1
|
Jacques LS, Pereira JPC, Santos BM, Barrioni BR, Del Bianco Borges B. Flaxseed and mulberry extract improve trabecular bone quality in estrogen-deficient rats. Climacteric 2025; 28:175-183. [PMID: 39937165 DOI: 10.1080/13697137.2025.2457988] [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: 08/19/2024] [Revised: 10/31/2024] [Accepted: 01/14/2025] [Indexed: 02/13/2025]
Abstract
Many hormones, including estrogens, modulate bone metabolism, which plays a crucial role in maintaining bone health. Estrogen depletion, as occurs in menopause, leads to increased bone resorption and decreased formation, resulting in osteopenia/osteoporosis. This study investigates the effects of flaxseed (Linum usitatissimum) and mulberry (Morus nigra L.) extracts, known for their phenolic compounds and antioxidant properties, against estrogen deficiency-induced bone loss in female Wistar rats. These extracts were administered to ovariectomized rats for 60 days. High-performance liquid chromatography analysis revealed the presence of some phenolic compounds in the extracts, including trigonelline, gallic acid, theobromine, chlorogenic acid, syringic acid and p-coumaric acid. The extracts improved bone microstructure with higher trabecular bone, bone mineral density, calcium, phosphorus and magnesium levels, and lower porosity and intertrabecular space in bone tissue. Furthermore, plasma alkaline phosphatase activity was elevated in extract-treated animals, indicating enhanced bone tissue formation. Although serum carboxy-terminal fragment levels showed no significant change, the data suggest that flaxseed and mulberry extracts may protect against trabecular bone loss and support bone formation in estrogen-deficient conditions. These results suggest that supplementing these natural extracts holds promise in preventing or alleviating the signs and symptoms associated with estrogenic deficiency.
Collapse
Affiliation(s)
- Larissa Sampaio Jacques
- Medicine Department, Health Science Faculty, Lavras Federal University - UFLA, Lavras, Brazil
| | | | - Beatriz Menegate Santos
- Medicine Department, Health Science Faculty, Lavras Federal University - UFLA, Lavras, Brazil
| | - Breno Rocha Barrioni
- Postgraduate Program in Metallurgical Engineering, Mines and Materials, Minas Gerais Federal University- UFMG, Belo Horizonte, Brazil
| | - Bruno Del Bianco Borges
- Medicine Department, Health Science Faculty, Lavras Federal University - UFLA, Lavras, Brazil
| |
Collapse
|
2
|
Sukkho T, Khanongnuch C, Lumyong S, Ruangsuriya J, Apichai S, Surh YJ, Pattananandecha T, Saenjum C. Osteoprotective Activity of Sambucus javanica Reinw Ex Blume subsp. javanica Leaf Extracts by Suppressing ROS Production. Antioxidants (Basel) 2025; 14:252. [PMID: 40227226 PMCID: PMC11939775 DOI: 10.3390/antiox14030252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 04/15/2025] Open
Abstract
Sambucus javanica subsp. javanica (SJ) has been used in traditional medicine in the northern region of Thailand for healing bone fractures; however, studies on how this plant stimulates bone formation are still scarce. The present study aimed to investigate the potential of crude extracts and fractions obtained from SJ leaves for osteoporotic protection. All samples were investigated in murine preosteoblast MC3T3-E1 cells for bone formation and resorption biomarkers, namely alkaline phosphatase (ALP), osteocalcin (OC), osteoprotegerin (OPG), receptor activator of nuclear factor-κB ligand (RANKL), and the OPG/RANKL ratio. Additionally, calcium deposits were determined using the alizarin red S staining technique. The results indicated that the crude water and the crude ethanol extracts contained gallic acid, rutin, and chlorogenic acid as major compounds. The extracts stimulated osteoblastic cell differentiation and enhanced osteoprotective activity, as measured by a significant increase in ALP activity, OC, OPG, the OPG/RANKL ratio, and the degree of calcification. Additionally, they exhibited a negative impact on bone resorption by significantly reducing RANKL and reactive oxygen species (ROS) production. Therefore, our findings add novel evidence indicating that the SJ crude extracts from water and ethanol extraction could be further utilized as a natural active pharmaceutical ingredient (NAPI) for the development of bone health products.
Collapse
Affiliation(s)
- Treethip Sukkho
- Department of Biotechnology, Multidisciplinary and Interdisciplinary School, Chiang Mai University, Chiang Mai 50200, Thailand;
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
| | - Chartchai Khanongnuch
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Research Center for Multidisciplinary Approaches to Miang, Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand
| | - Saisamorn Lumyong
- Department of Biology, Faculty of Sciences, Chiang Mai University, Chiang Mai 50200, Thailand;
- Research Center of Microbial Diversity and Sustainable Utilization, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Jetsada Ruangsuriya
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sutasinee Apichai
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Young-Joon Surh
- College of Pharmacy, Seoul National University, Seoul 08828, Republic of Korea;
| | - Thanawat Pattananandecha
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Chalermpong Saenjum
- Research Center for Innovation in Analytical Science and Technology for Biodiversity-Based Economic and Society (I-ANALY-S-T_B.BES-CMU), Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand; (C.K.); (J.R.); (S.A.)
- Research Center for Multidisciplinary Approaches to Miang, Multidisciplinary Research Institute (MDRI), Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
3
|
Abilkassymova A, Aldana-Mejía JA, Katragunta K, Kozykeyeva R, Omarbekova A, Avula B, Turgumbayeva A, Datkhayev UM, Khan IA, Ross SA. Phytochemical Screening Using LC-MS to Study Antioxidant and Toxicity Potential of Methanolic Extracts of Atraphaxis pyrifolia Bunge. Molecules 2024; 29:4478. [PMID: 39339473 PMCID: PMC11434437 DOI: 10.3390/molecules29184478] [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: 08/06/2024] [Revised: 09/13/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Atraphaxis pyrifolia, a native medicinal plant of Central Asia, has a long history of traditional medicinal use; however, scientific research on its phytochemical and biological properties remains scarce. This paper aims to elucidate its chemical profile and assess its pharmacological potential through a comprehensive investigation of the phytochemical composition of stems and leaves using Liquid Chromatography-Mass Spectrometry (LC-MS), in conjunction with the assessment of its antioxidant (DPPH and ABTS) and cytotoxicity test on Artemia salina. Predominantly, glycosylated flavonoids were detected in stems and leaves extracts, notably including 8-Acetoxy-3',4',5,5'-tetrahydroxy-7-methoxy-3-α-L-rhamno-pyranosyloxyflavone, pyrifolin, and dehydroxypyrifolin. While the latter compound is exclusive to A. pyrifolia, the former compounds serve as shared chemical markers with other Atraphaxis species. The methanolic extracts of A. pyrifolia leaves exhibited significant antioxidant capacity without toxicity against Artemia salina. This study contributes to current research through providing valuable insights into the chemical diversity and potential medicinal properties of this plant species.
Collapse
Affiliation(s)
- Alima Abilkassymova
- Higher School of Medicine, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (A.A.); (A.T.)
- School of Pharmacy, Asfendiyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.O.); (U.M.D.)
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (J.A.A.-M.); (K.K.); (R.K.); (B.A.); (I.A.K.)
| | - Jennyfer A. Aldana-Mejía
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (J.A.A.-M.); (K.K.); (R.K.); (B.A.); (I.A.K.)
| | - Kumar Katragunta
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (J.A.A.-M.); (K.K.); (R.K.); (B.A.); (I.A.K.)
| | - Raushan Kozykeyeva
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (J.A.A.-M.); (K.K.); (R.K.); (B.A.); (I.A.K.)
- Faculty of Pharmacy, South Kazakhstan Medical Academy, Shymkent 160019, Kazakhstan
| | - Ardak Omarbekova
- School of Pharmacy, Asfendiyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.O.); (U.M.D.)
- Faculty of Pharmacy, South Kazakhstan Medical Academy, Shymkent 160019, Kazakhstan
| | - Bharathi Avula
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (J.A.A.-M.); (K.K.); (R.K.); (B.A.); (I.A.K.)
| | - Aknur Turgumbayeva
- Higher School of Medicine, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (A.A.); (A.T.)
- School of Pharmacy, Asfendiyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.O.); (U.M.D.)
| | - Ubaidilla M. Datkhayev
- School of Pharmacy, Asfendiyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.O.); (U.M.D.)
| | - Ikhlas A. Khan
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (J.A.A.-M.); (K.K.); (R.K.); (B.A.); (I.A.K.)
- Department of Biomolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| | - Samir A. Ross
- School of Pharmacy, Asfendiyarov Kazakh National Medical University, Almaty 050012, Kazakhstan; (A.O.); (U.M.D.)
- National Center for Natural Products Research, School of Pharmacy, The University of Mississippi, Oxford, MS 38677, USA; (J.A.A.-M.); (K.K.); (R.K.); (B.A.); (I.A.K.)
- Department of Biomolecular Sciences, Division of Pharmacognosy, School of Pharmacy, University of Mississippi, Oxford, MS 38677, USA
| |
Collapse
|
4
|
Cai Y, Jiang J, Yue C, Zhang Z, Liu W. Gallic acid promotes macrophage phagosome acidification and phagolysosome formation by activating NLRP3/mTOR signaling pathway. J Infect Chemother 2024; 30:867-875. [PMID: 38462174 DOI: 10.1016/j.jiac.2024.02.030] [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: 11/20/2023] [Revised: 02/20/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Gallic acid (GA) has a good therapeutic effect in bacteriological inhibition and plays a variety of functions in maintaining the stability of the immune system. The aim of the present study was to investigate the effect of GA on the bactericidal activity of macrophages against Vibrio vulnificus (Vv). METHODS A cell counting kit-8 (CCK-8) assay was carried out to test the cytotoxicity of GA on J774A.1 cells. Concentration of proinflammatory cytokines in J774A.1 cells were evaluated by ELISA. The internalization and degradation of Vv in the phagosomes were observed by transmission electron microscopy (TEM). The phagosome acidification and phagolysosome formation were detected to evaluate the bacteria-clearing function of J774A.1 cells. The bactericidal activity of GA in vivo was also investigated by collecting the survival time of Vv infected mice and observing the inflammatory infiltration of organs. RESULTS Our results demonstrated that GA at 50 μM significantly inhibited the proinflammatory cytokines levels, promoted phagosome acidification and phagolysosome formation in J774A.1 cells with Vv infection. This may be related to the activation of NLRP3/mTOR signaling pathway. Additionally, GA treatment improves the survival and bactericidal activity of mice infected with Vv. CONCLUSIONS In summary, GA exerts bactericidal activity against Vv infection by regulating the formation and acidification of phagocytic lysosomes in macrophages.
Collapse
Affiliation(s)
- Yanqu Cai
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Guangzhou Universities Town Campus, Guangzhou, China; Key Laboratory of Modern Chinese Medicine of Education Department of Guangdong Province, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Guangzhou Universities Town Campus, Guangzhou, China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery Systems, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Guangzhou Universities Town Campus, Guangzhou, China; Class III Laboratory of Modern Chinese Medicine Preparation, State Administration of Traditional Chinese Medicine of the P.R.C, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Guangzhou Universities Town Campus, Guangzhou, China.
| | - Jinzhu Jiang
- Center for New Drug Research and Development, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Guangzhou Universities Town Campus, Guangzhou, China
| | - Chunhua Yue
- College of Pharmacy, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Guangzhou Universities Town Campus, Guangzhou, China
| | - Zhipeng Zhang
- College of Pharmacy, Hubei University of Science & Technology, No. 88, Xianning Avenue, Xianning, China
| | - Wenbin Liu
- School of Basic Medical Sciences, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Guangzhou Universities Town Campus, Guangzhou, China; Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, No. 280, East Waihuan Road, Guangzhou Universities Town Campus, Guangzhou, China.
| |
Collapse
|
5
|
Zhang T, Zhang X, Fei Y, Lu J, Zhou D, Zhang L, Fan S, Zhou J, Liang C, Su Y. Gallic acid suppresses the progression of clear cell renal cell carcinoma through inducing autophagy via the PI3K/Akt/Atg16L1 signaling pathway. Int J Oncol 2024; 65:70. [PMID: 38818827 PMCID: PMC11173374 DOI: 10.3892/ijo.2024.5658] [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: 06/10/2023] [Accepted: 11/22/2023] [Indexed: 06/01/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC), the most common type of renal cell carcinoma (RCC), is not sensitive to traditional radiotherapy and chemotherapy. The polyphenolic compound Gallic acid (GA) can be naturally found in a variety of fruits, vegetables and plants. Autophagy, an intracellular catabolic process, regulates the lysosomal degradation of organelles and portions in cytoplasm. It was reported that autophagy and GA could affect the development of several cancers. Therefore, the aim of the present study was to evaluate the effects of GA on ccRCC development and clarify the role of autophagy in this process. In the present study, the effects of GA on the proliferation, migration and invasion of ccRCC cells were investigated in vitro by Cell Counting Kit‑8, colony formation, flow cytometry, wound healing and Transwell migration assays, respectively. Additionally, the effects of GA on ccRCC growth and metastasis were evaluated using hematoxylin‑eosin and immunohistochemical staining in vivo. Moreover, it was sought to explore the underlying molecular mechanisms using transmission electron microscopy, western blotting and reverse transcription‑quantitative PCR analyses. In the present study, it was revealed that GA had a more potent viability inhibitory effect on ccRCC cells (786‑O and ACHN) than the effect on normal renal tubular epithelial cell (HK‑2), which demonstrated that GA selectively inhibits the viability of cancer cells. Furthermore, it was identified that GA dose‑dependently inhibited the proliferation, migration and invasion of ccRCC cells in vitro and in vivo. It was demonstrated that GA promoted the release of autophagy markers, which played a role in regulating the PI3K/Akt/Atg16L1 signaling pathway. All the aforementioned data provided evidence for the great potential of GA in the treatment of ccRCC.
Collapse
Affiliation(s)
- Tianxiang Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032
- State Key Laboratory of Systems Medicine for Cancer, Department of Urology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127
| | - Xi Zhang
- Department of Urology, The Second Hospital of Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Yang Fei
- State Key Laboratory of Systems Medicine for Cancer, Department of Urology, Shanghai Cancer Institute, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127
| | - Jinsen Lu
- Nuffield Department of Orthopedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Dairan Zhou
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, Shanghai 200003
| | - Li Zhang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032
- Institute of Urology, Anhui Medical University, Hefei, Anhui 230032
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui 230032, P.R. China
| | - Song Fan
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032
- Institute of Urology, Anhui Medical University, Hefei, Anhui 230032
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui 230032, P.R. China
| | - Jun Zhou
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032
- Institute of Urology, Anhui Medical University, Hefei, Anhui 230032
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui 230032, P.R. China
| | - Chaozhao Liang
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032
- Institute of Urology, Anhui Medical University, Hefei, Anhui 230032
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui 230032, P.R. China
| | - Yang Su
- Department of Urology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230032
- Institute of Urology, Anhui Medical University, Hefei, Anhui 230032
- Anhui Province Key Laboratory of Urological and Andrological Diseases Research and Medical Transformation, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
6
|
Lv S, Zhang G, Lu Y, Zhong X, Huang Y, Ma Y, Yan W, Teng J, Wei S. Pharmacological mechanism of natural antidepressants: The role of mitochondrial quality control. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 129:155669. [PMID: 38696923 DOI: 10.1016/j.phymed.2024.155669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/15/2024] [Accepted: 04/21/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND Depression is a mental illness characterized by persistent sadness and a reduced capacity for pleasure. In clinical practice, SSRIs and other medications are commonly used for therapy, despite their various side effects. Natural products present distinct advantages, including synergistic interactions among multiple components and targeting multiple pathways, suggesting their tremendous potential in depression treatment. Imbalance in mitochondrial quality control (MQC) plays a significant role in the pathology of depression, emphasizing the importance of regulating MQC as a potential intervention strategy in addressing the onset and progression of depression. However, the role and mechanism through which natural products regulate MQC in depression treatments still need to be comprehensively elucidated, particularly in clinical and preclinical settings. PURPOSE This review was aimed to summarize the findings of recent studies and outline the pharmacological mechanisms by which natural products modulate MQC to exert antidepressant effects. Additionally, it evaluated current research limitations and proposed new strategies for future preclinical and clinical applications in the depression domain. METHODS To study the main pharmacological mechanisms underlying the regulation of MQC by natural products in the treatment of depression, we conducted a thorough search across databases such as PubMed, Web of Science, and ScienceDirect databases to classify and summarize the relationship between MQC and depression, as well as the regulatory mechanisms of natural products. RESULTS Numerous studies have shown that irregularities in the MQC system play an important role in the pathology of depression, and the regulation of the MQC system is involved in antidepressant treatments. Natural products mainly regulate the MQC system to induce antidepressant effects by alleviating oxidative stress, balancing ATP levels, promoting mitophagy, maintaining calcium homeostasis, optimizing mitochondrial dynamics, regulating mitochondrial membrane potential, and enhancing mitochondrial biogenesis. CONCLUSIONS We comprehensively summarized the regulation of natural products on the MQC system in antidepressants, providing a unique perspective for the application of natural products within antidepressant therapy. However, extensive efforts are imperative in clinical and preclinical investigations to delve deeper into the mechanisms underlying how antidepressant medications impact MQC, which is crucial for the development of effective antidepressant treatments.
Collapse
Affiliation(s)
- Shimeng Lv
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Guangheng Zhang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yitong Lu
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Xia Zhong
- Institute of Child and Adolescent Health, School of Public Health, Peking University, Beijing 100191, China
| | - Yufei Huang
- Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200025, China
| | - Yuexiang Ma
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355,China
| | - Wei Yan
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Jing Teng
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan 250014, China.
| | - Sheng Wei
- Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; High Level Key Disciplines of Traditional Chinese Medicine: Basic Theory of Traditional Chinese Medicine, National Administration of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; Shandong Provincial Engineering Research Center for the Prevention and Treatment of Major Brain Diseases with Traditional Chinese Medicine (PTMBD), Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
7
|
Abilkassymova A, Turgumbayeva A, Sarsenova L, Tastambek K, Altynbay N, Ziyaeva G, Blatov R, Altynbayeva G, Bekesheva K, Abdieva G, Ualieva P, Shynykul Z, Kalykova A. Exploring Four Atraphaxis Species: Traditional Medicinal Uses, Phytochemistry, and Pharmacological Activities. Molecules 2024; 29:910. [PMID: 38398660 PMCID: PMC10891555 DOI: 10.3390/molecules29040910] [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: 12/15/2023] [Revised: 02/03/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Atraphaxis is a genus of flowering plants in the family Polygonaceae, with approximately 60 species. Species of Atraphaxis are much-branched woody plants, forming shrubs or shrubby tufts, primarily inhabiting arid zones across the temperate steppe and desert regions of Central Asia, America, and Australia. Atraphaxis species have been used by diverse groups of people all over the world for the treatment of various diseases. However, their biologically active compounds with therapeutic properties have not been investigated well. Studying the biologically active components of Atraphaxis laetevirens, Atraphaxis frutescens, Atraphaxis spinosa L., and Atraphaxis pyrifolia is crucial for several reasons. Firstly, it can unveil the therapeutic potential of these plants, aiding in the development of novel medicines or natural remedies for various health conditions. Understanding their bioactive compounds enables scientists to explore their pharmacological properties, potentially leading to the discovery of new drugs or treatments. Additionally, investigating these components contributes to preserving traditional knowledge and validating the historical uses of these plants in ethnomedicine, thus supporting their conservation and sustainable utilization. These herbs have been used as an anti-inflammatory and hypertension remedies since the dawn of time. Moreover, they have been used to treat a variety of gastrointestinal disorders and problems related to skin in traditional Kazakh medicine. Hence, the genus Atraphaxis can be considered as a potential medicinal plant source that is very rich in biologically active compounds that may exhibit great pharmacological properties, such as antioxidant, antibacterial, antiulcer, hypoglycemic, wound healing, neuroprotective, antidiabetic, and so on. This study aims to provide a collection of publications on the species of Atraphaxis, along with a critical review of the literature data. This review will constitute support for further investigations on the pharmacological activity of these medicinal plant species.
Collapse
Affiliation(s)
- Alima Abilkassymova
- Higher School of Medicine, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (L.S.); (Z.S.); (A.K.)
| | - Aknur Turgumbayeva
- Higher School of Medicine, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (L.S.); (Z.S.); (A.K.)
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK
| | - Lazzat Sarsenova
- Higher School of Medicine, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (L.S.); (Z.S.); (A.K.)
| | - Kuanysh Tastambek
- Institute of Ecology, Khoja Akhmet Yassawi International Kazakh-Turkish University, Turkistan 161200, Kazakhstan;
| | - Nazym Altynbay
- Institute of Ecological Problems, Al-Farabi Kazakh National University, Al-Farabi Ave. 71, Almaty 050040, Kazakhstan;
| | - Gulnar Ziyaeva
- Department of Biology, Taraz Regional University Named after M.Kh.Dulaty, Taraz 080000, Kazakhstan;
| | - Ravil Blatov
- Department of Pharmacy, Kazakh-Russian Medical University, Almaty 050000, Kazakhstan;
| | - Gulmira Altynbayeva
- School of Pharmacy, JSC “S.D. Asfendiyarov Kazakh National Medical University”, Almaty 050000, Kazakhstan;
- Neonatology and Neonatal Surgery Department, JSC “Scientific Center of Pediatrics and Pediatric Surgery”, Almaty 050060, Kazakhstan
| | - Kuralay Bekesheva
- JSC “Scientific Centre for Anti-Infectious Drugs”, Almaty 010000, Kazakhstan;
| | - Gulzhamal Abdieva
- Department of Biotechnology, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Al-Farabi 71, Almaty 050040, Kazakhstan; (G.A.); (P.U.)
| | - Perizat Ualieva
- Department of Biotechnology, Faculty of Biology and Biotechnology, Al-Farabi Kazakh National University, Al-Farabi 71, Almaty 050040, Kazakhstan; (G.A.); (P.U.)
| | - Zhanserik Shynykul
- Higher School of Medicine, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (L.S.); (Z.S.); (A.K.)
| | - Assem Kalykova
- Higher School of Medicine, Al-Farabi Kazakh National University, Almaty 050040, Kazakhstan; (L.S.); (Z.S.); (A.K.)
- School of Life Sciences, University of Westminster, 115 New Cavendish Street, London W1W 6UW, UK
| |
Collapse
|
8
|
Chen Y, Gan W, Cheng Z, Zhang A, Shi P, Zhang Y. Plant molecules reinforce bone repair: Novel insights into phenol-modified bone tissue engineering scaffolds for the treatment of bone defects. Mater Today Bio 2024; 24:100920. [PMID: 38226013 PMCID: PMC10788623 DOI: 10.1016/j.mtbio.2023.100920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/17/2024] Open
Abstract
Bone defects have become a major cause of disability and death. To overcome the limitations of natural bone implants, including donor shortages and immune rejection risks, bone tissue engineering (BTE) scaffolds have emerged as a promising therapy for bone defects. Despite possessing good biocompatibility, these metal, ceramic and polymer-based scaffolds are still challenged by the harsh conditions in bone defect sites. ROS accumulation, bacterial infection, excessive inflammation, compromised blood supply deficiency and tumor recurrence negatively impact bone tissue cells (BTCs) and hinder the osteointegration of BTE scaffolds. Phenolic compounds, derived from plants and fruits, have gained growing application in treating inflammatory, infectious and aging-related diseases due to their antioxidant ability conferred by phenolic hydroxyl groups. The prevalent interactions between phenols and functional groups also facilitate their utilization in fabricating scaffolds. Consequently, phenols are increasingly incorporated into BTE scaffolds to boost therapeutic efficacy in bone defect. This review demonstrated the effects of phenols on BTCs and bone defect microenvironment, summarized the intrinsic mechanisms, presented the advances in phenol-modified BTE scaffolds and analyzed their potential risks in practical applications. Overall, phenol-modified BTE scaffolds hold great potential for repairing bone defects, offering novel patterns for BTE scaffold construction and advancing traumatological medicine.
Collapse
Affiliation(s)
| | | | | | - Anran Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengzhi Shi
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yukun Zhang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
9
|
Tang X, Huang Y, Fang X, Tong X, Yu Q, Zheng W, Fu F. Cornus officinalis: a potential herb for treatment of osteoporosis. Front Med (Lausanne) 2023; 10:1289144. [PMID: 38111697 PMCID: PMC10725965 DOI: 10.3389/fmed.2023.1289144] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 11/17/2023] [Indexed: 12/20/2023] Open
Abstract
Osteoporosis (OP) is a systemic metabolic skeletal disorder characterized by a decline in bone mass, bone mineral density, and deterioration of bone microstructure. It is prevalent among the elderly, particularly postmenopausal women, and poses a substantial burden to patients and society due to the high incidence of fragility fractures. Kidney-tonifying Traditional Chinese medicine (TCM) has long been utilized for OP prevention and treatment. In contrast to conventional approaches such as hormone replacement therapy, TCM offers distinct advantages such as minimal side effects, low toxicity, excellent tolerability, and suitability for long-term administration. Extensive experimental evidence supports the efficacy of kidney-tonifying TCM, exemplified by formulations based on the renowned herb Cornus officinalis and its bioactive constituents, including morroniside, sweroside, flavonol kaempferol, Cornuside I, in OP treatment. In this review, we provide a comprehensive elucidation of the underlying pathological principles governing OP, with particular emphasis on bone marrow mesenchymal stem cells, the homeostasis of osteogenic and osteoclastic, and the regulation of vascular and immune systems, all of which critically influence bone homeostasis. Furthermore, the therapeutic mechanisms of Cornus officinalis-based TCM formulations and Cornus officinalis-derived active constituents are discussed. In conclusion, this review aims to enhance understanding of the pharmacological mechanisms responsible for the anti-OP effects of kidney-tonifying TCM, specifically focusing on Cornus officinalis, and seeks to explore more efficacious and safer treatment strategies for OP.
Collapse
Affiliation(s)
- Xinyun Tang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, China
| | - Yuxin Huang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, China
| | - Xuliang Fang
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, China
| | - Xuanying Tong
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
- The Third Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, China
| | - Qian Yu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
- The First Clinical Medical College, Zhejiang Chinese Medical University, Zhejiang, China
| | - Wenbiao Zheng
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou, China
| | - Fangda Fu
- Institute of Orthopaedics and Traumatology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| |
Collapse
|
10
|
Kim JW, Choi J, Park MN, Kim B. Apoptotic Effect of Gallic Acid via Regulation of p-p38 and ER Stress in PANC-1 and MIA PaCa-2 Cells Pancreatic Cancer Cells. Int J Mol Sci 2023; 24:15236. [PMID: 37894916 PMCID: PMC10607041 DOI: 10.3390/ijms242015236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Pancreatic cancer (PC) is currently recognized as the seventh most prevalent cause of cancer-related mortality among individuals of both genders. It is projected that a significant number of individuals will succumb to this disease in the forthcoming years. Extensive research and validation have been conducted on both gemcitabine and 5-fluorouracil as viable therapeutic options for PC. Nevertheless, despite concerted attempts to enhance treatment outcomes, PC continues to pose significant challenges in terms of achieving effective treatment alone through chemotherapy. Gallic acid, an endogenous chemical present in various botanical preparations, has attracted considerable attention due to its potential as an anticancer agent. The results of the study demonstrated that gallic acid exerted a decline in cell viability that was dependent on its concentration. Furthermore, it efficiently suppressed cell proliferation in PC cells. This study observed a positive correlation between gallic acid and the production of reactive oxygen species (ROS). Additionally, it confirmed the upregulation of proteins associated with the protein kinase-like endoplasmic reticulum kinase (PERK) pathway, which is one of the pathways involved in endoplasmic reticulum (ER) stress. Moreover, the administration of gallic acid resulted in verified alterations in the transmission of mitogen-activated protein kinase (MAPK) signals. Notably, an elevation in the levels of p-p38, which represents the phosphorylated state of p38 MAPK was detected. The scavenger of reactive oxygen species (ROS), N-Acetyl-L-cysteine (NAC), has shown inhibitory effects on phosphorylated p38 (p-p38), whereas the p38 inhibitor SB203580 inhibited C/EBP homologous protein (CHOP). In both instances, the levels of PARP have been successfully reinstated. In other words, the study discovered a correlation between endoplasmic reticulum stress and the p38 signaling pathway. Consequently, gallic acid induces the activation of both the p38 pathway and the ER stress pathway through the generation of ROS, ultimately resulting in apoptosis. The outcomes of this study provide compelling evidence to support the notion that gallic acid possesses considerable promise as a viable therapeutic intervention for pancreatic cancer.
Collapse
Affiliation(s)
- Jeong Woo Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea; (J.W.K.); (J.C.); (M.N.P.)
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea
| | - Jinwon Choi
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea; (J.W.K.); (J.C.); (M.N.P.)
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea
| | - Moon Nyeo Park
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea; (J.W.K.); (J.C.); (M.N.P.)
| | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea; (J.W.K.); (J.C.); (M.N.P.)
- Korean Medicine-Based Drug Repositioning Cancer Research Center, College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05253, Republic of Korea
| |
Collapse
|
11
|
Gelen V, Sengul E, Yildirim S, Cinar İ. The role of GRP78/ATF6/IRE1 and caspase-3/Bax/Bcl2 signaling pathways in the protective effects of gallic acid against cadmium-induced liver damage in rats. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:1326-1333. [PMID: 37886005 PMCID: PMC10598816 DOI: 10.22038/ijbms.2023.71343.15525] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/30/2023] [Indexed: 10/28/2023]
Abstract
Objectives Cadmium (CD) causes widespread and severe toxic effects on various tissues. Studies have shown that apoptosis, inflammation, and endoplasmic reticulum stress play a role in organ damage caused by CD. Phenolic compounds with strong antioxidant effects are found in various fruits and vegetables. One of these compounds is Gallic acid (GA), which is found both free and hydrolyzable in grapes, pomegranate, tea, hops, and oak bark. Result of various studies show that GA has active antioxidant, anti-inflammatory, and anti-apoptotic properties. In our study, we investigated the mechanism of the protective effect of GA on CD-induced hepatotoxicity in rats. Materials and Methods In this study, 50 adult male Sprague Dawley rats weighing approximately 200-250 g were used and the rats were divided into 5 groups: Control, CD, GA50+CD, GA100+CD, and GA100. The rats were treated with GA (50 and 100 mg/kg body weight), and Cd (6.5 mg/kg) was administrated to the rats for 5 consecutive days. The liver enzymes, TB levels in serum samples, oxidative stress, inflammation, ER stresses, apoptosis marker, histopathology, 8-OHDG, and caspase-3 positivity were analyzed. Results CD administration significantly increased liver enzyme levels (AST, ALT, ALP, and LDH), MDA, IL-1-β, IFN-γ, TNF-α, IL-10, IL-6, GRP78, CHOP, ATF6, p -IRE1, sXBP, Bax mRNA expression, Caspase 3, and 8-OHdG expression (P<0.05). These values were found to be significantly lower in the Control, GA100+CD, and GA100 groups compared to the CD group (P<0.05). CD administration significantly decreased the expression levels of TB, IL-4, SOD, GSH, CAT, GPX, and Bcl-2 mRNA (P<0.05). These values were found to be significantly higher in the Control, GA100+CD, and GA100 groups compared to the CD group (P<0.05). Conclusion The results of the present study indicated that GA prevented Cd-induced hepatic oxidative stress, inflammation, ER stress, apoptosis, and tissue damage in rats.
Collapse
Affiliation(s)
- Volkan Gelen
- Department of Physiology, Faculty of Veterinary Medicine, Kafkas University, Kars, Turkey
| | - Emin Sengul
- Department of Physiology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
- Department of Pharmacology, Faculty of Medicine, Atatürk University, Erzurum, Turkey
| | - Serkan Yildirim
- Department of Pathology, Faculty of Veterinary Medicine, Atatürk University, Erzurum, Turkey
| | - İrfan Cinar
- Department of Pharmacology, Faculty of Medicine, Kastamonu University, Kastamonu, Turkey
| |
Collapse
|