Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 473
Filter
1.
Neuroscience ; 555: 213-221, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39089569

ABSTRACT

Anxiety disorders are prevalent chronic psychological disease with complex pathogenic mechanisms. Current anxiolytics have limited efficacy and numerous side effects in many anxiety patients, highlighting the urgent need for new therapies. Recent research has been focusing on nutritional supplements, particularly amino acids, as potential therapies for anxiety disorders. Among these, L-Cysteine plays a crucial role in various biological processes. L-Cysteine exhibits antioxidant properties that can enhance the antioxidant functions of the central nervous system (CNS). Furthermore, metabolites of L-cysteine, such as glutathione and hydrogen sulfide have been shown to alleviate anxiety through distinct molecular mechanisms. Long-term administration of L-Cysteine has anxiolytic, antidepressant, and memory-improving effects. L-Cysteine depletion can lead to increased oxidative stress in the brain. This review delves into the potential mechanisms of L-Cysteine and its main products, glutathione (GSH) and hydrogen sulfide (H2S) in the management of anxiety and related diseases.


Subject(s)
Anxiety Disorders , Cysteine , Dietary Supplements , Cysteine/pharmacology , Humans , Anxiety Disorders/drug therapy , Animals , Anti-Anxiety Agents/pharmacology , Anti-Anxiety Agents/therapeutic use , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Glutathione/metabolism , Antioxidants/pharmacology , Antioxidants/administration & dosage , Oxidative Stress/drug effects
2.
Biomolecules ; 14(7)2024 Jun 22.
Article in English | MEDLINE | ID: mdl-39062455

ABSTRACT

Ischemia-reperfusion (I/R) injury, a prevalent pathological condition in medical practice, presents significant treatment challenges. Hydrogen sulfide (H2S), acknowledged as the third gas signaling molecule, profoundly impacts various physiological and pathophysiological processes. Extensive research has demonstrated that H2S can mitigate I/R damage across multiple organs and tissues. This review investigates the protective effects of H2S in preventing I/R damage in the heart, brain, liver, kidney, intestines, lungs, stomach, spinal cord, testes, eyes, and other tissues. H2S provides protection against I/R damage by alleviating inflammation and endoplasmic reticulum stress; inhibiting apoptosis, oxidative stress, and mitochondrial autophagy and dysfunction; and regulating microRNAs. Significant advancements in understanding the mechanisms by which H2S reduces I/R damage have led to the development and synthesis of H2S-releasing agents such as diallyl trisulfide-loaded mesoporous silica nanoparticles (DATS-MSN), AP39, zofenopril, and ATB-344, offering a new therapeutic avenue for I/R injury.


Subject(s)
Hydrogen Sulfide , Reperfusion Injury , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/pharmacology , Humans , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism , Animals , Oxidative Stress/drug effects , Apoptosis/drug effects , Endoplasmic Reticulum Stress/drug effects
3.
Neuromolecular Med ; 26(1): 26, 2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38907170

ABSTRACT

Spinal cord injury (SCI) causes irreversible cell loss and neurological dysfunctions. Presently, there is no an effective clinical treatment for SCI. It can be the only intervention measure by relieving the symptoms of patients such as pain and fever. Free radical-induced damage is one of the validated mechanisms in the complex secondary injury following primary SCI. Hydrogen sulfide (H2S) as an antioxidant can effectively scavenge free radicals, protect neurons, and improve SCI by inhibiting the p38MAPK/mTOR/NF-κB signaling pathway. In this report, we analyze the pathological mechanism of SCI, the role of free radical-mediated the p38MAPK/mTOR/NF-κB signaling pathway in SCI, and the role of H2S in scavenging free radicals and improving SCI.


Subject(s)
Free Radical Scavengers , Hydrogen Sulfide , NF-kappa B , Signal Transduction , Spinal Cord Injuries , TOR Serine-Threonine Kinases , p38 Mitogen-Activated Protein Kinases , Spinal Cord Injuries/drug therapy , Spinal Cord Injuries/metabolism , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , NF-kappa B/metabolism , Animals , Free Radical Scavengers/therapeutic use , Free Radical Scavengers/pharmacology , Signal Transduction/drug effects , Rats , Mice , Free Radicals/metabolism , Antioxidants/therapeutic use , Antioxidants/pharmacology , Spinal Cord/drug effects , Spinal Cord/metabolism , Humans
4.
Biomolecules ; 14(5)2024 Apr 30.
Article in English | MEDLINE | ID: mdl-38785947

ABSTRACT

Hydrogen sulfide (H2S), previously regarded as a toxic exhaust and atmospheric pollutant, has emerged as the third gaseous signaling molecule following nitric oxide (NO) and carbon monoxide (CO). Recent research has revealed significant biological effects of H2S in a variety of systems, such as the nervous, cardiovascular, and digestive systems. Additionally, H2S has been found to impact reproductive system function and may have therapeutic implications for reproductive disorders. This paper explores the relationship between H2S and male reproductive disorders, specifically erectile dysfunction, prostate cancer, male infertility, and testicular damage. Additionally, it examines the impact of H2S regulation on the pathophysiology of the female reproductive system, including improvements in preterm birth, endometriosis, pre-eclampsia, fetal growth restriction, unexplained recurrent spontaneous abortion, placental oxidative damage, embryo implantation, recovery of myometrium post-delivery, and ovulation. The study delves into the regulatory functions of H2S within the reproductive systems of both genders, including its impact on the NO/cGMP pathway, the activation of K+ channels, and the relaxation mechanism of the spongy smooth muscle through the ROCK pathway, aiming to broaden the scope of potential therapeutic strategies for treating reproductive system disorders in clinical settings.


Subject(s)
Hydrogen Sulfide , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/therapeutic use , Humans , Female , Male , Pregnancy , Animals , Nitric Oxide/metabolism , Reproduction/drug effects
5.
J Nanobiotechnology ; 22(1): 277, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38783332

ABSTRACT

Spinal Cord Injury (SCI) is a condition characterized by complete or incomplete motor and sensory impairment, as well as dysfunction of the autonomic nervous system, caused by factors such as trauma, tumors, or inflammation. Current treatment methods primarily include traditional approaches like spinal canal decompression and internal fixation surgery, steroid pulse therapy, as well as newer techniques such as stem cell transplantation and brain-spinal cord interfaces. However, the above methods have limited efficacy in promoting axonal and neuronal regeneration. The challenge in medical research today lies in promoting spinal cord neuron regeneration and regulating the disrupted microenvironment of the spinal cord. Studies have shown that gas molecular therapy is increasingly used in medical research, with gasotransmitters such as hydrogen sulfide, nitric oxide, carbon monoxide, oxygen, and hydrogen exhibiting neuroprotective effects in central nervous system diseases. The gas molecular protect against neuronal death and reshape the microenvironment of spinal cord injuries by regulating oxidative, inflammatory and apoptotic processes. At present, gas therapy mainly relies on inhalation for systemic administration, which cannot effectively enrich and release gas in the spinal cord injury area, making it difficult to achieve the expected effects. With the rapid development of nanotechnology, the use of nanocarriers to achieve targeted enrichment and precise control release of gas at Sites of injury has become one of the emerging research directions in SCI. It has shown promising therapeutic effects in preclinical studies and is expected to bring new hope and opportunities for the treatment of SCI. In this review, we will briefly outline the therapeutic effects and research progress of gasotransmitters and nanogas in the treatment of SCI.


Subject(s)
Gasotransmitters , Spinal Cord Injuries , Spinal Cord Injuries/therapy , Humans , Animals , Gasotransmitters/therapeutic use , Gasotransmitters/metabolism , Nitric Oxide/metabolism , Neuroprotective Agents/therapeutic use , Neuroprotective Agents/pharmacology , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Carbon Monoxide/metabolism , Carbon Monoxide/therapeutic use , Oxygen/metabolism , Spinal Cord , Hydrogen/therapeutic use , Hydrogen/pharmacology
6.
Pharmacol Res ; 203: 107180, 2024 May.
Article in English | MEDLINE | ID: mdl-38599468

ABSTRACT

Primary mitochondrial diseases (PMD) are amongst the most common inborn errors of metabolism causing fatal outcomes within the first decade of life. With marked heterogeneity in both inheritance patterns and physiological manifestations, these conditions present distinct challenges for targeted drug therapy, where effective therapeutic countermeasures remain elusive within the clinic. Hydrogen sulfide (H2S)-based therapeutics may offer a new option for patient treatment, having been proposed as a conserved mitochondrial substrate and post-translational regulator across species, displaying therapeutic effects in age-related mitochondrial dysfunction and neurodegenerative models of mitochondrial disease. H2S can stimulate mitochondrial respiration at sites downstream of common PMD-defective subunits, augmenting energy production, mitochondrial function and reducing cell death. Here, we highlight the primary signalling mechanisms of H2S in mitochondria relevant for PMD and outline key cytoprotective proteins/pathways amenable to post-translational restoration via H2S-mediated persulfidation. The mechanisms proposed here, combined with the advent of potent mitochondria-targeted sulfide delivery molecules, could provide a framework for H2S as a countermeasure for PMD disease progression.


Subject(s)
Hydrogen Sulfide , Mitochondria , Mitochondrial Diseases , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/therapeutic use , Humans , Animals , Mitochondrial Diseases/drug therapy , Mitochondrial Diseases/metabolism , Mitochondria/metabolism , Mitochondria/drug effects , Dietary Supplements , Signal Transduction/drug effects
7.
Exp Neurol ; 376: 114758, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38513970

ABSTRACT

Impaired long-term memory, a complication of traumatic stress including hemorrhage shock and resuscitation (HSR), has been reported to be associated with multiple neurodegenerations. The ventral tegmental area (VTA) participates in both learned appetitive and aversive behaviors. In addition to being prospective targets for the therapy of addiction, depression, and other stress-related diseases, VTA glutamatergic neurons are becoming more widely acknowledged as powerful regulators of reward and aversion. This study revealed that HSR exposure induces memory impairments and decreases the activation in glutamatergic neurons, and decreased ß power in the VTA. We also found that optogenetic activation of glutamatergic neurons in the VTA mitigated HSR-induced memory impairments, and restored ß power. Moreover, hydrogen sulfide (H2S), a gasotransmitter with pleiotropic roles, has neuroprotective functions at physiological concentrations. In vivo, H2S administration improved HSR-induced memory deficits, elevated c-fos-positive vesicular glutamate transporters (Vglut2) neurons, increased ß power, and restored the balance of γ-aminobutyric acid (GABA) and glutamate in the VTA. This work suggests that glutamatergic neuron stimulation via optogenetic assay and exogenous H2S may be useful therapeutic approaches for improving memory deficits following HSR.


Subject(s)
Disease Models, Animal , Glutamic Acid , Hydrogen Sulfide , Memory Disorders , Mice, Inbred C57BL , Neurons , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Mice , Memory Disorders/drug therapy , Memory Disorders/etiology , Memory Disorders/therapy , Male , Neurons/drug effects , Neurons/metabolism , Glutamic Acid/metabolism , Glutamic Acid/toxicity , Shock, Hemorrhagic , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism , Optogenetics/methods
8.
ACS Nano ; 18(14): 9871-9885, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38545939

ABSTRACT

As an ideal drug carrier, it should possess high drug loading and encapsulation efficiency and precise drug targeting release. Herein, we utilized a template-guided self-weaving technology of phase-separated silk fibroin (SF) in reverse microemulsion (RME) to fabricate a kind of hyaluronic acid (HA) coated SF nanocage (HA-gNCs) for drug delivery of cancer immunotherapy. Due to the hollow structure, HA-gNCs were capable of simultaneous encapsulation of the anti-inflammatory drug betamethasone phosphate (BetP) and the immune checkpoint blockade (ICB) agent PD-L1 antibody (αPD-L1) efficiently. Another point worth noting was that the thiocarbonate cross-linkers used to strengthen the SF shell of HA-gNCs could be quickly broken by overexpressed glutathione (GSH) to reach responsive drug release inside tumor tissues accompanied by hydrogen sulfide (H2S) production in one step. The synergistic effect of released BetP and generated H2S guaranteed chronological modulation of the immunosuppressive tumor microenvironment (ITME) to amplify the therapeutic effect of αPD-L1 for the growth, metastasis, and recurrence of tumors. This study highlighted the exceptional prospect of HA-gNCs as a self-assistance platform for cancer drug delivery.


Subject(s)
Antineoplastic Agents , Hydrogen Sulfide , Nanoparticles , Neoplasms , Humans , Hydrogen Sulfide/therapeutic use , Antineoplastic Agents/therapeutic use , Drug Delivery Systems/methods , Neoplasms/drug therapy , Neoplasms/pathology , Glutathione , Immunotherapy , Tumor Microenvironment , Cell Line, Tumor , Nanoparticles/chemistry
9.
J Neuroimmunol ; 389: 578324, 2024 04 15.
Article in English | MEDLINE | ID: mdl-38422691

ABSTRACT

Sickness behavior reflects a state of altered physiology and central nervous system function that occurs during systemic infection or inflammation, serving as an adaptive response to illness. This study aims to elucidate the role of hydrogen sulfide (H2S) in regulating sickness behavior and neuroinflammatory responses in a rat model of systemic inflammation. Adult male Wistar rats were treated with lipopolysaccharide (LPS) to induce sickness behavior. Intracerebroventricular (i.c.v.) pretreatments included aminooxyacetic acid (AOAA), an inhibitor of H2S synthesis, and sodium sulfide (NaHS), an H2S donor. Behavioral assays were conducted, along with the assessment of astrocyte activation, as indicated by GFAP expression in the hypothalamus. Pretreatment with NaHS mitigated LPS-induced behavioral changes, including hypophagia, social and exploratory deficits, without affecting peripheral cytokine levels, indicating a central modulatory effect. AOAA, conversely, accentuated certain behavioral responses, suggesting a complex role of endogenous H2S in sickness behavior. These findings were reinforced by a lack of effect on plasma interleukin levels but significant reduction in GFAP expression. Our findings support the central role of H2S in modulating neuroinflammation and sickness behavior, highlighting the therapeutic potential of targeting H2S signaling in neuroinflammatory conditions.


Subject(s)
Hydrogen Sulfide , Sulfides , Rats , Male , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Lipopolysaccharides/toxicity , Illness Behavior , Rats, Wistar , Inflammation/chemically induced , Inflammation/drug therapy , Inflammation/metabolism , Aminooxyacetic Acid/pharmacology , Neurotransmitter Agents
10.
Eur J Pharm Sci ; 195: 106721, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38331005

ABSTRACT

Hydrogen sulfide (H2S), a gasotransmitter, plays a crucial role in vasorelaxation, anti-inflammatory processes and mitigating myocardial ischemia/reperfusion-induced injury by regulating various signaling processes. We designed a water soluble H2S-releasing ascorbic acid derivative, BM-164, to combine the beneficial cardiovascular and anti-inflammatory effects of H2S with the excellent water solubility and antioxidant properties of ascorbic acid. DPPH antioxidant assay revealed that the antioxidant activity of BM-164 in the presence of a myocardial tissue homogenate (extract) increased continuously over the 120 min test interval due to the continuous release of H2S from BM-164. The cytotoxicity of BM-164 was tested by MTT assay on H9c2 cells, which resulted in no cytotoxic effect at concentrations of 10 to 30 µM. The possible beneficial effects of BM-164 (30 µM) was examined in isolated 'Langendorff' rat hearts. The incidence of ventricular fibrillation (VF) was significantly reduced from its control value of 79 % to 31 % in the BM-164 treated group, and the infarct size was also diminished from the control value of 28 % to 14 % in the BM-164 treated group. However, coronary flow (CF) and heart rate (HR) values in the BM-164 treated group did not show significantly different levels in comparison with the drug-free control, although a non-significant recovery in both CF and HR was observed at each time point. We attempted to reveal the mechanism of action of BM-164, focusing on the processes of autophagy and apoptosis. The expression of key autophagic and apoptotic markers in isolated rat hearts were detected by Western blot analysis. All the examined autophagy-related proteins showed increased expression levels in the BM-164 treated group in comparison to the drug-free control and/or ascorbic acid treated groups, while the changes in the expression of apoptotic markers were not obvious. In conclusion, the designed water soluble H2S releasing ascorbic acid derivative, BM-164, showed better cardiac protection against ischemia/reperfusion-induced injury compared to the untreated and ascorbic acid treated hearts, respectively.


Subject(s)
Hydrogen Sulfide , Myocardial Reperfusion Injury , Rats , Animals , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Antioxidants/pharmacology , Rats, Wistar , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Ischemia , Anti-Inflammatory Agents/therapeutic use , Water , Reperfusion , Hydrogen Sulfide/metabolism , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use
11.
Cells ; 13(2)2024 01 15.
Article in English | MEDLINE | ID: mdl-38247849

ABSTRACT

Hydrogen sulfide (H2S) belongs to the family of gasotransmitters and can modulate a myriad of biological signaling pathways. Among others, its cardioprotective effects, through antioxidant, anti-inflammatory, anti-fibrotic, and proangiogenic activities, are well-documented in experimental studies. Cardiorespiratory failure, predominantly cardiomyopathy, is a life-threatening complication that is the number one cause of death in patients with Duchenne muscular dystrophy (DMD). Although recent data suggest the role of H2S in ameliorating muscle wasting in murine and Caenorhabditis elegans models of DMD, possible cardioprotective effects have not yet been addressed. In this review, we summarize the current understanding of the role of H2S in animal models of cardiac dysfunctions and cardiac cells. We highlight that DMD may be amenable to H2S supplementation, and we suggest H2S as a possible factor regulating DMD-associated cardiomyopathy.


Subject(s)
Cardiomyopathies , Hydrogen Sulfide , Muscular Dystrophy, Duchenne , Humans , Animals , Mice , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Muscular Dystrophy, Duchenne/drug therapy , Cardiomyopathies/drug therapy , Cardiomyopathies/etiology , Antioxidants , Caenorhabditis elegans
12.
Eur J Pharmacol ; 966: 176344, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38280462

ABSTRACT

Diabetes-associated cognitive dysfunction (DACD) is a complication of diabetes mellitus that leads to an increased risk of cognitive impairment and dementia. However, the molecular mechanism underlying DACD has not been elucidated, and a promising therapy for this disease remains to be established. Hydrogen sulfide (H2S), a significant antioxidative and anti-inflammatory gasotransmitter, has emerged as a neuroprotective agent. In this study, we investigated the protective effects of H2S on DACD in a streptozotocin (STZ)-induced diabetic rat model. We applied the Morris water maze to evaluate spatial learning and memory abilities. We used Western blotting and immunohistochemical staining to investigate the expression of the Nrf-2/HO-1 axis and the NLRP3 inflammasome. After NaHS (H2S donor) administration, diabetic rats exhibited improved spatial learning and memory retrieval abilities in the Morris water maze. In STZ-induced diabetic rats, the protein expression levels of the Nrf-2/HO-1 axis, the NLRP3 inflammasome and subsequent inflammatory cytokines in the hippocampal region were elevated compared to those in control rats. Exogenous H2S triggered Nrf-2/HO-1 antioxidant activity and inhibited NLRP3 inflammasome activation and proinflammatory cytokine expression. These findings suggested that exogenous H2S has neuroprotective effects by modulating the Nrf-2/HO-1 axis and the NLRP3 inflammasome pathway, which were found to be associated with DACD. H2S treatment may be a promising therapeutic strategy for preventing the progression of tissue damage caused by DACD.


Subject(s)
Cognitive Dysfunction , Diabetes Mellitus, Experimental , Hydrogen Sulfide , Rats , Animals , Inflammasomes/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/metabolism , Cognitive Dysfunction/drug therapy , Cognitive Dysfunction/etiology
13.
Cell Commun Signal ; 22(1): 33, 2024 01 12.
Article in English | MEDLINE | ID: mdl-38217003

ABSTRACT

BACKGROUND: Spinal cord ischemia‒reperfusion injury (SCIRI) can lead to paraplegia, which leads to permanent motor function loss. It is a disastrous complication of surgery and causes tremendous socioeconomic burden. However, effective treatments for SCIRI are still lacking. PANoptosis consists of three kinds of programmed cell death, pyroptosis, apoptosis, and necroptosis, and may contribute to ischemia‒reperfusion-induced neuron death. Previous studies have demonstrated that hydrogen sulfide (H2S) exerts a neuroprotective effect in many neurodegenerative diseases. However, whether H2S is anti-PANoptosis and neuroprotective in the progression of acute SCIRI remains unclear. Thus, in this study we aimed to explore the role of H2S in SCIRI and its underlying mechanisms. METHODS: Measurements of lower limb function, neuronal activity, microglia/macrophage function histopathological examinations, and biochemical levels were performed to examine the efficacy of H2S and to further demonstrate the mechanism and treatment of SCIRI. RESULTS: The results showed that GYY4137 (a slow-releasing H2S donor) treatment attenuated the loss of Nissl bodies after SCIRI and improved the BBB score. Additionally, the number of TUNEL-positive and cleaved caspase-3-positive cells was decreased, and the upregulation of expression of cleaved caspase-8, cleaved caspase-3, Bax, and Bad and downregulation of Bcl-2 expression were reversed after GYY4137 administration. Meanwhile, both the expression and activation of p-MLKL, p-RIP1, and p-RIP3, along with the number of PI-positive and RIP3-positive neurons, were decreased in GYY4137-treated rats. Furthermore, GYY4137 administration reduced the expression of NLRP3, cleaved caspase-1 and cleaved GSDMD, decreased the colocalization NeuN/NLRP3 and Iba1/interleukin-1ß-expressing cells, and inhibited proinflammatory factors and microglia/macrophage polarization. CONCLUSIONS: H2S ameliorated spinal cord neuron loss, prevented motor dysfunction after SCIRI, and exerted a neuroprotective effect via the inhibition of PANoptosis and overactivated microglia-mediated neuroinflammation in SCIRI.


Subject(s)
Hydrogen Sulfide , Morpholines , Neuroprotective Agents , Organothiophosphorus Compounds , Reperfusion Injury , Rats , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Caspase 3/metabolism , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Spinal Cord/metabolism , Spinal Cord/pathology , Apoptosis , Reperfusion Injury/complications , Reperfusion Injury/drug therapy , Reperfusion Injury/metabolism
14.
Antioxid Redox Signal ; 40(1-3): 168-185, 2024 01.
Article in English | MEDLINE | ID: mdl-37917094

ABSTRACT

Significance: Gasotransmitters are small gas molecules that are endogenously generated and have well-defined physiological functions. The most well-defined gasotransmitters currently are nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), while other potent gasotransmitters include ammonia, methane, cyanide, hydrogen gas, and sulfur dioxide. Gasotransmitters play a role in various respiratory diseases such as asthma, chronic obstructive pulmonary disease, obstructive sleep apnea, lung infection, bronchiectasis, cystic fibrosis, primary ciliary dyskinesia, and COVID-19. Recent Advances: Gasotransmitters can act as biomarkers that facilitate disease diagnosis, indicate disease severity, predict disease exacerbation, and evaluate disease outcomes. They also have cell-protective properties, and many studies have been conducted to explore their pharmacological applications. Innovative drug donors and drug delivery methods have been invented to amplify their therapeutic effects. Critical Issues: In this article, we briefly reviewed the physiological and pathophysiological functions of some gasotransmitters in the respiratory system, the progress in detecting exhaled gasotransmitters, as well as innovative drugs derived from these molecules. Future Directions: The current challenge for gasotransmitter research includes further exploring their physiological and pathological functions, clarifying their complicated interactions, exploring suitable drug donors and delivery devices, and characterizing new members of gasotransmitters. Antioxid. Redox Signal. 40, 168-185.


Subject(s)
Gasotransmitters , Hydrogen Sulfide , Respiratory Tract Diseases , Humans , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/pharmacology , Nitric Oxide , Carbon Monoxide , Respiratory Tract Diseases/diagnosis , Respiratory Tract Diseases/drug therapy
15.
Naunyn Schmiedebergs Arch Pharmacol ; 397(3): 1853-1864, 2024 03.
Article in English | MEDLINE | ID: mdl-37773523

ABSTRACT

H2S-donors are cardioprotective in ischemia/reperfusion (I/R) injury. Some H2S-donors exert their beneficial effects in a nitric oxide (NO)-dependent manner, while others act using NO-independent pathways. The aims of the present study were to (i) evaluate whether H2S-donors with distinct pharmacodynamic properties act synergistically in I/R injury and (ii) determine if H2S-donors remain cardioprotective in obese mice. C57BL/6 mice were subjected to 30 min of ischemia followed by 120 min of reperfusion. Donors were administered intravenously at the end of ischemia (Na2S: 1 µmol/kg, GYY4137: 25 µmol/kg, AP39: 0,25 µmol/kg), while the 3-mercaptopyruvate sulfurtransferase (10 mg/kg) inhibitor was given intraperitonially 1 h prior to ischemia. Infarct size was estimated by 2,3,5-triphenyltetrazolium staining, while the area at risk was calculated using Evans blue. All three donors reduced infarct size when administered as a sole treatment. Co-administration of Na2S/GYY4137, as well as Na2S/AP39 reduced further the I/R injury, beyond what was observed with each individual donor. Since inhibition of the H2S-producing enzyme 3-mercaptopyruvate sulfurtransferase is known to reduce infarct size, we co-administered C3 with Na2S to determine possible additive effects between the two agents. In this case, combination of C3 with Na2S did not yield superior results compared to the individual treatments. Similarly, to what was observed in healthy mice, administration of a H2S-donor (Na2S or AP39) reduced I/R injury in mice rendered obese by consumption of a high fat diet. We conclude that combining a NO-dependent with a NO-independent H2S-donor leads to enhanced cardioprotection and that H2S-donors remain effective in obese animals.


Subject(s)
Hydrogen Sulfide , Morpholines , Organothiophosphorus Compounds , Reperfusion Injury , Mice , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/metabolism , Mice, Inbred C57BL , Ischemia , Infarction , Obesity
16.
Macromol Biosci ; 24(1): e2300138, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37326828

ABSTRACT

Gasotransmitters, gaseous signaling molecules including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2 S), maintain myriad physiological processes. Low levels of gasotransmitters are often associated with specific problems or diseases, so NO, CO, and H2 S hold potential in treating bacterial infections, chronic wounds, myocardial infarction, ischemia, and various other diseases. However, their clinical applications as therapeutic agents are limited due to their gaseous nature, short half-life, and broad physiological roles. One route toward the greater application of gasotransmitters in medicine is through localized delivery. Hydrogels are attractive biomedical materials for the controlled release of embedded therapeutics as they are typically biocompatible, possess high water content, have tunable mechanical properties, and are injectable in certain cases. Hydrogel-based gasotransmitter delivery systems began with NO, and hydrogels for CO and H2 S have appeared more recently. In this review, the biological importance of gasotransmitters is highlighted, and the fabrication of hydrogel materials is discussed, distinguishing between methods used to physically encapsulate small molecule gasotransmitter donor compounds or chemically tether them to a hydrogel scaffold. The release behavior and potential therapeutic applications of gasotransmitter-releasing hydrogels are also detailed. Finally, the authors envision the future of this field and describe challenges moving forward.


Subject(s)
Gasotransmitters , Hydrogen Sulfide , Gasotransmitters/physiology , Gasotransmitters/therapeutic use , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Nitric Oxide , Carbon Monoxide/pharmacology , Carbon Monoxide/therapeutic use , Hydrogels/pharmacology , Hydrogels/therapeutic use
17.
Antioxid Redox Signal ; 40(4-6): 272-291, 2024 02.
Article in English | MEDLINE | ID: mdl-36974358

ABSTRACT

Significance: Nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) are, to date, the identified members of the gasotransmitter family, which consists of gaseous signaling molecules that play central roles in the regulation of a wide variety of physiological and pathophysiological processes, including inflammatory edema. Recent Advances: Recent studies show the potential anti-inflammatory and antiedematogenic effects of NO-, CO-, and H2S-donors in vivo. In general, it has been observed that the therapeutical effects of NO-donors are more relevant when administered at low doses at the onset of the inflammatory process. Regarding CO-donors, their antiedematogenic effects are mainly associated with inhibition of proinflammatory mediators (such as inducible NO synthase [iNOS]-derived NO), and the observed protective effects of H2S-donors seem to be mediated by reducing some proinflammatory enzyme activities. Critical Issues: The most recent investigations focus on the interactions among the gasotransmitters under different pathophysiological conditions. However, the biochemical/pharmacological nature of these interactions is neither general nor fully understood, although specifically dependent on the site where the inflammatory edema occurs. Future Directions: Considering the nature of the involved mechanisms, a deeper knowledge of the interactions among the gasotransmitters is mandatory. In addition, the development of new pharmacological tools, either donors or synthesis inhibitors of the three gasotransmitters, will certainly aid the basic investigations and open new strategies for the therapeutic treatment of inflammatory edema. Antioxid. Redox Signal. 40, 272-291.


Subject(s)
Gasotransmitters , Hydrogen Sulfide , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/pharmacology , Nitric Oxide , Signal Transduction , Carbon Monoxide
18.
Neurosci Res ; 199: 21-29, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37442198

ABSTRACT

Epilepsy is a common neurological disorder worldwide. Hydrogen sulfide (H2S) has been found to have anti-seizure effects. However, its mechanism remains to be explored. In the present study, we showed that a novel H2S donor attenuated neuroinflammation by up-regulating ATP-sensitive potassium channel (KATP) expression to reduce seizures. The novel H2S donor significantly reduced the expression of TNF-α and increased the expression of IL-10 in LPS-treated BV2 cells and the hippocampus of pilocarpine-induced epileptic mice. The modulatory effects of the H2S donor on inflammatory cytokines were prevented by glibenclamide, a common KATP channels blocker. The H2S donor promoted the expression of KATP channel subunits SUR2 and Kir6.1 in LPS-treated BV2 cells and the hippocampus of pilocarpine-induced epileptic mice. In addition, the H2S donor reduced the electroencephalography amplitude of hippocampal epileptic waves and reduced seizures in pilocarpine-induced epileptic mice, which were also attenuated by glibenclamide. These results indicated that the novel H2S donor reduced seizures and regulated microglial inflammatory cytokines by activating KATP channels, which may provide a prospective therapeutic strategy for the anti-seizure effects of H2S donor.


Subject(s)
Hydrogen Sulfide , Mice , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/metabolism , KATP Channels/metabolism , Neuroinflammatory Diseases , Glyburide/pharmacology , Lipopolysaccharides , Pilocarpine , Adenosine Triphosphate , Cytokines/metabolism
19.
Eur J Pharmacol ; 963: 176265, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38070636

ABSTRACT

Hydrogen sulfide (H2S), a colorless exhaust gas, has been traditionally considered an air pollutant. However, recent studies have revealed that H2S functions as a novel gas signaling molecule, exerting diverse biological effects on various systems, including the cardiovascular, digestive, and nervous systems. Thus, H2S is involved in various pathophysiological processes. As H2S affects reproductive function, it has potential therapeutic implications in reproductive system diseases. This review examined the role of H2S in various female reproductive organs, including the ovary, fallopian tube, vagina, uterus, and placenta. Additionally, the regulatory function of H2S in the female reproductive system has been discussed to provide useful insights for developing clinical therapeutic strategies for reproductive diseases.


Subject(s)
Cardiovascular System , Hydrogen Sulfide , Female , Humans , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Ovary , Uterus , Signal Transduction
20.
Pharmacol Biochem Behav ; 234: 173677, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37967673

ABSTRACT

BACKGROUND: Depression is a highly prevalent comorbidity arising in patients with Parkinson's disease (PD). However, depression in patients with PD is poorly treated. Hydrogen sulfide (H2S), a neuromodulator, has the potential to relieve depression. OBJECTIVE: To investigate whether H2S attenuates depression-like behaviours in a rat model of PD and examine the underlying mechanisms. METHODS: We utilised rotenone to develop a PD model with subcutaneous injections in the dorsal cervical region of Sprague-Dawley rats. The depression-like behaviours in the rotenone-induced PD model rats were assessed through forced swimming, tail suspension, open field, novelty-suppressed feeding, and elevated plus-maze tests. The expression of postsynaptic density protein-95 and synapsin-1, related to synaptic plasticity, was detected using Western blot in the hippocampus. The hippocampal ultrastructure, including the synaptic density, length of the synaptic active zone, postsynaptic density thickness, and synaptic gap width, was detected using transmission electron microscopy. RESULTS: We proved that sodium hydrosulfide (NaHS; a donor of H2S) significantly attenuated the depression-like behaviours and disorders of hippocampal synaptic plasticity in rotenone-induced PD rats. Furthermore, inhibition of the hippocampal Warburg effect by 2-deoxyglucose abolished NaHS-enhanced hippocampal synaptic plasticity and reversed NaHS-attenuated depression-like behaviours in the rotenone-induced PD rats. CONCLUSION: H2S attenuates PD-associated depression by improving the hippocampal synaptic plasticity in a hippocampal Warburg effect-dependent manner.


Subject(s)
Hydrogen Sulfide , Parkinson Disease , Humans , Rats , Animals , Hydrogen Sulfide/pharmacology , Hydrogen Sulfide/therapeutic use , Hydrogen Sulfide/metabolism , Depression/drug therapy , Parkinson Disease/metabolism , Rats, Sprague-Dawley , Rotenone/pharmacology , Rotenone/metabolism , Neuronal Plasticity , Hippocampus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL