This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
1. Introduction
Sepsis is a life-threatening clinical syndrome characterized by multiple organ dysfunction caused by the body’s dysregulated response to infection. Despite better supportive therapy and care, sepsis and sepsis shock have led to a high morbidity and mortality rate in recent years [1]. Just as intricated immune or inflammatory factors drive multiple metabolic diseases [2, 3], the exact mechanisms responsible for sepsis are also complex, involving coagulation abnormalities, uncontrolled release of inflammatory mediators, excessive innate immune response, endothelial capillary leakage syndrome, and organ dysfunction [4]. Though progress has been made regarding recognizing and managing clinical sepsis, incidence and mortality rates remain high. For example, in 2017, an estimated 48.9 million incident cases of sepsis and 11.0 million sepsis-related deaths were reported, representing 19.7% of all global deaths [5, 6]. Furthermore, clinical trials of therapeutics have failed to obtain promising results, and it likely got even worse during the coronavirus disease 2019 (COVID-19) pandemic due to the limited intensive care unit resources [7]. Given the lack of effective treatment for sepsis, Chinese herbal medicines, i.e., Chinese patent medicines, Chinese herbal prescriptions, and single Chinese herbs, have therapeutic actions promising in treating sepsis through multicomponent, multipathway, and multitargeting abilities [8].
Berberine (BBR), an isoquinoline alkaloid, is widely distributed in many medicinal plants, such as Coptis chinensis and Berberis vulgaris, and is usually regarded as an ingredient of Chinese herbal medicines [9]. It has a molecular weight of 336.37 Da and can be obtained through de novo synthesis. Though BBR was approved to be an antibacterial agent, it also plays a crucial therapeutic role in infectious diseases, cardiovascular diseases, neurodegenerative diseases, and rheumatoid arthritis due to its antimicrobial, antioxidant, anti-inflammatory, and metabolic regulation effects [10, 11]. BBR can act on a diverse range of molecular targets by binding to the active cavities with specific structural and physiochemical properties. Among the related conditions, sepsis is characterized by the host’s excessive and complex inflammatory response and is linked with bacterial and viral infections during its occurrence and progression. In recent years, though no studies about BBR for treating sepsis in clinical practice, its protective effect and mechanism on sepsis have been explored extensively in cellular and animal models [12–15]. Whether BBR could be a potential therapeutic drug for sepsis and septic complications remains unclear in the real world. Given that dysregulated host systemic inflammatory and immune response during sepsis usually lead to life-threatening organ dysfunction, this paper summarizes the protective role of BBR for organ injury and related mechanism and, finally, prospects the potential BBR-based therapeutic strategies in sepsis.
2. Effects and Mechanisms of BBR in Sepsis-Related Organ Dysfunction
2.1. BBR in Sepsis-Related Intestinal Barrier Injury
The intestinal barrier and the subsequent translocation of intestinal bacteria play a critical role in multiple organ dysfunction syndromes during sepsis [16]. Lipopolysaccharide (LPS) produced by intestinal bacteria might damage gut-vascular integrity, increase permeability, and reduce tight junction and adherens junction protein production [17]. Several studies have shown that pretreatment with BBR protects against intestinal injury in sepsis-challenged animals with various mechanisms. For example, in male Sprague-Dawley (SD) rats undergoing cecal ligation and puncture (CLP), TNF-α and IL-6 were significantly lower, while the tight junction protein level, the percentage of cell death in intestinal epithelial cells, and the mucosal permeability were significantly elevated after treatment by BBR [12]. The overall effects were consistent with other studies [18, 19]. Further investigation showed that the mRNA level of intestinal Toll-like receptors 2 (TLR2) and TLR4 was also increased [18, 20], indicating that BBR might protect the intestinal mucosal barrier in the early phase of sepsis through the TLR-NF-κB signaling pathway [20]. Moreover, BBR protected the damaged gut-vascular barrier (GVB) via modulation of the hepatic apolipoprotein M/S1P pathway in the polymicrobial sepsis model [21] and attenuated the impairment of glutamine transport and glutaminase activity in rats [19]. BBR also inhibited inducible cyclooxygenase-2 (COX-2) overexpression in rat small intestinal mucosa via activation of the peroxisome proliferator-activated receptor-gamma (PPARγ) pathway and inhibited the effects of LPS in a rat model of endotoxemia [13]. However, although NOD-like receptors (NLR) play a crucial role in host defense against intestinal infection, there is no evidence that the NLR pathway was related to the preventive effect of BBR for sepsis [12]. In contrast, Wnt/beta-catenin signaling pathway was related to the protective effect of BBR on the gut-vascular barrier during sepsis [17].
Zinc deficiency is prevalent in the gut and might drive bacterial translocation and toxin dissemination, while zinc redistribution could protect the intestinal barrier in sepsis [22–24]. It was reported that BBR could regulate the trace elements of zinc’s metabolism and exert intestinal protection role. He et al. demonstrated that BBR (50 mg/kg/day, gavage) improved survival rates of septic rats in the rat CLP sepsis model (BBR vs. CLP group: 80% vs. 46.7% at 48 hours) and reduced plasma endotoxin levels and intestinal mucosal permeability of septic rats, which were accompanied by the increased zinc levels [25]. In cellular studies, BBR treatment increased intracellular zinc transporter Zrt-Irt-like protein 14 (ZIP14) mRNA and protein expression in LPS-treated human colorectal adenocarcinoma cell line Caco-2 cells and upregulated LPS-decreased claudin-1 and occludin generation [25]. From the above evidence, we could conclude that BBR has a definite protective effect on intestinal barrier dysfunction, and this aspect might be the first breakthrough of BBR in treating organ injury in sepsis.
2.2. BBR in Sepsis-Related Lung Injury
Lung injury and acute respiratory distress syndrome (ARDS) are common challenges in sepsis. Microbial infections are usually responsible for pneumonia or sepsis, which cause altered respiratory function, pulmonary edema, and accumulation of inflammatory cells, including polymorphonuclear neutrophils (PMNs), circulating monocytes, and tissue-resident macrophages. The protective role of BBR for lung injury in sepsis was mainly derived from its anti-inflammatory effect. BBR was endowed with pronounced anti-inflammatory property, which was probably associated with suppressing the activation of the NF-κB signaling pathway and the subsequent gene expressions and productions of proinflammatory mediators [26, 27], as well as suppressing the phosphorylation of MAPKs, such as p38, ERK, and JNK, and the level of reactive oxygen species in macrophages [28]. Berberine pretreatment maintained the integrity of endothelial glycocalyx, inhibited NF-κB signaling pathway activation, and decreased the production of proinflammatory cytokines TNF-α, IL-1β, and IL-6 in mice with LPS-induced ARDS [29]. Pretreatment with neutral sulfate berberine attenuates lung and small intestine tissue injury and improves survival in endotoxemic mice, which may be mediated, at least in part, by the inhibition of proinflammatory mediators TNF-α and IFN-γ and NO production and upregulation of IL-10 release [1]. Wang et al. also reported that BBR inhibited LPS or CpG-ODN-induced proinflammatory cytokine expression at 12 and 24 h post BBR treatment in bone marrow-derived macrophages (BMDM) and LPS/D-galactosamine-challenged septic mouse model in vivo. They demonstrated that BBR significantly attenuated lung tissue injury and potently increased the survival rate in the septic mice with negligible side effects by suppressing NF-κB- and IL-6-mediated STAT3 activation [30]. Additionally, BBR could protect the permeability of pulmonary microvascular endothelial cells (HPMECs) and decrease the expression of IL-1β and IL-18 by suppressing the expression of phosphorylated NF-κB and further restraining the downstream gene nucleotide-binding domain and leucine-rich repeat protein-3 (Nlrp3) [31]. This limited evidence hints that the anti-inflammatory roles might be the core mechanisms for BBR in treating sepsis-related lung injury.
2.3. BBR in Sepsis-Related Acute Kidney Injury
Septic acute kidney injury (S-AKI), which is characterized by a rapid deterioration of renal function, accounts for about half of all patients with AKI syndrome treated in the intensive care unit (ICU) [32]. The mechanisms of sepsis-associated acute kidney injury involve ischemia-reperfusion, inflammation, oxidative stress, tubular cell damage, dysregulated microcirculation, morphofunctional alterations in the mitochondria, and apoptosis. The pathogen-associated molecular patterns (PAMP) and damage-associated molecular patterns (DAMP) participate in the pathological process [33, 34]. Sepsis-causing pathogens include bacteria, fungi, and viruses, and the widely used models of S-AKI in basic research were usually induced by bacteria or isolated bacterial pathogenic substances [33]. As an alkaloid with well antibacterial activity and even an alternative to antibiotics, few studies reported the application of BBR in S-AKI. Al-Kuraishy et al. revealed that BBR has no significant effect on AKI biomarkers except on serum kidney injury molecules (KIM-1) in diclofenac-induced AKI, whereas combined use of BBR and pentoxifylline significantly reduced renal biomarkers such as blood urea, serum creatinine, and glomerular filtration rate (eGFR) [35]. Berberine is the main bioactive component in Rhizoma Coptidis [36]. Integrating metabolomics and network pharmacology showed that Rhizoma Coptidis extracts increased the nuclear translocation of nuclear factor-erythroid 2-related factor-2 (Nrf2), the protein expression of heme oxygenase-1 (HO-1), and the mRNA expression of PPARα and reduced nitric oxide synthase 2 (NOS2) activity [37]. Of note, BBR also exerts renoprotective effects caused by other inducers such as cisplatin and gentamicin in rodent models through upregulating mitophagy or its antioxidant, anti-inflammatory, and antiapoptotic properties [38–40].
2.4. BBR in Sepsis-Related Liver Injury
Disturbed liver function and drastic changes in hepatic gene and protein expression during sepsis induce inflammatory pathway activation and downregulate house-keeping functions such as metabolic, biotransformation, or bile transport activities [41]. During systemic infections, the liver regulates immune defenses via bacterial clearance, production of acute-phase proteins (APPs) and cytokines, and metabolic adaptation to inflammation. The release of APPs with various biological functions in the systemic circulation contributes to the systemic activation of immune responses, while mitochondrial and endoplasmic reticulum (ER) dysfunctions during the acute phase response elicited by systemic inflammation lead to liver failure in sepsis [3, 41, 42]. To date, the molecular mechanism studies for hepatoprotection of BBR in sepsis were insufficient. Limited investigations revealed that BBR inhibits LPS-induced oxidative stress markers NO protein and iNOS expression in RAW 264.7 and THP-1 macrophage cells [14] and further alleviates ALT, AST, malondialdehyde (MDA), and myeloperoxidase (MPO) activity in sepsis rat model [15]. Moreover, BBR reduced proapoptotic protein cleaved caspase-3 and increased antiapoptotic protein Bcl-2 in CLP-induced liver injury, which were associated with a reduction in intestinal permeability and CLP-induced distant organ cell apoptosis as compared with mice on vehicle treatment [43]. Among the numerous mediators in sepsis, the high mobility group box 1 (HMGB1) is an endogenous DAMP that mediates downstream effects within the inflammatory cascade. It was reported that AMPK activation could significantly inhibit LPS-induced HMGB1 release in RAW264.7 cells [44]. A berberine derivative, 13-ethylberberine (13-EBR), could reduce HMGB1 release by activating AMPK under septic conditions and protect endotoxemic mice from liver damage [45]. Similarly, other researchers also have verified the inhibition of BBR-loaded nanostructured lipid carriers for HMGB1/TLR4/NF-κB signaling and protective roles for warm hepatic ischemia/reperfusion-induced lesion [46]. Therefore, based on this limited evidence, inhibiting the key inflammatory mediators and pathways is still a dominant mechanism of BBR in treating sepsis-related liver injury.
2.5. BBR in Sepsis-Induced Disseminated Intravascular Coagulation
Disseminated intravascular coagulation (DIC) was recognized as a deadly complication in sepsis. In addition to the activation of coagulation induced by pathogens, damage-associated molecular patterns, neutrophil extracellular traps, extracellular vesicles, and glycocalyx damage are also involved in the pathogenesis of sepsis-induced DIC. Tissue factor (TF) plays a crucial role in the coagulation of sepsis [47]. It was reported that BBR could inhibit LPS-induced TF activity and expression and downregulate NF-κB, Akt, and MAPK/JNK/p38/ERK pathways in THP-1 cells, which provides some new insights into its mechanism for sepsis treatment [48]. Yang et al.’s study showed that caspase-11, as a cytosolic LPS receptor, could enhance the activation of tissue factor and subsequent phosphatidylserine exposure independent of cell death, while deletion of caspase-11 or neutralization of phosphatidylserine or TF prevented LPS-induced DIC [49]. Berberine inhibits caspase-11-dependent coagulation in bacterial sepsis by blocking Msr1, thus preventing coagulation syndrome [50]. Mechanistically, Wang et al. first reported the possible targets and mechanisms of BBR and its primary metabolite berberrubine in inhibiting platelet activation. That is, BBR significantly inhibited ADP-induced integrin αIIbβ3 activation, reduced the level of P-selectin on the platelet membrane, and suppressed the binding of fibrinogen to the platelets, which was derived from the inhibition roles of BBR for the PI3K/Akt pathway, Rasa3 membrane translocation, and Rap1 activation [51]. These existing studies open a promising direction for BBR in treating sepsis-induced disseminated intravascular coagulation.
3. Berberine-Based Therapeutic Strategies in Sepsis
Although many studies have reported the protective effects and multiple mechanisms of BBR in sepsis-related animal or cell models (Table 1), unfortunately, there is no clinical practice at present. We speculated that to treat this kind of systemic severe inflammatory disease, it still needs to improve the drug concentration of BBR in blood, discover compounds with better efficacy based on the structure of BBR, and explore drug combination strategies in the future. Definitely, the poor oral bioavailability of BBR limited its therapeutic potential, and fortunately, BBR solid lipid nanoparticles (BBR-SLNs) were previously developed and showed significant improvement in bioavailability and reduction in systemic adverse reactions [52]. In recent years, BBR-SLNs have been applied for the management of ulcerative colitis [53, 54], bacterial infection [55], and infections of traumatic wounds [56], which are essential sepsis triggers. The effect of immunomodulatory activity and inhibition for the expression of multiple inflammatory factors were demonstrated, suggesting that BBR-SLNs might be developed to enhance the therapeutic effects of BBR as an antisepsis drug. Besides, several derivatives of BBR were reported to have better pharmaceutical effects. For example, the natural oxoderivative oxyberberine (OBB) was endowed with more pronounced anti-inflammatory properties than BBR, which was likely to be associated with suppressing the activation of the NF-κB, the subsequent gene expressions and productions of proinflammatory mediators [26]. Tetrahydroberberrubine (THBru), a berberine derivative, significantly decreased TNF-α and nitrate/nitrite content in the plasma, reduced the MPO activity in LPS-induced acute lung injury, and suppressed the activation of the MAPKs, AKT, and the NF-κB subunit p65 in LPS-induced THP-1 cells [57]. Similarly, another berberine derivative, 13-ethylberberine (13-EBR), reduces HMGB1 release through AMPK activation in LPS-activated RAW264.7 cells and protects endotoxemic mice from lung and liver damage [45]. Wang et al. considered that BBR might also serve as a promising adjuvant that could be used in conjunction with other medications for the treatment of septic patients [30]. For example, combinatorial liposomes of BBR and curcumin could inhibit biofilm formation and intracellular methicillin-resistant Staphylococcus aureus infections and associated inflammation, with five times more efficient than clindamycin [58]. The combined use of sodium new houttuyfonate and berberine chloride presented synergistic antibacterial effects against growing and persistent methicillin-resistant and vancomycin-intermediate Staphylococcus aureus, which is likely related to the interruption of the cell membrane [59]. These combined antibacterial effects of BBR with other agents predicted their potential application in sepsis. Taken together, the strategies for improving the bioavailability, discovering more powerful and effective berberine derivatives, and drug combinations might contribute to the application of BBR in treating sepsis in the future.
Table 1
The protective effects and mechanisms of berberine in sepsis-related animal or cell model.
In vivo/vitro | Animal or cell model | Therapeutic dose | Effects and mechanisms | Reference |
In vivo | Cecal slurry-induced neonatal sepsis in C57BL/6J mice (5-7 days) | 50/100 μg/mouse, i.p. | ↑Survival rate; ↓intestinal injury; ↓IL-6, IL-1β, and TNF-α; ↑miR-132-3p; ↓FOXA1, p-IκBα, and p65; and ↑IκBα | [60] |
In vivo | Mouse model of E. coli sepsis | 5 mg/kg, i.p. | ↑Survival rate; ↓IL-6, TNF-α, and CD4+ and CD8+ lymphocyte population in septic mice | [61] |
In vivo | LPS/D-gal-induced C57BL/6 septic mouse model (6-8 weeks) | 200 μg/mouse, i.v. | ↓Lung tissue injury; ↑survival rate; ↓IL-6, TNF-α, and IL-1β; (-) B cells%, T cells%, and subtypes of T cells in the spleen; and (-) CD11b + Gr-1+ cells in bone marrow cells | [30] |
In vivo | LPS-induced male BALB/c mice (6-8 weeks) | 50 mg/kg, i.g. | ↓Ileum injury (injury score, intestinal villus height, gut mucosal weight, and intestinal permeability); ↓enterocyte apoptosis; and ↓TLR4 mRNA, p-IκBα, MIP-2 production, and MPO activity | [18] |
In vivo | CLP-induced male Wistar rats (260-300 g) | 25/50/100 mg/kg/d, i.g. | ↓Sepsis-related mortality; ↑hepatic and plasma ApoM level; ↓hyperglycemia, TNF-α, and IL-1β; and ↓sepsis-induced gluconeogenesis expression | [62] |
In vivo | CLP-induced male Long-Evans rats (270-300 g) | 25/50 mg/kg/d, i.p. | ↓Microvascular permeability, serum endotoxemia levels | [17] |
In vivo | CLP-induced male Wistar rats (270-310 g) | 50 mg/kg/d, i.g. | ↑Survival rates; ↓endotoxin levels and intestinal mucosal permeability; and ↑Zn2+ in intestinal mucosa, ZIP14 expression | [25] |
In vivo | CLP-induced male SD rats (200-230 g) | 50 mg/kg, i.g. | ↓TNF-α, IL-6; ↓claudin-4, mucosal permeability; and ↓intestinal epithelial cell mortality | [12] |
In vivo | CLP-induced SD rat (200-230 g) | 50 mg/kg, i.g. | ↓Intestinal epithelial cell death; ↓TLR2/4, TNF-α, and IL-6; and ↑zonula occludens-1 | [20] |
In vivo | Outer membrane vesicles (used for delivering LPS) challenged mice (25-30 g) | 5 mg/kg | ↓Vessel occlusion and fibrin deposition in liver microvasculature; ↓DIC markers (TAT and PAI-1); ↓multiorgan dysfunction and death rate; and ↓caspase-11-mediated coagulation activation | [50] |
In vivo | LPS-induced male C57BL/6 mice (8 weeks) | 10 mg/kg, injected into peritoneal cavity | ↓Lung damage and injury scores; ↓injury of pulmonary permeability; ↓ROS level, IL-6/-18/-1β, and TNF-α; ↑IL-10; and ↓p-NF-κB, Nlrp3 | [31] |
In vitro | HepG2 cells | 5/10/20 μM | ↓TLR4 mRNA | [62] |
In vitro | LPS or CpG-ODN-induced BMDM | 5 μM | ↓IL-6, TNF-α, and IL-1β; ↓p-IκBα/β, p-IκBα, and NF-κB nuclear translocation | [30] |
In vitro | LPS-induced RIMECs | 10/20 μM | ↓RIMEC permeability; ↑transendothelial electrical resistance; and ↑β-catenin, claudin-12, and VE-cadherin | [17] |
In vitro | LPS-induced Caco-2 cells | 20 μM | ↑Intracellular zinc content; ↑ZIP14, claudin-1, and occludin expression | [25] |
In vitro | LPS-stimulated HPMECs | 2.5 μM | ↓ROS level, IL-6/-18/-1β, and TNF-α; ↑IL-10; and ↓Nlrp3 | [31] |
In vitro | E. coli-stimulated primary macrophages and THP-1 cells | 5 μM | ↓Cytotoxicity; ↓pyroptosis; and ↓caspase-11 pathway | [50] |
In vitro | LPS-induced HUVECs | 1.25/2.5/5 μM | ↓Endothelial glycocalyx degradation; ↓endothelial glycocalyx damage factors (ROS, heparanase, and MMP9) | [29] |
ApoM: apolipoprotein M; BMDM: bone marrow-derived macrophages; CLP: ligation and puncture; D-gal: D-galactosamine; HUVECs: human umbilical endothelial cells; HPMECs: human pulmonary microvascular endothelial cells; i.g.: intragastric gavage; i.p.: intraperitoneal injection; i.v.: intravenous injection; RIMECs: rat intestinal microvascular endothelial cells; ROS: reactive oxygen species.
4. Conclusion and Perspective
BBR, as a natural modulator of inflammatory signaling in multiple immune and infectious diseases, could also effectively protect the various organs in sepsis from severe damage and increase the survival rate in rodent models. Currently, BBR could be considered to act as a multitarget drug to antagonize sepsis. The primary mechanism mainly derives from the regulation for inflammatory signaling pathways (i.e., NF-κB, JAK/STAT, Akt, and MAPK), the key inflammatory mediators (i.e., HMGB1 and tissue factor), the subsequent anti-/proinflammatory cytokines (i.e., TNF-α, IL-6, and IL-10), and the dysregulated endoplasmic reticulum/oxidative stress and apoptosis (Figure 1). BBR exhibited potential therapeutic effects for sepsis-related intestinal barrier injury, lung injury, acute kidney injury, liver injury, and disseminated intravascular coagulation. Given the exact antibacterial effect of BBR and a large number of experimental reports in intestinal regulation, it is expected to be first used to protect against sepsis-induced intestinal injury in the clinic. Also, owing to its poor oral bioavailability and current unsatisfied therapeutic effects, as well as lack of practical application of BBR in sepsis, the nanostructured lipid carrier of BBR, more effective BBR derivatives, and drug combination strategies need to be investigated in various well-designed clinical trials in the future.
[figure(s) omitted; refer to PDF]
Acknowledgments
This study was supported by the Capital’s Funds for Health Improvement and Research (2020-2-2027), Funding Support for Key Clinical Projects in Beijing (WJQ), and Youyi Zhongzi Plan (YYZZ202043).
[1] Y. Y. Zhang, B. T. Ning, "Signaling pathways and intervention therapies in sepsis," Signal Transduction and Targeted Therapy, vol. 6 no. 1,DOI: 10.1038/s41392-021-00816-9, 2021.
[2] X. C. Dong, K. Chowdhury, M. Huang, H. G. Kim, "Signal transduction and molecular regulation in fatty liver disease," Antioxidants & Redox Signaling, vol. 35 no. 9, pp. 689-717, DOI: 10.1089/ars.2021.0076, 2021.
[3] H. Li, M. H. Huang, J. D. Jiang, Z. G. Peng, "Hepatitis C: from inflammatory pathogenesis to anti-inflammatory/hepatoprotective therapy," World Journal of Gastroenterology, vol. 24 no. 47, pp. 5297-5311, DOI: 10.3748/wjg.v24.i47.5297, 2018.
[4] T. van der Poll, F. L. van de Veerdonk, B. P. Scicluna, M. G. Netea, "The immunopathology of sepsis and potential therapeutic targets," Nature Reviews. Immunology, vol. 17 no. 7, pp. 407-420, DOI: 10.1038/nri.2017.36, 2017.
[5] K. E. Rudd, S. C. Johnson, K. M. Agesa, K. A. Shackelford, D. Tsoi, D. R. Kievlan, D. V. Colombara, K. S. Ikuta, N. Kissoon, S. Finfer, C. Fleischmann-Struzek, F. R. Machado, K. K. Reinhart, K. Rowan, C. W. Seymour, R. S. Watson, T. E. West, F. Marinho, S. I. Hay, R. Lozano, A. D. Lopez, D. C. Angus, C. J. L. Murray, M. Naghavi, "Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study," The Lancet, vol. 395 no. 10219, pp. 200-211, DOI: 10.1016/S0140-6736(19)32989-7, 2020.
[6] M. Cecconi, L. Evans, M. Levy, A. Rhodes, "Sepsis and septic shock," The Lancet, vol. 392 no. 10141, pp. 75-87, DOI: 10.1016/S0140-6736(18)30696-2, 2018.
[7] A. C. Kalil, "Sepsis and septic shock," Seminars in Respiratory and Critical Care Medicine, vol. 42 no. 5, pp. 639-640, DOI: 10.1055/s-0041-1735218, 2021.
[8] C. Cheng, X. Yu, "Research progress in Chinese herbal medicines for treatment of sepsis: pharmacological action, phytochemistry, and pharmacokinetics," International Journal of Molecular Sciences, vol. 22 no. 20,DOI: 10.3390/ijms222011078, 2021.
[9] H. Li, N. N. Liu, J. R. Li, B. Dong, M. X. Wang, J. L. Tan, X. K. Wang, J. Jiang, L. Lei, H. Y. Li, H. Sun, J. D. Jiang, Z. G. Peng, "Combined use of bicyclol and berberine alleviates mouse nonalcoholic fatty liver disease," Frontiers in Pharmacology, vol. 13, article 843872,DOI: 10.3389/fphar.2022.843872, 2022.
[10] W. J. Kong, C. Vernieri, M. Foiani, J. D. Jiang, "Berberine in the treatment of metabolism-related chronic diseases: a drug cloud (dCloud) effect to target multifactorial disorders," Pharmacology & Therapeutics, vol. 209, article 107496,DOI: 10.1016/j.pharmthera.2020.107496, 2020.
[11] Y. Wang, X. Zhou, D. Zhao, X. Wang, E. C. Gurley, R. Liu, X. Li, P. B. Hylemon, W. Chen, H. Zhou, "Berberine inhibits free fatty acid and LPS-induced inflammation via modulating ER stress response in macrophages and hepatocytes," PLoS One, vol. 15 no. 5, article e0232630,DOI: 10.1371/journal.pone.0232630, 2020.
[12] G. X. Li, X. M. Wang, T. Jiang, J. F. Gong, L. Y. Niu, N. Li, "Berberine prevents damage to the intestinal mucosal barrier during early phase of sepsis in rat through mechanisms independent of the NOD-like receptors signaling pathway," European Journal of Pharmacology, vol. 730,DOI: 10.1016/j.ejphar.2014.02.006, 2014.
[13] A. W. Feng, W. Gao, G. R. Zhou, R. Yu, N. Li, X. L. Huang, Q. R. Li, J. S. Li, "Berberine ameliorates COX-2 expression in rat small intestinal mucosa partially through PPAR γ pathway during acute endotoxemia," International Immunopharmacology, vol. 12 no. 1, pp. 182-188, DOI: 10.1016/j.intimp.2011.11.009, 2012.
[14] J. S. Shin, H. E. Choi, S. Seo, J. H. Choi, N. I. Baek, K. T. Lee, "Berberine decreased inducible nitric oxide synthase mRNA stability through negative regulation of human antigen R in lipopolysaccharide-induced macrophages," The Journal of Pharmacology and Experimental Therapeutics, vol. 358 no. 1,DOI: 10.1124/jpet.115.231043, 2016.
[15] Y. Lv, J. Wang, D. Xu, S. Liao, P. Li, Q. Zhang, M. Yang, L. Kong, "Comparative study of single/combination use of Huang-Lian-Jie-Du decoction and berberine on their protection on sepsis induced acute liver injury by NMR metabolic profiling," Journal of Pharmaceutical and Biomedical Analysis, vol. 145, pp. 794-804, DOI: 10.1016/j.jpba.2017.07.062, 2017.
[16] C. Wang, Q. Li, J. Ren, "Microbiota-immune interaction in the pathogenesis of gut-derived infection," Frontiers in Immunology, vol. 10,DOI: 10.3389/fimmu.2019.01873, 2019.
[17] Y. He, X. Yuan, H. Zuo, Y. Sun, A. Feng, "Berberine exerts a protective effect on gut-vascular barrier via the modulation of the Wnt/beta-catenin signaling pathway during sepsis," Cellular Physiology and Biochemistry, vol. 49 no. 4, pp. 1342-1351, DOI: 10.1159/000493412, 2018.
[18] H. M. Li, Y. Y. Wang, H. D. Wang, W. J. Cao, X. H. Yu, D. X. Lu, R. B. Qi, C. F. Hu, Y. X. Yan, "Berberine protects against lipopolysaccharide-induced intestinal injury in mice via alpha 2 adrenoceptor-independent mechanisms," Acta Pharmacologica Sinica, vol. 32 no. 11, pp. 1364-1372, DOI: 10.1038/aps.2011.102, 2011.
[19] L. Niu, W. Qiao, Z. Hu, N. Li, Q. Huang, J. Gong, Q. Li, W. Zhu, J. Li, "Berberine attenuates lipopolysaccharide-induced impairments of intestinal glutamine transport and glutaminase activity in rat," Fitoterapia, vol. 82 no. 3, pp. 323-330, DOI: 10.1016/j.fitote.2010.11.007, 2011.
[20] G. X. Li, X. M. Wang, T. Jiang, J. F. Gong, L. Y. Niu, N. Li, "Berberine prevents intestinal mucosal barrier damage during early phase of sepsis in rat through the Toll-like receptors signaling pathway," The Korean Journal of Physiology & Pharmacology, vol. 19 no. 1,DOI: 10.4196/kjpp.2015.19.1.1, 2015.
[21] Y. Li, J. Zhou, J. Qiu, Z. Huang, W. Wang, P. Wu, A. Feng, "Berberine reduces gut-vascular barrier permeability via modulation of ApoM/S1P pathway in a model of polymicrobial sepsis," Life Sciences, vol. 261, article 118460,DOI: 10.1016/j.lfs.2020.118460, 2020.
[22] J. Souffriau, C. Libert, "Mechanistic insights into the protective impact of zinc on sepsis," Cytokine & Growth Factor Reviews, vol. 39, pp. 92-101, DOI: 10.1016/j.cytogfr.2017.12.002, 2018.
[23] S. S. Siddiqui, C. Dhar, V. Sundaramurthy, A. Sasmal, H. Yu, E. Bandala-Sanchez, M. Li, X. Zhang, X. Chen, L. C. Harrison, D. Xu, A. Varki, "Sialoglycan recognition is a common connection linking acidosis, zinc, and HMGB1 in sepsis," Proceedings of the National Academy of Sciences of the United States of America, vol. 118 no. 10,DOI: 10.1073/pnas.2018090118, 2021.
[24] I. Wessels, R. J. Cousins, "Zinc dyshomeostasis during polymicrobial sepsis in mice involves zinc transporter Zip14 and can be overcome by zinc supplementation," American Journal of Physiology. Gastrointestinal and Liver Physiology, vol. 309 no. 9, pp. G768-G778, DOI: 10.1152/ajpgi.00179.2015, 2015.
[25] Y. He, X. Yuan, H. Zuo, X. Li, Y. Sun, A. Feng, "Berberine induces ZIP14 expression and modulates zinc redistribution to protect intestinal mucosal barrier during polymicrobial sepsis," Life Sciences, vol. 233, article 116697,DOI: 10.1016/j.lfs.2019.116697, 2019.
[26] C. L. Li, L. H. Tan, Y. F. Wang, C. D. Luo, H. B. Chen, Q. Lu, Y. C. Li, X. B. Yang, J. N. Chen, Y. H. Liu, J. H. Xie, Z. R. Su, "Comparison of anti-inflammatory effects of berberine, and its natural oxidative and reduced derivatives from Rhizoma Coptidis in vitro and in vivo," Phytomedicine, vol. 52, pp. 272-283, DOI: 10.1016/j.phymed.2018.09.228, 2019.
[27] K. K. Reddi, H. Li, W. Li, S. D. Tetali, "Berberine, a phytoalkaloid, inhibits inflammatory response induced by LPS through NF-kappa β pathway: possible involvement of the IKK α," Molecules, vol. 26 no. 16,DOI: 10.3390/molecules26164733, 2021.
[28] H. W. Jeong, K. C. Hsu, J. W. Lee, M. Ham, J. Y. Huh, H. J. Shin, W. S. Kim, J. B. Kim, "Berberine suppresses proinflammatory responses through AMPK activation in macrophages," American Journal of Physiology. Endocrinology and Metabolism, vol. 296 no. 4, pp. E955-E964, DOI: 10.1152/ajpendo.90599.2008, 2009.
[29] L. Huang, X. Zhang, X. Ma, D. Zhang, D. Li, J. Feng, X. Pan, J. Lü, X. Wang, X. Liu, "Berberine alleviates endothelial glycocalyx degradation and promotes glycocalyx restoration in LPS-induced ARDS," International Immunopharmacology, vol. 65, pp. 96-107, DOI: 10.1016/j.intimp.2018.10.001, 2018.
[30] Y. Wang, P. Du, D. Jiang, "Berberine functions as a negative regulator in lipopolysaccharide -induced sepsis by suppressing NF- κ B and IL-6 mediated STAT3 activation," Pathogens and Disease, vol. 78 no. 7,DOI: 10.1093/femspd/ftaa047, 2020.
[31] J. Chen, Y. Huang, X. Bian, Y. He, "Berberine ameliorates inflammation in acute lung injury via NF- κ B/Nlrp3 signaling pathway," Frontiers in Nutrition, vol. 9, article 851255,DOI: 10.3389/fnut.2022.851255, 2022.
[32] R. Bellomo, J. A. Kellum, C. Ronco, R. Wald, J. Martensson, M. Maiden, S. M. Bagshaw, N. J. Glassford, Y. Lankadeva, S. T. Vaara, A. Schneider, "Acute kidney injury in sepsis," Intensive Care Medicine, vol. 43 no. 6, pp. 816-828, DOI: 10.1007/s00134-017-4755-7, 2017.
[33] T. Li, J. Y. Liu, J. F. Liu, M. Duan, A. Li, "The correlation between the types of initial bacterial infection and clinical prognosis in patients with septic AKI," Frontiers in Medicine (Lausanne), vol. 8, article 800532, 2021.
[34] L. S. Chawla, R. L. Amdur, S. Amodeo, P. L. Kimmel, C. E. Palant, "The severity of acute kidney injury predicts progression to chronic kidney disease," Kidney International, vol. 79 no. 12, pp. 1361-1369, DOI: 10.1038/ki.2011.42, 2011.
[35] H. M. Al-Kuraishy, A. I. Al-Gareeb, N. R. Hussien, "Synergistic effect of berberine and pentoxifylline in attenuation of acute kidney injury," International Journal of Critical Illness and Injury Science, vol. 9 no. 2, pp. 69-74, DOI: 10.4103/IJCIIS.IJCIIS_85_18, 2019.
[36] B. L. Ma, C. Yin, B. K. Zhang, Y. Dai, Y. Q. Jia, Y. Yang, Q. Li, R. Shi, T. M. Wang, J. S. Wu, Y. Y. Li, G. Lin, Y. M. Ma, "Naturally occurring proteinaceous nanoparticles in _Coptidis Rhizoma_ extract act as concentration-dependent carriers that facilitate berberine absorption," Scientific Reports, vol. 6 no. 1,DOI: 10.1038/srep20110, 2016.
[37] Y. Zheng, X. Shi, J. Hou, S. Gao, Y. Chao, J. Ding, L. Chen, Y. Qian, G. Shao, Y. Si, W. Chen, "Integrating metabolomics and network pharmacology to explore _Rhizoma Coptidis_ extracts against sepsis-associated acute kidney injury," Journal of Chromatography. B, Analytical Technologies in the Biomedical and Life Sciences, vol. 1164, article 122525,DOI: 10.1016/j.jchromb.2021.122525, 2021.
[38] J. Shen, W. Wang, X. Shao, J. Wu, S. Li, X. Che, Z. Ni, "Integrated analysis of m6A methylome in cisplatin-induced acute kidney injury and berberine alleviation in mouse," Frontiers in Genetics, vol. 11, article 584460,DOI: 10.3389/fgene.2020.584460, 2020.
[39] J. Qi, Q. Xue, L. Kuang, L. Xie, R. Luo, X. Nie, "Berberine alleviates cisplatin-induced acute kidney injury by regulating mitophagy via PINK 1/Parkin pathway," Translational Andrology and Urology, vol. 9 no. 4, pp. 1712-1724, DOI: 10.21037/tau-20-1129, 2020.
[40] M. Adil, A. D. Kandhare, G. Dalvi, P. Ghosh, S. Venkata, K. S. Raygude, S. L. Bodhankar, "Ameliorative effect of berberine against gentamicin-induced nephrotoxicity in rats via attenuation of oxidative stress, inflammation, apoptosis and mitochondrial dysfunction," Renal Failure, vol. 38 no. 6, pp. 996-1006, DOI: 10.3109/0886022X.2016.1165120, 2016.
[41] P. Strnad, F. Tacke, A. Koch, C. Trautwein, "Liver -- guardian, modifier and target of sepsis," Nature Reviews. Gastroenterology & Hepatology, vol. 14 no. 1, pp. 55-66, DOI: 10.1038/nrgastro.2016.168, 2017.
[42] H. Li, J. R. Li, M. H. Huang, J. H. Chen, X. Q. Lv, L. L. Zou, J. L. Tan, B. Dong, Z. G. Peng, J. D. Jiang, "Bicyclol attenuates liver inflammation induced by infection of hepatitis C virus via repressing ROS-mediated activation of MAPK/NF- κ B signaling pathway," Frontiers in Pharmacology, vol. 9,DOI: 10.3389/fphar.2018.01438, 2018.
[43] D. Qiu, W. Zhang, Z. Song, M. Xue, Y. Zhang, Y. Yang, C. Tong, D. Cai, "Berberine suppresses cecal ligation and puncture induced intestinal injury by enhancing Treg cell function," International Immunopharmacology, vol. 106, article 108564,DOI: 10.1016/j.intimp.2022.108564, 2022.
[44] K. Tsoyi, H. J. Jang, I. T. Nizamutdinova, Y. M. Kim, Y. S. Lee, H. J. Kim, H. G. Seo, J. H. Lee, K. C. Chang, "Metformin inhibits HMGB1 release in LPS-treated RAW 264.7 cells and increases survival rate of endotoxaemic mice," British Journal of Pharmacology, vol. 162 no. 7, pp. 1498-1508, DOI: 10.1111/j.1476-5381.2010.01126.x, 2011.
[45] D. U. Lee, Y. S. Ko, H. J. Kim, K. C. Chang, "13-Ethylberberine reduces HMGB1 release through AMPK activation in LPS- activated RAW264.7 cells and protects endotoxemic mice from organ damage," Biomedicine & Pharmacotherapy, vol. 86, pp. 48-56, DOI: 10.1016/j.biopha.2016.11.099, 2017.
[46] A. M. Gendy, M. R. Elnagar, M. M. Allam, M. R. Mousa, A. E. Khodir, A. E. el-Haddad, O. S. Elnahas, S. M. Fayez, S. S. el-Mancy, "Berberine-loaded nanostructured lipid carriers mitigate warm hepatic ischemia/reperfusion-induced lesion through modulation of HMGB1/TLR4/NF- κ B signaling and autophagy," Biomedicine & Pharmacotherapy, vol. 145, article 112122,DOI: 10.1016/j.biopha.2021.112122, 2022.
[47] T. van der Poll, "Tissue factor as an initiator of coagulation and inflammation in the lung," Critical Care, vol. 12,DOI: 10.1186/cc7026, 2008.
[48] M. Y. Gao, L. Chen, L. Yang, X. Yu, J. P. Kou, B. Y. Yu, "Berberine inhibits LPS-induced TF procoagulant activity and expression through NF- κ B/p65, Akt and MAPK pathway in THP-1 cells," Pharmacological Reports, vol. 66 no. 3, pp. 480-484, DOI: 10.1016/j.pharep.2013.12.004, 2014.
[49] X. Yang, X. Cheng, Y. Tang, X. Qiu, Y. Wang, H. Kang, J. Wu, Z. Wang, Y. Liu, F. Chen, X. Xiao, N. Mackman, T. R. Billiar, J. Han, B. Lu, "Bacterial endotoxin activates the coagulation cascade through gasdermin D-dependent phosphatidylserine exposure," Immunity, vol. 51 no. 6, article e986, pp. 983-996.e6, DOI: 10.1016/j.immuni.2019.11.005, 2019.
[50] C. Yuan, M. Wu, Q. Xiao, W. Zhao, H. Li, Y. Zhong, M. Zhao, C. Li, Y. Li, X. Yang, "Blocking Msr1 by berberine alkaloids inhibits caspase-11-dependent coagulation in bacterial sepsis," Signal Transduction and Targeted Therapy, vol. 6 no. 1,DOI: 10.1038/s41392-021-00483-w, 2021.
[51] C. Wang, Y. Cheng, Y. Zhang, H. Jin, Z. Zuo, A. Wang, J. Huang, J. Jiang, W. Kong, "Berberine and its main metabolite berberrubine inhibit platelet activation through suppressing the class I PI3K β /Rasa3/Rap1 pathway," Frontiers in Pharmacology, vol. 12, article 734603,DOI: 10.3389/fphar.2021.734603, 2021.
[52] M. Xue, M. X. Yang, W. Zhang, X. M. Li, D. H. Gao, Z. M. Ou, Z. P. Li, S. H. Liu, X. J. Li, S. Y. Yang, "Characterization, pharmacokinetics, and hypoglycemic effect of berberine loaded solid lipid nanoparticles," International Journal of Nanomedicine, vol. 8, pp. 4677-4687, DOI: 10.2147/IJN.S51262, 2013.
[53] J. Deng, Z. Wu, Z. Zhao, C. Wu, M. Yuan, Z. Su, Y. Wang, Z. Wang, "Berberine-loaded nanostructured lipid carriers enhance the treatment of ulcerative colitis," International Journal of Nanomedicine, vol. 15, pp. 3937-3951, DOI: 10.2147/IJN.S247406, 2020.
[54] L. Zhao, X. Du, J. Tian, X. Kang, Y. Li, W. Dai, D. Li, S. Zhang, C. Li, "Berberine-loaded carboxylmethyl chitosan nanoparticles ameliorate DSS-induced colitis and remodel gut microbiota in mice," Frontiers in Pharmacology, vol. 12, article 644387,DOI: 10.3389/fphar.2021.644387, 2021.
[55] T. Li, P. Wang, W. Guo, X. Huang, X. Tian, G. Wu, B. Xu, F. Li, C. Yan, X. J. Liang, H. Lei, "Natural berberine-based Chinese herb medicine assembled nanostructures with modified antibacterial application," ACS Nano, vol. 13 no. 6, pp. 6770-6781, DOI: 10.1021/acsnano.9b01346, 2019.
[56] Z. Liu, Y. Liu, T. Fang, J. Xia, N. Ma, Y. Wang, "Application of berberine-loaded albumin nanoparticles in infections of traumatic wounds," International Journal of Burns and Trauma, vol. 12 no. 1, pp. 28-34, 2022.
[57] X. Yu, S. Yu, L. Chen, H. Liu, J. Zhang, H. Ge, Y. Zhang, B. Yu, J. Kou, "Tetrahydroberberrubine attenuates lipopolysaccharide-induced acute lung injury by down-regulating MAPK, AKT, and NF- κ B signaling pathways," Biomedicine & Pharmacotherapy, vol. 82, pp. 489-497, DOI: 10.1016/j.biopha.2016.05.025, 2016.
[58] E. Bhatia, S. Sharma, K. Jadhav, R. Banerjee, "Combinatorial liposomes of berberine and curcumin inhibit biofilm formation and intracellular methicillin resistant Staphylococcus aureus infections and associated inflammation," Journal of Materials Chemistry B, vol. 9 no. 3, pp. 864-875, DOI: 10.1039/D0TB02036B, 2021.
[59] X. Li, P. Wang, X. Hu, Y. Zhang, X. Lu, C. Li, T. Nie, G. Li, X. Wang, J. Pang, Y. Lu, X. Yang, X. You, "The combined antibacterial effects of sodium new houttuyfonate and berberine chloride against growing and persistent methicillin-resistant and vancomycin-intermediate Staphylococcus aureus," BMC Microbiology, vol. 20 no. 1,DOI: 10.1186/s12866-020-02003-2, 2020.
[60] B. Li, S. Niu, H. Geng, C. Yang, C. Zhao, "Berberine attenuates neonatal sepsis in mice by inhibiting FOXA1 and NF- κ B signal transduction via the induction of MiR-132-3p," Inflammation, vol. 44 no. 6, pp. 2395-2406, DOI: 10.1007/s10753-021-01510-2, 2021.
[61] E. Pierpaoli, O. Cirioni, O. Simonetti, F. Orlando, A. Giacometti, P. Lombardi, M. Provinciali, "Potential application of berberine in the treatment of Escherichia coli sepsis," Natural Product Research, vol. 35 no. 22, pp. 4779-4784, DOI: 10.1080/14786419.2020.1721729, 2021.
[62] A. A. Alalwan, A. Taher, A. H. Alaradi, "A hemodialysis patient with severe COVID-19 pneumonia," Cureus, vol. 12 no. 5, article e7995,DOI: 10.7759/cureus.7995, 2020.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer
Copyright © 2023 Tingxia Lv et al. This is an open access article distributed under the Creative Commons Attribution License (the “License”), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License. https://creativecommons.org/licenses/by/4.0/
Abstract
Sepsis is defined as a dysregulated immune response to infection that leads to multiple organ dysfunction. To date, though a growing body of knowledge has gained insight into the clinical risk factors, pathobiology, treatment response, and recovery methods, sepsis remains a significant concern and clinical burden. Therefore, further study is urgently needed to alleviate the acute and chronic outcomes. Berberine (BBR), a traditional Chinese medicine with multiple actions and mechanisms, has been investigated in cellular and rodent animal models of sepsis mainly based on its anti-inflammatory effect. However, the practical application of BBR in sepsis is still lacking, and it is imperative to systematically summarize the study of BBR in sepsis. This review summarized its pharmacological activities and mechanisms in septic-related organ injuries and the potential BBR-based therapeutic strategies for sepsis, which will provide comprehensive references for scientific research and clinical application.
You have requested "on-the-fly" machine translation of selected content from our databases. This functionality is provided solely for your convenience and is in no way intended to replace human translation. Show full disclaimer
Neither ProQuest nor its licensors make any representations or warranties with respect to the translations. The translations are automatically generated "AS IS" and "AS AVAILABLE" and are not retained in our systems. PROQUEST AND ITS LICENSORS SPECIFICALLY DISCLAIM ANY AND ALL EXPRESS OR IMPLIED WARRANTIES, INCLUDING WITHOUT LIMITATION, ANY WARRANTIES FOR AVAILABILITY, ACCURACY, TIMELINESS, COMPLETENESS, NON-INFRINGMENT, MERCHANTABILITY OR FITNESS FOR A PARTICULAR PURPOSE. Your use of the translations is subject to all use restrictions contained in your Electronic Products License Agreement and by using the translation functionality you agree to forgo any and all claims against ProQuest or its licensors for your use of the translation functionality and any output derived there from. Hide full disclaimer