Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 209
Filter
1.
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
2.
Med. clín (Ed. impr.) ; 162(7): 350-353, abril 2024.
Article in Spanish | IBECS | ID: ibc-232084

ABSTRACT

Introducción: El síndrome de pulmón encogido (SPE) es una manifestación rara del lupus eritematoso sistémico. Nuestro objetivo fue describir las características clínicas, radiológicas y funcionales de una cohorte con SPE y su evolución en el tiempo.MétodosEstudio retrospectivo entre 2009 y 2018. Se recogieron datos demográficos, clínicos, funcionales, radiológicos y de tratamiento.ResultadosDe un total de 225 pacientes, 11 presentaron SPE (prevalencia del 4,8%). Dos fueron excluidos. La edad media fue 39,33±16 años, 6 eran mujeres. Los síntomas principales fueron la disnea y el dolor pleurítico. La capacidad vital forzada media fue del 49%, la capacidad pulmonar total del 60%, la capacidad de difusión de monóxido de carbono del 66%, el factor de transferencia para el monóxido de carbono del 128%, la presión inspiratoria máxima del 66% y la presión espiratoria máxima del 82%. Todos los pacientes recibieron corticosteroides. Después de una mediana de seguimiento de 19 meses, 4 casos presentaron mejoría y 4 estabilización.ConclusionesEl SPE debe tenerse presente en todo paciente lúpico con disnea de causa no evidente. Si bien suele evolucionar con mejoría, la mayoría queda con deterioro persistente a pesar del tratamiento. (AU)


Introduction: Shrinking lung syndrome (SLS) is a rare manifestation of systemic lupus erythematosus. Our aim was to describe the clinical, radiological, and functional characteristics of a cohort with SLS and its evolution over time.MethodsA retrospective study was conducted between 2009 and 2018. Demographic, clinical, functional, radiological, and treatment data were collected.ResultsOut of a total of 225 patients, 11 presented with SLS (prevalence of 4.8%). Two patients were excluded. The mean age was 39.33±16 years, and 6 were female. The main symptoms were dyspnea and pleuritic pain. The mean forced vital capacity was 49%, total lung capacity was 60%, carbon monoxide diffusing capacity was 66%, carbon monoxide transference factor was 128%, maximal inspiratory pressure was 66%, and maximal expiratory pressure was 82%. All patients received corticosteroids. After a median follow-up of 19 months, 4 cases showed improvement, and 4 cases remained stable.ConclusionsSLS should be considered in every lupus patient with unexplained dyspnea. Although it often shows improvement, many cases experience persistent deterioration despite treatment. (AU)


Subject(s)
Humans , Carbon Monoxide/therapeutic use , Digestive System Diseases , Dyspnea/etiology , Lung Diseases/diagnosis , Lupus Erythematosus, Systemic/diagnosis , Lung/diagnostic imaging , Muscular Diseases
3.
Med Clin (Barc) ; 162(7): 350-353, 2024 04 12.
Article in English, Spanish | MEDLINE | ID: mdl-38195280

ABSTRACT

INTRODUCTION: Shrinking lung syndrome (SLS) is a rare manifestation of systemic lupus erythematosus. Our aim was to describe the clinical, radiological, and functional characteristics of a cohort with SLS and its evolution over time. METHODS: A retrospective study was conducted between 2009 and 2018. Demographic, clinical, functional, radiological, and treatment data were collected. RESULTS: Out of a total of 225 patients, 11 presented with SLS (prevalence of 4.8%). Two patients were excluded. The mean age was 39.33±16 years, and 6 were female. The main symptoms were dyspnea and pleuritic pain. The mean forced vital capacity was 49%, total lung capacity was 60%, carbon monoxide diffusing capacity was 66%, carbon monoxide transference factor was 128%, maximal inspiratory pressure was 66%, and maximal expiratory pressure was 82%. All patients received corticosteroids. After a median follow-up of 19 months, 4 cases showed improvement, and 4 cases remained stable. CONCLUSIONS: SLS should be considered in every lupus patient with unexplained dyspnea. Although it often shows improvement, many cases experience persistent deterioration despite treatment.


Subject(s)
Digestive System Diseases , Lung Diseases , Lupus Erythematosus, Systemic , Muscular Diseases , Humans , Female , Young Adult , Adult , Middle Aged , Male , Retrospective Studies , Carbon Monoxide/therapeutic use , Syndrome , Lung Diseases/diagnosis , Lupus Erythematosus, Systemic/diagnosis , Dyspnea/etiology , Lung/diagnostic imaging
4.
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
5.
Article in English | MEDLINE | ID: mdl-37904284

ABSTRACT

Gaseous signaling molecules such as nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2 S) have recently been recognized as essential signal mediators that regulate diverse physiological and pathological processes in the human body. With the evolution of gaseous signaling molecule biology, their therapeutic applications have attracted growing attention. One of the challenges in translational research of gaseous signaling molecules is the lack of efficient and safe delivery systems. To tackle this issue, researchers developed a library of gas donors, which are low molecular weight compounds that can release gaseous signaling molecules upon decomposition under physiological conditions. Despite the significant efforts to control gaseous signaling molecule release from gas donors, the therapeutic potential of gaseous signaling molecules cannot be fully explored due to their unfavorable pharmacokinetics and toxic side effects. Recently, the use of nanoparticle-based gas donors, especially self-assembled polymeric gas donors, have emerged as a promising approach. In this review, we describe the development of conventional small gas donors and the challenges in their therapeutic applications. We then illustrate the concepts and critical aspects for designing self-assembled polymeric gas donors and discuss the advantages of this approach in gasotransmistter delivery. We also highlight recent efforts to develop the delivery systems for those molecules based on self-assembled polymeric nanostructures. This article is categorized under: Therapeutic Approaches and Drug Discovery > Emerging Technologies.


Subject(s)
Gases , Hydrogen Sulfide , Humans , Nanomedicine , Signal Transduction , Hydrogen Sulfide/chemistry , Carbon Monoxide/therapeutic use , Nitric Oxide , Polymers
6.
Nano Lett ; 23(21): 9769-9777, 2023 11 08.
Article in English | MEDLINE | ID: mdl-37616496

ABSTRACT

Staphylococcus aureus (S. aureus) infection is a major infectious skin disease that is highly resistant to conventional antibiotic treatment and host immune defense, leading to recurrence and exacerbation of bacterial infection. Herein, we developed a photoresponsive carbon monoxide (CO)-releasing nanocomposite by integrating anion-π+ type-I photosensitizer (OMeTBP) and organometallic complex (FeCO) for the treatment of planktonic S. aureus and biofilm-associated infections. After optimizing the molar ratio of FeCO and OMeTBP, the prepared nanoparticles, OMeTBP@FeCONPs, not only ensured sufficient loading of CO donors and efficient CO generation but also showed negligible free ROS leakage under light irradiation, which helped to avoid tissue damage caused by excessive ROS. Both in vitro and in vivo results demonstrated that OMeTBP@FeCONPs could effectively inhibit S. aureus methicillin-resistant S. aureus (MRSA), and bacterial biofilm. Our design has the potential to overcome the resistance of conventional antibiotic treatment and provide a more effective option for bacterial infections.


Subject(s)
Methicillin-Resistant Staphylococcus aureus , Skin Diseases, Infectious , Staphylococcal Infections , Humans , Staphylococcus aureus , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use , Carbon Monoxide/pharmacology , Carbon Monoxide/therapeutic use , Reactive Oxygen Species , Staphylococcal Infections/drug therapy , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Biofilms , Microbial Sensitivity Tests
7.
Colloids Surf B Biointerfaces ; 230: 113513, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37579528

ABSTRACT

In situ mitochondrial oxidative stress amplification is an effective strategy to improve efficacy of cancer treatment. In this work, a tumor and mitochondria dual-targeted multifunctional nanoplatform CMS@AIPH@PDA@COTPP@FA (CAPCTF) was prepared, in which a thermally decomposable radical initiator AIPH was loaded inside the mesoporores of CuxMoySz (CMS) nanoparticles with polydopamine (PDA) covered films that were further covalently functionalized by a mitochondria-targeted CO donor (COTPP) and a directing group of folic acid (FA). The prepared CAPCTF nanoplatform selectively accumulated in cancer cells and further targeted the mitochondrial organelle where carbon monoxide (CO) and O2-independent free radicals (•OH/•R) were in situ generated upon 1064 nm laser irradiation. Furthermore, the CMS nanocarrier was capable of depleting the GSH overexpressed in the tumor microenvironment (TME), thus preventing free radical scavenging. As a result, the CAPCTF nanoplatform exhibited outstanding in vitro and in vivo antitumor efficacy under hypoxic conditions. This provides an innovative strategy that combines O2-independent free radicals (•OH/•R) generation, CO delivery and GSH consumption to amplify intracellular oxidative stresses and induce mitochondrial dysfunction, thus leading to cancer cells eradication, which may have significant implications for personalized hypoxic tumor treatment.


Subject(s)
Nanoparticles , Neoplasms , Photochemotherapy , Humans , Carbon Monoxide/pharmacology , Carbon Monoxide/therapeutic use , Neoplasms/pathology , Free Radicals , Mitochondria/pathology , Cell Line, Tumor , Tumor Microenvironment
8.
Int J Mol Sci ; 24(15)2023 Aug 05.
Article in English | MEDLINE | ID: mdl-37569855

ABSTRACT

Ischemia is the underlying mechanism in a wide variety of acute and persistent pathologies. As such, understanding the fine intracellular events occurring during (and after) the restriction of blood supply is pivotal to improving the outcomes in clinical settings. Among others, gaseous signaling molecules constitutively produced by mammalian cells (gasotransmitters) have been shown to be of potential interest for clinical treatment of ischemia/reperfusion injury. Nitric oxide (NO and its sibling, HNO), hydrogen sulfide (H2S), and carbon monoxide (CO) have long been proven to be cytoprotective in basic science experiments, and they are now awaiting confirmation with clinical trials. The aim of this work is to review the literature and the clinical trials database to address the state of development of potential therapeutic applications for NO, H2S, and CO and the clinical scenarios where they are more promising.


Subject(s)
Cardiovascular System , Gasotransmitters , Hydrogen Sulfide , Animals , Nitric Oxide , Signal Transduction , Carbon Monoxide/therapeutic use , Mammals
9.
Angew Chem Int Ed Engl ; 62(23): e202302525, 2023 06 05.
Article in English | MEDLINE | ID: mdl-36930411

ABSTRACT

Carbon monoxide (CO) is an endogenous signaling molecule with broad therapeutic effects. Here, a multifunctional X-ray-triggered carbon monoxide (CO) and manganese dioxide (MnO2 ) generation nanoplatform based on metal carbonyl and scintillating nanoparticles (SCNPs) is reported. Attributed to the radioluminescent characteristic of SCNPs, UV-responsive Mn2 (CO)10 is not only indirectly activated to release CO by X-ray but can also be degraded into MnO2 . A high dose of CO can be used as a glycolytic inhibitor for tumor suppression; it will also sensitize tumor cells to radiotherapy. Meanwhile MnO2 , as the photolytic byproduct of Mn2 (CO)10 , has both glutathione (GSH) depletion and Fenton-like Mn2+ delivery properties to produce highly toxic hydroxyl radical (⋅OH) in tumors. Thus, this strategy can realize X-ray-activated CO release, GSH depletion, and ⋅OH generation for cascade cancer radiosensitization. Furthermore, X-ray-activated Mn2+ in vivo demonstrates an MRI contrast effect, making it a potential theranostic nanoplatform.


Subject(s)
Nanoparticles , Neoplasms , Humans , Manganese Compounds/pharmacology , Manganese Compounds/therapeutic use , Oxides/pharmacology , Carbon Monoxide/pharmacology , Carbon Monoxide/therapeutic use , X-Rays , Neoplasms/drug therapy , Neoplasms/metabolism , Cell Line, Tumor , Glutathione/metabolism , Hydrogen Peroxide/therapeutic use
10.
Am J Cardiovasc Drugs ; 23(3): 329-338, 2023 May.
Article in English | MEDLINE | ID: mdl-36995544

ABSTRACT

BACKGROUND: Pulmonary veno-occlusive disease (PVOD) and pulmonary capillary hemangiomatosis (PCH) are rare types of pulmonary arterial hypertension with dismal prognoses; there is no established medical treatment for these conditions. Possible efficacy of imatinib against these conditions has been reported in 15 cases; however, how and in whom imatinib is effective remain unknown. METHODS: We retrospectively evaluated clinical data from consecutive patients with PVOD/PCH treated with imatinib at our institution. The diagnosis of PVOD/PCH was established using the following criteria: pre-capillary pulmonary hypertension; diffusion capacity of the lung for carbon monoxide < 60%; and two or more high-resolution computed tomography findings of interlobular septal thickening, centrilobular opacities, and mediastinal lymphadenopathy. The dose of pulmonary vasodilators remained unchanged during the assessment of imatinib. RESULTS: The medical records of five patients with PVOD/PCH were reviewed. The patients were aged 67 ± 13 years, their diffusion capacity of the lung for carbon monoxide was 29 ± 8%, and their mean pulmonary artery pressure was 40 ± 7 mmHg. Imatinib was administered at 50-100 mg/day; consequently, the World Health Organization functional class improved in one patient. In addition, imatinib improved the arterial oxygen partial pressure in this and another patient (these two also experienced a decreased mean pulmonary artery pressure and pulmonary vascular resistance after imatinib usage). CONCLUSIONS: This study indicated that imatinib improves the clinical condition, including pulmonary hemodynamics, of some patients with PVOD/PCH. In addition, patients with a certain high-resolution computed tomography pattern or PCH-dominant vasculopathy may respond favorably to imatinib.


Subject(s)
Hemangioma, Capillary , Lung Neoplasms , Pulmonary Veno-Occlusive Disease , Humans , Retrospective Studies , Pulmonary Artery , Imatinib Mesylate/therapeutic use , Pilot Projects , Pulmonary Veno-Occlusive Disease/drug therapy , Pulmonary Veno-Occlusive Disease/diagnosis , Lung Neoplasms/diagnosis , Lung Neoplasms/drug therapy , Carbon Monoxide/therapeutic use , Hemangioma, Capillary/drug therapy , Hemangioma, Capillary/diagnosis , Hemodynamics
11.
Small ; 19(14): e2206749, 2023 04.
Article in English | MEDLINE | ID: mdl-36599631

ABSTRACT

As a gasotransmitter, carbon monoxide (CO) possesses antitumor activity by reversing the Warburg effect at higher concentrations. The targeted delivery of carbon monoxide-releasing molecules (CORMs) using nanomaterials is an appealing option for CO administration, but how to maintain CO above the threshold concentration in tumor tissue remains a challenge. Herein, a nanozyme-catalyzed cascade reaction is proposed to promote CO release for high-efficacy photothermal therapy (PTT)-combined CO therapy of cancer. A gold-based porphyrinic coordination polymer nanosheet (Au0 -Por) is synthesized to serve as a carrier for CORM. It also possesses excellent glucose oxygenase-like activity owing to ultrasmall zero-valent gold atoms on the nanosheet. The catalytically generated H2 O2 can efficiently catalyze CORM decomposition, which enables in situ generation of sufficient CO for gas therapy. In vivo, the Au0 -Por nanosheets-enhanced photoacoustic imaging (PAI) and fluorescence imaging collectively demonstrate high tumor-targeting efficiency and nanomaterial retention. Proven to have augmented therapeutic efficacy, the nanoplatform can also be easily degraded and excreted through the kidney, indicating good biocompatibility. Thus, the application of rational designed Au0 -Por nanosheet with facile approach and biodegradable property to PAI-guided synergistic gas therapy can provide a strategy for the development of biocompatible and highly effective gaseous nanomedicine.


Subject(s)
Hyperthermia, Induced , Neoplasms , Porphyrins , Humans , Polymers/therapeutic use , Photothermal Therapy , Carbon Monoxide/therapeutic use , Porphyrins/therapeutic use , Hyperthermia, Induced/methods , Neoplasms/drug therapy , Gold/therapeutic use , Cell Line, Tumor
12.
ACS Biomater Sci Eng ; 9(6): 2937-2948, 2023 06 12.
Article in English | MEDLINE | ID: mdl-36444758

ABSTRACT

Carbon monoxide (CO) is a therapeutic gas with therapeutic potential in intestinal bowel disease. Therapeutic efficacy in the gastrointestinal tract (GIT) must be paired with safe and convenient use. Therefore, we designed an oral CO releasing system (OCORS) pairing tunable CO release into the GIT while preventing the release of any other molecule from within the device, causing safety concerns. The dimensions of the device, which is manufactured from 3D printed components, are within compendial limits. This is achieved by controlling CO decarbonylation from a molybdenum complex with a FeCl3 solution. OCORS' surrounding silicon membranes control release rates, as does the loading with carbonylated molybdenum complex and FeCl3 solution. Herein we describe the development of the system, the characterization of the CO releasing molecule (CORM), and the CO release kinetics of the overall system. Neither the CORM nor isocyanoacetate as a potential reaction byproduct were cytotoxic. Finally, we demonstrated by design validation in an in vivo porcine model that, except for the release of the therapeutic CO, OCORS isolates all components during transit through the stomach. We could show that OCORS generated and released CO locally into the stomach of the animals without systemic exposure, measured as the carboxyhemoglobin content in the blood of the pigs. In conclusion, OCORS derisks oral development by limiting patient exposure to (desirable) CO while preventing contact with any further (undesirable) chemical, by-, or degradation products. CO generating devices come in reach, which now can be used by anyone, anywhere, and anytime.


Subject(s)
Carbon Monoxide , Molybdenum , Animals , Swine , Carbon Monoxide/therapeutic use , Carbon Monoxide/metabolism
13.
Int Immunopharmacol ; 113(Pt B): 109441, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36435060

ABSTRACT

BACKGROUND: Intestinal ischemia-reperfusion injury (IRI) occurs in multiple clinical settings and contributes to systemic inflammatory response syndrome (SIRS) and multiple organ dysfunction syndrome (MODS). Due to the innate inflammatory immune nature, T cells play a crucial role in the pathogenesis of IRI, which includes not only CD4 + T cells, but also CD8 + and γδ T cells. Carbon monoxide (CO) plays an important role in regulating CD4 + T cell responses and has been proven to be a novel therapeutic target in a variety of inflammatory disease models. OBJECTIVE: This study aimed to assess whether pretreatment with carbon monoxide-releasing molecule-2 (CORM-2) could ameliorate inflammation by regulating differentiation of CD4 + T cells in intestinal mucosa of rats undergoing hemorrhagic shock. METHODS: A hemorrhagic shock model was established to study intestinal IRI. Morphological changes were investigated using light microscopes. Fluorescein isothiocyanate-dextran (FITC-dextran) was used as an indicator of intestinal paracellular permeability. Transcription factors involved in differentiation of CD4 + T cells in intestinal mucosa were detected by immunofluorescence, and the expression levels of related cytokines were determined by Western blotting. RESULTS: The results of hematoxylin-eosin (H-E) staining and FITC-dextran intestinal paracellular permeability assay revealed that CORM-2 maintained the integrity of intestinal mucosal barrier and inhibited the changes of intestinal mucosal permeability. In addition, activation of T helper type 1 (Th1) and T helper type 17 (Th17) cells, and the increased expression levels of proinflammatory cytokines, such as interleukin-17 (IL-17) and interferon-gamma (IFN-γ), were observed in intestinal IRI process. In contrast, pretreatment with CORM-2 weakened changes of the abovementioned observations, in which inhibited activation of Th1 and Th17 cells. However, CORM-2 did not influence differentiation of regulatory T (Treg) cells in intestinal IRI progress. Notably, CORM-2 significantly upregulated the expression level of interleukin-10 (IL-10) protein and downregulated the expression levels of IL-17 and IFN-γ proteins in ileal tissues of rats. CONCLUSION: CORM-2 possessed anti-inflammatory effects in the progress of intestinal IRI by inhibiting activation of Th1 and Th17 cells in rats undergoing hemorrhagic shock.


Subject(s)
Reperfusion Injury , Shock, Hemorrhagic , Animals , Rats , Carbon Monoxide/therapeutic use , Cytokines , Inflammation , Interferon-gamma , Interleukin-17 , Reperfusion Injury/drug therapy , Shock, Hemorrhagic/drug therapy , Systemic Inflammatory Response Syndrome
14.
Small ; 18(40): e2204244, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36055775

ABSTRACT

As a promising therapeutic modality targeting cancer, gas therapy still faces critical challenges, especially in enhancing therapeutic efficacy and avoiding gas poisoning risks. Here, a pH/glutathione (GSH) dual stimuli-responsive CRISPR/Cas9 gene-editing nanoplatform combined with calcium-enhanced CO gas therapy for precise anticancer therapy, is established. In the tumor microenvironment (TME), the fast biodegradation of the CaCO3 layer via pH-induced hydrolyzation allows glucose oxidase (GOx) to catalyze glucose for H2 O2 production, which further reacts with manganese carbonyl (MnCO) and achieves the precise release of CO gas. Simultaneously, in situ Ca2+ overload from CaCO3 degradation disturbs mitochondrial Ca2+ homeostasis, resulting in Ca2+ -driven reactive oxygen species (ROS) formation and subsequent mitochondrial apoptosis signaling pathway activation. Subsequently, by GSH-induced cleavage of a disulfide bond, the released Cas9/sgRNA (RNP) can achieve nuclear factor E2-related factor 2 (Nrf2) gene ablation to sensitize gas therapy by interfering with ROS signaling. This therapeutic modality endows codelivery of CRISPR, ions, and gas with smart control features, which demonstrates great potential for future clinical applications in precise nanomedicine.


Subject(s)
Nanoparticles , Neoplasms , Calcium , Carbon Monoxide/therapeutic use , Cell Line, Tumor , Disulfides , Gene Editing/methods , Glucose , Glucose Oxidase , Glutathione , Humans , Ions , Manganese , NF-E2-Related Factor 2/therapeutic use , Nanoparticles/chemistry , Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Tumor Microenvironment
15.
J Control Release ; 351: 81-101, 2022 11.
Article in English | MEDLINE | ID: mdl-36116579

ABSTRACT

Gasotransmitters like nitric oxide, carbon monoxide, and hydrogen sulfide with unique pleiotropic pharmacological effects in mammals are an emerging therapeutic modality for different human diseases including cancer, infection, ischemia-reperfusion injuries, and inflammation; however, their clinical translation is hampered by the lack of a reliable delivery form, which delivers such gasotransmitters to the action site with precisely controlled dosage. The external stimuli-responsive prodrug strategy has shown tremendous potential in developing gasotransmitter prodrugs, which affords precise temporospatial control and better dose control compared with endogenous stimuli-sensitive prodrugs. The promising external stimuli employed for gasotransmitter activation range from photo, ultrasound, and bioorthogonal click chemistry to exogenous enzymes. Herein, we highlight the recent development of external stimuli-mediated decaging chemistry for the temporospatial delivery of gasotransmitters including nitric oxide, carbon monoxide, hydrogen sulfide and sulfur dioxide, and discuss the pros and cons of different designs.


Subject(s)
Gasotransmitters , Hydrogen Sulfide , Prodrugs , Animals , Humans , Carbon Monoxide/therapeutic use , Hydrogen Sulfide/therapeutic use , Nitric Oxide , Mammals
16.
J Control Release ; 350: 652-667, 2022 10.
Article in English | MEDLINE | ID: mdl-36063960

ABSTRACT

Carbon monoxide (CO) has been regarded as a "silent killer" for its toxicity toward biological systems. However, a low concentration of endogenously produced CO has shown a number of therapeutic benefits such as anti-inflammatory, anti-proliferative, anti-apoptosis, and cytoprotective activities. Carbon monoxide-releasing molecules (CORMs) have been developed as alternatives to direct CO inhalation, which requires a specialized setting for strict dose control. CORMs are efficient CO donors, with central transition metals (such as ruthenium, iron, cobalt, and manganese) surrounded by CO as a ligand. CORMs can stably store and subsequently release their CO payload in the presence of certain triggers including solvent, light, temperature, and ligand substitution. However, CORMs require appropriate delivery strategies to improve short CO release half-life and target specificity. Herein, we highlighted the therapeutic potential of inhalation and CORMs-delivered CO. The applications of conjugate and nanocarrier systems for controlling CO release and improving therapeutic efficacy of CORMs are also described in detail. The review concludes with some of the hurdles that limit clinical translation of CORMs. Keeping in mind the tremendous potential and growing interest in CORMs, this review would be helpful for designing controlled CO release systems for clinical applications.


Subject(s)
Carbon Monoxide , Ruthenium , Anti-Inflammatory Agents , Carbon Monoxide/therapeutic use , Cobalt , Iron , Ligands , Manganese , Solvents
17.
Front Immunol ; 13: 991256, 2022.
Article in English | MEDLINE | ID: mdl-36119110

ABSTRACT

Antineutrophil cytoplasmic antibody (ANCA)-associated vasculitis (AAV) is a group of diseases characterized by inflammation and destruction of small and medium-sized blood vessels. Clinical disease phenotypes include microscopic polyangiitis (MPA), granulomatosis with polyangiitis (GPA), and eosinophilic granulomatosis with polyangiitis (EGPA). The incidence of AAV has been on the rise in recent years with advances in ANCA testing. The etiology and pathogenesis of AAV are multifactorial and influenced by both genetic and environmental factors, as well as innate and adaptive immune system responses. Multiple case reports have shown that sustained exposure to silica in an occupational environment resulted in a significantly increased risk of ANCA positivity. A meta-analysis involving six case-control studies showed that silica exposure was positively associated with AAV incidence. Additionally, exposure to air pollutants, such as carbon monoxide (CO), is a risk factor for AAV. AAV has seasonal trends. Studies have shown that various environmental factors stimulate the body to activate neutrophils and expose their own antigens, resulting in the release of proteases and neutrophil extracellular traps, which damage vascular endothelial cells. Additionally, the activation of complement replacement pathways may exacerbate vascular inflammation. However, the role of environmental factors in the etiology of AAV remains unclear and has received little attention. In this review, we summarized the recent literature on the study of environmental factors, such as seasons, air pollution, latitude, silica, and microbial infection, in AAV with the aim of exploring the relationship between environmental factors and AAV and possible mechanisms of action to provide a scientific basis for the prevention and treatment of AAV.


Subject(s)
Air Pollutants , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis , Churg-Strauss Syndrome , Granulomatosis with Polyangiitis , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/drug therapy , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/epidemiology , Anti-Neutrophil Cytoplasmic Antibody-Associated Vasculitis/etiology , Antibodies, Antineutrophil Cytoplasmic , Carbon Monoxide/therapeutic use , Churg-Strauss Syndrome/complications , Endothelial Cells/pathology , Humans , Inflammation/complications , Peptide Hydrolases , Silicon Dioxide
18.
Clin Cardiol ; 45(10): 1053-1059, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35920821

ABSTRACT

INTRODUCTION: Chronic heart failure (CHF) is associated with elevated total blood volume (BV) and distinct phenotypes of total red cell volume (RCV) and plasma volume (PV) elevations. Especially PV expansion during clinical decompensation is linked with adverse clinical outcomes. The role of PV expansion in compensated CHF patients is less clear. Aim of the present study is to investigate the impact of BV parameters on long-term mortality in CHF patients investigated at a compensated state. METHODS AND RESULTS: BV, PV and RCV were determined in 44 (9 female) compensated CHF patients using an abbreviated carbon monoxide method, who were followed up for 6.0 years, (range: 3.7-6.5 years) for all-cause mortality. In univariate analysis PV expansion but not BV and RCV predicted all-cause mortality (p = .021). A cutoff of 1800 ml PV/m² body-surface area allows stratification for all-cause mortality (p = .044). PV expansion but not RCV reduction explains the significantly lower hematocrit values of nonsurvivors. DISCUSSION: In this pilot study, PV expansion, which was unnoticed from a clinician's perspective, but is indicated by significantly lower hematocrit, appears to be a relevant predictor of long-term all-cause mortality. Whether PV expansion constitutes an adverse CHF phenotype and can be targeted by diuretic therapy is currently unclear.


Subject(s)
Heart Failure , Plasma Volume , Carbon Monoxide/therapeutic use , Chronic Disease , Diuretics/therapeutic use , Female , Heart Failure/diagnosis , Heart Failure/drug therapy , Humans , Pilot Projects
20.
BMC Psychiatry ; 22(1): 536, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35941573

ABSTRACT

BACKGROUND: Inflammation, autoimmunity, and gut-brain axis have been implicated in the pathogenesis of autism spectrum disorder (ASD). Carboxyhemoglobin (SpCO) as a non-invasive measurement of inflammation has not been studied in individuals with ASD. We conducted this post-hoc study based on our published clinical trial to explore SpCO and its association with ASD severity, autoimmunity, and response to daily Lactobacillus plantarum probiotic supplementation. METHODS: In this study, we included 35 individuals with ASD aged 3-20 years from a previously published clinical trial of the probiotic Lactobacillus plantarum. Subjects were randomly assigned to receive daily Lactobacillus plantarum probiotic (6 × 1010 CFUs) or a placebo for 16 weeks. The outcomes in this analysis include Social Responsiveness Scale (SRS), Aberrant Behavior Checklist second edition (ABC-2), Clinical Global Impression (CGI) scale, SpCO measured by CO-oximetry, fecal microbiome by 16 s rRNA sequencing, blood serum inflammatory markers, autoantibodies, and oxytocin (OT) by ELISA. We performed Kendall's correlation to examine their interrelationships and used Wilcoxon rank-sum test to compare the means of all outcomes between the two groups at baseline and 16 weeks. RESULTS: Elevated levels of serum anti-tubulin, CaM kinase II, anti-dopamine receptor D1 (anti-D1), and SpCO were found in the majority of ASD subjects. ASD severity is correlated with SpCO (baseline, R = 0.38, p = 0.029), anti-lysoganglioside GM1 (R = 0.83, p = 0.022), anti-tubulin (R = 0.69, p = 0.042), and anti-D1 (R = 0.71, p = 0.045) in treatment group. CONCLUSIONS: The findings of the present study suggests that the easily administered and non-invasive SpCO test offers a potentially promising autoimmunity and inflammatory biomarker to screen/subgroup ASD and monitor the treatment response to probiotics. Furthermore, we propose that the associations between autoantibodies, gut microbiome profile, serum OT level, GI symptom severity, and ASD core symptom severity scores are specific to the usage of probiotic treatment in our subject cohort. Taken together, these results warrant further studies to improve ASD early diagnosis and treatment outcomes. TRIAL REGISTRATION: ClinicalTrials.gov NCT03337035 , registered November 8, 2017.


Subject(s)
Autism Spectrum Disorder , Probiotics , Autism Spectrum Disorder/drug therapy , Autoantibodies , Autoimmunity , Biomarkers , Carbon Monoxide/therapeutic use , Child , Humans , Inflammation , Probiotics/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...