1
|
Yuan Y, Zhang Q, Qiu F, Kang N, Zhang Q. Targeting TRPs in autophagy regulation and human diseases. Eur J Pharmacol 2024; 977:176681. [PMID: 38821165 DOI: 10.1016/j.ejphar.2024.176681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Transient receptor potential channels (TRPs) are widely recognized as a group of ion channels involved in various sensory perceptions, such as temperature, taste, pressure, and vision. While macroautophagy (hereafter referred to as autophagy) is primarily regulated by core machinery, the ion exchange mediated by TRPs between intracellular and extracellular compartments, as well as within organelles and the cytoplasm, plays a crucial role in autophagy regulation as an important signaling transduction mechanism. Moreover, certain TRPs can directly interact with autophagy regulatory proteins to participate in autophagy regulation. In this article, we provide an in-depth review of the current understanding of the regulatory mechanisms of autophagy, with a specific focus on TRPs. Furthermore, we highlight the potential prospects for drug development targeting TRPs in autophagy for the treatment of human diseases.
Collapse
Affiliation(s)
- Yongkang Yuan
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Qiuju Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China
| | - Feng Qiu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China; Tianjin Key Laboratory of Therapeutic Substance of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China; School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| | - Ning Kang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| | - Qiang Zhang
- School of Medical Technology, Tianjin University of Traditional Chinese Medicine, Tianjin, People's Republic of China.
| |
Collapse
|
2
|
Bai J, Feng Z, Chen Y, Li Y, Zhang L, Li L. Lycorine attenuated proliferation and induced apoptosis on imatinib-resistant K562 cell by inhibiting autophagy. Discov Oncol 2024; 15:217. [PMID: 38856766 PMCID: PMC11164850 DOI: 10.1007/s12672-024-01080-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Tyrosine kinase inhibitor (TKI) resistance is a significant factor exacerbating the burden on chronic myeloid leukemia (CML) patients and impacting clinical efficacy. The main goal is to offer new insights into overcoming drug resistance in treating CML. METHODS Imatinib (IM) resistant K562/IM cells were generated using gradient induction. Responses to IM, lycorine, and autophagy modulators were assessed using CCK-8. Protein expression of Beclin-1, Atg5, LC3, Caspase-3, P62, Bax, Bcl-2, and P-gp was detected using Western blot. Lycorine-induced apoptosis and cell cycle changes were evaluated through flow cytometry, while autophagy alterations were detected using monodansylcadaverine (MDC) staining. In the K562/IM mice model, non-obese diabetic severe combined immunodeficent (NOD-SCID) mice were subcutaneously inoculated with K562/IM cells. After 17 days of lycorine injection, assessments included tumor size, hematoxylin-eosin (HE) staining, and Ki67 expression. RESULTS After 72 h of IM treatment, K562/IM cells showed a 55.86-fold increase in drug resistance compared to K562 cells. Lycorine treatment for 24 h inhibited cell proliferation and induced G0/G1 phase cell cycle arrest and apoptosis in both K562 and K562/IM cells. MDC staining indicated reduced autophagy in K562/IM cells, mitigated by lycorine. In vivo experiments demonstrated reduced tumor size and Ki67 proliferation index in the lycorine treatment group (K562+L, K562/IM+L) compared to the control group, particularly in the drug-resistant group. However, no significant change in Ki67 was observed in the K562 group after lycorine treatment. CONCLUSION In summary, K562/IM cells displayed heightened autophagy levels compared to K562 cells. Lycorine effectively impeded the proliferation of K562/IM cells through diverse mechanisms, including reduced autophagy, enhanced apoptosis, and induced cell cycle arrest.
Collapse
Affiliation(s)
- Jun Bai
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Gansu Province Hematologic Disease Clinical Medical Research Center, Lanzhou, 730000, China
| | - Zuxi Feng
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, 730000, China
| | - Yaqiong Chen
- School of Basic Medical Sciences, Lanzhou University, Lanzhou, 730000, China
| | - Yanhong Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Gansu Province Hematologic Disease Clinical Medical Research Center, Lanzhou, 730000, China
| | - Liansheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Gansu Province Hematologic Disease Clinical Medical Research Center, Lanzhou, 730000, China.
| | - Lijuan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Gansu Province Hematologic Disease Clinical Medical Research Center, Lanzhou, 730000, China.
| |
Collapse
|
3
|
Xie X, Chen W, Xu M, Chen J, Yang T, Wang C, Su Y, Zhao J, Xu J, Liu Q. IKK/NF-κB and ROS signal axes are involved in Tenacissoside H mediated inhibitory effects on LPS-induced inflammatory osteolysis. Cell Prolif 2024; 57:e13535. [PMID: 37551727 PMCID: PMC10771108 DOI: 10.1111/cpr.13535] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 06/07/2023] [Accepted: 07/25/2023] [Indexed: 08/09/2023] Open
Abstract
Periodontal disease and arthroplasty prosthesis loosening and destabilization are both associated with osteolysis, which is predominantly caused by abnormal bone resorption triggered by pro-inflammatory cytokines. Osteoclasts (OCs) are critical players in the process. Concerns regarding the long-term efficacy and side effects of current frontline therapies, however, remain. Alternative therapies are still required. The aim of this work was to investigate the involvement of Tenacissoside H (TDH) in RANKL-mediated OC differentiation, as well as inflammatory osteolysis and associated processes. In vitro, bone marrow-derived macrophages (BMMs) cultured with RANKL and M-CSF were used to detect TDH in the differentiation and function of OCs. Real-time quantitative PCR was used to measure the expression of specific genes and inflammatory factors in OCs. Western blot was used to identify NFATc1, IKK, NF-κB, MAPK pathway, and oxidative stress-related components. Finally, an LPS-mediated calvarial osteolysis mouse model was employed to explore TDH's role in inflammatory osteolysis. The results showed that in vivo TDH inhibited the differentiation and resorption functions of OCs and down-regulated the transcription of osteoclast-specific genes, as well as Il-1β, Il-6 and Tnf-α. In addition, TDH inhibited the IKK and NF-κB signalling pathways and down-regulated the level of ROS. In vivo studies revealed that TDH improves the bone loss caused by LPS. TDH may be a new candidate or treatment for osteoclast-associated inflammatory osteolytic disease.
Collapse
Affiliation(s)
- Xiaoxiao Xie
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical UniversityNanningGuangxiChina
| | - Weiwei Chen
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical UniversityNanningGuangxiChina
| | - Minglian Xu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Junchun Chen
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Tao Yang
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical UniversityNanningGuangxiChina
| | - Chaofeng Wang
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical UniversityNanningGuangxiChina
| | - Yuangang Su
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jinmin Zhao
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
| | - Jiake Xu
- School of Biomedical SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Qian Liu
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic DepartmentThe First Affiliated Hospital of Guangxi Medical UniversityNanningGuangxiChina
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Co‐constructed by the Province and Ministry, Life Sciences Institute, Guangxi Medical UniversityNanningGuangxiChina
| |
Collapse
|
4
|
Sharma K, Kumar S, Prakash R, Khanka S, Mishra T, Rathur R, Biswas A, Verma SK, Bhatta RS, Narender T, Singh D. Chebulinic acid alleviates LPS-induced inflammatory bone loss by targeting the crosstalk between reactive oxygen species/NFκB signaling in osteoblast cells. Free Radic Biol Med 2023; 194:99-113. [PMID: 36423891 DOI: 10.1016/j.freeradbiomed.2022.11.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/22/2022]
Abstract
Chebulinic acid (CA), a plant ellagitannin derived from Triphala, is reported to exhibit both anti-inflammatory & anti-oxidant activity apart from anti-tumour property. However, its role in inflammatory bone loss conditions was unexplored. We hypothesized that CA may prevent the bone loss under inflammatory conditions induced by lipopolysaccharide (LPS) in 10-week-old male C57BL/6J mice. Micro-CT analysis and histomorphometric evaluations were carried out where it was found that CA significantly improved the bone micro-architectures by enhancing trabecular connectivity and strength of the bone. CA also increased the bone regeneration as examined by calcein labelling and ex-vivo mineralisation along with maintaining the bone serum markers. Further, CA ameliorated the reduction in osteoblast cell differentiation, proliferation and viability after LPS stimulation. DCFDA and Mitosox staining revealed that CA presented remarkable protective effects against LPS treatment by attenuating oxidative stress, both at cellular & mitochondrial levels. In addition, CA significantly decreased the production of pro-inflammatory cytokines, and down-regulated the phosphorylation of NFκB and IκBα, indicating that CA could attenuate the inflammatory impairment to primary osteoblast cells by suppressing the NFkB signalling pathway. Taken together, the protective role of CA against LPS-induced bone loss & inhibitory effect on total ROS levels hold promise as a potential novel therapeutic strategy for the inflammatory diseases in bones.
Collapse
Affiliation(s)
- Kriti Sharma
- Division of Endocrinology, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Shiv Kumar
- Division of Medicinal & Process Chemistry, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | | | - Sonu Khanka
- Division of Endocrinology, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | | | - Rajat Rathur
- Division of Endocrinology, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - Arpon Biswas
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - Sarvesh Kumar Verma
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India
| | - R S Bhatta
- Pharmaceutics and Pharmacokinetics Division, CSIR-Central Drug Research Institute, Lucknow, 226031, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| | - T Narender
- Division of Medicinal & Process Chemistry, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India.
| | - Divya Singh
- Division of Endocrinology, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India.
| |
Collapse
|
5
|
Bott KN, Feldman E, de Souza RJ, Comelli EM, Klentrou P, Peters SJ, Ward WE. Lipopolysaccharide-Induced Bone Loss in Rodent Models: A Systematic Review and Meta-Analysis. J Bone Miner Res 2023; 38:198-213. [PMID: 36401814 PMCID: PMC10107812 DOI: 10.1002/jbmr.4740] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/27/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
Abstract
Osteoporosis has traditionally been characterized by underlying endocrine mechanisms, though evidence indicates a role of inflammation in its pathophysiology. Lipopolysaccharide (LPS), a component of gram-negative bacteria that reside in the intestines, can be released into circulation and stimulate the immune system, upregulating bone resorption. Exogenous LPS is used in rodent models to study the effect of systemic inflammation on bone, and to date a variety of different doses, routes, and durations of LPS administration have been used. The study objective was to determine whether systemic administration of LPS induced inflammatory bone loss in rodent models. A systematic search of Medline and four other databases resulted in a total of 110 studies that met the inclusion criteria. Pooled standardized mean differences (SMDs) and corresponding 95% confidence intervals (CI) with a random-effects meta-analyses were used for bone volume fraction (BV/TV) and volumetric bone mineral density (vBMD). Heterogeneity was quantified using the I2 statistic. Shorter-term (<2 weeks) and longer-term (>2 weeks) LPS interventions were analyzed separately because of intractable study design differences. BV/TV was significantly reduced in both shorter-term (SMD = -3.79%, 95% CI [-4.20, -3.38], I2 62%; p < 0.01) and longer-term (SMD = -1.50%, 95% CI [-2.00, -1.00], I2 78%; p < 0.01) studies. vBMD was also reduced in both shorter-term (SMD = -3.11%, 95% CI [-3.78, -2.44]; I2 72%; p < 0.01) and longer-term (SMD = -3.49%, 95% CI [-4.94, -2.04], I2 82%; p < 0.01) studies. In both groups, regardless of duration, LPS negatively impacted trabecular bone structure but not cortical bone structure, and an upregulation in bone resorption demonstrated by bone cell staining and serum biomarkers was reported. This suggests systemically delivered exogenous LPS in rodents is a viable model for studying inflammatory bone loss, particularly in trabecular bone. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Kirsten N Bott
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Evelyn Feldman
- Lakehead University Library, Lakehead University, Thunder Bay, ON, Canada
| | - Russell J de Souza
- Department of Health Research Methods, Evidence, and Impact, Faculty of Health Sciences, McMaster University, Hamilton, ON, Canada.,Population Health Research Institute, Hamilton Health Sciences Corporation, Hamilton, ON, Canada
| | - Elena M Comelli
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.,Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada.,Joannah and Brian Lawson Centre for Child Nutrition, University of Toronto, Toronto, ON, Canada
| | - Panagiota Klentrou
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Sandra J Peters
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada
| | - Wendy E Ward
- Department of Kinesiology, Brock University, St. Catharines, ON, Canada.,Centre for Bone and Muscle Health, Brock University, St. Catharines, ON, Canada.,Department of Nutritional Sciences, University of Toronto, Toronto, ON, Canada.,Department of Health Sciences, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
6
|
Amaryllidaceae Alkaloids Decrease the Proliferation, Invasion, and Secretion of Clinically Relevant Cytokines by Cultured Human Colon Cancer Cells. Biomolecules 2022; 12:biom12091267. [PMID: 36139106 PMCID: PMC9496155 DOI: 10.3390/biom12091267] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/29/2022] [Accepted: 09/05/2022] [Indexed: 12/30/2022] Open
Abstract
Alkaloids isolated from members of the Amaryllidaceae plant family are promising anticancer agents. The purpose of the current study was to determine if the isocarbostyrils narciclasine, pancratistatin, lycorane, lycorine, crinane, and haemanthamine inhibit phenomena related to cancer progression in vitro. To achieve this, we examined the proliferation, adhesion, and invasion of cultured human colon cancer cells via MTT assay and Matrigel-coated Boyden chambers. In addition, Luminex assays were used to quantify the secretion of matrix metalloproteinases (MMP) and cytokines associated with poor clinical outcomes. We found that all alkaloids decreased cell proliferation regardless of TP53 status, with narciclasine exhibiting the greatest potency. The effects on cell proliferation also appear to be specific to cancer cells. Narciclasine, lycorine, and haemanthamine decrease both adhesion and invasion but with various potencies depending on the cell line. In addition, narciclasine, lycorine, and haemanthamine decreased the secretion of MMP-1, -2, and -7, as well as the secretion of the cytokines pentraxin 3 and vascular endothelial growth factor. In conclusion, the present study shows that Amaryllidaceae alkaloids decrease phenomena and cytokines associated with colorectal cancer progression, supporting future investigations regarding their potential as multifaceted drug candidates.
Collapse
|
7
|
Xiao H, Xu X, Du L, Li X, Zhao H, Wang Z, Zhao L, Yang Z, Zhang S, Yang Y, Wang C. Lycorine and organ protection: Review of its potential effects and molecular mechanisms. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2022; 104:154266. [PMID: 35752077 DOI: 10.1016/j.phymed.2022.154266] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/05/2022] [Accepted: 06/09/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Multiorgan dysfunction, especially sepsis-related multiorgan damage, remains a major cause of high mortality in the late stages of infection and a great clinical challenge. In recent years, natural drugs have received widespread attention because of their low cost, wide sources, high efficacy, low toxicity, and limited side effects. Lycorine, a natural compound extracted from Amaryllidaceae, exhibits multiple pharmacological activities, including in the regulation of autophagy and the induction of cancer cell apoptosis, and has anti-inflammatory, antifungal, antiviral, antimalarial, and antitumor activities. However, studies on lycorine have mainly focused on its antitumor properties, and research on its use for organ protection, especially in sepsis-related organ injury, is relatively limited. PURPOSE To review and discuss the effects and mechanisms of lycorine in the treatment of multi-organ dysfunction, especially sepsis. METHODS Literature searches in electronic databases, such as Web of Science, Science Direct, PubMed, Google Scholar, and Scopus, were performed using 'Lycorine', 'Amaryllidaceae', 'Pharmacology', 'Pharmacokinetics', 'Anti-inflammation', 'Autophagy', 'Apoptosis', 'Anti-microbial and anti-parasitic', 'Antitumor', 'Organ protection', and 'Sepsis' as keywords, the correlated literature was extracted and conducted from the databases mentioned above. RESULTS By summarizing the progress made in existing research, we found that the general effects of lycorine involve the regulation of autophagy and the induction of cancer cell apoptosis, and anti-inflammatory, antifungal, antiviral, antimalarial, and antitumor effects; through these pathways, the compound can ameliorate organ damage. In addition, lycorine was found to have an important effect on organ damage in sepsis. CONCLUSION Lycorine is a promising natural organ protective agent. This review will provide a new theoretical basis for the treatment of organ protection, especially in sepsis.
Collapse
Affiliation(s)
- Haoxiang Xiao
- Department of Cardiology, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Xuezeng Xu
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Luyang Du
- Department of Cardiology, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Xiyang Li
- Department of Cardiology, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Huadong Zhao
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zheng Wang
- Department of Cardiothoracic Surgery, Central Theater Command General Hospital of Chinese People's Liberation Army, Wuhan, China
| | - Lin Zhao
- Department of Cardiovascular Surgery, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhi Yang
- Department of General Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Shaofei Zhang
- Department of Cardiology, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, Xi'an, China
| | - Yang Yang
- Department of Cardiology, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, Xi'an, China.
| | - Changyu Wang
- Department of Cardiology, Xi'an No.3 Hospital, Faculty of Life Sciences and Medicine, Northwest University, Xi'an, China; Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education. Faculty of Life Sciences, Northwest University, Xi'an, China.
| |
Collapse
|
8
|
Doxorubicin Induces Bone Loss by Increasing Autophagy through a Mitochondrial ROS/TRPML1/TFEB Axis in Osteoclasts. Antioxidants (Basel) 2022; 11:antiox11081476. [PMID: 36009195 PMCID: PMC9404930 DOI: 10.3390/antiox11081476] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 12/10/2022] Open
Abstract
Doxorubicin (DOX), a widely used chemotherapeutic agent, has been linked to an increased risk of bone damage in human patients and induces bone loss in mice. DOX induces autophagy, which contributes to bone homeostasis and excess autophagy in osteoclasts (OCs), resulting in bone loss. We hypothesized that DOX-induced bone loss is caused by the induction of autophagy in OCs. In vitro, DOX significantly increased the area of OCs and bone resorption activity, whereas it decreased OC number through apoptosis. DOX enhanced the level of LC3II and acidic vesicular organelles-containing cells in OCs, whereas an autophagy inhibitor, 3-methyladenine (3-MA), reversed these, indicating that enhanced autophagy was responsible for the effects of DOX. Increased mitochondrial reactive oxygen species (mROS) by DOX oxidized transient receptor potential mucolipin 1 (TRPML1) on the lysosomal membrane, which led to nuclear localization of transcription factor EB (TFEB), an autophagy-inducing transcription factor. In vivo, micro-computerized tomography analysis revealed that the injection of 3-MA reversed DOX-induced bone loss, and tartrate-resistant acid phosphatase staining showed that 3-MA reduced the area of OCs on the bone surface, which was enhanced upon DOX administration. Collectively, DOX-induced bone loss is at least partly attributable to autophagy upregulation in OCs via an mROS/TRPML1/TFEB axis.
Collapse
|
9
|
Park HJ, Park JN, Yoon SY, Yu R, Suh JH, Choi HS. Morin Disrupts Cytoskeleton Reorganization in Osteoclasts through an ROS/SHP1/c-Src Axis and Grants Protection from LPS-Induced Bone Loss. Antioxidants (Basel) 2022; 11:963. [PMID: 35624827 PMCID: PMC9137647 DOI: 10.3390/antiox11050963] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Morin is a naturally occurring flavonoid with anti-inflammatory and antioxidative properties. Therefore, we hypothesized that morin may prevent inflammatory bone loss by reducing oxidative stress. To investigate the effect of morin on inflammatory bone loss, mice were injected with lipopolysaccharide (LPS). Osteoclasts (OCs) were analyzed by tartrate-resistant acid phosphatase (TRAP) staining and actin ring formation. Micro-computerized tomography analysis indicated that morin prevented LPS-induced bone loss in mice. In vivo TRAP staining indicated that morin decreased the number and surface of the OCs that were increased in LPS-treated mice. Furthermore, in vitro experiments indicated that morin decreased the number and activity of OCs upon LPS stimulation. Morin decreased actin ring-containing OCs with decreased activation of c-Src (Y416)/vav guanine nucleotide exchange factor 3/Ras-related C3 botulinum toxin substrate 1 compared with LPS alone. Morin decreased cytosolic reactive oxygen species (ROS), thus preventing the oxidation of Src homology region 2 domain-containing phosphatase 1 (SHP-1), followed by the inactivation of c-Src via direct interaction with SHP1. Conversely, SHP1 knockdown abolished the inhibitory effect of morin on OCs. Therefore, our findings suggest that morin disrupted cytoskeletal reorganization via an ROS/SHP1/c-Src axis in OCs, thereby granting protection from LPS-induced bone loss, which demonstrates its therapeutic potential against inflammatory bone loss.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Biological Sciences (BK21 Program), University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (J.-N.P.); (S.-Y.Y.)
| | - Jung-Nam Park
- Department of Biological Sciences (BK21 Program), University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (J.-N.P.); (S.-Y.Y.)
| | - Sun-Young Yoon
- Department of Biological Sciences (BK21 Program), University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (J.-N.P.); (S.-Y.Y.)
| | - Rina Yu
- Department of Food and Nutrition, University of Ulsan, Ulsan 44610, Korea;
| | - Jae-Hee Suh
- Department of Pathology, Ulsan University Hospital, Ulsan 44030, Korea;
| | - Hye-Seon Choi
- Department of Biological Sciences (BK21 Program), University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (J.-N.P.); (S.-Y.Y.)
| |
Collapse
|
10
|
Jian T, Zhang Y, Zhang G, Ling J. Metabolomic comparison between natural Huaier and artificial cultured Huaier. Biomed Chromatogr 2022; 36:e5355. [PMID: 35156219 DOI: 10.1002/bmc.5355] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Revised: 01/22/2022] [Accepted: 01/24/2022] [Indexed: 11/06/2022]
Abstract
Vanderbylia robiniophila (Murrill) B.K. (Huaier) is a kind of higher fungal fruiting body parasitic on the trunk of Sophora japonica and Robinia pseudoacacia L.. As a traditional Chinese medicine with a history of more than 1600 years, Huaier has attracted wide attention for its excellent anticancer activity. A systematic study on the metabolome differences between natural Huaier and artificial cultured Huaier was conducted using liquid chromatography-mass spectrometry in this study. Principal component analysis and orthogonal projection on latent structure-discriminant analysis results showed that cultured Huaier evidently separated and individually separated from natural Huaier, indicating metabolome difference between natural Huaier and cultured Huaier. Hierarchical clustering analysis was further performed to cluster the differential metabolites and samples based on their metabolic similarity. The higher content of amino acids, alkaloids and terpenoids in natural Huaier makes it an excellent choice as a traditional Chinese medicine for anti-cancer or nutritional supplementation. The results of the Bel-7402 and A549 cells cytotoxicity test showed that the anticancer activity of natural Huaier was better than that of cultured Huaier. This may be due to the difference in chemical composition, which makes the anticancer activity of natural and cultured Huaier different.
Collapse
Affiliation(s)
- Tongtong Jian
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yu Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guoying Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jianya Ling
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| |
Collapse
|
11
|
Lu Q, Li R, Yang Y, Zhang Y, Zhao Q, Li J. Ingredients with anti-inflammatory effect from medicine food homology plants. Food Chem 2022; 368:130610. [PMID: 34419798 DOI: 10.1016/j.foodchem.2021.130610] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/18/2021] [Accepted: 07/13/2021] [Indexed: 02/09/2023]
Abstract
Inflammation occurs when the immune system responses to external harmful stimuli and infection. Chronic inflammation induces various diseases. A variety of foods are prescribed in the traditional medicines of many countries all over the world, which gave birth to the concept of medicine food homology. Over the past few decades, a number of secondary metabolites from medicine food homology plants have been demonstrated to have anti-inflammatory effects. In the present review, the effects and mechanisms of the medicine food homology plants-derived active components on relieving inflammation and inflammation-mediated diseases were summarized and discussed. The information provided in this review is valuable to future studies on anti-inflammatory ingredients derived from medicine food homology plants as drugs or food supplements.
Collapse
Affiliation(s)
- Qiuxia Lu
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China
| | - Rui Li
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China
| | - Yixi Yang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China
| | - Yujin Zhang
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Qi Zhao
- College of Food and Biological Engineering, Chengdu University, Chengdu 610106, China
| | - Jian Li
- School of Medicine, Chengdu University, Chengdu 610106, China; Institute of Cancer Biology and Drug Discovery, Chengdu University, Chengdu 610106, China.
| |
Collapse
|
12
|
Ion Channels and Pumps in Autophagy: A Reciprocal Relationship. Cells 2021; 10:cells10123537. [PMID: 34944044 PMCID: PMC8700256 DOI: 10.3390/cells10123537] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 12/24/2022] Open
Abstract
Autophagy, the process of cellular self-degradation, is intrinsically tied to the degradative function of the lysosome. Several diseases have been linked to lysosomal degradative defects, including rare lysosomal storage disorders and neurodegenerative diseases. Ion channels and pumps play a major regulatory role in autophagy. Importantly, calcium signaling produced by TRPML1 (transient receptor potential cation channel, mucolipin subfamily) has been shown to regulate autophagic progression through biogenesis of autophagic-lysosomal organelles, activation of mTORC1 (mechanistic target of rapamycin complex 1) and degradation of autophagic cargo. ER calcium channels such as IP3Rs supply calcium for the lysosome, and lysosomal function is severely disrupted in the absence of lysosomal calcium replenishment by the ER. TRPML1 function is also regulated by LC3 (microtubule-associated protein light chain 3) and mTORC1, two critical components of the autophagic network. Here we provide an overview of the current knowledge about ion channels and pumps-including lysosomal V-ATPase (vacuolar proton-ATPase), which is required for acidification and hence proper enzymatic activity of lysosomal hydrolases-in the regulation of autophagy, and discuss how functional impairment of some of these leads to diseases.
Collapse
|
13
|
Estrogen Decreases Cytoskeletal Organization by Forming an ERα/SHP2/c-Src Complex in Osteoclasts to Protect against Ovariectomy-Induced Bone Loss in Mice. Antioxidants (Basel) 2021; 10:antiox10040619. [PMID: 33920630 PMCID: PMC8073670 DOI: 10.3390/antiox10040619] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/30/2021] [Accepted: 04/15/2021] [Indexed: 12/16/2022] Open
Abstract
Loss of ovarian function is closely related to estrogen (E2) deficiency, which is responsible for increased osteoclast (OC) differentiation and activity. We aimed to investigate the action mechanism of E2 to decrease bone resorption in OCs to protect from ovariectomy (OVX)-induced bone loss in mice. In vivo, tartrate-resistant acid phosphatase (TRAP) staining in femur and serum carboxy-terminal collagen crosslinks-1 (CTX-1) were analyzed upon E2 injection after OVX in mice. In vitro, OCs were analyzed by TRAP staining, actin ring formation, carboxymethylation, determination of reactive oxygen species (ROS) level, and immunoprecipitation coupled with Western blot. In vivo and in vitro, E2 decreased OC size more dramatically than OC number and Methyl-piperidino-pyrazole hydrate dihydrochloride (MPPD), an estrogen receptor alpha (ERα) antagonist, augmented the OC size. ERα was found in plasma membranes and E2/ERα signaling affected receptor activator of nuclear factor κB ligand (RANKL)-induced actin ring formation by rapidly decreasing a proto-oncogene tyrosine-protein kinase, cellular sarcoma (c-Src) (Y416) phosphorylation in OCs. E2 exposure decreased physical interactions between NADPH oxidase 1 (NOX1) and the oxidized form of c-Src homology 2 (SH2)-containing protein tyrosine phosphatase 2 (SHP2), leading to higher levels of reduced SHP2. ERα formed a complex with the reduced form of SHP2 and c-Src to decrease c-Src activation upon E2 exposure, which blocked a signal for actin ring formation by decreased Vav guanine nucleotide exchange factor 3 (Vav3) (p-Y) and Ras-related C3 botulinum toxin substrate 1 (Rac1) (GTP) activation in OCs. E2/ERα signals consistently inhibited bone resorption in vitro. In conclusion, our study suggests that E2-binding to ERα forms a complex with SHP2/c-Src to attenuate c-Src activation that was induced upon RANKL stimulation in a non-genomic manner, resulting in an impaired actin ring formation and reducing bone resorption.
Collapse
|
14
|
Meng L, Lu C, Wu B, Lan C, Mo L, Chen C, Wang X, Zhang N, Lan L, Wang Q, Zeng X, Li X, Tang S. Taurine Antagonizes Macrophages M1 Polarization by Mitophagy-Glycolysis Switch Blockage via Dragging SAM-PP2Ac Transmethylation. Front Immunol 2021; 12:648913. [PMID: 33912173 PMCID: PMC8071881 DOI: 10.3389/fimmu.2021.648913] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Accepted: 03/22/2021] [Indexed: 12/11/2022] Open
Abstract
The excessive M1 polarization of macrophages drives the occurrence and development of inflammatory diseases. The reprogramming of macrophages from M1 to M2 can be achieved by targeting metabolic events. Taurine promotes for the balance of energy metabolism and the repair of inflammatory injury, preventing chronic diseases and complications. However, little is known about the mechanisms underlying the action of taurine modulating the macrophage polarization phenotype. In this study, we constructed a low-dose LPS/IFN-γ-induced M1 polarization model to simulate a low-grade pro-inflammatory process. Our results indicate that the taurine transporter TauT/SlC6A6 is upregulated at the transcriptional level during M1 macrophage polarization. The nutrient uptake signal on the membrane supports the high abundance of taurine in macrophages after taurine supplementation, which weakens the status of methionine metabolism, resulting in insufficient S-adenosylmethionine (SAM). The low availability of SAM is directly sensed by LCMT-1 and PME-1, hindering PP2Ac methylation. PP2Ac methylation was found to be necessary for M1 polarization, including the positive regulation of VDAC1 and PINK1. Furthermore, its activation was found to promote the elimination of mitochondria by macrophages via the mitophagy pathway for metabolic adaptation. Mechanistically, taurine inhibits SAM-dependent PP2Ac methylation to block PINK1-mediated mitophagy flux, thereby maintaining a high mitochondrial density, which ultimately hinders the conversion of energy metabolism to glycolysis required for M1. Our findings reveal a novel mechanism of taurine-coupled M1 macrophage energy metabolism, providing novel insights into the occurrence and prevention of low-grade inflammation, and propose that the sensing of taurine and SAM availability may allow communication to inflammatory response in macrophages.
Collapse
Affiliation(s)
- Ling Meng
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Cailing Lu
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Bin Wu
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Chunhua Lan
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Laiming Mo
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,School of Public Health, Guangxi Medical University, Nanning, China
| | - Chengying Chen
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xinhang Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Ning Zhang
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Li Lan
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Qihui Wang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xia Zeng
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| | - Xiyi Li
- School of Public Health, Guangxi Medical University, Nanning, China
| | - Shen Tang
- School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.,Guangxi Colleges and Universities Key Laboratory of Preclinical Medicine, Guangxi Medical University, Nanning, China
| |
Collapse
|
15
|
Su X, Zhuang D, Zhang Y, Lv H, Wang Y, Luan X, Bi L. Influence of photodynamic therapy on the periodontitis-induced bone resorption in rat. Lasers Med Sci 2021; 36:675-680. [PMID: 32808146 DOI: 10.1007/s10103-020-03126-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/10/2020] [Indexed: 11/29/2022]
Abstract
This study aimed to evaluate the effects of toluidine blue-mediated photodynamic therapy (TB-PDT) on the periodontitis-induced bone resorption in periodontitis in rats. Periodontal disease was induced by cotton ligature around the right second maxillary molar in 64 rats. After 4 weeks, the rats were randomly divided into four groups: sterile saline solution (control group); laser therapy (laser group); TB (100 μg/mL); TB plus laser (0.15 W/cm2) irradiation every other day for 240 s (PDT group). All rats were euthanized at 15 days postoperatively. Eight gingival tissue samples were collected from each group. The expressions of receptor activator of nuclear factor kappa-Β ligand (RANKL) and osteoprotegerin (OPG) in gingival tissue samples were detected by real-time quantitative polymerase chain reaction (RT-qPCR). The maxillae from the rest of the rats were taken for histological examination. In the PDT group, the analysis revealed less bone loss than in the control treatment (P < 0.05). No significant difference was found among the control group, TB group, and laser group (P > 0.05). Significantly higher and lower expressions of RANKL and OPG were revealed in the PDT group than that in control group, respectively (P < 0.01). When compared with the control group, the expression of RANKL was significantly reduced by 40.0% in periodontitis in rats treated with TB-PDT for 15 days (P < 0.01). The expression of OPG was increased in the PDT group with TB-PDT for 15 days, when compared with the control group (P < 0.05). TB-PDT treatment significantly reverses the abnormal expression of RANKL and OPG in periodontitis in rats.
Collapse
Affiliation(s)
- Xin Su
- Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Deshu Zhuang
- Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
- Faculty of Dentistry, Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Yi Zhang
- Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Han Lv
- Department of Stomatology, Centre Hospital of Longhua District, Shenzhen, 518000, China
| | - Yijing Wang
- Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Xiaomin Luan
- Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, China
| | - Liangjia Bi
- Department of Stomatology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, 150001, China.
| |
Collapse
|
16
|
Effects of Various Preextraction Treatments of Crinum asiaticum Leaf on Its Anti-Inflammatory Activity and Chemical Properties. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2021; 2021:8850744. [PMID: 33574885 PMCID: PMC7861935 DOI: 10.1155/2021/8850744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/20/2020] [Accepted: 01/04/2021] [Indexed: 11/17/2022]
Abstract
Crinum asiaticum Linn. has been used in Thai traditional medicine to relieve inflammatory symptoms and treat osteoarthritis. There have been reports on its potent anti-inflammatory property but nothing on the effects of different pretreatments on its chemical properties and anti-inflammatory activity. Pretreatment of herbal raw materials is an important step which affects the overall quality of Thai traditional medicine. The objectives of this study were to investigate different treatments of C. asiaticum leaves prior to ethanolic extraction and to compare the extracts for their anti-inflammatory activity and chemical properties. The treatments included hot air drying in an oven, microwave drying, traditional grilling on a charcoal stove before drying in an oven, and temperature shock in hot and cold water before hot air drying. The anti-inflammatory activity and chemical properties of the extracts were analyzed using the established methods. Results showed that 95% ethanolic extract of hot air oven-dried leaves had the highest anti-inflammatory activity and total phenolic and lycorine contents. We recommend hot air drying as a preextraction treatment for C. asiaticum leaves for its simplicity, best retention of the herbal quality, and suitability for scaling up to an industrial process.
Collapse
|
17
|
Li X, Xu J, Dai B, Wang X, Guo Q, Qin L. Targeting autophagy in osteoporosis: From pathophysiology to potential therapy. Ageing Res Rev 2020; 62:101098. [PMID: 32535273 DOI: 10.1016/j.arr.2020.101098] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/26/2020] [Accepted: 06/03/2020] [Indexed: 12/19/2022]
Abstract
Osteoporosis is a highly prevalent disorder characterized by the loss of bone mass and microarchitecture deterioration of bone tissue, attributed to various factors, including menopause (primary), aging (primary) and adverse effects of relevant medications (secondary). In recent decades, knowledge regarding the etiological mechanisms underpinning osteoporosis emphasizes that bone cellular homeostasis, including the maintenance of cell functions, differentiation, and the response to stress, is tightly regulated by autophagy, which is a cell survival mechanism for eliminating and recycling damaged proteins and organelles. With the important roles in the maintenance of cellular homeostasis and organ function, autophagy has emerged as a potential target for the prevention and treatment of osteoporosis. In this review, we update and discuss the pathophysiology of autophagy in normal bone cell life cycle and metabolism. Then, the alternations of autophagy in primary and secondary osteoporosis, and the accompanied pathological process are discussed. Finally, we discuss current strategies, limitations, and challenges involved in targeting relevant pathways and propose strategies by which such hurdles may be circumvented in the future for their translation into clinical validations and applications for the prevention and treatment of osteoporosis.
Collapse
|
18
|
Ge X, Meng X, Fei D, Kang K, Wang Q, Zhao M. Lycorine attenuates lipopolysaccharide-induced acute lung injury through the HMGB1/TLRs/NF-κB pathway. 3 Biotech 2020; 10:369. [PMID: 32818131 PMCID: PMC7395800 DOI: 10.1007/s13205-020-02364-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/25/2020] [Indexed: 12/20/2022] Open
Abstract
Lung injury associated with systemic inflammatory response is a common problem affecting human health. Previous studies have shown that lycorine exerts a anti-inflammatory effect. However, whether lycorine alleviates lung injury remains unclear. To explore this issue, BALB/c mice and MLE-12 cells were treated with lipopolysaccharide (LPS) to establish lung injury mouse model and cell model, respectively. Glycyrrhizic acid, known as an inhibitor of ALI, was also used to study the effects of lycorine in vitro. Our results showed that after LPS treatment, the lung injury score, lung wet-to-dry weight ratio, and malondialdehyde (MDA) production in the lung tissues and the expression levels of tumor necrosis factor-α, interleukin-1β, and interleukin-6 in bronchoalveolar lavage fluid were significantly increased, whereas their levels were decreased by lycorine. Additionally, LPS injection activated the high-mobility group box 1 (HMGB1)/Toll-like receptors (TLRs)/NF-κB pathway. However, lycorine treatment attenuated the activity of the HMGB1/TLRs/NF-κB pathway in the lung tissues. In vitro studies showed that lycorine administration significantly decreased the levels of inflammatory cytokines and MDA and attenuated the activity of the HMGB1/TLRs/NF-κB pathway in LPS-treated cells. Moreover, the inhibitory effects of lycorine on the inflammatory response and oxidative stress in LPS-treated lung cells were similar with that of glycyrrhizic acid, and this inhibition was intensified by both lycorine and glycyrrhizic acid treatment. We suggest that lycorine could alleviate LPS-induced lung injury of inflammation and oxidative stress by blocking the HMGB1/TLRs/NF-κB pathway, which gives a new perspective for ALI therapy to treat lycorine as a potential treatment clinically.
Collapse
Affiliation(s)
- Xin Ge
- Department of ICU, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001 Heilongjiang People’s Republic of China
- Department of ICU, Wuxi 9th Affiliated Hospital of Soochow University, Wuxi, 214000 Jiangsu People’s Republic of China
| | - Xianglin Meng
- Department of ICU, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001 Heilongjiang People’s Republic of China
| | - Dongsheng Fei
- Department of ICU, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001 Heilongjiang People’s Republic of China
| | - Kai Kang
- Department of ICU, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001 Heilongjiang People’s Republic of China
| | - Qiubo Wang
- Department of Clinical Laboratory, Wuxi 9th Affiliated Hospital of Soochow University, Wuxi, 214000 Jiangsu People’s Republic of China
| | - Mingyan Zhao
- Department of ICU, The First Affiliated Hospital of Harbin Medical University, No. 23 Youzheng Street, Harbin, 150001 Heilongjiang People’s Republic of China
| |
Collapse
|
19
|
Park HJ, Gholam Zadeh M, Suh JH, Choi HS. Dauricine Protects from LPS-Induced Bone Loss via the ROS/PP2A/NF-κB Axis in Osteoclasts. Antioxidants (Basel) 2020; 9:antiox9070588. [PMID: 32640590 PMCID: PMC7402093 DOI: 10.3390/antiox9070588] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/29/2020] [Accepted: 07/03/2020] [Indexed: 01/03/2023] Open
Abstract
Dauricine (DAC), an isoquinoline alkaloid, exhibits anti-inflammatory activity. We hypothesized that DAC may prevent the inflammatory bone loss induced by lipopolysaccharide (LPS). LPS-induced bone loss was decreased by DAC in female C57BL/6J mice as evaluated by micro-computerized tomography (μCT) analysis. In vivo tartrate-resistant acid phosphatase (TRAP) staining showed that the increased number of osteoclasts (OCs) in LPS-treated mice was attenuated by DAC, indicating that DAC exhibited bone sparing effects through acting on OCs. DAC also decreased the differentiation and activity of OCs after LPS stimulation in vitro. LPS-induced cytosolic reactive oxygen species (cROS) oxidized PP2A, a serine/threonine phosphatase, leading to the activation of IKKα/β, followed by the nuclear localization of p65. DAC decreased LPS-induced ROS, resulting in the recovery of the activity of PP2A by reducing its oxidized form. Consequently, DAC reduced the phosphorylation of IKKα/β to block the nuclear localization of p65, which decreased NF-κB activation. Taken together, DAC reduced the differentiation and activity of OCs by decreasing ROS via the ROS/PP2A/NF-κB axis, resulting in protection from LPS-induced bone loss. We have demonstrated that LPS-induced bone loss was inhibited by DAC via its action on OCs, implying the therapeutic potential of DAC against inflammatory bone loss.
Collapse
Affiliation(s)
- Hyun-Jung Park
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (M.G.Z.)
| | | | - Jae-Hee Suh
- Department of Pathology, Ulsan University Hospital, Ulsan 44030, Korea;
| | - Hye-Seon Choi
- Department of Biological Sciences, University of Ulsan, Ulsan 44610, Korea; (H.-J.P.); (M.G.Z.)
- Correspondence: ; Tel.: +82-52-259-1545; Fax: +82-52-259-1694
| |
Collapse
|
20
|
Research Progress on Anti-Inflammatory Effects and Mechanisms of Alkaloids from Chinese Medical Herbs. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:1303524. [PMID: 32256634 PMCID: PMC7104124 DOI: 10.1155/2020/1303524] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 02/17/2020] [Indexed: 12/18/2022]
Abstract
As the spectrum of diseases keeps changing and life pace keeps going faster, the probability and frequency of diseases caused by human inflammatory reactions also keep increasing. How to develop effective anti-inflammatory drugs has become the hotspot of researches. It has been found that alkaloids from Chinese medical herbs have anti-inflammatory, analgesic, antitumor, anticonvulsant, diuretic, and antiarrhythmic effects, among which the anti-inflammatory effect is very prominent and commonly used in the treatment of rheumatoid arthritis, ankylosing spondylitis, and other rheumatic immune diseases, but its mechanism of action has not been well explained. Based on this, this paper will classify alkaloids according to structural types and review the plant sources, applicable diseases, and anti-inflammatory mechanisms of 16 kinds of alkaloids commonly used in clinical treatment, such as berberine, tetrandrine, and stephanine, with the aim of providing a reference for drug researches and clinical applications.
Collapse
|