Next Article in Journal
Proof of Concept of Natural and Synthetic Antifouling Agents in Coatings
Previous Article in Journal
Genomic Analysis of Novel Sulfitobacter Bacterial Strains Isolated from Marine Biofilms
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Systematic Review

Fucoidan as a Promising Drug for Pain Treatment: Systematic Review and Meta-Analysis

by
Miguel Á. Huerta
1,2,3,
Miguel Á. Tejada
1,2,3 and
Francisco R. Nieto
1,2,3,*
1
Department of Pharmacology, University of Granada, 18016 Granada, Spain
2
Institute of Neuroscience, Biomedical Research Center, University of Granada, 18016 Granada, Spain
3
Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
*
Author to whom correspondence should be addressed.
Submission received: 25 May 2024 / Revised: 15 June 2024 / Accepted: 18 June 2024 / Published: 24 June 2024
(This article belongs to the Section Marine Pharmacology)

Abstract

:
Fucoidan is a polymer of L-fucose and L-fucose-4-sulphate naturally found in marine sources that inhibits p-selectin, preventing neutrophil recruitment to the site of injury. Fucoidan is employed in many studies as a tool to investigate the contribution of neutrophils to pain, showing analgesic effects. We performed a systematic review and meta-analysis to quantify the analgesic effects of pretreatment with fucoidan reported in the available preclinical studies. In addition, we summarized the articles which have studied the therapeutic effects of fucoidan in pathological pain at preclinical and clinical levels. The results of this systematic review reveal that pretreatment with fucoidan is a powerful tool which reduces neutrophil infiltration by 70–90% at early time points. This meta-analysis showed that preventative treatment with fucoidan produced a significant pain reduction. In addition, several preclinical studies have observed that fucoidan treatment reduces the pain that is associated with various pathologies. Finally, fucoidan has also been tested in several clinical trials, with some degree of analgesic efficacy, but they were mostly small pilot studies. Considering all the above information, it can be concluded that fucoidan is not only a preclinical tool for studying the role of neutrophils in pain but also a promising therapeutic strategy for pain treatment.

1. Introduction

The neutrophil is the first immune cell to respond to tissue damage or a potential aggressor [1]; in response to an injury, it migrates massively to the inflammatory focus, being clearly the most abundant cell [2]. Its role there is fundamental: on the one hand, thanks to its phagocytic capacity, it allows for the clearance of cellular debris associated with tissue damage and pathogens, along with degranulation and the release of nuclear material in the form of neutrophil extracellular traps (NETs), which also promote this process; on the other hand, it has the ability to release pro-inflammatory signaling molecules (mainly cytokines) that “call” other cells, favoring and accelerating their activation and migration to the affected area [3,4,5]. It can also sensitize sensory neurons, promoting their discharge [6,7]. These varied actions of neutrophils could participate in pain modulation.
The role of neutrophils in pain has been extensively studied in experimental research. In several studies, including early ones on the topic [8,9,10], a pronociceptive role has been proposed, contributing to the generation of hyperalgesia and allodynia in response to various stimuli [11,12,13]. Other authors demonstrate that under certain circumstances, neutrophils can produce analgesia [14,15,16,17], mainly associated with the production and release of endogenous opioid peptides (EOPs), such as β-endorphin, enkephalins, dynorphins, and endomorphins [14,18]. Additionally, some studies take a middle ground and argue that neutrophils do not contribute to pain [19,20]. Understanding the role of this immune population in pain could be of significant interest to the medical and scientific community, as it could lead to new therapeutic targets for pain of different etiologies and improve the pathophysiological understanding of pain and its neuroimmune modulation.
A significant portion of studies investigating the contribution of neutrophils to pain in animal models employ a strategy based on neutrophil depletion using pharmacological treatment. Immunological strategies are the standard treatment for achieving neutrophil depletion. In early studies, at the end of the century, anti-neutrophil sera were used [17,21,22]. A few years later, with the advent of monoclonal antibodies, the anti-Gr-1 antibody (RB6-8C5) was used, which achieves robust depletion but is not selective for neutrophils (it affects other immune cells, mainly monocytes) [20,23,24]. In contrast, the anti-Ly6G antibody has superior selectivity and generally only affects neutrophils [25,26,27], although some studies report a significant reduction in macrophages [11]. However, these strategies with biological treatments have several limitations [28]: (1) as they are generally new molecules with high production costs, the cost is high; (2) it has been reported that the potent and sudden depletion produced by these treatments can generate a rebound response where there is a prominent proliferation in the bone marrow to try to compensate for this deficiency state [29,30]; (3) because the same markers (GR-1 and Ly6-G) are used for the detection and quantification of neutrophils, there may be antigen masking, leading to misinterpretation of a good depletion efficiency [30]; (4) incomplete depletion of neutrophils [29]; and (5) organ- and strain-specific neutrophil depletion [28]. Based on these drawbacks, some authors have opted to use fucoidan to evaluate the role of neutrophils in their pain models.
In addition to being a tool for studying neuroimmune interactions in pain, some authors propose the use of fucoidan as a therapeutic strategy for pain related with several conditions [31,32]. Furthermore, fucoidan has also been proposed for blocking post-ischemic reperfusion injury, in which the cascade of inflammation causes tissue damage [33,34,35]. Although these activities are mainly related with the inhibition of neutrophil recruitment, other mechanisms of fucoidan may contribute to the analgesic and anti-inflammatory properties: selective inhibition of COX-2 [36], inhibition of hyaluronidase [36,37], mitogen-activated protein kinase p38 inhibition [38], protein denaturation inhibition [39], and stabilization of the cell membrane integrity of human red blood corpuscles [39]. Several articles evaluated the analgesic efficacy of fucoidan in different models of pain, and even a few clinical experiences have been reported [40]. Therefore, the number of studies evaluating fucoidan for various purposes has significantly increased in recent years [41].
The objective of this work is to gather all the studies that employ fucoidan to investigate the role of neutrophils in animal models of pain and quantify the effect over pain. For this purpose, we will perform a systematic review and meta-analysis to quantify the analgesic effects of pretreatment with fucoidan reported in the available preclinical studies. Additionally, we will also gather studies proposing fucoidan as a therapeutic strategy for pain associated with different pathological conditions, both at preclinical and clinical level.

2. Results and Discussion

2.1. Fucoidan: Mechanism of Action

Fucoidan (also called fucoidin) is a polymer of L-fucose and L-fucose-4-sulphate which belongs to a group of sugar analogs [42]. It is found naturally in marine sources, mainly in brown seaweeds (e.g., Fucus vesiculosus, Cladosiphon sp., or Undaria sp.), and some marine invertebrate tissues [43,44]. It is relevant to note that fucoidan abundance and composition may vary between these sources and even between growing conditions, geographic location, harvesting season, anatomical regions, and extraction procedures [41,42,45,46,47,48,49,50,51]. The mechanism of action of fucoidan on immune cells is based on the inhibition of cell adhesion molecules, P-selectin, and to a lesser extent, L-selectin [52,53,54,55,56]. Both are highly abundant on the neutrophil membrane and are necessary for their adhesion/extravasation during an acute inflammatory response [57]. P-selectin also promotes neutrophil extracellular trap formation [58]. However, these proteins are not exclusive to neutrophils and are found in other immune cell populations (e.g., macrophages [59], or lymphocytes [60]). Therefore, fucoidan treatment may directly affect the migration of other immune cells to the site of inflammation, such as macrophages/monocytes or lymphocytes [61]. Nevertheless, most models assessing the role of fucoidan on pain were acute models, where the pain response was generally evaluated in a short period after pain induction (e.g., 0.5–3 h after inflammatory substance injection [62,63,64,65,66,67,68]), when neutrophil migration is very prominent and the presence of other populations is residual. Therefore, fucoidan analgesic effects observed by these studies probably mainly account for neutrophil migration inhibition. In this sense, fucoidan can be considered appropriate for evaluating the contribution of neutrophils in these acute inflammatory models.

2.2. Fucoidan: A Tool to Study the Role of Neutrophils in Pain

The search results and selection strategy are detailed in the PRISMA flowchart (Figure 1). In total, 31 articles met the inclusion criteria, evaluating the role of neutrophils in pain using pretreatment with fucoidan as a tool. The main information of these articles is summarized in Table 1. All the evaluations were performed on rats (16/31/) or mice (15/31). The doses administered ranged between 10 and 100 mg/kg, and the administration time before injury was 10–30 min in most of the studies. Pain models evaluated were diverse. In majority, inflammation was induced by an intraplantar injection of complex pro-inflammatory substances (CFA [69,70] or carrageenan [63,65,68]), or isolated pro-inflammatory compounds such as 5-HT [71,72], TNF [64,73], IL-17 [73], and others [62,63,74,75,76]. The other studies employed arthritis models with an intra-articular administration of inflammatory substances such as zymosan [77], mBSA [78,79], LTB4 [77], IL-17 [79], MSU crystals [80,81], and others [82,83,84,85,86]. Moreover, two articles evaluated muscle pain [66,87], and one employed a neuropathic pain model (postherpetic neuralgia [88]). On the other hand, pain was mainly assessed by the evaluation of reflex-based responses to stimuli (evoked pain)—mechanical hyperalgesia, mechanical allodynia, and heat hyperalgesia—although non-evoked nociceptive behavior was also explored in some articles. The assessment of non-evoked pain has been suggested to be more translational as it better reflects the clinical situation of patients in which spontaneous pain is more clinically relevant than cutaneous hypersensitivity [89].

2.3. Efficacy of Fucoidan as a Neutrophil Depletion Strategy

The efficacy of neutrophil depletion was assessed by diverse quantification methods: immunohistochemistry, flow cytometry, or myeloperoxidase were the most common (see Table 1). Most of them (≈90%) employed the myeloperoxidase assay (MPO), which allows estimating the number of neutrophils per mg of tissue. Although this technique demonstrated a high correlation with the number of neutrophils [85], the enzyme is also present in monocytes/macrophages [95], so the quantification might not be totally selective for neutrophils. In contrast, quantification using flow cytometry is much more selective as specific markers are used to differentiate populations, yet the methodology is more complex [94]. However, the oldest studies employed dyes followed by a manual counting [69], which can be considered a semi-quantitative method. As an example, the oldest article included in this analysis observed that pretreatment with fucoidan induced a slight decrease (40%) in the number of immune cells containing β-endorphin (mainly neutrophils) after CFA injection [69]. This low efficacy observed can be associated with a too low of a dose being used (10 mg/kg) [69]. On the contrary, all the studies published thereafter employed higher doses (20–40 mg/kg), achieving a great neutrophil depletion efficacy (70–90%).
The use of fucoidan for neutrophil depletion presents several advantages: (1) it only blocks cell extravasation and does not affect the blood count of neutrophils [61], while other strategies such as antibodies anti-Ly6G (1A8) or anti-Gr-1 (RB6) also reduce blood-circulating neutrophils [28,96]; (2) fucoidan does not affect neutrophil tissular levels in naive animals, while antibodies cause a robust reduction observed in different organs (spleen, lungs or bone marrow) [28,97]. Moreover, neutrophil depletion has been associated with a decrease of edema in the injured area in several models [98,99]. Based on all the above, it can be concluded that preventive treatment with fucoidan is effective for neutrophil depletion and presents some advantages in comparison to other methods.

2.4. Analgesic Efficacy of Preventive Treatment with Fucoidan

In all the articles compiled in Table 1, the effect of pretreatment with fucoidan on pain is evaluated. As mentioned in the previous section, in all cases, neutrophil depletion was robust (generally, a reduction of 70–90%). In 95% of the studies, a significant and robust reduction in pain was observed. It is likely that the absence of an analgesic effect of fucoidan observed in some of the works was due to the dose of fucoidan being too low (e.g., 10 mg/kg [69]).
Next, the analgesic efficacy of neutrophil depletion with fucoidan was quantified by conducting a meta-analysis (Figure 2). Fucoidan clearly showed significant analgesic efficacy (p < 0.0001) with a large effect size (75.1, CI95: 66.6, 83.6). However, the heterogeneity was high (I² = 98.7%), which may be due to the large variety in the evaluations and experimental conditions.
According to these results, it can be concluded that neutrophils have a pronociceptive role in the acute pain models studied. Accordingly, treatment with fucoidan can be considered an interesting strategy for the treatment of pain in painful pathologies that involve an acute neutrophil response.
In addition, an important variable that can influence the analgesic efficacy is the administration route, as the bioavailability of fucoidan may vary between administration routes. Most of the studies employed the intravenous route 26/36, 9/36 of the studies used the intraperitoneal route, and only 1/36 used the oral route. A subgroup analysis stratified according to the administration route used concluded that the effect size for the analgesic efficacy was 69.06 (CI95: 61.15, 76.98) for the intravenous route, 87.85 (CI95: 65.86, 109.84) for the intraperitoneal route, and 115.87 (CI95: 103.75, 127.99) for the oral route. These results suggest that the intraperitoneal and oral bioavailability of fucoidan is high as the efficacy was equivalent to the one achieved by intravenous administration. Another variable which may influence the analgesic efficacy is the source of fucoidan, as its composition may vary between sources. However, the vast majority of studies employed the same commercial fucoidan extracted from Fucus vesiculosus (Sigma-Aldrich, St. Louis, MO, USA), so it was not necessary to conduct this analysis.

2.5. Fucoidan as a Tool to Study Opioid-Related Endogenous Analgesia

Endogenous analgesia has a very relevant role in the pathophysiology of pain and its treatment [100,101]. Among the different proposed mechanisms, the endogenous opioid system is one of the main players [102,103,104]. In fact, it is known to be involved in many well-known phenomena, such as the placebo effect [105]. Immune cells are the main actors in these mechanisms due to their ability to produce and release endogenous opioid peptides (EOPs) in response to different situations. As neutrophils are the primary cells during early inflammation, they constitute the main source of EOPs in the periphery.
Information on studies using fucoidan to evaluate endogenous opioid analgesia can be found in Table 2. These articles employed fucoidan together with an experimental analgesic strategy (e.g., ankle joint mobilization or light-emitting diode therapy) in order to evaluate the participation of neutrophils in the efficacy of the intervention being tested. In other words, in these studies, fucoidan was only a tool for evaluating if the analgesic efficacy of the experimental compound/intervention was dependent on OEP liberation by the recruited neutrophils. Already in 1998, the first study using fucoidan demonstrated that it was capable of blocking endogenous opioid analgesia associated with the release of EOP by immune cells (mainly neutrophils) after a stressful situation (cold water swim) [69]. A few years later, the same authors confirmed that the abolishment of endogenous opioid analgesia with the treatment of fucoidan was mediated by P-selectin and L-selectin inhibition, with the decrease in neutrophils and the consequent halving of EOP being confirmed by cytometry [61]. Subsequently, other authors have replicated these experiments with similar results, while others used this tool to elucidate whether the effect of an analgesic treatment was mediated by POEs [106,107,108]. In parallel, other authors have confirmed the effect of fucoidan in the reduction of opioid analgesia (induced by stress or corticotropin-releasing factor administration), employing other strategies for neutrophil depletion, such as anti-Ly6-G antibodies [17,22,109,110,111].

2.6. Fucoidan for Treating Pain-Related Conditions: Preclinical Studies

The previous analyses evaluated the effect of feeling pain after a pretreatment with fucoidan. Therefore, it is interesting to evaluate its analgesic efficacy when the treatment is therapeutic (once the pathology is already established) instead of prophylactic. Several studies evaluating the therapeutic use of fucoidan in pain-related diseases were found, and they are summarized in Table 3. It is interesting to highlight a good analgesic efficacy observed in two different models of neuropathic pain (nerve injury [112] and chemotherapy-induced injury [113]), in models of visceral inflammation (colitis [114], prostatitis [31]), and in models of joint degeneration [32].

2.7. Fucoidan for Treating Pain-Related Conditions: Human Studies

The promising preclinical results mentioned earlier prompted the initiation of various small to medium-sized clinical trials in humans to assess the effectiveness of fucoidan in treating pain-related conditions (summarized in Table 4). These studies were mainly performed in patients with joint pain associated with osteoarthritis, and promising results were observed. In addition to the anti-neutrophil effect, which may explain these positive results in pain and joint degradation, fucoidan has also demonstrated antifibrotic efficacy (specifically in the joint) by inhibiting the differentiation of fibroblast-like synoviocytes into myofibroblasts with tumor cell-like characteristics and restoring apoptosis [116]. Also, fucoidan was capable of ameliorating intervertebral disc degeneration in a preclinical model by restoring redox and matrix homeostasis of nucleus pulposus [32].

2.8. Deleterious Effects of Neutrophil Depletion

Neutropenic mice were found to have a larger quantity of large-sized Candida auris abscesses, suggesting an inability of the adaptive immune response to clear fungal infections in the absence of neutrophils [120], which may even cause death in the host [97]. However, it has been proposed that neutrophils are not that crucial for other infective pathologies [96]. A recent study found that early neutrophil depletion following an inflammatory injury could contribute to pain becoming chronic [121]. This suggests that an early neutrophilic response plays a protective role in preventing pain chronicity. Many authors have demonstrated that neutrophils are crucial cells in tissue regeneration processes, implying that inhibiting them could hinder tissue repair [122]. However, this notion is controversial, as other authors suggest the opposite—that neutrophils may cause further tissue damage, leading to delayed healing and excessive scar formation [123].

2.9. Disadvantages of Using Fucoidan for Neutrophil Depletion

The lack of specificity of fucoidan towards neutrophils in experimental setups might introduce ambiguity in delineating the pronociceptive role of neutrophils in pain, considering the potential involvement of other inflammatory cell types. Nevertheless, this circumstance could present a favorable prospect for fucoidan as a therapeutic intervention, as its anti-inflammatory and analgesic properties might be enhanced, given the reported pronociceptive nature of other inflammatory cell populations. Another disadvantage is that the composition and structure of fucoidan is dependent on seaweed species, geographic location, harvesting season, anatomical regions, and extraction procedures [41,42,45,46,47,48,49,50,51]. This structural variability (with molecular weight ranging from 10,000 to 100,000 Da) may generate variability in the bioactivity of fucoidan [42,45,124,125]. In addition, it is well known that fucoidan produces anticoagulant effects [126,127], which my interfere in its analgesic efficacy. In this line, some related adverse events, such as bleeding, may be associated with the fucoidan treatment.

3. Methods

3.1. Protocol and Registration

The methodology used in this review was specified in advance and documented in a protocol registered on Prospective Register of Systematic Reviews (PROSPERO) under the registration ID CRD42024534839. This review was performed in accordance with the latest version (2020) of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines on systematic reviews and meta-analyses [128].

3.2. PICO Research Question

(P) Animal models of pain.
(I) Pretreatment with fucoidan, also known as fucoidin.
(C) Control group treated with vehicle.
(O) Changes in pain-related parameters.
(S) Original preclinical studies.

3.3. Information Sources and Search Strategy

We performed a comprehensive systematic search in PubMed without restrictions for the language or date. The search was updated on 25 May 2024. The search strategy was as follows: (fucoidan OR fucoidin) AND ((pain OR hyperalgesia OR analgesia OR pain* OR nocicept* OR allodynia OR hyperalges*) OR (neutrophil* OR polymorphonuclear OR Ly6G OR GR-1)).

3.4. Inclusion and Exclusion Criteria

Inclusion criteria: original preclinical studies using animal models of pain in which the effect of the pretreatment (before injury) with fucoidan regarding pain outcomes was assessed.
Exclusion criteria: non-pain model, absence of pain measurement, review articles, human studies, in vitro or ex vivo experiments, studies including no relevant information or missing information, and violations of any of the above inclusion criteria.

3.5. Article Selection

Titles and abstracts of studies were retrieved using the search strategy by two review authors (MAH and MAT) in a blind manner to identify studies that potentially met the inclusion criteria. The full texts of these potentially eligible studies were retrieved and independently assessed for eligibility by the same team members (MAH and MAT). The selection process was carried out using the software Rayyan. Any disagreement between the authors was resolved through a discussion with a third reviewer (FRN).

3.6. Data Extraction

MAH and MAT independently extracted data from each included article by using a standardized, pre-piloted form. Discrepancies were identified and resolved through discussion (with a third author (FRN). Numerical data were extracted from tables, text, or figures. When they were not reported, data were extracted from graphs using digital ruler software. In case data were not reported or unclear, we contacted authors by e-mail. In case an outcome was measured at multiple time points, data from the time point where efficacy was highest were included. Extracted information included study setting; study population (animal model of pain used, including species and strains) and baseline characteristics; details of the fucoidan intervention, the timing, the dose used, and control conditions; study methodology; pain assessment test used; and main results of the intervention.

3.7. Meta-Analysis and Statistics

Meta-analyses were conducted using the metafor package in R, version 4.1.2 [129,130]. Since different pain outcomes were measured with different scales in the included studies, outcome data were normalized using the normalized mean difference (NMD), which is a useful approach when conducing a meta-analysis of preclinical data because it relates the magnitude of effect in the treatment group to a group of healthy animals [131]. Then, a 95% confidence interval (CI95%) was computed. The inverse variance statistical analysis method was used to summarize the effect sizes of the treatment, and the combined results were analyzed using the random effect model, which accounted for the variance within and between studies [132]. Effects were considered statistically significant when the p value was less than 0.05. For assessing heterogeneity, the Cochrane’s Q test (with p < 0.10 indicating asymmetry) and the Higgins–Thompson I2 values (null or low, 0–30%; medium, 30–50%; moderate, 50–75%; and high heterogeneity, >75%) were used to assess the heterogeneity within the pooled studies [133].

4. Concluding Remarks

Fucoidan is an interesting preclinical tool for depleting neutrophils, which may be useful to study their role in pain and other diseases. Moreover, in preclinical studies, preventive treatment with fucoidan, which substantially reduced neutrophil infiltration, was very effective for reversing acute pain. Furthermore, the therapeutic treatment with fucoidan also showed a robust analgesic effect in models of neuropathic, visceral, and joint pain. These positive preclinical results led to the launch of small clinical trials which reported some positive results, mainly for joint pain associated with osteoarthritis, with a good safety profile. Based on our data, fucoidan may be an interesting analgesic strategy for some pair-related conditions. However, randomized placebo-controlled trials involving more patients should be performed to confirm these promising findings.

Author Contributions

Conceptualization, M.Á.H. and F.R.N.; methodology, M.Á.H.; software, M.Á.H.; validation, M.Á.T. and F.R.N.; formal analysis, F.R.N.; investigation, M.Á.H.; resources, M.Á.T.; data curation, M.Á.T.; writing—original draft preparation, M.Á.H.; writing—review and editing, M.Á.T. and F.R.N.; visualization, M.Á.T.; supervision, F.R.N.; project administration, F.R.N.; funding acquisition, F.R.N. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. M.Á. Huerta was supported by the Training University Lecturers program (FPU21/02736) of the Spanish Ministry of Economy and Competitiveness (MINECO).

Institutional Review Board Statement

Not applicable.

Data Availability Statement

The raw data supporting the conclusions of this article will be made available by the authors upon request.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Fine, N.; Tasevski, N.; McCulloch, C.A.; Tenenbaum, H.C.; Glogauer, M. The Neutrophil: Constant Defender and First Responder. Front. Immunol. 2020, 11, 571085. [Google Scholar] [CrossRef] [PubMed]
  2. Margraf, A.; Lowell, C.A.; Zarbock, A. Neutrophils in Acute Inflammation: Current Concepts and Translational Implications. Blood 2022, 139, 2130–2144. [Google Scholar] [CrossRef] [PubMed]
  3. Rosales, C. Neutrophil: A Cell with Many Roles in Inflammation or Several Cell Types? Front. Physiol. 2018, 9, 113. [Google Scholar] [CrossRef] [PubMed]
  4. Riaz, B.; Sohn, S. Neutrophils in Inflammatory Diseases: Unraveling the Impact of Their Derived Molecules and Heterogeneity. Cells 2023, 12, 2621. [Google Scholar] [CrossRef]
  5. Soehnlein, O.; Steffens, S.; Hidalgo, A.; Weber, C. Neutrophils as Protagonists and Targets in Chronic Inflammation. Nat. Rev. Immunol. 2017, 17, 248–261. [Google Scholar] [CrossRef] [PubMed]
  6. Calvo, M.; Dawes, J.M.; Bennett, D.L.H. The Role of the Immune System in the Generation of Neuropathic Pain. Lancet Neurol. 2012, 11, 629–642. [Google Scholar] [CrossRef]
  7. Momin, A.; McNaughton, P.A. Regulation of Firing Frequency in Nociceptive Neurons by Pro-Inflammatory Mediators. Exp. Brain Res. 2009, 196, 45–52. [Google Scholar] [CrossRef] [PubMed]
  8. Levine, J.D.; Gooding, J.; Donatoni, P.; Borden, L.; Goetzl, E.J. The Role of the Polymorphonuclear Leukocyte in Hyperalgesia. J. Neurosci. 1985, 5, 3025. [Google Scholar] [CrossRef]
  9. Bisgaard, H.; Kristensen, J.K. Leukotriene B4 Produces Hyperalgesia in Humans. Prostaglandins 1985, 30, 791–797. [Google Scholar] [CrossRef]
  10. Levine, J.D.; Lau, W.; Kwiat, G.; Goetzl, E.J. Leukotriene B4 Produces Hyperalgesia That Is Dependent on Polymorphonuclear Leukocytes. Science 1984, 225, 743–745. [Google Scholar] [CrossRef]
  11. Lee, M.-C.; McCubbin, J.A.; Christensen, A.D.; Poole, D.P.; Rajasekhar, P.; Lieu, T.; Bunnett, N.W.; Garcia-Caraballo, S.; Erickson, A.; Brierley, S.M.; et al. G-CSF Receptor Blockade Ameliorates Arthritic Pain and Disease. J. Immunol. 2017, 198, 3565–3575. [Google Scholar] [CrossRef] [PubMed]
  12. Harada, Y.; Zhang, J.; Imari, K.; Yamasaki, R.; Ni, J.; Wu, Z.; Yamamoto, K.; Kira, J.I.; Nakanishi, H.; Hayashi, Y. Cathepsin E in Neutrophils Contributes to the Generation of Neuropathic Pain in Experimental Autoimmune Encephalomyelitis. Pain 2019, 160, 2050–2062. [Google Scholar] [CrossRef] [PubMed]
  13. Carreira, E.U.; Carregaro, V.; Teixeira, M.M.; Moriconi, A.; Aramini, A.; Verri, W.A.; Ferreira, S.H.; Cunha, F.Q.; Cunha, T.M. Neutrophils Recruited by CXCR1/2 Signalling Mediate Post-Incisional Pain. Eur. J. Pain 2013, 17, 654–663. [Google Scholar] [CrossRef] [PubMed]
  14. Rittner, H.L.; Brack, A.; Stein, C. Pain and the Immune System. Br. J. Anaesth. 2008, 101, 40–44. [Google Scholar] [CrossRef] [PubMed]
  15. Rittner, H.L.; Hackel, D.; Yamdeu, R.S.; Mousa, S.A.; Stein, C.; Schäfer, M.; Brack, A. Antinociception by Neutrophil-Derived Opioid Peptides in Noninflamed Tissue—Role of Hypertonicity and the Perineurium. Brain Behav. Immun. 2009, 23, 548–557. [Google Scholar] [CrossRef] [PubMed]
  16. Shi, J.T.; Cao, W.Y.; Zhang, X.N.; Wan, H.Y.; Su, Y.S.; Qu, Z.Y.; Wang, R.; He, W.; Jing, X.H.; Wang, X.Y. Local Analgesia of Electroacupuncture Is Mediated by the Recruitment of Neutrophils and Released β-Endorphins. Pain 2023, 164, 1965–1975. [Google Scholar] [CrossRef] [PubMed]
  17. Brack, A.; Rittner, H.L.; Machelska, H.; Leder, K.; Mousa, S.A.; Schäfer, M.; Stein, C. Control of Inflammatory Pain by Chemokine-Mediated Recruitment of Opioid-Containing Polymorphonuclear Cells. Pain 2004, 112, 229–238. [Google Scholar] [CrossRef] [PubMed]
  18. Gupta, A.; Gullapalli, S.; Pan, H.; Ramos-Ortolaza, D.L.; Hayward, M.D.; Low, M.J.; Pintar, J.E.; Devi, L.A.; Gomes, I. Regulation of Opioid Receptors by Their Endogenous Opioid Peptides. Cell Mol. Neurobiol. 2021, 41, 1103. [Google Scholar] [CrossRef] [PubMed]
  19. Segelcke, D.; Pradier, B.; Reichl, S.; Schäfer, L.C.; Pogatzki-Zahn, E.M. Investigating the Role of Ly6G+ Neutrophils in Incisional and Inflammatory Pain by Multidimensional Pain-Related Behavioral Assessments: Bridging the Translational Gap. Front. Pain Res. 2021, 2, 735838. [Google Scholar] [CrossRef]
  20. Sahbaie, P.; Li, X.; Shi, X.; Clark, J.D. Roles of Gr-1+ Leukocytes in Postincisional Nociceptive Sensitization and Inflammation. Anesthesiology 2012, 117, 602–612. [Google Scholar] [CrossRef]
  21. Bennett, G.; Al-Rashed, S.; Hoult, J.R.S.; Brain, S.D. Nerve Growth Factor Induced Hyperalgesia in the Rat Hind Paw Is Dependent on Circulating Neutrophils. Pain 1998, 77, 315–322. [Google Scholar] [CrossRef] [PubMed]
  22. Labuz, D.; Berger, S.; Mousa, S.A.; Zöllner, C.; Rittner, H.L.; Shaqura, M.A.; Segovia-Silvestre, T.; Przewlocka, B.; Stein, C.; Machelska, H. Peripheral Antinociceptive Effects of Exogenous and Immune Cell-Derived Endomorphins in Prolonged Inflammatory Pain. J. Neurosci. 2006, 26, 4350–4358. [Google Scholar] [CrossRef] [PubMed]
  23. Chiu, I.M.; Heesters, B.A.; Ghasemlou, N.; Von Hehn, C.A.; Zhao, F.; Tran, J.; Wainger, B.; Strominger, A.; Muralidharan, S.; Horswill, A.R.; et al. Bacteria Activate Sensory Neurons That Modulate Pain and Inflammation. Nature 2013, 501, 52–57. [Google Scholar] [CrossRef] [PubMed]
  24. González-Rodríguez, S.; Álvarez, M.G.; García-Domínguez, M.; Lastra, A.; Cernuda-Cernuda, R.; Folgueras, A.R.; Fernández-García, M.T.; Hidalgo, A.; Baamonde, A.; Menéndez, L. Hyperalgesic and Hypoalgesic Mechanisms Evoked by the Acute Administration of CCL5 in Mice. Brain Behav. Immun. 2017, 62, 151–161. [Google Scholar] [CrossRef] [PubMed]
  25. Nadeau, S.; Filali, M.; Zhang, J.; Kerr, B.J.; Rivest, S.; Soulet, D.; Iwakura, Y.; Vaccari, J.P.d.R.; Keane, R.W.; Lacroix, S. Functional Recovery after Peripheral Nerve Injury Is Dependent on the Pro-Inflammatory Cytokines IL-1β and TNF: Implications for Neuropathic Pain. J. Neurosci. 2011, 31, 12533–12542. [Google Scholar] [CrossRef] [PubMed]
  26. Tejada, M.A.; Montilla-García, A.; Cronin, S.J.; Cikes, D.; Sánchez-Fernández, C.; González-Cano, R.; Ruiz-Cantero, M.C.; Penninger, J.M.; Vela, J.M.; Baeyens, J.M.; et al. Sigma-1 Receptors Control Immune-Driven Peripheral Opioid Analgesia during Inflammation in Mice. Proc. Natl. Acad. Sci. USA 2017, 114, 8396–8401. [Google Scholar] [CrossRef] [PubMed]
  27. Ruiz-Cantero, M.C.; Huerta, M.; Tejada, M.; Santos-Caballero, M.; Fernández-Segura, E.; Cañizares, F.J.; Entrena, J.M.; Baeyens, J.M.; Cobos, E.J. Sigma-1 Receptor Agonism Exacerbates Immune-Driven Nociception: Role of TRPV1 + Nociceptors. Biomed. Pharmacother. 2023, 167, 115534. [Google Scholar] [CrossRef] [PubMed]
  28. Pollenus, E.; Malengier-Devlies, B.; Vandermosten, L.; Pham, T.T.; Mitera, T.; Possemiers, H.; Boon, L.; Opdenakker, G.; Matthys, P.; Van den Steen, P.E. Limitations of Neutrophil Depletion by Anti-Ly6G Antibodies in Two Heterogenic Immunological Models. Immunol. Lett. 2019, 212, 30–36. [Google Scholar] [CrossRef] [PubMed]
  29. Moses, K.; Klein, J.C.; Männ, L.; Klingberg, A.; Gunzer, M.; Brandau, S. Survival of Residual Neutrophils and Accelerated Myelopoiesis Limit the Efficacy of Antibody-Mediated Depletion of Ly-6G+ Cells in Tumor-Bearing Mice. J. Leukoc. Biol. 2016, 99, 811–823. [Google Scholar] [CrossRef]
  30. Boivin, G.; Faget, J.; Ancey, P.B.; Gkasti, A.; Mussard, J.; Engblom, C.; Pfirschke, C.; Contat, C.; Pascual, J.; Vazquez, J.; et al. Durable and Controlled Depletion of Neutrophils in Mice. Nat. Commun. 2020, 11, 2762. [Google Scholar] [CrossRef]
  31. Zhang, C.; Chen, J.; Wang, H.; Chen, J.; Zheng, M.J.; Chen, X.G.; Zhang, L.; Liang, C.Z.; Zhan, C.S. IL-17 Exacerbates Experimental Autoimmune Prostatitis via CXCL1/CXCL2-Mediated Neutrophil Infiltration. Andrologia 2022, 54, e14455. [Google Scholar] [CrossRef]
  32. Li, Y.; Tian, X.; He, W.; Jin, C.; Yang, C.; Pan, Z.; Xu, Y.; Yang, H.; Liu, H.; Liu, T.; et al. Fucoidan-Functionalized Gelatin Methacryloyl Microspheres Ameliorate Intervertebral Disc Degeneration by Restoring Redox and Matrix Homeostasis of Nucleus Pulposus. Int. J. Biol. Macromol. 2023, 250, 126166. [Google Scholar] [CrossRef] [PubMed]
  33. Wang, Z.; Liu, T.; Chen, X.; You, H.; Zhang, Q.; Xue, J.; Zheng, Y.; Luo, D. Low Molecular Weight Fucoidan Ameliorates Hindlimb Ischemic Injury in Type 2 Diabetic Rats. J. Ethnopharmacol. 2018, 210, 434–442. [Google Scholar] [CrossRef] [PubMed]
  34. Ahn, J.H.; Shin, M.C.; Kim, D.W.; Kim, H.; Song, M.; Lee, T.K.; Lee, J.C.; Kim, H.; Cho, J.H.; Kim, Y.M.; et al. Antioxidant Properties of Fucoidan Alleviate Acceleration and Exacerbation of Hippocampal Neuronal Death Following Transient Global Cerebral Ischemia in High-Fat Diet-Induced Obese Gerbils. Int. J. Mol. Sci. 2019, 20, 554. [Google Scholar] [CrossRef] [PubMed]
  35. Manzo-Silberman, S.; Louedec, L.; Meilhac, O.; Letourneur, D.; Michel, J.B.; Elmadbouh, I. Therapeutic Potential of Fucoidan in Myocardial Ischemia. J. Cardiovasc. Pharmacol. 2011, 58, 626–632. [Google Scholar] [CrossRef] [PubMed]
  36. Pozharitskaya, O.N.; Obluchinskaya, E.D.; Shikov, A.N. Mechanisms of Bioactivities of Fucoidan from the Brown Seaweed Fucus vesiculosus L. of the Barents Sea. Mar. Drugs 2020, 18, 275. [Google Scholar] [CrossRef] [PubMed]
  37. Arunkumar, K.; Raj, R.; Raja, R.; Carvalho, I.S. Brown Seaweeds as a Source of Anti-Hyaluronidase Compounds. S. Afr. J. Bot. 2021, 139, 470–477. [Google Scholar] [CrossRef]
  38. Choo, G.S.; Lee, H.N.; Shin, S.A.; Kim, H.J.; Jung, J.Y. Anticancer Effect of Fucoidan on DU-145 Prostate Cancer Cells through Inhibition of PI3K/Akt and MAPK Pathway Expression. Mar. Drugs 2016, 14, 126. [Google Scholar] [CrossRef] [PubMed]
  39. Obluchinskaya, E.D.; Pozharitskaya, O.N.; Shikov, A.N. In Vitro Anti-Inflammatory Activities of Fucoidans from Five Species of Brown Seaweeds. Mar. Drugs 2022, 20, 606. [Google Scholar] [CrossRef] [PubMed]
  40. Myers, S.P.; Mulder, A.M.; Baker, D.G.; Robinson, S.R.; Rolfe, M.I.; Brooks, L.; Fitton, J.H. Effects of Fucoidan from Fucus Vesiculosus in Reducing Symptoms of Osteoarthritis: A Randomized Placebo-Controlled Trial. Biologics 2016, 10, 81–88. [Google Scholar] [CrossRef]
  41. Ale, M.T.; Mikkelsen, J.D.; Meyer, A.S. Important Determinants for Fucoidan Bioactivity: A Critical Review of Structure-Function Relations and Extraction Methods for Fucose-Containing Sulfated Polysaccharides from Brown Seaweeds. Mar. Drugs 2011, 9, 2106–2130. [Google Scholar] [CrossRef] [PubMed]
  42. Wang, Y.; Xing, M.; Cao, Q.; Ji, A.; Liang, H.; Song, S. Biological Activities of Fucoidan and the Factors Mediating Its Therapeutic Effects: A Review of Recent Studies. Mar. Drugs 2019, 17, 183. [Google Scholar] [CrossRef] [PubMed]
  43. Anastyuk, S.D.; Imbs, T.I.; Shevchenko, N.M.; Dmitrenok, P.S.; Zvyagintseva, T.N. ESIMS Analysis of Fucoidan Preparations from Costaria Costata, Extracted from Alga at Different Life-Stages. Carbohydr. Polym. 2012, 90, 993–1002. [Google Scholar] [CrossRef] [PubMed]
  44. Wang, J.; Zhang, Q.; Zhang, Z.; Zhang, H.; Niu, X. Structural Studies on a Novel Fucogalactan Sulfate Extracted from the Brown Seaweed Laminaria Japonica. Int. J. Biol. Macromol. 2010, 47, 126–131. [Google Scholar] [CrossRef] [PubMed]
  45. Yuan, Y.; Macquarrie, D. Microwave Assisted Extraction of Sulfated Polysaccharides (Fucoidan) from Ascophyllum Nodosum and Its Antioxidant Activity. Carbohydr. Polym. 2015, 129, 101–107. [Google Scholar] [CrossRef] [PubMed]
  46. Li, B.; Lu, F.; Wei, X.; Zhao, R. Fucoidan: Structure and Bioactivity. Molecules 2008, 13, 1671–1695. [Google Scholar] [CrossRef] [PubMed]
  47. Foley, S.A.; Mulloy, B.; Tuohy, M.G. An Unfractionated Fucoidan from Ascophyllum Nodosum: Extraction, Characterization, and Apoptotic Effects in Vitro. J. Nat. Prod. 2011, 74, 1851–1861. [Google Scholar] [CrossRef]
  48. Honya, M.; Mori, H.; Anzai, M.; Araki, Y.; Nisizawa, K. Monthly Changes in the Content of Fucans, Their Constituent Sugars and Sulphate in Cultured Laminaria Japonica. Hydrobiologia 1999, 398, 411–416. [Google Scholar] [CrossRef]
  49. Mak, W.; Hamid, N.; Liu, T.; Lu, J.; White, W.L. Fucoidan from New Zealand Undaria Pinnatifida: Monthly Variations and Determination of Antioxidant Activities. Carbohydr. Polym. 2013, 95, 606–614. [Google Scholar] [CrossRef]
  50. Kusaykin, M.; Bakunina, I.; Sovo, V.; Ermakova, S.; Kuznetsova, T.; Besednova, N.; Zaporozhets, T.; Zvyagintseva, T. Structure, Biological Activity, and Enzymatic Transformation of Fucoidans from the Brown Seaweeds. Biotechnol. J. 2008, 3, 904–915. [Google Scholar] [CrossRef]
  51. Chizhov, A.O.; Dell, A.; Morris, H.R.; Haslam, S.M.; McDowell, R.A.; Shashkov, A.S.; Nifant’ev, N.E.; Khatuntseva, E.A.; Usov, A.I. A Study of Fucoidan from the Brown Seaweed Chorda Filum. Carbohydr. Res. 1999, 320, 108–119. [Google Scholar] [CrossRef] [PubMed]
  52. Juenet, M.; Aid-Launais, R.; Li, B.; Berger, A.; Aerts, J.; Ollivier, V.; Nicoletti, A.; Letourneur, D.; Chauvierre, C. Thrombolytic Therapy Based on Fucoidan-Functionalized Polymer Nanoparticles Targeting P-Selectin. Biomaterials 2018, 156, 204–216. [Google Scholar] [CrossRef] [PubMed]
  53. Bachelet, L.; Bertholon, I.; Lavigne, D.; Vassy, R.; Jandrot-Perrus, M.; Chaubet, F.; Letourneur, D. Affinity of Low Molecular Weight Fucoidan for P-Selectin Triggers Its Binding to Activated Human Platelets. Biochim. Biophys. Acta 2009, 1790, 141–146. [Google Scholar] [CrossRef] [PubMed]
  54. Shamay, Y.; Elkabets, M.; Li, H.; Shah, J.; Brook, S.; Wang, F.; Adler, K.; Baut, E.; Scaltriti, M.; Jena, P.V.; et al. P-Selectin Is a Nanotherapeutic Delivery Target in the Tumor Microenvironment. Sci. Transl. Med. 2016, 8, 345ra87. [Google Scholar] [CrossRef] [PubMed]
  55. Ahmad, T.; Eapen, M.S.; Ishaq, M.; Park, A.Y.; Karpiniec, S.S.; Stringer, D.N.; Sohal, S.S.; Fitton, J.H.; Guven, N.; Caruso, V.; et al. Anti-Inflammatory Activity of Fucoidan Extracts in Vitro. Mar. Drugs 2021, 19, 702. [Google Scholar] [CrossRef] [PubMed]
  56. Carvalho, A.C.S.; Sousa, R.B.; Franco, Á.X.; Costa, J.V.G.; Neves, L.M.; Ribeiro, R.A.; Sutton, R.; Criddle, D.N.; Soares, P.M.G.; De Souza, M.H.L.P. Protective Effects of Fucoidan, a P- and L-Selectin Inhibitor, in Murine Acute Pancreatitis. Pancreas 2014, 43, 82–87. [Google Scholar] [CrossRef] [PubMed]
  57. Burns, A.R.; Bowden, R.A.; Abe, Y.; Walker, D.C.; Simon, S.I.; Entman, M.L.; Smith, C.W. P-Selectin Mediates Neutrophil Adhesion to Endothelial Cell Borders. J. Leukoc. Biol. 1999, 65, 299–306. [Google Scholar] [CrossRef] [PubMed]
  58. Etulain, J.; Martinod, K.; Wong, S.L.; Cifuni, S.M.; Schattner, M.; Wagner, D.D. P-Selectin Promotes Neutrophil Extracellular Trap Formation in Mice. Blood 2015, 126, 242–246. [Google Scholar] [CrossRef]
  59. Tchernychev, B.; Furie, B.; Furie, B.C. Peritoneal Macrophages Express Both P-Selectin and PSGL-1. J. Cell Biol. 2003, 163, 1145–1155. [Google Scholar] [CrossRef]
  60. Bonder, C.S.; Norman, M.U.; MacRae, T.; Mangan, P.R.; Weaver, C.T.; Bullard, D.C.; McCafferty, D.M.; Kubes, P. P-Selectin Can Support Both Th1 and Th2 Lymphocyte Rolling in the Intestinal Microvasculature. Am. J. Pathol. 2005, 167, 1647–1660. [Google Scholar] [CrossRef]
  61. Machelska, H.; Brack, A.; Mousa, S.A.; Schopohl, J.K.; Rittner, H.L.; Schäfer, M.; Stein, C. Selectins and Integrins but Not Platelet–Endothelial Cell Adhesion Molecule-1 Regulate Opioid Inhibition of Inflammatory Pain. Br. J. Pharmacol. 2004, 142, 772–780. [Google Scholar] [CrossRef] [PubMed]
  62. Chatterjea, D.; Wetzel, A.; Mack, M.; Engblom, C.; Allen, J.; Mora-Solano, C.; Paredes, L.; Balsells, E.; Martinov, T. Mast Cell Degranulation Mediates Compound 48/80-Induced Hyperalgesia in Mice. Biochem. Biophys. Res. Commun. 2012, 425, 237–243. [Google Scholar] [CrossRef] [PubMed]
  63. Finley, A.; Chen, Z.; Esposito, E.; Cuzzocrea, S.; Sabbadini, R.; Salvemini, D. Sphingosine 1-Phosphate Mediates Hyperalgesia via a Neutrophil-Dependent Mechanism. PLoS ONE 2013, 8, e55255. [Google Scholar] [CrossRef] [PubMed]
  64. Russell, F.A.; Fernandes, E.S.; Courade, J.P.; Keeble, J.E.; Brain, S.D. Tumour Necrosis Factor Alpha Mediates Transient Receptor Potential Vanilloid 1-Dependent Bilateral Thermal Hyperalgesia with Distinct Peripheral Roles of Interleukin-1beta, Protein Kinase C and Cyclooxygenase-2 Signalling. Pain 2009, 142, 264–274. [Google Scholar] [CrossRef] [PubMed]
  65. Verri, W.A.; Cunha, T.M.; Magro, D.A.; Guerrero, A.T.G.; Vieira, S.M.; Carregaro, V.; Souza, G.R.; Henriques, M.D.G.M.O.; Ferreira, S.H.; Cunha, F.Q. Targeting Endothelin ETA and ETB Receptors Inhibits Antigen-Induced Neutrophil Migration and Mechanical Hypernociception in Mice. Naunyn Schmiedebergs Arch. Pharmacol. 2009, 379, 271–279. [Google Scholar] [CrossRef] [PubMed]
  66. Dos Santos, D.F.D.S.; de Melo Aquino, B.; Jorge, C.O.; de Azambuja, G.; Schiavuzzo, J.G.; Krimon, S.; dos Santos Neves, J.; Parada, C.A.; Oliveira-Fusaro, M.C.G. Muscle Pain Induced by Static Contraction in Rats Is Modulated by Peripheral Inflammatory Mechanisms. Neuroscience 2017, 358, 58–69. [Google Scholar] [CrossRef] [PubMed]
  67. Oliveira, M.C.G.; Pelegrini-da-Silva, A.; Tambeli, C.H.; Parada, C.A. Peripheral Mechanisms Underlying the Essential Role of P2X3,2/3 Receptors in the Development of Inflammatory Hyperalgesia. Pain 2009, 141, 127–134. [Google Scholar] [CrossRef] [PubMed]
  68. Cunha, T.M.; Verri, W.A.; Schivo, I.R.; Napimoga, M.H.; Parada, C.A.; Poole, S.; Teixeira, M.M.; Ferreira, S.H.; Cunha, F.Q. Crucial Role of Neutrophils in the Development of Mechanical Inflammatory Hypernociception. J. Leukoc. Biol. 2008, 83, 824–832. [Google Scholar] [CrossRef] [PubMed]
  69. Machelska, H.; Cabot, P.J.; Mousa, S.A.; Zhang, Q.; Stein, C. Pain Control in Inflammation Governed by Selectins. Nat. Med. 1998, 4, 1425–1428. [Google Scholar] [CrossRef]
  70. Dell’Antonio, G.; Quattrini, A.; Dal Cin, E.; Fulgenzi, A.; Ferrero, M.E. Relief of Inflammatory Pain in Rats by Local Use of the Selective P2X7 ATP Receptor Inhibitor, Oxidized ATP. Arthritis Rheum. 2002, 46, 3378–3385. [Google Scholar] [CrossRef]
  71. Tambeli, C.H.; Oliveira, M.C.G.; Clemente, J.T.; Pelegrini-da-Silva, A.; Parada, C.A. A Novel Mechanism Involved in 5-Hydroxytryptamine-Induced Nociception: The Indirect Activation of Primary Afferents. Neuroscience 2006, 141, 1517–1524. [Google Scholar] [CrossRef] [PubMed]
  72. Oliveira, M.C.G.; Pelegrini-da-Silva, A.; Parada, C.A.; Tambeli, C.H. 5-HT Acts on Nociceptive Primary Afferents through an Indirect Mechanism to Induce Hyperalgesia in the Subcutaneous Tissue. Neuroscience 2007, 145, 708–714. [Google Scholar] [CrossRef] [PubMed]
  73. McNamee, K.E.; Alzabin, S.; Hughes, J.P.; Anand, P.; Feldmann, M.; Williams, R.O.; Inglis, J.J. IL-17 Induces Hyperalgesia via TNF-Dependent Neutrophil Infiltration. Pain 2011, 152, 1838–1845. [Google Scholar] [CrossRef] [PubMed]
  74. Perin-Martins, A.; Teixeira, J.M.; Tambeli, C.H.; Parada, C.A.; Fischer, L. Mechanisms Underlying Transient Receptor Potential Ankyrin 1 (TRPA1)-Mediated Hyperalgesia and Edema. J. Peripher. Nerv. Syst. 2013, 18, 62–74. [Google Scholar] [CrossRef] [PubMed]
  75. Zarpelon, A.C.; Cunha, T.M.; Alves-Filho, J.C.; Pinto, L.G.; Ferreira, S.H.; McInnes, I.B.; Xu, D.; Liew, F.Y.; Cunha, F.Q.; Verri, W.A. IL-33/ST2 Signalling Contributes to Carrageenin-Induced Innate Inflammation and Inflammatory Pain: Role of Cytokines, Endothelin-1 and Prostaglandin E2. Br. J. Pharmacol. 2013, 169, 90–101. [Google Scholar] [CrossRef] [PubMed]
  76. Teixeira, J.M.; de Oliveira-Fusaro, M.C.G.; Parada, C.A.; Tambeli, C.H. Peripheral P2X7 Receptor-Induced Mechanical Hyperalgesia Is Mediated by Bradykinin. Neuroscience 2014, 277, 163–173. [Google Scholar] [CrossRef] [PubMed]
  77. Guerrero, A.T.G.; Verri, W.A.; Cunha, T.M.; Silva, T.A.; Schivo, I.R.S.; Dal-Secco, D.; Canetti, C.; Rocha, F.A.C.; Parada, C.A.; Cunha, F.Q.; et al. Involvement of LTB4 in Zymosan-Induced Joint Nociception in Mice: Participation of Neutrophils and PGE2. J. Leukoc. Biol. 2008, 83, 122–130. [Google Scholar] [CrossRef] [PubMed]
  78. Sachs, D.; Coelho, F.M.; Costa, V.V.; Lopes, F.; Pinho, V.; Amaral, F.A.; Silva, T.A.; Teixeira, A.L.; Souza, D.G.; Teixeira, M.M. Cooperative Role of Tumour Necrosis Factor-α, Interleukin-1β and Neutrophils in a Novel Behavioural Model That Concomitantly Demonstrates Articular Inflammation and Hypernociception in Mice. Br. J. Pharmacol. 2011, 162, 72–83. [Google Scholar] [CrossRef]
  79. Pinto, L.G.; Cunha, T.M.; Vieira, S.M.; Lemos, H.P.; Verri, W.A.; Cunha, F.Q.; Ferreira, S.H. IL-17 Mediates Articular Hypernociception in Antigen-Induced Arthritis in Mice. Pain 2010, 148, 247–256. [Google Scholar] [CrossRef]
  80. Amaral, F.A.; Costa, V.V.; Tavares, L.D.; Sachs, D.; Coelho, F.M.; Fagundes, C.T.; Soriani, F.M.; Silveira, T.N.; Cunha, L.D.; Zamboni, D.S.; et al. NLRP3 Inflammasome–Mediated Neutrophil Recruitment and Hypernociception Depend on Leukotriene B4 in a Murine Model of Gout. Arthritis Rheum. 2012, 64, 474–484. [Google Scholar] [CrossRef]
  81. Yin, C.; Liu, B.; Li, Y.; Li, X.; Wang, J.; Chen, R.; Tai, Y.; Shou, Q.; Wang, P.; Shao, X.; et al. IL-33/ST2 Induces Neutrophil-Dependent Reactive Oxygen Species Production and Mediates Gout Pain. Theranostics 2020, 10, 12189–12203. [Google Scholar] [CrossRef] [PubMed]
  82. Guerrero, A.T.; Zaperlon, A.C.; Vieira, S.M.; Pinto, L.G.; Ferreira, S.H.; Cunha, F.Q.; Verri, W.A.; Cunha, T.M. The Role of PAF/PAFR Signaling in Zymosan-Induced Articular Inflammatory Hyperalgesia. Naunyn Schmiedebergs Arch. Pharmacol. 2013, 386, 51–59. [Google Scholar] [CrossRef]
  83. Oliveira-Fusaro, M.C.G.; Clemente-Napimoga, J.T.; Teixeira, J.M.; Torres-Chávez, K.E.; Parada, C.A.; Tambeli, C.H. 5-HT Induces Temporomandibular Joint Nociception in Rats through the Local Release of Inflammatory Mediators and Activation of Local β Adrenoceptors. Pharmacol. Biochem. Behav. 2012, 102, 458–464. [Google Scholar] [CrossRef] [PubMed]
  84. Zanelatto, F.B.; Dias, E.V.; Teixeira, J.M.; Sartori, C.R.; Parada, C.A.; Tambeli, C.H. Anti-Inflammatory Effects of Propranolol in the Temporomandibular Joint of Female Rats and Its Contribution to Antinociceptive Action. Eur. J. Pain 2018, 22, 572–582. [Google Scholar] [CrossRef] [PubMed]
  85. Teixeira, J.M.; Pimentel, R.M.; Abdalla, H.B.; de Sousa, H.M.X.; Macedo, C.G.; Napimoga, M.H.; Tambeli, C.H.; Oliveira-Fusaro, M.C.G.; Clemente-Napimoga, J.T. P2X7-Induced Nociception in the Temporomandibular Joint of Rats Depends on Inflammatory Mechanisms and C-Fibres Sensitization. Eur. J. Pain 2021, 25, 1107–1118. [Google Scholar] [CrossRef] [PubMed]
  86. Teixeira, J.M.; Parada, C.A.; Tambeli, C.H. P2X3 and P2X2/3 Receptors Activation Induces Articular Hyperalgesia by an Indirect Sensitization of the Primary Afferent Nociceptor in the Rats’ Knee Joint. Eur. J. Pharmacol. 2020, 879, 173054. [Google Scholar] [CrossRef] [PubMed]
  87. Schiavuzzo, J.G.; Teixeira, J.M.; Melo, B.; da Silva dos Santos, D.F.; Jorge, C.O.; Oliveira-Fusaro, M.C.G.; Parada, C.A. Muscle Hyperalgesia Induced by Peripheral P2X3 Receptors Is Modulated by Inflammatory Mediators. Neuroscience 2015, 285, 24–33. [Google Scholar] [CrossRef] [PubMed]
  88. Silva, J.R.; Lopes, A.H.; Talbot, J.; Cecilio, N.T.; Rossato, M.F.; Silva, R.L.; Souza, G.R.; Silva, C.R.; Lucas, G.; Fonseca, B.A.; et al. Neuroimmune-Glia Interactions in the Sensory Ganglia Account for the Development of Acute Herpetic Neuralgia. J. Neurosci. 2017, 37, 6408–6422. [Google Scholar] [CrossRef] [PubMed]
  89. Huerta, M.Á.; Cisneros, E.; Alique, M.; Roza, C. Strategies for Measuring Non-Evoked Pain in Preclinical Models of Neuropathic Pain: Systematic Review. Neurosci. Biobehav. Rev. 2024, 163, 105761. [Google Scholar] [CrossRef]
  90. Lavich, T.R.; Siqueira, R.d.A.; Farias-Filho, F.A.; Balão Cordeiro, R.S.; Rodrigues e Silva, P.M.; Martins, M.A. Neutrophil Infiltration Is Implicated in the Sustained Thermal Hyperalgesic Response Evoked by Allergen Provocation in Actively Sensitized Rats. Pain 2006, 125, 180–187. [Google Scholar] [CrossRef]
  91. Marotta, D.M.; Costa, R.; Motta, E.M.; Fernandes, E.S.; Medeiros, R.; Quintão, N.L.M.; Campos, M.M.; Calixto, J.B. Mechanisms Underlying the Nociceptive Responses Induced by Platelet-Activating Factor (PAF) in the Rat Paw. Biochem. Pharmacol. 2009, 77, 1223–1235. [Google Scholar] [CrossRef] [PubMed]
  92. McNamee, K.E.; Burleigh, A.; Gompels, L.L.; Feldmann, M.; Allen, S.J.; Williams, R.O.; Dawbarn, D.; Vincent, T.L.; Inglis, J.J. Treatment of Murine Osteoarthritis with TrkAd5 Reveals a Pivotal Role for Nerve Growth Factor in Non-Inflammatory Joint Pain. Pain 2010, 149, 386–392. [Google Scholar] [CrossRef]
  93. Albuquerque, I.R.L.; Cordeiro, S.L.; Gomes, D.L.; Dreyfuss, J.L.; Filgueira, L.G.A.; Leit, E.L.; Nader, H.B.; Rocha, H.A.O. Evaluation of Anti-Nociceptive and Anti-Inflammatory Activities of a Heterofucan from Dictyota Menstrualis. Mar. Drugs 2013, 11, 2722–2740. [Google Scholar] [CrossRef]
  94. Dornelas-Filho, A.F.; Pereira, V.B.M.; Wong, D.V.T.; Nobre, L.M.S.; Melo, A.T.; Silva, C.M.S.; Wanderley, C.W.S.; Nour, M.L.; Araújo, L.C.N.C.; Silva, R.O.; et al. Neutrophils Contribute to the Pathogenesis of Hemorrhagic Cystitis Induced by Ifosfamide. Int. Immunopharmacol. 2018, 62, 96–108. [Google Scholar] [CrossRef] [PubMed]
  95. Shaeib, F.; Khan, S.N.; Thakur, M.; Kohan-Ghadr, H.R.; Drewlo, S.; Saed, G.M.; Pennathur, S.; Abu-Soud, H.M. The Impact of Myeloperoxidase and Activated Macrophages on Metaphase II Mouse Oocyte Quality. PLoS ONE 2016, 11, e0151160. [Google Scholar] [CrossRef] [PubMed]
  96. Xu, D.; Wang, P.; Yang, J.; Qian, Q.; Li, M.; Wei, L.; Xu, W. Gr-1+ Cells Other Than Ly6G+ Neutrophils Limit Virus Replication and Promote Myocardial Inflammation and Fibrosis Following Coxsackievirus B3 Infection of Mice. Front. Cell Infect. Microbiol. 2018, 8, 157. [Google Scholar] [CrossRef] [PubMed]
  97. Torres, S.R.; Pichowicz, A.; Torres-Velez, F.; Song, R.; Singh, N.; Lasek-Nesselquist, E.; De Jesus, M. Impact of Candida Auris Infection in a Neutropenic Murine Model. Antimicrob. Agents Chemother. 2020, 64, 10-1128. [Google Scholar] [CrossRef] [PubMed]
  98. Wanderley, C.W.S.; Silva, C.M.S.; Wong, D.V.T.; Ximenes, R.M.; Morelo, D.F.C.; Cosker, F.; Aragão, K.S.; Fernandes, C.; Palheta-Júnior, R.C.; Havt, A.; et al. Bothrops Jararacussu Snake Venom-Induces a Local Inflammatory Response in a Prostanoid- and Neutrophil-Dependent Manner. Toxicon 2014, 90, 134–147. [Google Scholar] [CrossRef] [PubMed]
  99. Park, J.; Cha, J.D.; Choi, K.M.; Lee, K.Y.; Han, K.M.; Jang, Y.S. Fucoidan Inhibits LPS-Induced Inflammation in Vitro and during the Acute Response in Vivo. Int. Immunopharmacol. 2017, 43, 91–98. [Google Scholar] [CrossRef]
  100. Ossipov, M.H.; Morimura, K.; Porreca, F. Descending Pain Modulation and Chronification of Pain. Curr. Opin. Support. Palliat. Care 2014, 8, 143–151. [Google Scholar] [CrossRef]
  101. Lv, Q.; Wu, F.; Gan, X.; Yang, X.; Zhou, L.; Chen, J.; He, Y.; Zhang, R.; Zhu, B.; Liu, L. The Involvement of Descending Pain Inhibitory System in Electroacupuncture-Induced Analgesia. Front. Integr. Neurosci. 2019, 13, 466401. [Google Scholar] [CrossRef]
  102. Kapitzke, D.; Vetter, I.; Cabot, P.J. Endogenous Opioid Analgesia in Peripheral Tissues and the Clinical Implications for Pain Control. Ther. Clin. Risk Manag. 2005, 1, 279. [Google Scholar] [PubMed]
  103. Ossipov, M.H. The Perception and Endogenous Modulation of Pain. Scientifica 2012, 2012, 561761. [Google Scholar] [CrossRef] [PubMed]
  104. Holden, J.E.; Jeong, Y.; Forrest, J.M. The Endogenous Opioid System and Clinical Pain Management. AACN Clin. Issues 2005, 16, 291–301. [Google Scholar] [CrossRef] [PubMed]
  105. Benedetti, F. Placebo and Endogenous Mechanisms of Analgesia. Handb. Exp. Pharmacol. 2006, 177, 393–413. [Google Scholar] [CrossRef]
  106. Martins, D.F.; Bobinski, F.; Mazzardo-Martins, L.; Cidral-Filho, F.J.; Nascimento, F.P.; Gadotti, V.M.; Santos, A.R.S. Ankle Joint Mobilization Decreases Hypersensitivity by Activation of Peripheral Opioid Receptors in a Mouse Model of Postoperative Pain. Pain Med. 2012, 13, 1049–1058. [Google Scholar] [CrossRef]
  107. Cidral-Filho, F.J.; Mazzardo-Martins, L.; Martins, D.F.; Santos, A.R.S. Light-Emitting Diode Therapy Induces Analgesia in a Mouse Model of Postoperative Pain through Activation of Peripheral Opioid Receptors and the L-Arginine/Nitric Oxide Pathway. Lasers Med. Sci. 2014, 29, 695–702. [Google Scholar] [CrossRef] [PubMed]
  108. Martins, D.F.; Turnes, B.L.; Cidral-Filho, F.J.; Bobinski, F.; Rosas, R.F.; Danielski, L.G.; Petronilho, F.; Santos, A.R.S. Light-Emitting Diode Therapy Reduces Persistent Inflammatory Pain: Role of Interleukin 10 and Antioxidant Enzymes. Neuroscience 2016, 324, 485–495. [Google Scholar] [CrossRef] [PubMed]
  109. Rittner, H.L.; Labuz, D.; Schaefer, M.; Mousa, S.A.; Schulz, S.; Schäfer, M.; Stein, C.; Brack, A.; Rittner, H.L.; Labuz, D.; et al. Pain Control by CXCR2 Ligands through Ca2+-Regulated Release of Opioid Peptides from Polymorphonuclear Cells. FASEB J. 2006, 20, 2627–2629. [Google Scholar] [CrossRef]
  110. Giorgi, R.; Pagano, R.L.; Amorim Dias, M.A.; Aguiar-Passeti, T.; Sorg, C.; Mariano, M. Antinociceptive Effect of the Calcium-Binding Protein MRP-14 and the Role Played by Neutrophils on the Control of Inflammatory Pain. J. Leukoc. Biol. 1998, 64, 214–220. [Google Scholar] [CrossRef]
  111. Pagano, R.L.; Dias, M.A.A.; Dale, C.S.; Giorgi, R. Neutrophils and the Calcium-Binding Protein MRP-14 Mediate Carrageenan-Induced Antinociception in Mice. Mediat. Inflamm. 2002, 11, 203–210. [Google Scholar] [CrossRef]
  112. Hu, C.; Zhang, G.; Zhao, Y. tao Fucoidan Attenuates the Existing Allodynia and Hyperalgesia in a Rat Model of Neuropathic Pain. Neurosci. Lett. 2014, 571, 66–71. [Google Scholar] [CrossRef] [PubMed]
  113. Hu, C.; Zhao, Y.T.; Zhang, G.; Xu, M.F. Antinociceptive Effects of Fucoidan in Rat Models of Vincristine-Induced Neuropathic Pain. Mol. Med. Rep. 2017, 15, 975–980. [Google Scholar] [CrossRef] [PubMed]
  114. Zhang, X.W.; Liu, Q.; Thorlacius, H. Inhibition of Selectin Function and Leukocyte Rolling Protects against Dextran Sodium Sulfate-Induced Murine Colitis. Scand. J. Gastroenterol. 2001, 36, 270–275. [Google Scholar] [CrossRef] [PubMed]
  115. Obluchinskaya, E.D.; Pozharitskaya, O.N.; Flisyuk, E.V.; Shikov, A.N. Formulation, Optimization and In Vivo Evaluation of Fucoidan-Based Cream with Anti-Inflammatory Properties. Mar. Drugs 2021, 19, 643. [Google Scholar] [CrossRef] [PubMed]
  116. Piñeiro-Ramil, M.; Flórez-Fernández, N.; Ramil-Gómez, O.; Torres, M.D.; Dominguez, H.; Blanco, F.J.; Meijide-Faílde, R.; Vaamonde-García, C. Antifibrotic Effect of Brown Algae-Derived Fucoidans on Osteoarthritic Fibroblast-like Synoviocytes. Carbohydr. Polym. 2022, 282, 119134. [Google Scholar] [CrossRef] [PubMed]
  117. Myers, S.P.; O’Connor, J.; Fitton, J.H.; Brooks, L.; Rolfe, M.; Connellan, P.; Wohlmuth, H.; Cheras, P.A.; Morris, C. A Combined Phase I and II Open Label Study on the Effects of a Seaweed Extract Nutrient Complex on Osteoarthritis. Biologics 2010, 4, 33–44. [Google Scholar] [CrossRef] [PubMed]
  118. Kan, J.; Cheng, J.; Xu, L.; Hood, M.; Zhong, D.; Cheng, M.; Liu, Y.; Chen, L.; Du, J. The Combination of Wheat Peptides and Fucoidan Protects against Chronic Superficial Gastritis and Alters Gut Microbiota: A Double-Blinded, Placebo-Controlled Study. Eur. J. Nutr. 2020, 59, 1655–1666. [Google Scholar] [CrossRef] [PubMed]
  119. Tay, A.; Jiang, Y.; Signal, N.; O’Brien, D.; Chen, J.; Murphy, R.; Lu, J. Combining Mussel with Fucoidan as a Supplement for Joint Pain and Prediabetes: Study Protocol for a Randomized, Double-Blinded, Placebo-Controlled Trial. Front. Nutr. 2022, 9, 1000510. [Google Scholar] [CrossRef]
  120. Chadwick, C.; De Jesus, M.; Ginty, F.; Martinez, J.S. Pathobiology of Candida Auris Infection Analyzed by Multiplexed Imaging and Single Cell Analysis. PLoS ONE 2024, 19, e0293011. [Google Scholar] [CrossRef]
  121. Parisien, M.; Lima, L.V.; Dagostino, C.; El-Hachem, N.; Drury, G.L.; Grant, A.V.; Huising, J.; Verma, V.; Meloto, C.B.; Silva, J.R.; et al. Acute Inflammatory Response via Neutrophil Activation Protects against the Development of Chronic Pain. Sci. Transl. Med. 2022, 14, eabj9954. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, J. Neutrophils in Tissue Injury and Repair. Cell Tissue Res. 2018, 371, 531. [Google Scholar] [CrossRef] [PubMed]
  123. Wilgus, T.A.; Roy, S.; McDaniel, J.C. Neutrophils and Wound Repair: Positive Actions and Negative Reactions. Adv. Wound Care 2013, 2, 379–388. [Google Scholar] [CrossRef] [PubMed]
  124. Vo, T.S.; Kim, S.K. Fucoidans as a Natural Bioactive Ingredient for Functional Foods. J. Funct. Foods 2013, 5, 16–27. [Google Scholar] [CrossRef]
  125. Hu, T.; Liu, D.; Chen, Y.; Wu, J.; Wang, S. Antioxidant Activity of Sulfated Polysaccharide Fractions Extracted from Undaria Pinnitafida in Vitro. Int. J. Biol. Macromol. 2010, 46, 193–198. [Google Scholar] [CrossRef] [PubMed]
  126. Mourao, P. Use of Sulfated Fucans as Anticoagulant and Antithrombotic Agents: Future Perspectives. Curr. Pharm. Des. 2004, 10, 967–981. [Google Scholar] [CrossRef] [PubMed]
  127. Luthuli, S.; Wu, S.; Cheng, Y.; Zheng, X.; Wu, M.; Tong, H. Therapeutic Effects of Fucoidan: A Review on Recent Studies. Mar. Drugs 2019, 17, 487. [Google Scholar] [CrossRef] [PubMed]
  128. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 Statement: An Updated Guideline for Reporting Systematic Reviews. BMJ 2021, 372, n71. [Google Scholar] [CrossRef] [PubMed]
  129. Viechtbauer, W. Conducting Meta-Analyses in R with the Metafor Package. J. Stat. Softw. 2010, 36, 1–48. [Google Scholar] [CrossRef]
  130. Harrer, M.; Cuijpers, P.; Furukawa, T.; Ebert, D. Doing Meta-Analysis with R: A Hands-On Guide; Chapman and Hall/CRC: London, UK; Boca Raton, FL, USA, 2021; ISBN 9780367610074. [Google Scholar]
  131. Vesterinen, H.M.; Sena, E.S.; Egan, K.J.; Hirst, T.C.; Churolov, L.; Currie, G.L.; Antonic, A.; Howells, D.W.; Macleod, M.R. Meta-Analysis of Data from Animal Studies: A Practical Guide. J. Neurosci. Methods 2014, 221, 92–102. [Google Scholar] [CrossRef]
  132. Cumpston, M.; Li, T.; Page, M.J.; Chandler, J.; Welch, V.A.; Higgins, J.P.; Thomas, J. Updated Guidance for Trusted Systematic Reviews: A New Edition of the Cochrane Handbook for Systematic Reviews of Interventions. Cochrane Database Syst. Rev. 2019, 2019, ED000142. [Google Scholar] [CrossRef] [PubMed]
  133. Higgins, J.P.T.; Thompson, S.G.; Deeks, J.J.; Altman, D.G. Measuring Inconsistency in Meta-Analyses. BMJ 2003, 327, 557–560. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Study selection PRISMA flow diagram.
Figure 1. Study selection PRISMA flow diagram.
Marinedrugs 22 00290 g001
Figure 2. Forest plot of the pain reduction efficacy of pretreatment with fucoidan. CI, confidence interval; RE, random effects [62,63,64,65,66,68,69,71,72,73,74,75,76,77,78,79,80,82,85,86,87,90,91,92,94].
Figure 2. Forest plot of the pain reduction efficacy of pretreatment with fucoidan. CI, confidence interval; RE, random effects [62,63,64,65,66,68,69,71,72,73,74,75,76,77,78,79,80,82,85,86,87,90,91,92,94].
Marinedrugs 22 00290 g002
Table 1. Information on studies using fucoidan to evaluate the role of neutrophils in pain.
Table 1. Information on studies using fucoidan to evaluate the role of neutrophils in pain.
StudySpecies; StrainDose; Route; Administration Time before Injury; Positive ControlNeutrophil Count; Efficacy/Selectivity, % Neutrophil DepletionPain ModelPain OutcomeAnalgesic Efficacy (p > 0.05)
Machelsa et al., 1998 [69]Rat; Wistar10 mg/kg; i.v.; 10 min; fentanylManual count (β-endorphin) (≈40%)CFA (i.pl.)MHNo
* Dell’Antonio et al., 2002 [70]Rat; Wistar10 mg/kg; i.v.; 30 min; oxidized ATPNon-evaluatedCFA (i.pl.)MHNo
Tambeli et al., 2006 [71]Rat; Wistar20 mg/kg; i.v.; 30 min; tropisetron, indomethacin, and atenololMPO; yes/no (≈60%)5-HT (i.pl.)NB (Paw)Yes
Lavich et al., 2006 [90]Rat; Wistar10 mg/kg, i.v.; 15 min; anti-neutrophil serumManual count and MPO; (>90%)Ovalbumin probe in sensitized ratsHHYes
Oliveira et al., 2007 [72]Rat; Wistar20 mg/kg, i.v.; 20 min; tropisetron, indomethacin, guanethidine, and atenololNon-evaluated5-HT; PGE; epinephrine (i.pl.)MHYes; no; no
Cunha et al., 2008 [68]Rat; Wistar20 mg/kg; i.v.; 10 min; no positive controlMPO; yes/no (=90%)Carrageenan (i.pl.)MHYes
Guerrero et al., 2008 [77]Mouse; C57BL/620 mg/kg; i.v.; 20 min; indomethacin, MK886, celecoxib, anti-neutrophil antibodyMPO; yes/yesLTB4; zymosan (i.a.)MHYes
Russell et al., 2009 [64]Mouse; CD140 mg/kg; i.v.; 20 min; SB366791, TNF inhibitor, PKC inhibitor, indomethacin, COX2 inhibitor, and nimesulideMPO; yes/no (=70%)TNF (i.pl.)HHYes
Verri et al., 2009 [65]Mouse; BALB/c20 mg/kg; i.v.; 15 min; bosentan, BQ123, and BQ788MPO; yes/no (=70%)Carrageenan (i.pl.)MHYes
Marotta et al., 2009 [91]Rat; Wistar10 mg/kg; i.v.; 15 min; WEB2086, anti-TNF, IL-1 antagonist, indomethacin, and celecoxibMPO; yes/no (=90%)PAF (i.pl.)MH; NB (Paw)Yes; no
McNamee et al., 2010 [92]Mouse; C57BL/620 mg/kg; i.p.; 24 h
before every 2 d; anti-NGF, and anti-TNF
MPO; yes/no (50%)DMM surgery; OANB (WB)Yes
Pinto et al., 2010 [79]Mouse; BALB/c20 mg/kg; i.v.; 15 min before and 3.5 h after; infliximab, IL-1 antagonist, and DF2156Manual count (May–Grünwald–Giemsa); yes/no (80%)AIA-mBSA or IL-17 (i.a.)MHYes
Sachs et al., 2011 [78]Mouse; C57Bl/6J20 mg/kg; i.v.; 10 min; lidocaine, morphine, dexamethasone, anti-TNF, and IL-1 antagonistMPO; yes/no (>90%)AIA-mBSA (i.a.)MHYes
McNamee et al., 2011 [73]Mouse; C57BL/620 mg/kg; i.p.; 2 h; anti-TNFMPO; yes/no (>90%)TNF; IL-17 (i.pl.)HH; NB (WB)Yes
Amaral et al., 2012 [80]Mouse; C57BL/6J20 mg/kg; i.v.; 15 min; IL-1 antagonist, DF2162, CP105,696, and MK886MPO; yes/no (>90%)MSU crystals (i.a.)MHYes
Chatterjea et al., 2012 [62]Mouse; ND420 mg/kg; p.o.; 30 min; sodium cromoglycateMPO; Hematoxylin-
Eosin; yes/no (>90%)
Compound 48/80 (i.pl.)HHYes
* Oliveira-Fusaro et al., 2012 [83]Rat; Wistar20 mg/kg; i.v.; 20 min; tropisetron, guanethidine, atenolol, and indomethacinMPO; yes/no (>90%)5-HT (i.a.)NB (Face)No
Finley et al., 2013 [63]Rat; Sprague Dawley40 mg/kg; i.p.; 30 min; sphingosine-1 antagonists and fingolimodMPO; Hematoxylin-
Eosin; yes/no (>60%)
Carrageenan; S1P; SEW2871 (i.pl.)HHYes
Guerrero et al., 2013 [82]Mouse; BALB/C20 mg/kg; i.v.; 15 min; indomethacin, MK886, selective PAFR antagonistsMPO; no/noPAF (i.a.)MHYes
Perin-Martins et al., 2013 [74]Rat; Wistar20 mg/kg; i.v.; 30 min; TRPA1 antagonist, CGRP antagonist, indomethacinMPO; yes/no (100%)Allyl isothiocyanate (i.pl.)MHYes
Zarpelon et al., 2013 [75]Mouse; BALB/C20 mg/kg; i.v.; 15 min; infliximab, IL1 antagonist, anti-CXCL1, indomethacin, BQ788, and clazosentanMPO; yes/no (>90%)IL–33 (i.pl.)MHYes
* Albuquerque et al., 2013 [93]Mouse; BALB/C20 mg/kg; i.v.; 30 min; morphine and dipyroneManual counting; yes/no (>90%)Acetic acid (i.p.)NB (writhing)Yes
Teixeira et al., 2014 [76]Rat; Wistar25 mg/kg, i.v., 20 min; P2X7 and P2X1,3,2/3 antagonists, atenolol, and indomethacinNon-evaluatedBzATP (i.pl.)MHYes
Schiavuzzo et al., 2015 [87]Rat; Wistar25 mg/kg; i.v.; 20 min; P2X1,3,2/3 antagonist, lidocaine, bradykinin antagonist, and indomethacinMPO; no/noα,β-meATP (i.m.)MHYes
Silva et al., 2017 [88]Mouse; C57BL/620 mg/kg; i.v.; daily from 2 to 7 d; dexamethasone, morphine, lidocaine, infliximab, etanercept, clodronate, and indomethacinFlow cytometry; yes/yes
(>90% reduced CD45+ cells)
HSV-1 (postherpetic neuralgia)MAYes
Santos et al., 2017 [66]Rat; Wistar25 mg/kg; i.v.; 20 min; dexamethasone, bradykinin antagonist, atenolol, indomethacin, and ICI118MPO; no/noSustained muscle contractionMHYes
Dornelas-Filho et al., 2018 [94]Mouse; Swiss100 mg/kg; i.v.; 30 min; anti-neutrophil antibodyMPO; flow cytometry (>90%)Ifosfamide-induced hemorrhagic cystitisMHYes
Zanelatto et al., 2018 [84]Rat; Wistar20 mg/kg; i.v.; 20 min; propranolol and thalidomideMPO; no/noIsoproterenol (i.a.)NB (Face)Yes
Yin et al., 2020 [81]Mouse; C57BL/6J20 mg/kg; i.v.; 8 h y 23 h after; clodronateMPO; yes/no (>95%)MSU crystals (i.a.)MAYes
Teixeira et al., 2020 [86]Rat; Wistar25 mg/kg, i.v.; 30 min; P2X3 and P2X2/3 antagonistMPO; yes/no (100%)αβ-meATP (i.a.)NB (WB)Yes
Teixeira et al., 2021 [85]Rat; Wistar25 mg/kg; i.v.; 20 min; P2X7 antagonist, lidocaine, atenolol, and indomethacinManual count; no/noBzATP (i.a.)NB (Face)Yes
Abbreviations: 5-HT: 5-hydroxytryptamine; α,β-meATP: α,β-methylene ATP; AIA: antigen-induced arthritis; BzATP: 3′-O-(4-Benzoyl)benzoyladenosine 5′-triphosphate; CFA: complete Freund’s adjuvant; DMM: destabilization of the medial meniscus; Face: facial grooming; HH: heat hyperalgesia; HSV-1: herpes simplex virus type 1; i.a.: intra-articular; IL-17: interleukin 17; i.m.: intramuscular; i.p.: intraperitoneal; i.pl.: intraplantar; i.v.: intravenous; LTB4: leukotriene B4; MA: mechanical allodynia; mBSA: methylated bovine serum albumin; MH: mechanical hyperalgesia; MPO: myeloperoxidase assay; MSU: monosodium urate; NB: nociceptive behavior; PAF: platelet-activating factor; p.o.: oral; Paw: paw flinching; S1P: sphingosine-1-phosphate; TNF: tumor necrosis factor; WB: weight bearing distribution. * Articles not included in this meta-analysis as one of the comparison groups was missing.
Table 2. Information on studies using fucoidan to evaluate endogenous opioid analgesia.
Table 2. Information on studies using fucoidan to evaluate endogenous opioid analgesia.
StudySpecies; StrainFucoidan Dose; Route; %
Neutrophil Reduction
Pain ModelPain OutcomeMain Results
Machelsa et al., 1998 [69]Rat; Wistar10 mg/kg; i.v.; ≈40% indirect reduction (β-endorphin)CFA (i.pl.)MHDecreased analgesia induced by cold water swim stress and CRF
Machelsa et al., 2004 [17]Rat; Wistar10 mg/kg; i.v.; ≈50% (macrophage and T cell reduction)CFA (i.pl.)MHAbolished peripheral stress-induced antinociception
Martins et al., 2012 [106]Mouse; Swiss100 µg/mouse; i.p.; non-evaluatedPIMHDid not reverse analgesia induced by ankle joint mobilization
Cidral-Filho et al., 2013 [107]Mouse; Swiss100 μg/mouse; i.p.; non-evaluatedPIMHAbolished the analgesic effect of light-emitting diode therapy
Martins et al., 2016 [108]Mouse; Swiss100 μg/mouse; i.p.; non-evaluatedCFA (i.pl.)MHAbolished the analgesic effect of light-emitting diode therapy
Abbreviations: CFA: complete Freund’s adjuvant; CRF: corticotropin-releasing factor; i.p.: intraperitoneal; i.pl.: intraplantar; i.v.: intravenous; MH: mechanical hyperalgesia.
Table 3. Therapeutic use of fucoidan in pain-related diseases.
Table 3. Therapeutic use of fucoidan in pain-related diseases.
StudyPathological ModelIntervention; Positive Control Main Results
Zhang et al., 2001 [114]Dextran sodium sulfate-induced murine colitisFucoidan daily (25 mg/kg, i.v.) starting immediately prior to the 5-day challenge; DF2162 and MK886 and CP105,696Fucoidan can reduce mucosal damage and crypt destruction in the colon of dextran sodium sulfate-treated mice, relieving chronic colitis.
Hu et al., 2014 [112]Peripheral nerve injury produced by spinal nerve ligationFucoidan (15, 50 and 100 mg/kg, i.t.) once daily during the period of days 11–20, inclusively; no positive controlAttenuated the existing allodynia and hyperalgesia induced by nerve injury. Also inhibited cytokines production, glial. and ERK activation.
Hu et al., 2017 [113]Chemotherapy-induced peripheral neuropathy (vincristine)Fucoidan (50, 100 or 200 mg/kg, i.p.) on single treatment (day 14) or repeated treatment once daily for 14 days; pregabalinRepeated. However, no single treatment attenuated vincristine-induced mechanical and cold allodynia in a dose-dependent manner.
Wang et al., 2018 [33]Peripheral arterial disease by injection of sodium laurate into femoral arteryLow molecular weight fucoidan (20, 40 or 80 mg/kg/day; p.o.) for 4 weeks; cilostazolAmeliorated foot ulceration and improved plantar perfusion. Suppressed the upregulation of inflammatory factors (ICAM-1 and IL-1β) in the gastrocnemius muscles of ischemic hindlimb.
Ahn et al., 2019 [34]Transient global cerebral ischemia in obese gerbilsFucoidan (50 mg/kg; p.o.) daily for the last 5 days; no positive controlRelieved acceleration and exacerbation of ischemic brain injury in an obese state via the attenuation of obesity-induced severe oxidative damage.
Obluchinskaya et al., 2021 [115]Peripheral inflammation carrageenan (i.pl.)Fucoidan-based cream 100, 200, and 400 mg/rat/day (topical) for 5 days; diclofenacInhibited carrageenan-induced edema and ameliorated mechanical allodynia (efficacy comparable with diclofenac).
Zhang et al., 2022 [31]Experimental autoimmune prostatitisFucoidan 20 mg/kg (i.p.) one day before disease induction and then once a week; anti-neutrophil antibodyHistological appearance of prostate tissues improved, and chronic pain development was ameliorated.
Li et al., 2023 [32]Intervertebral disc degenerationSingle treatment with 5 μL of fucoidan (10 μg/mL; intra-disc injection); no positive controlAmeliorated intervertebral disc degeneration 4 and 8 days after injury, and preserved disc height, extracellular matrix components, and nucleus pulposus hydration.
Abbreviations: ERK: Extracellular-Signal-Regulated Kinase; ICAM-1: Intercellular Adhesion Molecule 1; IL-1β: interleukin 1β; i.p.: intraperitoneal; i.pl.: intraplantar; i.v.: intravenous; p.o.: oral administration.
Table 4. Human studies evaluating fucoidan for pain-related diseases.
Table 4. Human studies evaluating fucoidan for pain-related diseases.
StudyStudy Type; PopulationIntervention; Route; DosageMain Efficacy ResultsMain Safety Results
Myers et al., 2010 [117]Open-label, randomized, combined Phase I and II; knee osteoarthritis (n = 10 adults 9F/1M)100 and 1000 mg of fucoidan; p.o.; daily for 4 weeksReduced COAT score by 18% for the 100 mg treatment and 52% for the 1000 mg dose. Clear dose response in all COAT subscales (pain, stiffness, physical activity and overall symptom severity)Well tolerated (few adverse events related to treatment). No changes in blood parameters
Myers et al., 2016 [40]Randomized placebo-controlled trial; hip/knee osteoarthritis (n = 96 adults 56F/40M)300 mg dose of a Fucus vesiculosus extract (85% fucoidan); p.o.; daily for 12 weeksFucoidan improved by 29% COAT score (knee), while placebo improved it by 30.6%. No significant differences in symptom reduction vs. placebo. No difference in the usage of paracetamolIt was safe and well tolerated
Kan et al., 2020 [118]Randomized, double-blind, placebo-controlled trial; chronic gastritis (n = 101 adults)Wheat peptides and fucoidan; p.o.; once daily for 45 daysReduced gastric mucosal damage in 70% of subjects. Significantly less stomach pain, belching, bloating, acid reflux, appetite loss, increased food intake, and higher quality of life (p > 0.05 for all)No adverse event reported
Tay et al., 2022 [119]Randomized, double-blind, placebo-controlled trial; prediabetes and hip or knee joint pain
(n = 150 adults)
20 g of chocolate (1000 mg mussel powder and 1000 mg of fucoidan); p.o.; once daily for 100 daysResults not available; the primary endpoints are change in insulin resistance and patient-reported joint pain. Secondary endpoints include anthropometry, fasting glucose and insulin, HbA1c, inflammatory markers, satiety, quality of life, physical function, pain intensity, and analgesic medication useResults not available; complications reported and described by duration, severity, outcome, treatment, and relation to study treatment or cause
Abbreviations: COAT: comprehensive arthritis test; p.o.: oral administration.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Huerta, M.Á.; Tejada, M.Á.; Nieto, F.R. Fucoidan as a Promising Drug for Pain Treatment: Systematic Review and Meta-Analysis. Mar. Drugs 2024, 22, 290. https://0-doi-org.brum.beds.ac.uk/10.3390/md22070290

AMA Style

Huerta MÁ, Tejada MÁ, Nieto FR. Fucoidan as a Promising Drug for Pain Treatment: Systematic Review and Meta-Analysis. Marine Drugs. 2024; 22(7):290. https://0-doi-org.brum.beds.ac.uk/10.3390/md22070290

Chicago/Turabian Style

Huerta, Miguel Á., Miguel Á. Tejada, and Francisco R. Nieto. 2024. "Fucoidan as a Promising Drug for Pain Treatment: Systematic Review and Meta-Analysis" Marine Drugs 22, no. 7: 290. https://0-doi-org.brum.beds.ac.uk/10.3390/md22070290

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop