Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Phytomedicine ; 100: 154074, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35397283

ABSTRACT

RATIONALE: Alveolar epithelial cell death, inflammation, and oxidative stress are typical features of acute lung injury (ALI). Aloperine (Alo), an alkaloid isolated from Sophora alopecuroides, has been reported to display various biological effects, such as anti-inflammatory, immunoregulatory, and anti-oxidant properties. In this study, we investigated the effects and mechanisms of Alo in treating a lipopolysaccharide (LPS)-induced ALI in a murine model. METHODS: The effects of Alo in LPS-induced ALI were investigated in C57BL/6 mice. The RIPK1 inhibitor (Nec-1) and the RIPK3 inhibitor (GSK'872) were used to evaluate the relationship of necroptosis, NF-κB activation, and PDC subunits in LPS-treated mouse alveolar epithelial cells (MLE-12). Then the effects of Alo on necroptosis, inflammation, and oxidative stress of LPS-stimulated MLE-12 cells were evaluated. RESULTS: Alo significantly attenuated histopathological lung injuries and reduced lung wet/dry ratio in LPS-induced ALI mice. Alo also remarkedly reduced total protein and neutrophils recruitment in bronchoalveolar lavage fluid of ALI mice. Meanwhile, Alo ameliorated the LPS-induced necroptosis in the lungs of ALI mice. The RIPK3 inhibitor GSK'872, but not the RIPK1 inhibitor Nec-1, reversed LPS-induced p65 phosphorylation and translocation to the nucleus in MLE-12 cells. GSK'872 also reversed the LPS-induced increase in ROS and binding of RIPK3 and PDC subunits in MLE-12 cells. Moreover, Alo down-regulated the levels of p-RIPK1, p-RIPK3, p-MLKL, p-p65, the translocation of p65 to the nucleus, and reduced the expression of IL-6 and IL-8 in LPS-stimulated MLE-12 cells. Alo also inhibited the binding of RIPK3 and PDC-E1α, PDC-E1ß, PDC-E2, and PDC-E3 and the ROS production in LPS-treated MLE-12 cells. CONCLUSION: The present study validated the beneficial effects of Alo on LPS-induced ALI , suggesting Alo may be a new drug candidate against ALI.


Subject(s)
Acute Lung Injury , Lipopolysaccharides , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/metabolism , Animals , Disease Models, Animal , Inflammation/drug therapy , Inflammation/pathology , Lipopolysaccharides/pharmacology , Lung/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Necroptosis , Oxidative Stress , Piperidines/pharmacology , Quinolizidines , Reactive Oxygen Species
2.
Eur J Pharmacol ; 916: 174721, 2022 Feb 05.
Article in English | MEDLINE | ID: mdl-34954231

ABSTRACT

Type 1 diabetes (T1D) is a metabolic dysfunction characterized by the selective destruction of islet ß-cells, with oxidative stress playing an essential role in the manifestation of this disease state. Aloperine (ALO) represents the main active alkaloid extracted from the traditional Chinese herbal Sophora alopecuroides L. and features outstanding antioxidative properties. In this study, T1D was induced by a single high dose streptozotocin (STZ, 150 mg/kg, intraperitoneal) in mice. Diabetic animals were intragastrically administered ALO at a dose of 50 mg/kg/day. Notably, treatment of ALO (50 mg/kg/day) for seven consecutive days could observably reverse the onset of diabetes induced by STZ accompanied by weight gain, lower blood glucose levels, and relief of ß-cells damage. Our in vitro study further demonstrated that ALO protected ß-cells from STZ/hydrogen peroxide-induced oxidative damage as manifested by increased expression of MnSOD and CAT. Furthermore, a network pharmacology study revealed that NOS1 represented the main target of ALO. Mechanistic studies subsequently showed that treatment of ALO increased the expression of NOS1, whereas NOS2 was decreased. Moreover, a docking study carried out suggested that ALO could fit into the binding pocket of human NOS1 and molecular dynamics simulation further validated this docking event. Collectively, the administration of ALO prior to diabetes could be a viable approach to the prevention of ß-cell injury. This study may offer a novel potential herbal medicine against T1D and may further help improve the understanding of the underlying molecular mechanisms of ALO-mediated protection against oxidative stress.


Subject(s)
Diabetes Mellitus, Experimental , Diabetes Mellitus, Type 1 , Insulin-Secreting Cells , Nitric Oxide Synthase Type I , Quinolizidines , Animals , Blood Glucose/metabolism , Cytoprotection , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 1/drug therapy , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/pathology , Mice , Nitric Oxide Synthase Type I/metabolism , Oxidative Stress , Piperidines/pharmacology , Quinolizidines/administration & dosage , Quinolizidines/pharmacology , Quinolizidines/therapeutic use , Streptozocin
3.
Chin J Nat Med ; 19(11): 815-824, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34844720

ABSTRACT

Cervical cancer (CC) is recognized as the most common neoplasm in the female reproductive system worldwide. The lack of chemotherapeutic agents with outstanding effectiveness and safety severely compromises the anti-cipated prognosis of patients. Aloperine (ALO) is a natural quinolizidine alkaloid with marked anti-cancer effects on multiple malignancies as well as favorable activity in relieving inflammation, allergies and infection. However, its therapeutic efficacy and underlying mechanism in CC are still unclear. In the current study, MTT assay was employed to evaluate the viability of HeLa cells exposed to ALO to preliminarily estimate the effectiveness of ALO in CC. Then, the effects of ALO on the proliferation and apoptosis of HeLa cells were further investigated by plate colony formation and flow cytometry, respectively, while the migration and invasion of ALO-treated HeLa cells were evaluated using Transwell assay. Moreover, nude mice were subcutaneously inoculated with HeLa cells to demonstrate the anti-CC properties of ALO in vivo. The molecular mechanisms underlying these effects of ALO were evaluated by Western blot and immunohistochemical analysis. This study experimentally demonstrated that ALO inhibited the proliferation of HeLa cells via G2 phase cell cycle arrest. Simultaneously, ALO promoted an increase in the percentage of apoptotic HeLa cells by increasing the Bax/Bcl-2 ratio. Additionally, the migration and invasion of HeLa cells were attenuated by ALO treatment, which was considered to result from inhibition of epithelial-to-mesenchymal transition. For molecular mechanisms, the expression and activation of the IL-6-JAK1-STAT3 feedback loop were markedly suppressed by ALO treatment. This study indicated that ALO markedly suppresses the proliferation, migration and invasion and enhances the apoptosis of HeLa cells. In addition, these prominent anti-CC properties of ALO are associated with repression of the IL-6-JAK1-STAT3 feedback loop.


Subject(s)
Quinolizidines/pharmacology , Uterine Cervical Neoplasms , Animals , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Feedback , Female , HeLa Cells , Humans , Interleukin-6/genetics , Janus Kinase 1/genetics , Mice , Mice, Nude , STAT3 Transcription Factor/genetics , Signal Transduction , Uterine Cervical Neoplasms/drug therapy
4.
Int Immunopharmacol ; 97: 107720, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33945918

ABSTRACT

Presently, postmenopausal osteoporosis mainly caused by excessive activation of in vivo osteoclasts has become a global public health burden. Natural compounds have gradually become the potential drugs for the treatment of postmenopausal osteoporosis. Aloperine is a new alkaloid extracted from the leaves and seeds of sophora bean. The current studies have proved that aloperine has many biological activities, including anti-inflammatory, antiviral and anticancer activities. This study shows that aloperine can inhibit activity and formation of osteoclast mediated by RANKL in a dose-dependent manner without affecting the activity of bone marrow macrophages (BMM). In addition, it is found that aloperine can inhibit the expression of osteoclast specific marker genes, including nuclear factor of activated T cells cytoplasmic 1 (NFATc1), tartrate resistant acid phosphatase (TRAcP), matrix metallopeptidase 9 (MMP9), cathepsin K (Ctsk), V-ATPase d2 and calcitonin receptor. The in vitro experiment of aloperine proved that aloperine can inhibit the degradation of IκBα and the phosphorylation of P65, ERK and JNK. Additionally, aloperine improves bone loss in ovariectomized (OVX) mice by inhibiting osteoclast activity. This project proved that aloperine can affect the formation of osteoclasts by inhibiting RANKL signaling channel, and it is indicated that aloperine has the potential to be developed as a new drug for the prevention and treatment of postmenopausal osteoporosis.


Subject(s)
Osteogenesis/drug effects , Osteoporosis, Postmenopausal/drug therapy , Quinolizidines/pharmacology , Animals , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Femur/drug effects , Femur/pathology , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , MAP Kinase Signaling System/drug effects , Mice , NF-kappa B/antagonists & inhibitors , NF-kappa B/metabolism , Osteoclasts/drug effects , Osteoporosis, Postmenopausal/diagnosis , Osteoporosis, Postmenopausal/etiology , Osteoporosis, Postmenopausal/pathology , Ovariectomy , Quinolizidines/therapeutic use , RANK Ligand/antagonists & inhibitors , RANK Ligand/metabolism , X-Ray Microtomography
5.
Protein Pept Lett ; 27(9): 860-869, 2020.
Article in English | MEDLINE | ID: mdl-32196436

ABSTRACT

BACKGROUND: Ovarian cancer is the most lethal gynecologic malignancy worldwide with poor prognosis owing to chemotherapy resistance and cancer relapse. Hence, there is an urgent need to develop novel anticancer agents against ovarian cancer. OBJECTIVE: The aim of this research is to investigate the possible anticancer activity of aloperine, an active ingredient from a traditional Chinese medicine Sophora alopecuroides, and to explore the possible Reactive Oxygen Species (ROS)-related mechanism. METHODS: Cell viability, cytotoxicity, apoptosis, ROS generation, and oxidant stress indicators were analyzed. RESULTS: Our results demonstrated that aloperine significantly induced inhibition of cell viability, promoted cytotoxicity and mitochondrial-related apoptosis, and increased ROS generation in ovarian cancer cells. Furthermore, the antioxidant α-lipoic acid reversed apoptosis in aloperinetreated cells. In addition, we identified hydrogen peroxide as the main type of ROS, and the antioxidant catalase suppressed the apoptotic inducing effect of aloperine whereas hydrogen peroxide supplement exacerbated the effect of aloperine in ovarian cancer cells. CONCLUSION: Taken together, our results indicated that aloperine could exert anti-ovarian cancer cell activity through a reactive oxygen species activation mechanism and suggested aloperine as a potential agent against ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Ovarian Neoplasms/metabolism , Piperidines/pharmacology , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Quinolizidines
6.
Front Pharmacol ; 11: 538137, 2020.
Article in English | MEDLINE | ID: mdl-33536900

ABSTRACT

Aloperine, a quinolizidine-type alkaloid, was first isolated from the seeds and leaves of herbal plant, Sophora alopecuroides L. Empirically, Sophora alopecuroides L. is appreciated for its anti-dysentry effect, a property that is commonly observed in other Sophora Genus phytomedicines. Following the rationale of reductionism, subsequent biochemical analyses attribute such anti-dysentry effect to the bactericidal activity of aloperine. From then on, the multiple roles of aloperine are gradually revealed. Accumulating evidence suggests that aloperine possesses multiple pharmacological activities and holds a promising potential in clinical conditions including skin hyper-sensitivity, tumor and inflammatory disorders etc.; however, the current knowledge on aloperine is interspersed and needs to be summarized. To facilitate further investigation, herein, we conclude the key pharmacological functions of aloperine, and most importantly, the underlying cellular and molecular mechanisms are clarified in detail to explain the functional mode of aloperine.

7.
J Ethnopharmacol ; 248: 112172, 2020 Feb 10.
Article in English | MEDLINE | ID: mdl-31442619

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Sophora alopecuroides L., which is called Kudouzi in China, is a medicinal plant distributed in Western and Central Asia, especially in China, and has been used for decades to treat fever, bacterial infection, heart disease, rheumatism, and gastrointestinal diseases. AIM OF THE REVIEW: This review aims to provide up-to-date information on S. alopecuroides, including its botanical characterization, medicinal resources, traditional uses, phytochemistry, pharmacological research, and toxicology, in exploring future therapeutic and scientific potentials. MATERIALS AND METHODS: The information related to this article was systematically collected from the scientific literature databases including PubMed, Google Scholar, Web of Science, Science Direct, Springer, China National Knowledge Infrastructure, published books, PhD and MS dissertations, and other web sources, such as the official website of Flora of China and Yao Zhi website (https://db.yaozh.com/). RESULTS: A total of 128 compounds, such as alkaloids, flavonoids, steroids, and polysaccharides, were isolated from S. alopecuroides. Among these compounds, the effects of alkaloids, such as matrine and oxymatrine, were extensively studied and developed into new drugs. S. alopecuroides and its active components had a wide range of pharmacological activities, such as anticancer, antiviral, anti-inflammatory, antimicrobial, analgesic, and neuroprotective functions, as well as protective properties against pulmonary fibrosis and cardiac fibroblast proliferation. CONCLUSIONS: As an important traditional Chinese medicine, modern pharmacological studies have demonstrated that S. alopecuroides has prominent bioactivities, especially on gynecological inflammation and hepatitis B, and anticancer activities. These activities provide prospects for novel drug development for cancer and some chronic diseases. Nevertheless, the comprehensive evaluation, quality control, understanding of the multitarget network pharmacology, long-term in vivo toxicity, and clinical efficacy of S. alopecuroides require further detailed research.


Subject(s)
Sophora , Agriculture , Animals , Ethnobotany , Ethnopharmacology , Humans , Phytochemicals/analysis , Phytochemicals/pharmacology , Phytochemicals/therapeutic use , Phytochemicals/toxicity , Plant Preparations/chemistry , Plant Preparations/pharmacology , Plant Preparations/therapeutic use , Plant Preparations/toxicity , Quality Control
8.
PeerJ ; 7: e7652, 2019.
Article in English | MEDLINE | ID: mdl-31534865

ABSTRACT

OBJECTIVE: Aloperine (ALO), an alkaloid isolated from the leaves of Sophora alopecuroides, has been suggested to exhibit anti-inflammatory and anti-tumor properties and is traditionally used to treat various human diseases, including cancer. However, limited information is available about the mechanisms that determine the anti-tumor activities of ALO. METHODS: Herein, through comprehensive bioinformatics methods and in vitro functional analyses, we evaluated the detailed anti-tumor mechanisms of ALO. RESULTS: Using the databases Bioinformatics analysis tool for molecular mechanism of traditional Chinese medicine and PubChem Project, we identified the potential targets of ALO. A protein-protein interaction network was constructed to determine the relationship among these probable targets. Functional enrichment analysis revealed that ALO is potentially involved in the induction of apoptosis. In addition, molecular docking demonstrated that ALO expectedly docks into the active pocket of the Bcl2 protein, suggesting Bcl2 as a direct target of ALO. Moreover, western blot and qPCR analysis showed that ALO downregulated Bcl2 expression in human glioma cell lines, SK-N-AS and U118. Using flow cytometry methods, we further confirmed that ALO significantly promotes apoptosis in SK-N-AS and U118 cell lines, similar to the effect induced by ABT-737, a well-known Bcl2 inhibitor. In addition, Bcl-2 overexpression could rescue ALO-induced Bcl-2 inhibition and suppress pro-apoptotic effects in glioma cells. CONCLUSION: Taken together, these findings suggest that the natural agent ALO effectively enhances apoptosis by acting as a potential Bcl2 inhibitor in human glioma cells.

9.
Chin J Physiol ; 62(4): 157-165, 2019.
Article in English | MEDLINE | ID: mdl-31535631

ABSTRACT

Abnormal pulmonary arterial vascular smooth muscle cells (PASMCs) proliferation is critical pathological feature of pulmonary vascular remodeling that acts as driving force in the initiation and development of pulmonary arterial hypertension (PAH), ultimately leading to pulmonary hypertension. Aloperine is a main active alkaloid extracted from the traditional Chinese herbal Sophora alopecuroides and possesses outstanding antioxidation and anti-inflammatory effects. Our group found Aloperine has protective effects on monocroline-induced pulmonary hypertension in rats by inhibiting oxidative stress in previous researches. However, the anti-inflammation effects of Aloperine on PAH remain unclear. Therefore, to further explore whether the beneficial role of Aloperine on PAH was connected with its anti-inflammatory effects, we performed experiments in vitro. Aloperine significantly inhibited the proliferation and DNA synthesis of human pulmonary artery smooth muscle cells (HPASMCs) induced by platelet-derived growth factor-BB, blocked progression through G0/G1to S phase of the cell cycle and promoted total ratio of apoptosis. In summary, these results suggested that Aloperine negatively regulated nuclear factor-κB signaling pathway activity to exert protective effects on PAH and suppressed HPASMCs proliferation therefore has a potential value in the treatment of pulmonary hypertension by negatively modulating pulmonary vascular remodeling.


Subject(s)
Hypertension, Pulmonary , Muscle, Smooth, Vascular , Animals , Cell Proliferation , Humans , Myocytes, Smooth Muscle , Piperidines , Pulmonary Artery , Quinolizidines , Rats
10.
Phytomedicine ; 61: 152843, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31039533

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) ranks third among the most common causes of cancer-related deaths worldwide. The chemotherapy for HCC is still insufficient, so far. In searching for effective anti-HCC agents from traditional Chinese medicine, we discovered that aloperine (ALO), a quinolizidine alkaloid from Sophora alopecuroides L., exerts anti-HCC activities. However, the effects of ALO on HCC have been rarely studied, and its underlying mechanisms remain unknown. PURPOSE: This study aims to evaluate the anti-HCC activities of ALO and explore its underlying mechanisms. METHODS: MTT assay and colony formation assay were used to investigate the anti-proliferative effects of ALO on human HCC Hep3B and Huh7 cells. Hoechst 33258 staining was used to observe the morphological changes of cells after ALO treatment. Flow cytometry was used to analyze apoptosis induction, the collapse of the mitochondrial membrane potential and cell cycle distribution. Western blotting was used to examine the expression levels of proteins associated with apoptosis and cell cycle arrest, and key proteins in the PI3K/Akt signaling pathway. Small interfering RNA (siRNA) transfection was used to investigate the role of Akt in ALO-induced apoptosis and cell cycle arrest. Zebrafish tumor model was used to evaluate the anti-HCC effects of ALO in vivo. RESULTS: ALO inhibited the proliferation of Hep3B and Huh7 cells. ALO induced apoptosis in HCC cells, which was accompanied by the loss of mitochondrial potential, the release of cytochrome c into cytosol, as well as the increased cleavages of caspase-9, caspase-3 and PARP. Moreover, ALO induced G2/M cell cycle arrest by downregulating the expression levels of cdc25C, cdc2 and cyclin B1. In addition, ALO inhibited activation of the PI3K/Akt signaling pathway by decreasing the expression levels of p110α, p85, Akt and p-Akt (Ser473). Further study showed that inhibition of Akt by siRNA augmented ALO-mediated apoptosis and G2/M cell cycle arrest in HCC cells. Critically, ALO inhibited the growth of Huh7 cells in vivo. CONCLUSION: We first demonstrated that ALO induced apoptosis and G2/M cell cycle arrest in HCC cells through inhibition of the PI3K/Akt signaling pathway. This study provides a rationale for ALO as a potential chemotherapeutic agent for HCC.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Piperidines/pharmacology , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Embryo, Nonmammalian , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Membrane Potential, Mitochondrial/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinolizidines , Xenograft Model Antitumor Assays , Zebrafish/embryology
11.
Biomed Pharmacother ; 95: 1161-1168, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28926926

ABSTRACT

Pulmonary hypertension (PH) is fatal disease which closely involves Rho A/ Rho kinsase (ROCK) pathway. Aloperine is a main active alkaloid extracted from Sophora alopecuroides, which is a traditional Chinese herbal medicine that has been used widely. However, the effects of this alkaloid on pulmonary hypertension and its mechanisms remain unclear. Therefore, this study is designed to investigate whether aloperine has protective effects on PH induced by monocrotaline, whether these effects may be related to regulation of RhoA/ROCK pathway in rats. Pulmonary hypertension was induced by monocrotaline (60mg/kg), and subsequently oral administration of aloperine (25, 50, 100mg/kg/day) for 21 days. At the end of the experiment, rats were underwent hemodynamic and morphologic assessments. At same time, the expression of Rho A, ROCK1, ROCK2, as well as activities of ROCK in the lung of rat has been detected. Afterwards, the expression of p27kip1, Bax, Bcl-2, which was the downstream proliferation and apoptosis factors of ROCK, were tested. The result indicted that aloperine treatment showed significantly improvement in hemodynamic and pathomorphologic data. Moreover, the reduction in expression of Rho A, ROCK1, ROCK2, and suppression in activities of ROCK were found in rat lungs after aloperine treatment. Furthermore, aloperine also alleviated the MCT-induced changes of p27kip1, Bax and Bcl-2. In summary, this study indicates that aloperine have protective effects on monocrotaline-induced PH. And these effects may be partially related to RhoA/ROCK pathway. Thus, aloperine could be considered a possible therapeutic strategy for PH.


Subject(s)
Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Piperidines/therapeutic use , Protective Agents/therapeutic use , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Cardiomegaly/complications , Cardiomegaly/drug therapy , Cardiomegaly/physiopathology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Electrocardiography , Hemodynamics/drug effects , Hypertension, Pulmonary/diagnostic imaging , Hypertension, Pulmonary/physiopathology , Lung/drug effects , Lung/pathology , Lung/physiopathology , Male , Monocrotaline , Piperidines/pharmacology , Proliferating Cell Nuclear Antigen/metabolism , Protective Agents/pharmacology , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Quinolizidines , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley , Vascular Remodeling/drug effects , bcl-2-Associated X Protein/metabolism , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL