1
|
Okechukwu Paul-Chima U, Chinyere Nkemjika A, Melvin Nnaemeka U, Onohuean H. Harnessing plant metabolic pathways for innovative diabetes management: unlocking the therapeutic potential of medicinal plants. PLANT SIGNALING & BEHAVIOR 2025; 20:2486076. [PMID: 40191975 PMCID: PMC11980501 DOI: 10.1080/15592324.2025.2486076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/24/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
The exploration of plant signaling pathways is transforming the way diabetes is managed, providing new, multi-target strategies for controlling this complex metabolic disorder. Medicinal plants are rich in bioactive compounds like phytohormones, flavonoids and polyphenols, which regulate key pathways including oxidative stress, inflammation, insulin resistance, and gut microbiota modulation. Research is emerging on the therapeutic potential of Momordica charantia, Cinnamomum verum and Trigonella foenum-graecum, which enhance insulin secretion, sensitivity and glucose homeostasis. These plant derived compounds, resveratrol and plant based insulin mimetics, not only address metabolic dysfunction but also offer holistic treatment for long term complications such as neuropathy and retinopathy. The development of precision medicine advances the tailoring of plant based therapies to individual metabolic responses, increasing efficacy and decreasing reliance on synthetic drugs with adverse side effects. Despite challenges of standardization, regulatory barriers, and limited clinical trials, incorporating medicinal plants into national diabetes management guidelines represents a cost effective and accessible option, particularly in resource limited settings. In this review, we highlight the importance of collaborative work across disciplines and the use of technologies such as artificial intelligence to speed research and optimize patient specific applications. The therapeutic power of plant signaling pathways is harnessed to develop sustainable, inclusive, and effective diabetes management strategies.
Collapse
Affiliation(s)
| | - Anyanwu Chinyere Nkemjika
- Department of Microbiology and Immunology, Kampala International University, Ishaka-Bushenyi, Uganda
| | - Ugwu Melvin Nnaemeka
- Department of Medical Biochemistry, Faculty of Basic Medical Sciences, State University of Medical and Applied Science, Enugu, Nigeria
| | - Hope Onohuean
- Biomolecules, Metagenomics, Endocrine and Tropical Disease Research Group (BMETDREG), Kampala International University, Ishaka-Bushenyi, Uganda
- Biopharmaceutics unit, Department of Pharmacology and Toxicology, School of Pharmacy, Kampala International University, Ishaka-Bushenyi, Uganda
| |
Collapse
|
2
|
Shen S, Shen W, Wang L, Sun B, Zhang Y, Zhang Y, Jia R, Wu Y, Chen X, Cao K, Fang Y, Xia H. Berberine hydrochloride-loaded liposomes-in-hydrogel microneedles achieve the efficient treatment for psoriasis. Mater Today Bio 2025; 32:101795. [PMID: 40343170 PMCID: PMC12059721 DOI: 10.1016/j.mtbio.2025.101795] [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: 02/07/2025] [Revised: 04/13/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Psoriasis is a common immune-mediated squamous skin disease, primarily characterized by the over proliferation of keratinocytes and a significant thickening of the stratum corneum. Traditional systemic drug delivery therapies often fall short due to low drug bioavailability and significant toxic side effects. Topical medications, while capable of achieving local or systemic treatment via transdermal routes, face limitations in psoriasis patients due to the abnormal thickening of the epidermis, which reduces skin permeability and hampers drug penetration efficiency. Hydrogel microneedles, as an emerging transdermal drug delivery technology, offer significant advantages such as high permeability, ease of use, low toxicity and side effects, and controlled release. Therefore, this study developed a liposome-hydrogel microneedle delivery system for the administration of berberine hydrochloride. We successfully prepared berberine hydrochloride-loaded liposomes (Ber-LPs) with high encapsulation efficiency and good stability, and integrated them into hydrogel microneedles crosslinked with PVA and PEGDA (Ber-LPs-PEGDA&PVA MNs) through a photocuring method. These microneedles exhibit an intact structure, high mechanical strength, and effective skin penetration. In vivo studies on anti-psoriatic effects showed that, compared to the model group, Ber-LPs-PEGDA&PVA MNs significantly alleviated imiquimod-induced psoriasis-like symptoms in mice, reduced skin epidermal thickness, decreased the expression levels of inflammatory cytokines, and lowered the expression of CD31 and VEGF, demonstrating excellent therapeutic efficacy. Additionally, the microneedles exhibited good drug release properties, antioxidant capacity, and biocompatibility. The novel hydrogel microneedle drug delivery system developed in this study offers a safe and effective solution for the treatment of psoriasis, with significant potential for clinical application.
Collapse
Affiliation(s)
- Si Shen
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | | | | | - Bin Sun
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | - Yongli Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | - Yong Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | - Ruoyang Jia
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | - Yang Wu
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | - Xue Chen
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | - Keang Cao
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | - Yuqing Fang
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| | - Hongmei Xia
- College of Pharmacy, Anhui University of Chinese Medicine, No. 350, Long Zi Hu Road, Hefei, 230012, China
| |
Collapse
|
3
|
Gangwar T, Poonia N, Subudhi RN, Arora V. Therapeutic potential and underlying mechanisms of phytoconstituents: emphasizing on resveratol, curcumin, quercetin, berberine, and hesperidin in ulcerative colitis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6579-6596. [PMID: 39878817 DOI: 10.1007/s00210-025-03811-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 01/10/2025] [Indexed: 01/31/2025]
Abstract
Ulcerative colitis is a long-term inflammatory colon illness that significantly affects patients quality of life. Traditional medicines and therapies often come with challenges such as side effects, instability, unpredictability, and high costs. This has captured interest in natural products that have huge health benefits. Various natural compounds, including resveratrol, curcumin, quercetin, berberine, and hesperidin demonstrate immunomodulatory and oxido-inflammatory properties inside the gut epithelium, showing potential in managing ulcerative colitis. These compounds attenuate inflammatory mediators, NF-κB, and TLR4 signaling leading to a reduction in the production of inflammation-related cytokines, including TNF-α and IL-6. They also augment the activity of internal defense compounds, including superoxide radical dismutase enzyme and heme oxygenase-1, thereby alleviating oxidative damage. In addition, natural compounds have a profound effect on the endogenous microbiota and thus, support mucosal healing and intercellular barrier integrity. Both experimental and clinical analyses provide evidence that these bioactive compounds may help reduce clinical manifestations, induce and sustain remission, and improve the well-being of individuals suffering from ulcerative colitis. This review seeks to discuss various aspects of natural compounds in the management of ulcerative colitis, including mechanisms, therapeutic prospects, and hurdles, and hence the basis for future research and practice.
Collapse
Affiliation(s)
- Tanuj Gangwar
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| | - Neelam Poonia
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India.
| | - Rudra Narayan Subudhi
- Institute of Pharmaceutical Sciences, J.S. University, Shikohabad, Uttar Pradesh, India
| | - Vimal Arora
- University Institute of Pharma Sciences, Chandigarh University, Gharuan, Mohali, Punjab, India
| |
Collapse
|
4
|
Wang H, Peng H, Ding W, Zhang Z, Zheng Y, Chen S, Chen X, Qu Q, Liu Y, Li Y. Optimization of ultrasound enzyme-assisted trehalose lipid extraction of Coptis alkaloids and evaluation of its anti-inflammatory effects. ULTRASONICS SONOCHEMISTRY 2025; 119:107396. [PMID: 40424690 DOI: 10.1016/j.ultsonch.2025.107396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 03/23/2025] [Accepted: 05/21/2025] [Indexed: 05/29/2025]
Abstract
Coptidis Rhizoma (CR), a traditional Chinese medicine, has extensive pharmacological activity because it is rich in isoquinoline alkaloids. Nonetheless, the limited solubility of these alkaloids presents a significant challenge, hindering the full realization of CR's therapeutic potential through conventional extraction methods. To address this issue and enhance the solubility of the target compounds while optimizing extraction efficiency, this study employed an environmentally sustainable extraction technique, Ultrasound-Enzyme-Trehalose Lipid (UET), to synergistically extract five key alkaloids from CR. Based on the results of single-factor experiments, Box-Behnken design (BBD) was employed to optimize the selected model variables. The optimal extraction conditions were determined: extraction pH of 4.3, extraction temperature of 40 ℃, solid-liquid ratio of 1:27 g/mL, and ultrasonication time of 48 min. UET extracts were compared with other extracts, and it was proved that UET extracts had higher alkaloid extraction rate, comprehensive evaluation value (CEV) and lower energy consumption and CO2 emission. Then scanning electron microscopy (SEM), dynamic light scattering (DLS) and transmission electron microscopy (TEM) were used to explore the extraction mechanism of UET extraction. In addition, UET extraction has more excellent anti-inflammatory activity. The establishment of UET method of CR provides a method reference for green and efficient extraction of alkaloid components from natural drugs.
Collapse
Affiliation(s)
- Huiwen Wang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Haixin Peng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Wenwen Ding
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Zhiyun Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yadan Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Sikai Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Xueying Chen
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Qianwei Qu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; The Laboratory of Molecular Nutrition and Immunity, College of Animal Science and Technol, Northeast Agricultural University, Harbin 150030, China.
| | - Yanyan Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| | - Yanhua Li
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.
| |
Collapse
|
5
|
Zhaleh N, Sharifi M, Samari E, Halvagar MR, Zeidi SH. A comparative study of alkaloid and phenolic compounds in different organs and tissues of Berberis integerrima. PLoS One 2025; 20:e0321255. [PMID: 40388442 PMCID: PMC12087984 DOI: 10.1371/journal.pone.0321255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/04/2025] [Indexed: 05/21/2025] Open
Abstract
Berberis integerrima Bunge is a valuable plant of the Berberidaceae family used in medicine and industry. The properties of the different parts of this plant vary and depend on the distribution of its secondary metabolites, including alkaloids and phenolic compounds. Here, we aimed to evaluate and compare phenolics and alkaloids of the different organs and tissues of B. integerrima. Our results showed that the highest content of phenolic compounds was found in the fruit and leaf, while the total alkaloid level was higher in the root. In the fruit, leaf and stem, the main phenolic acids were caffeic acid, cinnamic acid and gallic acid, respectively. In contrast, the highest levels of ferulic acid, catechin, resveratrol and luteolin were detected in the root. The highest content of berberine, one of the most important alkaloids of barberry, was found in the root, especially in the bark tissue. Further experiments showed that phenolic compounds and berberine, in the aerial organs and root of B. integerrima, respectively, are likely responsible for the antioxidant capacity of these organs. Given the high berberine content of the root (6.26 mg g-1 Dry Weight), and after trying to find a simple yet effective method to extract berberine, it was found that 80% ethanol containing 2% acetic acid at 25 °C with 72 h of maceration gave the highest berberine yield. Overall, the distribution and accumulation patterns of the secondary metabolites in the different organs of B. integerrima lead to their different applications.
Collapse
Affiliation(s)
- Nastaran Zhaleh
- Department of Plant Biology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohsen Sharifi
- Department of Plant Biology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Elaheh Samari
- Department of Plant Biology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Reza Halvagar
- Department of Inorganic Chemistry, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran
| | - Seyedeh Hanieh Zeidi
- Department of Plant Biology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
6
|
Sharma S, Kaur I, Dubey N, Goswami N, Tanwar SS. Berberine can be a Potential Therapeutic Agent in Treatment of Huntington's Disease: A Proposed Mechanistic Insight. Mol Neurobiol 2025:10.1007/s12035-025-05054-6. [PMID: 40377895 DOI: 10.1007/s12035-025-05054-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025]
Abstract
Huntington's disease (HD) is a genetic neurodegenerative disorder caused by CAG repeat expansion in the HTT gene, producing mutant huntingtin (mHTT) protein. This leads to neuronal damage through protein aggregation, transcriptional dysregulation, excitotoxicity, and mitochondrial dysfunction. mHTT impairs protein clearance and alters gene expression, energy metabolism, and synaptic function. Therapeutic strategies include enhancing mHTT degradation, gene silencing via antisense oligonucleotides and RNAi, promoting neuroprotection through BDNF signaling, and modulating neurotransmitters like glutamate and dopamine. Berberine, a natural isoquinoline alkaloid, has emerged as a promising therapeutic option for HD due to its multifaceted neuroprotective properties. Research indicates that berberine can mitigate the progression of neurodegenerative diseases, including HD, by targeting various molecular pathways. It exhibits antioxidant, anti-inflammatory, and autophagy-enhancing effects, which are crucial in reducing neuronal damage and apoptosis associated with HD. These properties make berberine a potential candidate for therapeutic intervention in HD, as demonstrated in both cellular and animal models. Berberine activates the PI3K/Akt pathway, which is vital for cell survival and neuroprotection. It reduces oxidative stress and neuroinflammation, both of which are implicated in HD pathology. Berberine enhances autophagic processes, promoting the degradation of mutant huntingtin protein, a key pathological feature of HD. In transgenic HD mouse models, berberine administration has been shown to alleviate motor dysfunction and prolong survival. It effectively reduces the accumulation of mutant huntingtin in cultured cells, suggesting a direct impact on the disease's molecular underpinnings. Berberine's safety profile, established through its use in treating other conditions, supports its potential for clinical trials in HD patients. Its ability to modulate neurotransmitter levels and engage multiple signaling pathways further underscores its therapeutic promise. While berberine shows significant potential as a therapeutic agent for HD, further research is necessary to fully elucidate its mechanisms and optimize its clinical application. The current evidence in the review paper, primarily from preclinical studies, provides a strong foundation for future investigations into berberine's efficacy and safety in human HD patients.
Collapse
Affiliation(s)
- Seema Sharma
- Department of Pharmacy, Shri Vaishnav Vidyapeeth Vishwavidyalaya, Indore, M.P, India
- Research Scholar, Department of Pharmacology, SAGE University, Indore, M.P, India
| | - Inderpreet Kaur
- Department of Pharmacy, Shivalik College of Pharmacy, Nangal, Punjab, India
| | - Naina Dubey
- Department of Pharmaceutical Sciences, SAGE University, Bhopal, M.P, India
| | - Neelima Goswami
- Department of Pharmaceutics, Sagar Institute of Research Technology and Science-Pharmacy, Bhopal, M.P, India
| | - Sampat Singh Tanwar
- Department of Pharmacy, Shri Vaishnav Vidyapeeth Vishwavidyalaya, Indore, M.P, India.
| |
Collapse
|
7
|
Xu HJ, Su Y. Potential of Berberine for Rheumatoid Arthritis Prevention and Treatment. Chin J Integr Med 2025:10.1007/s11655-025-4217-y. [PMID: 40366564 DOI: 10.1007/s11655-025-4217-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/17/2025] [Indexed: 05/15/2025]
Affiliation(s)
- Hao-Jie Xu
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, 100044, China
| | - Yin Su
- Department of Rheumatology and Immunology, Peking University People's Hospital, Beijing, 100044, China.
- Department of Rheumatology and Immunology, Peking University People's Hospital, Qingdao, Shandong Province, 266111, China.
| |
Collapse
|
8
|
Zhang Y, Feng G, Zhang W, Liu X. Natural Compounds Exert Anti-Obesity Effects by Regulating Cytokines. Phytother Res 2025. [PMID: 40312999 DOI: 10.1002/ptr.8508] [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: 08/02/2024] [Revised: 01/13/2025] [Accepted: 03/29/2025] [Indexed: 05/03/2025]
Abstract
Obesity, along with its associated health risks such as hypertension, hyperlipidemia, Type 2 diabetes, stroke, metabolic syndrome, asthma, and cancer, constitutes a significant global health burden, contributing substantially to morbidity and mortality. Cytokines, a group of secreted signaling proteins, are crucial in initiating, maintaining, and resolving immune and metabolic responses. Although cytokines have unique advantages in regulating immune and metabolic functions, their therapeutic application for obesity remains limited in clinical practice. Natural compounds, known for their structural diversity and low toxicity, have become a valuable resource for drug development. Many natural compounds have shown anti-obesity effects. This review comprehensively examines the mechanisms underlying obesity, with a specific focus on the roles of cytokines, such as inflammatory cytokines, adipokines, and growth factors. Additionally, it highlights the regulatory interactions between gut microbiota and cytokines in obesity. The review critically analyzes current anti-obesity pharmacological interventions and summarizes advanced methodologies for identifying potential natural compounds. Finally, it identifies promising natural compounds that modulate cytokine activity to prevent or treat obesity and assesses their potential as complementary or alternative therapies.
Collapse
Affiliation(s)
- Yu Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Guize Feng
- School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Weidong Zhang
- School of Pharmacy, Second Military Medical University, Shanghai, China
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xia Liu
- School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
9
|
Zhao H, Chen S, Cao N, Wu W, Liu G, Gao J, Chen J, Li T, Lu D, Zeng L, Zhu H, Zhang W, Xia Q, Li T, Zhou T, Zhang X, Li A, Pan X. Berberine is a Novel Mitochondrial Calcium Uniporter Inhibitor that Disrupts MCU-EMRE Assembly. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412311. [PMID: 39921279 PMCID: PMC12061237 DOI: 10.1002/advs.202412311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/27/2025] [Indexed: 02/10/2025]
Abstract
The mitochondrial calcium uniporter (MCU) complex mediates Ca2+ entry into mitochondria, which plays a crucial role in regulating cellular energy metabolism and apoptosis. Dysregulation of MCU is implicated in various diseases, such as neurodegenerative disorders, cardiac diseases, and cancer. Despite its importance, developing specific and clinically viable MCU inhibitors is challenging. Here, Berberine, a well-established drug with a documented safety profile, is identified as a potent MCU inhibitor through a virtual screening of an FDA-approved drug library. Berberine localizes within mitochondria and directly binds to the juxtamembrane loop domain of MCU. This binding disrupts the interaction of MCU with its essential regulator, EMRE, thereby inhibiting rapid Ca2+ entry into the mitochondria. Notably, Berberine pretreatment reduces mitochondrial Ca2+ overload and mitigates ischemia/reperfusion-induced myocardial injury in mice. These findings establish Berberine as a potent MCU inhibitor, offering a safe therapeutic strategy for diseases associated with dysregulated mitochondrial calcium homeostasis.
Collapse
Affiliation(s)
- Haixin Zhao
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
- State Key Laboratory of Experimental HaematologyFifth Medical Center of Chinese PLA General HospitalBeijing100071China
| | - Siqi Chen
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
- School of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Nian Cao
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
- Department of Cardiologythe Sixth Medical CentreChinese PLA General HospitalBeijing100048China
| | - Wenjun Wu
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Guangqin Liu
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
- School of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Jun Gao
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Jiayi Chen
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Ting Li
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Dingyi Lu
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Lingmin Zeng
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Haizhen Zhu
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Weina Zhang
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Qing Xia
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Teng Li
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Tao Zhou
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
| | - Xue‐Min Zhang
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
- School of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Ai‐Ling Li
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
- School of Basic Medical SciencesFudan UniversityShanghai200032China
| | - Xin Pan
- Nanhu LaboratoryNational Center of Biomedical Analysis27 Tai‐Ping RoadBeijing100039China
- School of Basic Medical SciencesFudan UniversityShanghai200032China
| |
Collapse
|
10
|
Efati M, Sahebkar A, Tavallaei S, Alidadi S, Hosseini H, Hamidi-Alamdari D. Protective effect of Leuco-methylene blue against acetaminophen-induced liver injury: an experimental study. Drug Chem Toxicol 2025:1-13. [PMID: 40207489 DOI: 10.1080/01480545.2025.2485347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/21/2025] [Accepted: 03/22/2025] [Indexed: 04/11/2025]
Abstract
Acetaminophen is a commonly used drug for mild to moderate pain relief; however, acetaminophen toxicity due to the formation of toxic metabolites is a major cause of drug-induced liver injury. Methylene blue is an FDA-approved drug for the treatment of methemoglobinemia and has potential applications in the treatment of carbon monoxide and cyanide poisoning. Leuco-methylene blue, a colorless form of methylene blue, is more effective in entering cells and counteracting oxidative stress, making it a valuable option in regulating mitochondrial function and ATP production. In this study, we aimed to evaluate the effect of LMB on liver damage caused by acetaminophen toxicity. Thirty-six rats were divided into six groups: control, APAP, NAC, LMB, MB, and NAC+LMB. All groups except the control received acetaminophen (1500 mg/kg), followed by treatments with NAC (100 mg/kg), LMB (5 mg/kg), MB (5 mg/kg), and NAC+LMB after 3 hours. The rats were sacrificed 24 hours post-acetaminophen administration. LMB significantly reduced serum levels of liver enzymes (ALT, AST, and ALP) and increased the expression of genes involved in mitochondrial biogenesis and antioxidant defense (PGC-1, Nrf2, and Tfam). Additionally, LMB significantly increased total antioxidant capacity and glutathione reductase levels, decreased the prooxidant-antioxidant balance (PAB), and reduced the expression of inflammatory cytokines (IL-6 and TNF-α) in the liver tissue. LMB effectively reduced the severity of acetaminophen-induced liver damage through antioxidant and anti-inflammatory effects. LMB can effectively ameliorate APAP-induced toxicity in rats, with comparable efficacy to N-acetylcysteine with respect to most complications of acetaminophen-induced toxicity in rats.
Collapse
Affiliation(s)
- Majid Efati
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Shima Tavallaei
- Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Soodeh Alidadi
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Hossein Hosseini
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Daryoush Hamidi-Alamdari
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Surgical Oncology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
11
|
Zhao M, Deng D, Liu H, Guo R, Wu J, Hao Y, Yang M. Berberine Suppresses Influenza A Virus-Triggered Pyroptosis in Macrophages via Intervening in the mtROS-MAVS-NLRP3 Inflammasome Pathway. Viruses 2025; 17:539. [PMID: 40284982 PMCID: PMC12030943 DOI: 10.3390/v17040539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 04/02/2025] [Indexed: 04/29/2025] Open
Abstract
Infection with influenza A virus (IAV) may trigger excessive inflammatory responses, leading to severe viral pneumonia and accelerating disease progression. Therefore, controlling these excessive inflammatory responses is crucial for the prevention and treatment of pneumonia caused by IAV. Berberine (BBR), an isoquinoline alkaloid extracted from traditional Chinese medicine, possesses extensive pharmacological activities. However, its immunoregulatory effects and molecular mechanisms in the context of IAV infection require further investigation. This study explored the impact of BBR on macrophage pyroptosis and inflammatory responses induced by IAV infection. Our findings revealed that BBR effectively inhibits the release of IL-1β and TNF-α induced by IAV infection and suppresses gasdermin D (GSDMD)-mediated pyroptosis in a dose-dependent manner. Further research indicates that BBR alleviates macrophage pyroptosis and inflammatory responses in IAV-infected cells by reducing the release of mitochondrial reactive oxygen species (mtROS), inhibiting mitochondrial antiviral signaling protein (MAVS) expression and blocking the activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome. Experiments using siRNA to knockdown MAVS further confirmed the pivotal role of MAVS in BBR's inhibition of IAV-induced macrophage pyroptosis. This study provides a scientific basis for the application of BBR as an anti-inflammatory drug in the treatment of inflammatory diseases caused by IAV infection and directs future research endeavors.
Collapse
Affiliation(s)
| | | | | | | | | | - Yu Hao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (M.Z.); (D.D.); (H.L.); (R.G.); (J.W.)
| | - Mingrui Yang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing 102488, China; (M.Z.); (D.D.); (H.L.); (R.G.); (J.W.)
| |
Collapse
|
12
|
Lu F, Li E, Gao Y, Zhang Y, Kong L, Yang X. Dapagliflozin modulates hepatic lipid metabolism through the proprotein convertase subtilisin/kexin type 9/low density lipoprotein receptor pathway. Diabetes Obes Metab 2025; 27:2096-2109. [PMID: 39831337 DOI: 10.1111/dom.16202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/22/2025]
Abstract
BACKGROUND Proprotein convertase subtilisin/kexin type 9 (PCSK9) is mainly secreted by the liver, and plays a crucial role in lipid metabolism disorder. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) can regulate lipid metabolism through various pathways, including reducing visceral fat accumulation, modulating serum lipoprotein levels and alleviating hepatic steatosis. However, the specific regulatory mechanisms remain unclear. METHODS We built a model of glucose and lipid metabolism disorder in vivo and in vitro, and explored the regulatory mechanism of dapagliflozin in regulating liver lipid metabolism. RESULTS We found that the SGLT2i dapagliflozin significantly reduced serum levels of PCSK9, total cholesterol (TC), low density lipoprotein cholesterol (LDL-C) in high-fat diet (HFD)-fed mice, while also improving hepatic steatosis. In vitro studies confirmed that dapagliflozin increased LDL receptor (LDLR) expression in HepG2 cells, enhancing their ability to uptake LDL-C. CONCLUSIONS Further mechanistic studies revealed that the hepatocyte nuclear factor-1-alpha (HNF1α)/PCSK9/LDLR signalling pathway may be involved in dapagliflozin's regulation of lipid metabolism homeostasis.
Collapse
Affiliation(s)
- Fengyuan Lu
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - En Li
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yifeng Gao
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yan Zhang
- Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lijuan Kong
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xiaoyu Yang
- The Second Affiliated Hospital, Zhengzhou University, Zhengzhou, China
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
13
|
Wang Y, Gao P, Wu Z, Jiang B, Wang Y, He Z, Zhao B, Tian X, Gao H, Cai L, Li W. Exploring the therapeutic potential of Chinese herbs on comorbid type 2 diabetes mellitus and Parkinson's disease: A mechanistic study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119095. [PMID: 39537117 DOI: 10.1016/j.jep.2024.119095] [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: 08/21/2024] [Revised: 10/12/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Type 2 diabetes mellitus (T2DM) and Parkinson's disease (PD) are chronic conditions that affect the aging population, with increasing prevalence globally. The rising prevalence of comorbidity between these conditions, driven by demographic shifts, severely impacts the quality of life of patients, posing a significant burden on healthcare resources. Chinese herbal medicine has been used to treat T2DM and PD for millennia. Pharmacological studies have demonstrated that medicinal herbs effectively lower blood glucose levels and exert neuroprotective effects, suggesting their potential as adjunctive therapy for concurrent management of T2DM and PD. AIM OF THE STUDY To elucidate the shared mechanisms underlying T2DM and PD, particularly focusing on the potential mechanisms by which medicinal herbs (including herbal formulas, single herbs, and active compounds) may treat these diseases, to provide valuable insights for developing therapeutics targeting comorbid T2DM and PD. MATERIALS AND METHODS Studies exploring the mechanisms underlying T2DM and PD, as well as the treatment of these conditions with medicinal herbs, were extracted from several electronic databases, including PubMed, Web of Science, Google Scholar, and China National Knowledge Infrastructure (CNKI). RESULTS Numerous studies have shown that inflammation, oxidative stress, insulin resistance, impaired autophagy, gut microbiota dysbiosis, and ferroptosis are shared mechanisms underlying T2DM and PD mediated through the NLRP3 inflammasome, NF-κB, MAPK, Keap1/Nrf2/ARE, PI3K/AKT, AMPK/SIRT1, and System XC--GSH-GPX4 signaling pathways. Thirty-four medicinal herbs, including 2 herbal formulas, 4 single herbs, and 28 active compounds, have been reported to potentially exert anti-T2DM and anti-PD effects by targeting these shared mechanisms. CONCLUSIONS Traditional Chinese medicine effectively combats T2DM and PD through shared pathological mechanisms, highlighting their potential for application in treating these comorbid conditions.
Collapse
Affiliation(s)
- Yan Wang
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China; Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Pengpeng Gao
- Department of Preventive Treatment, Ningxia Integrated Chinese and Western Medicine Hospital, Yinchuan, 750004, China
| | - Zicong Wu
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Bing Jiang
- Department of Integrated Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Yanru Wang
- Gansu University Key Laboratory for Molecular Medicine & Chinese Medicine Prevention and Treatment of Major Diseases, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Zhaxicao He
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Bing Zhao
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, 730000, China
| | - Xinyun Tian
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Han Gao
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Li Cai
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| | - Wentao Li
- Encephalopathy Department, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China.
| |
Collapse
|
14
|
Zhang SL, Fu H, Ma Y, Lin Q, Xu Y, Yang Q, He P, Wei Z. A novel platinum(II) complex with a berberine derivative as a potential antitumor agent targeting G-quadruplex DNA. Org Biomol Chem 2025; 23:1112-1119. [PMID: 39654427 DOI: 10.1039/d4ob01705f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
G-quadruplexes are considered attractive targets for various human diseases, including cancer therapy, owing to their potential therapeutic applications. Understanding the interaction between ligands and G-quadruplexes is crucial for the development of novel anticancer agents. In this study, we designed a novel platinum(II) complex (Pt1), with a berberine derivative (L) serving as a bioactive ligand. The structures of both ligand L and Pt1 were fully characterized using NMR, ESI-MS, and IR. UV-visible spectroscopy, fluorescence spectroscopy, circular dichroism spectroscopy, electrostatic surface potential, frontier molecular orbital and molecular docking experiments were employed to investigate the interaction between Pt1 and G-quadruplexes. The results suggested that Pt1 interacted favorably with G-quadruplex DNA over double-stranded DNA (DS26). Among them, Pt1 interacts with the bcl-2 G-quadruplex with a binding affinity of 17.9 μM and did not induce conformational changes in the topology of the bcl-2 G-quadruplex. Moreover, we evaluated its antiproliferative activities on tumor cells (HeLa, A549 and T24), which demonstrated that Pt1 inhibited tumor cell proliferation and induced HeLa cell apoptosis. Overall, this study offers novel insights for the development of promising platinum(II) antitumor agents based on G-quadruplex structures.
Collapse
Affiliation(s)
- Shu-Lin Zhang
- College of Marine Sciences, Beibu Gulf University, Qinzhou, China.
| | - Haimei Fu
- College of Marine Sciences, Beibu Gulf University, Qinzhou, China.
| | - Yingxia Ma
- College of Marine Sciences, Beibu Gulf University, Qinzhou, China.
| | - Qifu Lin
- College of Marine Sciences, Beibu Gulf University, Qinzhou, China.
| | - Yanli Xu
- College of Marine Sciences, Beibu Gulf University, Qinzhou, China.
| | - Qiyuan Yang
- College of Chemistry and Materials, Nanning Normal University, Nanning, China.
| | - Peng He
- College of Marine Sciences, Beibu Gulf University, Qinzhou, China.
- Guangxi Key Laboratory of Marine Environmental Change and Disaster in Beibu Gulf, College of Marine Sciences, Beibu Gulf University, China.
| | - Zuzhuang Wei
- College of Marine Sciences, Beibu Gulf University, Qinzhou, China.
| |
Collapse
|
15
|
Zhuang Y, Gao D, Jiang W, Xu Y, Liu G, Hou G, Chen T, Li S, Zhang S, Liu S, Wang J, Xiao J, Li M, Wang W, Li S, Cao Z. Core microbe Bifidobacterium in the hindgut of calves improves the growth phenotype of young hosts by regulating microbial functions and host metabolism. MICROBIOME 2025; 13:13. [PMID: 39819813 PMCID: PMC11740343 DOI: 10.1186/s40168-024-02010-9] [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: 06/12/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025]
Abstract
BACKGROUND The growth and health of young ruminants are regulated by their gut microbiome, which can have lifelong consequences. Compared with subjective grouping, phenotypic clustering might be a more comprehensive approach to revealing the relationship between calf growth state and core gut microbes. However, the identification of beneficial gut bacteria and its internal mechanisms of shaping host phenotype differentiation remains unclear. RESULTS In this study, calves were divided into two clusters, cluster1 and cluster2, based on 29 phenotypic indicators using cluster analysis. Calves in cluster2 showed better growth performance, including higher body weight (BW), average daily gain (ADG), and dry matter intake (DMI), as well as better serum indicators with a high level of total superoxide dismutase (T-SOD), interleukin-6 (IL-6), and insulin-like growth factor-1 (IGF-1) compared to those in cluster1. Multi-omics was used to detect microbial features among calves in different phenotypic clusters. Distinct differences were observed between the two clustered gut microbiomes, including microbial diversity and composition. The close relationships between growth performance, blood metabolites, and microbiome were also confirmed. In cluster2, Bifidobacterium members were the dominant contributors to microbial metabolic functions with a higher abundance. Furthermore, pathways involved in carbohydrate degradation, glycolysis, and biosynthesis of propionate and proteins were active, while methane production was inhibited. In addition, the diversity and richness of hindgut resistome in cluster2 were lower than those in cluster1. The isolation and culture of Bifidobacterium strain, as well as the mice experiment, indicated that B. longum 1109 from calf feces in cluster2 could promote the growth of young hosts, enhance their blood immunity and antioxidation, and improve the development of hindgut. CONCLUSIONS In summary, cluster analysis has proved to be a feasible and reliable approach for identifying phenotypic subgroups of calves, prompting further exploration of host-microbiome interactions. Bifidobacterium as a core microbe in the hindgut of calves may play a crucial probiotic role in host phenotypic differentiation. This study enhances our comprehension of how gut core microbe shapes the host phenotype and provides new insights into the manipulation of beneficial gut colonizers to improve the growth performance and productivity of young ruminants. Video Abstract.
Collapse
Affiliation(s)
- Yimin Zhuang
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Duo Gao
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wen Jiang
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- College of Animal Science, Xinjiang Uygur Autonomous Region 830052, Xinjiang Agricultural University, Urumqi, China
| | - Yiming Xu
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- College of Animal Science, Xinjiang Uygur Autonomous Region 830052, Xinjiang Agricultural University, Urumqi, China
| | - Guanglei Liu
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Guobin Hou
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing, 102206, China
| | - Tianyu Chen
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shangru Li
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Siyuan Zhang
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- College of Animal Science, Xinjiang Uygur Autonomous Region 830052, Xinjiang Agricultural University, Urumqi, China
| | - Shuai Liu
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jingjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jianxin Xiao
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mengmeng Li
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Wei Wang
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Shengli Li
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhijun Cao
- State Key Laboratory of Animal Nutrition and Feeding, International Calf and Heifer Organization, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
16
|
Du M, Liu X, Ji X, Wang Y, Liu X, Zhao C, Jin E, Gu Y, Wang H, Zhang F. Berberine alleviates enterotoxigenic Escherichia coli-induced intestinal mucosal barrier function damage in a piglet model by modulation of the intestinal microbiome. Front Nutr 2025; 11:1494348. [PMID: 39877539 PMCID: PMC11772193 DOI: 10.3389/fnut.2024.1494348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/13/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Enterotoxic Escherichia coli (ETEC) is the main pathogen that causes diarrhea, especially in young children. This disease can lead to substantial morbidity and mortality and is a major global health concern. Managing ETEC infections is challenging owing to the increasing prevalence of antibiotic resistance. Berberine, categorized as a substance with similarities in "medicine and food," has been used in China for hundreds of years to treat gastrointestinal disorders and bacteria-induced diarrhea. This study investigated the preventive effect of dietary berberine on the intestinal mucosal barrier induced by ETEC and the microbial community within the intestines of weaned piglets. Methods Twenty-four piglets were randomly divided into four groups. Piglets were administered either a standard diet or a standard diet supplemented with berberine at concentrations of 0.05 and 0.1%. and orally administered ETEC or saline. Results Dietary supplementation with berberine reduced diamine oxidase, d-lactate, and endotoxin levels in piglets infected with ETEC (P < 0.05). Berberine increased jejunal villus height, villus/crypt ratio, mucosal thickness (P < 0.05), and goblet cell numbers in the villi and crypts (P < 0.05). Furthermore, berberine increased the optical density of mucin 2 and the mucin 2, P-glycoprotein, and CYP3A4 mRNA expression levels (P < 0.05). Berberine increased the expressions of zonula occludins-1 (ZO-1), zonula occludins-2 (ZO-2), Claudin-1, Occludin, and E-cadherin in the ileum (P < 0.05). Moreover, berberine increased the expression of BCL2, reduced intestinal epithelial cell apoptosis (P < 0.05) and decreased the expression of BAX and BAK in the duodenum and jejunum, as well as that of CASP3 and CASP9 in the duodenum and ileum (P < 0.05). Berberine decreased the expression of IL-1β, IL-6, IL-8, TNF-α, and IFN-γ (P < 0.05) and elevated total volatile fatty acids, acetic acid, propionic acid, valeric acid, and isovaleric acid concentrations (P < 0.05). Notably, berberine enhanced the abundance of beneficial bacteria including Enterococcus, Holdemanella, Weissella, Pediococcus, Muribaculum, Colidextribacter, Agathobacter, Roseburia, Clostridium, Fusicatenibacter, and Bifidobacterium. Simultaneously, the relative abundance of harmful and pathogenic bacteria, such as Prevotella, Paraprevotella, Corynebacterium, Catenisphaera, Streptococcus, Enterobacter, and Collinsella, decreased (P < 0.05). Discussion Berberine alleviated ETEC-induced intestinal mucosal barrier damage in weaned piglets models. This is associated with enhancement of the physical, chemical, and immune barrier functions of piglets by enhancing intestinal microbiota homeostasis.
Collapse
Affiliation(s)
- Min Du
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xinran Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Yue Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xiaodan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Chunfang Zhao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Youfang Gu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| | - Hongyu Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Chuzhou, China
| |
Collapse
|
17
|
Hirakawa K, Matsuura T, Nishimura Y, Mori H, Takagi S. Relaxation process of photoexcited berberine via aggregation and dissociation state-dependent intramolecular electron transfer. Photochem Photobiol Sci 2025; 24:79-87. [PMID: 39730844 DOI: 10.1007/s43630-024-00673-9] [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: 09/17/2024] [Accepted: 12/06/2024] [Indexed: 12/29/2024]
Abstract
The fluorescence quantum yield of berberine in aqueous solution is significantly smaller than those of organic solution. The time profile of fluorescence intensity of berberine was analyzed by a bi-exponential function, showing that two kinds of states of berberine exist in the solutions. The observed fluorescence lifetime of shorter lifetime species of berberine in water (0.08 ns) was markedly smaller than those of organic solvents and the relative amplitude of the shorter lifetime was dominated in the aqueous solution. Thus, this shorter lifetime can be explained by the deactivation via intramolecular electron transfer. These two states of berberine were independent of pH. The enthalpy and entropy changes between these two states were - 23.2 kJ mol-1 and - 90 J K-1 mol-1, supporting the aggregation of berberine. In the aggregation state, an electrostatic interaction between cationic berberine and chloride ion decreases the electron accepting ability of the isoquinoline moiety of berberine, resulting in the suppression of intramolecular electron transfer. Furthermore, in the presence of clay, the interaction between berberine and clay increased the fluorescence intensity of berberine and its lifetime, showing that the negative charge of clay suppresses the intramolecular electron transfer. Since the electron transfer quenching of the photo-excited berberine is advantageous for suppressing the phototoxic effect of berberine, the inhibition of berberine aggregation is an important process for the phototoxicity prevention.
Collapse
Affiliation(s)
- Kazutaka Hirakawa
- Applied Chemistry and Biochemical Engineering Course, Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Johoku 3-5-1, Chuo-Ku, Hamamatsu, Shizuoka, 432-8561, Japan.
- Department of Optoelectronics and Nanostructure Science, Graduate School of Science and Technology, Shizuoka University, Johoku 3-5-1, Chuo-Ku, Hamamatsu, Shizuoka, 432-8561, Japan.
- Cooperative Major in Medical Photonics, Shizuoka University, Johoku 3-5-1, Chuo-Ku, Hamamatsu, Shizuoka, 432-8561, Japan.
| | - Toji Matsuura
- Applied Chemistry and Biochemical Engineering Course, Department of Engineering, Graduate School of Integrated Science and Technology, Shizuoka University, Johoku 3-5-1, Chuo-Ku, Hamamatsu, Shizuoka, 432-8561, Japan
| | - Yoshinobu Nishimura
- Department of Chemistry, University of Tsukuba, Tennoudai 1-1-1, Tsukuba, Ibaraki, 305-8571, Japan
| | - Hakan Mori
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-Shi, Tokyo, 192-0397, Japan
| | - Shinsuke Takagi
- Department of Applied Chemistry for Environment, Graduate School of Urban Environmental Sciences, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-Shi, Tokyo, 192-0397, Japan
| |
Collapse
|
18
|
Chen T, Zhan X, Zhu J, Zhou C, Huang C, Wu S, Yao Y, Zhang B, Feng S, Chen J, Xue J, Yang Z, Liu C. Integrating multiomics and Single-Cell communication analysis to uncover Ankylosing spondylitis mechanisms. Int Immunopharmacol 2024; 143:113276. [PMID: 39357209 DOI: 10.1016/j.intimp.2024.113276] [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: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND Ankylosing spondylitis (AS) is a chronic inflammatory joint disorder, necessitating early diagnosis and effective treatment. The specific mechanism of action of Cassia twigs in the treatment of AS is not fully understood. METHODS Blood samples and clinical data from 28,458 individuals (6,101 with AS, 22,357 without AS) were collected. To construct a predictive model, we utilized logistic regressions and machine learning techniques to create a dynamic nomogram. Immune cell infiltration was evaluated using the GSE73754 dataset. Subsequently, we obtained vertebral bone marrow blood from AS patients for 10X single-cell sequencing. We also extracted and purified total RNA from hip joint ligament tissue samples from six AS patients and six non-AS patients. The genes related to the expression of AS and Cassia twigs were analyzed comprehensively, and the specific drug targets were identified by molecular docking. The interactions between immune cells through cell communication analysis were elucidated. RESULTS We developed a dynamic nomogram incorporating the neutrophil count (NEUT) and other variables. Neutrophil immune responses were confirmed through immune infiltration analysis utilizing GSE73754. We observed the early involvement of neutrophils in the pathology of AS. The CAT-expressing Cassia twigs gene could be used as a drug target for the treatment of AS. Moreover, comprehensive RNA analysis revealed notable CAT expression in neutrophils and various other immune cells. CONCLUSIONS Neutrophils play dual roles in AS, regulating inflammation and initiating differentiation signals to other cells. The CAT gene, which is expressed in Cassia twigs, has emerged as a potential therapeutic target for AS treatment.
Collapse
Affiliation(s)
- Tianyou Chen
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Xinli Zhan
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Jichong Zhu
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Chenxing Zhou
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Chengqian Huang
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Shaofeng Wu
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Yuanlin Yao
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Bin Zhang
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Sitan Feng
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Jiarui Chen
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Jiang Xue
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Zhenwei Yang
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| | - Chong Liu
- The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Nanning, Guangxi, 530021, P.R. China.
| |
Collapse
|
19
|
Wang Y, Chen Y, Zhang H, Yu S, Yuan G, Hu H. Colon-targeted self-assembled nanoparticles loaded with berberine double salt ameliorate ulcerative colitis by improving intestinal mucosal barrier and gut microbiota. Colloids Surf B Biointerfaces 2024; 245:114353. [PMID: 39509850 DOI: 10.1016/j.colsurfb.2024.114353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/30/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024]
Abstract
Ulcerative colitis (UC) is a chronic, recurrent inflammatory bowel disease marked by disturbances in intestinal mucosal barriers, persistent inflammation, oxidative stress, and dysbiosis of the intestinal microbiota. Traditional treatments often fail to adequately address these issues, primarily targeting inflammation. To address these limitations, this study developed an innovative approach using self-assembled nanoparticles for oral administration that target colonic inflammation. Berberine hydrochloride and ursodeoxycholic acid were combined to form a double salt (BeU), enhancing solubility and encapsulation. An amphiphilic polymer (FU-PA) was created by esterifying fucoidan with palmitic acid. FU-PA/BeU nanoparticles were prepared using the nanoprecipitation method and further encapsulated in acid-resistant sodium alginate microspheres (FU-PA/BeU NPs@MS) for targeted delivery to colonic lesions. The aggregation rate of nanoparticles with mucus was significantly reduced to 59 % of free berberine, while the apparent permeability coefficient increased by 2.4 times. In vitro, FU-PA/BeU NPs effectively targeted inflammatory macrophages, reducing IL-6 and NO levels while increasing IL-10 level (to 42.5 %, 26.8 %, and 539 % of the LPS-treated group, respectively). Additionally, the ABTS and DPPH radical scavenging capabilities of FU-PA/BeU NPs were 177.8 % and 151.7 % of BeU, respectively. In dextran sulphate sodium-induced UC mice, oral FU-PA/BeU NPs@MS significantly improved epithelial and mucosal barriers, restored gut microbiota diversity, reduced inflammation and oxidative stress. Remarkably, the mean colon length in the FU-PA/BeU NPs@MS group was 1.2 times longer than that in the sulfasalazine group. These dual-targeted FU-PA/BeU NPs@MS show great potential for UC treatment.
Collapse
Affiliation(s)
- Yalong Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Yan Chen
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Hongjuan Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shihui Yu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Gang Yuan
- Department of Geriatrics, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China.
| | - Haiyan Hu
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Chiral Molecule and Drug Discovery, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
20
|
Kamali M, Sarhadynejad Z, Tajadini H, Keikha M, Salari Z, Dehesh T, Lashkarizadeh M, Esmaili M. Evaluation of the Efficacy of Medicinal Plants in Treating Bacterial Vaginosis: A Comprehensive Systematic Review of Interventional Studies. IRANIAN JOURNAL OF NURSING AND MIDWIFERY RESEARCH 2024; 29:649-659. [PMID: 39759925 PMCID: PMC11694588 DOI: 10.4103/ijnmr.ijnmr_311_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 01/07/2025]
Abstract
Background Bacterial Vaginosis (BV) is the most prevalent cause of vaginal infection among women. This study aimed to summarize the evidence related to the effectiveness of medicinal plants as an alternative therapy for the management of BV. Materials and Methods PubMed, Scopus, Cochrane Library, Web of Science, and Medline PubMed were systematically searched. Moreover, we searched Google Scholar to explore the possible effects of herbal treatments on BV in women of childbearing age up to 2022. All randomized clinical trials investigating the effects of medicinal plants as oral or vaginal monotherapy or in combination for BV treatment in women of childbearing age were included in this systematic review. Results In total, 20 studies comprising 2685 participants were included in our review. The results show that combinations of herbal medicines such as Prangos ferulacea, Berberis vulgaris, Myrtus communis, and Quercus Brantii with metronidazole can have better results in the treatment of BV. Moreover, the main results show that some medicinal plant products alone such as Forzejehe (Tribulus terrestris + Myrtus communis + Foeniculum vulgare + Tamarindus indica), Zataria multiflora, and Calendula officinalis had therapeutic effects similar to metronidazole. Propolis and Brazilian pepper tree (Schinus) were effective in the treatment of BV, but they have less therapeutic effect than metronidazole. Conclusions To reduce the complications caused by chemical treatments and also the resistance of patients to these treatments, it seems necessary to use supportive treatments along with chemical drugs after the necessary approvals have been obtained.
Collapse
Affiliation(s)
- Mohadese Kamali
- Faculty of Persian Medicine, Herbal and Traditional Medicines Research Center, Department of Traditional Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Zarrin Sarhadynejad
- Faculty of Persian Medicine, Herbal and Traditional Medicines Research Center, Department of Traditional Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
| | - Haleh Tajadini
- Faculty of Persian Medicine, Medical Mycology and Bacteriology Research Center, Department of Traditional Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mojtaba Keikha
- Department of Biostatistics and Epidemiology, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Zohreh Salari
- School of Medicine, Obstetrics and Gynecology Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Tania Dehesh
- Department of Biostatistics and Epidemiology, Faculty of Public Health, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahdiyeh Lashkarizadeh
- School of Medicine, Department of Pathology and Stem Cell Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mitra Esmaili
- Faculty of Persian Medicine, Herbal and Traditional Medicines Research Center, Department of Traditional Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
21
|
Ali SA, Datusalia AK. Berberine attenuates ECM accumulation and the progression of acute liver failure through inhibition of NLRP3 inflammasome signalling. Toxicol Appl Pharmacol 2024; 492:117129. [PMID: 39428072 DOI: 10.1016/j.taap.2024.117129] [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: 06/19/2024] [Revised: 09/10/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Acute liver failure (ALF) is a life-threatening disease, characterized by upregulated extracellular matrix deposition and inflammatory signalling, with no effective treatment options and targets. The present study was designed to investigate the preventive and therapeutic effects of berberine (BBR) and its underlying mechanism in thioacetamide (TAA)-induced ALF. Male SD rats were administered with TAA 300 mg/kg, i.p., thrice to induce ALF and pre- or post-treated with BBR. To decipher the effects of BBR LFT markers, histopathological analysis of key fibrotic and inflammatory proteins was performed. In addition, the levels of pro-inflammatory cytokines IL-1β, IL-6, and TNF-α were assessed by ELISA. Our work showed TAA-induced ALF animals were associated with increased ALT, AST, bilirubin (LFT markers) and histopathological alterations with profuse infiltration of inflammatory cells in the liver tissue. Treatment with BBR has significantly inhibited LFT markers and histological alterations triggered by TAA. In addition, TAA animals demonstrated increased collagen accumulation and upregulated expression of TGF-β1, vimentin, and α-SMA compared to control. The excessive accumulation of collagen, TGF-β1, vimentin, and α-SMA were significantly modulated with BBR treatment. Further, the fluorescence intensity of ROS an activator of NLRP3 including the NLRP3 inflammasome, and its downstream signalling ASC, cleaved IL-1β, and other pro-inflammatory cytokines like TNF-α and IL-6 stimulated by TAA were attenuated by BBR treatment. The current work indicated that BBR significantly ameliorated TAA-induced ALF by inhibiting the extracellular matrix accumulation associated with the NLRP3/IL-1β signalling pathway and could be a viable therapeutic option to treat ALF and other fibroinflammatory diseases.
Collapse
Affiliation(s)
- Syed Afroz Ali
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Uttar Pradesh 226002, India
| | - Ashok Kumar Datusalia
- Laboratory of Molecular NeuroTherapeutics, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Uttar Pradesh 226002, India; Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Uttar Pradesh 226002, India.
| |
Collapse
|
22
|
Li Z, Lan J, Liu L, Wang Y, Chen L, Zeng R, Gu D, Hu R, Zhang T, Ding Y. Versatile Thermo-Sensitive liposomes with HSP inhibition and Anti-Inflammation for synergistic Chemo-Photothermal to inhibit breast cancer metastasis. Int J Pharm 2024; 664:124583. [PMID: 39153642 DOI: 10.1016/j.ijpharm.2024.124583] [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: 04/26/2024] [Revised: 07/13/2024] [Accepted: 08/10/2024] [Indexed: 08/19/2024]
Abstract
Photothermal therapy (PTT) is a prospective therapeutic method for breast cancer. However, excess inflammatory response induced by PTT may aggravate tumor metastasis. Meanwhile, the overexpressed heat shock proteins (HSPs) by cancer cells can protect them from hyperthermia during PTT. Therefore, to attenuate the PTT-induced inflammation and inhibit tumor metastasis, a folate receptor-targeted thermo-sensitive liposome (BI-FA-LP) co-loading Berberine (BBR) and Indocyanine green (ICG) was developed. BI-FA-LP utilized enhanced permeability and retention (EPR) effect and FA receptor-mediated endocytosis to selectively accumulate at tumor, reducing off-target toxicity during the treatment. After targeting to the tumor site, BBR and ICG were released from BI-FA-LP upon laser irradiation, and ICG showed good photothermal performance, while BBR inhibited HSP70 and HSP90 expression during PTT, exerting chemo-photothermal synergetic anti-tumor effect. Moreover, BBR could suppress the PTT induced inflammation, thus inhibiting tumor metastasis and ameliorating tissue injury. Thus, this versatile liposome provided a new strategy to enhance PTT and anti-inflammatory effects for breast cancer treatment.
Collapse
Affiliation(s)
- Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Li Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yu Wang
- Shanghai Children's Medical Center, Shanghai 200127, China
| | - Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruifeng Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Donghao Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ruolu Hu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; National Innovation Platform for medical industry-education integration, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
23
|
García-Muñoz AM, Victoria-Montesinos D, Ballester P, Cerdá B, Zafrilla P. A Descriptive Review of the Antioxidant Effects and Mechanisms of Action of Berberine and Silymarin. Molecules 2024; 29:4576. [PMID: 39407506 PMCID: PMC11478310 DOI: 10.3390/molecules29194576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Oxidative stress is a key factor in the development of chronic diseases such as type 2 diabetes, cardiovascular diseases, and liver disorders. Antioxidant therapies that target oxidative damage show significant promise in preventing and treating these conditions. Berberine, an alkaloid derived from various plants in the Berberidaceae family, enhances cellular defenses against oxidative stress through several mechanisms. It activates the AMP-activated protein kinase (AMPK) pathway, which reduces mitochondrial reactive oxygen species (ROS) production and improves energy metabolism. Furthermore, it boosts the activity of key antioxidant enzymes like superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), thus protecting cells from oxidative damage. These actions make berberine effective in managing diseases like type 2 diabetes, cardiovascular conditions, and neurodegenerative disorders. Silymarin, a flavonolignan complex derived from Silybum marianum, is particularly effective for liver protection. It activates the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway, enhancing antioxidant enzyme expression and stabilizing mitochondrial membranes. Additionally, silymarin reduces the formation of ROS by chelating metal ions, and it also diminishes inflammation. This makes it beneficial for conditions like non-alcoholic fatty liver disease (NAFLD) and alcohol-related liver disorders. This review aims to highlight the distinct mechanisms by which berberine and silymarin exert their antioxidant effects.
Collapse
Affiliation(s)
| | | | - Pura Ballester
- Faculty of Pharmacy and Nutrition, UCAM Universidad Católica de Murcia, 30107 Murcia, Spain; (A.M.G.-M.); (D.V.-M.); (B.C.); (P.Z.)
| | | | | |
Collapse
|
24
|
Wang Y, Zhang Z, Du M, Ji X, Liu X, Zhao C, Pang X, Jin E, Wen A, Li S, Zhang F. Berberine alleviates ETEC-induced intestinal inflammation and oxidative stress damage by optimizing intestinal microbial composition in a weaned piglet model. Front Immunol 2024; 15:1460127. [PMID: 39351242 PMCID: PMC11440249 DOI: 10.3389/fimmu.2024.1460127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 08/27/2024] [Indexed: 10/04/2024] Open
Abstract
Introduction Enterotoxigenic Escherichia coli (ETEC) is the main diarrhea-causing pathogen in children and young animals and has become a global health concern. Berberine is a type of "medicine and food homology" and has a long history of use in China, particularly in treating gastrointestinal disorders and bacterial diarrhea. Methods In this study, we explored the effects of berberine on growth performance, intestinal inflammation, oxidative damage, and intestinal microbiota in a weaned piglet model of ETEC infection. Twenty-four piglets were randomly divided into four groups-a control group (fed a basal diet [BD] and infused with saline), a BD+ETEC group (fed a basal diet and infused with ETEC), a LB+ETEC group (fed a basal diet with 0.05% berberine and infused with ETEC infection), and a HB+ETEC group (fed a basal diet with 0.1% berberine and infused with ETEC). Results Berberine significantly improved the final body weight (BW), average daily gain (ADG), and average daily feed intake (ADFI) (P<0.05) of piglets, and effectively decreased the incidence of diarrhea among the animals (P<0.05). Additionally, berberine significantly downregulated the expression levels of the genes encoding TNF-α, IL-1β, IL-6, IL-8, TLR4, MyD88, NF-κB, IKKα, and IKKβ in the small intestine of piglets (P<0.05). ETEC infection significantly upregulated the expression of genes coding for Nrf2, CAT, SOD1, GPX1, GST, NQO1, HO-1, GCLC, and GCLM in the small intestine of the animals (P<0.05). Berberine significantly upregulated 12 functional COG categories and 7 KEGG signaling pathways. A correlation analysis showed that berberine significantly increased the relative abundance of beneficial bacteria (Gemmiger, Pediococcus, Levilactobacillus, Clostridium, Lactiplantibacillus, Weissella, Enterococcus, Blautia, and Butyricicoccus) and decreased that of pathogenic bacteria (Prevotella, Streptococcus, Parabacteroides, Flavonifractor, Alloprevotella) known to be closely related to intestinal inflammation and oxidative stress in piglets. In conclusion, ETEC infection disrupted the intestinal microbiota in weaned piglets, upregulating the TLR4/MyD88/NF-κB and Nrf2 signaling pathways, and consequently leading to intestinal inflammation and oxidative stress-induced damage. Discussion Our data indicated that berberine can optimize intestinal microbiota balance and modulate the TLR4/MyD88/NF-κB and Nrf2 signaling pathways, thus helping to alleviate intestinal inflammation and oxidative damage caused by ETEC infection in weaned piglets.
Collapse
Affiliation(s)
- Yue Wang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Ziting Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Min Du
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Xu Ji
- Anhui Province Key Laboratory of Livestock and Poultry Product Safety Engineering, Institute of Animal Science and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, China
| | - Xiaodan Liu
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Chunfang Zhao
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Anhui Science and Technology University, Chuzhou, China
| | - Xunsheng Pang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Erhui Jin
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Anhui Science and Technology University, Chuzhou, China
| | - Aiyou Wen
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
| | - Shenghe Li
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Anhui Science and Technology University, Chuzhou, China
| | - Feng Zhang
- College of Animal Science, Anhui Science and Technology University, Chuzhou, China
- Anhui Province Key Laboratory of Animal Nutrition Regulation and Health, Anhui Science and Technology University, Chuzhou, China
| |
Collapse
|
25
|
Wang M, Qu L, Du X, Song P, Ng JPL, Wong VKW, Law BYK, Fu X. Natural Products and Derivatives Targeting Metabolic Reprogramming in Colorectal Cancer: A Comprehensive Review. Metabolites 2024; 14:490. [PMID: 39330497 PMCID: PMC11433951 DOI: 10.3390/metabo14090490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024] Open
Abstract
Metabolic reprogramming is a critical pathogenesis of colorectal cancer (CRC), referring to metabolic disorders that cancer cells make in response to the stimulating pressure. Metabolic reprogramming induces changes in genetic material and promotes CRC progression and has been proven to be an efficient target of CRC. As natural products have garnered interest due to notable pharmacological effects and potential in counteracting chemoresistance, an increasing body of research is delving into the impact of these natural products on the metabolic reprogramming associated with CRC. In this review, we collected published data from the Web of Science and PubMed, covering the period from January 1980 to October 2023. This article focuses on five central facets of metabolic alterations in cancer cells, glucose metabolism, mitochondrial oxidative phosphorylation (OXPHOS), amino acid metabolism, fatty acid synthesis, and nucleotide metabolism, to provide an overview of recent advancements in natural product interventions targeting metabolic reprogramming in CRC. Our analysis underscores the potential of natural products in disrupting the metabolic pathways of CRC, suggesting promising therapeutic targets for CRC and expanding treatment options for metabolic-associated ailments.
Collapse
Affiliation(s)
- Mengyu Wang
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Liqun Qu
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| | - Xinying Du
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| | - Peng Song
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jerome P. L. Ng
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Vincent Kam Wai Wong
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Betty Yuen Kwan Law
- Nehr’s Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China; (M.W.)
| | - Xianjun Fu
- Research Institute for Marine Traditional Chinese Medicine, Key Laboratory of Marine Traditional Chinese Medicine in Shandong Universities, Shandong Engineering and Technology Research Center on Omics of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medical Sciences Shandong University of Traditional Chinese Medicine, Qingdao Key Laboratory of Research in Marine Traditional Chinese Medicine, Qingdao Key Technology Innovation Center of Marine Traditional Chinese Medicine’s Deep Development and Industrialization, Qingdao 266114, China
| |
Collapse
|