Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
1.
Stroke ; 49(3): 700-709, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29382802

RESUMO

BACKGROUND AND PURPOSE: Human amnion epithelial cells (hAECs) are nonimmunogenic, nontumorigenic, anti-inflammatory cells normally discarded with placental tissue. We reasoned that their profile of biological features, wide availability, and the lack of ethical barriers to their use could make these cells useful as a therapy in ischemic stroke. METHODS: We tested the efficacy of acute (1.5 hours) or delayed (1-3 days) poststroke intravenous injection of hAECs in 4 established animal models of cerebral ischemia. Animals included young (7-14 weeks) and aged mice (20-22 months) of both sexes, as well as adult marmosets of either sex. RESULTS: We found that hAECs administered 1.5 hours after stroke in mice migrated to the ischemic brain via a CXC chemokine receptor type 4-dependent mechanism and reduced brain inflammation, infarct development, and functional deficits. Furthermore, if hAECs administration was delayed until 1 or 3 days poststroke, long-term functional recovery was still augmented in young and aged mice of both sexes. We also showed proof-of-principle evidence in marmosets that acute intravenous injection of hAECs prevented infarct development from day 1 to day 10 after stroke. CONCLUSIONS: Systemic poststroke administration of hAECs elicits marked neuroprotection and facilitates mechanisms of repair and recovery.


Assuntos
Âmnio/transplante , Células Epiteliais/transplante , Neuroproteção , Acidente Vascular Cerebral/terapia , Animais , Callithrix , Modelos Animais de Doenças , Feminino , Xenoenxertos , Humanos , Masculino , Camundongos , Acidente Vascular Cerebral/metabolismo , Acidente Vascular Cerebral/patologia
2.
J Pharmacol Exp Ther ; 367(2): 335-347, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30104322

RESUMO

Renal podocyte survival depends upon the dynamic regulation of a complex cell architecture that links the glomerular basement membrane to integrins, ion channels, and receptors. Alport syndrome is a heritable chronic kidney disease where mutations in α3, α4, or α5 collagen genes promote podocyte death. In rodent models of renal failure, activation of the calcium-sensing receptor (CaSR) can protect podocytes from stress-related death. In this study, we assessed CaSR function in podocyte-like cells derived from induced-pluripotent stem cells from two patients with Alport Syndrome (AS1 & AS2) and a renal disease free individual [normal human mesangial cell (NHMC)], as well as a human immortalized podocyte-like (HIP) cell line. Extracellular calcium elicited concentration-dependent elevations of intracellular calcium in all podocyte-like cells. NHMC and HIP, but not AS1 or AS2 podocyte-like cells, also showed acute reductions in intracellular calcium prior to elevation. In NHMC podocyte-like cells this acute reduction was blocked by the large-conductance potassium channel (KCNMA1) inhibitors iberiotoxin (10 nM) and tetraethylammonium (5 mM), as well as the focal adhesion kinase inhibitor PF562271 (N-methyl-N-(3-((2-(2-oxo-2,3-dihydro-1H-indol-5-ylamino)-5-trifluoromethyl-pyrimidin-4-ylamino)-methyl)-pyridin-2-yl)-methanesulfonamide, 10 nM). Quantitative polymerase chain reaction (qPCR) and immunolabeling showed the presence of KCNMA1 transcript and protein in all podocyte-like cells tested. Cultivation of AS1 podocytes on decellularized plates of NHMC podocyte-like cells partially restored acute reductions in intracellular calcium in response to extracellular calcium. We conclude that the AS patient-derived podocyte-like cells used in this study showed dysfunctional integrin signaling and potassium channel function, which may contribute to podocyte death seen in Alport syndrome.


Assuntos
Células-Tronco Pluripotentes Induzidas/metabolismo , Nefrite Hereditária/metabolismo , Podócitos/metabolismo , Canais de Potássio/metabolismo , Adolescente , Cálcio/metabolismo , Linhagem Celular , Colágeno Tipo IV/metabolismo , Proteína-Tirosina Quinases de Adesão Focal/metabolismo , Membrana Basal Glomerular/metabolismo , Humanos , Subunidades alfa do Canal de Potássio Ativado por Cálcio de Condutância Alta/metabolismo , Masculino , Pessoa de Meia-Idade , Fenótipo , Receptores de Detecção de Cálcio/metabolismo , Transdução de Sinais/fisiologia
3.
Adv Funct Mater ; 25(6): 977-986, 2015 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-26327819

RESUMO

Hydrogels are often employed as temporary platforms for cell proliferation and tissue organization in vitro. Researchers have incorporated photodegradable moieties into synthetic polymeric hydrogels as a means of achieving spatiotemporal control over material properties. In this study protein-based photodegradable hydrogels composed of methacrylated gelatin (GelMA) and a crosslinker containing o-nitrobenzyl ester groups have been developed. The hydrogels are able to degrade rapidly and specifically in response to UV light and can be photopatterned to a variety of shapes and dimensions in a one-step process. Micropatterned photodegradable hydrogels are shown to improve cell distribution, alignment and beating regularity of cultured neonatal rat cardiomyocytes. Overall this work introduces a new class of photodegradable hydrogel based on natural and biofunctional polymers as cell culture substrates for improving cellular organization and function.

4.
Mol Pharm ; 12(3): 794-809, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25644368

RESUMO

The lymphatic system plays a major role in the metastatic dissemination of cancer and has an integral role in immunity. PEGylation enhances drainage and lymphatic uptake following subcutaneous (sc) administration of proteins and protein-like polymers, but the impact of PEGylation of very large proteins (such as antibodies) on subcutaneous and lymphatic pharmacokinetics is unknown. This study therefore aimed to evaluate the impact of PEGylation on the sc absorption and lymphatic disposition of the anti-HER2 antibody trastuzumab in rats. PEG-trastuzumab was generated via the conjugation of a single 40 kDa PEG-NHS ester to trastuzumab. PEG-trastuzumab showed a 5-fold reduction in HER2 binding affinity, however the in vitro growth inhibitory effects were preserved as a result of changes in cellular trafficking when compared to native trastuzumab. The lymphatic pharmacokinetics of PEG-trastuzumab was evaluated in thoracic lymph duct cannulated rats after iv and sc administration and compared to the pharmacokinetics of native trastuzumab. The iv pharmacokinetics and lymphatic exposure of PEG-trastuzumab was similar when compared to trastuzumab. After sc administration, initial plasma pharmacokinetics and lymphatic exposure were also similar between PEG-trastuzumab and trastuzumab, but the absolute bioavailability of PEG-trastuzumab was 100% when compared to 86.1% bioavailability for trastuzumab. In contrast to trastuzumab, PEG-trastuzumab showed accelerated plasma clearance beginning approximately 7 days after sc, but not iv, administration, presumably as a result of the generation of anti-PEG IgM. This work suggests that PEGylation does not significantly alter the lymphatic disposition of very large proteins, and further suggests that it is unlikely to benefit therapy with monoclonal antibodies.


Assuntos
Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Trastuzumab/administração & dosagem , Trastuzumab/metabolismo , Administração Intravenosa , Animais , Antineoplásicos/química , Biofarmácia , Permeabilidade Capilar , Linhagem Celular Tumoral , Células Endoteliais da Veia Umbilical Humana , Humanos , Imunoglobulina M/biossíntese , Imunoglobulina M/sangue , Injeções Subcutâneas , Linfa/metabolismo , Sistema Linfático/metabolismo , Masculino , Taxa de Depuração Metabólica , Modelos Biológicos , Polietilenoglicóis/efeitos adversos , Polietilenoglicóis/química , Ratos , Ratos Sprague-Dawley , Trastuzumab/química
5.
Mol Pharm ; 12(2): 432-43, 2015 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-25485615

RESUMO

The current study sought to explore whether the subcutaneous administration of lymph targeted dendrimers, conjugated with a model chemotherapeutic (methotrexate, MTX), was able to enhance anticancer activity against lymph node metastases. The lymphatic pharmacokinetics and antitumor activity of PEGylated polylysine dendrimers conjugated to MTX [D-MTX(OH)] via a tumor-labile hexapeptide linker was examined in rats and compared to a similar system where MTX was α-carboxyl O-tert-butylated [D-MTX(OtBu)]. The latter has previously been shown to exhibit longer plasma circulation times. D-MTX(OtBu) was well absorbed from the subcutaneous injection site via the lymph, and 3 to 4%/g of the dose was retained by sentinel lymph nodes. In contrast, D-MTX(OH) showed limited absorption from the subcutaneous injection site, but absorption was almost exclusively via the lymph. The retention of D-MTX(OH) by sentinel lymph nodes was also significantly elevated (approximately 30% dose/g). MTX alone was not absorbed into the lymph. All dendrimers displayed lower lymph node targeting after intravenous administration. Despite significant differences in the lymph node retention of D-MTX(OH) and D-MTX(OtBu) after subcutaneous and intravenous administration, the growth of lymph node metastases was similarly inhibited. In contrast, the administration of MTX alone did not significantly reduce lymph node tumor growth. Subcutaneous administration of drug-conjugated dendrimers therefore provides an opportunity to improve drug deposition in downstream tumor-burdened lymph nodes. In this case, however, increased lymph node biodistribution did not correlate well with antitumor activity, possibly suggesting constrained drug release at the site of action.


Assuntos
Dendrímeros/química , Dendrímeros/farmacocinética , Linfonodos/metabolismo , Metotrexato/química , Metotrexato/farmacocinética , Polietilenoglicóis/química , Animais , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Masculino , Microscopia Confocal , Neoplasias/metabolismo , Ratos , Ratos Endogâmicos F344 , Ratos Sprague-Dawley
6.
Nat Methods ; 8(12): 1037-40, 2011 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-22020065

RESUMO

NKX2-5 is expressed in the heart throughout life. We targeted eGFP sequences to the NKX2-5 locus of human embryonic stem cells (hESCs); NKX2-5(eGFP/w) hESCs facilitate quantification of cardiac differentiation, purification of hESC-derived committed cardiac progenitor cells (hESC-CPCs) and cardiomyocytes (hESC-CMs) and the standardization of differentiation protocols. We used NKX2-5 eGFP(+) cells to identify VCAM1 and SIRPA as cell-surface markers expressed in cardiac lineages.


Assuntos
Separação Celular/métodos , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Proteínas de Homeodomínio/metabolismo , Mioblastos Cardíacos/citologia , Miócitos Cardíacos/citologia , Fatores de Transcrição/metabolismo , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Biomarcadores/análise , Diferenciação Celular , Perfilação da Expressão Gênica , Proteína Homeobox Nkx-2.5 , Proteínas de Homeodomínio/genética , Humanos , Mioblastos Cardíacos/metabolismo , Miócitos Cardíacos/metabolismo , Reação em Cadeia da Polimerase , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Fatores de Transcrição/genética , Molécula 1 de Adesão de Célula Vascular/genética , Molécula 1 de Adesão de Célula Vascular/metabolismo
7.
Stem Cells ; 30(7): 1349-61, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22495882

RESUMO

LIM homeobox transcription factor 1 alpha (Lmx1a) is required for the development of midbrain dopaminergic neurons, roof plate formation, and cortical hem development. We generated a reporter embryonic stem cell (ESC) line for Lmx1a and used it to track differentiation and extract neural progenitors from differentiating mouse ESCs. Lmx1a(+) cells gave rise to functional cortical upper layer GABAergic neurons or dopaminergic neurons depending on the culture conditions used for differentiation. Under chemically defined neurobasal conditions, ESC differentiation resulted in widespread and transient expression of Lmx1a, without the addition of exogenous factors such as sonic hedgehog (Shh), Wnts, and/or bone morphogenic proteins (BMPs). Under neutral conditions, Lmx1a(+) cells express genes known to be downstream of Lmx1a and cortical hem markers Wnt3a and p73. The majority of these cells did not express the ventral midbrain dopaminergic marker Foxa2 or dorsal roof plate marker BMP-2. Lmx1a(+) -Foxa2(-) cells were primed to become SatB2(+) GABAergic neurons and appeared to be resistant to dopaminergic patterning cues. PA6 coculture produced a substantial population of Lmx1a(+) progenitors that also expressed Foxa2 and on further differentiation gave rise to dopaminergic neurons at high frequency. We conclude that Lmx1a is a useful marker for the extraction of progenitors of GABAergic or dopaminergic neurons. We caution against the assumption that it indicates dopaminergic commitment during in vitro differentiation of ESCs. Indeed, in monolayer culture under neurobasal conditions, with or without the addition of Shh and fibroblast growth factor 8 (FGF8), Lmx1a(+) cells were predominantly progenitors of forebrain GABAergic neurons. We obtained dopaminergic cells in large numbers only by coculture with PA6 cells.


Assuntos
Neurônios Dopaminérgicos/citologia , Neurônios Dopaminérgicos/metabolismo , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Neurônios GABAérgicos/citologia , Neurônios GABAérgicos/metabolismo , Proteínas com Homeodomínio LIM/metabolismo , Fator de Transcrição MSX1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Citometria de Fluxo , Imuno-Histoquímica , Proteínas com Homeodomínio LIM/genética , Fator de Transcrição MSX1/genética , Camundongos , Reação em Cadeia da Polimerase , Fatores de Transcrição/genética
8.
Front Cell Dev Biol ; 11: 1111705, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36819101

RESUMO

The generation of midbrain dopaminergic neurons (mDAs) from pluripotent stem cells (hPSC) holds much promise for both disease modelling studies and as a cell therapy for Parkinson's disease (PD). Generally, dopaminergic neuron differentiation paradigms rely on inhibition of smad signalling for neural induction followed by hedgehog signalling and an elevation of ß-catenin to drive dopaminergic differentiation. Post-patterning, differentiating dopaminergic neuron cultures are permitted time for maturation after which the success of these differentiation paradigms is usually defined by expression of tyrosine hydroxylase (TH), the rate limiting enzyme in the synthesis of dopamine. However, during maturation, culture media is often supplemented with additives to promote neuron survival and or promote cell differentiation. These additives include dibutyryl cyclic adenosine monophosphate (dbcAMP), transforming growth factor ß3 (TGFß3) and or the γ-secretase inhibitor (DAPT). While these factors are routinely added to cultures, their impact upon pluripotent stem cell-derived mDA phenotype is largely unclear. In this study, we differentiate pluripotent stem cells toward a dopaminergic phenotype and investigate how the omission of dbcAMP, TGFß3 or DAPT, late in maturation, affects the regulation of multiple dopaminergic neuron phenotype markers. We now show that the removal of dbcAMP or TGFß3 significantly and distinctly impacts multiple markers of the mDA phenotype (FOXA2, EN1, EN2, FOXA2, SOX6), while commonly increasing both MSX2 and NEUROD1 and reducing expression of both tyrosine hydroxylase and WNT5A. Removing DAPT significantly impacted MSX2, OTX2, EN1, and KCNJ6. In the absence of any stressful stimuli, we suggest that these culture additives should be viewed as mDA phenotype-modifying, rather than neuroprotective. We also suggest that their addition to cultures is likely to confound the interpretation of both transplantation and disease modelling studies.

9.
Stem Cells ; 29(6): 928-39, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21472823

RESUMO

Investigation of serotonergic neuronal activity and its relationship to disease has been limited by a lack of physiologically relevant in vitro cell models. Serotonergic neurons derived from embryonic stem cells (ESCs) could provide a platform for such studies and provide models for use in drug discovery. Here, we report enhancement of serotonergic differentiation using a genetic approach. Expression of Gata2 increased the yield of serotonergic neurons. Enhancement was only achieved when Gata2 was expressed under the control of the tissue-specific promoter of the transcription factor Nkx6.1. High levels of Gata2 expression in ESCs compromised pluripotency and induced non-neuronal differentiation. Combined directed expression of Gata2, proneural gene Mash1, and forkhead transcription factor Foxa2 further enhanced serotonergic neural differentiation, resulting in a 10-fold increase in serotonin content. These neurons were also capable of depolarization (KCl, 30 mM)-induced elevations of intracellular Ca(2+) . The presence of sonic hedgehog during differentiation produced a further modest increase in numbers (1.5-fold). Transgene expression did not influence the number of tyrosine hydroxylase positive neurons in the cultures after 20 days, implying that Gata2, Mash1, and Foxa2 modulate in vitro differentiation at a time beyond the decision-point for dopaminergic or nondopaminergic commitment. This study demonstrates that the directed expression of specific transcription factors enhances serotonergic neuron differentiation in vitro and highlights the importance of transgene expression at the right stage of ESC differentiation to effect the generation of a desired neural subtype.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Células-Tronco Embrionárias/citologia , Fator de Transcrição GATA2/metabolismo , Fator 3-beta Nuclear de Hepatócito/metabolismo , Neurônios/metabolismo , Serotonina/biossíntese , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Clonagem Molecular , Células-Tronco Embrionárias/metabolismo , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA3/genética , Fator de Transcrição GATA3/metabolismo , Genes Reporter , Proteínas de Fluorescência Verde/biossíntese , Proteínas de Fluorescência Verde/genética , Proteínas Hedgehog/farmacologia , Fator 3-beta Nuclear de Hepatócito/genética , Proteínas de Homeodomínio/genética , Camundongos , Fator 1 de Elongação de Peptídeos/genética , Fenótipo , Regiões Promotoras Genéticas , Biossíntese de Proteínas/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , beta-Lactamases/biossíntese , beta-Lactamases/genética
10.
Stem Cells ; 29(3): 462-73, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21425409

RESUMO

We have used homologous recombination in human embryonic stem cells (hESCs) to insert sequences encoding green fluorescent protein (GFP) into the NKX2.1 locus, a gene required for normal development of the basal forebrain. Generation of NKX2.1-GFP(+) cells was dependent on the concentration, timing, and duration of retinoic acid treatment during differentiation. NKX2.1-GFP(+) progenitors expressed genes characteristic of the basal forebrain, including SHH, DLX1, LHX6, and OLIG2. Time course analysis revealed that NKX2.1-GFP(+) cells could upregulate FOXG1 expression, implying the existence of a novel pathway for the generation of telencephalic neural derivatives. Further maturation of NKX2.1-GFP(+) cells gave rise to γ-aminobutyric acid-, tyrosine hydroxylase-, and somatostatin-expressing neurons as well as to platelet-derived growth factor receptor α-positive oligodendrocyte precursors. These studies highlight the diversity of cell types that can be generated from human NKX2.1(+) progenitors and demonstrate the utility of NKX2.1(GFP/w) hESCs for investigating human forebrain development and neuronal differentiation.


Assuntos
Linhagem da Célula/genética , Rastreamento de Células/métodos , Células-Tronco Embrionárias/metabolismo , Proteínas Nucleares/genética , Prosencéfalo/embriologia , Fatores de Transcrição/genética , Animais , Animais Recém-Nascidos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células Cultivadas , Células-Tronco Embrionárias/citologia , Citometria de Fluxo/métodos , Genes Reporter , Humanos , Camundongos , Camundongos Transgênicos , Terapia de Alvo Molecular/métodos , Neurogênese/genética , Neurogênese/fisiologia , Proteínas Nucleares/metabolismo , Prosencéfalo/citologia , Prosencéfalo/fisiologia , Fator Nuclear 1 de Tireoide , Fatores de Transcrição/metabolismo
11.
Cells ; 11(12)2022 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-35741036

RESUMO

Molecular transport between the nucleus and cytoplasm of the cell is mediated by the importin superfamily of transport receptors, of which the bidirectional transporter Importin 13 (IPO13) is a unique member, with a critical role in early embryonic development through nuclear transport of key regulators, such as transcription factors Pax6, Pax3, and ARX. Here, we examined the role of IPO13 in neuronal differentiation for the first time, using a mouse embryonic stem cell (ESC) model and a monolayer-based differentiation protocol to compare IPO13-/- to wild type ESCs. Although IPO13-/- ESCs differentiated into neural progenitor cells, as indicated by the expression of dorsal forebrain progenitor markers, reduced expression of progenitor markers Pax6 and Nestin compared to IPO13-/- was evident, concomitant with reduced nuclear localisation/transcriptional function of IPO13 import cargo Pax6. Differentiation of IPO13-/- cells into neurons appeared to be strongly impaired, as evidenced by altered morphology, reduced expression of key neuronal markers, and altered response to the neurotransmitter glutamate. Our findings establish that IPO13 has a key role in ESC neuronal differentiation, in part through the nuclear transport of Pax6.


Assuntos
Carioferinas/metabolismo , Células-Tronco Neurais , Transporte Ativo do Núcleo Celular , Animais , Diferenciação Celular , Núcleo Celular/metabolismo , Citoplasma/metabolismo , Camundongos , Células-Tronco Neurais/metabolismo
12.
Pharmaceuticals (Basel) ; 15(3)2022 Feb 24.
Artigo em Inglês | MEDLINE | ID: mdl-35337083

RESUMO

The aim of this study was to assess the utility of inexpensive techniques in evaluating the interactions of risperidone (Ris) with different traditional π-acceptors, with subsequent application of the findings into a Ris pharmaceutical formulation with improved therapeutic properties. Molecular docking calculations were performed using Ris and its different charge-transfer complexes (CT) with picric acid (PA), 2,3-dichloro-5,6-dicyanop-benzoquinon (DDQ), tetracyanoquinodimethane (TCNQ), tetracyano ethylene (TCNE), tetrabromo-pquinon (BL), and tetrachloro-p-quinon (CL), as donors, and three receptors (serotonin, dopamine, and adrenergic) as acceptors to study the comparative interactions among them. To refine the docking results and further investigate the molecular processes of receptor-ligand interactions, a molecular dynamics simulation was run with output obtained from AutoDock Vina. Among all investigated complexes, the [(Ris) (PA)]-serotonin (CTcS) complex showed the highest binding energy. Molecular dynamics simulation of the 100 ns run revealed that both the Ris-serotonin (RisS) and CTcS complexes had a stable conformation; however, the CTcS complex was more stable.

13.
J Pharmacol Exp Ther ; 339(3): 870-7, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21885618

RESUMO

Increased smooth muscle tone in the human prostate contributes to the symptoms associated with benign prostatic hyperplasia. In the mouse prostate gland, cholinergic innervation is responsible for a component of the nerve-mediated contractile response. This study investigates the muscarinic receptor subtype responsible for the cholinergic contractile response in the mouse prostate gland. To characterize the muscarinic receptor subtype, mouse prostates taken from wild-type or M(3) muscarinic receptor knockout mice were mounted in organ baths. The isometric force that tissues developed in response to electrical-field stimulation or exogenously applied cholinergic agonists in the presence or absence of a range of muscarinic receptor antagonists was evaluated. Carbachol elicited reproducible and concentration-dependent contractions of the isolated mouse prostate, which were antagonized by the presence of muscarinic receptor antagonists. Calculation of antagonist affinities (pA(2) values) indicated a rank order of antagonist potencies in the mouse prostate of: darifenacin (9.08) = atropine (9.07) = 1,1-dimethyl-4-diphenylacetoxypiperidinium iodide (9.02) > cyclohexyl-hydroxy-phenyl-(3-piperidin-1-ylpropyl)silane (7.85) > cyclohexyl-(4-fluorophenyl)-hydroxy-(3-piperidin-1-ylpropyl)silane (7.39) > himbacine (7.19) > pirenzipine (6.88) > methoctramine (6.20). Furthermore, genetic deletion of the M(3) muscarinic receptor inhibited prostatic contractions to electrical-field stimulation or exogenous administration of acetylcholine. In this study we identified that the cholinergic component of contraction in the mouse prostate is mediated by the M(3) muscarinic receptor subtype. Pharmacological antagonism of the M(3) muscarinic receptor may be a beneficial additional target for the treatment of benign prostatic hyperplasia in the human prostate gland.


Assuntos
Acetilcolina/farmacologia , Agonistas Colinérgicos/farmacologia , Mecamilamina/farmacologia , Antagonistas Muscarínicos/farmacologia , Contração Muscular/efeitos dos fármacos , Antagonistas Nicotínicos/farmacologia , Próstata/fisiologia , Receptor Muscarínico M3/antagonistas & inibidores , Receptor Muscarínico M3/metabolismo , Antagonistas de Receptores Adrenérgicos alfa 1/farmacologia , Animais , Benzofuranos/farmacologia , Peso Corporal/efeitos dos fármacos , Carbacol/farmacologia , Interações Medicamentosas , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Contração Muscular/fisiologia , Prazosina/farmacologia , Próstata/efeitos dos fármacos , Pirrolidinas/farmacologia , Receptor Muscarínico M3/genética
14.
Lab Chip ; 21(20): 4016-4030, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34487130

RESUMO

Compartmentalized microfluidic devices are becoming increasingly popular and have proven to be valuable tools to probe neurobiological functions that are inherently difficult to study using traditional approaches. The ability of microfluidic devices to compartmentalize neurons offers considerable promise for disease modeling and drug discovery. Rodent cortical neurons/neural progenitors are commonly used in such studies but, while these cells mature rapidly, they do not possess the same receptors, ion channels and transport proteins found in human cortical neurons. Human pluripotent stem cell derived neurons offer a human phenotype, but their slow maturation offsets this phenotypic advantage, particularly over long-term culture where overgrowth and subsequent death of neurons may be a problem. In this work, we integrate the use of Matrigel as a 3D cell culture scaffold that enables high cell seeding density over a small fraction of the culture surface. This approach, in an open chamber microfluidic system, enables culture over a five-month period without the use of growth inhibitors. Matrigel was also uniquely utilized to hinder agonist diffusion across microchannels. We demonstrate the development of neuron-to-neuron communication networks by showing that electrical stimulation or the unilateral addition of agonists to one chamber resulted in activation of neurons in the adjacent chamber. Lastly, using a delayed neuron seeding strategy, we show that we can foster essentially one-way communication between separate populations of human forebrain and midbrain dopaminergic neuron containing cultures.


Assuntos
Microfluídica , Células-Tronco Pluripotentes , Diferenciação Celular , Neurônios Dopaminérgicos , Humanos , Mesencéfalo , Prosencéfalo
15.
PLoS One ; 16(12): e0261730, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34941945

RESUMO

In this study we investigate how ß-catenin-dependent WNT signalling impacts midbrain dopaminergic neuron (mDA) specification. mDA cultures at day 65 of differentiation responded to 25 days of the tankyrase inhibitor XAV969 (XAV, 100nM) with reduced expression of markers of an A9 mDA phenotype (KCNJ6, ALDH1A1 and TH) but increased expression of the transcriptional repressors NR0B1 and NR0B2. Overexpression of NR0B1 and or NR0B2 promoted a loss of A9 dopaminergic neuron phenotype markers (KCNJ6, ALDH1A1 and TH). Overexpression of NR0B1, but not NR0B2 promoted a reduction in expression of the ß-catenin-dependent WNT signalling pathway activator RSPO2. Analysis of Parkinson's disease (PD) transcriptomic databases shows a profound PD-associated elevation of NR0B1 as well as reduced transcript for RSPO2. We conclude that reduced ß-catenin-dependent WNT signalling impacts dopaminergic neuron identity, in vitro, through increased expression of the transcriptional repressor, NR0B1. We also speculate that dopaminergic neuron regulatory mechanisms may be perturbed in PD and that this may have an impact upon both existing nigral neurons and also neural progenitors transplanted as PD therapy.


Assuntos
Receptor Nuclear Órfão DAX-1/biossíntese , Neurônios Dopaminérgicos/metabolismo , Regulação para Baixo , Células-Tronco Embrionárias Humanas/metabolismo , Doença de Parkinson/metabolismo , Regulação para Cima , Via de Sinalização Wnt , beta Catenina/metabolismo , Biomarcadores/metabolismo , Receptor Nuclear Órfão DAX-1/genética , Humanos , Doença de Parkinson/genética , beta Catenina/genética
16.
Nat Commun ; 12(1): 3015, 2021 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-34021136

RESUMO

The role of microglia cells in Alzheimer's disease (AD) is well recognized, however their molecular and functional diversity remain unclear. Here, we isolated amyloid plaque-containing (using labelling with methoxy-XO4, XO4+) and non-containing (XO4-) microglia from an AD mouse model. Transcriptomics analysis identified different transcriptional trajectories in ageing and AD mice. XO4+ microglial transcriptomes demonstrated dysregulated expression of genes associated with late onset AD. We further showed that the transcriptional program associated with XO4+ microglia from mice is present in a subset of human microglia isolated from brains of individuals with AD. XO4- microglia displayed transcriptional signatures associated with accelerated ageing and contained more intracellular post-synaptic material than XO4+ microglia, despite reduced active synaptosome phagocytosis. We identified HIF1α as potentially regulating synaptosome phagocytosis in vitro using primary human microglia, and BV2 mouse microglial cells. Together, these findings provide insight into molecular mechanisms underpinning the functional diversity of microglia in AD.


Assuntos
Doença de Alzheimer/metabolismo , Microglia/metabolismo , Fagocitose/fisiologia , Placa Amiloide/metabolismo , Idoso , Idoso de 80 Anos ou mais , Animais , Encéfalo/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Redes Reguladoras de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Placa Amiloide/genética , Transcriptoma
17.
Prostate ; 70(11): 1222-32, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20564424

RESUMO

BACKGROUND: Increased smooth muscle tone is a significant component of benign prostatic hyperplasia, the onset of which correlates with age and declining serum testosterone levels. This study investigates the effects of androgens on key regulators of smooth muscle tone: intracellular calcium ([Ca(2+)](i)) and cyclic adenosine monophosphate (cAMP) in human cultured prostatic stromal cells (HCPSC). METHODS: HCPSC were cultured in the absence or presence of dihydrotestosterone (DHT; 3, 30, and 300 nM) or testosterone (0.3-300 nM) alone or in the presence of flutamide (10 microM). Changes in [Ca(2+)](i) were determined in FURA-2AM (10 microM) loaded cells. Changes in cAMP were determined by Alpha Screen(R) assay. RESULTS: Up to 32% of cultured cells exhibited spontaneous elevations of [Ca(2+)](i). The frequency of these elevations was reduced by nifedipine (10 microM), ryanodine (1 microM), and the adenylate cyclase inhibitor MDL 12,330A (20 microM). Compared to steroid-free cells, a 3-day incubation of cells with testosterone (only 3 nM) elevated basal, but not peak [Ca(2+)](i). In the presence of flutamide, all concentrations of testosterone tested elevated basal, but not peak [Ca(2+)](i). DHT (30 and 300, but not 3 nM) lowered peak and basal [Ca(2+)](i). Increased testosterone concentration dependently decreased resting cell cAMP (pIC(50): 7.64 +/- 0.29 nM). CONCLUSIONS: These findings demonstrate that some HCPSC have the ability to spontaneously and transiently elevate [Ca(2+)](i). The magnitude of these [Ca(2+)](i) peaks, along with resting levels of calcium and cAMP, appear to be regulated by androgens.


Assuntos
Adenilil Ciclases/metabolismo , Androgênios/metabolismo , Cálcio/metabolismo , AMP Cíclico/metabolismo , Hiperplasia Prostática/metabolismo , Inibidores de Adenilil Ciclases , Idoso , Antagonistas de Androgênios/farmacologia , Inibidores Enzimáticos/farmacologia , Flutamida/farmacologia , Humanos , Iminas/farmacologia , Imuno-Histoquímica , Masculino , Microscopia de Fluorescência , Nifedipino/farmacologia , Hiperplasia Prostática/enzimologia , Hiperplasia Prostática/patologia , Rianodina/farmacologia , Células Estromais/enzimologia , Células Estromais/metabolismo , Células Estromais/patologia
18.
J Pharmacol Exp Ther ; 335(2): 489-96, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20724483

RESUMO

Neuronal release of noradrenaline is primarily responsible for the contraction of prostatic smooth muscle in all species, and this forms the basis for the use of α(1)-adrenoceptor antagonists as pharmacotherapies for benign prostatic hyperplasia. Previous studies in mice have demonstrated that a residual nonadrenergic component to nerve stimulation remains after α(1)-adrenoceptor antagonism. In the guinea pig and rat prostate and the vas deferens of guinea pigs, rats, and mice, ATP is the mediator of this residual contraction. This study investigates the mediator of residual contraction in the mouse prostate. Whole prostates from wild-type, α(1A)-adrenoceptor, and P2X1-purinoceptor knockout mice were mounted in organ baths, and the isometric force that tissues developed in response to electrical field stimulation or exogenously applied agonists was recorded. Deletion of the P2X1 purinoceptor did not affect nerve-mediated contraction. Furthermore, the P2-purinoceptor antagonist suramin (30 µM) failed to attenuate nerve-mediated contractions in wild-type, α(1A)-adrenoceptor, or P2X1-purinoceptor knockout mice. Atropine (1 µM) attenuated contraction in prostates taken from wild-type mice. In the presence of prazosin (0.3 µM) or guanethidine (10 µM), or in prostates taken from α(1A)-adrenoceptor knockout mice, residual nerve-mediated contraction was abolished by atropine (1 µM), but not suramin (30 µM). Exogenously administered acetylcholine elicited reproducible concentration-dependent contractions of the mouse prostate that were atropine-sensitive (1 µM), but not prazosin-sensitive (0.3 µM). Acetylcholine, but not ATP, mediates the nonadrenergic component of contraction in the mouse prostate. This cholinergic component of prostatic contraction is mediated by activation of muscarinic receptors.


Assuntos
Acetilcolina/metabolismo , Trifosfato de Adenosina/metabolismo , Contração Muscular/fisiologia , Próstata/metabolismo , Ducto Deferente/metabolismo , Agonistas de Receptores Adrenérgicos alfa 1 , Antagonistas de Receptores Adrenérgicos alfa 1 , Animais , Atropina/farmacologia , Relação Dose-Resposta a Droga , Estimulação Elétrica , Masculino , Camundongos , Camundongos Knockout , Contração Muscular/efeitos dos fármacos , Músculo Liso/efeitos dos fármacos , Músculo Liso/inervação , Músculo Liso/metabolismo , Técnicas de Cultura de Órgãos , Prazosina/farmacologia , Próstata/efeitos dos fármacos , Próstata/inervação , Próstata/fisiologia , Receptores Adrenérgicos alfa 1/genética , Receptores Purinérgicos P2X1/agonistas , Receptores Purinérgicos P2X1/antagonistas & inibidores , Receptores Purinérgicos P2X1/genética , Ducto Deferente/efeitos dos fármacos , Ducto Deferente/inervação , Ducto Deferente/fisiologia
19.
J Cardiovasc Pharmacol ; 56(3): 282-92, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20571427

RESUMO

The cardioprotective effects of a novel adenosine A1 receptor agonist N6-(2,2,5,5-tetramethylpyrrolidin-1-yloxyl-3-ylmethyl) adenosine (VCP28) were compared with the selective adenosine A1 receptor agonist N6-cyclopentyladenosine (CPA) in a H9c2(2-1) cardiac cell line-simulated ischemia (SI) model (12 hours) and a global ischemia (30 minutes) and reperfusion (60 minutes) model in isolated rat heart model. H9c2(2-1) cells were treated with CPA and VCP28 at the start of ischemia for entire ischemic duration, whereas isolated rat hearts were treated at the onset of reperfusion for 15 minutes. In the H9c2(2-1) cells SI model, CPA and VCP28 (100 nM) significantly (P < 0.05, n = 5-6) reduced the proportion of nonviable cells (30.88% +/- 2.49% and 16.17% +/- 3.77% of SI group, respectively) and lactate dehydrogenase efflux. In isolated rat hearts, CPA and VCP28 significantly (n = 6-8, P < 0.05) improved post-ischemic contractility (dP/dt(max), 81.69% +/- 10.96%, 91.07% +/- 19.87% of baseline, respectively), left ventricular developed pressure, and end diastolic pressure and reduced infarct size. The adenosine A1 receptor antagonist abolished the cardioprotective effects of CPA and VCP28 in SI model and isolated rat hearts. In conclusion, the adenosine A1 receptor agonist VCP28 has equal cardioprotective effects to the prototype A1 agonist CPA at concentrations that have no effect on heart rate.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Adenosina/análogos & derivados , Isquemia Miocárdica/metabolismo , Pirrolidinas/farmacologia , Traumatismo por Reperfusão/metabolismo , Adenosina/farmacologia , Animais , Linhagem Celular , Frequência Cardíaca/efeitos dos fármacos , Técnicas In Vitro , Contração Miocárdica/efeitos dos fármacos , Isquemia Miocárdica/patologia , Isquemia Miocárdica/fisiopatologia , Miocárdio/patologia , Ratos , Traumatismo por Reperfusão/patologia , Traumatismo por Reperfusão/fisiopatologia
20.
Nat Rev Drug Discov ; 6(8): 605-16, 2007 08.
Artigo em Inglês | MEDLINE | ID: mdl-17667955

RESUMO

Embryonic stem cells (ESCs) will become a source of models for a wide range of adult differentiated cells, providing that reliable protocols for directed differentiation can be established. Stem-cell technology has the potential to revolutionize drug discovery, making models available for primary screens, secondary pharmacology, safety pharmacology, metabolic profiling and toxicity evaluation. Models of differentiated cells that are derived from mouse ESCs are already in use in drug discovery, and are beginning to find uses in high-throughput screens. Before analogous human models can be obtained in adequate numbers, reliable methods for the expansion of human ESC cultures will be needed. For applications in drug discovery, involving either species, protocols for directed differentiation will need to be robust and affordable. Here, we explore current challenges and future opportunities in relation to the use of stem-cell technology in drug discovery, and address the use of both mouse and human models.


Assuntos
Desenho de Fármacos , Células-Tronco Embrionárias/metabolismo , Tecnologia Farmacêutica/métodos , Animais , Técnicas de Cultura de Células/métodos , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Microscopia de Fluorescência , Modelos Biológicos , Estrutura Molecular , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Preparações Farmacêuticas/administração & dosagem , Preparações Farmacêuticas/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa