Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 235
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Am Chem Soc ; 146(28): 18927-18937, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-38968420

RESUMEN

Hydrogen sulfide (H2S) is an endogenously produced gasotransmitter involved in many physiological processes that are integral to proper cellular functioning. Due to its profound anti-inflammatory and antioxidant properties, H2S plays important roles in preventing inflammatory skin disorders and improving wound healing. Transdermal H2S delivery is a therapeutically viable option for the management of such disorders. However, current small-molecule H2S donors are not optimally suited for transdermal delivery and typically generate electrophilic byproducts that may lead to undesired toxicity. Here, we demonstrate that H2S release from metal-organic frameworks (MOFs) bearing coordinatively unsaturated metal centers is a promising alternative for controlled transdermal delivery of H2S. Gas sorption measurements and powder X-ray diffraction (PXRD) studies of 11 MOFs support that the Mg-based framework Mg2(dobdc) (dobdc4- = 2,5-dioxidobenzene-1,4-dicarboxylate) is uniquely well-suited for transdermal H2S delivery due to its strong yet reversible binding of H2S, high capacity (14.7 mmol/g at 1 bar and 25 °C), and lack of toxicity. In addition, Rietveld refinement of synchrotron PXRD data from H2S-dosed Mg2(dobdc) supports that the high H2S capacity of this framework arises due to the presence of three distinct binding sites. Last, we demonstrate that transdermal delivery of H2S from Mg2(dobdc) is sustained over a 24 h period through porcine skin. Not only is this significantly longer than sodium sulfide but this represents the first example of controlled transdermal delivery of pure H2S gas. Overall, H2S-loaded Mg2(dobdc) is an easily accessible, solid-state source of H2S, enabling safe storage and transdermal delivery of this therapeutically relevant gas.


Asunto(s)
Administración Cutánea , Sulfuro de Hidrógeno , Estructuras Metalorgánicas , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/administración & dosificación , Estructuras Metalorgánicas/química , Animales , Porcinos , Piel/metabolismo
2.
Pharm Res ; 39(2): 341-352, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-35088236

RESUMEN

PURPOSE: Hydrogen sulphide (H2S) is an important signalling molecule involved in the regulation of several physiological and pathophysiological processes. The objective of this study was to investigate the feasibility of transdermal delivery of ADT-OH, a H2S donor, by investigating the transdermal flux of aqueous gels loaded with penetration enhancers or liposomes. Furthermore, we explored the ability of permeated ADT-OH to promote angiogenesis and mitochondrial bioenergetics in HUVEC cells. METHODS: Aqueous hypromellose gels (5% w/v) were prepared with up to 10% v/v propylene glycol (PG) or deformable liposomes with 0.025% w/w ADT-OH. ADT-OH permeation from formulations across excised murine skin into PBS was quantified over 24 h using HPLC-UV detection. Media was collected and applied to HUVEC cells to evidence ADT-OH functionality following permeation. Tube formation assays were performed as indicative of angiogenesis and mitochondrial oxygen consumption was evaluated using a Seahorse XF24. RESULTS: Increasing the loading of PG caused an increase in ADT-OH permeation rate across skin and a decrease in dermal drug retention whereas liposomal gels produced a slow-release profile. Treatment of HUVEC's using conditioned media collected from the ADT-OH loaded permeation studies enhanced tube formation and the basal oxygen consumption rates after 30 min of treatment. CONCLUSIONS: These findings demonstrate that transdermal delivery of ADT-OH may provide a promising approach in the treatment of impaired vascular function. Gels prepared with 10% v/v PG have the potential for use in conditions requiring rapid H2S release whereas liposomal loaded gels for treatment requiring sustained H2S release.


Asunto(s)
Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Sulfuro de Hidrógeno/administración & dosificación , Absorción Cutánea , Tionas/administración & dosificación , Administración Cutánea , Animales , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Composición de Medicamentos , Metabolismo Energético/efectos de los fármacos , Femenino , Geles , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/metabolismo , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Neovascularización Fisiológica/efectos de los fármacos , Consumo de Oxígeno/efectos de los fármacos , Tionas/química , Tionas/metabolismo
3.
Dig Dis Sci ; 66(8): 2661-2668, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-32918175

RESUMEN

Hydrogen sulfide (H2S) is a recently discerned endogenous signaling molecule that modulates the vascular system. Endogenous hydrogen sulfide has been shown to dilate both the mesenteric and portal vasculature. Gut microbiome, via sulfur reducing bacteria, is another source of H2S production within the gut lumen; this source of H2S is primarily produced and detoxified in the colon under physiologic conditions. Nitric oxide (NO), a major endogenous vasodilator in the portal circulation, participates in H2S-induced vasodilation in some vascular beds. We hypothesize that jejunal but not colonic H2S increases portal vein flow in a NO-dependent fashion. To evaluate the effects of luminal H2S, venous blood flow, portal venous pressure, and systemic venous pressure were measured in rats after administration of either vehicle or an H2S donor (NaHS) into the jejunum or the colon. We found that portal venous pressure and systemic pressure did not change and were similar between the three study groups. However, portal venous blood flow significantly increased following jejunal administration of NaHS but not in response to colonic NaHS or vehicle administration. To test the contribution of NO production to this response, another group of animals was treated with either an NO synthase inhibitor (N-Ω-nitro-L-arginine, L-NNA) or saline prior to jejunal NaHS infusion. After L-NNA pretreatment, NaHS caused a significant fall rather than increase in portal venous flow compared to saline pretreatment. These data demonstrate that H2S within the small intestine significantly increases portal venous blood flow in a NO-dependent fashion.


Asunto(s)
Colon/metabolismo , Sulfuro de Hidrógeno/farmacología , Yeyuno/metabolismo , Óxido Nítrico/metabolismo , Vena Porta/fisiología , Animales , Colon/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Sulfuro de Hidrógeno/administración & dosificación , Yeyuno/efectos de los fármacos , Óxido Nítrico Sintasa/genética , Óxido Nítrico Sintasa/metabolismo , Nitroarginina/farmacología , Ratas , Ratas Sprague-Dawley
4.
J Nanobiotechnology ; 19(1): 40, 2021 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-33549092

RESUMEN

BACKGROUND: Cardiac arrest (CA) is a leading cause of death worldwide. Even after successful cardiopulmonary resuscitation (CPR), the majorities of survivals are companied with permanent myocardial and cerebral injury. Hydrogen sulfide (H2S) has been recognized as a novel gasotransmitter exerting multiple organ protection; however, the lacks of ideal H2S donors which can controlled release H2S to targeted organs such as heart and brain limits its application. RESULTS: This work utilized mesoporous iron oxide nanoparticle (MION) as the carriers of diallyl trisulfide (DATS), with polyethylene glycol (PEG) and lactoferrin (LF) modified to MIONs to acquire the prolonged circulation time and brain-targeting effects, and a novel targeted H2S releasing system was constructed (DATS@MION-PEG-LF), which exhibited excellent biocompatibility, controlled-releasing H2S pattern, heart and brain targeting features, and the ability to be non-invasive traced by magnetic resonance imaging. DATS@MION-PEG-LF presented potent protective effects against cerebral and cardiac ischemic injury after CA in both in vitro hypoxia/reoxygenation models and in vivo CA/CPR models, which mainly involves anti-apoptosis, anti-inflammatory and anti-oxidant mechanisms. Accordingly, the cardiac and cerebral functions were obviously improved after CA/CPR, with potentially improved survival. CONCLUSIONS: The present work provides a unique platform for targeted controlled release of H2S based on MIONs, and offers a new method for combinational myocardial and cerebral protection from ischemic injury, bringing considerable benefits for CA patients.


Asunto(s)
Isquemia Encefálica/prevención & control , Preparaciones de Acción Retardada/química , Paro Cardíaco/complicaciones , Sulfuro de Hidrógeno/administración & dosificación , Daño por Reperfusión Miocárdica/prevención & control , Sustancias Protectoras/administración & dosificación , Compuestos Alílicos/administración & dosificación , Compuestos Alílicos/uso terapéutico , Animales , Antioxidantes/administración & dosificación , Antioxidantes/uso terapéutico , Isquemia Encefálica/etiología , Células Cultivadas , Sistemas de Liberación de Medicamentos , Sulfuro de Hidrógeno/uso terapéutico , Nanopartículas Magnéticas de Óxido de Hierro/química , Masculino , Ratones Endogámicos BALB C , Daño por Reperfusión Miocárdica/etiología , Sustancias Protectoras/uso terapéutico , Ratas Sprague-Dawley , Sulfuros/administración & dosificación , Sulfuros/uso terapéutico
5.
Int J Mol Sci ; 22(4)2021 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-33672103

RESUMEN

Hydrogen sulfide (H2S) has long been considered as a toxic gas, but as research progressed, the idea has been updated and it has now been shown to have potent protective effects at reasonable concentrations. H2S is an endogenous gas signaling molecule in mammals and is produced by specific enzymes in different cell types. An increasing number of studies indicate that H2S plays an important role in cardiovascular homeostasis, and in most cases, H2S has been reported to be downregulated in cardiovascular diseases (CVDs). Similarly, in preclinical studies, H2S has been shown to prevent CVDs and improve heart function after heart failure. Recently, many H2S donors have been synthesized and tested in cellular and animal models. Moreover, numerous molecular mechanisms have been proposed to demonstrate the effects of these donors. In this review, we will provide an update on the role of H2S in cardiovascular activities and its involvement in pathological states, with a special focus on the roles of exogenous H2S in cardiac protection.


Asunto(s)
Fármacos Cardiovasculares/farmacología , Enfermedades Cardiovasculares/tratamiento farmacológico , Sulfuro de Hidrógeno/administración & dosificación , Sulfuro de Hidrógeno/metabolismo , Administración por Inhalación , Animales , Enfermedades Cardiovasculares/metabolismo , Movimiento Celular , Humanos , Neovascularización Fisiológica/fisiología , Estrés Oxidativo/fisiología
6.
Pharmacol Res ; 161: 105121, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32798649

RESUMEN

Acute kidney injury (AKI) is a syndrome affecting most patients hospitalized due to kidney disease; it accounts for 15 % of patients hospitalized in intensive care units worldwide. AKI is mainly caused by ischemia and reperfusion (IR) injury, which temporarily obstructs the blood flow, increases inflammation processes and induces oxidative stress. AKI treatments available nowadays present notable disadvantages, mostly for patients with other comorbidities. Thus, it is important to investigate different approaches to help minimizing side effects such as the ones observed in patients subjected to the aforementioned treatments. Therefore, the aim of the current review is to highlight the potential of two endogenous gasotransmitters - hydrogen sulfide (H2S) and nitric oxide (NO) - and their crosstalk in AKI treatment. Both H2S and NO are endogenous signalling molecules involved in several physiological and pathophysiological processes, such as the ones taking place in the renal system. Overall, these molecules act by decreasing inflammation, controlling reactive oxygen species (ROS) concentrations, activating/inactivating pro-inflammatory cytokines, as well as promoting vasodilation and decreasing apoptosis, hypertrophy and autophagy. Since these gasotransmitters are found in gaseous state at environmental conditions, they can be directly applied by inhalation, or in combination with H2S and NO donors, which are compounds capable of releasing these molecules at biological conditions, thus enabling higher stability and slow release of NO and H2S. Moreover, the combination between these donor compounds and nanomaterials has the potential to enable targeted treatments, reduce side effects and increase the potential of H2S and NO. Finally, it is essential highlighting challenges to, and perspectives in, pharmacological applications of H2S and NO to treat AKI, mainly in combination with nanoparticulated delivery platforms.


Asunto(s)
Lesión Renal Aguda/tratamiento farmacológico , Gasotransmisores/administración & dosificación , Sulfuro de Hidrógeno/administración & dosificación , Donantes de Óxido Nítrico/uso terapéutico , Óxido Nítrico/administración & dosificación , Lesión Renal Aguda/metabolismo , Lesión Renal Aguda/patología , Administración por Inhalación , Animales , Portadores de Fármacos , Quimioterapia Combinada , Gasotransmisores/efectos adversos , Gasotransmisores/metabolismo , Humanos , Sulfuro de Hidrógeno/efectos adversos , Sulfuro de Hidrógeno/metabolismo , Nanomedicina , Nanoestructuras , Óxido Nítrico/efectos adversos , Óxido Nítrico/metabolismo , Donantes de Óxido Nítrico/efectos adversos , Donantes de Óxido Nítrico/metabolismo , Transducción de Señal
7.
Acta Pharmacol Sin ; 41(2): 218-228, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31316179

RESUMEN

Endothelial angiogenesis plays a vital role in recovery from chronic ischemic injuries. ZYZ-803 is a hybrid donor of hydrogen sulfide (H2S) and nitric oxide (NO). Previous studies showed that ZYZ-803 stimulated endothelial cell angiogenesis both in vitro and in vivo. In this study, we investigated whether the signal transducer and activator of transcription 3 (STAT3) and Ca2+/CaM-dependent protein kinase II (CaMKII) signaling was involved in ZYZ-803-induced angiogenesis. Treatment with ZYZ-803 (1 µM) significantly increased the phosphorylation of STAT3 (Tyr705) and CaMKII (Thr286) in human umbilical vein endothelial cells (HUVECs), these two effects had a similar time course. Pretreatment with WP1066 (STAT3 inhibitor) or KN93 (CAMKII inhibitor) blocked ZYZ-803-induced STAT3/CAMKII activation and significantly suppressed the proliferation and migration of HUVECs. In addition, pretreatment with the inhibitors significantly decreased ZYZ-803-induced tube formations along with the outgrowths of branch-like microvessels in aortic rings. In the mice with femoral artery ligation, administration of ZYZ-803 significantly increased the blood perfusion and vascular density in the hind limb, whereas co-administration of WP1066 or KN93 abrogated ZYZ-803-induced angiogenesis. By using STAT3 siRNA, we further explored the cross-talk between STAT3 and CaMKII in ZYZ-803-induced angiogenesis. We found that STAT3 knockdown suppressed ZYZ-803-induced HUVEC angiogenesis and affected CaMKII expression. ZYZ-803 treatment markedly enhanced the interaction between CaMKII and STAT3. ZYZ-803 treatment induced the nuclear translocation of STAT3. We demonstrated that both STAT3 and CaMKII functioned as positive regulators in ZYZ-803-induced endothelial angiogenesis and STAT3 was important in ZYZ-803-induced CaMKII activation, which highlights the beneficial role of ZYZ-803 in STAT3/CaMKII-related cardiovascular diseases.


Asunto(s)
Inductores de la Angiogénesis/farmacología , Sulfuro de Hidrógeno/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Óxido Nítrico/farmacología , Inductores de la Angiogénesis/administración & dosificación , Inductores de la Angiogénesis/química , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Humanos , Sulfuro de Hidrógeno/administración & dosificación , Sulfuro de Hidrógeno/química , Masculino , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico/administración & dosificación , Óxido Nítrico/química , Ratas , Ratas Sprague-Dawley , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal/efectos de los fármacos
8.
Int J Mol Sci ; 21(4)2020 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-32053981

RESUMEN

The social and economic impact of chronic inflammatory diseases, such as arthritis, explains the growing interest of the research in this field. The antioxidant and anti-inflammatory properties of the endogenous gasotransmitter hydrogen sulfide (H2S) were recently demonstrated in the context of different inflammatory diseases. In particular, H2S is able to suppress the production of pro-inflammatory mediations by lymphocytes and innate immunity cells. Considering these biological effects of H2S, a potential role in the treatment of inflammatory arthritis, such as rheumatoid arthritis (RA), can be postulated. However, despite the growing interest in H2S, more evidence is needed to understand the pathophysiology and the potential of H2S as a therapeutic agent. Within this review, we provide an overview on H2S biological effects, on its role in immune-mediated inflammatory diseases, on H2S releasing drugs, and on systems of tissue repair and regeneration that are currently under investigation for potential therapeutic applications in arthritic diseases.


Asunto(s)
Artritis/tratamiento farmacológico , Gasotransmisores/inmunología , Gasotransmisores/uso terapéutico , Sulfuro de Hidrógeno/inmunología , Sulfuro de Hidrógeno/uso terapéutico , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/inmunología , Antiinflamatorios no Esteroideos/uso terapéutico , Artritis/inmunología , Artritis Reumatoide/tratamiento farmacológico , Artritis Reumatoide/inmunología , Gasotransmisores/administración & dosificación , Humanos , Sulfuro de Hidrógeno/administración & dosificación , Inflamación/tratamiento farmacológico , Inflamación/inmunología , Estrés Oxidativo/efectos de los fármacos
9.
Int J Mol Sci ; 21(19)2020 Oct 08.
Artículo en Inglés | MEDLINE | ID: mdl-33050005

RESUMEN

Osteoarthritis (OA) is the most common articular chronic disease. However, its current treatment is limited and mostly symptomatic. Hydrogen sulfide (H2S) is an endogenous gas with recognized physiological activities. The purpose here was to evaluate the effects of the intraarticular administration of a slow-releasing H2S compound (GYY-4137) on an OA experimental model. OA was induced in Wistar rats by the transection of medial collateral ligament and the removal of the medial meniscus of the left joint. The animals were randomized into three groups: non-treated and intraarticularly injected with saline or GYY-4137. Joint destabilization induced articular thickening (≈5% increment), the loss of joint mobility and flexion (≈12-degree angle), and increased levels of pain (≈1.5 points on a scale of 0 to 3). Animals treated with GYY-4137 presented improved motor function of the joint, as well as lower pain levels (≈75% recovery). We also observed that cartilage deterioration was attenuated in the GYY-4137 group (≈30% compared with the saline group). Likewise, these animals showed a reduced presence of pro-inflammatory mediators (cyclooxygenase-2, inducible nitric oxide synthase, and metalloproteinase-13) and lower oxidative damage in the cartilage. The increment of the nuclear factor-erythroid 2-related factor 2 (Nrf-2) levels and Nrf-2-regulated gene expression (≈30%) in the GYY-4137 group seem to be underlying its chondroprotective effects. Our results suggest the beneficial impact of the intraarticular administration of H2S on experimental OA, showing a reduced cartilage destruction and oxidative damage, and supporting the use of slow H2S-producing molecules as a complementary treatment in OA.


Asunto(s)
Artralgia/tratamiento farmacológico , Sulfuro de Hidrógeno/administración & dosificación , Morfolinas/administración & dosificación , Compuestos Organotiofosforados/administración & dosificación , Osteoartritis/tratamiento farmacológico , Sustancias Protectoras/administración & dosificación , Animales , Cartílago Articular/metabolismo , Ciclooxigenasa 2/metabolismo , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Inyecciones Intraarticulares , Metaloproteinasa 13 de la Matriz/metabolismo , Actividad Motora/efectos de los fármacos , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Estrés Oxidativo/efectos de los fármacos , Ratas , Ratas Wistar , Prueba de Desempeño de Rotación con Aceleración Constante , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
10.
Int J Mol Sci ; 21(4)2020 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-32093102

RESUMEN

Decreased circulating levels of hydrogen sulfide (H2S) are associated with higher mortality following myocardial ischemia. This study aimed at determining the long-term dose-dependent effects of sodium hydrosulfide (NaSH) administration on myocardial ischemia-reperfusion (IR) injury. Male rats were divided into control and NaSH groups that were treated for 9 weeks with daily intraperitoneal injections of normal saline or NaSH (0.28, 0.56, 1.6, 2.8, and 5.6 mg/kg), respectively. At the end of the study, hearts from all rats were isolated and hemodynamic parameters were recorded during baseline and following IR. In isolated hearts, infarct size, oxidative stress indices as well as mRNA expression of H2S-, nitric oxide (NO)-producing enzymes, and inflammatory markers were measured. In heart tissue following IR, low doses of NaSH (0.28 and 0.56 mg/kg) had no effect, whereas an intermediate dose (1.6 mg/kg), improved recovery of hemodynamic parameters, decreased infarct size, and decreased oxidative stress. It also increased expression of cystathionine γ-lyase (CSE), Raf kinase inhibitor protein (RKIP), endothelial NO synthase (eNOS), and neuronal NOS (nNOS), as well as decreased expression of inducible NOS (iNOS) and nuclear factor kappa-B (NF-κB). At the high dose of 5.6 mg/kg, NaSH administration was associated with worse recovery of hemodynamic parameters and increased infarct size as well as increased oxidative stress. This dose also decreased expression of CSE, RKIP, and eNOS and increased expression of iNOS and NF-κB. In conclusion, chronic treatment with NaSH has a U-shaped concentration effect on IR injury in heart tissue. An intermediate dose was associated with higher CSE-derived H2S, lower iNOS-derived NO, lower oxidative stress, and inflammation in heart tissue following IR.


Asunto(s)
Sulfuro de Hidrógeno/administración & dosificación , Daño por Reperfusión Miocárdica/tratamiento farmacológico , FN-kappa B/metabolismo , Estrés Oxidativo/efectos de los fármacos , Proteínas de Unión a Fosfatidiletanolamina/metabolismo , Animales , Cistationina gamma-Liasa/genética , Cistationina gamma-Liasa/metabolismo , Hemodinámica/efectos de los fármacos , Sulfuro de Hidrógeno/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Masculino , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , FN-kappa B/genética , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo I/genética , Óxido Nítrico Sintasa de Tipo I/metabolismo , Óxido Nítrico Sintasa de Tipo III/genética , Óxido Nítrico Sintasa de Tipo III/metabolismo , Proteínas de Unión a Fosfatidiletanolamina/genética , Ratas , Ratas Wistar
11.
J Am Chem Soc ; 141(34): 13610-13618, 2019 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-31373809

RESUMEN

Hydrogen sulfide (H2S) is an important signaling molecule that provides protective activities in a variety of physiological and pathological processes. Among the different types of H2S donor compounds, thioamides have attracted attention due to prior conjugation to nonsteroidal anti-inflammatory drugs (NSAIDs) to access H2S-NSAID hybrids with significantly reduced toxicity, but the mechanism of H2S release from thioamides remains unclear. Herein, we reported the synthesis and evaluation of a class of thioamide-derived sulfenyl thiocarbamates (SulfenylTCMs) that function as a new class of H2S donors. These compounds are efficiently activated by cellular thiols to release carbonyl sulfide (COS), which is quickly converted to H2S by carbonic anhydrase (CA). In addition, through mechanistic investigations, we establish that COS-independent H2S release pathways are also operative. In contrast to the parent thioamide-based donors, the SulfenylTCMs exhibit excellent H2S releasing efficiencies of up to 90% and operate through mechanistically well-defined pathways. In addition, we demonstrate that the sulfenyl thiocarbamate group is readily attached to common NSAIDs, such as naproxen, to generate YZ-597 as an efficient H2S-NSAID hybrid, which we demonstrate releases H2S in cellular environments. Taken together, this new class of H2S donor motifs provides an important platform for new donor development.


Asunto(s)
Sulfuro de Hidrógeno/administración & dosificación , Compuestos de Sulfhidrilo/metabolismo , Óxidos de Azufre/administración & dosificación , Tiocarbamatos/síntesis química , Tiocarbamatos/farmacología , Antiinflamatorios no Esteroideos/síntesis química , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/farmacología , Anhidrasas Carbónicas/metabolismo , Ciclización , Células HeLa , Humanos , Sulfuro de Hidrógeno/química , Sulfuro de Hidrógeno/metabolismo , Naproxeno/análogos & derivados , Naproxeno/síntesis química , Naproxeno/farmacología , Óxidos de Azufre/química , Óxidos de Azufre/metabolismo , Tiocarbamatos/química
12.
Anal Chem ; 91(24): 15382-15386, 2019 12 17.
Artículo en Inglés | MEDLINE | ID: mdl-31741384

RESUMEN

Hydrogen sulfide (H2S) is dichotomous in nature as it is lethal at higher concentrations, but at lower concentrations it shows a more cytoprotective nature. Due to its involvement in many physiological processes, it has recently risen to significance as a gasotransmitter alongside carbon monoxide (CO) and nitric oxide (NO). Experimentation using H2S is thus important to generate in vitro and in vivo models for this ubiquitous gasotransmitter. However, laboratory studies involving H2S are challenging due to the special handling conditions required to work with such a toxic gas. The use of chemical donors in the cell culture also show temporally varying release profiles, which are not optimal when trying to control H2S in cell cultures lasting several days. Previously we applied microfluidics to deliver stable concentrations of H2S, and in this work, we demonstrate a novel experimental method incorporating feedback control to precisely deliver H2S that accounts for donor solution concentration decay.


Asunto(s)
Sulfuro de Hidrógeno/química , Animales , Técnicas de Cultivo de Célula , Relación Dosis-Respuesta a Droga , Humanos , Sulfuro de Hidrógeno/administración & dosificación , Sulfuro de Hidrógeno/toxicidad , Morfolinas , Compuestos Organotiofosforados , Impresión Tridimensional
13.
Int J Exp Pathol ; 100(5-6): 330-336, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31777145

RESUMEN

One of the most important causes of visual loss (blindness) is glaucoma, which occurs due to the degeneration of the ganglion cells in retina. It has been shown that hydrogen sulphide (H2 S) acts an antioxidant, neuroprotective and neuromodulator and provides protection against oxidative stress and apoptosis. This study aims to examine through which apoptotic pathway H2 S acts in experimental glaucoma model. Twenty-two male wistar albino rats were used in this study. Group 1 (n = 6, control group): Intravitreal saline was given in the third week without inducing ocular hypertension (OHT) with laser photocoagulation. Group 2 (n = 8): After the induction of OHT with laser photocoagulation, intravitreal saline was given in the third week. Group 3 (n = 8): After the induction of OHT with laser photocoagulation, intravitreal H2 S's donor sodium hydrosulphide (NaSH) 100 nmol/L was given in the third week. At the end of the 6th week, the eyes of the rats were sacrified under anaesthesia and extracted and then routine tissue follow-up was undertaken. Besides haematoxylin & eosin (H&E) staining, Bax, Bcl-2, p53 and caspase-3 activation were examined immunohistochemically in the retina and the cornea. This showed that ocular hypertension caused apoptosis through the intrinsic pathway, due to Bax and caspase-3 activation, in both retina and cornea, and that this led to DNA damage due to p53 activation. Also, we found that H2 S exposure in glaucoma distinctly suppressed Bax, caspase-3 and p53 activations in retina but that it has a limited effect on the cornea. According to these results, glaucoma caused apoptosis in the retina through intrinsic pathway, and the damage to the retina could be compensated partially by H2 S but would have limited on the cornea.


Asunto(s)
Apoptosis/efectos de los fármacos , Córnea/diagnóstico por imagen , Glaucoma/tratamiento farmacológico , Sulfuro de Hidrógeno/farmacología , Sustancias Protectoras/farmacología , Retina/efectos de los fármacos , Animales , Biomarcadores/metabolismo , Córnea/metabolismo , Córnea/fisiopatología , Glaucoma/metabolismo , Glaucoma/fisiopatología , Sulfuro de Hidrógeno/administración & dosificación , Sulfuro de Hidrógeno/uso terapéutico , Inyecciones Intravítreas , Masculino , Sustancias Protectoras/administración & dosificación , Sustancias Protectoras/uso terapéutico , Ratas , Ratas Wistar , Retina/metabolismo , Retina/fisiopatología
14.
Bioconjug Chem ; 30(3): 614-620, 2019 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-30609890

RESUMEN

H2S donors are currently emerging as promising therapeutic agents in a wide variety of pathologies, including tumors. Cancer cells are characterized by an enhanced uptake of sugars, such as glucose. Therefore, novel glycoconjugated H2S donors were synthesized so that high concentrations of H2S can be selectively achieved therein. Dithiolethione portions or isothiocyanate portions were selected for their well-known H2S-releasing properties in the presence of biological substrates. A synthetic procedure employing trichloroacetimidate glycosyl donors was applied to produce, in a stereoselective fashion, C1-glycoconjugates, whereas C6-glycoconjugates were obtained by a Mitsunobu-based transformation. The resulting molecules were then tested for their anticancer effects on human pancreas adenocarcinoma ascites metastasis cell line AsPC-1. The most potent inhibitors of cell viability (6aß and 7b) proved to release H2S inside the AsPC-1 cells and to alter the basal cell cycle.


Asunto(s)
Antineoplásicos/química , Antineoplásicos/farmacología , Glicoconjugados/química , Glicoconjugados/farmacología , Sulfuro de Hidrógeno/farmacología , Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/síntesis química , Línea Celular Tumoral , Glicoconjugados/síntesis química , Humanos , Sulfuro de Hidrógeno/administración & dosificación , Isotiocianatos/síntesis química , Isotiocianatos/química , Isotiocianatos/farmacología , Neoplasias Pancreáticas/tratamiento farmacológico , Estereoisomerismo , Tionas/síntesis química , Tionas/química , Tionas/farmacología , Neoplasias Pancreáticas
15.
Nitric Oxide ; 92: 26-33, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31401106

RESUMEN

Hepatic fibrosis is caused by chronic inflammation and characterized as the excessive accumulation of extracellular matrix (ECM) by activated hepatic stellate cells (HSCs). Gasotransmitters like NO and CO are known to modulate inflammation and fibrosis, however, little is known about the role of the gasotransmitter hydrogen sulfide (H2S) in liver fibrogenesis and stellate cell activation. Endogenous H2S is produced by the enzymes cystathionine ß-synthase (CBS), cystathionine γ-lyase (CTH) and 3-mercaptopyruvate sulfur transferase (MPST) [1]. The aim of this study was to elucidate the role of endogenously produced and/or exogenously administered H2S on rat hepatic stellate cell activation and fibrogenesis. Primary rat HSCs were culture-activated for 7 days and treated with different H2S releasing donors (slow releasing donor GYY4137, fast releasing donor NaHS) or inhibitors of the H2S producing enzymes CTH and CBS (DL-PAG, AOAA). The main message of our study is that mRNA and protein expression level of H2S synthesizing enzymes are low in HSCs compared to hepatocytes and Kupffer cells. However, H2S promotes hepatic stellate cell activation. This conclusion is based on the fact that production of H2S and mRNA and protein expression of its producing enzyme CTH are increased during hepatic stellate cell activation. Furthermore, exogenous H2S increased HSC proliferation while inhibitors of endogenous H2S production reduce proliferation and fibrotic makers of HSCs. The effect of H2S on stellate cell activation correlated with increased cellular bioenergetics. Our results indicate that the H2S generation in hepatic stellate cells is a target for anti-fibrotic intervention and that systemic interventions with H2S should take into account cell-specific effects of H2S.


Asunto(s)
Células Estrelladas Hepáticas/efectos de los fármacos , Sulfuro de Hidrógeno/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Células Estrelladas Hepáticas/metabolismo , Sulfuro de Hidrógeno/administración & dosificación , Sulfuro de Hidrógeno/análisis , Masculino , Ratas , Ratas Wistar
16.
Biol Pharm Bull ; 42(2): 273-279, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30713258

RESUMEN

Hydrogen sulfide (H2S) is an endogenous gaseous transmitter known to play an important role in biological functions. For the hepatic and intrahepatic targeting of H2S prodrug at the cellular level, we developed two types of sulfo-albumins, in which five sulfide groups (source of H2S) were covalently bound to succinylated (Suc) or galactosylated (Gal) bovine serum albumin (BSA). Sulfo-BSA-Suc and polyethylene glycol (PEG)-Sulfo-BSA-Gal, both released H2S in the 5 mM glutathione solution, but not in the plasma. Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal were taken up by RAW264.7 cells (mouse macrophage-like cells) and Hep G2 cells (human hepatocellular carcinoma cells), respectively, and H2S was released. These results indicate that Sulfo-BSA-Suc and PEG -Sulfo-BSA-Gal selectively released H2S intracellularly. In a biodistribution study, up to 80% of 111In-labeled Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal rapidly accumulated in the liver, 30 min after intravenous injection in mice. Furthermore, 111In-labeled Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal predominantly accumulated in liver nonparenchymal (endothelial cells and Kupffer cells) and parenchymal cells (hepatocytes), respectively. These findings suggest that targeted delivery of H2S prodrug to a specific type of liver cells was successfully achieved by bioconjugation.


Asunto(s)
Hepatocitos/metabolismo , Sulfuro de Hidrógeno/administración & dosificación , Hígado/metabolismo , Profármacos/administración & dosificación , Albúmina Sérica Bovina/administración & dosificación , Animales , Células Hep G2 , Hepatocitos/efectos de los fármacos , Humanos , Sulfuro de Hidrógeno/química , Hígado/efectos de los fármacos , Masculino , Ratones , Profármacos/química , Células RAW 264.7 , Albúmina Sérica/administración & dosificación , Albúmina Sérica/química , Albúmina Sérica Bovina/química
17.
Can J Physiol Pharmacol ; 97(1): 47-54, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30521368

RESUMEN

Hydrogen sulfide (H2S) is found in both the plasma and synovial fluid of patients with gonarthrosis. In the present study, we investigated whether intra-articular injection of sodium hydrosulfide (NaSH) (1 mM, 30 µL), a H2S donor, might affect gonarthrosis in rats. Gonarthrosis was induced surgically in the left knees of rats and left for 6 weeks for the development of disease. Then, intra-articular injections of NaSH or methylprednisolone (1 mg/kg, 30 µL) were administered to rats. Half of each group was sacrificed at the end of the first day and the other half was sacrificed at the end of 4 weeks to evaluate early and later effects of injections on gonarthrosis. The injury induced by anterior cruciate ligament resection and medial meniscectomy in rats caused the development of gonarthrosis. As the duration lengthened after gonarthrosis induction, the progression of the disease continued. According to the modified Mankin Scoring System, intra-articular injection of NaSH histopathologically slowed the progression of gonarthrosis, whereas methylprednisolone was ineffective. In addition, NaSH decreased apoptosis in rat knees with gonarthrosis. Each treatment did not cause injury to healthy knees. Our results lead to the consideration that intra-articular NaSH administration may be effective in the progression of gonarthrosis.


Asunto(s)
Progresión de la Enfermedad , Sulfuro de Hidrógeno/administración & dosificación , Osteoartritis de la Rodilla/diagnóstico por imagen , Osteoartritis de la Rodilla/prevención & control , Animales , Gasotransmisores/administración & dosificación , Inyecciones Intraarticulares , Masculino , Osteoartritis de la Rodilla/patología , Ratas , Ratas Wistar
18.
Ecotoxicol Environ Saf ; 182: 109459, 2019 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-31344591

RESUMEN

The utilization of forages grown on metal-contaminated soil can increase the risk of heavy metals entering the food chain and affecting human health because of elevated toxic metal concentrations. Meanwhile, hydrogen sulfide (H2S) and nitric oxide (NO) as signaling molecules are known to promote plant growth in metal-contaminated soils. However, the regulatory mechanisms of such molecules in plant physiology and soil biochemistry have not been well-documented. Hence, we investigate the role of the exogenous application of H2S and NO on alfalfa growth in lead/cadmium (Pb/Cd)-contaminated soil. Our results indicate that the signaling molecules increase the alfalfa chlorophyll and biomass content and improve alfalfa growth. Further, H2S and NO reduce the translocation and bioconcentration factors of Pb and Cd, potentially reducing the risk of heavy metals entering the food chain. These signaling molecules reduce metal-induced oxidative damage to alfalfa by mitigating reactive oxygen species accumulation and increasing antioxidant enzyme activities. Their exogenous application increases soil enzymatic activities, particularly of catalase and polyphenol oxidase, without significantly changing the composition and structure of rhizosphere bacterial communities. Interestingly, H2S addition enriches the abundance of plant-growth-promoting rhizobacteria in soil, including Nocardioides, Rhizobium, and Glycomyces. H2S is more effective than NO in improving alfalfa growth and reducing heavy-metal contamination of the food chain. These results provide new insights into the exogenous application of signaling molecules in alleviating metal-induced phytotoxicity, including an efficient strategy for the safe use of forages.


Asunto(s)
Cadmio/análisis , Sulfuro de Hidrógeno/farmacología , Plomo/análisis , Medicago sativa/efectos de los fármacos , Óxido Nítrico/farmacología , Contaminantes del Suelo/análisis , Riego Agrícola , Biomasa , Cadmio/toxicidad , Fumigación , Sulfuro de Hidrógeno/administración & dosificación , Plomo/toxicidad , Medicago sativa/química , Medicago sativa/crecimiento & desarrollo , Microbiota/efectos de los fármacos , Óxido Nítrico/administración & dosificación , Contaminantes del Suelo/toxicidad
19.
Med Res Rev ; 38(1): 57-100, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28240384

RESUMEN

Hydrogen sulfide (H2 S), together with nitric oxide (NO) and carbon monoxide (CO), belongs to the gasotransmitter family and plays important roles in mammals as a signaling molecule. Many studies have also shown the various therapeutic effects of H2 S, which include protection against myocardial ischemia injury, cytoprotection against oxidative stress, mediation of neurotransmission, inhibition of insulin signaling, regulation of inflammation, inhibition of the hypoxia-inducible pathway, and dilation of blood vessels. One major challenge in the development of H2 S-based therapeutics is its delivery. In this manuscript, we assess the various drug delivery strategies in the context of being used research tools and eventual developability as therapeutic agents.


Asunto(s)
Sulfuro de Hidrógeno/administración & dosificación , Sulfuro de Hidrógeno/metabolismo , Animales , Sistemas de Liberación de Medicamentos , Humanos , Sulfuro de Hidrógeno/farmacocinética , Profármacos/administración & dosificación , Profármacos/farmacocinética
20.
J Cell Physiol ; 233(12): 9786-9798, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30078216

RESUMEN

BACKGROUND: Previous studies have suggested that exogenous hydrogen sulfide can alleviate the development of diabetic cardiomyopathy (DCM) by inhibiting oxidative stress, inflammation, and apoptosis. However, the underlying mechanism is not fully understood. Nuclear expression and function of the transcription factor Forkhead box protein O (FoxO1) have been associated with cardiovascular diseases, and thus, the importance of FoxO1 in DCM has gained increasing attention. This study was designed to investigate the interactions between hydrogen sulfide (H2 S) and nuclear FoxO1 in DCM. METHODS: Diabetes was induced in adult male C57BL/6J mice by intraperitoneal injection of streptozotocin and was treated with H2 S donor sodium hydrosulfide for 12 weeks. The H9C2 cardiomyoblast cell line and neonatal rat cardiomyocytes (NRCMs) were treated with the slow-releasing H2 S donor GYY4137 before high-glucose (HG) exposure with or without pretreatment with the Akt inhibitor MK-2206 2HCl. Changes in FoxO1 protein phosphorylation and subcellular localization were determined in H9C2 cells, NRCMs, and cardiac tissues from normal and diabetic mice. Cardiac structure and function in the diabetic mice were evaluated by echocardiography and histological analysis and compared with those in control animals. RESULTS: The echocardiographic and histopathological data indicated that exogenous H2 S improved cardiac function and attenuated cardiac hypertrophy and myocardial fibrosis in diabetic mice. H2 S also improved HG-induced oxidative stress and apoptosis in cardiac tissue and NRCMs. In addition, H2 S induced FoxO1 phosphorylation and nuclear exclusion in vitro and in vivo, and this function was not inhibited by MK-2206 2HCl. Alanine substitution mutation of three sites in FoxO1-enhanced FoxO1 transcriptional activity, and subsequent treatment with exogenous H2 S could not prevent HG-induced nuclear retention. CONCLUSIONS: Our data indicate that H2 S is a novel regulator of FoxO1 in cardiac cells and provide evidence supporting the potential of H2 S in inhibiting the progression of DCM.


Asunto(s)
Cardiomiopatías Diabéticas/tratamiento farmacológico , Proteína Forkhead Box O1/genética , Sulfuro de Hidrógeno/administración & dosificación , Inflamación/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/fisiopatología , Modelos Animales de Enfermedad , Corazón/efectos de los fármacos , Corazón/fisiopatología , Humanos , Inflamación/genética , Inflamación/fisiopatología , Ratones , Ratones Endogámicos NOD , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Ratas , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA