Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters










Publication year range
1.
J Nanobiotechnology ; 22(1): 241, 2024 May 12.
Article in English | MEDLINE | ID: mdl-38735933

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) incidence is increasing in recent years due to intestinal flora imbalance, making oral probiotics a hotspot for research. However, numerous studies related to intestinal flora regulation ignore its internal mechanisms without in-depth research. RESULTS: Here, we developed a probiotic microgel delivery system (L.r@(SA-CS)2) through the layer-by-layer encapsulation technology of alginate (SA) and chitosan (CS) to improve gut microbiota dysbiosis and enhance anti-tumor therapeutic effect. Short chain fatty acids (SCFAs) produced by L.r have direct anti-tumor effects. Additionally, it reduces harmful bacteria such as Proteobacteria and Fusobacteriota, and through bacteria mutualophy increases beneficial bacteria such as Bacteroidota and Firmicutes which produce butyric acid. By binding to the G protein-coupled receptor 109A (GPR109A) on the surface of colonic epithelial cells, butyric acid can induce apoptosis in abnormal cells. Due to the low expression of GPR109A in colon cancer cells, MK-6892 (MK) can be used to stimulate GPR109A. With increased production of butyrate, activated GPR109A is able to bind more butyrate, which further promotes apoptosis of cancer cells and triggers an antitumor response. CONCLUSION: It appears that the oral administration of L.r@(SA-CS)2 microgels may provide a treatment option for CRC by modifying the gut microbiota.


Subject(s)
Fatty Acids, Volatile , Gastrointestinal Microbiome , Limosilactobacillus reuteri , Probiotics , Gastrointestinal Microbiome/drug effects , Probiotics/pharmacology , Humans , Fatty Acids, Volatile/metabolism , Animals , Limosilactobacillus reuteri/metabolism , Mice , Chitosan/chemistry , Alginates/chemistry , Alginates/pharmacology , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Administration, Oral , Colorectal Neoplasms/drug therapy , Cell Line, Tumor , Receptors, G-Protein-Coupled/metabolism , Microgels/chemistry , Mice, Inbred BALB C , Butyric Acid/pharmacology , Butyric Acid/metabolism
2.
World J Gastroenterol ; 30(15): 2155-2174, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38681991

ABSTRACT

BACKGROUND: Necroptosis has emerged as a novel molecular pathway that can be targeted by chemotherapy agents in the treatment of cancer. OSW-1, which is derived from the bulbs of Ornithogalum saundersiae Baker, exerts a wide range of pharmacological effects. AIM: To explore whether OSW-1 can induce necroptosis in colorectal cancer (CRC) cells, thereby expanding its range of clinical applications. METHODS: We performed a sequence of functional experiments, including Cell Counting Kit-8 assays and flow cytometry analysis, to assess the inhibitory effect of OSW-1 on CRC cells. We utilized quantitative proteomics, employing tandem mass tag labeling combined with liquid chromatography-tandem mass spectrometry, to analyze changes in protein expression. Subsequent bioinformatic analysis was conducted to elucidate the biological processes associated with the identified proteins. Transmission electron microscopy (TEM) and immunofluorescence studies were also performed to examine the effects of OSW-1 on necroptosis. Finally, western blotting, siRNA experiments, and immunoprecipitation were employed to evaluate protein interactions within CRC cells. RESULTS: The results revealed that OSW-1 exerted a strong inhibitory effect on CRC cells, and this effect was accompanied by a necroptosis-like morphology that was observable via TEM. OSW-1 was shown to trigger necroptosis via activation of the RIPK1/RIPK3/MLKL pathway. Furthermore, the accumulation of p62/SQSTM1 was shown to mediate OSW-1-induced necroptosis through its interaction with RIPK1. CONCLUSION: We propose that OSW-1 can induce necroptosis through the RIPK1/RIPK3/MLKL signaling pathway, and that this effect is mediated by the RIPK1-p62/SQSTM1 complex, in CRC cells. These results provide a theoretical foundation for the use of OSW-1 in the clinical treatment of CRC.


Subject(s)
Colorectal Neoplasms , Necroptosis , Plant Extracts , Receptor-Interacting Protein Serine-Threonine Kinases , Sequestosome-1 Protein , Signal Transduction , Humans , Cell Line, Tumor , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/genetics , HCT116 Cells , Necroptosis/drug effects , Plant Extracts/pharmacology , Protein Kinases/metabolism , Proteomics/methods , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Sequestosome-1 Protein/metabolism , Sequestosome-1 Protein/genetics
3.
Food Funct ; 15(9): 4970-4982, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38606509

ABSTRACT

Inhibition of ferroptosis in intestinal epithelial cells serves as an attractive target for the development of therapeutic strategies for colitis. Pinobanksin, one of the main flavonoids derived from propolis, possesses significant anti-inflammatory effects and inhibits the cell death of several cell lines. Here, we evaluated whether pinobanksin influenced colitis by modulation of epithelial ferroptosis. Mice treated with 2.5% DSS dissolved in sterile distilled water were established for an acute colitis model. The mitochondrial morphology, colonic iron level, lipid peroxidation products MDA/4-HNE, and lipid reactive oxygen species levels were measured to assess ferroptosis in epithelial cells. RNA-seq and functional analyses were performed to reveal key genes mediating pinobanksin-exerted modulation of ferroptosis. We found that pinobanksin, at different doses, induced significant anti-colitis effects and inhibited the elevated ferroptosis in colonic epithelial cells isolated from DSS-treated mice largely by activating GPX4 (negative regulator of ferroptosis). Furthermore, RNA-seq assays indicated that pinobanksin significantly increased the cystine transporter SLC7A11 in colonic tissues from mice with colitis. Depletion of SLC7A11 largely blocked pinobanksin-induced promotion of cystine uptake/glutathione biosynthesis and suppression of ferroptosis in epithelial cells from mice with colitis or IEC-6 cells pretreated with RSL3. Altogether, pinobanksin alleviated DSS-induced colitis largely by inhibition of ferroptosis in epithelial cells. Activation of SLC7A11 by pinobanksin resulted in the promotion of cystine uptake and enhancement of glutathione biosynthesis. This work will provide novel guidance for the clinical use of pinobanksin to treat colitis through inhibition of epithelial ferroptosis.


Subject(s)
Amino Acid Transport System y+ , Colitis , Ferroptosis , Glutathione , Animals , Humans , Male , Mice , Amino Acid Transport System y+/metabolism , Amino Acid Transport System y+/genetics , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Ferroptosis/drug effects , Flavonoids/pharmacology , Glutathione/metabolism , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Reactive Oxygen Species/metabolism
4.
Int J Pharm ; 652: 123810, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38244648

ABSTRACT

Transforming growth factor ß (TGF-ß), a versatile immunosuppressive cytokine, has gained increasing attention as a potential target for cancer immunotherapy. However, current strategies are constrained by tumor heterogeneity and drug resistance. Therapeutic probiotics, such as Escherichia coli Nissle1917 (EcN), not only regulate the gut microbiota to increase beneficial bacteria with anti-tumor effects, but also modulate immune factors within the body, thereby enhancing immunity. In this study, we developed an oral microgel delivery system of EcN@(CS-SA)2 by electrostatic interaction between chitosan (CS) and sodium alginate (SA), aiming to enhance its bioavailability in the gastrointestinal tract (GIT). Notably, EcN@(CS-SA)2 microgel showed a synergistic enhancement of the anti-tumor efficacy of Galunisertib (Gal, a TGF-ß inhibitor) by inducing apoptosis and immunogenic cell death (ICD) in tumor cells, as well as promoting increased infiltration of CD8+ T cells into the tumor microenvironment (TME).


Subject(s)
Colorectal Neoplasms , Microgels , Probiotics , Pyrazoles , Quinolines , Humans , Transforming Growth Factor beta/metabolism , CD8-Positive T-Lymphocytes , Immunotherapy , Colorectal Neoplasms/drug therapy , Immunity , Tumor Microenvironment , Cell Line, Tumor
5.
Asian J Pharm Sci ; 18(5): 100850, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37920651

ABSTRACT

The high nutrient and energy demand of tumor cells compared to normal cells to sustain rapid proliferation offer a potentially auspicious avenue for implementing starvation therapy. However, conventional starvation therapy, such as glucose exhaustion and vascular thrombosis, can lead to systemic toxicity and exacerbate tumor hypoxia. Herein, we developed a new "valve-off" starvation tactic, which was accomplished by closing the valve of glucose transporter protein 1 (GLUT1). Specifically, dihydroartemisinin (DHA), 2,20-azobis [2-(2-imidazolin-2-yl) propane] dihydrochloride (AI), and Ink were co-encapsulated in a sodium alginate (ALG) hydrogel. Upon irradiation with the 1064 nm laser, AI rapidly disintegrated into alkyl radicals (R•), which exacerbated the DHA-induced mitochondrial damage through the generation of reactive oxygen species and further reduced the synthesis of adenosine triphosphate (ATP). Simultaneously, the production of R• facilitated DHA-induced starvation therapy by suppressing GLUT1, which in turn reduced glucose uptake. Systematic in vivo and in vitro results suggested that this radical-enhanced "valve-off" strategy for inducing tumor cell starvation was effective in reducing glucose uptake and ATP levels. This integrated strategy induces tumor starvation with efficient tumor suppression, creating a new avenue for controlled, precise, and concerted tumor therapy.

7.
Food Funct ; 13(18): 9470-9480, 2022 Sep 22.
Article in English | MEDLINE | ID: mdl-35983876

ABSTRACT

Inhibition of ferroptosis in intestinal epithelial cells ameliorates clinical symptoms and improves endoscopic presentations in inflammatory bowel disease (IBD). Licorice is used worldwide in food and medicine fields. Liquiritin, a flavonoid component in licorice, is an effective substance used as an anti-inflammatory, antioxidant food that has been shown to improve chemically induced colitis. Herein we evaluated the therapeutic effects of liquiritin on colitis and determined whether liquiritin could affect colitis by modulating ferroptosis in epithelial cells. A colitis model was induced in mice by oral administration with 2.5% DSS dissolved in drinking water. The results showed that liquiritin significantly alleviated symptoms, suppressed intestinal inflammation and restored the epithelial barrier function in the colitis mouse model. Liquiritin supplementation upregulated colonic ferritin expression, increased the storage of cellular iron, reduced the cellular iron level and further inhibited ferroptosis in epithelial cells from the colitis model. Pharmacological stimulation of ferroptosis largely blocked liquiritin-induced alleviation of colitis. Peroxiredoxin-6 (Prdx6) expression was significantly decreased in the DSS group, which was reversed by liquiritin treatment. Genetic or pharmacological silencing of Prdx6 largely reversed liquiritin-induced modulation of the ferritin/iron level and ferroptosis in epithelial cells. Molecular docking results showed that liquiritin could bind to Prdx6 through the hydrogen bond interaction with amino acid residues Thr208, Val206 and Pro203. In conclusion, liquiritin treatment largely alleviated DSS induced colitis by inhibiting ferroptosis in epithelial cells. Liquiritin negatively regulated ferroptosis in epithelial cells in colitis by activating Prdx6, increasing the expression of ferritin and subsequently reducing the cellular iron level.


Subject(s)
Colitis , Ferroptosis , Flavanones , Peroxiredoxin VI , Amino Acids/metabolism , Animals , Anti-Inflammatory Agents/metabolism , Antioxidants/metabolism , Colitis/chemically induced , Colitis/drug therapy , Colitis/genetics , Dextran Sulfate/adverse effects , Disease Models, Animal , Epithelial Cells/metabolism , Ferritins/metabolism , Flavanones/pharmacology , Glucosides/pharmacology , Iron/metabolism , Mice , Mice, Inbred C57BL , Molecular Docking Simulation , Peroxiredoxin VI/metabolism
8.
J Colloid Interface Sci ; 614: 436-450, 2022 May 15.
Article in English | MEDLINE | ID: mdl-35108635

ABSTRACT

Although low-temperature photothermal therapy (PTT) can sensitize tumors to immune checkpoint inhibition, its efficacy is still restricted in the deep and internal tumors without enough oxygen and lymphocytic infiltration. Non-oxygen-dependent alkyl radicals have been demonstrated to synergistically enhance PTT through up-regulating lipid peroxidation and reactive oxygen species (ROS). Herein, an innovative strategy based on alkyl radicals to augment immunogenetic cell death (ICD) caused by mild PTT was proposed to improve poor efficacy of immunotherapy, which composed of a photothermal material of Chinse ink, an azo-initiator of 2,2-azobis[2-(2-imidazoline-2-acyl)propane]dihydrochloride (AIPH) and a PD-L1 inhibitor of HY19991 (HY). Upon near-infrared-II laser irradiation, low-temperature (<45℃) stimulation induced a high expression of immune checkpoint receptor (PD-L1) in tumors and triggered a large amount alkyl radicals generated by AIPH. Significantly, the alkyl radicals augmented the ICD and increased the recruitment of tumor-infiltrating lymphocytes against tumors after transformation of the immunologically cold tumor microenvironment into hot by mild PTT. The released HY further enhanced the immunotherapy effect by blocking the binding of activated T lymphocytes and PD-L1. In vivo studies exhibited that the all-in-one hydrogel with synergistic mechanisms had an extraordinary ability to reverse the immunosuppressive microenvironment, stimulate innate and adaptive immune responses to eliminate tumors and prevent metastasis.


Subject(s)
Immunotherapy , Neoplasms , Cell Line, Tumor , Humans , Phototherapy , Temperature , Tumor Microenvironment
9.
Mater Today Bio ; 12: 100154, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34778741

ABSTRACT

Ferroptosis has received ever-increasing attention due to its unparalleled mechanism in eliminating resistant tumor cells. Nevertheless, the accumulation of toxic lipid peroxides (LPOs) at the tumor site is limited by the level of lipid oxidation. Herein, by leveraging versatile sodium alginate (ALG) hydrogel, a localized ferroptosis trigger consisting of gambogic acid (GA), 2,2'-azobis [2-(2-imidazolin-2-yl) propane] dihydrochloride (AIPH), and Ink (a photothermal agent), was constructed via simple intratumor injection. Upon 1064 â€‹nm laser irradiation, the stored AIPH rapidly decomposed into alkyl radicals (R•), which aggravated LPOs in tumor cells. Meanwhile, GA could inhibit heat shock protein 90 (HSP90) to reduce the heat resistance of tumor cells, and forcefully consume glutathione (GSH) to weaken the antioxidant capacity of cells. Systematic in vitro and in vivo experiments have demonstrated that synchronous consumption of GSH and increased reactive oxygen species (ROS) facilitated reduced expression of glutathione peroxidase 4 (GPX4), which further contributed to disruption of intracellular redox homeostasis and ultimately boosted ferroptosis. This all-in-one strategy has a highly effective tumor suppression effect by depleting and generating fatal active compounds at tumor sites, which would pave a new route for the controllable, accurate, and coordinated tumor treatments.

10.
Drug Des Devel Ther ; 14: 5623-5632, 2020.
Article in English | MEDLINE | ID: mdl-33376306

ABSTRACT

INTRODUCTION: Paeoniflorin is a main active component in traditional Chinese medicine. Paeoniae alba radix is widely used as a spasmolytic and pain-relieving agent for abdominal spasmodic pain. Functional dyspepsia (FD) is characterized by pain or burning in the epigastrium, fullness, bloating and nausea. However, limited information is available about the effect of paeoniflorin on FD. MATERIALS AND METHODS: In this study, iodoacetamide or clonidine-induced FD rat models were established to investigate the impacts of paeoniflorin on FD induced by different pathophysiologic disturbances. RESULTS: We found the therapeutic effect of paeoniflorin through assessing the gastric emptying, gastric accommodation and visceral hypersensitivity. This function of paeoniflorin was related to the release of acetylcholine (ACh), which was accompanied by reduced acetylcholinesterase (AchE) activity in stomach and hypothalamus. Paeoniflorin administration inhibited the cyclo-oxygenase-2 (COX-2) expression and increased the level of ghrelin in the stomach. Besides, the levels of occludin and ZO-1 were elevated in the duodenum from paeoniflorin-treated rats, suggesting the impaired duodenal barrier was ameliorated. DISCUSSION: These results indicate that paeoniflorin possesses the ability to alleviate functional dyspepsia.


Subject(s)
Acetylcholine/metabolism , Dyspepsia/drug therapy , Glucosides/pharmacology , Monoterpenes/pharmacology , Animals , Clonidine , Disease Models, Animal , Dyspepsia/chemically induced , Dyspepsia/metabolism , Iodoacetamide , Male , Rats , Rats, Sprague-Dawley
11.
J Nutr Biochem ; 75: 108261, 2020 01.
Article in English | MEDLINE | ID: mdl-31710934

ABSTRACT

Aging-related cholinergic dysfunction, extensive neuroinflammation and oxidative stress in brain are predominant pathogenic factors for dementia. In the present study, we aimed to evaluate the protective effects of piperine, an alkaloid nutrient component of Piper nigrum, against cognitive impairment in a senescent mouse model induced by D-galactose (D-Gal) and to explore the underlying mechanisms. Senescent mouse model was established by repeated subcutaneous injection of D-Gal (150 mg/kg, once daily for 42 days). Fourteen days after the first D-Gal exposure, piperine (2.5, 5, 10 mg/kg) or vehicle was intraperitoneally administered once daily for 28 days. The cognitive function of mice was evaluated by Morris water maze test (MWM). Twenty-four hours after behavioral test, the cholinergic function and oxidative stress level in mouse hippocampus were measured by spectrophotometric assays. In addition, the hippocampal levels of proinflammatory cytokines, including tumor necrosis factor-α, interleukin-1ß and interleukin-6, were quantified using enzyme-linked immunosorbent assay. Expressions of glycogen synthase kinase-3ß (GSK-3ß) and its upstream or downstream molecules including phosphatidylinositol 3-kinase (PI3K),protein kinase B (AKT), protein kinase C (PKC), NF-E2-related factor 2, nuclear factor-κB and microtubule-associated protein tau in hippocampus were determined by western blotting, immunohistochemical or immunofluorescent staining. Our data revealed that chronic D-Gal exposure in mice led to cognitive impairment in MWM, along with cholinergic malfunction, extensive oxidative stress and neuroinflammation, as well as hyperphosphorylation of tau protein in hippocampus. All these neurochemical, neuroinflammatory and cognitive alterations could be ameliorated by 4-week repeated piperine administration. Moreover, piperine also reversed D-Gal-induced GSK-3ß activation through modulating PKC and PI3K/AKT pathways in senescent mouse hippocampus, suggesting GSK-3ß-related signaling might be involved in the benefits of piperine against D-Gal-induced cognitive decline in mice.


Subject(s)
Aging , Alkaloids/pharmacology , Benzodioxoles/pharmacology , Cognition Disorders/drug therapy , Glycogen Synthase Kinase 3 beta/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Signal Transduction , Animals , Behavior, Animal , Cellular Senescence , Cognition , Cytokines/metabolism , Galactose , Glutathione/metabolism , Hippocampus/metabolism , Inflammation , Lipid Peroxidation , Male , Maze Learning , Mice , Models, Animal , Neurons/metabolism , Oxidative Stress , Protein Kinase C/metabolism , Reactive Oxygen Species/metabolism
12.
ACS Appl Mater Interfaces ; 11(42): 38555-38567, 2019 Oct 23.
Article in English | MEDLINE | ID: mdl-31558017

ABSTRACT

As a novel treatment modality of tumors, hypothermal hyperthermia employed relatively lower temperature (<45 °C) to damage cancer cells with mild toxicity to normal tissues. However, beyond that inducible heat resistance of tumor cells, the discounted therapeutic effect of low temperature hyperthermia was also ascribed to poor penetration of exogenous light stimulation and low accumulation of photothermal agents in tumor sites. Herein, we constructed a multifunctional in situ hydrogel of sodium alginate (ALG) via Ca2+ coordinated with ALG to encapsulate the photothermal agent of Ink and azo initiator of 2,2'-azobis[2-(2-imidazolin-2-yl)propane]dihydrochloride (AIPH) for effective tumor treatment. The designed ALG hydrogel was used to improve the therapeutic effect by increased accumulation of Ink and AIPH and avoid potential side-effects caused by the unexpected spread to the surrounding normal tissues. After injection, local low temperature stimulation was generated with near-infrared-II irradiation by a 1064 nm laser, triggering rapid decomposition of AIPH to produce alkyl radicals. The synergistic low temperature photothermal therapy and cytotoxic-free radicals enhanced the apoptosis of tumor cells via physical heat damage and lipid peroxidation. Thus, remarkable inhibition of tumor growth was observed in a subcutaneous colorectal cancer with negligible side effects. Furthermore, the formulation could also exert strong photoacoustic signals, which were utilized to monitor the stability of the composite hydrogel.


Subject(s)
Azo Compounds/chemistry , Free Radicals/chemistry , Imidazoles/chemistry , Infrared Rays , Alginates/chemistry , Animals , Apoptosis/drug effects , Azo Compounds/pharmacology , Azo Compounds/therapeutic use , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Biocompatible Materials/therapeutic use , Chromatography, High Pressure Liquid , HCT116 Cells , Humans , Hydrogels/chemistry , Imidazoles/pharmacology , Imidazoles/therapeutic use , Ink , Lipids/analysis , Mass Spectrometry , Membrane Potential, Mitochondrial , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/therapy , Oxidative Stress/drug effects , Temperature
13.
Pharmacol Res ; 148: 104461, 2019 10.
Article in English | MEDLINE | ID: mdl-31542404

ABSTRACT

Intestinal epithelial barrier dysfunction is a key pathology of colitis. Autophagy of epithelial cells maintains homeostasis of the intestinal barrier by inhibiting apoptosis and stimulating degradation of the tight junction protein claudin-2. This study investigated the effects and mechanism of activity of sinensetin, a polymethylated flavonoid isolated from tangerine peel and citrus, on intestinal barrier dysfunction in colitis. Animal model of colitis were established by intracolonic administration of 2, 4, 6-trinitrobenzene sulfonic acid and oral treatment with dextran sulfate sodium. Epithelial barrier function was evaluated by measuring the serum recovery of fluorescein isothiocyanate-4 kD dextran in vivo and transepithelial electrical resistance in Caco-2 cells, respectively. Epithelial cell autophagy assayed by autophagosome formation and expression of autophagy-related protein. Sinensetin reversed colitis-associated increase in intestinal permeability, significantly promoted epithelial cell autophagy, and further decreased epithelial cell apoptosis, and reduced mucosal claudin-2. Sinenstetin alleviated colitis symptoms rats and mice with colitis. Knockdown of 5' adenosine monophosphate-activated protein kinase (AMPK) reversed the promotion of epithelial autophagy by sinensetin. In conclusion, sinensetin significantly alleviated intestinal barrier dysfunction in colitis by promoting epithelial cell autophagy, and further inhibiting apoptosis and promoting claudin-2 degradation. The results highlighted novel potential benefits of sinensetin in colitis.


Subject(s)
Autophagy/drug effects , Colitis/drug therapy , Epithelial Cells/drug effects , Flavonoids/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Caco-2 Cells , Claudin-2/metabolism , Colitis/metabolism , Dextran Sulfate/pharmacology , Disease Models, Animal , Epithelial Cells/metabolism , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/metabolism , Intestines/drug effects , Male , Mice , Mice, Inbred C57BL , Permeability/drug effects , Rats , Rats, Sprague-Dawley , Tight Junctions/drug effects , Tight Junctions/metabolism
14.
J Nutr Biochem ; 70: 147-155, 2019 08.
Article in English | MEDLINE | ID: mdl-31207354

ABSTRACT

Piperine, the major alkaloid constituent of black pepper, has been reported to possess a wide range of pharmacological effects on the central nervous system, including antidepressant, anticonvulsant and anti-ischemic activities. In the present study, we aimed to investigate the therapeutic potential and neuroprotective mechanisms of piperine in an experimental mouse model of sporadic Alzheimer's disease (sAD) induced by intracerebroventricular (ICV) infusion of streptozotocin (STZ). STZ was infused bilaterally at a dose of 1.5 mg/kg/day on day 1 and day 3. From day 8, piperine (2.5-10 mg/kg body weight) was administered intraperitoneally once daily for 15 consecutive days. The locomotor activity and cognitive performance of mice were evaluated using open field test and Morris water maze test, respectively. On day 23, all animals were sacrificed, and the hippocampus was used for biochemical, neurochemical and neuroinflammatory determinations. Our data revealed that the ICV-STZ-infused sAD mouse showed an increased oxidative-nitrosative stress, an altered neurotransmission and an elevated neuroinflammation in hippocampus, as well as significant cognitive deficits. All these alterations can be ameliorated by piperine in a dose-dependent manner. In summary, our findings predict a therapeutic potential of piperine against cognitive deficits in sAD mouse. This effect might be due to its abilities to ameliorate oxidative-nitrosative stress, restore neurotransmission and reduce neuroinflammation.


Subject(s)
Alkaloids/pharmacology , Alzheimer Disease/drug therapy , Benzodioxoles/pharmacology , Cognition Disorders/drug therapy , Neuroprotective Agents/pharmacology , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Alzheimer Disease/chemically induced , Animals , Cognition Disorders/chemically induced , Disease Models, Animal , Hippocampus/metabolism , Inflammation , Infusions, Intraventricular , Male , Maze Learning , Memory Disorders , Mice , Nitrogen/chemistry , Oxidative Stress , Streptozocin
15.
Biomaterials ; 195: 51-62, 2019 03.
Article in English | MEDLINE | ID: mdl-30610993

ABSTRACT

Carbon monoxide (CO) gas therapy combined with chemotherapy and photothermal therapy (PTT) is a promising treatment mode for malignant tumor. Herein, we firstly reported doxorubicin (DOX) loaded Mn carbonyl modified Fe (Ⅲ)-based nanoMOFs (MIL-100) coated PEGylated magnetic carbon nanoparticles (denoted as MCM@PEG-CO-DOX NPs) as theranostics nanoplatforms for near-infrared (NIR)-responded CO-DOX combination therapy. MIL-100 as a good nanocarrier of DOX with high loading capacity can also chelate the Mn carbonyl after a smart modification. Meanwhile, magnetic carbon core possessed photothermal effect, which can convert the NIR light to heat by an 808 nm laser irradiation, resulting in the on-demand release of CO and DOX. As a result, combining with PTT, MCM@PEG-CO-DOX NPs killed tumor efficiently. Moreover, our synthesized MCM@PEG-CO-DOX NPs were capable of realizing tumor dual-mode imaging including magnetic resonance imaging (MRI) and photoacoustic imaging (PAI).


Subject(s)
Doxorubicin/chemistry , Drug Delivery Systems/methods , Nanoparticles/chemistry , Drug Liberation , Mannose/chemistry
16.
Front Immunol ; 9: 83, 2018.
Article in English | MEDLINE | ID: mdl-29472916

ABSTRACT

Activation of phosphatase and tensin homolog (PTEN) is known to induce cell apoptosis. MicroRNA-374a (miR-374a), which can suppress PTEN expression, has been found abnormally expressed in inflammatory bowel disease (IBD). Fortunellin is a citrus flavonoid that is a potential anti-inflammation agent in inflammatory diseases. The present study investigated the effects and mechanisms underlying fortunellin-induced inhibition of PTEN in IBD. Colitis was established in rats by the intracolonic administration of 2,4,6-trinitrobenzene sulfonic acid to mimic human ulcerative colitis, which is the main type of IBD. miR-374a expression was measured by quantitative real-time polymerase chain reaction, and the regulation of PTEN by miR-374a was evaluated by dual luciferase reporter assay. Western blotting was used to measure the corresponding protein expression. Fortunellin ameliorated colitis symptoms, including excessive inflammation and oxidative stress. Fortunellin decreased epithelial cell apoptosis through inhibiting PTEN expression in colitis. Fortunellin-induced downregulation of PTEN could be counteracted by miR-374a depletion. Moreover, knockdown of miR-374a in vivo partly inhibited the effects of fortunellin on rat colitis. In conclusion, PTEN inhibition contributes to the amelioration effects of fortunellin on colitis. It was confirmed that fortunellin targets miR-374a, which is a negative regulator of PTEN. This study provides novel insights into the pathological mechanisms and treatment alternatives of colitis.


Subject(s)
Colitis/etiology , Colitis/metabolism , Flavonoids/pharmacology , Glycosides/pharmacology , Intestinal Mucosa/metabolism , MicroRNAs/genetics , PTEN Phosphohydrolase/genetics , Animals , Antagomirs/genetics , Apoptosis/drug effects , Apoptosis/genetics , Biopsy , Cell Membrane Permeability , Colitis/pathology , Disease Models, Animal , Flavonoids/adverse effects , Gene Expression Regulation , Glycosides/adverse effects , Intestinal Mucosa/pathology , Male , Oxidative Stress , RNA Interference , Rats , Reactive Oxygen Species/metabolism , Symptom Assessment
17.
J Mater Chem B ; 6(43): 7033-7041, 2018 Nov 21.
Article in English | MEDLINE | ID: mdl-32254586

ABSTRACT

Polypyrrole nanoparticles (PPy NPs) have been extensively studied for photothermal therapy (PTT) of tumors because they can generate heat upon near-infrared (NIR) irradiation. Developing non-toxic, self-targeting and long-circulating PPy biomaterials to maximize photothermal effects remains challenging. Here, we show that PPy NPs camouflaged with fusing red blood cells (RBC) and platelet (PLT) membranes can kill tumor cells under direct near infrared irradiation (NIR). The resulting RBC-PLT hybrid membrane-coated PPy NPs (PPy@[R-P] NPs) possess characteristics of both RBC and PLT, exhibiting long circulation times and self-targeting properties. After administration of PPy@[R-P] NPs via tail vein, tumor vessels were injured by photothermal stimulation under NIR laser exposure, which induced a large amount of microthrombosis. Due to the existence of PLT membranes, a large number of PPy@[R-P] NPs were successfully recruited to the microthrombosis sites. As a result, the distribution of nanomaterials in the tumor tissues was improved, and excellent photothermal treatment was achieved. The resulting PPy@[R-P] NPs may contribute to anti-tumor PTT.

18.
Biochem Biophys Res Commun ; 495(1): 867-872, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29170134

ABSTRACT

Combined use of different therapies is conducive to improve the effectiveness of cancer therapy. The chemo-photothermal therapy is commonly used in one nanocarrier to provide an excellent synergistic effect for cancer therapy over photothermal therapy (PTT) or chemotherapy alone. In this study, biocompatible and monodisperse hollow carbon nanospheres (HCNs) were developed as a multifunctional platform for the delivery of paclitaxel (PTX) and PTT of cancer simultaneously. The mesoporous HCNs have large pore volume and proper channels for loading and release of PTX. Upon near-infrared (NIR) laser illumination, the photothermal mediator of HCNs could effectively convert absorbed light into heat, which triggered rapid release of chemotherapeutic drug from HCNs through dissociating the interactions between PTX and HCNs by heat energy. A large number of tumor cells were significantly destroyed when hct116 cells treated with PTX@HCNs were irradiated, which was mainly attribute to the synergistic result of HCNs-mediated photothermal damage and cytotoxicity of light-triggered PTX release.


Subject(s)
Nanocapsules/administration & dosage , Nanopores/ultrastructure , Nanospheres/administration & dosage , Neoplasms, Experimental/therapy , Paclitaxel/administration & dosage , Phototherapy/methods , Antineoplastic Agents/administration & dosage , Carbon/chemistry , Combined Modality Therapy/methods , HCT116 Cells , Humans , Hyperthermia, Induced/methods , Laser Therapy/methods , Nanocapsules/chemistry , Nanocapsules/ultrastructure , Nanospheres/chemistry , Neoplasms, Experimental/pathology , Treatment Outcome
19.
Biomaterials ; 143: 130-141, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28800434

ABSTRACT

In this study, we reported a strategy to improve delivery efficiency of a long-circulation biomimetic photothermal nanoagent for enhanced photothermal therapy through selectively dilating tumor vasculature. By using a simply nanocoating technology, a biomimetic layer of natural red blood cell (RBC) membranes was camouflaged on the surface of photothermal polypyrrole nanoparticles (PPy@RBC NPs). The erythrocyte-mimicking PPy NPs inherited the immune evasion ability from natural RBC resulting in superior prolonged blood retention time. Additionally, excellent photothermal and photoacoustic imaging functionalities were all retained attributing to PPy NPs cores. To further improve the photothermal outcome, the endothelin A (ETA) receptor antagonist BQ123 was jointly employed to regulate tumor microenvironment. The BQ123 could induce tumor vascular relaxation and increase blood flow perfusion through modulating an ET-1/ETA transduction pathway and blocking the ETA receptor, whereas the vessel perfusion of normal tissues was not altered. Through our well-designed tactic, the concentration of biomimetic PPy NPs in tumor site was significantly improved when administered systematically. The study documented that the antitumor efficiency of biomimetic PPy NPs combined with specific antagonist BQ123 was particularly prominent and was superior to biomimetic PPy NPs (P < 0.05) and PEGylated PPy NPs with BQ123 (P < 0.01), showing that the greatly enhanced photothermal treatment could be achieved with low-dose administration of photothermal agents. Our findings would provide a promising procedure for other similar enhanced photothermal treatment by blocking ETA receptor to dramatically increase the delivery of biomimetic photothermal nanomaterials.


Subject(s)
Endothelin Receptor Antagonists/therapeutic use , Hyperthermia, Induced/methods , Nanoparticles/therapeutic use , Neoplasms/therapy , Peptides, Cyclic/therapeutic use , Phototherapy/methods , Polymers/therapeutic use , Pyrroles/therapeutic use , Animals , Biomimetic Materials/chemistry , Biomimetic Materials/therapeutic use , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/therapeutic use , Endothelin Receptor Antagonists/chemistry , Erythrocyte Membrane/chemistry , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Nanoparticles/chemistry , Neoplasms/blood supply , Peptides, Cyclic/chemistry , Polymers/chemistry , Pyrroles/chemistry , RAW 264.7 Cells
20.
Front Pharmacol ; 8: 292, 2017.
Article in English | MEDLINE | ID: mdl-28588494

ABSTRACT

Myosin light chain kinase (MLCK) induces contraction of the perijunctional apical actomyosin ring in response to phosphorylation of the myosin light chain. Abnormal expression of MLCK has been observed in respiratory diseases, pancreatitis, cardiovascular diseases, cancer, and inflammatory bowel disease. The signaling pathways involved in MLCK activation and triggering of endothelial barrier dysfunction are discussed in this review. The pharmacological effects of regulating MLCK expression by inhibitors such as ML-9, ML-7, microbial products, naturally occurring products, and microRNAs are also discussed. The influence of MLCK in inflammatory diseases starts with endothelial barrier dysfunction. The effectiveness of anti-MLCK treatment may depend on alleviation of that primary pathological mechanism. This review summarizes evidence for the potential benefits of anti-MLCK agents in the treatment of inflammatory disease and the importance of avoiding treatment-related side effects, as MLCK is widely expressed in many different tissues.

SELECTION OF CITATIONS
SEARCH DETAIL
...