Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 5.341
Filter
1.
ACS Appl Mater Interfaces ; 16(19): 24295-24307, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38697643

ABSTRACT

Pyroptosis has garnered increasing attention because of its ability to trigger robust antitumor immunity. Pyroptosis is initiated by the activation of inflammasomes, which are regulated by various organelles. The collaboration among organelles offers several protective mechanisms to prevent activation of the inflammasome, thereby limiting the induction of efficient pyroptosis. Herein, a multiorganelle homeostasis disruptor (denoted BLL) is constructed by encapsulating liposomes and bortezomib (BTZ) within a layered double hydroxide (LDH) nanocage to continuously activate inflammasomes for inducing efficient pyroptosis. In lysosomes, the negatively charged liposomes are released to recruit the NLRP3 inflammasomes through electrostatic interactions. ER stress is induced by BTZ to enhance the activation of the NLRP3 inflammasome. Meanwhile, the BLL nanocage exhibited H+-scavenging ability due to the weak alkalinity of LDH, thus disrupting the homeostasis of the lysosome and alleviating the degradation of the NLRP3 inflammasome by lysosomal-associated autophagy. Our results suggest that the BLL nanocage induces homeostatic imbalance in various organelles and efficient pyroptosis. We hope this work can provide new insights into the design of an efficient pyroptosis inducer by disrupting the homeostatic balance of multiple organelles and promote the development of novel antineoplastic platforms.


Subject(s)
Homeostasis , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Pyroptosis , Pyroptosis/drug effects , Inflammasomes/metabolism , Inflammasomes/drug effects , Homeostasis/drug effects , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Humans , Mice , Bortezomib/pharmacology , Bortezomib/chemistry , Liposomes/chemistry , Animals , Lysosomes/metabolism , Lysosomes/drug effects , Hydroxides/chemistry , Hydroxides/pharmacology , Nanostructures/chemistry , Nanoparticles/chemistry
2.
J Pharmacol Exp Ther ; 389(3): 313-314, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38772716

ABSTRACT

We thank Dr. Weimer and her colleagues for their comments related to our recent work (Anding et al., 2023) and are grateful for the opportunity to further discuss the importance of efficient lysosomal targeting of enzyme-replacement therapies (ERT) for the treatment of Pompe disease. Patients with Pompe disease have mutations in the gene that encodes for acid α glucosidase (GAA), a lysosomal enzyme necessary for the breakdown of glycogen. The first-generation ERT, alglucosidase alfa, provides a lifesaving therapy for the severe form of the disease (infantile onset Pompe disease) and improves or stabilizes respiratory and motor function in patients with less severe disease (late onset Pompe disease). Despite these gains, significant unmet need remains, particularly in patients who display respiratory and motor decline following years of treatment. Poor tissue uptake and lysosomal targeting via inefficient binding of the cation-independent mannose-6-phosphate (M6P) receptor (CIMPR) in skeletal muscle contributed to this suboptimal treatment response, prompting the development of new ERTs with increased levels of M6P.


Subject(s)
1-Deoxynojirimycin , Enzyme Replacement Therapy , Glycogen Storage Disease Type II , Mannosephosphates , alpha-Glucosidases , Glycogen Storage Disease Type II/drug therapy , Animals , Enzyme Replacement Therapy/methods , Mannosephosphates/metabolism , Mice , alpha-Glucosidases/therapeutic use , alpha-Glucosidases/metabolism , alpha-Glucosidases/administration & dosage , 1-Deoxynojirimycin/analogs & derivatives , 1-Deoxynojirimycin/administration & dosage , 1-Deoxynojirimycin/therapeutic use , Humans , Lysosomes/drug effects , Lysosomes/metabolism
3.
J Am Heart Assoc ; 13(9): e033700, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38700005

ABSTRACT

BACKGROUND: The only clinically approved drug that reduces doxorubicin cardiotoxicity is dexrazoxane, but its application is limited due to the risk of secondary malignancies. So, exploring alternative effective molecules to attenuate its cardiotoxicity is crucial. Colchicine is a safe and well-tolerated drug that helps reduce the production of reactive oxygen species. High doses of colchicine have been reported to block the fusion of autophagosomes and lysosomes in cancer cells. However, the impact of colchicine on the autophagy activity within cardiomyocytes remains inadequately elucidated. Recent studies have highlighted the beneficial effects of colchicine on patients with pericarditis, postprocedural atrial fibrillation, and coronary artery disease. It remains ambiguous how colchicine regulates autophagic flux in doxorubicin-induced heart failure. METHODS AND RESULTS: Doxorubicin was administered to establish models of heart failure both in vivo and in vitro. Prior studies have reported that doxorubicin impeded the breakdown of autophagic vacuoles, resulting in damaged mitochondria and the accumulation of reactive oxygen species. Following the administration of a low dose of colchicine (0.1 mg/kg, daily), significant improvements were observed in heart function (left ventricular ejection fraction: doxorubicin group versus treatment group=43.75%±3.614% versus 57.07%±2.968%, P=0.0373). In terms of mechanism, a low dose of colchicine facilitated the degradation of autolysosomes, thereby mitigating doxorubicin-induced cardiotoxicity. CONCLUSIONS: Our research has shown that a low dose of colchicine is pivotal in restoring the autophagy activity, thereby attenuating the cardiotoxicity induced by doxorubicin. Consequently, colchicine emerges as a promising therapeutic candidate to improve doxorubicin cardiotoxicity.


Subject(s)
Autophagy , Cardiotoxicity , Colchicine , Doxorubicin , Lysosomes , Myocytes, Cardiac , Colchicine/toxicity , Colchicine/pharmacology , Doxorubicin/toxicity , Cardiotoxicity/prevention & control , Autophagy/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , Animals , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Disease Models, Animal , Male , Heart Failure/chemically induced , Heart Failure/drug therapy , Heart Failure/metabolism , Antibiotics, Antineoplastic/toxicity , Reactive Oxygen Species/metabolism , Mice , Mice, Inbred C57BL , Ventricular Function, Left/drug effects
4.
Ecotoxicol Environ Saf ; 276: 116318, 2024 May.
Article in English | MEDLINE | ID: mdl-38626609

ABSTRACT

Perfluorooctane sulfonate (PFOS), an officially listed persistent organic pollutant, is a widely distributed perfluoroalkyl substance. Epidemiological studies have shown that PFOS is intimately linked to the occurrence of insulin resistance (IR). However, the detailed mechanism remains obscure. In previous studies, we found that mitochondrial calcium overload was concerned with hepatic IR induced by PFOS. In this study, we found that PFOS exposure noticeably raised lysosomal calcium in L-02 hepatocytes from 0.5 h. In the PFOS-cultured L-02 cells, inhibiting autophagy alleviated lysosomal calcium overload. Inhibition of mitochondrial calcium uptake aggravated the accumulation of lysosomal calcium, while inhibition of lysosomal calcium outflowing reversed PFOS-induced mitochondrial calcium overload and IR. Transient receptor potential mucolipin 1 (TRPML1), the calcium output channel of lysosomes, interacted with voltage-dependent anion channel 1 (VDAC1), the calcium intake channel of mitochondria, in the PFOS-cultured cells. Moreover, we found that ATP synthase F1 subunit beta (ATP5B) interacted with TRPML1 and VDAC1 in the L-02 cells and the liver of mice under PFOS exposure. Inhibiting ATP5B expression or restraining the ATP5B on the plasma membrane reduced the interplay between TRPML1 and VDAC1, reversed the mitochondrial calcium overload and deteriorated the lysosomal calcium accumulation in the PFOS-cultured cells. Our research unveils the molecular regulation of the calcium crosstalk between lysosomes and mitochondria, and explains PFOS-induced IR in the context of activated autophagy.


Subject(s)
Alkanesulfonic Acids , Autophagy , Calcium , Fluorocarbons , Insulin Resistance , Liver , Lysosomes , Mitochondria , Mitochondrial Proton-Translocating ATPases , Alkanesulfonic Acids/toxicity , Fluorocarbons/toxicity , Animals , Lysosomes/drug effects , Lysosomes/metabolism , Autophagy/drug effects , Calcium/metabolism , Mice , Mitochondrial Proton-Translocating ATPases/metabolism , Liver/drug effects , Liver/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Male , Voltage-Dependent Anion Channel 1/metabolism , Cell Line , Hepatocytes/drug effects , Hepatocytes/metabolism , Environmental Pollutants/toxicity , TRPM Cation Channels/metabolism , Mice, Inbred C57BL
5.
Ecotoxicol Environ Saf ; 276: 116322, 2024 May.
Article in English | MEDLINE | ID: mdl-38636258

ABSTRACT

Lead is a widespread environmental pollutant with serious adverse effects on human health, but the mechanism underlying its toxicity remains elusive. This study aimed to investigate the role of miR-584-5p / Ykt6 axis in the toxic effect of lead on HK-2 cells and the related mechanism. Our data suggested that lead exposure caused significant cytotoxicity, DNA and chromosome damage to HK-2 cells. Mechanistically, lead exposure down-regulated miR-584-5p and up-regulated Ykt6 expression, consequently, autophagosomal number and autophagic flux increased, lysosomal number and activity decreased, exosomal secretion increased. Interestingly, when miR-584-5p level was enhanced with mimic, autophagosomal number and autophagic flux decreased, lysosomal number and activity increased, ultimately, exosomal secretion was down-regulated, which resulted in significant aggravated toxic effects of lead. Further, directly blocking exosomal secretion with inhibitor GW4869 also resulted in exacerbated toxic effects of lead. Herein, we conclude that miR-584-5p / Ykt6 - mediated autophagy - lysosome - exosome pathway may be a critical route affecting the toxic effects of lead on HK-2 cells. We provide a novel insight into the mechanism underlying the toxicity of lead on human cells.


Subject(s)
Autophagy , Exosomes , Lead , Lysosomes , MicroRNAs , Humans , Autophagy/drug effects , MicroRNAs/genetics , MicroRNAs/metabolism , Exosomes/drug effects , Exosomes/metabolism , Lysosomes/drug effects , Cell Line , Lead/toxicity , Environmental Pollutants/toxicity , Vacuolar Proton-Translocating ATPases/genetics , DNA Damage
6.
Int J Mol Sci ; 25(8)2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38673807

ABSTRACT

Fluorescence lifetime imaging (FLIM) and confocal fluorescence studies of a porphyrin-based photosensitiser (meso-tetraphenylporphine disulfonate: TPPS2a) were evaluated in 2D monolayer cultures and 3D compressed collagen constructs of a human ovarian cancer cell line (HEY). TPPS2a is known to be an effective model photosensitiser for both Photodynamic Therapy (PDT) and Photochemical Internalisation (PCI). This microspectrofluorimetric study aimed firstly to investigate the uptake and subcellular localisation of TPPS2a, and evaluate the photo-oxidative mechanism using reactive oxygen species (ROS) and lipid peroxidation probes combined with appropriate ROS scavengers. Light-induced intracellular redistribution of TPPS2a was observed, consistent with rupture of endolysosomes where the porphyrin localises. Using the same range of light doses, time-lapse confocal imaging permitted observation of PDT-induced generation of ROS in both 2D and 3D cancer models using fluorescence-based ROS together with specific ROS inhibitors. In addition, the use of red light excitation of the photosensitiser to minimise auto-oxidation of the probes was investigated. In the second part of the study, the photophysical properties of TPPS2a in cells were studied using a time-domain FLIM system with time-correlated single photon counting detection. Owing to the high sensitivity and spatial resolution of this system, we acquired FLIM images that enabled the fluorescence lifetime determination of the porphyrin within the endolysosomal vesicles. Changes in the lifetime dynamics upon prolonged illumination were revealed as the vesicles degraded within the cells.


Subject(s)
Photosensitizing Agents , Porphyrins , Reactive Oxygen Species , Humans , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Porphyrins/pharmacology , Porphyrins/chemistry , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Photochemotherapy/methods , Optical Imaging/methods , Lysosomes/metabolism , Lysosomes/drug effects , Female , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/drug therapy
7.
Int J Mol Sci ; 25(8)2024 Apr 16.
Article in English | MEDLINE | ID: mdl-38673964

ABSTRACT

Hyponatremia is the prevalent electrolyte imbalance in cancer patients, and it is associated with a worse outcome. Notably, emerging clinical evidence suggests that hyponatremia adversely influences the response to anticancer treatments. Therefore, this study aims to investigate how reduced extracellular [Na+] affects the responsiveness of different cancer cell lines (from human colon adenocarcinoma, neuroblastoma, and small cell lung cancer) to cisplatin and the underlying potential mechanisms. Cisplatin dose-response curves revealed higher IC50 in low [Na+] than normal [Na+]. Accordingly, cisplatin treatment was less effective in counteracting the proliferation and migration of tumor cells when cultured in low [Na+], as demonstrated by colony formation and invasion assays. In addition, the expression analysis of proteins involved in autophagosome-lysosome formation and the visualization of lysosomal areas by electron microscopy revealed that one of the main mechanisms involved in chemoresistance to cisplatin is the promotion of autophagy. In conclusion, our data first demonstrate that the antitumoral effect of cisplatin is markedly reduced in low [Na+] and that autophagy is an important mechanism of drug escape. This study indicates the role of hyponatremia in cisplatin chemoresistance and reinforces the recommendation to correct this electrolyte alteration in cancer patients.


Subject(s)
Antineoplastic Agents , Autophagy , Cell Proliferation , Cisplatin , Sodium , Humans , Cisplatin/pharmacology , Autophagy/drug effects , Sodium/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/drug effects , Hyponatremia/metabolism , Cell Movement/drug effects , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Lysosomes/metabolism , Lysosomes/drug effects
8.
Toxicology ; 504: 153795, 2024 May.
Article in English | MEDLINE | ID: mdl-38574842

ABSTRACT

The mechanistic target of rapamycin (RAPA) complex 1 (mTORC1) - transcription factor EB (TFEB) pathway plays a crucial role in response to nutritional status, energy and environmental stress for maintaining cellular homeostasis. But there is few reports on its role in the toxic effects of arsenic exposure and the related mechanisms. Here, we show that the exposure of bronchial epithelial cells (BEAS-2B) to sodium arsenite promoted the activation of mTORC1 (p-mTORC1) and the inactivation of TFEB (p-TFEB), the number and activity of lysosomes decreased, the content of reduced glutathione (GSH) and superoxide dismutase (SOD) decreased, the content of malondialdehyde (MDA) increased, the DNA and chromosome damage elevated. Further, when mTORC1 was inhibited with RAPA, p-mTORC1 and p-TFEB down-regulated, GSH and SOD increased, MDA decreased, the DNA and chromosome damage reduced significantly, as compared with the control group. Our data revealed for the first time that mTORC1 - TFEB pathway was involved in sodium arsenite induced lysosomal alteration, oxidative stress and genetic damage in BEAS-2B cells, and it may be a potential intervention target for the toxic effects of arsenic.


Subject(s)
Arsenites , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , DNA Damage , Lysosomes , Mechanistic Target of Rapamycin Complex 1 , Oxidative Stress , Sodium Compounds , Arsenites/toxicity , Sodium Compounds/toxicity , Oxidative Stress/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Lysosomes/drug effects , Lysosomes/metabolism , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Cell Line , DNA Damage/drug effects , TOR Serine-Threonine Kinases/metabolism , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Signal Transduction/drug effects , Bronchi/drug effects , Bronchi/metabolism , Bronchi/cytology , Bronchi/pathology , Glutathione/metabolism , Superoxide Dismutase/metabolism , Multiprotein Complexes/metabolism , Malondialdehyde/metabolism
9.
Aging (Albany NY) ; 16(9): 7683-7703, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38683121

ABSTRACT

Ferroptosis, an iron-triggered modality of cellular death, has been reported to closely relate to human aging progression and aging-related diseases. However, the involvement of ferroptosis in the development and maintenance of senescent cells still remains elusive. Here, we established a doxorubicin-induced senescent HSkM cell model and found that both iron accumulation and lipid peroxidation increase in senescent cells. Moreover, such iron overload in senescent cells has changed the expression panel of the ferroptosis-response proteins. Interestingly, the iron accumulation and lipid peroxidation does not trigger ferroptosis-induced cell death. Oppositely, senescent cells manifest resistance to the ferroptosis inducers, compared to the proliferating cells. To further investigate the mechanism of ferroptosis-resistance for senescent cells, we traced the iron flux in cell and found iron arrested in lysosome. Moreover, disruption of lysosome functions by chloroquine and LLOMe dramatically triggered the senescent cell death. Besides, the ferroitinophagy-related proteins FTH1/FTL and NCOA4 knockdown also increases the senescent cell death. Thus, we speculated that iron retardation in lysosome of senescent cells is the key mechanism for ferroptosis resistance. And the lysosome is a promising target for senolytic drugs to selectively clear senescent cells and alleviate the aging related diseases.


Subject(s)
Cellular Senescence , Ferroptosis , Iron , Lysosomes , Ferroptosis/drug effects , Humans , Cellular Senescence/drug effects , Cellular Senescence/physiology , Lysosomes/metabolism , Lysosomes/drug effects , Iron/metabolism , Lipid Peroxidation/drug effects , Doxorubicin/pharmacology , Nuclear Receptor Coactivators/metabolism , Nuclear Receptor Coactivators/genetics , Cell Line , Chloroquine/pharmacology , Iron Overload/metabolism , Ferritins , Oxidoreductases
10.
J Ethnopharmacol ; 330: 118196, 2024 Aug 10.
Article in English | MEDLINE | ID: mdl-38631488

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Rosmarinic acid (RA), a natural polyphenol abundant in numerous herbal remedies, has been attracting growing interest owing to its exceptional ability to protect the liver. Toosendanin (TSN), a prominent bioactive compound derived from Melia toosendan Siebold & Zucc., boasts diverse pharmacological properties. Nevertheless, TSN possesses remarkable hepatotoxicity. Intriguingly, the potential of RA to counteract TSN-induced liver damage and its probable mechanisms remain unexplored. AIM OF THE STUDY: This study is aimed at exploring whether RA can alleviate TSN-induced liver injury and the potential mechanisms involved autophagy. MATERIALS AND METHODS: CCK-8 and LDH leakage rate assay were used to evaluate cytotoxicity. Balb/c mice were intraperitoneally administered TSN (20 mg/kg) for 24 h after pretreatment with RA (0, 40, 80 mg/kg) by gavage for 5 days. The autophagic proteins P62 and LC3B expressions were detected using western blot and immunohistochemistry. RFP-GFP-LC3B and transmission electron microscopy were applied to observe the accumulation levels of autophagosomes and autolysosomes. LysoTracker Red and DQ-BSA staining were used to evaluate the lysosomal acidity and degradation ability respectively. Western blot, immunohistochemistry and immunofluorescence staining were employed to measure the expressions of JAK2/STAT3/CTSC pathway proteins. Dual-luciferase reporter gene was used to measure the transcriptional activity of CTSC and RT-PCR was used to detect its mRNA level. H&E staining and serum biochemical assay were employed to determine the degree of damage to the liver. RESULTS: TSN-induced damage to hepatocytes and livers was significantly alleviated by RA. RA markedly diminished the autophagic flux blockade and lysosomal dysfunction caused by TSN. Mechanically, RA alleviated TSN-induced down-regulation of CTSC by activating JAK2/STAT3 signaling pathway. CONCLUSION: RA could protect against TSN-induced liver injury by activating the JAK2/STAT3/CTSC pathway-mediated autophagy and lysosomal function.


Subject(s)
Autophagy , Chemical and Drug Induced Liver Injury , Cinnamates , Depsides , Janus Kinase 2 , Lysosomes , Mice, Inbred BALB C , Rosmarinic Acid , STAT3 Transcription Factor , Signal Transduction , Animals , Depsides/pharmacology , STAT3 Transcription Factor/metabolism , Janus Kinase 2/metabolism , Cinnamates/pharmacology , Autophagy/drug effects , Mice , Lysosomes/drug effects , Lysosomes/metabolism , Male , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Chemical and Drug Induced Liver Injury/prevention & control , Signal Transduction/drug effects , Liver/drug effects , Liver/metabolism , Liver/pathology , Drugs, Chinese Herbal/pharmacology , Humans
11.
J Hazard Mater ; 471: 134357, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38643584

ABSTRACT

The compound 6PPD is widely acknowledged for its antioxidative properties; however, concerns regarding its impact on aquatic organisms have spurred comprehensive investigations. In our study, we advanced our comprehension by revealing that exposure to 6PPD could induce cardiac dysfunction, myocardial injury and DNA damage in adult zebrafish. Furthermore, our exploration unveiled that the exposure of cardiomyocytes to 6PPD resulted in apoptosis and mitochondrial injury, as corroborated by analyses using transmission electron microscopy and flow cytometry. Significantly, our study demonstrated the activation of the autophagy pathway in both the heart of zebrafish and cardiomyocytes, as substantiated by transmission electron microscopy and immunofluorescent techniques. Importantly, the increased the expression of P62 in the heart and cardiomyocytes suggested an inhibition of the autophagic process. The reduction in autophagy flux was also verified through in vivo experiments involving the infection of mCherry-GFP-LC3. We further identified that the fusion of autophagosomes and lysosomes was impaired in the 6PPD treatment group. In summary, our findings indicated that the impaired fusion of autophagosomes and lysosomes hampered the autophagic degradation process, leading to apoptosis and ultimately resulting in cardiac dysfunction and myocardial injury. This study discovered the crucial role of the autophagy pathway in regulating 6PPD-induced cardiotoxicity. SYNOPSIS: 6PPD exposure inhibited the autophagic degradation process and induced mitochondrial injury and apoptosis in the heart of adult zebrafish.


Subject(s)
Apoptosis , Autophagy , Mitochondria , Myocytes, Cardiac , Zebrafish , Animals , Autophagy/drug effects , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Apoptosis/drug effects , Mitochondria/drug effects , Lysosomes/drug effects , Lysosomes/metabolism , DNA Damage , Heart/drug effects
12.
J Photochem Photobiol B ; 255: 112919, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38677261

ABSTRACT

Endolysosomes perform a wide range of cellular functions, including nutrient sensing, macromolecule digestion and recycling, as well as plasma membrane repair. Because of their high activity in cancerous cells, endolysosomes are attractive targets for the development of novel cancer treatments. Light-activated compounds termed photosensitizers (PS) can catalyze the oxidation of specific biomolecules and intracellular organelles. To selectively damage endosomes and lysosomes, HT-29 colorectal cancer cells were incubated with nanomolar concentrations of meso-tetraphenylporphine disulfonate (TPPS2a), an amphiphilic PS taken up via endocytosis and activated by green light (522 nm, 2.1 J.cm-1). Several cellular responses were characterized by a combination of immunofluorescence and immunoblotting assays. We showed that TPPS2a photosensitization blocked autophagic flux without extensive endolysosomal membrane rupture. Nevertheless, there was a severe functional failure of endolysosomes due to a decrease in CTSD (cathepsin D, 55%) and CTSB (cathepsin B, 52%) maturation. PSAP (prosaposin) processing (into saposins) was also considerably impaired, a fact that could be detrimental to glycosphingolipid homeostasis. Therefore, photosensitization of HT-29 cells previously incubated with a low concentration of TPPS2a promotes endolysosomal dysfunction, an effect that can be used to improve cancer therapies.


Subject(s)
Autophagy , Lysosomes , Photosensitizing Agents , Humans , HT29 Cells , Lysosomes/metabolism , Lysosomes/drug effects , Autophagy/drug effects , Autophagy/radiation effects , Photosensitizing Agents/pharmacology , Photosensitizing Agents/chemistry , Endosomes/metabolism , Endosomes/drug effects , Cathepsins/metabolism , Cathepsins/antagonists & inhibitors , Light , Porphyrins/pharmacology , Porphyrins/chemistry , Cathepsin D/metabolism , Cathepsin B/metabolism
13.
Antiviral Res ; 226: 105891, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38649071

ABSTRACT

Zoonoses such as ZIKV and SARS-CoV-2 pose a severe risk to global health. There is urgent need for broad antiviral strategies based on host-targets filling gaps between pathogen emergence and availability of therapeutic or preventive strategies. Significant reduction of pathogen titers decreases spread of infections and thereby ensures health systems not being overloaded and public life to continue. Based on previously observed interference with FGFR1/2-signaling dependent impact on interferon stimulated gene (ISG)-expression, we identified Pim kinases as promising druggable cellular target. We therefore focused on analyzing the potential of pan-Pim kinase inhibition to trigger a broad antiviral response. The pan-Pim kinase inhibitor AZD1208 exerted an extraordinarily high antiviral effect against various ZIKV isolates, SARS-CoV-2 and HBV. This was reflected by strong reduction in viral RNA, proteins and released infectious particles. Especially in case of SARS-CoV-2, AZD1208 led to a complete removal of viral traces in cells. Kinome-analysis revealed vast changes in kinase landscape upon AZD1208 treatment, especially for inflammation and the PI3K/Akt-pathway. For ZIKV, a clear correlation between antiviral effect and increase in ISG-expression was observed. Based on a cell culture model with impaired ISG-induction, activation of the PI3K-Akt-mTOR axis, leading to major changes in the endolysosomal equilibrium, was identified as second pillar of the antiviral effect triggered by AZD1208-dependent Pim kinase inhibition, also against HBV. We identified Pim-kinases as cellular target for a broad antiviral activity. The antiviral effect exerted by inhibition of Pim kinases is based on at least two pillars: innate immunity and modulation of the endolysosomal system.


Subject(s)
Antiviral Agents , Immunity, Innate , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-pim-1 , SARS-CoV-2 , Signal Transduction , TOR Serine-Threonine Kinases , Humans , Immunity, Innate/drug effects , Antiviral Agents/pharmacology , TOR Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-pim-1/antagonists & inhibitors , Proto-Oncogene Proteins c-pim-1/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/immunology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Lysosomes/drug effects , Lysosomes/metabolism , Zika Virus/drug effects , Protein Kinase Inhibitors/pharmacology , Animals , Hepatitis B virus/drug effects , Endosomes/drug effects , Endosomes/metabolism , Cell Line , COVID-19/immunology , COVID-19/virology , COVID-19 Drug Treatment , Virus Replication/drug effects , Biphenyl Compounds , Thiazolidines
14.
Chem Biol Interact ; 394: 110990, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38579922

ABSTRACT

Swainsonine (SW) is the main toxic component of locoweed. Previous studies have shown that kidney damage is an early pathologic change in locoweed poisoning in animals. Trehalose induces autophagy and alleviates lysosomal damage, while its protective effect and mechanism against the toxic injury induced by SW is not clear. Based on the published literature, we hypothesize that transcription factor EB(TFEB) -regulated is targeted by SW and activating TFEB by trehalose would reverse the toxic effects. In this study, we investigate the mechanism of protective effects of trehalose using renal tubular epithelial cells. The results showed that SW induced an increase in the expression level of microtubule-associated protein light chain 3-II and p62 proteins and a decrease in the expression level of ATPase H+ transporting V1 Subunit A, Cathepsin B, Cathepsin D, lysosome-associated membrane protein 2 and TFEB proteins in renal tubular epithelial cells in a time and dose-dependent manner suggesting TFEB-regulated lysosomal pathway is adversely affected by SW. Conversely, treatment with trehalose, a known activator of TFEB promote TFEB nuclear translocation suggesting that TFEB plays an important role in protection against SW toxicity. We demonstrated in lysosome staining that SW reduced the number of lysosomes and increased the luminal pH, while trehalose could counteract these SW-induced effects. In summary, our results demonstrated for the first time that trehalose could alleviate the autophagy degradation disorder and lysosomal damage induced by SW. Our results provide an interesting method for reversion of SW-induced toxicity in farm animals and furthermore, activation of TFEB by trehalose suggesting novel mechanism of treating lysosomal storage diseases.


Subject(s)
Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Epithelial Cells , Kidney Tubules , Lysosomes , Swainsonine , Trehalose , Animals , Autophagy/drug effects , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Cell Line , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Kidney Tubules/drug effects , Kidney Tubules/pathology , Kidney Tubules/metabolism , Kidney Tubules/cytology , Lysosomes/metabolism , Lysosomes/drug effects , Swainsonine/toxicity , Trehalose/pharmacology
15.
Nanoscale ; 16(17): 8597-8606, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38602353

ABSTRACT

Frequently, subcellular-targeted drugs tend to accumulate in lysosomes after cellular absorption, a process termed the lysosomal trap. This accumulation often interferes with the drug's ability to bind to its target, resulting in decreased efficiency. Existing methods for addressing lysosome-induced drug resistance mainly involve improving the structures of small molecules or enveloping drugs in nanomaterials. Nonetheless, these approaches can lead to changes in the drug structure or potentially trigger unexpected reactions within organisms. To address these issues, we introduced a strategy that involves inactivating the lysosome with the use of Ag nanoparticles (Cy3.5@Ag NPs). In this method, the Cy3.5@Ag NPs gradually accumulate inside lysosomes, leading to permeation of the lysosomal membrane and subsequent lysosomal inactivation. In addition, Cy3.5@Ag NPs also significantly affected the motility of lysosomes and induced the occurrence of lysosome passivation. Importantly, coincubating Cy3.5@Ag NPs with various subcellular-targeted drugs was found to significantly increase the efficiency of these treatments. Our strategy illustrates the potential of using lysosomal inactivation to enhance drug efficacy, providing a promising therapeutic strategy for cancer.


Subject(s)
Lysosomes , Metal Nanoparticles , Silver , Lysosomes/metabolism , Lysosomes/drug effects , Silver/chemistry , Silver/pharmacology , Metal Nanoparticles/chemistry , Humans , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Drug Delivery Systems , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology
16.
J Am Chem Soc ; 146(18): 12836-12849, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38683943

ABSTRACT

The biological properties of two water-soluble organic cations based on polypyridyl structures commonly used as ligands for photoactive transition metal complexes designed to interact with biomolecules are investigated. A cytotoxicity screen employing a small panel of cell lines reveals that both cations show cytotoxicity toward cancer cells but show reduced cytotoxicity to noncancerous HEK293 cells with the more extended system being notably more active. Although it is not a singlet oxygen sensitizer, the more active cation also displayed enhanced potency on irradiation with visible light, making it active at nanomolar concentrations. Using the intrinsic luminescence of the cations, their cellular uptake was investigated in more detail, revealing that the active compound is more readily internalized than its less lipophilic analogue. Colocalization studies with established cell probes reveal that the active cation predominantly localizes within lysosomes and that irradiation leads to the disruption of mitochondrial structure and function. Stimulated emission depletion (STED) nanoscopy and transmission electron microscopy (TEM) imaging reveal that treatment results in distinct lysosomal swelling and extensive cellular vacuolization. Further imaging-based studies confirm that treatment with the active cation induces lysosomal membrane permeabilization, which triggers lysosome-dependent cell-death due to both necrosis and caspase-dependent apoptosis. A preliminary toxicity screen in the Galleria melonella animal model was carried out on both cations and revealed no detectable toxicity up to concentrations of 80 mg/kg. Taken together, these studies indicate that this class of synthetically easy-to-access photoactive compounds offers potential as novel therapeutic leads.


Subject(s)
Antineoplastic Agents , Cations , Phenazines , Humans , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Cations/chemistry , Cations/pharmacology , Phenazines/chemistry , Phenazines/pharmacology , Lysosomes/metabolism , Lysosomes/drug effects , HEK293 Cells , Apoptosis/drug effects , Drug Screening Assays, Antitumor , Cell Line, Tumor , Animals , Theranostic Nanomedicine , Molecular Structure
17.
Ecotoxicol Environ Saf ; 274: 116195, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38479315

ABSTRACT

Fluoride is known to induce nephrotoxicity; however, the underlying mechanisms remain incompletely understood. Therefore, this study aims to explore the roles and mechanisms of lysosomal membrane permeabilization (LMP) and the GSDME/HMGB1 axis in fluoride-induced nephrotoxicity and the protective effects of rutin. Rutin, a naturally occurring flavonoid compound known for its antioxidative and anti-inflammatory properties, is primarily mediated by inhibiting oxidative stress and reducing proinflammatory markers. To that end, we established in vivo and in vitro models. In the in vivo study, rats were exposed to sodium fluoride (NaF) throughout pregnancy and up until 2 months after birth. In parallel, we employed in vitro models using HK-2 cells treated with NaF, n-acetyl-L-cysteine (NAC), or rutin. We assessed lysosomal permeability through immunofluorescence and analyzed relevant protein expression via western blotting. Our findings showed that NaF exposure increased ROS levels, resulting in enhanced LMP and increased cathepsin B (CTSB) and D (CTSD) expression. Furthermore, the exposure to NaF resulted in the upregulation of cleaved PARP1, cleaved caspase-3, GSDME-N, and HMGB1 expressions, indicating cell death and inflammation-induced renal damage. Rutin mitigates fluoride-induced nephrotoxicity by suppressing ROS-mediated LMP and the GSDME/HMGB1 axis, ultimately preventing fluoride-induced renal toxicity occurrence and development. In conclusion, our findings suggest that NaF induces renal damage through ROS-mediated activation of LMP and the GSDME/HMGB1 axis, leading to pyroptosis and inflammation. Rutin, a natural antioxidative and anti-inflammatory dietary supplement, offers a novel approach to prevent and treat fluoride-induced nephrotoxicity.


Subject(s)
Fluorides , HMGB1 Protein , Kidney Diseases , Rutin , Animals , Rats , Anti-Inflammatory Agents/pharmacology , Antioxidants/metabolism , Caspase 3/metabolism , Fluorides/metabolism , Fluorides/toxicity , HMGB1 Protein/drug effects , HMGB1 Protein/metabolism , Inflammation/metabolism , Lysosomes/drug effects , Pyroptosis/drug effects , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/toxicity , Rutin/pharmacology , Sodium Fluoride/toxicity , Kidney Diseases/chemically induced , Kidney Diseases/drug therapy , Gasdermins/drug effects , Gasdermins/metabolism
18.
Acta Pharmacol Sin ; 45(5): 900-913, 2024 May.
Article in English | MEDLINE | ID: mdl-38225393

ABSTRACT

Autophagy impairment is a key factor in Alzheimer's disease (AD) pathogenesis. TFEB (transcription factor EB) and TFE3 (transcription factor binding to IGHM enhancer 3) are nuclear transcription factors that regulate autophagy and lysosomal biogenesis. We previously showed that corynoxine (Cory), a Chinese medicine compound, protects neurons from Parkinson's disease (PD) by activating autophagy. In this study, we investigated the effect of Cory on AD models in vivo and in vitro. We found that Cory improved learning and memory function, increased neuronal autophagy and lysosomal biogenesis, and reduced pathogenic APP-CTFs levels in 5xFAD mice model. Cory activated TFEB/TFE3 by inhibiting AKT/mTOR signaling and stimulating lysosomal calcium release via transient receptor potential mucolipin 1 (TRPML1). Moreover, we demonstrated that TFEB/TFE3 knockdown abolished Cory-induced APP-CTFs degradation in N2aSwedAPP cells. Our findings suggest that Cory promotes TFEB/TFE3-mediated autophagy and alleviates Aß pathology in AD models.


Subject(s)
Alzheimer Disease , Autophagy , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors , Disease Models, Animal , Transient Receptor Potential Channels , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Animals , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Autophagy/drug effects , Mice , Lysosomes/metabolism , Lysosomes/drug effects , Humans , Mice, Transgenic , Amyloid beta-Peptides/metabolism , Mice, Inbred C57BL , TOR Serine-Threonine Kinases/metabolism , Male , Proto-Oncogene Proteins c-akt/metabolism , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Signal Transduction/drug effects , Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/genetics
19.
J Biol Inorg Chem ; 29(2): 265-278, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38189962

ABSTRACT

Transition metal complexes with characteristics of unique packaging in nanoparticles and remarkable cancer cell cytotoxicity have emerged as potential alternatives to platinum-based antitumor drugs. Here we report the synthesis, characterization, and antitumor activities of three new Ruthenium complexes that introduce 5-fluorouracil-derived ligands. Notably, encapsulation of one such metal complex, Ru3, within pluronic® F-127 micelles (Ru3-M) significantly enhanced Ru3 cytotoxicity toward A549 cells by a factor of four. To determine the mechanisms underlying Ru3-M cytotoxicity, additional in vitro experiments were conducted that revealed A549 cell treatment with lysosome-targeting Ru3-M triggered oxidative stress, induced mitochondrial membrane potential depolarization, and drastically reduced intracellular ATP levels. Taken together, these results demonstrated that Ru3-M killed cells mainly via a non-apoptotic pathway known as oncosis, as evidenced by observed Ru3-M-induced cellular morphological changes including cytosolic flushing, cell swelling, and cytoplasmic vacuolation. In turn, these changes together caused cytoskeletal collapse and activation of porimin and calpain1 proteins with known oncotic functions that distinguished this oncotic process from other cell death processes. In summary, Ru3-M is a potential anticancer agent that kills A549 cells via a novel mechanism involving Ru(II) complex triggering of cell death via oncosis.


Subject(s)
Antineoplastic Agents , Coordination Complexes , Lysosomes , Poloxamer , Ruthenium , Humans , Poloxamer/chemistry , Poloxamer/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , A549 Cells , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Ruthenium/chemistry , Ruthenium/pharmacology , Coordination Complexes/pharmacology , Coordination Complexes/chemistry , Coordination Complexes/chemical synthesis , Membrane Potential, Mitochondrial/drug effects , Drug Screening Assays, Antitumor , Oxidative Stress/drug effects
20.
Ecotoxicol Environ Saf ; 249: 114381, 2023 Jan 01.
Article in English | MEDLINE | ID: mdl-36508801

ABSTRACT

Black carbon (BC) is an important component of atmospheric PM 2.5 and the second largest contributor to global warming. 1,4-naphthoquinone-coated BC (1,4 NQ-BC) is a secondary particle with great research value, so we chose 1,4 NQ-BC as the research object. In our study, mitochondria and lysosomes were selected as targets to confirm whether they were impaired by 1,4 NQ-BC, label free proteomics technology, fluorescent probes, qRT-PCR and western blots were used to investigate the mechanism of 1,4 NQ-BC toxicity. We found 494 differentially expressed proteins (DEPs) in mitochondria and 86 DEPs in lysosomes using a proteomics analysis of THP1 cells after 1,4 NQ-BC exposure for 24 h. Through proteomics analysis and related experiments, we found that 1,4 NQ-BC can damage THP-1-M cells by obstructing autophagy, increasing lysosomal membrane permeability, disturbing the balance of ROS, and reducing the mitochondrial membrane potential. It is worth noting that 1,4 NQ-BC prevented the removal of FTL by inhibiting autophagy, and increased IL-33 level by POR/FTL/IL-33 axis. We first applied proteomics to study the damage mechanism of 1,4 NQ-BC on THP1 cells. Our research will enrich knowledge of the mechanism by which 1,4 NQ-BC damages human macrophages and identify important therapeutic targets and adverse outcome pathways for 1,4 NQ-BC-induced damage.


Subject(s)
Apoferritins , Autophagy , Interleukin-33 , Lysosomes , Naphthoquinones , Soot , Humans , Apoferritins/metabolism , Autophagy/drug effects , Interleukin-33/metabolism , Macrophages/drug effects , Naphthoquinones/toxicity , Soot/toxicity , Up-Regulation , Lysosomes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...