International Journal of Molecular Medicine is an international journal devoted to molecular mechanisms of human disease.
International Journal of Oncology is an international journal devoted to oncology research and cancer treatment.
Covers molecular medicine topics such as pharmacology, pathology, genetics, neuroscience, infectious diseases, molecular cardiology, and molecular surgery.
Oncology Reports is an international journal devoted to fundamental and applied research in Oncology.
Experimental and Therapeutic Medicine is an international journal devoted to laboratory and clinical medicine.
Oncology Letters is an international journal devoted to Experimental and Clinical Oncology.
Explores a wide range of biological and medical fields, including pharmacology, genetics, microbiology, neuroscience, and molecular cardiology.
International journal addressing all aspects of oncology research, from tumorigenesis and oncogenes to chemotherapy and metastasis.
Multidisciplinary open-access journal spanning biochemistry, genetics, neuroscience, environmental health, and synthetic biology.
Open-access journal combining biochemistry, pharmacology, immunology, and genetics to advance health through functional nutrition.
Publishes open-access research on using epigenetics to advance understanding and treatment of human disease.
An International Open Access Journal Devoted to General Medicine.
Psoriasis is a chronic immune-mediated inflammatory skin disease characterized by keratinocyte hyperproliferation and infiltration of inflammatory cells, including CD4+ T and mast cells, into the skin (1). Although the precise molecular mechanisms underlying psoriasis pathogenesis are not completely understood, accumulating evidence highlights a key role for T helper 17 (Th17) cells as the primary source of IL-17, a key cytokine in psoriasis development (2,3). Th17 cells differentiate from naïve CD4+ T cells primarily in response to IL-23 through activation of the JAK2/STAT3 signaling pathway (4,5). Thus, the IL-23/IL-17 axis is key to the immunopathogenesis of psoriasis (4). During disease onset, proinflammatory cytokines such as tumor necrosis factor (TNF)-α and IL-1β promote abnormal keratinocyte proliferation and differentiation (6). These cytokines suppress keratin 10 expression while upregulating involucrin and keratins 6, 16 and 17, which serve as molecular markers of psoriatic lesions (7). This dysregulation reflects keratinocyte activation and contributes to hallmark histopathological features of psoriasis, including epidermal hyperplasia, parakeratosis and acanthosis (8).
Although various therapeutic agents for psoriasis such as methotrexate, cyclosporine, retinoic acid, vitamin D3 analogs and biological agents such as alefacept are widely used, a definitive cure remains elusive (2,3). Moreover, long-term use of certain agents, including cyclosporine and retinoic acid, is associated with significant adverse effects (9). As a result, alternative strategies, including combination therapies involving conventional and biological agents, have been explored. However, these approaches are limited by cumulative toxicity and safety concerns (10). Therefore, the development of novel, safe and effective therapeutic options for psoriasis remains a clinical need.
Marine algae are regarded as the ‘plant-based therapy of the future’ due to their abundance of health-promoting compounds with activity against various diseases and disorders (11). One of the most notable components of seaweeds is their high content of sulfated polysaccharides (SPs). SPs derived from seaweeds exhibit a broad spectrum of biological activities, including anti-inflammatory, immunomodulatory, antioxidant and antiviral effects, making them promising candidates for pharmaceutical and nutraceutical applications (12). Gracilaria fisheri, a species commonly found in Thailand, is rich in sulfated galactan (SG), a type of SP known for its diverse biological properties, especially its anti-inflammatory and immunomodulatory effects (13). Determining molecular characteristics such as molecular weight is key for enhancing the biological activity of SPs (14). Our previous study prepared low molecular weight SG (LSG) from G. fisheri and demonstrated its superior antioxidant activity compared with SG (15). Oligosaccharides derived from G. fisheri modulate immune responses by inhibiting proinflammatory cytokines such as TNF-α and IL-1β, which are key mediators in psoriasis pathogenesis (13). Moreover, SG has been shown to interfere with signaling pathways such as NF-κB and JAK/STAT, which serve central roles in the regulation of inflammatory gene expression (16,17).
Given these properties, LSG derived from G. fisheri presents a promising candidate for the development of novel anti-psoriatic agents. The present study employed an imiquimod (IMQ)-induced psoriasis-like mouse model to evaluate the therapeutic potential of LSG from G. fisheri.
LSG was prepared using a previously established method (18). SG was isolated from the red seaweed G. fisheri using a cold-water extraction method as described by Wongprasert et al (19). SG was stirred in 0.1 M HCl (RCI Labscan Ltd.; ratio 10:1) for 6 h at room temperature. The mixture was neutralized to pH 8 and precipitated with 95% ethanol (RCILabscan). The resulting pellet was collected, re-suspended in distilled water and dialyzed against distilled water in a dialysis bag for 24 h. LSG was obtained by freeze-drying overnight at -60˚C. The chemical structure and molecular weight of LSG were confirmed by fourier transform infrared (FTIR), nuclear magnetic resonance (NMR) and gel permeation chromatography (GPC) analyses. LSG (2 mg) was analyzed using FTIR spectroscopy with an IRAffinity-1S (IR) and MIRacle 10 (ATR; Shimadzu). The compound was transferred to a film and analyzed. Spectral baselines were corrected to the 400-4,000 cm-¹ range. For NMR analysis, LSG (10 mg) was dissolved in deuterium oxide (D2O, 99.9%; 0.5 ml) in 5 mm NMR tubes and analyzed using a Varian Bruker 400 MHz spectrometer. For GPC, LSG was dissolved in deionized water (1:1 w/v), and 20 µl of the solution was analyzed using a Shimadzu LC-20AD system with an LC-20A oven column and a RID-10A detector, equipped with a TSKgel Guard PWH Size Exclusion Guard column. The column temperature was maintained at 60.0±0.1˚C.
Male BALB/c mice (n=48; age, 6-8 weeks; average weight, 23.96±1.34 grams) were purchased from Nomura International Siam (Bangkok, Thailand). All animals were housed under SPF conditions with controlled humidity (50-70%), temperature (25±2˚C) and a 12/12-h light/dark cycle. Mice had free access to a standard pellet diet and water ad libitum throughout the experiment. All experimental procedures were approved by the Ethical Committee of the University of Phayao, Phayao, Thailand (approval no. 1-024-65).
The dorsal hair of BALB/c mice was removed 1 day before starting the experiment. IMQ cream (5% IMQ; Aldara™, Ensign Laboratories Pty Ltd.) was applied daily at a topical dose of 62.5 mg for 7 consecutive days to establish an IMQ-induced psoriasis mouse model characterized by moderate-to-severe psoriasis-like skin lesions, including pronounced erythema, scale formation, epidermal thickening and inflammatory cell infiltration, corresponding to Psoriasis Area and Severity Index (PASI) scores of 2-3 for each parameter (20,21). MTX was used as a positive control because it is commonly used as a first-line treatment for moderate-to-severe plaque psoriasis (22). The concentration of 5.0 mg/kg LSG was selected based on previous studies (23,24). The mice were randomly divided into six groups (n=8/group) as follows: i) Normal control (NC), mice were intraperitoneally injected with 200 µl normal saline, followed by the topical application of petroleum cream (Chemipan Corporation Co., Ltd.); ii) methotrexate (MTX) control, mice were intraperitoneally injected with 200 µl 1.0 mg/kg MTX, followed by the topical application of petroleum cream; iii) LSG control, mice were intraperitoneally injected with 200 µl 5.0 mg/kg LSG, followed by the topical application of petroleum cream; iv) IMQ, mice were intraperitoneally injected with 200 µl normal saline, followed by the topical application of IMQ cream; v) MTX + IMQ, mice were intraperitoneally injected with 200 µl 1.0 mg/kg MTX, followed by the topical application of IMQ cream; and vi) LSG + IMQ, mice were intraperitoneally injected with 200 µl 5.0 mg/kg LSG, followed by the topical application of IMQ cream (Fig. S1). After 7 days of treatment, all mice were anesthetized with 5% isoflurane in oxygen and maintained at 1.5-3%, followed by cervical dislocation. The thoracic cavity was opened, and blood was collected via cardiac puncture. Skin lesions, lymph nodes, spleen, kidney and liver were harvested for further analysis.
The severity of skin lesions was assessed using a modified human scoring system based on PASI (21,25). Mice were evaluated daily for erythema, scaling and thickening. Each parameter was scored independently on a scale from 0 to 4 (0, none; 1, mild; 2, moderate; 3, marked; 4, obvious; Table I) (21). The cumulative score was used to evaluate the overall severity of skin inflammation.
Table IDescriptive Psoriasis Area and Severity Index scores in a mouse model following imiquimod induction. |
Body weight was recorded on days 1, 3, 5 and 7 of administration. On day 8, the mice were euthanized, and right axillary lymph nodes and spleens were removed, cleaned and weighed. The weights of the right axillary lymph nodes and spleens were normalized to body weight to calculate the organ index.
Skin lesions were collected, fixed in 4% paraformaldehyde for 48 h at 4˚C and embedded in paraffin. Paraffin blocks were sectioned at 4 µm thickness using a rotary microtome. Tissue sections were deparaffinized with xylene and rehydrated through a descending graded ethanol series. Sections were stained with Mayer's hematoxylin (cat. no. 05-06002/L) and Eosin Y alcoholic solution (cat. no. 05-11007/L; both Bio Optica Milano SpA). Sections were dehydrated, cleared, mounted and cover slipped for histopathological observation under a light microscope. Epidermal and dermal thickness were measured in four randomly selected areas of view/section at 10X magnification using ImageJ software version 1.32j (National Institutes of Health). To visualize blood vessels in the skin lesions, Periodic Acid-Schiff (PAS) staining was performed using a PAS staining kit (cat. no. 1.01646.0001, Sigma-Aldrich; Merck KGaA) according to the manufacturer's protocol (26). Additionally, Giemsa staining was performed using Giemsa solution (cat. no. RA-002-05, Biotechnical Co., Ltd.), following the method described by Pudgerd et al (27). Stained sections were observed and photographed under a light microscope. Mast cells were counted manually in four randomly selected fields of view/section at 10X magnification.
Skin lesions were fixed in 4% paraformaldehyde for 48 h at 4˚C and embedded in paraffin. Paraffin blocks were sectioned at 4 µm thickness. Tissue sections were deparaffinized with xylene and rehydrated through a descending graded ethanol series. Tissue sections were incubated in 3% H2O2 for 15 min to block endogenous peroxidase activity, followed by antigen retrieval using Tris-EDTA buffer (pH 9.0) at 100˚C for 20 min, and washed with 1X PBS-T (0.05% Tween 20). Tissue sections were incubated with protein blocking solution (0.5% BSA, 0.5% casein in PBS; cat. no. AB64226, Abcam) for 1 h at room temperature. Sections were incubated with primary antibodies against CD4+ (rabbit monoclonal anti-CD4+, cat. no. ab183685, Abcam; 1:50) and Ki67 (rabbit polyclonal anti-Ki67, cat. no. ab15580, Abcam; 1:100) for 3 h at room temperature. Following washing with 1X PBS-T, sections were incubated with peroxidase-conjugated secondary antibody (AffiniPure goat anti-rabbit IgG H&L, cat. no. 111-035-003, Jackson ImmunoResearch Laboratories, Inc.; 1:500) for 1 h at room temperature. Positive signals were developed using NOVA Red substrate (cat. no. SK-4800, Vector Laboratories, Inc.; Maravai LifeSciences) and counterstained with Mayer's hematoxylin for 20 sec at room temperature. For quantitative analysis, slides were imaged at 10X magnification under a light microscope. CD4+ and Ki67-positive cells were counted manually in four randomly selected areas of view/section.
Skin tissue and blood serum samples were collected to assess cytokine levels using ELISA kits. Dorsal skin was excised and any attached connective tissue was removed. A 0.1 cm2 section of tissue was homogenized in 1 ml normal saline using a tissue homogenizer. The homogenate was centrifuged at 2,500 x g for 15 min at 4˚C. The supernatant was collected and stored at -20˚C until analysis. For blood serum, samples were allowed to clot at room temperature for 30 min, followed by centrifugation at 2,500 x g for 15 min at 4˚C. The serum was separated and stored at -20˚C until use (27). Cytokine concentrations including IL-1β (ELISA MAX™ Deluxe Set Mouse IL-1β kit, cat. no. 432604), TNF-α (cat. no. 430904), IL-17A (cat. no. 432504), and IL-23 (cat. no. 433704), were measured using commercial ELISA kits, according to the manufacturer's protocols (all BioLegend, Inc.). Absorbance was measured at 450 nm using a VersaMax™ microplate reader and data were analyzed with SoftMax Pro software version 6 (Molecular Devices, LLC).
Total RNA was extracted from skin tissue using TRI Reagent® (Molecular Research Center, Inc., cat. no. TR118) according to the manufacturer's instructions. A total of 1 µg total RNA was reverse-transcribed into cDNA using the iScript™ Reverse Transcription Supermix for RT-qPCR (Bio-Rad Laboratories, Inc.; cat. no. 1708840). All RT-qPCR assays were conducted using the QIAquant Real-Time PCR Thermal Cycler (Qiagen, Inc.) with the SensiFAST™ SYBR® No-ROX Kit (Bioline, cat. no. BIO-98050), according to the manufacturer's protocols. Thermocycling conditions were as follows: Initial denaturation at 95˚C for 2 min, followed by 40 cycles of 95˚C for 5 sec, 60˚C for 10 sec of annealing, and 72˚C for 20 sec of extension. Relative gene expression of IL-1β, TNF-α, keratin 6, 16 and 17, involucrin, JAK1, 2 and 3, STAT1, 2 and 3, BCL2 and CCND1 was normalized to β-actin and calculated using the 2-ΔΔCq method (28). Primer sequences for all target genes are listed in Table II.
Total protein from skin tissue was extracted using lysis buffer containing 20 mM Tris-HCL, 100 mM NaCl, 5 mM phenylmethylsulfonyl fluoride and 100X protease inhibitor solution. Protein concentration was determined with a NanoDrop 2000 spectrophotometer (Thermo Fisher Scientific, Inc.). A total of 50 µg protein/lane was separated on a 12.5% SDS-polyacrylamide gel and transferred to a nitrocellulose membrane. The membrane was blocked with 2% BSA (cat. no. 112018, Merck KGaA) in TBS-T (100 mM Tris-base, 150 mM NaCl, 0.1% Tween 20) for 2 h at room temperature and incubated overnight at 4˚C with primary antibodies (1:1,000) against mouse anti-Ki67 (cat. no. 14569982, Invitrogen, Thermo Fisher Scientific, Inc.), rabbit anti-TNF-α (cat. no. 3707S), mouse anti-IL-6 (cat. no. 12019S), rabbit anti-IL-10 (cat. no. 12163S; all Cell Signaling Technology Inc.) and rabbit anti-β-actin (cat. no. 4970S, Cell Signaling Technology, Inc.). After three washes with TBS-T for 10 min each, the membranes were incubated with HRP-conjugated secondary antibody (1:2,000; cat. nos. 31460 and 626520; Invitrogen, Thermo Fisher Scientific, Inc.) for 1 h at room temperature. Membranes were washed with TBS-T and immunoreactivity was visualized using the Clarity™ Western ECL substrate kit (Bio-Rad Laboratories, Inc.). Protein bands were detected using the Amersham™ ImageQuant™ 800 biomolecular imager (Cytiva). Relative protein expression was quantified using ImageJ software version 1.32j (National Institutes of Health), with band intensities normalized to β-actin.
All data are presented as mean ± SD of triplicate experiments. Statistical analysis was performed using one-way ANOVA followed by Tukey's post hoc multiple comparison test. P<0.05 was considered to indicate a statistically significant difference. All statistical analyses were performed using GraphPad Prism software, version 10.3.1.509 (Dotmatics).
LSG had a molecular weight of 7.87 kDa and consisted of complex structures with alternating 3-linked β-D-galactopyranose and 4-linked 3,6-anhydro-α-L-galactopyranose or α-L-galactopyranose-6- sulfate units (Fig. S2) (18).
To investigate whether LSG ameliorated skin lesions in psoriasis, IMQ was applied to induce a psoriasis-like mouse model, with and without LSG treatment, and compared with MTX treatment. Mice treated with IMQ exhibited rapid proliferation of immune cells and keratinocytes, leading to increased cytokine production. This mimics the features of human plaque psoriasis, which is characterized by erythema, skin thickening, scaling, and epidermal changes associated with increased inflammation and vascular alterations (29). Symptoms of the psoriatic condition were observed over 7 days of continuous IMQ application in the IMQ group. However, LSG + IMQ group showed a decrease in IMQ-induced psoriatic traits, similar to MTX + IMQ group (Figs. 1A and S3).
Subcutaneous vasculature in the lesional area was visualized post-mortem to evaluate inflammatory angiogenesis. Mice in the IMQ group displayed dense and complex vascular networks, characterized by prominent branching and bifurcation, indicative of increased neovascularization and vascular remodeling commonly associated with psoriatic inflammation (8,29). By contrast, both the MTX + IMQ and LSG + IMQ groups exhibited decreased vascular complexity, suggesting that these treatments effectively suppressed IMQ-induced angiogenic responses. NC, MTX and LSG groups exhibited sparse and minimally branched vasculature, consistent with normal skin lacking inflammatory stimulation (Fig. 1B and C).
PASI scores were calculated to provide a qualitative assessment of disease progression and therapeutic efficacy (Figs. 1D and S4). Mice in the LSG + IMQ group showed significantly lower PASI scores in all parameters compared with the IMQ group, indicating a marked decrease in psoriatic severity. There were significant differences between the LSG + IMQ and MTX + IMQ groups. Collectively, the PASI data supported the protective effect of LSG against IMQ-induced psoriatic skin inflammation.
Histopathological analysis of dorsal skin was performed to evaluate the effect of LSG in IMQ-induced psoriasis. Mice in the IMQ group developed typical psoriasis-like changes, including hyperkeratosis, parakeratosis and thickening of the epidermal stratum spinosum, whereas the LSG + IMQ group showed decreased psoriasis-like alterations (Fig. 2A). Quantitative analysis confirmed that epidermal thickness was significantly decreased in the LSG + IMQ group compared with the IMQ group (Fig. 2B). No significant difference in epidermal thickness was observed between the LSG + IMQ and MTX + IMQ groups. Although dermal thickness did not differ significantly between the IMQ-treated groups, both the MTX + IMQ and LSG + IMQ groups showed decreased dermal thickness compared with the IMQ group (Fig. 2C).
Epidermal thickening in psoriasis results from increased keratinocyte proliferation, with Ki67 serving as a proliferation marker (30). Immunohistochemistry showed fewer Ki67-positive cells in the epidermis of the MTX + IMQ and LSG + IMQ groups compared with the IMQ group (Fig. 2A and D). Ki67 protein expression was significantly decreased in the LSG + IMQ compared with the IMQ group (Fig. 2E). To assess keratinocyte differentiation, the expression of keratin 6, 16 and 17 and involucrin (established biomarkers of keratinocyte differentiation) (7) was evaluated by RT-qPCR. Expression levels of keratin 6, 16 and 17 were significantly decreased in the LSG + IMQ compared with the IMQ group. No significant differences were observed between the LSG + IMQ and MTX + IMQ groups (Fig. 2F-H). Expression of involucrin, a structural protein involved in psoriatic plaque formation, was also decreased in the LSG + IMQ group compared with the IMQ group (Fig. 2I). Taken together, these findings indicated that LSG treatment reduces keratinocyte proliferation and differentiation.
Psoriasis is a T cell-mediated autoimmune skin disease characterized by overexpression of proinflammatory cytokines (31). TNF-α and IL-1β serve key roles in regulating inflammation and promoting Th17 cell development, which is central to the pathogenesis of psoriasis (32). In addition, IL-23 activates CD4+ T cells and promotes the release of IL-17A, which drives keratinocyte proliferation in psoriasis (4). ELISA and western blot analyses were performed to determine whether LSG modulates inflammatory cytokine expression during psoriasis development. ELISA showed that levels of TNF-α, IL-1β, IL-17A, and IL-23 were significantly decreased in the skin lesions of the LSG + IMQ group compared with the IMQ group. These effects were consistent with those observed in the MTX + IMQ group (Fig. 3A-H). Western blotting further demonstrated that protein expression of TNF-α and IL-6 proinflammatory cytokines was significantly decreased in the LSG + IMQ compared with the IMQ group. By contrast, IL-10, an anti-inflammatory cytokine, showed increased expression in the LSG + IMQ group (Fig. 3I-L). These findings highlight the efficacy of LSG in attenuating cutaneous inflammation in psoriatic mice.
By reducing levels of TNF-α, IL-1β, and IL-17A, LSG decreased the recruitment of inflammatory Th17 cells. To assess the effect of LSG on inflammatory cell infiltration in psoriatic skin lesions, immunohistochemical staining for CD4+ cells was performed. IMQ group demonstrated accumulation of CD4+ cells in psoriatic skin lesions. By contrast, CD4+ cell infiltration was significantly decreased in both the MTX + IMQ and LSG + IMQ groups compared with the IMQ group (Fig. 4A and B). Giemsa staining of skin sections revealed that mast cell infiltration was significantly lower in the MTX + IMQ and LSG + IMQ groups compared with the IMQ group (Fig. 5A and B). These findings suggested that LSG reduces inflammatory cell infiltration in psoriatic skin.
Proinflammatory cytokines are mediated by IL-17A and IL-23 via activation of the JAK/STAT signaling pathway (5). The decreased expression of IL-17A and IL-23 in the LSG + IMQ group may affect JAK/STAT pathway activation. To investigate this, RT-qPCR was used to analyze mRNA expression of JAK/STAT pathway components, demonstrating downregulation of JAK2, STAT2 and STAT3 (Fig. 6A-F). Consistent with these findings, expression of the JAK/STAT target genes BCL2 and CCND1 was also decreased in the LSG + IMQ group to similar levels observed in the MTX + IMQ group, compared with the IMQ group (Fig. 6G and H).
Psoriasis, as an inflammatory disease, leads to immune system activation and is associated with increased lymph node and spleen weight (33). To assess systemic inflammation, lymph nodes and spleens were collected and weighed. IMQ treatment resulted in increased size and weight of lymph nodes and spleens in the IMQ group compared with controls (NC, MTX, and LSG; Fig. 7). By contrast, the LSG + IMQ group showed a significant decrease in both lymph node (Fig. 7A and B) and spleen (Fig. 7C and D) size and weight compared with the IMQ group. No significant differences were observed between the MTX + IMQ and LSG + IMQ groups. These findings suggested LSG decreased systemic inflammation by suppressing IMQ-induced organomegaly.
To evaluate the potential toxicity of LSG on major internal organs, histopathological examination of the kidney and liver was performed using H&E staining (Fig. 8). Representative micrographs of renal (Fig. 8A) and hepatic tissue (Fig. 8B) showed preserved normal architecture without evidence of degeneration, necrosis, inflammatory infiltration or vascular congestion. Renal histology revealed intact glomeruli and renal tubules with no morphological abnormalities across all groups, including those receiving MTX or LSG with or without IMQ. Similarly, hepatic tissue exhibited normal lobular architecture with well-preserved hepatocyte morphology and no signs of hepatocellular damage. These findings suggested that LSG at the administered dose did not induce detectable hepatic or renal toxicity in IMQ-induced psoriatic mice, supporting its safety for systemic use.
There is increasing research on seaweed-derived SPs for the treatment of numerous ailments (11,12), particularly inflammatory conditions, due to their efficacy and minimal side effects. Studies have shown that oligosaccharides derived from G. fisheri modulate immune responses by inhibiting proinflammatory cytokines such as TNF-α and IL-1β, key mediators in psoriasis pathogenesis (13,34). Furthermore, modification of SG to obtain LSG from G. fisheri has been found to significantly enhance its biological activity (15,18). To the best of our knowledge, the present study is the first to provide in vivo evidence that intraperitoneal administration of LSG exerts potent anti-inflammatory and anti-psoriatic effects in an IMQ-induced psoriasis-like mouse model. Administration methods include oral, injectable and topical routes. Intraperitoneal administration allows rapid absorption of large volumes of substances and is the preferred injection route for non-irritant, isotonic solutions (35). Numerous studies have investigated the intraperitoneal injection of polysaccharides for their anti-inflammatory and anti-psoriatic effects (23,36,37). For example, psoriatic mice that received β-glucan intraperitoneally show decreased psoriatic arthritis-like symptoms (23). In another study, mice treated with intraperitoneal acitretin-dextran nanoparticles for six consecutive days exhibited amelioration of psoriasis-like skin disease (36). Additionally, the intraperitoneal injection of fucoidans at doses of 10, 50 and 250 mg/kg significantly reduced inflammation induced by sodium carboxymethyl cellulose in mice (37).
The IMQ-induced psoriasis animal model is used as it replicates key features of human psoriatic lesions, including epidermal thickening, erythema, scaling, vascular proliferation and infiltration of T and other immune cells (38). The PASI score is a standard tool for evaluating disease severity and treatment response, with a reduction >50% generally considered a significant improvement (39). Administration of LSG significantly decreased erythema, scaling and skin thickening, as reflected by decreased PASI scores throughout the treatment period, indicating its anti-psoriatic effect. An additional factor that may have influenced lesion severity was hair regrowth during IMQ application. New hair growth appeared earlier in certain treatment groups, particularly in IMQ group, which decreased the effective skin surface area directly exposed to IMQ. Nevertheless, the overall amount of IMQ applied and the treated area remained unchanged; some of the compound may have initially adhered to the hair shafts. However, as the hair was relatively short, the drug was still able to permeate the skin over time. This was supported by the observation that the mice exhibited itching and scratching in these regions, similar to the areas without hair. However, to maintain a fully exposed dorsal skin surface for photographic observation, mice in control groups were re-shaved mid-protocol, whereas IMQ-treated groups were not re-shaved to avoid additional irritation. Hair regrowth in treated groups may also indicate a biological effect of the intervention. IMQ is associated with the activation of Th17 cells, leading to skin inflammation (40), while at the same time Th17-mediated mechanisms have been implicated in alopecia areata (41). The earlier reappearance of hair in treatment groups compared with untreated controls suggests that the treatment may have alleviated local inflammation and restored a skin microenvironment permissive for hair growth. Although hair regrowth was not quantitatively assessed, this supports the hypothesis that the therapeutic regimens not only attenuate psoriasiform inflammation but may also promote recovery of skin homeostasis. Future studies incorporating both surface area control and systematic monitoring of hair regrowth may clarify the dual role of this phenomenon. Histopathological and immunohistochemical staining (Ki67 and CD4+) supported these findings, showing attenuated epidermal hyperplasia, normalized epidermal architecture and decreased vascular development and infiltration of inflammatory cells, including CD4+ T and mast cells in the dermis (27). In psoriatic lesions, CD4+ T cells are typically concentrated in the upper dermis (31). Mast cells also serve a key role in the development and progression of psoriasis, with increased infiltration contributing to heightened inflammation (42). The decrease in dermal CD4+ T and mast cells following LSG treatment suggested LSG may help inhibit disease progression.
Ki67 is a protein used as a marker of cell proliferation, particularly to assess keratinocyte activity in skin conditions (43). In healthy skin, Ki67 expression is typically low but increases in conditions involving active keratinocyte division, such as wound healing or inflammatory skin disorders such as psoriasis (44). Previous studies have shown that Ki67 upregulation in psoriatic epidermis is decreased following treatment with acitretin-conjugated dextran, MTX and oxymatrine (36,45). Consistent with these findings, the present results demonstrated that LSG significantly reduced both the number of Ki67-positive cells and overall Ki67 expression in IMQ-induced psoriatic mice. These effects were comparable with those observed with MTX, a standard anti-psoriatic agent. Additional biomarkers associated with keratinocyte proliferation and differentiation in psoriasis include keratin 6, 16 and 17 and involucrin (7). In the present study, IMQ-induced upregulation of keratin 6, 16 and 17 and involucrin was significantly decreased following LSG treatment. This aligns with previous work demonstrating that tryptophol-containing emulgel, a topical drug delivery system that combines the properties of an emulsion and a gel, ameliorates IMQ-induced psoriasis in mice by reducing the expression of these markers (27).
The pathogenesis and progression of psoriasis involve the activation of immune cells, including T and antigen-presenting cells. Among these, Th cells, particularly Th1 and Th17 subsets, serve a key role in driving the immunological alterations characteristic of the disease (46). Activation of Th1 and Th17 cells leads to the aberrant production of proinflammatory cytokines such as TNF-α, IL-1β, IL-17A, IL-23 and IL-6, all of which are key mediators in psoriasis (6). In IMQ-induced psoriatic mice, LSG exhibited immunomodulatory effects by decreasing both systemic and cutaneous levels of TNF-α, IL-1β, IL-17A, IL-23 and IL-6 (a key cytokine in psoriasis pathogenesis) (3,8). These findings suggest that LSG suppresses inflammatory cytokine expression under psoriatic conditions. Mechanistically, the present findings indicated that LSG administration interferes with key signaling pathways involved in psoriasis. RT-qPCR revealed that LSG significantly decreased mRNA expression of JAK2, STAT2 and STAT3, along with downregulation of downstream targets BCL2 and CCND1 in psoriatic skin. The decreased dermal expression of BCL2 suggests inhibition of the JAK2/STAT3 pathway (27). The downregulation of JAK2, STAT2 and STAT3 mRNA following LSG administration may be a consequence of upstream cytokine suppression, particularly IL-23 and IL-17A, which drive JAK/STAT activation. While direct inhibition of JAK/STAT signaling by LSG cannot be excluded, evidence suggests an indirect mechanism mediated by decreased proinflammatory cytokine levels (47). Future mechanistic studies, including kinase activity assays, are warranted to determine whether LSG directly interacts with JAK/STAT components. This pathway serves a critical role in IL-23-mediated STAT3 activation, which promotes Th17 cell differentiation and subsequent IL-17A production, contributing to psoriatic inflammation and keratinocyte dysfunction (4). Although NF-κB activity was not directly assessed in the present study, its role as a master regulator of psoriatic inflammation is well established (4,48). Notably, SPs from other marine sources inhibit NF-κB activation (49), suggesting that LSG may exert similar effects.
Psoriasis, as an inflammatory disease, leads to immune system activation and is commonly associated with organomegaly, manifested by increased lymph node and spleen weight, which serves as an indicator of disease severity (33). LSG in IMQ-induced psoriatic mice significantly decreased lymph node and spleen size and weight compared with untreated psoriatic mice, suggesting LSG alleviated disease progression driven by immune activation (50). Additionally, no signs of hepatic or renal toxicity were observed in LSG-treated mice during the 7-day treatment period, as assessed by histopathological analysis. Serum biochemical markers for renal and hepatic function (alanine aminotransferase, aspartate transaminase; AST, creatinine) were not measured due to limited serum availability, as the samples were primarily used for inflammatory cytokine analysis. Therefore, potential subclinical organ toxicity cannot be fully excluded, although histological analysis did not reveal any renal or hepatic damage. However, in a previous study, intraperitoneal administration of SP from Caulerpa cupressoides var. lycopodium at a concentration of 9 mg/kg for 14 days produced no signs of systemic toxicity in mice (51). Thus, short-term LSG is unlikely to induce systemic toxicity, but longer-term studies and comprehensive toxicological evaluations are required to confirm the absence of cumulative or delayed adverse effects. This contrasts with conventional treatments such as MTX and cyclosporine, which are associated with cumulative toxicity and systemic side effects (9).
LSG promotes wound healing by promoting fibroblast proliferation, collagen synthesis and re-epithelialization (52). These regenerative effects, combined with its anti-inflammatory properties, suggest LSG may be effective in managing inflammatory skin conditions such as psoriasis. Additionally, LSG derived from G. fisheri exhibits strong free radical scavenging activity and enhances antioxidant capacity via activation of the nuclear factor erythroid 2-related factor 2/antioxidant response element signaling pathway, indicating broader therapeutic potential beyond dermatological applications (15). Previous studies on G. fisheri derived SG have primarily focused on wound-healing models (18,52). An octanoyl-esterified SG accelerates wound closure in rats, improves collagen deposition, increases α-smooth muscle actin (SMA) and vimentin expression and decreases expression of TNF-α at the wound site. Likewise, our recent study reported that a SG derivative improves histopathology and modulates wound healing proteins such as Ki67, α-SMA, E-cadherin and vimentin in a rat excision wound model (52). These findings highlight the regenerative and anti-fibrotic potential of G. fisheri-derived polysaccharides in tissue repair. To the best of our knowledge, the present study is the first to demonstrate that LSG from G. fisheri attenuates IMQ-induced psoriasis, an immune-mediated dermatosis characterized by keratinocyte hyperproliferation and Th17-driven inflammation. This extends the therapeutic scope of G. fisheri derived SG beyond wound repair to the modulation of immune-driven skin inflammation. Bioactivity of Gracilaria-derived polysaccharides is influenced by molecular weight and degree of sulfation, with lower molecular weight fractions exhibiting anti-inflammatory effects (53); consistent with this, the LSG preparation used in the present study displayed potent immunomodulatory effects in vivo. In comparison with standard therapies such as cyclosporine and acitretin, MTX exerts anti-proliferative and immunosuppressive effects primarily via the inhibition of dihydrofolate reductase (54), whereas LSG acts via immunomodulation, downregulation of inflammatory cytokines and suppression of JAK/STAT signaling. Unlike MTX, which can cause cumulative hepatotoxicity and bone marrow suppression with long-term use, LSG demonstrated no detectable hepatic or renal toxicity over the 7-day treatment period. From a pharmacological perspective, LSG represents a class of marine-derived polysaccharides with potent bioactivity and a favorable safety profile, making it a promising candidate for further drug development (12). Collectively, these findings suggest that LSG from G. fisheri exerts strong anti-psoriatic effects by inhibiting key inflammatory pathways and decreasing keratinocyte hyperproliferation and abnormal differentiation. However, the optimal delivery route, long-term efficacy and large-scale production feasibility remain to be further investigated.
In conclusion, the present study demonstrated the efficacy of LSG derived from G. fisheri in alleviating IMQ-induced psoriasis by decreasing psoriatic skin lesions and attenuating inflammatory responses. These effects were evidenced by decreased production of proinflammatory cytokines and infiltration of inflammatory cells in both skin lesions and systemic tissue. In addition, LSG treatment downregulated mRNA expression of key components of the JAK/STAT signaling pathway. Although the precise molecular mechanisms underlying its therapeutic effects remain to be elucidated, the immunomodulatory and anti-inflammatory properties suggest that LSG has potential as a marine-derived therapeutic agent for psoriasis management. Further studies are warranted to clarify the specific signaling pathways involved and validate its clinical applicability in human models.
The authors would like to thank the Laboratory Animal Research Center, University of Phayao, for providing the animal facility, and the School of Medical Sciences, University of Phayao, Phayao, Thailand for access to the laboratory facility. The authors would also to thank Dr Dylan Southard for language editing of the manuscript via the Khon Kaen University Publication Clinic (Khon Kaen, Thailand).
Funding: The present study was supported by the University of Phayao and the Thailand Science Research and Innovation Fund (Fundamental Fund 2024; grant no. 256/2567) and the Faculty of Medicine, Khon Kaen University, Thailand (grant no. IN67066).
The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.
KJ, AP and TR conceived and designed the study. KJ, AP, LP, SJ, TC, PP and TR performed the experiments. KJ, AP, KW and TR analyzed data. KJ, AP and TR wrote the manuscript. KJ, AP, KW, and TR edited the manuscript. KW and TR supervised the study. KJ, AP and TR confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.
The present study was approved by the Ethical Committee of the University of Phayao, Phayao, Thailand in accordance with the Ethics of Animal Experimentation guidelines established by the National Research Council (approval no. 1-024-65).
Not applicable.
The authors declare that they have no competing interests.
|
Moon CI, Lee J, Baek YS and Lee O: Psoriasis severity classification based on adaptive multi-scale features for multi-severity disease. Sci Rep. 13(17331)2023.PubMed/NCBI View Article : Google Scholar | |
|
Rendon A and Schäkel K: Psoriasis pathogenesis and treatment. Int J Mol Sci. 20(1475)2019.PubMed/NCBI View Article : Google Scholar | |
|
Griffiths CEM, Armstrong AW, Gudjonsson JE and Barker JNWN: Psoriasis. Lancet. 397:1301–1315. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Bugaut H and Aractingi S: Major role of the IL17/23 axis in psoriasis supports the development of new targeted therapies. Front Immunol. 12(621956)2021.PubMed/NCBI View Article : Google Scholar | |
|
Xue Y, Zhang L, Chu L, Song Z, Zhang B, Su X, Liu W and Li X: JAK2/STAT3 pathway inhibition by AG490 ameliorates experimental autoimmune encephalomyelitis via regulation of Th17 cells and autophagy. Neuroscience. 552:65–75. 2024.PubMed/NCBI View Article : Google Scholar | |
|
Dutta S, Chawla S and Kumar S: Psoriasis: A review of existing therapies and recent advances in treatment. J Rational Pharmacother Res. 4:12–23. 2018. | |
|
Qiao P, Zhi D, Yu C, Zhang C, Wu K, Fang H, Shao S, Yin W, Dang E, Li K and Wang G: Activation of the C3a anaphylatoxin receptor inhibits keratinocyte proliferation by regulating keratin 6, keratin 16, and keratin 17 in psoriasis. FASEB J. 36(e22322)2022.PubMed/NCBI View Article : Google Scholar | |
|
Lowes MA, Bowcock AM and Krueger JG: Pathogenesis and therapy of psoriasis. Nature. 445:866–873. 2007.PubMed/NCBI View Article : Google Scholar | |
|
Balak DMW, Gerdes S, Parodi A and Salgado-Boquete L: Long-term safety of oral systemic therapies for psoriasis: A comprehensive review of the literature. Dermatol Ther (Heidelb). 10:589–613. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Woo YR, Cho DH and Park HJ: Molecular mechanisms and management of a cutaneous inflammatory disorder: Psoriasis. Int J Mol Sci. 18(2684)2017.PubMed/NCBI View Article : Google Scholar | |
|
Kumar A, Soratur A, Kumar S and Maran BAV: A review of marine algae as a sustainable source of antiviral and anticancer compounds. Macromol. 5(11)2025. | |
|
B J and R R: A critical review on pharmacological properties of sulfated polysaccharides from marine macroalgae. Carbohydr Polym. 344(122488)2024.PubMed/NCBI View Article : Google Scholar | |
|
Siringoringo B, Huipao N, Tipbunjong C, Nopparat J, Wichienchot S, Hutapea AM and Khuituan P: Gracilaria fisheri oligosaccharides ameliorate inflammation and colonic epithelial barrier dysfunction in mice with acetic acid-induced colitis. Asian Pac J Trop Biomed. 11:440–449. 2021. | |
|
Kang J, Jia X, Wang N, Xiao M, Song S, Wu S, Li Z, Wang S, Cui SW and Guo Q: Insights into the structure-bioactivity relationships of marine sulfated polysaccharides: A review. Food Hydrocoll. 123(107049)2021. | |
|
Rudtanatip T, Pariwatthanakun C, Somintara S, Sakaew W and Wongprasert K: Structural characterization, antioxidant activity, and protective effect against hydrogen peroxide-induced oxidative stress of chemically degraded Gracilaria fisheri sulfated galactans. Int J Biol Macromol. 206:51–63. 2022.PubMed/NCBI View Article : Google Scholar | |
|
Chen YF, Zheng JJ, Qu C, Xiao Y, Li FF, Jin QX, Li HH, Meng FP, Jin GH and Jin D: Inonotus obliquus polysaccharide ameliorates dextran sulphate sodium induced colitis involving modulation of Th1/Th2 and Th17/Treg balance. Artif Cells Nanomed Biotechnol. 47:757–766. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Pai SK, Chakraborty K, Pai AA and Dhara S: Therapeutic promise of a sulfated (1→4) galactan from edible sea grape Caulerpa racemosa: Modulation of cytokine expression in lipopolysaccharide-induced CALU-1 cells. Int J Biol Macromol. 313(144117)2025.PubMed/NCBI View Article : Google Scholar | |
|
Rudtanatip T, Somintara S, Sakaew W, El-Abid J, Cano ME, Jongsomchai K, Wongprasert K and Kovensky J: Sulfated galactans from Gracilaria fisheri with supplementation of octanoyl promote wound healing activity in vitro and in vivo. Macromol Biosci. 22(2200172)2022.PubMed/NCBI View Article : Google Scholar | |
|
Wongprasert K, Rudtanatip T and Praiboon J: Immunostimulatory activity of sulfated galactans isolated from the red seaweed Gracilaria fisheri and development of resistance against white spot syndrome virus (WSSV) in shrimp. Fish Shellfish Immunol. 36:52–60. 2014.PubMed/NCBI View Article : Google Scholar | |
|
Karamani C, Antoniadou IT, Dimou A, Andreou E, Kostakis G, Sideri A, Vitsos A, Gkavanozi A, Sfiniadakis I, Skaltsa H, et al: Optimization of psoriasis mouse models. J Pharmacol Toxicol Methods. 108(107054)2021.PubMed/NCBI View Article : Google Scholar | |
|
Neu SD, Strzepa A, Martin D, Sorci-Thomas MG, Pritchard KA Jr and Dittel BN: Myeloperoxidase inhibition ameliorates plaque psoriasis in mice. Antioxidants (Basel). 10(1338)2021.PubMed/NCBI View Article : Google Scholar | |
|
Wang A, Ma X, Wei F, Li Y, Liu Q and Zhang H: Evidence on the therapeutic role of thiolutin in imiquimod-induced psoriasis-like skin inflammation in mice. Immun Inflamm Dis. 11(e877)2023.PubMed/NCBI View Article : Google Scholar | |
|
Fahlquist-Hagert C, Sareila O, Rosendahl S and Holmdahl R: Variants of beta-glucan polysaccharides downregulate autoimmune inflammation. Commun Biol. 5(449)2022.PubMed/NCBI View Article : Google Scholar | |
|
dos Santos Ferreira J, da Silva Prudêncio R, de Sousa AK, Sousa SG, de Sousa de Lima FM, dos Santos Carvalho A, da Costa ACC, Silva DMM, da Graça Sales Furtado M, Rocha DML, et al: Sulfated polysaccharide extracted from the alga Gracilaria domingensis modified with propionic anhydride negatively modulates acute inflammation and experimental hypernociception. Bioact Carbohydr Diet Fibre. 32(100459)2024. | |
|
van der Fits L, Mourits S, Voerman JSA, Kant M, Boon L, Laman JD, Cornelissen F, Mus AM, Florencia E, Prens EP and Lubberts E: Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis. J Immunol. 182:5836–5845. 2009.PubMed/NCBI View Article : Google Scholar | |
|
Lindford A, Juteau S, Jaks V, Klaas M, Lagus H, Vuola J and Kankuri E: Case report: Unravelling the mysterious Lichtenberg figure skin response in a patient with a high-voltage electrical injury. Front Med (Lausanne). 8(663807)2021.PubMed/NCBI View Article : Google Scholar | |
|
Pudgerd A, Jongsomchai K, Muangkaew W, Phuapittayalert L, Jamsuwan S, Chanmanee T, Vanichviriyakit R and Sukphopetch P: A tryptophol-containing emulgel ameliorates imiquimod-induced mice psoriasis. Sci Rep. 15(19398)2025.PubMed/NCBI View Article : Google Scholar | |
|
Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar | |
|
Aratwar A, Maji I, Chilvery S, Mahajan S, Aalhate M, Gupta U, Godugu C and Singh PK: Contemplating novel W/O emulsion-based gel for anti-psoriatic activity of Tofacitinib in imiquimod-induced Balb/C mice model. AAPS PharmSciTech. 26(12)2024.PubMed/NCBI View Article : Google Scholar | |
|
Sezer E, Böer-Auer A, Cetin E, Tokat F, Durmaz E, Sahin S and Ince U: Diagnostic utility of Ki-67 and Cyclin D1 immunostaining in differentiation of psoriasis vs other psoriasiform dermatitis. Dermatol Pract Concept. 5:7–13. 2015.PubMed/NCBI View Article : Google Scholar | |
|
Zhang P, Su Y, Li S, Chen H, Wu R and Wu H: The roles of T cells in psoriasis. Front Immunol. 14(1081256)2023.PubMed/NCBI View Article : Google Scholar | |
|
Li L, Lu J, Liu J, Wu J, Zhang X, Meng Y, Wu X, Tai Z, Zhu Q and Chen Z: Immune cells in the epithelial immune microenvironment of psoriasis: Emerging therapeutic targets. Front Immunol. 14(1340677)2024.PubMed/NCBI View Article : Google Scholar | |
|
Hatem S and El-Kayal M: Novel anti-psoriatic nanostructured lipid carriers for the cutaneous delivery of luteolin: A comprehensive in-vitro and in-vivo evaluation. Eur J Pharm Sci. 191(106612)2023.PubMed/NCBI View Article : Google Scholar | |
|
Khongthong S, Theapparat Y, Roekngam N, Tantisuwanno C, Otto M and Piewngam P: Characterization and immunomodulatory activity of sulfated galactan from the red seaweed Gracilaria fisheri. Int J Biol Macromol. 189:705–714. 2021.PubMed/NCBI View Article : Google Scholar | |
|
Lambert LJ, Muzumdar MD, Rideout WM III and Jacks T: Chapter 15-Basic mouse methods for clinician researchers: Harnessing the mouse for biomedical research. In: Basic Science Methods for Clinical Researchers. Academic Press, pp291-312, 2017. | |
|
Lan J, Li Y, Wen J, Chen Y, Yang J, Zhao L, Xia Y, Du H, Tao J, Li Y and Zhu J: Acitretin-conjugated dextran nanoparticles ameliorate psoriasis-like skin disease at low dosages. Front Bioeng Biotechnol. 9(816757)2022.PubMed/NCBI View Article : Google Scholar | |
|
Manggau M, Kasim S, Fitri N, Aulia S, Agustiani AN, Raihan M and Nurdin WB: Antioxidant, anti-inflammatory and anticoagulant activities of sulfate polysaccharide isolate from brown alga Sargassum polycystum. IOP Conf Ser Earth Environ Sci. 967(012029)2022. | |
|
Tripathi D, Srivastava M, Rathour K, Rai AK, Wal P, Sahoo J, Tiwari KR and Pandey P: A promising approach of dermal targeting of antipsoriatic drugs via engineered nanocarriers drug delivery systems for tackling psoriasis. Drug Metab Bioanal Lett. 16:89–104. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Dawoud ADH and Abdalbagi M: Evaluation of anti-psoriatic activity of Aloe sinkatana extract on imiquimod induced psoriatic-like dermatitis in mice. Pharm Biomed Res. 11:41–48. 2025. | |
|
Cho Y, Kwon J and Kim TS: Imiquimod promotes Th1 and Th17 responses via NF-κB-driven IL-12 and IL-6 production in an in vitro co-culture model. Exp Ther Med. 30(175)2025.PubMed/NCBI View Article : Google Scholar | |
|
Minokawa Y, Sawada Y and Nakamura M: Lifestyle factors involved in the pathogenesis of alopecia areata. Int J Mol Sci. 23(1038)2022.PubMed/NCBI View Article : Google Scholar | |
|
Zhou XY, Chen K and Zhang JA: Mast cells as important regulators in the development of psoriasis. Front Immunol. 13(1022986)2022.PubMed/NCBI View Article : Google Scholar | |
|
Assawawongkasem N, Techangamsuwan S, Piyaviriyakul P, Puchadapirom P and Sailasuta A: Involucrin, cytokeratin 10 and Ki67 expression in a threedimensional cultured canine keratinocyte cell line in comparison to canine skin and cutaneous squamous cell carcinoma and a pilot study on custom-designed siRNA-INV transfection. Thai J Vet Med. 50:43–55. 2020. | |
|
Ramezani M, Shamshiri A, Zavattaro E, Khazaei S, Rezaei M, Mahmoodi R and Sadeghi M: Immunohistochemical expression of P53, Ki-67, and CD34 in psoriasis and psoriasiform dermatitis. Biomedicine (Taipei). 9(26)2019.PubMed/NCBI View Article : Google Scholar | |
|
Shi HJ, Zhou H, Ma AL, Wang L, Gao Q, Zhang N, Song HB, Bo KP and Ma W: Oxymatrine therapy inhibited epidermal cell proliferation and apoptosis in severe plaque psoriasis. Br J Dermatol. 181:1028–1037. 2019.PubMed/NCBI View Article : Google Scholar | |
|
Armstrong AW and Read C: Pathophysiology, clinical presentation, and treatment of psoriasis: A review. JAMA. 323:1945–1960. 2020.PubMed/NCBI View Article : Google Scholar | |
|
Fu S, Bao X, Mao Z, Lv Y, Zhu B, Chen Y, Zhou M, Tain S, Zhou F and Ding Z: Tetrastigma hemsleyanum polysaccharide ameliorates cytokine storm syndrome via the IFN-γ-JAK2/STAT pathway. Int J Biol Macromol. 275(133427)2024.PubMed/NCBI View Article : Google Scholar | |
|
Blauvelt A and Chiricozzi A: The immunologic role of IL-17 in psoriasis and psoriatic arthritis pathogenesis. Clin Rev Allergy Immunol. 55:379–390. 2018.PubMed/NCBI View Article : Google Scholar | |
|
Jayawardhana HHACK, Lee HG, Liyanage NM, Nagahawatta DP, Ryu B and Jeon YJ: Structural characterization and anti-inflammatory potential of sulfated polysaccharides from Scytosiphon lomentaria; attenuate inflammatory signaling pathways. J Funct Foods. 102(105446)2023. | |
|
Takuathung MN, Wongnoppavich A, Panthong A, Khonsung P, Chiranthanut N, Soonthornchareonnon N and Sireeratawong S: Antipsoriatic effects of Wannachawee recipe on imiquimod-induced psoriasis-like dermatitis in BALB/c mice. Evid Based Complement Alternat Med. 2018(7931031)2018.PubMed/NCBI View Article : Google Scholar | |
|
Rodrigues JAG, Vanderlei ESO, de Araújo IWF, Quinderé ALG, Coura CO and Benevides NMB: In vivo toxicological evaluation of crude sulfated polysaccharide from the green seaweed Caulerpa cupressoides var. lycopodium in Swiss mice. Acta Sci Technol. 35:603–610. 2013. | |
|
Jongsomchai K, Pudgerd A, Sakaew W, Wongprasert K, Kovensky J and Rudtanatip T: Sulfated galactan derivative from Gracilaria fisheri improves histopathology and alters wound healing-related proteins in the skin of excision rats. Front Biosci (Landmark Ed). 29(388)2024.PubMed/NCBI View Article : Google Scholar | |
|
Khandwal D, Patel S, Pandey AK and Mishra A: A comprehensive, analytical narrative review of polysaccharides from the red seaweed Gracilaria: Pharmaceutical applications and mechanistic insights for human health. Nutrients. 17(744)2025.PubMed/NCBI View Article : Google Scholar | |
|
Johnston A, Gudjonsson JE, Sigmundsdottir H, Ludviksson BR and Valdimarsson H: The anti-inflammatory action of methotrexate is not mediated by lymphocyte apoptosis, but by the suppression of activation and adhesion molecules. Clin Immunol. 114:154–163. 2005.PubMed/NCBI View Article : Google Scholar | |
|
Chen Y, Song S, Wang Y, Zhang X, Zhang J, Wu L, Wu J and Li X: Topical application of berberine ameliorates imiquimod-induced psoriasis-like dermatitis in BALB/c mice via suppressing JAK1/STAT1 signaling pathway. Arab J Chem. 17(105612)2024. | |
|
Maglakelidze N, Gao T, Feehan RP and Hobbs RP: AIRE deficiency leads to the development of alopecia areata-like lesions in mice. J Invest Dermatol. 143:578–587.e3. 2023.PubMed/NCBI View Article : Google Scholar | |
|
Plaza R, Vidal S, Rodriguez-Sanchez JL and Juarez C: Implication of STAT1 and STAT3 transcription factors in the response to superantigens. Cytokine. 25:1–10. 2004.PubMed/NCBI View Article : Google Scholar |