in their therapeutic actions and side-effects, notably gastrointestinal (GI) ulceration/bleeding, have not been clearly defined. In this study, we have used a number of biochemical, structural, computational and biological systems including; Fourier Transform InfraRed (FTIR). Nuclear Magnetic Resonance (NMR) and Surface Plasmon Resonance (SPR) spectroscopy, and cell culture using a specific fluorescent membrane probe, to demonstrate that NSAIDs have a strong affinity to form ionic and hydrophobic associations with zwitterionic phospholipids, and specifically phosphatidylcholine (PC), that are reversible and non-covalent in nature. We propose that the pH-dependent partition of these potent anti-inflammatory drugs into the phospholipid bilayer, and possibly extracellular mono/multilayers present on the luminal interface of the mucus gel layer, may result in profound changes in the hydrophobicity, fluidity, permeability, biomechanical properties and stability of these membranes and barriers. These changes may not only provide an explanation of how NSAIDs induce surface injury to the GI mucosa as a component in the pathogenic mechanism leading to peptic ulceration and bleeding, but potentially an explanation for a number of (COX-independent) biological actions of this family of pharmaceuticals. This insight also has proven useful in the design and development of a novel class of PC-associated NSAIDs that have reduced GI toxicity while maintaining their essential therapeutic efficacy to inhibit pain and inflammation.


Assuntos
Anti-Inflamatórios não Esteroides/química , Aspirina/química , Mucosa Gástrica/efeitos dos fármacos , Ibuprofeno/química , Naproxeno/química , Fosfatidilcolinas/química , Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Corantes Fluorescentes , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Humanos , Concentração de Íons de Hidrogênio , Interações Hidrofóbicas e Hidrofílicas , Ibuprofeno/farmacologia , Bicamadas Lipídicas/química , Espectroscopia de Ressonância Magnética , Simulação de Dinâmica Molecular , Naproxeno/farmacologia , Compostos de Piridínio , Espectroscopia de Infravermelho com Transformada de Fourier , Ressonância de Plasmônio de Superfície
9.
Pflugers Arch ; 465(2): 177-86, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23207579

RESUMO

Kidneys are complex highly organized paired organs of nearly one million nephrons each. They rigorously process about 180 l of plasma daily to keep whole body homeostasis. To effectively perform such a titanic work, kidneys rely on mechanisms able to sense dynamic changes in composition and flow rates of protourine along the renal tubule. It is envisioned that Ca(2+)-permeable transient receptor potential (TRP) channels, and specifically mechanosensitive TRPV4, can serve to interpret these external mechanical cues in the form of elevated intracellular Ca(2+) concentration. This, in turn, initiates multiple cellular responses and adaptation mechanisms. The current review summarizes up-to-date knowledge about the sites of TRPV4 expression in renal tissue as well as discusses the functional role of the channel in cellular responses to hypotonicity and tubular flow. We will also provide insights as to how TRPV4 fits into classical polycystin mechanosensory complex in cilia and will speculate about previously underappreciated clinical implication of pharmacological TRPV4 targeting in treatment of polycystic kidney disease.


Assuntos
Rim/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Cálcio/metabolismo , Expressão Gênica , Humanos , Rim/fisiologia , Doenças Renais Policísticas/metabolismo , Canais de Cátion TRPV/genética
10.
Am J Physiol Renal Physiol ; 300(5): F1105-15, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21325499

RESUMO

Activation of the renal kallikrein-kinin system results in natriuresis and diuresis, suggesting its possible role in renal tubular sodium transport regulation. Here, we used patch-clamp electrophysiology to directly assess the effects of bradykinin (BK) on the epithelial Na(+) channel (ENaC) activity in freshly isolated split-opened murine aldosterone-sensitive distal nephrons (ASDNs). BK acutely inhibits ENaC activity by reducing channel open probability (P(o)) in a dose-dependent and reversible manner. Inhibition of B2 receptors with icatibant (HOE-140) abolished BK actions on ENaC. In contrast, activation of B1 receptors with the selective agonist Lys-des-Arg(9)-BK failed to reproduce BK actions on ENaC. This is consistent with B2 receptors playing a critical role in mediating BK signaling to ENaC. BK has little effect on ENaC P(o) when G(q/11) was inhibited with Gp antagonist 2A. Moreover, inhibition of phospholipase C (PLC) with U73122, but not saturation of cellular cAMP levels with the membrane-permeable nonhydrolysable cAMP analog 8-cpt-cAMP, prevents BK actions on ENaC activity. This argues that BK stimulates B2 receptors with subsequent activation of G(q/11)-PLC signaling cascade to acutely inhibit ENaC activity. Activation of BK signaling acutely depletes apical PI(4,5)P(2) levels. However, inhibition of Ca(2+) pump SERCA of the endoplasmic reticulum with thapsigargin does not prevent BK signaling to ENaC. Furthermore, caffeine, while producing a similar rise in [Ca(2+)](i) as in response to BK stimulation, fails to recapitulate BK actions on ENaC. Therefore, we concluded that BK acutely inhibits ENaC P(o) in mammalian ASDN via stimulation of B2 receptors and following depletion of PI(4,5)P(2), but not increases in [Ca(2+)](i).


Assuntos
Aldosterona/metabolismo , Bradicinina/farmacologia , Bloqueadores do Canal de Sódio Epitelial , Ativação do Canal Iônico/efeitos dos fármacos , Natriurese/efeitos dos fármacos , Néfrons/efeitos dos fármacos , Bloqueadores dos Canais de Sódio/farmacologia , Sódio/metabolismo , Absorção , Animais , Técnicas Biossensoriais , Cafeína/farmacologia , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacologia , Relação Dose-Resposta a Droga , Canais Epiteliais de Sódio/metabolismo , Estrenos/farmacologia , Subunidades alfa Gq-G11 de Proteínas de Ligação ao GTP/metabolismo , Hidrólise , Calidina/análogos & derivados , Calidina/farmacologia , Masculino , Potenciais da Membrana , Camundongos , Camundongos Endogâmicos C57BL , Néfrons/metabolismo , Técnicas de Patch-Clamp , Fosfatidilinositol 4,5-Difosfato/metabolismo , Inibidores de Fosfodiesterase/farmacologia , Pirrolidinonas/farmacologia , Receptor B1 da Bradicinina/agonistas , Receptor B1 da Bradicinina/metabolismo , Receptor B2 da Bradicinina/agonistas , Receptor B2 da Bradicinina/metabolismo , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/antagonistas & inibidores , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tapsigargina/farmacologia , Tionucleotídeos/farmacologia , Técnicas de Cultura de Tecidos , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo
11.
Am J Physiol Cell Physiol ; 298(6): C1583-93, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20164382

RESUMO

Hypoxia-induced disruption of the blood-brain barrier (BBB) is the result of many different mechanisms, including alterations to the cytoskeleton. In this study, we identified actin-binding proteins involved in cytoskeletal dynamics with quantitative proteomics and assessed changes in subcellular localization of two proteins involved in actin polymerization [vasodilator-stimulated phosphoprotein (VASP)] and cytoskeleton-plasma membrane cross-linking (moesin). We found significant redistribution of both VASP and moesin to the cytoskeletal and membrane fractions of BBB endothelial cells after 1-h hypoxic stress. We also investigated activation of actin-myosin contraction through assessment of phosphorylated myosin light chain (pMLC) with confocal microscopy. Hypoxia caused a rapid and transient increase in pMLC. Blocking MLC phosphorylation through inhibition of myosin light chain kinase (MLCK) with ML-7 prevented hypoxia-induced BBB disruption and relocalization of the tight junction protein ZO-1. Finally, we implicate the transient receptor potential (TRP)C family of channels in mediating these events since blockade of TRPC channels and the associated calcium influx with SKF-96365 prevents hypoxia-induced permeability changes and the phosphorylation of MLC needed for actin-myosin contraction. These data suggest that hypoxic stress triggers alterations to cytoskeletal structure that contribute to BBB disruption and that calcium influx through TRPC channels contributes to these events.


Assuntos
Actinas/metabolismo , Barreira Hematoencefálica/metabolismo , Forma Celular , Células Endoteliais/metabolismo , Miosinas/metabolismo , Estresse Fisiológico , Canais de Cátion TRPC/metabolismo , Animais , Azepinas/farmacologia , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/patologia , Cálcio/metabolismo , Bloqueadores dos Canais de Cálcio/farmacologia , Moléculas de Adesão Celular/metabolismo , Hipóxia Celular , Linhagem Celular , Forma Celular/efeitos dos fármacos , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/patologia , Inibidores Enzimáticos/farmacologia , Imidazóis/farmacologia , Camundongos , Proteínas dos Microfilamentos/metabolismo , Cadeias Leves de Miosina/metabolismo , Quinase de Cadeia Leve de Miosina/antagonistas & inibidores , Quinase de Cadeia Leve de Miosina/metabolismo , Naftalenos/farmacologia , Permeabilidade , Fosfoproteínas/metabolismo , Fosforilação , Transporte Proteico , Proteômica/métodos , Junções Íntimas/metabolismo , Fatores de Tempo
12.
J Nucl Med ; 49(4): 517-23, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18344436

RESUMO

UNLABELLED: PET molecular imaging of 15O-labeled water is the gold standard for measuring blood flow in humans. However, this requires an on-site cyclotron to produce the short-lived 15O tracer, which is cost-prohibitive for most clinical PET centers. The purpose of this study was to determine if the early uptake of 18F-FDG could be used to measure regional blood flow in tumors in the absence of 15O-water. METHODS: PET scans were obtained in patients being evaluated for tumor perfusion and glucose metabolism in a phase I dose-escalating protocol for endostatin, a novel antiangiogenic agent. A 2-min perfusion scan was performed with a bolus injection of 2,220 MBq (60 mCi) of 15O-water, which was followed by a 370-MBq (10 mCi) dose of 18F-FDG. Four sequential scans of 18F-FDG uptake were acquired, consisting of an early 2-min uptake scan-or first-pass scan-and 3 sequential 15-min late 18F-FDG uptake scans. Regions of interest (ROIs) were drawn on 2 or more tumor sites and on back muscle, as a control ROI, for each patient. Arterial blood concentration was derived from the PET scans by drawing an ROI over a large artery in the field of view. Blood flow was computed with a simple 1-compartment blood flow model using the first 2 min of data after injection. RESULTS: Blood flow estimated from the early uptake of 18F-FDG was linearly correlated with 15O-measured blood flow, with an intercept of 0.01, a slope of 0.86, and an R2 regression coefficient of 0.74 (r = 0.86). The 18F-FDG tumor extraction fraction relative to 15O-water averaged 0.86. A preliminary case study of a patient with prostate cancer confirms the utility of the first-pass 18F-FDG blood flow analysis in tumor diagnosis. CONCLUSION: These results suggest that the first-pass uptake of 18F-FDG may provide an estimate of perfusion in a tumor within the limitations of incomplete extraction of 18F-FDG compared with 15O-water.


Assuntos
Fluordesoxiglucose F18 , Radioisótopos de Oxigênio , Tomografia por Emissão de Pósitrons/métodos , Neoplasias da Próstata/irrigação sanguínea , Humanos , Masculino , Fluxo Sanguíneo Regional
13.
Brain Res ; 1130(1): 17-30, 2007 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-17169347

RESUMO

Understanding the molecular and biochemical mechanisms regulating the blood-brain barrier is aided by in vitro model systems. Many studies have used primary cultures of brain microvessel endothelial cells for this purpose. However, primary cultures limit the generation of material for molecular and biochemical assays since cells grow slowly, are prone to contamination by other neurovascular unit cells, and lose blood-brain barrier characteristics when passaged. To address these issues, immortalized cell lines have been generated. In these studies, we assessed the suitability of the immortalized mouse brain endothelial cell line, bEnd3, as a blood-brain barrier model. RT-PCR and immunofluorescence indicated expression of multiple tight junction proteins. bEnd3 cells formed barriers to radiolabeled sucrose, and responded like primary cultures to disrupting stimuli. Exposing cells to serum-free media on their basolateral side significantly decreased paracellular permeability; astrocyte-conditioned media did not enhance barrier properties. The serum-free media-induced decrease in permeability was correlated with an increase in claudin-5 and zonula occludens-1 immunofluorescence at cell-cell contracts. We conclude that bEnd3 cells are an attractive candidate as a model of the blood-brain barrier due to their rapid growth, maintenance of blood-brain barrier characteristics over repeated passages, formation of functional barriers and amenability to numerous molecular interventions.


Assuntos
Barreira Hematoencefálica/fisiologia , Permeabilidade Capilar/fisiologia , Células Endoteliais/metabolismo , Proteínas de Membrana/metabolismo , Junções Íntimas/metabolismo , Animais , Barreira Hematoencefálica/metabolismo , Encéfalo/irrigação sanguínea , Encéfalo/metabolismo , Capilares/citologia , Capilares/fisiologia , Capilares/ultraestrutura , Técnicas de Cultura de Células , Linhagem Celular , Claudina-5 , Células Endoteliais/citologia , Células Endoteliais/ultraestrutura , Proteínas de Membrana/genética , Camundongos , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , RNA Mensageiro/análise , Junções Íntimas/ultraestrutura , Proteína da Zônula de Oclusão-1
15.
PLoS One ; 11(5): e0155006, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27159616

RESUMO

The voltage- and Ca2+-activated, large conductance K+ channel (BK, maxi-K) is expressed in the collecting duct system of kidney where it underlies flow- and Ca2+-dependent K+ excretion. To determine if other Ca2+-activated K+ channels (KCa) may participate in this process, mouse kidney and the K+-secreting mouse cortical collecting duct (CCD) cell line, mCCDcl1, were assessed for TRPV4 and KCa channel expression and cross-talk. qPCR mRNA analysis and immunocytochemical staining demonstrated TRPV4 and KCa expression in mCCDcl1 cells and kidney connecting tubule (CNT) and CCD. Three subfamilies of KCa channels were revealed: the high Ca2+-binding affinity small-conductance SK channels, SK1and SK3, the intermediate conductance channel, IK1, and the low Ca2+-binding affinity, BK channel (BKα subunit). Apparent expression levels varied in CNT/CCD where analysis of CCD principal cells (PC) and intercalated cells (IC) demonstrated differential staining: SK1:PCIC, IK1:PC>IC, BKα:PC = IC, and TRPV4:PC>IC. Patch clamp analysis and fluorescence Ca2+ imaging of mCCDcl1 cells demonstrated potent TRPV4-mediated Ca2+ entry and strong functional cross-talk between TRPV4 and KCa channels. TRPV4-mediated Ca2+ influx activated each KCa channel, as evidenced by selective inhibition of KCa channels, with each active KCa channel enhancing Ca2+ entry (due to membrane hyperpolarization). Transepithelial electrical resistance (TEER) analysis of confluent mCCDcl1 cells grown on permeable supports further demonstrated this cross-talk where TRPV4 activation induce a decrease in TEER which was partially restored upon selective inhibition of each KCa channel. It is concluded that SK1/SK3 and IK1 are highly expressed along with BKα in CNT and CCD and are closely coupled to TRPV4 activation as observed in mCCDcl1 cells. The data support a model in CNT/CCD segments where strong cross talk between TRPV4-mediated Ca2+ influx and each KCa channel leads to enhance Ca2+ entry which will support activation of the low Ca2+-binding affinity BK channel to promote BK-mediated K+ secretion.


Assuntos
Túbulos Renais Coletores/metabolismo , Canais de Potássio Cálcio-Ativados/metabolismo , Canais de Cátion TRPV/metabolismo , Animais , Linhagem Celular , Camundongos , Camundongos Endogâmicos C57BL
16.
Mol Imaging Biol ; 7(6): 388-92, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-16284704

RESUMO

PURPOSE: A new fluorescent analog of D -glucose was recently developed by [Yoshioka K, Takahashi H, Homma T, Sato M, Ki Bong O, Nemoto Y, Matsuoka H (1996) A novel fluorescent derivative of glucose applicable to the assessment of glucose uptake activity of Escherichia coli. Biochim Biophys Acta 1289:5-9] and shown to be transported into normal cells. The purpose of this preliminary study was to assess the use of this fluorescent 2-deoxyglucose analog, 2-[N-(7-nitrobenz-2-oxa-1,3-diaxol-4-yl)amino]-2-deoxyglucose (2-NBDG), as a sensitive probe for monitoring glucose uptake into malignant tumor cells. PROCEDURES: MCF-7 breast cancer epithelial cells were grown and plated on coverslips for analysis of 2-NBDG uptake via fluorescence imaging microscopy. RESULTS: Steady-state fluorescence analysis of 2-NBDG uptake displayed rapid uptake for the first one to five minutes, then slowed, reaching an apparent maximum uptake near 20-30 minutes. Addition of 5 mM D -glucose to the media markedly reduced 2-NBDG uptake. Uptake of 2-NBDG in nonmalignant epithelial cells (M-1 epithelial cells) was slow, averaging less than 20% of that observed for tumorigenic cells, the MCF-7 breast cancer cells and the HepG2 liver cancer cell line. CONCLUSIONS: The preliminary data clearly demonstrate a rapid uptake of 2-NBDG into tumor cells that can be monitored by fluorescence imaging analysis. The uptake displays saturation and competition with D -glucose, all properties expected for 2-NBDG uptake and retention in cancer cells. Additional studies, including comparisons among other malignant cell lines and control cells, will be needed to fully characterize the kinetic properties of 2-NBDG uptake and the potential use of this 2-DG analog as a probe for glucose uptake in malignant cells.


Assuntos
4-Cloro-7-nitrobenzofurazano/análogos & derivados , Desoxiglucose/análogos & derivados , Neoplasias/metabolismo , 4-Cloro-7-nitrobenzofurazano/metabolismo , 4-Cloro-7-nitrobenzofurazano/farmacocinética , Transporte Biológico , Linhagem Celular , Linhagem Celular Tumoral , Desoxiglucose/metabolismo , Desoxiglucose/farmacocinética , Humanos
17.
PLoS One ; 9(4): e95149, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24762817

RESUMO

The Ca2+-activated, maxi-K (BK) K+ channel, with low Ca2+-binding affinity, is expressed in the distal tubule of the nephron and contributes to flow-dependent K+ secretion. In the present study we demonstrate that the Ca2+-activated, SK3 (KCa2.3) K+ channel, with high Ca2+-binding affinity, is also expressed in the mouse kidney (RT-PCR, immunoblots). Immunohistochemical evaluations using tubule specific markers demonstrate significant expression of SK3 in the distal tubule and the entire collecting duct system, including the connecting tubule (CNT) and cortical collecting duct (CCD). In CNT and CCD, main sites for K+ secretion, the highest levels of expression were along the apical (luminal) cell membranes, including for both principal cells (PCs) and intercalated cells (ICs), posturing the channel for Ca2+-dependent K+ secretion. Fluorescent assessment of cell membrane potential in native, split-opened CCD, demonstrated that selective activation of the Ca2+-permeable TRPV4 channel, thereby inducing Ca2+ influx and elevating intracellular Ca2+ levels, activated both the SK3 channel and the BK channel leading to hyperpolarization of the cell membrane. The hyperpolarization response was decreased to a similar extent by either inhibition of SK3 channel with the selective SK antagonist, apamin, or by inhibition of the BK channel with the selective antagonist, iberiotoxin (IbTX). Addition of both inhibitors produced a further depolarization, indicating cooperative effects of the two channels on Vm. It is concluded that SK3 is functionally expressed in the distal nephron and collecting ducts where induction of TRPV4-mediated Ca2+ influx, leading to elevated intracellular Ca2+ levels, activates this high Ca2+-affinity K+ channel. Further, with sites of expression localized to the apical cell membrane, especially in the CNT and CCD, SK3 is poised to be a key pathway for Ca2+-dependent regulation of membrane potential and K+ secretion.


Assuntos
Túbulos Renais Distais/metabolismo , Canais de Potássio Ativados por Cálcio de Condutância Baixa/metabolismo , Canais de Cátion TRPV/fisiologia , Animais , Aquaporina 2/metabolismo , Sinalização do Cálcio , Expressão Gênica , Rim/citologia , Rim/fisiologia , Potenciais da Membrana , Camundongos Endogâmicos C57BL , Canais de Potássio Ativados por Cálcio de Condutância Baixa/genética
18.
Nat Struct Mol Biol ; 21(2): 180-8, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24440983

RESUMO

FGFR2-expressing human cancer cells with low concentrations of the adaptor protein Grb2 show high prevalence for metastatic outcome. In nonstimulated cells, the SH3 domain (and not the SH2 domains) of Plcγ1 directly competes for a binding site at the very C terminus of FGFR2 with the C-terminal SH3 domain of Grb2. Reduction of Grb2 concentration permits Plcγ1 access to the receptor. Recruitment of Plcγ1 in this way is sufficient to upregulate phospholipase activity. This results in elevated phosphatidylinositol 4,5-bisphosphate turnover and intracellular calcium levels, thus leading to increased cell motility and promotion of cell-invasive behavior in the absence of extracellular receptor stimulation. Therefore, metastatic outcome can be dictated by the constitutive competition between Grb2 and Plcγ1 for the phosphorylation-independent binding site on FGFR2.


Assuntos
Proteína Adaptadora GRB2/fisiologia , Fosfolipase C gama/fisiologia , Fosfolipases/fisiologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Sítios de Ligação , Ligação Competitiva , Linhagem Celular Tumoral , Proteína Adaptadora GRB2/metabolismo , Células HEK293 , Humanos , Modelos Genéticos , Invasividade Neoplásica/genética , Fosfolipase C gama/metabolismo , Estrutura Terciária de Proteína
20.
Methods Mol Biol ; 998: 371-84, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23529445

RESUMO

Transient receptor potential (TRP) channels are expressed in almost every segment of renal nephron from the glomerulus to the inner medullary collecting duct. Serving as a route for Ca(2+) entry from the intratubular space into cells in response to external cues, TRP channels modulate water-electrolyte transport, thus determining functional properties of the renal tubule. In this chapter, we discuss technical aspects of using Ca(2+) imaging to monitor activity of TRP channels in situ, namely, in the freshly isolated distal nephrons, with a special emphasis on the mechanosensitive TRPV4 channel and its role in tubular flow sensing.


Assuntos
Cálcio/metabolismo , Imagem Molecular/métodos , Néfrons/metabolismo , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Imuno-Histoquímica , Camundongos , Néfrons/citologia , Permeabilidade , Ratos , Estresse Mecânico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa