Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 94
Filtrar
Mais filtros

Medicinas Complementares
Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Adv Healthc Mater ; 12(10): e2202562, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36610060

RESUMO

Ferroptosis-based treatment strategies display the potential to suppress some malignant tumors with intrinsic apoptosis resistance. However, current related cancer treatments are still hampered by insufficient intracellular reactive oxygen species (ROS) levels and Fe2+ contents, posing considerable challenges for their clinical translation. Herein, an intracellular acid-biodegradable iridium-coordinated nanosheets (Ir-Hemin) with sonodynamic therapy (SDT) properties to effectively induce ferroptosis in tumor cells through multiple regulatory pathways are proposed. Under ultrasound (US) irradiation, Ir-Hemin nanosheets act as nanosonosensitizers to effectively generate ROS, subsequently causing the accumulation of lipid peroxides (LPO) and inducing ferroptotic cell death. Furthermore, these Ir-Hemin nanosheets decompose quickly to release hemin and Ir(IV), which deplete intracellular glutathione (GSH) to deactivate the enzyme glutathione peroxidase 4 (GPX4) and initiate the ferroptosis pathway. Specifically, the released hemin enables heme oxygenase 1 (HO-1) upregulation for endogenous ferrous ion supplementation, which compensates for the toxicity concerns brought about by the large uptake of exogenous iron. Surprisingly, Ir-Hemin nanosheets exhibit high tumor accumulation and trigger effective ferroptosis for tumor therapy. These Ir-Hemin nanosheets display pronounced synergistic anticancer efficacy under US stimulation both in vitro and in vivo, providing a strong rationale for the application of ferroptosis in cancer treatment.


Assuntos
Irídio , Neoplasias , Humanos , Irídio/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Hemina/farmacologia , Hemina/uso terapêutico , Morte Celular , Apoptose , Neoplasias/tratamento farmacológico , Glutationa/metabolismo
2.
Oxid Med Cell Longev ; 2023: 1464853, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36647427

RESUMO

Background and Aims: Endotoxemia (ET) is a common critical illness in patients receiving intensive care and is associated with high mortality and prolonged hospital stay. The intestinal epithelial cell dysfunction is regarded as the "engine" of deteriorated ET. Although electroacupuncture (EA) can mitigate endotoxin-induced intestinal epithelial cell dysfunction in ET, the mechanism through which EA improves endotoxin-induced intestinal injury for preventing ET deterioration needs further investigation. Methods: An in vivo ET model was developed by injecting lipopolysaccharide (LPS) in wild-type and PINK1-knockout mice. An in vitro model was also established by incubating epithelial cells in the serum samples obtained from both groups of mice. Hemin and zinc protoporphyrin IX (ZnPP) were applied to activate or inhibit heme oxygenase 1 (HO-1) production. EA treatment was performed for 30 min consecutively for 5 days before LPS injection, and on the day of the experiment, EA was performed throughout the process. Samples were harvested at 6 h after LPS induction for analyzing tissue injury, oxidative stress, ATP production, activity of diamine oxidase (DAO), and changes in the levels of HO-1, PTEN-induced putative kinase 1 (PINK1), mitochondrial fusion and fission marker gene, caspase-1, and interleukin 1 beta (IL-1ß). Results: In the wild-type models (both in vivo and vitro), EA alleviated LPS-induced intestinal injury and mitochondrial dysfunction, as indicated by decreased reactive oxygen species (ROS) production and oxygen consumption rate (OCR) and reduced levels of mitochondrial fission proteins. EA treatment also boosted histopathological morphology, ATP levels, DAO activity, and levels of mitochondrial fusion proteins in vivo and vitro. The effect of EA was enhanced by hemin but suppressed by Znpp. However, EA + AP, Znpp, or hemin had no effects on the LPS-induced, PINK1-knocked out mouse models. Conclusion: EA may improve the HO-1/PINK1 pathway-mediated mitochondrial dynamic balance to protect the intestinal barrier in patients with ET.


Assuntos
Eletroacupuntura , Endotoxemia , Heme Oxigenase-1 , Proteínas Quinases , Animais , Camundongos , Trifosfato de Adenosina , Endotoxemia/induzido quimicamente , Endotoxemia/terapia , Endotoxinas , Heme Oxigenase-1/metabolismo , Hemina/farmacologia , Lipopolissacarídeos/toxicidade , Dinâmica Mitocondrial
3.
Adv Healthc Mater ; 12(6): e2202663, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36653312

RESUMO

Ferroptosis, characterized by the accumulation of reactive oxygen species and lipid peroxides, has emerged as an attractive strategy to reverse drug resistance. Of particular interest is the ferroptosis-apoptosis combination therapy for cancer treatment. Herein, a nanoplatform is reported for effective co-delivery of the anticancer drug sorafenib (S) and the ferroptosis inducer hemin (H), toward synergistic ferroptosis-apoptosis therapy of advanced hepatocellular carcinoma (HCC) as a proof-of-concept study. Liposome is an excellent delivery system; however, it is not sufficiently responsive to the acidic tumor microenvironment (TME) for tumor-targeted drug delivery. The pH-sensitive vesicles are therefore developed (SH-AD-L) by incorporating amphiphilic dendrimers (AD) into liposomes for controlled and pH-stimulated release of sorafenib and hemin in the acidic TME, thanks to the protonation of numerous amine functionalities in AD. Importantly, SH-AD-L not only blocked glutathione synthesis to disrupt the antioxidant system, but also increased intracellular Fe2+ and ·OH concentrations to amplify oxidative stress, both of which contribute to enhanced ferroptosis. Remarkably, high levels of ·OH also augmented sorafenib-mediated apoptosis in tumor cells. This study demonstrates the efficacy of ferroptosis-apoptosis combination therapy, as well as the promise of the AD-doped TME-responsive vesicles for drug delivery in combination therapy to treat advanced HCC.


Assuntos
Carcinoma Hepatocelular , Dendrímeros , Ferroptose , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/patologia , Sorafenibe/farmacologia , Sorafenibe/uso terapêutico , Dendrímeros/farmacologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Hemina/farmacologia , Hemina/uso terapêutico , Apoptose , Lipossomos/farmacologia , Polímeros/farmacologia , Concentração de Íons de Hidrogênio , Linhagem Celular Tumoral , Microambiente Tumoral
4.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 48(10): 1437-1444, 2023 Oct 28.
Artigo em Inglês, Chinês | MEDLINE | ID: mdl-38432874

RESUMO

OBJECTIVES: Heme chloride (Hemin) is an in vitro purified form of natural heme and an important raw material for anti-anemia and antitumor drugs. This study aims to analyze the protective effect of Hemin on tissue damage in low-pressure oxygen chamber simulated plateau hypoxic mice, and explore its role in anti-plateau hypoxia. METHODS: Thirty male BALB/c mice were randomly divided into a blank group, a positive drug group (acetazolomide, 200 mg/kg), a Hemin low-dose group (15 mg/kg), a Hemin medium-dose group (30 mg/kg), and a Hemin high-dose group (60 mg/kg) with intraperitoneal injection. The anti-hypoxic activity of Hemin was explored by atmospheric closed hypoxia experiment and the optimal dose was screened. Thirty-six male BALB/c mice were randomly divided into a blank group, a hypoxia group, a positive drug group, and a Hemin high-dose group. The plasma inflammatory factor levels and oxidative stress indicators malondialdehyde (MDA), glutataione (GSH), and superoxide dismutase (SOD) levels of myocardium, brain, lung, and liver tissues were measured in different groups with hypoxia for 24 h. The degree of histopathological damage of mice was observed with HE staining. The degree of protection of Hemin against tissue hypoxia injury was detected with the hypoxia probe piperidazole. RESULTS: Compared with the blank group, the survival time of mice in the positive drug group, the Hemin medium-dose group, and high-dose group was significantly extended (all P<0.05), with the highest prolongation rate in the Hemin high-dose group. Compared with the hypoxia group, mice in the Hemin high-dose group showed a significant increase in SOD level and GSH content of brain tissue, and a significant decrease in MDA content of lung tissue (all P<0.05). The results of HE staining and hypoxia probe showed that Hemin had a significant protective effect on the damage of liver, heart, brain and lung tissues of mice with hypoxia, and the most obvious effect on that of the brain tissue. CONCLUSIONS: Hemin has an effect of improvement on oxidative stress and inflammatory response caused by hypoxia, and has obvious protective effect on tissue damage caused by hypoxia.


Assuntos
Heme , Hemina , Masculino , Camundongos , Animais , Hemina/farmacologia , Cloretos , Hipóxia , Camundongos Endogâmicos BALB C , Superóxido Dismutase
5.
Biomaterials ; 290: 121832, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36228518

RESUMO

Photodynamic therapy (PDT) induces apoptosis of cancer cells by generating cytotoxic reactive oxygen species, the therapeutic effect of which, however, is impeded by intrinsic/inducible apoptosis-resistant mechanisms in cancer cells and hypoxia of tumor microenvironment (TME); also, PDT-induced anti-tumor immunity activation is insufficient. To deal with these obstacles, a novel biomimetic nanoplatform is fabricated for the precise delivery of photosensitizer chlorin e6 (Ce6), hemin and PEP20 (CD47 inhibitory peptide), integrating oxygen-boosted PDT, ferroptosis activation and CD47-SIRPα blockade. Hemin's catalase-mimetic activity alleviates TME hypoxia and enhances PDT. The nanoplatform activates ferroptosis via both classical (down-regulating glutathione peroxidase 4 pathway) and non-classical (inducing Fe2+ overload) modes. Besides the role of hemin in consuming glutathione and up-regulating heme oxygenase-1 expression, interestingly, we observe that Ce6 enhance ferroptosis activation via both classical and non-classical modes. The anti-cancer immunity is reinforced by combining PEP20-mediated CD47-SIRPα blockade and PDT-mediated T cell activation, efficiently suppressing primary tumor growth and metastasis. PEP20 has been revealed for the first time to sensitize ferroptosis by down-regulating system Xc-. This work sheds new light on the mechanisms of PDT-ferroptosis activation interplay and bridges immunotherapy and ferroptosis activation, laying the theoretical foundation for novel combinational modes of cancer treatment.


Assuntos
Clorofilídeos , Ferroptose , Fotoquimioterapia , Porfirinas , Humanos , Antígeno CD47 , Microambiente Tumoral , Oxigênio/farmacologia , Biomimética , Hemina/farmacologia , Clorofilídeos/farmacologia , Linhagem Celular Tumoral , Fármacos Fotossensibilizantes/farmacologia , Fármacos Fotossensibilizantes/uso terapêutico , Fototerapia , Hipóxia/tratamento farmacológico
6.
J Vet Sci ; 23(5): e74, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36174978

RESUMO

BACKGROUND: Previous studies have presented evidence to support the significant association between red meat intake and colon cancer, suggesting that heme iron plays a key role in colon carcinogenesis. Epigallocatechin-3-gallate (EGCG), the major constituent of green tea, exhibits anti-oxidative and anti-cancer effects. However, the effect of EGCG on red meat-associated colon carcinogenesis is not well understood. OBJECTIVES: We aimed to investigate the regulatory effects of hemin and EGCG on colon carcinogenesis and the underlying mechanism of action. METHODS: Hemin and EGCG were treated in Caco2 cells to perform the water-soluble tetrazolium salt-1 assay, lactate dehydrogenase release assay, reactive oxygen species (ROS) detection assay, real-time quantitative polymerase chain reaction and western blot. We investigated the regulatory effects of hemin and EGCG on an azoxymethane (AOM) and dextran sodium sulfate (DSS)-induced colon carcinogenesis mouse model. RESULTS: In Caco2 cells, hemin increased cell proliferation and the expression of cell cycle regulatory proteins, and ROS levels. EGCG suppressed hemin-induced cell proliferation and cell cycle regulatory protein expression as well as mitochondrial ROS accumulation. Hemin increased nuclear factor erythroid-2-related factor 2 (Nrf2) expression, but decreased Keap1 expression. EGCG enhanced hemin-induced Nrf2 and antioxidant gene expression. Nrf2 inhibitor reversed EGCG reduced cell proliferation and cell cycle regulatory protein expression. In AOM/DSS mice, hemin treatment induced hyperplastic changes in colon tissues, inhibited by EGCG supplementation. EGCG reduced the hemin-induced numbers of total aberrant crypts and malondialdehyde concentration in the AOM/DSS model. CONCLUSIONS: We demonstrated that EGCG reduced hemin-induced proliferation and colon carcinogenesis through Nrf2-inhibited mitochondrial ROS accumulation.


Assuntos
Fator 2 Relacionado a NF-E2 , Doenças dos Roedores , Animais , Antioxidantes , Azoximetano , Células CACO-2 , Carcinogênese , Catequina/análogos & derivados , Proteínas de Ciclo Celular , Colo , Dextranos , Hemina/farmacologia , Humanos , Ferro , Proteína 1 Associada a ECH Semelhante a Kelch , Lactato Desidrogenases , Malondialdeído , Camundongos , Espécies Reativas de Oxigênio , Chá , Sais de Tetrazólio
7.
Sci Total Environ ; 853: 158665, 2022 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-36096218

RESUMO

Exposure to lead (Pb), a known toxin causing developmental neurotoxicity, can impair neurogenesis and oxidative phosphorylation (OXPHOS), but the mechanism is not clarified. In the current study, we aim to explore the effects of Pb on the differentiation of SH-SY5Y cells and investigate the role of heme and heme-binding protein BACH1 during differentiation. We found that Pb exposure caused a shift from OXPHOS to glycolysis, resulting in neurogenesis impairment by decreasing neurite growth and downregulation of PSD95 and Synapsin-1 in differentiated SH-SY5Y cells. Heme reduction mediated this mitochondria metabolism repression caused by Pb depending on BACH1 activation. Hemin supplement alleviated Pb-induced OXPHOS damage and adenosine triphosphate (ATP) reduction in differentiated SH-SY5Y cells, and further protected for Pb-induced damage of synapse. Heme binding factor BACH1 was negatively regulated by heme content and BACH1 knockout rescued the Pb-induced transcription and expression decline of genes related to OXPHOS and abrogated Pb-induced growth inhibition of axon promotion and synapse formation. Collectively, the present study demonstrates that heme deficiency mediates OXPHOS damage caused by Pb through BACH1 activation, resulting in neurogenesis impairment.


Assuntos
Hemina , Neuroblastoma , Humanos , Hemina/metabolismo , Hemina/farmacologia , Chumbo/toxicidade , Chumbo/metabolismo , Proteínas Ligantes de Grupo Heme , Sinapsinas/metabolismo , Sinapsinas/farmacologia , Neuroblastoma/metabolismo , Mitocôndrias , Heme/metabolismo , Trifosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Fatores de Transcrição de Zíper de Leucina Básica/genética , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/farmacologia
8.
Biomater Adv ; 140: 213091, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-36041322

RESUMO

The biocompatible nanosystem integrating hemin into black phosphorus nanosheets was ingeniously constructed through the easy modified strategy. Taking advantage of the enhanced permeability and retention (EPR) effect, the designed nanosystem could accumulate into the tumor location, leading to attractive cytotoxicity through the enhanced photodynamic therapy (PDT) ascribing to the catalytic oxygen supply and GSH depletion of hemin. Simultaneously, combining PDT and photothermal therapy (PTT) showed an apparent promotion in anti-tumor effect. Moreover, inflammatory response and immune activation amplified anti-tumor effect, which could compensate limitations of exogenous therapy (i.e., limited tissue depth and intensity-dependent curation effect) and potentiate the efficiency of the endogenous immune-activating behavior. Especially, the designed nanosystem degraded followed by being metabolized in the blood circulation. By and large, this constructed nanosystem provides the new insight into designing biocompatible nanomaterials and paves the ideal way for anti-tumor therapy. STATEMENT OF SIGNIFICANCE: Biocompatible nanomaterials-based synergistic tumor therapy offers the potential application prospect. Taking advantage of degradable black phosphorus, the nanosystem integrating hemin into black phosphorus for the enhanced photodynamic therapy and synergistic photothermal-photodynamic activating inflammation-immune response was developed and the results demonstrate that tumor growth was inhibited followed by activating inflammatory factors and leading to satisfactory immune response.


Assuntos
Neoplasias , Fotoquimioterapia , Hemina/farmacologia , Humanos , Imunidade , Neoplasias/tratamento farmacológico , Fósforo , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/farmacologia
9.
Transfus Apher Sci ; 61(2): 103319, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34801431

RESUMO

BACKGROUND: In transfusion-related iron overload, haem-derived iron accumulation in monocytes/macrophages is the initial event. When iron loading exceeds the ferritin storage capacity, iron is released into the plasma. When iron loading exceeds transferrin binding capacity, labile, non-transferrin-bound iron (NTBI) appears and causes organ injury. Haemin-induced cell death has already been investigated; however, whether NTBI induces cell death in monocytes/macrophages remains unclear. MATERIAL AND METHODS: Human monocytic THP-1 cells were treated with haemin or NTBI, particularly ferric ammonium citrate (FAC) or ferrous ammonium sulfate (FAS). The intracellular labile iron pool (LIP) was measured using an iron-sensitive fluorescent probe. Ferritin expression was measured by western blotting. RESULTS: LIP was elevated after haemin treatment but not after FAC or FAS treatment. Reactive oxygen species (ROS) generation and cell death induction were remarkable after haemin treatment but not after FAC or FAS treatment. Ferritin expression was not different between the FAC and haemin treatments. The combination of an iron chelator and a ferroptosis inhibitor significantly augmented the suppression of haemin cytotoxicity (p = 0.011). DISCUSSION: The difference in LIP suggests the different iron traffic mechanisms for haem-derived iron and NTBI. The Combination of iron chelators and antioxidants is beneficial for iron overload therapy.


Assuntos
Sobrecarga de Ferro , Ferro , Morte Celular , Ferritinas , Hemina/farmacologia , Humanos , Ferro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transferrina/metabolismo , Transferrina/farmacologia
10.
J Zhejiang Univ Sci B ; 22(11): 941-953, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34783224

RESUMO

Ganoderic triterpenoids (GTs) are the primary bioactive constituents of the Basidiomycotina fungus, Ganoderma lucidum. These compounds exhibit antitumor, anti-hyperlipidemic, and immune-modulatory pharmacological activities. This study focused on GT accumulation in mycelia of G. lucidum mediated by the heme oxygenase-1 (HO-1)/carbon monoxide (CO) signaling. Compared with the control, hemin (10 µmol/L) induced an increase of 60.1% in GT content and 57.1% in HO-1 activity. Moreover, carbon monoxide-releasing molecule-2 (CORM-2), CO donor, increased GT content by 56.0% and HO-1 activity by 18.1%. Zn protoporphyrin IX (ZnPPIX), a specific HO-1 inhibitor, significantly reduced GT content by 26.0% and HO-1 activity by 15.8%, while hemin supplementation reversed these effects. Transcriptome sequencing showed that HO-1/CO could function directly as a regulator involved in promoting GT accumulation by regulating gene expression in the mevalonate pathway, and modulating the reactive oxygen species (ROS) and Ca2+ pathways. The results of this study may help enhance large-scale GT production and support further exploration of GT metabolic networks and relevant signaling cross-talk.


Assuntos
Monóxido de Carbono/fisiologia , Heme Oxigenase-1/fisiologia , Reishi/metabolismo , Triterpenos/metabolismo , Sinalização do Cálcio , Ontologia Genética , Hemina/farmacologia , Protoporfirinas/farmacologia , RNA Mensageiro/análise , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/fisiologia
11.
Biochem Biophys Res Commun ; 556: 149-155, 2021 06 04.
Artigo em Inglês | MEDLINE | ID: mdl-33839410

RESUMO

Secondary injury is the main cause of high mortality and poor prognosis of TBI, which has recently been suggested to be related to ferroptosis. Polydatin, a monocrystalline compound extracted from the rhizome of Polygonum, has been shown to exert potential neuroprotective effects. However, its role and mechanism in the secondary injury of TBI has not been elucidated. In this study, the inhibition of Polydatin on ferroptosis was observed both in the hemoglobin treated Neuro2A cells in vitro and in TBI mouse model in vivo, characterized by reversion of accumulation or deposition of free Fe2+, increased content of MDA, decreased activity of key REDOX enzyme GPx4, cell death and tissues loss. Although Polydatin corrected the increased mRNA levels of ferroptosis signaling molecules GPX4, SLC7A11, PTGS2, and ATP5G3 after TBI, TBI and Polydatin treatment had no significant effect on their protein expression. Notably, Polydatin could completely reverse the decrease of GPx4 activity after TBI in vivo and in vitro, and the effect was stronger than that of the classical ferroptosis inhibitor FER-1 in vitro. Further, Polydatin has been shown to reduce the severity of acute neurological impairment and significantly improve subacute motor dysfunction in TBI mice. Our findings provided translational insight into neuroprotection with Polydatin in TBI by inhibiting ferroptosis mainly depending on the maintenance of GPx4 activity.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Lesões Encefálicas Traumáticas/prevenção & controle , Ferroptose/efeitos dos fármacos , Glucosídeos/farmacologia , Glucosídeos/uso terapêutico , Neuroproteção/efeitos dos fármacos , Fármacos Neuroprotetores/farmacologia , Estilbenos/farmacologia , Estilbenos/uso terapêutico , Animais , Encéfalo/efeitos dos fármacos , Encéfalo/fisiologia , Lesões Encefálicas Traumáticas/fisiopatologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Hemina/farmacologia , Ferro/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/efeitos dos fármacos , Neurônios/patologia , Fármacos Neuroprotetores/uso terapêutico , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/metabolismo
13.
Lab Invest ; 99(12): 1795-1809, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31570770

RESUMO

Sepsis-related acute lung injury (ALI) remains a major cause of mortality in critically ill patients and lacks specific therapy. Mitochondrial dysfunction is involved in the progression of septic lung injury. Mitochondrial dynamics, mitophagy, and biogenesis converge to constitute the assiduous quality control of mitochondria (MQC). Heme oxygenase-1 (HO-1) protects against sepsis-induced ALI through the modulation of mitochondrial dynamics. However, the causal relationship between HO-1 and the general processes of MQC, and their associated cellular pathways in sepsis-related ALI remain ill-defined. Herein, lipopolysaccharide (LPS)-induced ALI in Sprague-Dawley rats together with LPS-induced oxidative injury in RAW264.7 macrophages were used to investigate whether the PI3K/Akt pathway-mediated induction of HO-1 preserves MQC and alleviates septic lung injury. After pretreatment with hemin, a potent inducer of HO-1, LPS-induced cell apoptosis, enhanced mitochondrial fragmentation, and mitochondrial membrane potential damage were significantly reduced in macrophages. In rats, these effects were accompanied by a higher survival rate, less damage to lung tissue, a 28.5% elevation in lung mitochondria MnSOD activity, and a 39.2% increase in respiratory control ratios. Concomitantly, HO-1 induction preserved the dynamic process of mitochondrial fusion/fission (Mfn2, OPA1, Drp1), promoted mitochondrial biogenesis (NRF1, PGC1α, Tfam), and facilitated the key mediators of mitochondrial mitophagy (Parkin, PINK1) at mRNA and protein levels. Notably, LY294002, a PI3K inhibitor, or knockdown of PI3K by small interfering RNA significantly suppressed Akt phosphorylation, attenuated HO-1 induction, and further reversed these beneficial effects evoked by hemin pretreatment in RAW264.7 cells or rats received LPS, indicating a direct involvement of PI3K/Akt pathway. Taken together, our results indicated that HO-1 activation, through PI3K/Akt pathway, plays a critical role in protecting lung from oxidative injury in the setting of sepsis by regulating MQC. HO-1 may therefore be a therapeutic target for the prevention sepsis-related lung injury.


Assuntos
Lesão Pulmonar Aguda/prevenção & controle , Heme Oxigenase-1/metabolismo , Hemina/uso terapêutico , Renovação Mitocondrial/efeitos dos fármacos , Sepse/complicações , Lesão Pulmonar Aguda/etiologia , Animais , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática , Hemina/farmacologia , Lipopolissacarídeos , Pulmão/enzimologia , Masculino , Camundongos , Fator 2 Relacionado a NF-E2/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células RAW 264.7 , Ratos Sprague-Dawley , Transdução de Sinais
14.
J Inorg Biochem ; 193: 166-172, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30772693

RESUMO

Due to the traditional therapies of cancer inducing huge pains to patients, the non-invasive photo-guided therapies are attracting massive attentions of researchers. Herein, the intelligent-designed carbon-dots/hemin nanoplatforms (HCDs NPs) were developed, owning high-authority photo-therapy for cancer. The fluorescence resonance energy transfer (FRET) effect enhanced the photo-thermal ability of HCDs NPs, endowing the synthesized nanoplatforms with photo-dynamic property simultaneously. Therefore, the obtained HCDs NPs could achieve synergetic photo-thermal and photo-dynamic therapies for cancer. Basing on the experimental results, the prepared HCDs NPs could induce the temperature enhancement high to ca 26 °C under laser irradiation, also with the outstanding photo-dynamic efficacy. More than 90% of cancer cells die after 10 min laser treatment. Thus, the dual-modal photo-therapeutic HCDs NPs are promising and excellent nanomaterials for potential application in synergistic cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Hemina/farmacologia , Pontos Quânticos/química , Antineoplásicos/efeitos da radiação , Antineoplásicos/toxicidade , Carbono/química , Carbono/efeitos da radiação , Carbono/toxicidade , Transferência Ressonante de Energia de Fluorescência , Hemina/efeitos da radiação , Hemina/toxicidade , Células Hep G2 , Humanos , Hipertermia Induzida , Luz , Tamanho da Partícula , Fotoquimioterapia , Pontos Quânticos/efeitos da radiação , Pontos Quânticos/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Solubilidade , Temperatura
15.
Nutrients ; 9(8)2017 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-28933767

RESUMO

Hemin is a breakdown product of hemoglobin. It has been reported that the injection of hemin improves lipid metabolism and insulin sensitivity in various genetic models. However, the effect of hemin supplementation in food on lipid metabolism and insulin sensitivity is still unclear, and whether hemin directly affects cellular insulin sensitivity is yet to be elucidated. Here we show that hemin enhances insulin-induced phosphorylation of insulin receptors, Akt, Gsk3ß, FoxO1 and cytoplasmic translocation of FoxO1 in cultured primary hepatocytes under insulin-resistant conditions. Furthermore, hemin diminishes the accumulation of triglyceride and increases in free fatty acid content in primary hepatocytes induced by palmitate. Oral administration of hemin decreases body weight, energy intake, blood glucose and triglyceride levels, and improves insulin and glucose tolerance as well as hepatic insulin signaling and hepatic steatosis in male mice fed a high-fat diet. In addition, hemin treatment decreases the mRNA and protein levels of some hepatic genes involved in lipogenic regulation, fatty acid synthesis and storage, and increases the mRNA level and enzyme activity of CPT1 involved in fatty acid oxidation. These data demonstrate that hemin can improve lipid metabolism and insulin sensitivity in both cultured hepatocytes and mice fed a high-fat diet, and show the potential beneficial effects of hemin from food on lipid and glucose metabolism.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/prevenção & controle , Hemina/farmacologia , Hepatócitos/efeitos dos fármacos , Resistência à Insulina , Insulina/farmacologia , Metabolismo dos Lipídeos/efeitos dos fármacos , Animais , Biomarcadores/sangue , Glicemia/efeitos dos fármacos , Glicemia/metabolismo , Células Cultivadas , Relação Dose-Resposta a Droga , Metabolismo Energético/efeitos dos fármacos , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Intolerância à Glucose/sangue , Intolerância à Glucose/etiologia , Intolerância à Glucose/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Hepatócitos/metabolismo , Metabolismo dos Lipídeos/genética , Masculino , Camundongos Endogâmicos C57BL , Ácido Palmítico/farmacologia , Fosforilação , Transporte Proteico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor de Insulina/agonistas , Receptor de Insulina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Triglicerídeos/sangue , Redução de Peso/efeitos dos fármacos
16.
Biol Pharm Bull ; 39(6): 1007-12, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27251503

RESUMO

In this study we investigated the effect of free heme, the local level of which was increased by bleeding, on the intestinal barrier function, using human epithelial colorectal adenocarcinoma cells (Caco-2). Our results show that the addition of hemin to the culture medium markedly disrupted the barrier function, which was significantly improved by glutamine supplementation. Although hemin treatment caused the increased expression of heme oxygenase (HO)-1, the inhibition of HO activity resulted in the aggravation of hemin-induced barrier dysfunction. Up-regulation of HO-1 by pretreatment with a low concentration of hemin almost completely prevented hemin-induced barrier dysfunction. Taken together, these observations indicate that an abnormally high level of intracellular free heme causes barrier dysfunction, probably through the modulation of proteins forming tight junctions.


Assuntos
Hemorragia Gastrointestinal/metabolismo , Heme Oxigenase-1/metabolismo , Mucosa Intestinal/metabolismo , Ácido Ascórbico/farmacologia , Células CACO-2 , Glutamina/farmacologia , Heme Oxigenase-1/antagonistas & inibidores , Hemina/farmacologia , Humanos , Malondialdeído/metabolismo , Metaloporfirinas/farmacologia
17.
Proc Natl Acad Sci U S A ; 113(3): E348-57, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26729876

RESUMO

Mycobacterium tuberculosis (Mtb) encodes five type VII secretion systems (T7SS), designated ESX-1-ESX-5, that are critical for growth and pathogenesis. The best characterized is ESX-1, which profoundly impacts host cell interactions. In contrast, the ESX-3 T7SS is implicated in metal homeostasis, but efforts to define its function have been limited by an inability to recover deletion mutants. We overcame this impediment using medium supplemented with various iron complexes to recover mutants with deletions encompassing select genes within esx-3 or the entire operon. The esx-3 mutants were defective in uptake of siderophore-bound iron and dramatically accumulated cell-associated mycobactin siderophores. Proteomic analyses of culture filtrate revealed that secretion of EsxG and EsxH was codependent and that EsxG-EsxH also facilitated secretion of several members of the proline-glutamic acid (PE) and proline-proline-glutamic acid (PPE) protein families (named for conserved PE and PPE N-terminal motifs). Substrates that depended on EsxG-EsxH for secretion included PE5, encoded within the esx-3 locus, and the evolutionarily related PE15-PPE20 encoded outside the esx-3 locus. In vivo characterization of the mutants unexpectedly showed that the ESX-3 secretion system plays both iron-dependent and -independent roles in Mtb pathogenesis. PE5-PPE4 was found to be critical for the siderophore-mediated iron-acquisition functions of ESX-3. The importance of this iron-acquisition function was dependent upon host genotype, suggesting a role for ESX-3 secretion in counteracting host defense mechanisms that restrict iron availability. Further, we demonstrate that the ESX-3 T7SS secretes certain effectors that are important for iron uptake while additional secreted effectors modulate virulence in an iron-independent fashion.


Assuntos
Proteínas de Bactérias/metabolismo , Ferro/metabolismo , Mycobacterium tuberculosis/metabolismo , Mycobacterium tuberculosis/patogenicidade , Aerossóis , Animais , Polaridade Celular/efeitos dos fármacos , Genótipo , Hemina/farmacologia , Proteínas de Homeodomínio/metabolismo , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Humanos , Ferro/farmacologia , Macrófagos/citologia , Macrófagos/microbiologia , Espectrometria de Massas , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos SCID , Mutação/genética , Mycobacterium tuberculosis/efeitos dos fármacos , Mycobacterium tuberculosis/crescimento & desenvolvimento , Oxazóis/metabolismo , Fenótipo , Proteômica , Sideróforos/metabolismo , Especificidade por Substrato/efeitos dos fármacos , Virulência/efeitos dos fármacos
18.
Sci Rep ; 4: 6941, 2014 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-25373594

RESUMO

Although it is recognized that the abnormal accumulation of amino acid is a cause of the symptoms in metabolic disease such as phenylketonuria (PKU), the relationship between disease severity and serum amino acid levels is not well understood due to the lack of experimental model. Here, we present a novel in vitro cellular model using K562-D cells that proliferate slowly in the presence of excessive amount of phenylalanine within the clinically observed range, but not phenylpyruvate. The increased expression of the L-type amino acid transporter (LAT2) and its adapter protein 4F2 heavy chain appeared to be responsible for the higher sensitivity to phenylalanine in K562-D cells. Supplementation with valine over phenylalanine effectively restored cell proliferation, although other amino acids did not improve K562-D cell proliferation over phenylalanine. Biochemical analysis revealed mammalian target of rapamycin complex (mTORC) as a terminal target of phenylalanine in K562-D cell proliferation, and supplementation of valine restored mTORC1 activity. Our results show that K562-D cell can be a potent tool for the investigation of PKU at the molecular level and to explore new therapeutic approaches to the disease.


Assuntos
Modelos Biológicos , Fenilalanina/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Transporte Biológico , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proteína-1 Reguladora de Fusão/genética , Proteína-1 Reguladora de Fusão/metabolismo , Regulação da Expressão Gênica , Hemina/farmacologia , Humanos , Células K562 , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Fenilalanina/farmacologia , Fenilcetonúrias/genética , Fenilcetonúrias/metabolismo , Fenilcetonúrias/patologia , Ácidos Fenilpirúvicos/metabolismo , Ácidos Fenilpirúvicos/farmacologia , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Valina/metabolismo , Valina/farmacologia
19.
PLoS One ; 9(5): e97906, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24830678

RESUMO

Typically, most nephropathies can be categorized as complex human diseases in which the cumulative effect of multiple minor genes, combined with environmental and lifestyle factors, determines the disease phenotype. Thus, multi-target drugs would be more likely to facilitate comprehensive renoprotection than single-target agents. In this study, functional chemical-protein association analysis was performed to retrieve multi-target drugs of high pathway wideness from the STITCH 3.1 database. Pathway wideness of a drug evaluated the efficiency of regulation of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways in quantity. We identified nine experimentally validated renoprotectants that exerted remarkable impact on KEGG pathways by targeting a limited number of proteins. We selected curcumin as an illustrative compound to display the advantage of multi-pathway drugs on renoprotection. We compared curcumin with hemin, an agonist of heme oxygenase-1 (HO-1), which significantly affects only one KEGG pathway, porphyrin and chlorophyll metabolism (adjusted p = 1.5×10-5). At the same concentration (10 µM), both curcumin and hemin equivalently mitigated oxidative stress in H2O2-treated glomerular mesangial cells. The benefit of using hemin was derived from its agonistic effect on HO-1, providing relief from oxidative stress. Selective inhibition of HO-1 completely blocked the action of hemin but not that of curcumin, suggesting simultaneous multi-pathway intervention by curcumin. Curcumin also increased cellular autophagy levels, enhancing its protective effect; however, hemin had no effects. Based on the fact that the dysregulation of multiple pathways is implicated in the etiology of complex diseases, we proposed a feasible method for identifying multi-pathway drugs from compounds with validated targets. Our efforts will help identify multi-pathway agents capable of providing comprehensive protection against renal injuries.


Assuntos
Curcumina/farmacologia , Hemina/farmacologia , Substâncias Protetoras/farmacologia , Protoporfirinas/farmacologia , Autofagia , Células Cultivadas , Avaliação Pré-Clínica de Medicamentos , Heme Oxigenase-1/antagonistas & inibidores , Heme Oxigenase-1/metabolismo , Humanos , Peróxido de Hidrogênio , Nefropatias/tratamento farmacológico , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Associadas aos Microtúbulos/farmacologia
20.
PLoS One ; 9(4): e95079, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24733557

RESUMO

We investigated the effect of chronic nicotine on cholinergically-mediated renal vasodilations in female rats and its modulation by the nitric oxide synthase (NOS)/heme oxygenase (HO) pathways. Dose-vasodilatory response curves of acetylcholine (0.01-2.43 nmol) were established in isolated phenylephrine-preconstricted perfused kidneys obtained from rats treated with or without nicotine (0.5-4.0 mg/kg/day, 2 weeks). Acetylcholine vasodilations were potentiated by low nicotine doses (0.5 and 1 mg/kg/day) in contrast to no effect for higher doses (2 and 4 mg/kg/day). The facilitatory effect of nicotine was acetylcholine specific because it was not observed with other vasodilators such as 5'-N-ethylcarboxamidoadenosine (NECA, adenosine receptor agonist) or papaverine. Increases in NOS and HO-1 activities appear to mediate the nicotine-evoked enhancement of acetylcholine vasodilation because the latter was compromised after pharmacologic inhibition of NOS (L-NAME) or HO-1 (zinc protoporphyrin, ZnPP). The renal protein expression of phosphorylated Akt was not affected by nicotine. We also show that the presence of the two ovarian hormones is necessary for the nicotine augmentation of acetylcholine vasodilations to manifest because nicotine facilitation was lost in kidneys of ovariectomized (OVX) and restored after combined, but not individual, supplementation with medroxyprogesterone acetate (MPA) and estrogen (E2). Together, the data suggests that chronic nicotine potentiates acetylcholine renal vasodilation in female rats via, at least partly, Akt-independent HO-1 upregulation. The facilitatory effect of nicotine is dose dependent and requires the presence of the two ovarian hormones.


Assuntos
Heme Oxigenase (Desciclizante)/metabolismo , Hormônios/farmacologia , Rim/irrigação sanguínea , Nicotina/farmacologia , Óxido Nítrico Sintase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Vasodilatação/efeitos dos fármacos , Acetilcolina/farmacologia , Adenosina-5'-(N-etilcarboxamida)/farmacologia , Animais , Área Sob a Curva , Arginina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Estradiol/farmacologia , Feminino , Hemina/farmacologia , Técnicas In Vitro , Rim/efeitos dos fármacos , Acetato de Medroxiprogesterona/farmacologia , Mifepristona/farmacologia , Papaverina/farmacologia , Fenilefrina/farmacologia , Ratos Wistar , Transdução de Sinais/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA