1
|
Melato J, Goldoni FC, Benvenutti L, Corrêa TP, Remor AP, Varela KG, Stoeberl LC, Fernandes GG, de Lima Rasga G, Passos GF, Apel MA, Klein-Junior LC, Santin JR, da Costa R, Quintão NLM. Omega-3-Enriched Fish oil reduces the chemotherapy-induced peripheral neuropathy in mice. Neuropharmacology 2025; 271:110384. [PMID: 40015508 DOI: 10.1016/j.neuropharm.2025.110384] [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: 10/31/2024] [Revised: 02/21/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
Cancer is a leading cause of mortality and morbidity worldwide and conventional chemotherapy frequently induce irreversible adverse effects in patients. Chemotherapy-induced peripheral neuropathy (CIPN) is a common adverse effect of both paclitaxel (PTX) and oxaliplatin (OXA) chemotherapies, affecting approximately 30-50% of patients. As cancer survival rates have improved, the efforts of scientific community to develop new strategies for preventing CIPN are also growing. This study presents the effects of omega-3 (ω-3)-enriched fish oil supplementation on the hypersensitivity induced by PTX or OXA in mice. GC-MS analysis of the fish oil revealed an amount of EPA and DHA corresponding to 55.2% and 37.4% of total oil composition, respectively. The thirty-day supplementation with the fish oil prevented the cold hypersensitivity induced by the acute OXA injection protocol, with reduction of spinal cord microglia activation, as well as decreased levels of cytokines and BDNF in the spinal cord and brain. A similar effect was observed with the chronic OXA administration, reducing both mechanical and thermal hypersensitivity. The fish oil also prevented PTX-induced neuropathy, accompanied by a reduction in cytokine levels. It is important to mention that biochemical parameters such as total cholesterol, triglycerides and glucose were also normalised. The fish oil supplementation prevented the development of hypersensitivity in both OXA and PTX models, with reduced neuroinflammation likely being the main mechanism behind this effect. The fish oil supplementation, either before or during chemotherapy, could be an important ally to prevent and treat CIPN, improving the patients' quality of life post-cancer treatment.
Collapse
Affiliation(s)
- Jessica Melato
- Postgraduate Program in Pharmaceutical Science, School of Health Sciences, Universidade do Vale do Itajaí (UNIVALI), SC, Brazil
| | - Fernanda Capitanio Goldoni
- Postgraduate Program in Pharmaceutical Science, School of Health Sciences, Universidade do Vale do Itajaí (UNIVALI), SC, Brazil
| | - Larissa Benvenutti
- Postgraduate Program in Pharmaceutical Science, School of Health Sciences, Universidade do Vale do Itajaí (UNIVALI), SC, Brazil
| | - Thiago Patrício Corrêa
- Postgraduate Program in Pharmaceutical Science, School of Health Sciences, Universidade do Vale do Itajaí (UNIVALI), SC, Brazil
| | - Aline Pertile Remor
- Postgraduate Program in Bioscience and Health, Universidade do Oeste de Santa Catarina (UNOESC), SC, Brazil
| | - Karina Giacomini Varela
- Postgraduate Program in Bioscience and Health, Universidade do Oeste de Santa Catarina (UNOESC), SC, Brazil
| | | | | | | | | | - Miriam Anders Apel
- Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Luiz Carlos Klein-Junior
- Postgraduate Program in Pharmaceutical Science, School of Health Sciences, Universidade do Vale do Itajaí (UNIVALI), SC, Brazil
| | - José Roberto Santin
- Postgraduate Program in Pharmaceutical Science, School of Health Sciences, Universidade do Vale do Itajaí (UNIVALI), SC, Brazil
| | - Robson da Costa
- Faculty of Pharmacy, Federal University of Rio de Janeiro (UFRJ), RJ, Brazil
| | - Nara Lins Meira Quintão
- Postgraduate Program in Pharmaceutical Science, School of Health Sciences, Universidade do Vale do Itajaí (UNIVALI), SC, Brazil.
| |
Collapse
|
2
|
Zhang L, Tian Y, Zhang L, Zhang H, Yang J, Wang Y, Lu N, Guo W, Wang L. A comprehensive review on the plant sources, pharmacological activities and pharmacokinetic characteristics of Syringaresinol. Pharmacol Res 2025; 212:107572. [PMID: 39742933 DOI: 10.1016/j.phrs.2024.107572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/09/2024] [Accepted: 12/29/2024] [Indexed: 01/04/2025]
Abstract
Syringaresinol, a phytochemical constituent belonging to lignan, is formed from two sinapyl alcohol units linked via a β-β linkage, which can be found in a wide variety of cereals and medicinal plants. Medical researches revealed that Syringaresinol possesses a broad spectrum of biological activities, including anti-inflammatory, anti-oxidation, anticancer, antibacterial, antiviral, neuroprotection, and vasodilation effects. These pharmacological properties lay the foundation for its use in treating various diseases such as inflammatory diseases, neurodegenerative disorders, diabetes and its complication, skin disorders, cancer, cardiovascular, and cerebrovascular diseases. As the demand for natural therapeutics increases, Syringaresinol has garnered significant attention for its pharmacological properties. Despite the extensive literature that highlights the various biological activities of this molecule, the underlying mechanisms and the interrelationships between these activities are rarely addressed from a comprehensive perspective. Moreover, no thorough comprehensive summary and evaluation of Syringaresinol has been conducted to offer recommendations for potential future clinical trials and therapeutic applications of this bioactive compound. Thus, a comprehensive review on Syringaresinol is essential to advance scientific understanding, assess its therapeutic applications, ensure safety, and guide future research efforts. This will ultimately contribute to its potential integration into clinical practice and public health. This study aims to provide a comprehensive overview of Syringaresinol on its sources and biological activities to provide insights into its therapeutic potential, and to provide a basis for high-quality studies to determine the clinical efficacy of this compound. Additionally, we explored the pharmacokinetics, toxicology, and drug development aspects of Syringaresinol to guide future research efforts. The review also discussed the limitations of current research on Syringaresinol and put forward some new perspectives and challenges, which laid a solid foundation for further study on clinical application and new drug development of Syringaresinol in the future.
Collapse
Affiliation(s)
- Lei Zhang
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Yuqing Tian
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Lingling Zhang
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Huanyu Zhang
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Jinghua Yang
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Yi Wang
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China
| | - Na Lu
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China.
| | - Wei Guo
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China.
| | - Liang Wang
- Research Center of Traditional Chinese Medicine and Clinical Pharmacy, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan 250014, China.
| |
Collapse
|
3
|
Zheng S, Teng Y, Liu H, He J, Zhang S, Xiong H. Syringaresinol attenuates Tau phosphorylation and ameliorates cognitive dysfunction induced by sevoflurane in aged rats. J Neuropathol Exp Neurol 2024; 83:596-605. [PMID: 38622895 PMCID: PMC11187417 DOI: 10.1093/jnen/nlae026] [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] [Indexed: 04/17/2024] Open
Abstract
Cognitive dysfunction following anesthesia with agents such as sevoflurane is a significant clinical problem, particularly in elderly patients. This study aimed to explore the protective effects of the phytochemical syringaresinol (SYR) against sevoflurane-induced cognitive deficits in aged Sprague-Dawley rats and to determine the underlying mechanisms involved. We assessed the impact of SYR on sevoflurane-induced cognitive impairment, glial activation, and neuronal apoptosis through behavioral tests (Morris water maze), immunofluorescence, Western blotting for key proteins involved in apoptosis and inflammation, and enzyme-linked immunosorbent assays for interleukin-1β, tumor necrosis factor-α, and interleukin-6. SYR treatment mitigated sevoflurane-induced cognitive decline, reduced microglial and astrocyte activation (decreased Iba-1 and GFAP expression), and countered neuronal apoptosis (reduced Bax, cleaved-caspase3, and cleaved-PARP expression). SYR also enhanced Sirtuin-1 (SIRT1) expression and reduced p-Tau phosphorylation; these effects were reversed by the SIRT1 inhibitor EX527. SYR exerts neuroprotective effects on sevoflurane-induced cognitive dysfunction by modulating glial activity, apoptotic signaling, and Tau phosphorylation through the SIRT1 pathway. These findings could inform clinical strategies to safeguard cognitive function in patients undergoing anesthesia.
Collapse
Affiliation(s)
- Simin Zheng
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shannxi, China
| | - Yunpeng Teng
- Department of Anesthesia and Comfort Health Center, Xi’an International Medical Center Hospital, Xi’an, Shannxi, China
| | - Hongtao Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shannxi, China
| | - Jiaxuan He
- Department of Anesthesia and Comfort Health Center, Xi’an International Medical Center Hospital, Xi’an, Shannxi, China
| | - Shaobo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shannxi, China
| | - Hongfei Xiong
- Department of Anesthesia and Comfort Health Center, Xi’an International Medical Center Hospital, Xi’an, Shannxi, China
| |
Collapse
|
4
|
Han J, Lai H, Li W, Liao H, Xiao C, Li X, You F, Guo J. Efficacy and safety of traditional plant-based medicines for preventing chronic oxaliplatin-induced peripheral neurotoxicity in patients with colorectal cancer: A systematic review and meta-analysis with core herb contribution. JOURNAL OF ETHNOPHARMACOLOGY 2024; 326:117735. [PMID: 38211824 DOI: 10.1016/j.jep.2024.117735] [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: 07/12/2023] [Revised: 10/05/2023] [Accepted: 01/07/2024] [Indexed: 01/13/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Traditional plant-based medicines (TMs) have been widely used to prevent chronic oxaliplatin-induced peripheral neurotoxicity (OIPN). However, the prevention and safety of TMs for chronic OIPN remain ambiguous. Furthermore, diverse TM prescriptions and complicated components limit in-depth research on the mechanisms of TMs. AIM OF THIS STUDY To determine core TMs and potential pharmacological pathways on the basis of a thorough investigation into the preventive benefits and safety of oral TMs for chronic OIPN in colorectal cancer (CRC). METHODS A search of the PubMed, Cochrane, Embase, CNKI, VIP, and Wanfang databases for RCTs reporting on TMs for chronic OIPN was conducted through December 1, 2022. Subgroup analysis, sensitivity analysis and meta-regression were applied to assess the impacts of influencing variables. The assessment of Risk of Bias was relied on Cochrane Risk of Bias tool. The funnel plot, Egger's test, and the Trim and Fill method were applied to identify potential publication bias. Trial sequential analyses (TSA) were carried out by the TSA tool to increase the robustness. The assessment of the quality of evidence was according to the GRADE system. System pharmacology analysis was employed to screen core herbal combinations to elucidate possible mechanisms for preventing chronic OIPN in CRC. RESULTS The pooled effect estimate with robustness increased by TSA analysis demonstrated that oral TMs appeared to significantly decrease the incidence of chronic OIPN (RR = 0.66, 95% CI (0.56, 0.78); P<0.00001), leukocytopenia (RR = 0.65, 95% CI (0.54,0.79); P<0.00001), and nausea and vomiting (RR = 0.72, 95% CI (0.61,0.84); P<0.0001) as well as improve the Objective Response Rate (ORR) (RR = 1.31, 95% CI (1.09,1.56); P = 0.003). The incidence of severe chronic OIPN was revealed a significant reduction, particularly when chemotherapy was administered for periods of time shorter than six months (RR = 0.33, 95% CI (0.15,0.71); P = 0.005; actuation duration<3 months; RR = 0.33, 95% CI (0.17,0.62); P = 0.0007; actuation duration≥3 months, <6 months). The considerable heterogeneity among studies may be attributable to the severity of dysfunction categorized by grade and accumulated dosage. Using core TMs consisting of Astragalus membranaceus (Fisch.) Bunge, Atractylodes Macrocephala Koidz., Poria cocos (Schw.) Wolf, and Codonopsis pilosula (Franch.) Nannf. To regulate nuclear factor-kappa B against inflammation caused by activation of microglia might be an approach to preventing chronic OIPN. CONCLUSIONS TMs appear to be effective and safe in the prevention of chronic OIPN, especially severe chronic OIPN. Additionally, core TMs consisting of Astragalus membranaceus (Fisch.) Bunge, Atractylodes Macrocephala Koidz., Poria cocos (Schw.) Wolf, and Codonopsis pilosula (Franch.) Nannf were presumably responsible for reducing the incidence of chronic OIPN, and the mechanism may be related to relieving inflammation. However, quality-assured trials with long-term follow-up for exploring inflammatory factors and preliminary research on core TMs and pharmacological pathways are needed.
Collapse
Affiliation(s)
- Jierong Han
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China.
| | - Hengzhou Lai
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China.
| | - Wenyuan Li
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China; Evidence-based Traditional Chinese Medicine Center of Sichuan Province, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China.
| | - Huarui Liao
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China.
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China; Cancer Institute of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China.
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China; Tumor Teaching and Research Office of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China.
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China; Cancer Institute of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China.
| | - Jing Guo
- Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shierqiao Road, Jinniu District, Chengdu, Sichuan, 610075, China.
| |
Collapse
|
5
|
Ollodart J, Steele LR, Romero-Sandoval EA, Strowd RE, Shiozawa Y. Contributions of neuroimmune interactions to chemotherapy-induced peripheral neuropathy development and its prevention/therapy. Biochem Pharmacol 2024; 222:116070. [PMID: 38387528 PMCID: PMC10964384 DOI: 10.1016/j.bcp.2024.116070] [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: 09/06/2023] [Revised: 12/04/2023] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Chemotherapy-induced peripheral neuropathy (CIPN) is a debilitating sequela that is difficult for both clinicians and cancer patients to manage. Precise mechanisms of CIPN remain elusive and current clinically prescribed therapies for CIPN have limited efficacy. Recent studies have begun investigating the interactions between the peripheral and central nervous systems and the immune system. Understanding these neuroimmune interactions may shift the paradigm of elucidating CIPN mechanisms. Although the contribution of immune cells to CIPN pathogenesis represents a promising area of research, its fully defined mechanisms have not yet been established. Therefore, in this review, we will discuss (i) current shortcoming of CIPN treatments, (ii) the roles of neuroimmune interactions in CIPN development and (iii) potential neuroimmune interaction-targeting treatment strategies for CIPN. Interestingly, monocytes/macrophages in dorsal root ganglia; microglia and astrocytes in spinal cord; mast cells in skin; and Schwann cell near peripheral nerves have been identified as inducers of CIPN behaviors, whereas T cells have been found to contribute to CIPN resolution. Additionally, nerve-resident immune cells have been targeted as prevention and/or therapy for CIPN using traditional herbal medicines, small molecule inhibitors, and intravenous immunoglobulins in a preclinical setting. Overall, unveiling neuroimmune interactions associated with CIPN may ultimately reduce cancer mortality and improve cancer patients' quality of life.
Collapse
Affiliation(s)
- Jenna Ollodart
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | - Laiton R Steele
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | | | - Roy E Strowd
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA
| | - Yusuke Shiozawa
- Department of Cancer Biology, Wake Forest University School of Medicine, and Atrium Health Wake Forest Baptist Comprehensive Cancer, Winston-Salem, NC, USA.
| |
Collapse
|
6
|
Maia JRLCB, Machado LKA, Fernandes GG, Vitorino LC, Antônio LS, Araújo SMB, Colodeti LC, Fontes-Dantas FL, Zeidler JD, Saraiva GN, Da Poian AT, Figueiredo CP, Passos GF, da Costa R. Mitotherapy prevents peripheral neuropathy induced by oxaliplatin in mice. Neuropharmacology 2024; 245:109828. [PMID: 38158014 DOI: 10.1016/j.neuropharm.2023.109828] [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: 08/28/2023] [Revised: 12/01/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
Oxaliplatin (OXA) is an antineoplastic agent used for the treatment of cisplatin-resistant tumours, presenting lower incidence of nephrotoxicity and myelotoxicity than other platinum-based drugs. However, OXA treatment is highly associated with painful peripheral neuropathy, a well-known and relevant side effect caused by mitochondrial dysfunction. The transfer of functional exogenous mitochondria (mitotherapy) is a promising therapeutic strategy for mitochondrial diseases. We investigated the effect of mitotherapy on oxaliplatin-induced painful peripheral neuropathy (OIPN) in male mice. OIPN was induced by i.p. injections of oxaliplatin (3 mg/kg) over 5 consecutive days. Mechanical (von Frey test) and cold (acetone drop test) allodynia were evaluated between 7 and 17 days after the first OXA treatment. Mitochondria was isolated from donor mouse livers and mitochondrial oxidative phosphorylation was assessed with high resolution respirometry. After confirming that the isolated mitochondria were functional, the organelles were administered at the dose of 0.5 mg/kg of mitochondrial protein on days 1, 3 and 5. Treatment with OXA caused both mechanical and cold allodynia in mice that were significant 7 days after the initial injection of OXA and persisted for up to 17 days. Mitotherapy significantly prevented the development of both sensory alterations, and attenuated body weight loss induced by OXA. Mitotherapy also prevented spinal cord ERK1/2 activation, microgliosis and the increase in TLR4 mRNA levels. Mitotherapy prevented OIPN by inhibiting neuroinflammation and the consequent cellular overactivity in the spinal cord, presenting a potential therapeutic strategy for pain management in oncologic patients undergoing OXA treatment.
Collapse
Affiliation(s)
- João R L C B Maia
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Loreena K A Machado
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Gabriel G Fernandes
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Louise C Vitorino
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Letícia S Antônio
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Suzana Maria B Araújo
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lilian C Colodeti
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Fabrícia L Fontes-Dantas
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Departamento de Farmacologia e Psicobiologia, Instituto de Biologia Roberto Alcântara Gomes, Universidade Estadual do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Julianna D Zeidler
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Georgia N Saraiva
- Instituto de Microbiologia Paulo de Goés, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Andrea T Da Poian
- Instituto de Bioquímica Médica Leopoldo de Meis, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Claudia P Figueiredo
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Giselle F Passos
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Robson da Costa
- Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
7
|
Wang K, Zhu Y, Liu K, Zhu H, Ouyang M. Adverse events of biologic or small molecule therapies in clinical trials for inflammatory bowel disease: A systematic review and meta-analysis. Heliyon 2024; 10:e25357. [PMID: 38370239 PMCID: PMC10869791 DOI: 10.1016/j.heliyon.2024.e25357] [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: 03/19/2023] [Revised: 09/20/2023] [Accepted: 01/25/2024] [Indexed: 02/20/2024] Open
Abstract
Background Biologic or small-molecule therapies are highly effective for the treatment of inflammatory bowel disease (IBD), and approval by the FDA has significantly increased both their clinical use and the development of novel regimens. However, the identification and management of their associated toxicities poses challenges for clinicians and researchers. Methods A systematic review and meta-analysis of randomized controlled trials (RCTs) published from January 1, 2000, to October 15, 2022, and in the databases. A random-effects model with logit transformation was applied to the analysis heterogeneity between studies was evaluated using the I2 statistic with incidence and 95 % confidence interval (CI) for any adverse events (AEs), and serious AEs (SAEs). Results In Crohn's disease (CD), the total AE incidence was 67.0 % (95 % CI, 66.2%-67.8 %; I2 = 97.2 %) for any AEs and 7.3 % (6.9-7.7; 97.2) for serious AEs. In ulcerative colitis (UC), the overall incidence of any and serious AEs was 63.6 % (63.0-64.3; 98.1) and 5.7 % (5.4-6.0; 88.9), respectively. The most common AEs were infections (21.5 [20.3-22.8], 32.6 [31.0-34.2], 25.9 [24.5-27.2], and 13.7 [10.7-16.7]) in CD patients that were treated with TNF antagonists, anti-integrins, anti-IL agents, and JAK inhibitors, respectively, and in UC patients also were infections (22.8 [21.7-24.0], 27.4 [25.9-28.9], and 18.4 [16.7-20.2]), respectively, as well as increases in lactic dehydrogenase levels (23.1 [20.8-25.4]) with JAK inhibitors. Conclusion This study offers a comprehensive summary of toxic side effects of IBD treatments and a useful reference for both patients and clinicians.
Collapse
Affiliation(s)
- Kailing Wang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Youwen Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Kun Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hong Zhu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Miao Ouyang
- Department of Gastroenterology, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| |
Collapse
|
8
|
Zhang ZL, Wu ZY, Liu FY, Hang-YuChen, Zhai SD. Tetrandrine alleviates oxaliplatin-induced mechanical allodynia via modulation of inflammation-related genes. Front Mol Neurosci 2024; 17:1333842. [PMID: 38419796 PMCID: PMC10899404 DOI: 10.3389/fnmol.2024.1333842] [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: 11/06/2023] [Accepted: 01/18/2024] [Indexed: 03/02/2024] Open
Abstract
Oxaliplatin, a platinum-based chemotherapy drug, causes neuropathic pain, yet effective pharmacological treatments are lacking. Previously, we showed that tetrandrine (TET), with anti-inflammatory properties, reduces mechanical allodynia in nerve-injured mice. This study explores the effect of TET on oxaliplatin-induced mechanical allodynia and gene changes in mice. Male C57BL/6J mice received oxaliplatin intraperitoneally to induce mechanical allodynia. Post-treatment with TET or vehicle, the mechanical withdrawal threshold (WMT) was assessed using von Frey filaments. TET alleviated oxaliplatin-induced mechanical allodynia. RNA sequencing identified 365 differentially expressed genes (DEGs) in the Control vs. Oxaliplatin group and 229 DEGs in the Oxaliplatin vs. TET group. Pearson correlation analysis of co-regulated DEGs and inflammation-related genes (IRGs) revealed 104 co-regulated inflammation-related genes (Co-IRGs) (|cor| > 0.8, P < 0.01). The top 30 genes in the PPI network were identified. Arg2, Cxcl12, H2-Q6, Kdr, and Nfkbia were highlighted based on ROC analysis. Subsequently, Arg2, Cxcl12, Kdr, and Nfkbia were further verified by qRCR. Immune infiltration analysis indicated increased follicular CD4 T cell infiltration in oxaliplatin-treated mice, reduced by TET. Molecular docking showed strong binding affinity between TET and proteins encoded by Arg2, Cxcl12, Kdr, and Nfkbia. In summary, TET may alleviate oxaliplatin-induced peripheral neuropathy in clinical conditions.
Collapse
Affiliation(s)
- Zhi-Ling Zhang
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - Zi-Yang Wu
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - Feng-Yu Liu
- Key Laboratory for Neuroscience, Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Ministry of Education/National Health Commission, Peking University, Beijing, China
| | - Hang-YuChen
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| | - Suo-Di Zhai
- Department of Pharmacy, Peking University Third Hospital, Beijing, China
| |
Collapse
|
9
|
Qing X, Dou R, Wang P, Zhou M, Cao C, Zhang H, Qiu G, Yang Z, Zhang J, Liu H, Zhu S, Liu X. Ropivacaine-loaded hydrogels for prolonged relief of chemotherapy-induced peripheral neuropathic pain and potentiated chemotherapy. J Nanobiotechnology 2023; 21:462. [PMID: 38041074 PMCID: PMC10693114 DOI: 10.1186/s12951-023-02230-5] [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: 07/20/2023] [Accepted: 11/20/2023] [Indexed: 12/03/2023] Open
Abstract
Chemotherapy can cause severe pain for patients, but there are currently no satisfactory methods of pain relief. Enhancing the efficacy of chemotherapy to reduce the side effects of high-dose chemotherapeutic drugs remains a major challenge. Moreover, the treatment of chemotherapy-induced peripheral neuropathic pain (CIPNP) is separate from chemotherapy in the clinical setting, causing inconvenience to cancer patients. In view of the many obstacles mentioned above, we developed a strategy to incorporate local anesthetic (LA) into a cisplatin-loaded PF127 hydrogel for painless potentiated chemotherapy. We found that multiple administrations of cisplatin-loaded PF127 hydrogels (PFC) evoked severe CIPNP, which correlated with increased pERK-positive neurons in the dorsal root ganglion (DRG). However, incorporating ropivacaine into the PFC relieved PFC-induced CIPNP for more than ten hours and decreased the number of pERK-positive neurons in the DRG. Moreover, incorporating ropivacaine into the PFC for chemotherapy is found to upregulate major histocompatibility complex class I (MHC-I) expression in tumor cells and promote the infiltration of cytotoxic T lymphocytes (CD8+ T cells) in tumors, thereby potentiating chemotherapy efficacy. This study proposes that LA can be used as an immunemodulator to enhance the effectiveness of chemotherapy, providing new ideas for painless cancer treatment.
Collapse
Affiliation(s)
- Xin Qing
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Renbin Dou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Peng Wang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Mengni Zhou
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China
| | - Chenchen Cao
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Huiwen Zhang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Gaolin Qiu
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Zhilai Yang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China
| | - Jiqian Zhang
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| | - Hu Liu
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| | - Shasha Zhu
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, 230032, China.
| | - Xuesheng Liu
- Key Laboratory of Anesthesia and Perioperative Medicine of Anhui Higher Education Institutes, Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
10
|
Zhang S, Yang L, Hu D, He S, Cui L, Zhao J, Zhuo Y, Zhang L, Wang X. Syringaresinol alleviates IgG immune complex induced acute lung injury via activating PPARγ and suppressing pyroptosis. Int Immunopharmacol 2023; 124:111071. [PMID: 37857123 DOI: 10.1016/j.intimp.2023.111071] [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: 08/05/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/21/2023]
Abstract
Acute lung injury (ALI) is a life-threatening condition characterized by severe lung inflammation and tissue damage. In this study, we investigate the potential therapeutic efficacy of (+)-Syringaresinol (SYG), a natural compound known for its antioxidant and anti-inflammatory properties, in alleviating ALI induced by IgG immune complexes (IgG-IC). Using MH-S cells as a model, we explore SYG's ability to target peroxisome proliferator-activated receptor gamma (PPARγ) and its anti-inflammatory properties. Our comprehensive investigation aims to elucidate the specific molecular mechanisms underlying SYG's effects against pyroptosis, as revealed through transcriptomic analysis. Validation in C57BL/6 mice provides in vivo support. Our findings indicate that SYG effectively mitigates IgG-IC-induced lung damage, as evidenced by a significant reduction in lung inflammation and tissue injury. SYG treatment notably decreases pro-inflammatory cytokine levels (TNF-α, IL-6, IL-1β) in both lung tissue and cells. Molecular docking analysis reveals SYG's robust binding to PPARγ, leading to the inhibition of IgG-IC-induced inflammatory signaling pathways. Additionally, transcriptomic analysis unveils SYG's potential in suppressing macrophage pyroptosis, potentially through the downregulation of key inflammatory mediators (NLRP3, GSDMD, Caspase-1). In summary, our study presents compelling evidence supporting SYG as an effective therapeutic agent for ALI. SYG's activation of PPARγ contributes to the suppression of NF-κB and C/EBPs expression, thereby mitigating inflammation. Moreover, SYG demonstrates the ability to inhibit macrophage pyroptosis by targeting the NLRP3/GSDMD/caspase-1 axis.
Collapse
Affiliation(s)
- Sijia Zhang
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China
| | - Dongsheng Hu
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Siqi He
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Lingzhi Cui
- Graduate School, Tianjin Medical University, Tianjin, China
| | - Jiuling Zhao
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China
| | - Yuzhen Zhuo
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China.
| | - Lanqiu Zhang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China.
| | - Ximo Wang
- Graduate School, Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Tianjin Integrated Traditional Chinese and Western Medicine Hospital, Tianjin Nankai Hospital, Tianjin University, Tianjin, China.
| |
Collapse
|