logo
ResearchBunny Logo
Celastrol suppresses neovascularization in rat aortic vascular endothelial cells stimulated by inflammatory tenocytes via modulating the NLRP3 pathway

Medicine and Health

Celastrol suppresses neovascularization in rat aortic vascular endothelial cells stimulated by inflammatory tenocytes via modulating the NLRP3 pathway

Y. Yang, H. Wang, et al.

Discover how inflamatory processes in tenocyte injury impact angiogenesis during tendon-bone healing. This research, conducted by Yong Yang and colleagues, reveals that Celastrol can suppress excessive angiogenesis, leading to improved tendon healing outcomes.

00:00
00:00
~3 min • Beginner • English
Introduction
Rotator cuff injury is prevalent and burdensome, with high re-tear rates after surgery. Tendon-bone healing involves stem cells, growth factors, immune-inflammatory responses, and vascular regeneration. Inflammation is closely linked to tendinopathy, with elevated mediators (e.g., PGE2, IL-1, IL-6) and activation of signaling pathways (IFN, NF-κB, STAT-6). Excessive angiogenesis during repair can exacerbate scarring and impair functional recovery. VEGFA is a central angiogenic factor and is upregulated by inflammatory cytokines (e.g., TNF-α, IL-6), linking inflammation to angiogenesis. Celastrol, a bioactive component from Tripterygium wilfordii, has immunosuppressive and anti-inflammatory properties across diseases, but its role in rotator cuff injury and inflammation-induced angiogenesis is unclear. This study investigates whether celastrol can suppress LPS-induced inflammatory activation in tenocytes, reduce pro-angiogenic signaling (VEGFA), inhibit endothelial tube formation, and improve tendon-to-bone healing in a rat rotator cuff tear (RCT) model, potentially via modulation of the NLRP3 pathway.
Literature Review
The paper contextualizes angiogenesis as a double-edged process in tendon healing: while necessary, excessive neovascularization worsens scarring and mechanical outcomes. VEGFA is highlighted as a key mediator, influenced by inflammatory cytokines and hypoxic factors (e.g., HIF-1α). Prior studies report dynamic VEGF expression during tendon healing, with early peaks after acute injury and context-dependent roles where limiting angiogenesis can improve later healing stages. The NLRP3 inflammasome is implicated in tendon pathology, fibrosis, and regulation of pro-inflammatory cytokines such as IL-1β. Celastrol has documented anti-inflammatory and disease-modifying effects across rheumatoid arthritis, osteoarthritis, autoimmune disease, and cancer, acting through multiple pathways (e.g., NF-κB, TLR2/NF-κB, inhibition of NLRP3 activation). These data underpin the hypothesis that targeting NLRP3-driven inflammation could modulate VEGFA-mediated angiogenesis in tendon healing, and that celastrol may serve as a therapeutic.
Methodology
In vitro: Primary rat tenocytes were isolated from 5–6-week-old female Sprague-Dawley rats (~100 g). Tendons were minced, explant-adhered, cultured in DMEM + 10% FBS + 1% antibiotic-antimycotic, then passaged 3–4 times. Tenocytes were seeded at 1×10^5/mL in six-well plates, cultured 1 day, and treated with LPS at 0.5 or 1 μg/mL for 6 or 12 h. Conditioned media were collected. RAOECs (rat aortic vascular endothelial cells; YS2272C) were used for tube formation assays on Matrigel-coated 96-well plates; 10,000 cells/well were incubated for 6 h in LPS-tenocyte conditioned media or control media; images were acquired (Zeiss Axio Observer) and quantified with AngioTool. VEGFA secretion in tenocyte supernatants was quantified by ELISA (Beyotime PV960). Celastrol was administered to LPS-treated tenocytes to assess effects on inflammation and VEGFA secretion. Molecular assays: RNA was extracted (TRIzol), cDNA synthesized (TransGen kits), and RT-qPCR performed (TransStart Top Green qPCR SuperMix) using specified primers (IL-1β, TNF-α, Nlrp3, Vegfa, Gapdh). Protein analysis involved RIPA lysis, SDS-PAGE, PVDF transfer, and immunoblotting for NLRP3 (Abcam ab263899), IL-1β (Abcam EPR21086), with GAPDH as loading control; detection by ECL. Immunocytochemistry: tenocytes on poly-L-lysine coverslips were fixed, stained for NLRP3 with AlexaFluor 488 secondary, counterstained with DAPI, and imaged by confocal microscopy (Zeiss LSM 880). In vivo: Rat RCT model: a 1.5 cm anterolateral shoulder incision exposed the supraspinatus; half the tendon was detached from the greater tuberosity and repaired with 4-0 sutures. Groups: Sham, RCT, and RCT + Celastrol (1 mg/kg intra-articular weekly). At 4 or 8 weeks, biomechanical testing (MTS 858) measured ultimate load to failure and stiffness (preload up to 5 N; extension rate 14 mm/s). Tendon tissues were analyzed by RT-qPCR for Vegfa and Nlrp3 mRNA. Statistics: Experiments repeated ≥3 times; comparisons by Student’s t-test (two groups) or one-way ANOVA (≥3 groups); significance at P<0.05. In vivo sample size: three rats per group at each time point. Ethical approval: First Affiliated Hospital, Jinan University (202108246-25).
Key Findings
- LPS induced inflammatory activation in tenocytes: time- and dose-dependent increases in mRNA of NLRP3, TNF-α, IL-1β, and VEGFA at 0.5–1 μg/mL for 6–12 h; NLRP3 and IL-1β proteins also upregulated, with strongest response at 0.5 μg/mL for 12 h (selected for subsequent experiments). - LPS elevated secreted VEGFA in tenocyte conditioned media (ELISA; significant increase vs control), and this conditioned media significantly enhanced RAOEC tube formation (increased total tube length) (P<0.0001 in representative analyses). - Celastrol suppressed LPS-induced inflammation in tenocytes: significant reductions in NLRP3 and IL-1β mRNA and protein (immunoblot), and decreased NLRP3 immunostaining intensity (immunocytochemistry) compared with LPS alone (P<0.05 to P<0.001 depending on marker). - Celastrol reduced VEGFA secretion from LPS-induced tenocytes (ELISA; P<0.001) and inhibited RAOEC angiogenesis driven by inflammatory tenocyte conditioned media (significant reduction in total tube length; P<0.0001). - In RCT rats, celastrol improved biomechanics at 4 and 8 weeks: ultimate load to failure and stiffness were significantly higher in RCT + celastrol vs RCT controls (P<0.01 to P<0.0001), reversing injury-induced impairments. - Tendon tissues from RCT rats showed increased Vegfa and Nlrp3 mRNA, both of which were significantly reduced by celastrol treatment at 4 and 8 weeks (P<0.05 to P<0.0001). Overall, celastrol mitigates inflammation and pro-angiogenic signaling in vitro and improves structural-functional healing in vivo, consistent with modulation of the NLRP3–VEGFA axis.
Discussion
The study demonstrates that inflammatory activation of tenocytes (via LPS) elevates NLRP3/IL-1β and VEGFA expression and secretion, producing conditioned media that drives endothelial tube formation, implicating tenocyte-derived factors in neovascularization during tendon repair. Celastrol counteracts these effects by downregulating NLRP3 and IL-1β, reducing VEGFA secretion, and consequently inhibiting RAOEC angiogenesis. In vivo, celastrol enhances tendon-to-bone healing after RCT, restoring biomechanical strength and stiffness while lowering Vegfa and Nlrp3 expression in tendon, supporting the mechanistic link between NLRP3-driven inflammation, aberrant angiogenesis, and compromised healing. The findings align with literature indicating that excessive angiogenesis can worsen scarring and mechanical outcomes, and that modulating angiogenic pressure improves longer-term repair. By targeting the NLRP3 pathway, celastrol provides a dual anti-inflammatory and anti-angiogenic approach that may optimize the vascular milieu for tendon-bone integration and functional recovery.
Conclusion
Celastrol suppresses inflammation-induced neovascularization by modulating the NLRP3 pathway in tenocytes, reducing VEGFA secretion and endothelial tube formation in vitro, and improves biomechanical healing in a rat RCT model while lowering Vegfa and Nlrp3 expression in vivo. These results identify celastrol as a potential anti-angiogenic adjunct to promote tendon-to-bone healing. Future work should elucidate broader signaling networks affected by celastrol (e.g., via transcriptomics/proteomics), profile a wider range of inflammatory mediators and secretome components, and develop delivery strategies to overcome celastrol’s solubility and bioavailability limitations while ensuring safety for longer-term use.
Limitations
- Mechanistic depth is limited: downstream pathways beyond NLRP3/IL-1β (e.g., collagen production, ECM organization, cytoskeletal dynamics) were not interrogated; omics approaches (RNA-seq/proteomics) are needed. - Inflammatory profiling was incomplete: not all pro- and anti-inflammatory mediators (e.g., IL-2, IL-6, IL-10) or secreted factors beyond VEGFA were assessed. - Pharmacological constraints of celastrol: poor water solubility, low bioavailability, narrow therapeutic window, potential adverse and immunosuppressive effects; long-term safety and optimized delivery require further evaluation. - Sample sizes were modest (n=3 per group per timepoint in vivo), which may limit generalizability and statistical power for some endpoints.
Listen, Learn & Level Up
Over 10,000 hours of research content in 25+ fields, available in 12+ languages.
No more digging through PDFs, just hit play and absorb the world's latest research in your language, on your time.
listen to research audio papers with researchbunny