Open Access

Proanthocyanidins: A novel approach to Henoch‑Schonlein purpura through balancing immunity and arresting oxidative stress via TLR4/MyD88/NF‑κB signaling pathway (Review)

  • Authors:
    • Yuxin Xie
    • Qiyan Deng
    • Menglu Guo
    • Xiaolong Li
    • Deihai Xian
    • Jianqiao Zhong
  • View Affiliations

  • Published online on: May 9, 2023     https://doi.org/10.3892/etm.2023.11999
  • Article Number: 300
  • Copyright: © Xie et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Henoch‑Schonlein purpura (HSP), a recurrent and immunoglobulin (Ig)A‑mediated vasculitis, presents not only as skin lesions but also as systemic involvement that can be life‑threatening. Although the etiology of HSP remains unknown, immune imbalance and oxidative stress (OS) are primary contributors to its pathogenesis, alongside the abnormal activation of Toll‑like receptor (TLR)/myeloid differentiation primary response gene 88 (MyD88)/nuclear factor‑κB (NF‑κB) pathway. TLRs, especially TLR4, stimulate downstream signaling molecules such as NF‑κB and proinflammatory cytokines, which are released when TLRs combine with the key adapter molecule MyD88. This leads to the activation of T helper (Th) cell 2/Th17 and overproduction of reactive oxygen species (ROS). The function of regulatory T (Treg) cells is suppressed in the process. Th17/Treg imbalance then produces various inflammatory cytokines to promote proliferation and differentiation of B cells and the secretion of antibodies. IgA is secreted, and it binds to vascular endothelial surface receptors where the complex induces injury of the vascular endothelial cells. Additionally, excessive ROS creates OS that leads to an inflammatory response and vascular cell apoptosis or necrosis, thereby contributing to vascular endothelial damage and HSP occurrence. Proanthocyanidins are active compounds naturally enriched in fruits, vegetables and plants. Proanthocyanidins have diverse properties, including anti‑inflammatory, antioxidant, antibacterial, immunoregulatory, anticarcinogenic and vascular protective effects. Proanthocyanidins are used in the management of various diseases. Proanthocyanidins regulate T cells, equilibrate immunity and arrest OS by inhibiting the TLR4/MyD88/NF‑κB signaling pathway. Considering the pathogenesis of HSP and the properties of proanthocyanidins, the present study hypothesized that these compounds may potentially lead to HSP recovery through modulating the immune equilibrium and preventing OS by inhibiting the TLR4/MyD88/NF‑κB pathway. To the best of our knowledge, however, little is known about the positive effects of proanthocyanidins against HSP. The present review summarizes the potential of proanthocyanidins to treat HSP.

1. Introduction

Henoch-Schonlein purpura (HSP) is an immunoglobulin (Ig)A-mediated relapsing vasculitis that invades small arteries and capillaries of the skin and other organs, such as gastrointestinal tract and kidneys. It primarily manifests as cutaneous purpura, joint swelling, digestive symptoms (mainly abdominal pain) and kidney injury, which can be fatal (1,2). Based on clinical features, HSP is classified into five categories: Simple, arthritis, abdominal, renal and mixed types (3). HSP frequently occurs in children, affecting 3-27 children per 100,000 population; but there has been an increasing number of incidents in adults who possibly suffer more serious systemic issues due to age (4). To the best of our knowledge, the pathogenesis of HSP has not been completely elucidated; however, immune imbalance and oxidative stress (OS) that induce IgA-mediated vascular damage may be involved (5,6). It is also hypothesized that toll-like receptor (TLR)4/myeloid differentiation primary response gene 88 (MyD88)/nuclear factor-κB (NF-κB) signaling is involved (7). Various treatments have been applied to HSP, including anti-inflammatory drugs, anti-histamine, vitamin C, calcium, corticosteroids, cytotoxic drugs and immunosuppressants, as well as blood-activating and stasis-resolving medicines (8-10). However, these therapies exhibit poor efficacy and do not stop recurrence, research has shown that relapses occur in about 25% of patients and that children over age 8 or those with kidney-based disease are more prone to relapse (11); furthermore, most approaches to HSP are unsuitable for long-term application owing to high expenditures and adverse side effects, e.g. long-term and large-scale use of glucocorticoids could cause infections and Cushing's syndrome; long-term use of cytotoxic drugs that could result in gastrointestinal reactions, hepatotoxicity, bone marrow suppression, reproductive toxicity, cardiotoxicity (12). Thus, a novel and effective cure for HSP is required.

Proanthocyanidins are a class of natural polyphenolic compounds that consist of epicatechin and catechin. Proanthocyanidins are obtained from seeds, vegetables, fruits and plants. Previous studies have demonstrated that proanthocyanidins are beneficial to human health due to few side effects and multiple biological activities, such as immunoregulation, antioxidation, anti-inflammation, anti-angiogenesis, anti-proliferation and anti-tumor effects (13-17). Growing evidence from clinical and experimental studies indicates that proanthocyanidins are predominantly safe and effective against diseases, such as inflammatory disorders, OS-associated diseases, vascular disorders and immune complaints. Proanthocyanidins function by regulating immune balance, attenuating OS damage and promoting vascular endothelial integrity (14,16,18,19). Additionally, proanthocyanidins modulate the abnormal metabolism of the body to control metabolic complaints and neoplastic disorders through suppressing lipid peroxidation (LPO) and inflammatory responses (20-22). However, reports scarcely focus on the possible efficacy of proanthocyanidins in the treatment of HSP. Given that HSP is an immune-mediated and OS-induced vasculitis and proanthocyanidins possess immunomodulatory and antioxidative properties, the present study hypothesizes that proanthocyanidins could be a treatment for HSP. Therefore, the evidence that suggests the potential efficacy of proanthocyanidins in treating HSP is reviewed and discussed in the present study.

2. Possible pathogenesis of HSP

As a multi-factorial disease, the etiology of HSP is diverse and complex and includes microbial infection, foods, drugs and malignant tumors. Reports suggest that immune imbalance and OS that induce IgA-mediated vessel injury are involved in the pathogenesis of HSP (23,24). The abnormal activation of TLRs (primarily TLR4) and the downstream pathways, are implicated in this process (25-29).

Role of immune dysregulation in HSP

Immune imbalance, especially T helper 2 (Th2) and Th17 overactivity, facilitates secretion of numerous chemokines and inflammatory cytokines (such as TNF-α, IL-1, IL-4, IL-6, IL-17 and cell adhesion factors) and promotes the migration of leukocytes to inflammatory sites, thus aggravating vascular inflammation and exacerbating HSP (28,30). Donadio et al (27) demonstrated that overactivation of TLRs (primarily TLR4) and elevated levels of adaptive immunity appear in patients with HSP; these promote an imbalance of the T cells and a release of inflammatory cytokines. As the downstream molecule of TLRs, NF-κB is required for the development of T cells and is involved in the activation of CD4+ T cells, particularly Th2 and Th17 cells (31). TLRs initially promote NF-κB translocation and activation by recruiting MyD88; TLRs combined with MyD88 employ the IL-1 receptor-associated kinase 1 (IRAK1) and IRAK4 to induce the activation of the transforming growth factor-β-activated kinase (TAK) complex, which causes autophosphorylation of TAK1. TAK1 promotes phosphorylation of IκBα by degrading the inhibitor κ-B kinase, which facilitates dissociation of IκBα from NF-κB and the production of the NF-κB complex (a dimer of p50 and p65) (32). The activated NF-κB then promotes T cells to produce activation-associated molecules, thereby activating and increasing naive T cells. Stimulated naive T cells differentiate into the effector subpopulations, such as Th1, Th2 and Th17. NF-κB, by combining with enhancer sites of IL-4 and simultaneously binding with NF of activated T cells, stimulates IL-4 that then further promotes Th2 development and activation. By contrast, NF-κB facilitate the growth and development of Th17 cells through enhances the secretion of Th17 directive signals such as IL-17a, IL-6, TNF-α and TGF-β, and stimulating the expression of major transcription factors of Th17 (RORγt and RORγ) alongside c-Rel of the Rel family. In addition, activated NF-κB suppresses the function of regulatory T (Treg) cells, followed by initiation of immune imbalance and the inflammatory response (29,33-36). Due to activation of TLRs, a number of aberrant alterations emerge in patients with HSP, including a high-proportion of Th17 cells, Th1/Th2 cell imbalance and Th2-dominant immune response, as well as decreased IFN-γ and IL-4 release. These alterations contribute to production of pro-inflammatory/inflammatory factors and adhesion molecules, recruitment of immune/inflammatory cells and amplification of the immune/inflammatory response (28,37,38). Consequently, overactivated T cells in turn release inflammatory factors, especially Th2-released cytokines, to accelerate the proliferation and differentiation of B cells that synthesize and secrete antibodies (IgA, IgG and IgM). Igs bind to corresponding receptors and activate complement components 3/4 to trigger OS and initiate inflammation, which leads to vessel wall destruction and HSP (39).

Decreased Treg cells aggravates HSP. Based on the origin and differentiation, Treg cells are classified into three subtypes: Thymus-derived (t), also termed natural Treg cells; peripheral (p) and inducible (i). The tTregs are generated in thymus, while pTregs and iTregs are induced in peripheral sites (40,41). It is generally considered that an immune and inflammation imbalance weakens the function of Treg cells, which promotes inflammatory reactions and exacerbates the disease (34,42). Levels of CD4+CD25+Foxp3+ Treg cells markedly decrease in patients with HSP, whereas Th17 cell levels markedly increase; this imbalance of Th17/Treg promotes Treg cell dysfunction in HSP, amplifying inflammatory responses and destroying immune defense (35). Conversely, stable expression of Foxp3 induces Treg cells to produce anti-inflammatory factor IL-10, which inhibits the inflammatory response and maintains immune homeostasis. Furthermore, there is a decrease of follicular Treg cells in blood of pediatric patients with HSP, along with an increase in follicular Th cells; this imbalance leads to the overactivation of B cells, which then stimulate IgA production and inflammation (43).

As an antigen-antibody reaction, the inflammatory process mediated by IgA is the pathology of HSP. B cell-secreted IgA antibodies induce chemokine IL-8/leukotriene B4 (LTB4) production and promote chemotaxis/aggregation of neutrophils, which stimulates the inflammatory response and causes damage to endothelial cells (ECs) (44,45). Excessive numbers of IgA antibody interact with FcαRI (a prototypical IgA receptor) and bind to vascular ECs (VECs), thereby attracting neutrophils to migrate to the blood vessel wall in the presence of IL-8, in addition to eliciting ROS production and OS occurrence and membrane LPO) (46,47).

Biomarkers derived from investigation of OS activation in HSP

OS describes an imbalance where production of oxidative species exceeds the capacity of antioxidant defense systems. This then leads to macromolecular damage and redox homeostatic disorder. Free radical reaction is a widely recognized phenomenon that is greatly involved in macromolecular damage (48-51). The overproduction of oxygen radicals or oxides, including ROS [such as superoxide anion (O2-), hydrogen peroxide (H2O2), hydroxyl radical and singlet oxygen] and reactive nitrogen species [such as nitric oxide (NO), peroxynitrite anion and nitrite ion] (52,53), disturbs the equilibrium of the oxidant/antioxidant system. This imbalance creates OS that causes damage to the skin and VECs. As the primary contributor to OS, ROS production is triggered by exogenous factors [such as ultraviolet (UV) radiation, heavy metal ions, ozone, drugs or toxins] and endogenous factors including mitochondrial electron transport chain, membrane-bound NADPH oxidase isoforms 1-5 and endoplasmic reticulum. NADPH oxidases in particular are the primary source of ROS (53,54). The electrons generated by NADPH are transferred by an NADPH enzyme onto molecular oxygen that is reduced to O2•-. Superoxide dismutase (SOD) catalyzes O2-conversion into H2O2, and more ROS are formed (55-57).

Increasing evidence has supported OS as a promoter in the progression of vascular disorder, and in particular HSP (58-60). It has been revealed that OS often occurs in HSP, presenting as high levels of ROS and malondialdehyde (MDA) and low SOD levels and total antioxidant capacity. Serum MDA is an indicator of LPO, which increases in the active phase of HSP (massive purpura is seen on the skin and some patients experience abdominal and joint pain) and indirectly signifies the degree of VEC injury in patients with HSP (5,61,62). The dysfunction or damage of ECs impacts on the integrity of the vascular wall and the permeability of the vessel, which is attributed to the amount of OS (63-65). As a result, inflammatory cells adhere to the vessel wall to exacerbate damage of the vascular endothelium and deposit IgA at the vascular wall; these in turn promote the production of chemokines, accumulation/infiltration of inflammatory cells and injury of the vessel to exacerbate OS, thereby forming a positive feedback loop (66-68). Previous studies have reported higher levels of MDA in the serum of patients with HSP compared with levels in healthy individuals; MDA levels are highest in patients with renal or gastrointestinal involvement (21-23). Similar reports have indicated that the damage of ECs and aggravation of vascular inflammation frequently occur in the production of ROS from OS, while neutrophils in patients with HSP positively mediate oxygen radical production (61,69). Superfluous ROS destroy the antioxidant system and trigger OS, whereas the impaired antioxidant defense system aggravates the persistence of OS. This stimulates the release of diverse inflammatory factors and inflammatory reactions around the blood vessels. IL-1β, activated by TLRs, contributes to the HSP inflammatory reaction and is regulated by a biphasic redox response (70-72). The excess ROS and attenuated antioxidant defense stimulates the secretion of IL-1β, while IL-1β and cytokines involved in the inflammatory cascade response enhance the deposition of IgA on ECs (71,73). Conversely, damaged VECs and activated neutrophils produce large amounts of ROS, triggering the pathological and clinical manifestations of HSP (5,60,74).

The aforementioned processes are regulated by TLRs and downstream signaling pathways. Activating numerous inflammatory signals such as MyD88 and NF-κB, TLRs promote release of inflammatory mediators and the formation of immune complexes along with the overactivation of Th2/Th17 and the reduction of Treg cell populations and functional activity.

TLRs involvement in the induction of HSP

TLRs, belonging to a family of pattern recognition receptors, functionally recognize molecules that are associated with infection/tissue damage-associated molecules to trigger innate immunity and inflammation. The activation of TLRs promotes immune dysregulation, inflammatory responses and OS, thereby promoting vascular skin disorder (such as HSP) by stimulating TLR-mediated signaling pathways. TLRs comprise 13 members (TLR1-TLR13) (75,76). In all TLR members, the extracellular region is responsible for identifying the pathogen-associated molecular patterns (PAMPs) of the disease-causing microorganism, while the cytoplasmic region induces cellular immunological response (77). TLR4 localization on the cell surface is important in immune inflammatory disorders; it specifically recognizes the lipopolysaccharide (LPS) of pathogens and stimulates immune inflammatory reactions by activating adaptive immunity-associated genes, such as MyD88(78).

TLR4-mediated signaling pathways in immune responses primarily contain two categories: MyD88- and TIR-domain-containing adapter-inducing interferon-β (TRIF)-dependent pathway. Trough interacting with the MyD88, TRIF or TLR4 pathway that promotes downstream signaling molecule (NF-κB or activator protein-1) participation in immune inflammation (79). By stimulating MyD88, TLR4 activates NF-κB, thereby triggering signal transduction cascades and activating immune cells. MyD88 is an adaptor molecule recruited by almost all TLRs (TLR3-TLR13) (80,81). MyD88 has two domains, namely the TIR domain that interacts with the cognate domain in the cytoplasmic tails of TLRs and the death domain that binds to the corresponding domain of IRAK4. In the MyD88-dependent pathway, the signal pathway is exclusively activated by TLR4; signals are transmitted from TLR4 to MyD88, then to IRAK4; in the presence of phosphorylated IRAK1, IRAK4 activates TNF receptor-associated factor 6 and promotes NF-κB-dependent gene expression (82). NF-κB is activated after binding with TAK1 complex; activated NF-κB up-regulates the expression of pro-inflammatory factors (such as TNF-α and IL-1β) and increases the functional activities of NADPH oxidase and mitochondria. Both NADPH oxidase and mitochondria could promote the production of ROS, which touch off OS and inflammation, further contributing abnormal activation of Th17/Th2 cells, arresting Treg cell function and resulting in immune disturbance (25,28,83-85). Stimulated Th17 and Th2 cells generate chemokines and inflammatory cytokines to trigger the migration of neutrophils to inflammatory sites, thereby exacerbating vascular inflammation and facilitating HSP initiation (28,30).

HSP is caused by factors such as infection, drugs or tumors, which cause TLRs recognize PAMPs to activate the innate immunity. By binding to MyD88, TLR4 triggers signal transduction and stimulates the NF-κB signaling pathway, which promotes Th1/Th2 imbalance and release of pro-inflammatory mediators (25,74,83). These enhance B cell proliferation and differentiation, which contributes to IgA production and deposition on the vascular wall, as well as vascular endothelial damage (25,38). As a result, inflammatory cytokines are attracted to elicit ROS production and OS. Increased ROS in turn promotes TLR4 expression and the interplay between TLR4 and ROS stimulates the immuno-inflammatory response (47,68). The aforementioned molecular events cause further VEC injury and vascular permeability, ultimately leading to histopathological changes and clinical manifestations of HSP. The possible pathogenesis of HSP is presented in Fig. 1.

3. Biological properties and clinical applications of proanthocyanidins

Proanthocyanidins, a type of polyphenolic compound, are present in leaves, flowers, seeds, stems and roots, including common fruits and plants, such as grape, cranberry, black currant, pomegranate, cocoa and medlar (86). Proanthocyanidins comprise epicatechin, catechin, epigallocatechin or gallocatechin subunits connected by C4-C6 and C4-C8 bonds. Depending on the degree of polymerization, proanthocyanidins are primarily classified into two isoforms, namely oligomer proanthocyanidin (OPC) that have 2-5 monomers and polymer proanthocyanidin (PPC) that possess >5 monomers (87). Based on low molecular weight, OPCs exert prominent bioactivities such as immune modulatory, antioxidant, anti-inflammatory, antibacterial, anticarcinogenic, vascular protective and anti-hyperglycemic effects, which enable their application in clinical treatment (88-93). The bioactivity of PPCs is weaker due to the long carbon chain but increased by degeneration (94). Owing to their powerful immunomodulatory and antioxidant effects, proanthocyanidins are used to control immune-inflammatory and OS-associated disorder (95).

Previous studies have indicated that proanthocyanidins improving immune/inflammatory response and promoting lymphocyte transformation (96,97). Studies have confirmed that proanthocyanidins could modulate the balance of immune cells (especially T cells) and inflammatory cells to alleviate the immune/inflammatory reaction (96-99). Park et al (98) demonstrated in vitro and in vivo that proanthocyanidins notably increase gene transcription and protein levels of IFN-γ and decrease expression of IL-6. This results in activation of Th1 cells and inactivation of Th2 cells and a balance in humoral and cellular immunity. Furthermore, research on a murine model of auto-immune arthritis revealed that proanthocyanidins are effective in alleviating mouse arthritis symptoms by suppressing proliferation and differentiation of Th17 cells, increasing the activity and quantity of Treg cells and preventing secretion of various inflammatory cytokines, such as IL-17, IL-6, IL-21, IL-22 and TNF-α (96,99). Jhun et al (97) demonstrated that obesity and arthritis of both diet-induced obese mice and rheumatoid arthritis-like mice are relieved by proanthocyanidin treatment due to its ability to increase populations of the Treg cells and decrease the populations of the Th17 cells. Additionally, proanthocyanidins effectively prevent ovalbumin-induced allergic reactions by not only decreasing the release of inflammatory factors by regulating Th-derived cytokine expression (such as IL-4, IL-5 and IL-13), but also by inhibiting Ig (IgG1 and IgE) synthesis (100).

Furthermore, proanthocyanidins prevent OS, restore normal immune function and enhance antioxidant capacity by regulating multiple pathways, eliminating ROS/MDA, maintaining the balance of immune cells and inflammatory factors and upregulating detoxication enzyme and antioxidants (101-103). Kim et al (104) demonstrated that grape seed-derived proanthocyanidins effectively relieve joint swelling and the histopathological damage in a collagen-induced murine arthritis model through decreasing the levels of TLR4, MyD88 and NF-κB and inhibiting the TLR4/MyD88/NF-κB signaling pathway. Additionally, similar report has demonstrated that proanthocyanidins protectors against cisplatin-induced inflammatory and oxidative damage in the liver by suppressing TLR4/NF-κB (105). The proanthocyanidins not only downregulate gene expression of TLR4 and NF-κB and decrease levels of inflammatory factors such as IL-1β, IL-6 and TNF-α, but also decrease the production of OS-associated mediators (such as ROS and MDA) and upregulate antioxidants [such as heme oxygenase-1 (HO-1), glutathione (GSH), SOD and catalase] (105). Proanthocyanidins minimize the formation of MDA and its metabolites by inhibiting the LPO process (106); proanthocyanidins could protect the function of ECs and minimize risk of vascular complications in diabetes by decreasing LPO and inhibiting OS (107). Outcomes from numerous studies have demonstrated that proanthocyanidins mitigates photo-oxidative damage to prevent UV-induced melanoma and other types of skin cancer by regulating T cells and inhibiting MAPK/NF-κB signaling pathways (19,108,109). Furthermore, proanthocyanidins decrease LPS-stimulated inflammation and OS by inhibiting inflammatory cytokines (TNF-α and IL-6) and controlling MAPK/NF-κB signaling pathways (102,110,111). Jia et al (112) demonstrated that suppression of NF-κB by proanthocyanidins facilitates protect human lens epithelial B-3 cells from oxidative damage and inflammatory injury. Proanthocyanidins could reduce H2O2-induced OS damage and delay cataracts occurrence by inhibiting activation of NF-κB and MAPK pathways. In a previous study, proanthocyanidins were demonstrated to inhibit production of iNOS, which is key to lower NO levels, indicating the antioxidant ability of proanthocyanidins (110). Proanthocyanidins exerted a protective effect in the animal model of gingivitis with porphyromonas gingivalis infection via reducing the release of IL-1β, IL-6, ROS, NO and lipid peroxide, thereby inhibiting alveolar bone loss (91,113). Due to their potent antioxidant and anti-inflammatory capacities, proanthocyanidins enhance the function of VECs and maintain blood pressure stability by inhibiting inflammatory cytokine production (74). Proanthocyanidins protect VECs from OS damage through inhibiting NADPH oxidase activity and scavenging excess ROS (114,115). Pinna et al (116) demonstrated that proanthocyanidins prevent vascular endothelial damage in diabetic rats by serving as a strong ROS scavenger. Proanthocyanidins similarly attenuate OS and inflammatory responses by eliminating ROS and inhibiting NF-κB gene expression, thus decreasing capillary permeability and maintaining vascular homeostasis (117).

A number of clinical trials has verified that proanthocyanidins are a pharmacologically safe and effective agent without negative effects and are suitable for a range of populations, including pregnant people (118,119). Proanthocyanidins reliably relieve symptoms of patients that suffered from urinary tract infections or gingivitis (91,120). However, an in vivo study in female rats, reported that high-dose proanthocyanidin (>432 mg/kg) could cause delayed gastric emptying and pica behavior (121). At present, to the best of our knowledge, extensive research of proanthocyanidins has been performed in various disorders but reports regarding HSP are rare (91,105,109,122). Fig. 2 presents the molecular structure and multi-bioactivity of proanthocyanidins as well as their application in different types of disease.

4. Hypothesis of proanthocyanidins in the management of HSP

As immune imbalance and OS-induced IgA mediated vessel injury are implicated in HSP and proanthocyanidins have immunoregulatory, antioxidant and anti-inflammatory activity, it is hypothesized that proanthocyanidins may be beneficial to patients with HSP. HSP is a recurrent, immune-mediated vasculitis that invades arterioles and capillaries, typically featuring IgA deposition along vascular walls (23,45,46,123); immune imbalance and OS are responsible for HSP pathogenesis and involve signals from TLR4 and its downstream pathways MyD88/NF-κB (5,23,46,61,62); high proportions of Th2/Th1 as well as Th17/Treg and Th2 and Th17 overactivation are associated with increased ROS and redox imbalance in lesions and blood samples from patients with HSP; furthermore, Th2/Th17 promotion of Ig (primarily IgA) production and deposition are frequently observed in patients with HSP. The aforementioned factors stimulate and maintain the progression of HSP and, therefore may serve as potential targets for treatment of HSP (26,27,62,124). The aforementioned events are mediated by the TLR4/MyD88/NF-κB signaling pathway and a number of studies demonstrated high levels of TLR4 in patients with HSP accompanied by an increase in MyD88 and NF-κB, followed by release of inflammatory factors (such as IL-4, IL-6, IL-8, IL-17, IL-21 and LTB4) (5,25-29). Proanthocyanidins mitigate inflammatory and oxidative damage in inflammation/OS-associated disorder by inhibiting TLR4/MyD88/NF-κB pathways and inflammatory factors (IL-1β, IL-6 and TNF-α), scavenging NO, ROS and MDA and increasing release of HO-1, GSH, SOD and catalase (18,89,102,104,105,125). Proanthocyanidins improve vascular endothelial function, decrease vascular permeability and prevent vascular damage by inhibiting release of inflammatory factors, eliminating ROS and reducing the production of Igs and deposition of immune complexes (92,116,117,126). Thus, proanthocyanidins are currently applied in immune-mediated and vascular disorders to improving vascular endothelial function in hypertensive patients (92). The aforementioned hypothetical mechanisms of proanthocyanidins in treating HSP are based on inactivation of the TLR4/MyD88/NF-κB pathways, inhibition of OS, suppression of inflammation and balance of immunity (Fig. 3).

5. Clinical significance

As the most common form of systemic vasculitis in children, annual incidence rate is up to ~27/100,000(127). HSP presents as a skin impairment; multi-systemic involvement, which can be life threatening, is a research focus. Despite conventional medications to control HSP, such as anti-inflammatory drugs, corticosteroids, cytotoxic drugs and immunosuppressants (8,127,128). These measures are unsuitable for long-term use due to the high cost, transient efficacy and severe adverse effects, like long-term and large-scale use of glucocorticoids could cause infections and Cushing's syndrome; long-term use of cytotoxic drugs that could result in gastrointestinal reactions, hepatotoxicity, bone marrow suppression, reproductive toxicity, cardiotoxicity (12). Proanthocyanidins, as natural extracts from plants, have widespread sources and diverse bioactivity with minimal side effects and can be used by pregnant people (118). Therefore, proanthocyanidins are preferable options for treatment of HSP based on their immunoregulatory, antioxidant and anti-inflammatory properties and may prevent OS-induced damage and modulate immune balance by scavenging ROS, maintaining Th1/Th2 and Th17/Treg balance and decreasing the secretion of inflammatory factors by the inhibition of TLR4/MyD88/NF-κB signaling pathways (99,102,104). In addition, a number of factors may affect the efficacy of proanthocyanidins in treatment of patients with HSP, such as dose of medication, the duration of drug intervention, the quality of the experimental models (Models with insignificant pathological manifestations or molecular biological alterations could affect the judgement of the efficacy of proanthocyanidins), the physical status of patients and the presence of underlying pathological conditions.

6. Future research

Future research should evaluate the effects of proanthocyanidins on HSP-associated clinical manifestations, histopathological or immunopathological alterations, cytological and serological changes such as cutaneous or systemic symptoms, VEC swelling, fibrin degeneration/necrosis, inflammatory cell infiltration, IgA deposition along vascular walls, T/B cell quantities and inflammatory/OS-associated indicator levels, in the presence or absence of proanthocyanidins. The present study assumes that proanthocyanidins would be beneficial for HSP. Therefore, to verify this hypothesis and reveal the effectiveness of proanthocyanidins in the treatment of patients with HSP, animal experiments in vivo should be performed to evaluate the effects of proanthocyanidins in well-established HSP-like rats models (129). The present review describes the successful construction of HSP rat models based on the model research of Li et al (130). In the aforementioned study, Sprague Dawley rats were intraperitoneally injected with ovalbumin (OVA) emulsified solution (1:1 OVA solution mixed with Freund's Complete Adjuvant) once/week for 3 weeks. After 3 weeks, rats were injected with 1 ml 10 mg/ml OVA saline via the tail vein, and intradermal injection of 1 ml 0.3% saline to stimulate hypersensitivity type III; finally, a rat model of HSP was successfully built. The preliminary results indicated the skin of the abdomen and tail of the rats visually presented with scattered petechiae and ecchymosis; histopathology of the skin revealed subcutaneous hemorrhage and inflammatory infiltration; renal histopathology revealed mesangial cell and matrix proliferation and immunofluorescence of renal tissue exhibited granular deposition of IgA immune complexes in the mesangial area (Fig. 4) (129). Additionally, in vitro studies using HSP-like cell models should be performed to investigate the mechanism of proanthocyanidins in HSP. Furthermore, updated biological techniques should be used to determine the associated parameters. In addition, randomized placebo-controlled clinical trials may establish a scientific basis for further research and clinical application of proanthocyanidins.

7. Conclusion

HSP, an IgA-mediated vasculitis, commonly affects children, affects the skin and systemic organs and threatens life. The etiology and pathogenesis of HSP are unclear but currently it is hypothesized to be a multifactorial disorder involving immune imbalance, OS, infection, genes, foods, drugs and complex cytokine networks (23,27,124). Immune dysregulation and OS that induce IgA-mediated vascular injury are responsible for HSP and involve abnormal activation of TLR4/MyD88/NF-κB signaling. Thus, targeting these areas could lead to a treatment for patients with HSP. Treatments, such as targeting key inflammatory mediators, pathways or cells, have exhibited positive effects on HSP (5,131). Typically, corticosteroids have greater efficacy on severe cutaneous and visceral involvement such as gastrointestinal or kidney impairment by suppressing T/B cell activation, antibody production and inflammatory factor release (132-134); for example, patients with severe purpura nephritis are treated with methylprednisolone pulse therapy that effectively reduces kidney pathological injury and shortens treatment course (135). However, glucocorticoids merely ease the symptoms, while excessive use of glucocorticoids may lead to gastrointestinal bleeding or electrolyte disturbance (136,137). Furthermore, because of susceptibility to resistance to glucocorticoid treatment alone, the combination of glucocorticoids with an immunosuppressant tends to be more effective and safer, especially for patients with severe HSP nephritis (137,138); immunosuppressive agents such as mycophenolate mofetil act against purpura nephritis via selectively suppressing T/B lymphocytes (139). Additionally, plasma exchange has a notable effect on patients with recurrent or acute HSP by improving renal function and clearing immune complexes (140). Despite these treatments having efficacy against HSP, adverse reactions, short-term efficacy and high cost prevent widespread use. Therefore, safe, effective and cheaper treatments are needed for HSP. The present study explained the pathogenesis of HSP and applications of proanthocyanidins in clinical practice. Proanthocyanidins have potential as therapy of HSP, although factors, such as the dose of drug, the duration of drug intervention, quality of experimental models and patient's physical condition may affect proanthocyanidins for HSP treatment. Thus, the aforementioned factors should be investigated in future research. Additionally, studies in vitro and in vivo should be performed to verify the signaling pathways in HSP and to identify improvements in diagnostic biomarkers following proanthocyanidin treatment.

Acknowledgements

The authors would like to thank Professor Xia Xiong and Professor Yongqiong Deng (Department of Neurobiology, Southwest Medical University, Luzhou, China) for their valuable suggestions.

Funding

Funding: The present study was supported by the Healthcare for Cadre Research Project of Sichuan Province (grant no. 2020-1501) supported by the Sichuan Provincial Government.

Availability of data and materials

Not applicable.

Authors' contributions

QD revised the manuscript. MG conceived the study. XL and YX wrote the manuscript. YX, JZ and DX performed the literature review. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Wang K, Sun X, Cao Y, Dai L, Sun F, Yu P and Dong L: Risk factors for renal involvement and severe kidney disease in 2731 Chinese children with Henoch-Schönlein purpura: A retrospective study. Medicine (Baltimore). 97(e12520)2018.PubMed/NCBI View Article : Google Scholar

2 

Chimenz R, Cannavò L, Spinuzza A, Fede C, Cucinotta U, Pensabene L, Betta P, Gitto E, Concolino D and Cuppari C: Unusual presentation of Henoch-Schönlein purpura. J Biol Regul Homeost Agents. 33 (5 Suppl 1):S69–S74. 2019.PubMed/NCBI

3 

Gómez S, Pérez M, Pellegrini M, Isern E, Quintana C, Artacho P, Bertolini M, Pomerantz B and Gadda N: Henoch-Schonlein purpura in pediatrics: Ten years of experience at a moderate risk office of a general hospital. Arch Argent Pediatr. 118:31–37. 2020.PubMed/NCBI View Article : Google Scholar : (In English, Spanish).

4 

Yang HR: What we know about Henoch-Schönlein purpura in children up to date? J Korean Med Sci. 33(e199)2018.PubMed/NCBI View Article : Google Scholar

5 

Zhu Y, Dong Y, Wu L and Deng F: Changes of inflammatory mediators and oxidative stress indicators in children with Henoch-Schönlein purpura and clinical effects of hemoperfusion in the treatment of severe Henoch-Schönlein purpura with gastrointestinal involvement in children. BMC Pediatr. 19(409)2019.PubMed/NCBI View Article : Google Scholar

6 

Zhang N, Guo PJ, Liu PL, Yang HR, Xiao J, Li XP, Huang JB and Zheng YZ: Comparison of age-based clinical and abnormal immune parameters in patients with Henoch-Schönlein purpura. Zhonghua Xue Ye Xue Za Zhi. 38:60–64. 2017.PubMed/NCBI View Article : Google Scholar : (In Chinese).

7 

Niknam N, Ha L and Gautam-Goyal P: Adult onset immunoglobulin A vasculitis (Henoch-Schonlein purpura) with alveolar hemorrhage. IDCases. 12:47–48. 2018.PubMed/NCBI View Article : Google Scholar

8 

Tan J, Tang Y, Zhong Z, Yan S, Tan L, Tarun P and Qin W: The efficacy and safety of immunosuppressive agents plus steroids compared with steroids alone in the treatment of Henoch-Schönlein purpura nephritis: A meta-analysis. Int Urol Nephrol. 51:975–985. 2019.PubMed/NCBI View Article : Google Scholar

9 

Roman C, Dima B, Muyshont L, Schurmans T and Gilliaux O: Indications and efficiency of dapsone in IgA vasculitis (Henoch-Schonlein purpura): Case series and a review of the literature. Eur J Pediatr. 178:1275–1281. 2019.PubMed/NCBI View Article : Google Scholar

10 

Vonend C, Rifkin SI, Baliga RS and Weinstein SS: Henoch-Schönlein purpura and recurrent renal failure. Ren Fail. 32:888–891. 2010.PubMed/NCBI View Article : Google Scholar

11 

Oni L and Sampath S: Childhood IgA vasculitis (Henoch Schonlein Purpura)-advances and knowledge gaps. Front Pediatr. 7(257)2019.PubMed/NCBI View Article : Google Scholar

12 

Fan L, Yan H, Zhen X, Wu X, Hao J, Hou L and Han L: Safety and efficacy evaluation of traditional chinese medicine (Qingre-Lishi-Yishen Formula) based on treatment of regular glucocorticoid combined with cyclophosphamide pulse in children suffered from moderately Severe Henoch-Schonlein Purpura nephritis with nephrotic proteinuria. Evid Based Complement Alternat Med. 2020(3920735)2020.PubMed/NCBI View Article : Google Scholar

13 

Nichols JA and Katiyar SK: Skin photoprotection by natural polyphenols: Anti-inflammatory, antioxidant and DNA repair mechanisms. Arch Dermatol Res. 302:71–83. 2010.PubMed/NCBI View Article : Google Scholar

14 

Tong H, Song X, Sun X, Sun G and Du F: Immunomodulatory and antitumor activities of grape seed proanthocyanidins. J Agric Food Chem. 59:11543–1157. 2011.PubMed/NCBI View Article : Google Scholar

15 

Huang S, Yang N, Liu Y, Hu L, Zhao J, Gao J, Li Y, Li C, Zhang X and Huang T: Grape seed proanthocyanidins inhibit angiogenesis via the downregulation of both vascular endothelial growth factor and angiopoietin signaling. Nutr Res. 32:530–536. 2012.PubMed/NCBI View Article : Google Scholar

16 

Wen W, Lu J, Zhang K and Chen S: Grape seed extract inhibits angiogenesis via suppression of the vascular endothelial growth factor receptor signaling pathway. Cancer Prev Res (Phila). 1:554–561. 2008.PubMed/NCBI View Article : Google Scholar

17 

Wu L, Huang Z, Qin P, Yao Y, Meng X, Zou J, Zhu K and Ren G: Chemical characterization of a procyanidin-rich extract from sorghum bran and its effect on oxidative stress and tumor inhibition in vivo. J Agric Food Chem. 59:8609–8615. 2011.PubMed/NCBI View Article : Google Scholar

18 

Yang L, Xian D, Xiong X, Lai R, Song J and Zhong J: Proanthocyanidins against oxidative stress: From molecular mechanisms to clinical applications. Biomed Res Int. 2018(8584136)2018.PubMed/NCBI View Article : Google Scholar

19 

Mantena SK and Katiyar SK: Grape seed proanthocyanidins inhibit UV-radiation-induced oxidative stress and activation of MAPK and NF-kappaB signaling in human epidermal keratinocytes. Free Radic Biol Med. 40:1603–1614. 2006.PubMed/NCBI View Article : Google Scholar

20 

Koudoufio M, Feldman F, Ahmarani L, Delvin E, Spahis S, Desjardins Y and Levy E: Intestinal protection by proanthocyanidins involves anti-oxidative and anti-inflammatory actions in association with an improvement of insulin sensitivity, lipid and glucose homeostasis. Sci Rep. 11(3878)2021.PubMed/NCBI View Article : Google Scholar

21 

Baldivia DDS, Leite DF, Castro DTH, Campos JF, Santos UPD, Paredes-Gamero EJ, Carollo CA, Silva DB, de Picoli Souza K and Dos Santos EL: Evaluation of in vitro antioxidant and anticancer properties of the aqueous extract from the stem bark of stryphnodendron adstringens. Int J Mol Sci. 19(2432)2018.PubMed/NCBI View Article : Google Scholar

22 

Alsharairi NA: Insights into the mechanisms of action of proanthocyanidins and anthocyanins in the treatment of nicotine-induced non-small cell lung cancer. Int J Mol Sci. 23(7905)2022.PubMed/NCBI View Article : Google Scholar

23 

Lau KK, Suzuki H, Novak J and Wyatt RJ: Pathogenesis of Henoch-Schönlein purpura nephritis. Pediatr Nephrol. 25:19–26. 2010.PubMed/NCBI View Article : Google Scholar

24 

Keskin N, Civilibal M, Elevli M, Koldas M, Duru NS and Ozturk H: Elevated plasma advanced oxidation protein products in children with Henoch-Schonlein purpura. Pediatr Nephrol. 26:1989–1993. 2011.PubMed/NCBI View Article : Google Scholar

25 

Xu H, Jiang G, Shen H, Li W, Mao J and Pan Y: Association of TLR4 gene polymorphisms with childhood Henoch-Schönlein purpura in a Chinese population. Rheumatol Int. 37:1909–1915. 2017.PubMed/NCBI View Article : Google Scholar

26 

Canpinar H, Ozaltin F, Bilginer Y, Bakkaloğlu A and Ozen S: Toll-like receptors 2 and 4 cell surface expression reflects endotoxin tolerance in Henoch-Schönlein purpura. Turk J Pediatr. 52:22–27. 2010.PubMed/NCBI

27 

Donadio ME, Loiacono E, Peruzzi L, Amore A, Camilla R, Chiale F, Vergano L, Boido A, Conrieri M, Bianciotto M, et al: Toll-like receptors, immunoproteasome and regulatory T cells in children with Henoch-Schönlein purpura and primary IgA nephropathy. Pediatr Nephrol. 29:1545–1551. 2014.PubMed/NCBI View Article : Google Scholar

28 

Chang H, Zhang QY, Lin Y, Cheng N and Zhang SQ: Correlation of TLR2 and TLR4 expressions in peripheral blood mononuclear cells to Th1- and Th2-type immune responses in children with Henoch-Schönlein Purpura. Int J Clin Exp Med. 8:13532–13539. 2015.PubMed/NCBI

29 

Chang H, Yu DS, Liu XQ, Zhang QY, Cheng N, Zhang SQ and Qu ZH: Clinical significance of TLR3 and TLR4 in peripheral blood mononuclear cells from children with Henoch-Schönlein purpura nephritis. Exp Ther Med. 7:1703–1707. 2014.PubMed/NCBI View Article : Google Scholar

30 

Zhou L, Xu F, Chang C, Tao Y, Song L and Li X: Interleukin-17-producing CD4+ T lymphocytes are increased in patients with primary immune thrombocytopenia. Blood Coagul Fibrinolysis. 27:301–307. 2016.PubMed/NCBI View Article : Google Scholar

31 

Barnabei L, Laplantine E, Mbongo W, Rieux-Laucat F and Weil R: NF-κB: At the borders of autoimmunity and inflammation. Front Immunol. 12(716469)2021.PubMed/NCBI View Article : Google Scholar

32 

Yu H, Lin L, Zhang Z, Zhang H and Hu H: Targeting NF-κB pathway for the therapy of diseases: Mechanism and clinical study. Signal Transduct Target Ther. 5(209)2020.PubMed/NCBI View Article : Google Scholar

33 

Oh H and Ghosh S: NF-κB: Roles and regulation in different CD4(+) T-cell subsets. Immunol Rev. 252:41–51. 2013.PubMed/NCBI View Article : Google Scholar

34 

Pandiyan P and Zhu J: Origin and functions of pro-inflammatory cytokine producing Foxp3+ regulatory T cells. Cytokine. 76:13–24. 2015.PubMed/NCBI View Article : Google Scholar

35 

Chang H, Lin Y, Lei K, Wang F, Zhang QQ and Zhang QY: Role of hypomethylation of suppressor of cytokine signaling in T helper 17 cell/regulatory T cell imbalance in children with Henoch-Schönlein purpura. Zhongguo Dang Dai Er Ke Za Zhi. 21:38–44. 2019.PubMed/NCBI View Article : Google Scholar : (In Chinese).

36 

Li-Weber M, Giaisi M, Baumann S, Pálfi K and Krammer PH: NF-kappa B synergizes with NF-AT and NF-IL6 in activation of the IL-4 gene in T cells. Eur J Immunol. 34:1111–1118. 2004.PubMed/NCBI View Article : Google Scholar

37 

Li YY, Li CR, Wang GB, Yang J and Zu Y: Investigation of the change in CD4+ T cell subset in children with Henoch-Schonlein purpura. Rheumatol Int. 32:3785–3792. 2012.PubMed/NCBI View Article : Google Scholar

38 

Yang J, Li CR, Zu Y, Wang GB and Li YB: Role of regulatory T cells in pathogenesis of Henoch-Schonlein purpura in children. Zhonghua Er Ke Za Zhi. 44:411–414. 2006.PubMed/NCBI(In Chinese).

39 

Gujer C, Sundling C, Seder RA, Karlsson Hedestam GB and Loré K: Human and rhesus plasmacytoid dendritic cell and B-cell responses to Toll-like receptor stimulation. Immunology. 134:257–269. 2011.PubMed/NCBI View Article : Google Scholar

40 

Ohue Y and Nishikawa H: Regulatory T (Treg) cells in cancer: Can Treg cells be a new therapeutic target? Cancer Sci. 110:2080–2089. 2019.PubMed/NCBI View Article : Google Scholar

41 

Adeegbe DO and Nishikawa H: Natural and induced T regulatory cells in cancer. Front Immunol. 4(190)2013.PubMed/NCBI View Article : Google Scholar

42 

Weng L, Cao X, Han L, Zhao H, Qiu S, Yan Y, Wang X, Chen X, Zheng W, Xu X, et al: Association of increased Treg and Th17 with pathogenesis of moyamoya disease. Sci Rep. 7(3071)2017.PubMed/NCBI View Article : Google Scholar

43 

Wang CM, Luo Y, Wang YC and Sheng GY: Roles of follicular helper T cells and follicular regulatory T cells in pathogenesis of Henoch-Schönlein purpura in children. Zhongguo Dang Dai Er Ke Za Zhi. 17:1084–1087. 2015.PubMed/NCBI(In Chinese).

44 

Yang YH, Lai HJ, Huang CM, Wang LC, Lin YT and Chiang BL: Sera from children with active Henoch-Schönlein purpura can enhance the production of interleukin 8 by human umbilical venous endothelial cells. Ann Rheum Dis. 63:1511–1513. 2004.PubMed/NCBI View Article : Google Scholar

45 

Yang YH, Huang YH, Lin YL, Wang LC, Chuang YH, Yu HH, Lin YT and Chiang BL: Circulating IgA from acute stage of childhood Henoch-Schönlein purpura can enhance endothelial interleukin (IL)-8 production through MEK/ERK signalling pathway. Clin Exp Immunol. 144:247–253. 2006.PubMed/NCBI View Article : Google Scholar

46 

Heineke MH, Ballering AV, Jamin A, Ben Mkaddem S, Monteiro RC and Van Egmond M: New insights in the pathogenesis of immunoglobulin A vasculitis (Henoch-Schönlein purpura). Autoimmun Rev. 16:1246–1253. 2017.PubMed/NCBI View Article : Google Scholar

47 

Wu JJ, Zhu YT and Hu YM: Mechanism of feedback regulation of neutrophil inflammation in Henoch-Schönlein purpura. Eur Rev Med Pharmacol Sci. 20:4277–4285. 2016.PubMed/NCBI

48 

Ray PD, Huang BW and Tsuji Y: Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 24:981–990. 2012.PubMed/NCBI View Article : Google Scholar

49 

Schieber M and Chandel NS: ROS function in redox signaling and oxidative stress. Curr Biol. 24:R453–R462. 2014.PubMed/NCBI View Article : Google Scholar

50 

Cesselli D, Aleksova A, Sponga S, Cervellin C, Di Loreto C, Tell G and Beltrami AP: Cardiac cell senescence and redox signaling. Front Cardiovasc Med. 4(38)2017.PubMed/NCBI View Article : Google Scholar

51 

Ciccarese F, Raimondi V, Sharova E, Silic-Benussi M and Ciminale V: Nanoparticles as tools to target redox homeostasis in cancer cells. Antioxidants (Basel). 9(211)2020.PubMed/NCBI View Article : Google Scholar

52 

Nakai K and Tsuruta D: What are reactive oxygen species, free radicals, and oxidative stress in skin diseases? Int J Mol Sci. 22(10799)2021.PubMed/NCBI View Article : Google Scholar

53 

Sharifi-Rad M, Anil Kumar NV, Zucca P, Varoni EM, Dini L, Panzarini E, Rajkovic J, Tsouh Fokou PV, Azzini E, Peluso I, et al: Lifestyle, oxidative stress, and antioxidants: Back and forth in the pathophysiology of chronic diseases. Front Physiol. 11(694)2020.PubMed/NCBI View Article : Google Scholar

54 

Li Y and Pagano PJ: Microvascular NADPH oxidase in health and disease. Free Radic Biol Med. 109:33–47. 2017.PubMed/NCBI View Article : Google Scholar

55 

Schröder K: NADPH oxidases: Current aspects and tools. Redox Biol. 34(101512)2020.PubMed/NCBI View Article : Google Scholar

56 

Murphy MP: How mitochondria produce reactive oxygen species. Biochem J. 417:1–13. 2009.PubMed/NCBI View Article : Google Scholar

57 

Arazi H, Eghbali E and Suzuki K: Creatine supplementation, physical exercise and oxidative stress markers: A review of the mechanisms and effectiveness. Nutrients. 13(869)2021.PubMed/NCBI View Article : Google Scholar

58 

Kim YW, West XZ and Byzova TV: Inflammation and oxidative stress in angiogenesis and vascular disease. J Mol Med (Berl). 91:323–328. 2013.PubMed/NCBI View Article : Google Scholar

59 

Salazar G: NADPH Oxidases and mitochondria in vascular senescence. Int J Mol Sci. 19(1327)2018.PubMed/NCBI View Article : Google Scholar

60 

Zhu Y, Dong Y, Xu DL, Jiang JY, Wu L, Ke RJ, Fang SH and Peng Y: Clinical effect and mechanism of hemoperfusion in treatment of children with severe abdominal Henoch-Schönlein purpura. Zhongguo Dang Dai Er Ke Za Zhi. 20:378–382. 2018.PubMed/NCBI View Article : Google Scholar : (In Chinese).

61 

Gurses D, Parlaz N, Bor-Kucukatay M, Kucukatay V and Erken G: Evaluation of oxidative stress and erythrocyte properties in children with henoch-shoenlein purpura. Iran J Pediatr. 24:166–172. 2014.PubMed/NCBI

62 

Ece A, Kelekçi S, Kocamaz H, Hekimoğlu A, Balik H, Yolbaş I and Erel O: Antioxidant enzyme activities, lipid peroxidation, and total antioxidant status in children with Henoch-Schönlein purpura. Clin Rheumatol. 27:163–169. 2008.PubMed/NCBI View Article : Google Scholar

63 

Higashi Y, Maruhashi T, Noma K and Kihara Y: Oxidative stress and endothelial dysfunction: Clinical evidence and therapeutic implications. Trends Cardiovasc Med. 24:165–169. 2014.PubMed/NCBI View Article : Google Scholar

64 

Mai J, Virtue A, Shen J, Wang H and Yang XF: An evolving new paradigm: Endothelial cells-conditional innate immune cells. J Hematol Oncol. 6(61)2013.PubMed/NCBI View Article : Google Scholar

65 

Hou X, Yang S and Yin J: Blocking the REDD1/TXNIP axis ameliorates LPS-induced vascular endothelial cell injury through repressing oxidative stress and apoptosis. Am J Physiol Cell Physiol. 316:C104–C110. 2019.PubMed/NCBI View Article : Google Scholar

66 

Wu H, Wen Y, Yue C, Li X and Gao R: Serum TNF-α level is associated with disease severity in adult patients with immunoglobulin a vasculitis nephritis. Dis Markers. 2020(5514145)2020.PubMed/NCBI View Article : Google Scholar

67 

Yuan P, Bo Y, Ming G, Wen-Jun F, Qin Z and Bo H: Apoptosis of human umbilical vein endothelial cells (HUVEC) induced by IgA1 isolated from Henoch-Schonlein purpura children. Asian Pac J Allergy Immunol. 32:34–38. 2014.PubMed/NCBI View Article : Google Scholar

68 

Montezano AC and Touyz RM: Reactive oxygen species and endothelial function-role of nitric oxide synthase uncoupling and Nox family nicotinamide adenine dinucleotide phosphate oxidases. Basic Clin Pharmacol Toxicol. 110:87–94. 2012.PubMed/NCBI View Article : Google Scholar

69 

Elalfy MS, Elhenawy YI, Deifalla S, Hegazy M, Sabra A and Abdelaziz Y: Oxidant/antioxidant status in children and adolescents with immune thrombocytopenia (ITP) and the role of an adjuvant antioxidant therapy. Pediatr Blood Cancer. 62:830–837. 2015.PubMed/NCBI View Article : Google Scholar

70 

Carta S, Semino C, Sitia R and Rubartelli A: Dysregulated IL-1β secretion in autoinflammatory diseases: A matter of stress? Front Immunol. 8(345)2017.PubMed/NCBI View Article : Google Scholar

71 

Noval Rivas M, Wakita D, Franklin MK, Carvalho TT, Abolhesn A, Gomez AC, Fishbein MC, Chen S, Lehman TJ, Sato K, et al: Intestinal permeability and IgA provoke immune vasculitis linked to cardiovascular inflammation. Immunity. 51:508–521.e6. 2019.PubMed/NCBI View Article : Google Scholar

72 

Sugino H, Sawada Y and Nakamura M: IgA Vasculitis: Etiology, treatment, biomarkers and epigenetic changes. Int J Mol Sci. 22(7538)2021.PubMed/NCBI View Article : Google Scholar

73 

Lavieri R, Piccioli P, Carta S, Delfino L, Castellani P and Rubartelli A: TLR costimulation causes oxidative stress with unbalance of proinflammatory and anti-inflammatory cytokine production. J Immunol. 192:5373–5381. 2014.PubMed/NCBI View Article : Google Scholar

74 

Gong L, Lei Y, Liu Y, Tan F, Li S, Wang X, Xu M, Cai W, Du B, Xu F, et al: Vaccarin prevents ox-LDL-induced HUVEC EndMT, inflammation and apoptosis by suppressing ROS/p38 MAPK signaling. Am J Transl Res. 11:2140–2154. 2019.PubMed/NCBI

75 

Duan T, Du Y, Xing C, Wang HY and Wang RF: Toll-like receptor signaling and its role in cell-mediated immunity. Front Immunol. 13(812774)2022.PubMed/NCBI View Article : Google Scholar

76 

Aluri J, Cooper MA and Schuettpelz LG: Toll-Like receptor signaling in the establishment and function of the immune system. Cells. 10(1374)2021.PubMed/NCBI View Article : Google Scholar

77 

Vijay K: Toll-like receptors in immunity and inflammatory diseases: Past, present, and future. Int Immunopharmacol. 59:391–412. 2018.PubMed/NCBI View Article : Google Scholar

78 

Kuzmich NN, Sivak KV, Chubarev VN, Porozov YB, Savateeva-Lyubimova TN and Peri F: TLR4 signaling pathway modulators as potential therapeutics in inflammation and sepsis. Vaccines (Basel). 5(34)2017.PubMed/NCBI View Article : Google Scholar

79 

Yao C, Oh JH, Lee DH, Bae JS, Jin CL, Park CH and Chung JH: Toll-like receptor family members in skin fibroblasts are functional and have a higher expression compared to skin keratinocytes. Int J Mol Med. 35:1443–1450. 2015.PubMed/NCBI View Article : Google Scholar

80 

Krishnan JS, KSelvarajoo K, Tsuchiya M, Lee G and Choi S: Toll-like receptor signal transduction. Exp Mol Med. 39:421–438. 2007.PubMed/NCBI View Article : Google Scholar

81 

Rong Z, Huang Y, Cai H, Chen M, Wang H, Liu G, Zhang Z and Wu J: Gut microbiota disorders promote inflammation and aggravate spinal cord injury through the TLR4/MyD88 signaling pathway. Front Nutr. 8(702659)2021.PubMed/NCBI View Article : Google Scholar

82 

Yamamoto M, Sato S, Hemmi H, Uematsu S, Hoshino K, Kaisho T, Takeuchi O, Takeda K and Akira S: TRAM is specifically involved in the Toll-like receptor 4-mediated MyD88-independent signaling pathway. Nat Immunol. 4:1144–1150. 2003.PubMed/NCBI View Article : Google Scholar

83 

Mills KH: TLR-dependent T cell activation in autoimmunity. Nat Rev Immunol. 11:807–822. 2011.PubMed/NCBI View Article : Google Scholar

84 

Park HS, Jung HY, Park EY, Kim J, Lee WJ and Bae YS: Cutting edge: Direct interaction of TLR4 with NAD(P)H oxidase 4 isozyme is essential for lipopolysaccharide-induced production of reactive oxygen species and activation of NF-kappa B. J Immunol. 173:3589–3593. 2004.PubMed/NCBI View Article : Google Scholar

85 

Gill R, Tsung A and Billiar T: Linking oxidative stress to inflammation: Toll-like receptors. Free Radic Biol Med. 48:1121–1132. 2010.PubMed/NCBI View Article : Google Scholar

86 

Rauf A, Imran M, Abu-Izneid T, Iahtisham-Ul-Haq Patel S, Pan X, Naz S, Sanches Silva A, Saeed F and Rasul Suleria HA: Proanthocyanidins: A comprehensive review. Biomed Pharmacother. 116(108999)2019.PubMed/NCBI View Article : Google Scholar

87 

Bensa M, Glavnik V and Vovk I: Leaves of Invasive Plants-Japanese, bohemian and giant knotweed-the promising new nource of Flavan-3-ols and proanthocyanidins. Plants (Basel). 9(118)2020.PubMed/NCBI View Article : Google Scholar

88 

González-Quilen C, Rodríguez-Gallego E, Beltrán-Debón R, Pinent M, Ardévol A, Blay MT and Terra X: Health-promoting properties of proanthocyanidins for intestinal dysfunction. Nutrients. 12(130)2020.PubMed/NCBI View Article : Google Scholar

89 

Li S, Xu M, Niu Q, Xu S, Ding Y, Yan Y, Guo S and Li F: Efficacy of procyanidins against in vivo cellular oxidative damage: A systematic review and meta-analysis. PLoS One. 10(e0139455)2015.PubMed/NCBI View Article : Google Scholar

90 

Ravindranathan P, Pasham D, Balaji U, Cardenas J, Gu J, Toden S and Goel A: Mechanistic insights into anticancer properties of oligomeric proanthocyanidins from grape seeds in colorectal cancer. Carcinogenesis. 39:767–777. 2018.PubMed/NCBI View Article : Google Scholar

91 

Nawrot-Hadzik I, Matkowski A, Kubasiewicz-Ross P and Hadzik J: Proanthocyanidins and Flavan-3-ols in the prevention and treatment of periodontitis-immunomodulatory effects, animal and clinical Studies. Nutrients. 13(239)2021.PubMed/NCBI View Article : Google Scholar

92 

Odai T, Terauchi M, Kato K, Hirose A and Miyasaka N: Effects of grape seed proanthocyanidin extract on vascular endothelial function in participants with prehypertension: A randomized, double-blind, placebo-controlled study. Nutrients. 11(2844)2019.PubMed/NCBI View Article : Google Scholar

93 

Zhang R, Kang X, Liu L, Wang X, Li H, Zhu J, Cao Y and Zhu H: Gut microbiota modulation by plant polyphenols in koi carp (Cyprinus carpio L.). Front Microbiol. 13(977292)2022.PubMed/NCBI View Article : Google Scholar

94 

Suo H, Tian R, Li J, Zhang S, Cui Y, Li L and Sun B: Compositional characterization study on high-molecular-mass polymeric polyphenols in red wines by chemical degradation. Food Res Int. 123:440–449. 2019.PubMed/NCBI View Article : Google Scholar

95 

Yuan L, Wang Q, Zhang S and Zhang L: Correlation between serum inflammatory factors TNF-α, IL-8, IL-10 and Henoch-Schonlein purpura with renal function impairment. Exp Ther Med. 15:3924–3928. 2018.PubMed/NCBI View Article : Google Scholar

96 

Park MK, Park JS, Cho ML, Oh HJ, Heo YJ, Woo YJ, Heo YM, Park MJ, Park HS, Park SH, et al: Grape seed proanthocyanidin extract (GSPE) differentially regulates Foxp3(+) regulatory and IL-17(+) pathogenic T cell in autoimmune arthritis. Immunol Lett. 135:50–58. 2011.PubMed/NCBI View Article : Google Scholar

97 

Jhun JY, Moon SJ, Yoon BY, Byun JK, Kim EK, Yang EJ, Ju JH, Hong YS, Min JK, Park SH, et al: Grape seed proanthocyanidin extract-mediated regulation of STAT3 proteins contributes to Treg differentiation and attenuates inflammation in a murine model of obesity-associated arthritis. PLoS One. 8(e78843)2013.PubMed/NCBI View Article : Google Scholar

98 

Park IJ, Cha SY, Kang M, So YS, Go HG, Mun SP, Ryu KS and Jang HK: Effect of proanthocyanidin-rich extract from Pinus radiata bark on immune response of specific-pathogen-free White Leghorn chickens. Poult Sci. 90:977–982. 2011.PubMed/NCBI View Article : Google Scholar

99 

Min HK, Kim SM, Baek SY, Woo JW, Park JS, Cho ML, Lee J, Kwok SK, Kim SW and Park SH: Anthocyanin extracted from black soybean seed coats prevents autoimmune arthritis by suppressing the development of Th17 cells and synthesis of proinflammatory cytokines by such cells, via inhibition of NF-κB. PLoS One. 10(e0138201)2015.PubMed/NCBI View Article : Google Scholar

100 

Takano F, Takata T, Yoshihara A, Nakamura Y, Arima Y and Ohta T: Aqueous extract of peanut skin and its main constituent procyanidin A1 suppress serum IgE and IgG1 levels in mice-immunized with ovalbumin. Biol Pharm Bull. 30:922–927. 2007.PubMed/NCBI View Article : Google Scholar

101 

Long M, Yang SH, Han JX, Li P, Zhang Y, Dong S, Chen X, Guo J, Wang J and He JB: The protective effect of grape-seed proanthocyanidin extract on oxidative damage induced by zearalenone in kunming mice liver. Int J Mol Sci. 17(808)2016.PubMed/NCBI View Article : Google Scholar

102 

Han S, Gao H, Chen S, Wang Q, Li X, Du LJ, Li J, Luo YY, Li JX, Zhao LC, et al: Procyanidin A1 Alleviates Inflammatory Response induced by LPS through NF-κB, MAPK, and Nrf2/HO-1 Pathways in RAW264.7 cells. Sci Rep. 9(15087)2019.PubMed/NCBI View Article : Google Scholar

103 

Katiyar SK: Proanthocyanidins from grape seeds inhibit UV-radiation-induced immune suppression in mice: Detection and analysis of molecular and cellular targets. Photochem Photobiol. 91:156–162. 2015.PubMed/NCBI View Article : Google Scholar

104 

Kim SH, Bang J, Son CN, Baek WK and Kim JM: Grape seed proanthocyanidin extract ameliorates murine autoimmune arthritis through regulation of TLR4/MyD88/NF-κB signaling pathway. Korean J Intern Med. 33:612–621. 2018.PubMed/NCBI View Article : Google Scholar

105 

El-Shitany NA and Eid B: Proanthocyanidin protects against cisplatin-induced oxidative liver damage through inhibition of inflammation and NF-κβ/TLR-4 pathway. Environ Toxicol. 32:1952–1963. 2017.PubMed/NCBI View Article : Google Scholar

106 

Mittal A, Elmets CA and Katiyar SK: Dietary feeding of proanthocyanidins from grape seeds prevents photocarcinogenesis in SKH-1 hairless mice: Relationship to decreased fat and lipid peroxidation. Carcinogenesis. 24:1379–1388. 2003.PubMed/NCBI View Article : Google Scholar

107 

Rodríguez RM, Colom-Pellicer M, Blanco J, Calvo E, Aragonès G and Mulero M: Grape-Seed procyanidin extract (GSPE) Seasonal-Dependent modulation of glucose and lipid metabolism in the liver of healthy F344 rats. Biomolecules. 12(839)2022.PubMed/NCBI View Article : Google Scholar

108 

Sharma SD, Meeran SM and Katiyar SK: Dietary grape seed proanthocyanidins inhibit UVB-induced oxidative stress and activation of mitogen-activated protein kinases and nuclear factor-kappaB signaling in in vivo SKH-1 hairless mice. Mol Cancer Ther. 6:995–1005. 2007.PubMed/NCBI View Article : Google Scholar

109 

Divya SP, Wang X, Pratheeshkumar P, Son YO, Roy RV, Kim D, Dai J, Hitron JA, Wang L, Asha P, et al: Blackberry extract inhibits UVB-induced oxidative damage and inflammation through MAP kinases and NF-κB signaling pathways in SKH-1 mice skin. Toxicol Appl Pharmacol. 284:92–99. 2015.PubMed/NCBI View Article : Google Scholar

110 

Ma X, Wang R, Yu S, Lu G, Yu Y and Jiang C: Anti-Inflammatory activity of oligomeric proanthocyanidins via inhibition of NF-κB and MAPK in LPS-Stimulated MAC-T Cells. J Microbiol Biotechnol. 30:1458–1466. 2020.PubMed/NCBI View Article : Google Scholar

111 

Rajput SA, Sun L, Zhang NY, Khalil MM, Ling Z, Chong L, Wang S, Rajput IR, Bloch DM, Khan FA, et al: Grape seed proanthocyanidin extract alleviates aflatoxin B1-induced immunotoxicity and oxidative stress via modulation of NF-κB and Nrf2 signaling pathways in broilers. Toxins (Basel). 11(23)2019.PubMed/NCBI View Article : Google Scholar

112 

Jia Z, Song Z, Zhao Y, Wang X and Liu P: Grape seed proanthocyanidin extract protects human lens epithelial cells from oxidative stress via reducing NF-кB and MAPK protein expression. Mol Vis. 17:210–217. 2011.PubMed/NCBI

113 

Schmuch J, Beckert S, Brandt S, Löhr G, Hermann F, Schmidt TJ, Beikler T and Hensel A: Extract from Rumex acetosa L. for prophylaxis of periodontitis: Inhibition of bacterial in vitro adhesion and of gingipains of Porphyromonas gingivalis by epicatechin-3-O-(4β→8)-epicatechin-3-O-gallate (procyanidin-B2-Di-gallate). PLoS One. 10(e0120130)2015.PubMed/NCBI View Article : Google Scholar

114 

Steffen Y, Gruber C, Schewe T and Sies H: Mono-O-methylated flavanols and other flavonoids as inhibitors of endothelial NADPH oxidase. Arch Biochem Biophys. 469:209–219. 2008.PubMed/NCBI View Article : Google Scholar

115 

Álvarez E, Rodiño-Janeiro BK, Jerez M, Ucieda-Somoza R, Núñez MJ and González-Juanatey JR: Procyanidins from grape pomace are suitable inhibitors of human endothelial NADPH oxidase. J Cell Biochem. 113:1386–1396. 2012.PubMed/NCBI View Article : Google Scholar

116 

Pinna C, Morazzoni P and Sala A: Proanthocyanidins from Vitis vinifera inhibit oxidative stress-induced vascular impairment in pulmonary arteries from diabetic rats. Phytomedicine. 25:39–44. 2017.PubMed/NCBI View Article : Google Scholar

117 

Weseler AR and Bast A: Masquelier's grape seed extract: From basic flavonoid research to a well-characterized food supplement with health benefits. Nutr J. 16(5)2017.PubMed/NCBI View Article : Google Scholar

118 

Yang LJ, Zhu DN, Dang YL and Zhao X: Treatment of condyloma acuminata in pregnant women with cryotherapy combined with proanthocyanidins: Outcome and safety. Exp Ther Med. 11:2391–2394. 2016.PubMed/NCBI View Article : Google Scholar

119 

Sano A: Safety assessment of 4-week oral intake of proanthocyanidin-rich grape seed extract in healthy subjects. Food Chem Toxicol. 108:519–523. 2017.PubMed/NCBI View Article : Google Scholar

120 

Occhipinti A, Germano A and Maffei ME: Prevention of urinary tract infection with oximacro, a cranberry extract with a high content of A-type proanthocyanidins: A pre-clinical double-blind controlled study. Urol J. 13:2640–2649. 2016.PubMed/NCBI

121 

Serrano J, Casanova-Martí À, Gil-Cardoso K, Blay MT, Terra X, Pinent M and Ardévol A: Acutely administered grape-seed proanthocyanidin extract acts as a satiating agent. Food Funct. 7:483–490. 2016.PubMed/NCBI View Article : Google Scholar

122 

Zhao Y, Xie Y, Li X, Song J, Guo M, Xian D and Zhong J: The protective effect of proanthocyanidins on the psoriasis-like cell models via PI3K/AKT and HO-1. Redox Rep. 27:200–211. 2022.PubMed/NCBI View Article : Google Scholar

123 

Omma A, Colak S, Can Sandikci S, Yucel C, Erden A, Sertoglu E and Ozgurtas T: Serum neopterin and ischemia modified albumin levels are associated with the disease activity of adult immunoglobulin A vasculitis (Henoch-Schönlein purpura). Int J Rheum Dis. 22:1920–1925. 2019.PubMed/NCBI View Article : Google Scholar

124 

Wang B, Shao-Kuan FR and Dong C: Expression and significance of follicular helper T cells and galactose-deficient IgA1 in children with Henoch-Schönlein purpura. Zhongguo Dang Dai Er Ke Za Zhi. 22:473–477. 2020.PubMed/NCBI View Article : Google Scholar : (In Chinese).

125 

Long M, Zhang Y, Li P, Yang SH, Zhang WK, Han JX, Wang Y and He JB: Intervention of Grape seed proanthocyanidin extract on the subchronic immune injury in mice induced by Aflatoxin B1. Int J Mol Sci. 17(516)2016.PubMed/NCBI View Article : Google Scholar

126 

Liu X, Qiu J, Zhao S, You B, Ji X, Wang Y, Cui X, Wang Q and Gao H: Grape seed proanthocyanidin extract alleviates ouabain-induced vascular remodeling through regulation of endothelial function. Mol Med Rep. 6:949–954. 2012.PubMed/NCBI View Article : Google Scholar

127 

Di Pietro GM, Castellazzi ML, Mastrangelo A, Montini G, Marchisio P and Tagliabue C: Henoch-Schönlein Purpura in children: Not only kidney but also lung. Pediatr Rheumatol Online J. 17(75)2019.PubMed/NCBI View Article : Google Scholar

128 

Padeh S and Passwell JH: Successful treatment of chronic Henoch-Schonlein purpura with colchicine and aspirin. Isr Med Assoc J. 2:482–483. 2000.PubMed/NCBI

129 

Pan S and Zhong J: Construction and identification of simplified rat model of Henoch-Schönlein Purpura. Sichuan Med J. 42:669–672. 2021.(In Chinese).

130 

Li Y, Feng X, Huang L, Zhu H, Xu Y, Sui X, Xu Y, Han Y and Qin C: Hematologic and immunological characteristics of Henoch-Schönlein purpura in rat and rabbit models induced with ovalbumin based on type III hypersensitivity. Sci Rep. 5(8862)2015.PubMed/NCBI View Article : Google Scholar

131 

Kaegi C, Wuest B, Schreiner J, Steiner UC, Vultaggio A, Matucci A, Crowley C and Boyman O: Systematic review of safety and efficacy of rituximab in treating immune-mediated disorders. Front Immunol. 10(1990)2019.PubMed/NCBI View Article : Google Scholar

132 

Gohari A, Matsell DG, Mammen C and Goldman RD: Henoch-Schönlein purpura in children: Use of corticosteroids for prevention and treatment of renal disease. Can Fam Physician. 66:895–897. 2020.PubMed/NCBI View Article : Google Scholar

133 

Du Y, Hou L, Zhao C, Han M and Wu Y: Treatment of children with Henoch-Schönlein purpura nephritis with mycophenolate mofetil. Pediatr Nephrol. 27:765–771. 2012.PubMed/NCBI View Article : Google Scholar

134 

Nikibakhsh AA, Mahmoodzadeh H, Karamyyar M, Hejazi S, Noroozi M, Macooie AA, Gholizadeh A and Gholizadeh L: Treatment of complicated henoch-schönlein purpura with mycophenolate mofetil: A retrospective case series report. Int J Rheumatol. 2010(254316)2010.PubMed/NCBI View Article : Google Scholar

135 

Fujimoto M, Katayama K, Nishikawa K, Mizoguchi S, Oda K, Hirabayashi Y, Suzuki Y, Haruki A, Ito T, Murata T, et al: A Kidney transplant recipient with recurrent Henoch-Schönlein Purpura nephritis successfully treated with steroid pulse therapy and epipharyngeal abrasive therapy. Nephron. 144 (Suppl 1):S54–S48. 2020.PubMed/NCBI View Article : Google Scholar

136 

Kurnia B: Henoch-Schonlein purpura in children: The role of corticosteroids. Open Access Maced J Med Sci. 7:1812–1814. 2019.PubMed/NCBI View Article : Google Scholar

137 

Ozen S, Marks SD, Brogan P, Groot N, de Graeff N, Avcin T, Bader-Meunier B, Dolezalova P, Feldman BM, Kone-Paut I, et al: European consensus-based recommendations for diagnosis and treatment of immunoglobulin A vasculitis-the SHARE initiative. Rheumatology (Oxford). 58:1607–1616. 2019.PubMed/NCBI View Article : Google Scholar

138 

Han F, Chen LL, Ren PP, Le JY, Choong PJ, Wang HJ, Xu Y and Chen JH: Mycophenolate mofetil plus prednisone for inducing remission of Henoch-Schönlein purpura nephritis: A retrospective study. J Zhejiang Univ Sci B. 16:772–779. 2015.PubMed/NCBI View Article : Google Scholar

139 

Lu Z, Song J, Mao J, Xia Y and Wang C: Evaluation of mycophenolate mofetil and Low-dose steroid combined therapy in moderately severe Henoch-Schönlein Purpura nephritis. Med Sci Monit. 23:2333–2339. 2017.PubMed/NCBI View Article : Google Scholar

140 

Hamilton P, Ogundare O, Raza A, Ponnusamy A, Gorton J, Alachkar H, Choudhury J, Barratt J and Kalra PA: Long-term therapeutic plasma exchange to prevent End-stage kidney disease in adult severe resistant Henoch-Schonlein purpura nephritis. Case Rep Nephrol. 2015(269895)2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

June-2023
Volume 25 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Xie Y, Deng Q, Guo M, Li X, Xian D and Zhong J: Proanthocyanidins: A novel approach to Henoch‑Schonlein purpura through balancing immunity and arresting oxidative stress via TLR4/MyD88/NF‑κB signaling pathway (Review). Exp Ther Med 25: 300, 2023
APA
Xie, Y., Deng, Q., Guo, M., Li, X., Xian, D., & Zhong, J. (2023). Proanthocyanidins: A novel approach to Henoch‑Schonlein purpura through balancing immunity and arresting oxidative stress via TLR4/MyD88/NF‑κB signaling pathway (Review). Experimental and Therapeutic Medicine, 25, 300. https://doi.org/10.3892/etm.2023.11999
MLA
Xie, Y., Deng, Q., Guo, M., Li, X., Xian, D., Zhong, J."Proanthocyanidins: A novel approach to Henoch‑Schonlein purpura through balancing immunity and arresting oxidative stress via TLR4/MyD88/NF‑κB signaling pathway (Review)". Experimental and Therapeutic Medicine 25.6 (2023): 300.
Chicago
Xie, Y., Deng, Q., Guo, M., Li, X., Xian, D., Zhong, J."Proanthocyanidins: A novel approach to Henoch‑Schonlein purpura through balancing immunity and arresting oxidative stress via TLR4/MyD88/NF‑κB signaling pathway (Review)". Experimental and Therapeutic Medicine 25, no. 6 (2023): 300. https://doi.org/10.3892/etm.2023.11999