Skip to main content

GENERAL COMMENTARY article

Front. Pharmacol., 24 July 2024
Sec. Experimental Pharmacology and Drug Discovery

Commentary: Effect of curcumin nanoparticles on proliferation and migration of mouse airway smooth muscle cells and airway inflammatory infiltration

Fabien Beaufils,,
Fabien Beaufils1,2,3*Patrick Berger,,Patrick Berger1,2,3
  • 1Centre de Recherche Cardio-thoracique de Bordeaux, INSERM U1045, Bordeaux Imaging Center, University Bordeaux, Pessac, France
  • 2CHU Bordeaux, Département de Pédiatrie, CIC-P 1401, Service d’Anatomopathologie, Service d’Exploration Fonctionnelle Respiratoire, Bordeaux, France
  • 3INSERM, Centre de Recherche Cardio-thoracique de Bordeaux, U1045, Centre d’Investigation Clinique (CIC-P 1401), Pessac, France

Introduction

We read with interest the recent publication by Ma et al., Effect of curcumin nanoparticles on proliferation and migration of mouse airway smooth muscle cells and airway inflammatory infiltration (Ma et al., 2024) highlighting curcumin nanoparticles (CUR-NPs) as a potential new treatment of bronchial remodeling in asthma. Bronchial remodeling is a hallmark of asthma associated with both worse clinical outcomes and airflow obstruction related to structural alterations and thickening of the bronchial wall (Varricchi et al., 2022). Among the structural alterations of the bronchial wall, the increase in airway smooth muscle (ASM) mass appears as a key player by contributing to irreversible airflow limitation, poor symptom control and lack of response to treatment (Girodet et al., 2016; Varricchi et al., 2022). In asthma, ASM remodeling has been related to an increased ASM cell (ASMC) proliferation and migration enhanced by several factors (e.g., transforming growth factor (TGF)-β1) (Bara et al., 2010). In addition, ASMCs can produce themselves TGF-β1, collagen and fibronectin, also involved in bronchial fibrosis (Bara et al., 2010; Camoretti-Mercado and Lockey, 2021; Varricchi et al., 2022). However, usual anti-asthmatic treatments (i.e., corticosteroids and β2-adrenergic receptor agonists) remain unable to decrease ASMC proliferation and migration in vitro (Roth et al., 2004; Trian et al., 2007; Bara et al., 2010) as well as ASM mass in vivo in asthma (Camoretti-Mercado and Lockey, 2021), reinforcing the need of studies such as that of Ma et al. on curcumin, to identify new treatment that specifically target the ASM remodeling.

A novel method of curcumin administration

Curcumin is a polyphenolic compound derived from the curcuma plant exhibits anti-inflammatory and antioxidant properties, whose use in various forms (e.g., curcumin-containing poly (lactic-co-glycolic acid)-based microscale discoidal polymeric particles, curcumin-loaded niosomes or liposomal curcumin) has demonstrated promising preclinical results in asthma (Lelli et al., 2017; Ng et al., 2018; Park et al., 2020; Wong et al., 2020; Panknin et al., 2023). However, the poor bioavailability of curcumin delivered by oral ingestion may have limited its efficacy to improve lung function or symptoms in randomized clinical trials (Kim et al., 2011; Lelli et al., 2017; Panknin et al., 2023). Thus, the used of a novel method of administration, curcumin-coupled nanoparticles (CUR-NPs), represented a major advance and the strength of the study of Ma et al. Indeed, the authors clearly demonstrated that coupling curcumin to nanoparticles improved both curcumin intracellular uptake and accumulation in vitro (Ma et al., 2024). What is missing however, is the comparison of curcumin bioavailability in the lungs of mice treated either by coupled or uncoupled curcumin to fully convinced that CUR-NPs could fix the poor bioavailability issues identified in clinical trials (Kim et al., 2011; Lelli et al., 2017; Panknin et al., 2023).

Effects of curcumin-coupled nanoparticles

The other strong value of the study of Ma et al., is the in vivo validation of in vitro results. Indeed they also demonstrated, in lungs tissue from sensitized mice in vivo, that CUR-NPs decreased the protein expression of TGF-β1, Signal transducer and activator of transcription 3 (STAT3) and Connective tissue growth factor (CTGF) (Ma et al., 2024), whose roles in airway remodeling have been demonstrated (Bara et al., 2010; Gao et al., 2014; Gavino et al., 2016). They also showed, in vitro, that CUR-NPs may have a potential effect on both proliferation and migration of mouse-isolated tracheal cells described as ASMC. These effects of CUR-NPs are particularly relevant since both ASMC proliferation and migration are key mechanisms involved in ASM remodeling in asthma (Bara et al., 2010; Camoretti-Mercado and Lockey, 2021). Indeed, ASMC from asthmatic patients proliferate faster than those from controls (Bara et al., 2010; Esteves et al., 2021). This increased proliferation is mitochondria-dependent (Bara et al., 2010; Esteves et al., 2021) and can be enhanced by several factors increased in asthma such as growth factors/cytokines (e.g., TGFβ-1), inflammatory mediators (e.g., cysteinyl leukotriene) or enzymes (e.g., tryptase) (Bara et al., 2010; Trian et al., 2015; Camoretti-Mercado and Lockey, 2021). In addition, most of these mediators (e.g., TGFβ-1) can enhance the migration of ASMCs increasing ASM remodeling (Bara et al., 2010) and promote others characteristics of bronchial remodeling such as bronchial fibrosis (Camoretti-Mercado and Lockey, 2021; Varricchi et al., 2022). However, potential limitations of the study need to be considered.

Characterization of cells

Firstly, the authors described in the methods section the extraction and culture of murine ASMCs from cervical trachea and lung tissues (Ma et al., 2024) but there is no description regarding cell phenotype assessment to confirm the smooth muscle phenotype, as usually performed (Trian et al., 2007; Beaufils et al., 2021; Celle et al., 2022). This lack of information raises the question of what cell type has been studied, and how the authors concluded that these were indeed smooth muscle cells. In our hand, culturing murine ASMCs is more difficult than human ASMCs, because murine fibroblasts are more prone to proliferate than ASMCs.

Effect of CUR-NPs on isolated cell proliferation and migration

Secondly, ASMC proliferation was assessed using the Cell Counting Kit (CCK)-8 to the number of living cells. In the present study, this methods demonstrated approximately a doubling of the number of living cells after 48 h in cells stimulated by TGFβ-1 for 48 h compared to unstimulated cells or stimulated cells treated with CUR-NPs at 40 or 50 µM (Ma et al., 2024). Thus, this method confirmed an increased cell proliferation induced by TGFβ-1. However, the difference between TGFβ-1/CUR-NPs-treated cells and TGFβ-1-treated cells can suggest either a decreased cell proliferation, as mentioned by the authors, or an increased cell death, hence a possible toxicity, induced by CUR-NPs. Without any data regarding the effect of CUR-NPs on unstimulated cells to assess their toxicity, and/or specific assessment of cell death, it is thus difficult to conclude that CUR-NPs decreased the TGFβ-1 induced ASMC proliferation. In addition, if ASMC proliferation was effectively reduced or cell death increased upon CUR-NPs (40 µM) after 48 h of culture, the increased recovery of the gap by unstimulated ASMCs during migration assays using wound healing and transwell assays may be difficult to interpret. Indeed, after 48 h, the increased recovery of the gap by TGFβ-1-stimulated cells may result from an increased number of cells due to an increased proliferation or a decreased cell death of these cells compared with CUR-NPs-treated ASMCs instead of a difference in cell migration. As consequence, an actual decrease in ASMC migration remains to be demonstrated by repeating the migration assays over a shorter time (e.g., 24 h), less than the doubling time of both CUR-NPs-treated and untreated cells, and after comparison of cell death between CUR-NPs-treated and untreated cells.

Effect of CUR-NPs on airway remodeling

Thirdly, in their conclusion, the authors stated that CUR-NPs can effectively reverse airway remodeling in asthma (Ma et al., 2024). However, in their study, Ma et al. (2024) did not quantify the ASM mass to compare its remodeling between controls and asthmatic mice or between untreated and CUR-NPs-treated asthmatic mice. They also did not quantify the airway fibrosis whereas they clearly demonstrated the impacted CUR-NPs on TGFβ-1, a major player in this key aspect of bronchial remodeling. This lack of data regarding the ASM mass and airway fibrosis in mice limits the validity of their conclusion that CUR-NPs to effectively target airway remodeling in vivo despite the others promising in vitro and in vivo results. Indeed, among the several pharmacological components able to impact ASMC proliferation, migration and/or cell death in vitro, only two (i.e., gallopamil and fevipiprant) appeared able to decrease ASM mass in vivo (Camoretti-Mercado, 2009; Bara et al., 2010; Girodet et al., 2015; Saunders et al., 2019). In addition, bronchial thermoplasty has also been shown to decrease ASM mass in vivo (Cox et al., 2007; Girodet et al., 2015; Saunders et al., 2019).

In conclusion, by potentially decreasing the proliferation and the migration of the cells isolated from cervical trachea and lung tissues (Ma et al., 2024), CUR-NPs could target two major pathophysiological mechanisms involved in ASM remodeling and could represent a new promising treatment in asthma. Further studies on this issue, including studies in chronic mouse model studies to allow reversal of established remodeling to be assessed, obviously remain necessary and that, published by Ma et al. (2024), is thus an important contribution to identify potential new treatment of airway remodeling.

Author contributions

FB: Writing–original draft, Writing–review and editing. PB: Writing–review and editing.

Funding

The author(s) declare that no financial support was received for the research, authorship, and/or publication of this article.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

Bara, I., Ozier, A., Tunon de Lara, J. M., Marthan, R., and Berger, P. (2010). Pathophysiology of bronchial smooth muscle remodelling in asthma. Eur. Respir. J. 36 (5), 1174–1184. doi:10.1183/09031936.00019810

PubMed Abstract | CrossRef Full Text | Google Scholar

Beaufils, F., Esteves, P., Enaud, R., Germande, O., Celle, A., Marthan, R., et al. (2021). Mitochondria are involved in bronchial smooth muscle remodeling in severe preschool wheezers. J. Allergy Clin. Immunol. 148 (2), 645–651.e11. doi:10.1016/j.jaci.2021.03.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Camoretti-Mercado, B. (2009). Targeting the airway smooth muscle for asthma treatment. Transl. Res. 154 (4), 165–174. doi:10.1016/j.trsl.2009.06.008

PubMed Abstract | CrossRef Full Text | Google Scholar

Camoretti-Mercado, B., and Lockey, R. F. (2021). Airway smooth muscle pathophysiology in asthma. J. Allergy Clin. Immunol. 147 (6), 1983–1995. doi:10.1016/j.jaci.2021.03.035

PubMed Abstract | CrossRef Full Text | Google Scholar

Celle, A., Esteves, P., Cardouat, G., Beaufils, F., Eyraud, E., Dupin, I., et al. (2022). Rhinovirus infection of bronchial epithelium induces specific bronchial smooth muscle cell migration of severe asthmatic patients. J. Allergy Clin. Immunol. 150 (1), 104–113. doi:10.1016/j.jaci.2022.01.022

PubMed Abstract | CrossRef Full Text | Google Scholar

Cox, G., Thomson, N. C., Rubin, A. S., Niven, R. M., Corris, P. A., Siersted, H. C., et al. (2007). Asthma control during the year after bronchial thermoplasty. N. Engl. J. Med. 356 (13), 1327–1337. doi:10.1056/NEJMoa064707

PubMed Abstract | CrossRef Full Text | Google Scholar

Esteves, P., Blanc, L., Celle, A., Dupin, I., Maurat, E., Amoedo, N., et al. (2021). Crucial role of fatty acid oxidation in asthmatic bronchial smooth muscle remodelling. Eur. Respir. J. 58, 2004252. doi:10.1183/13993003.04252-2020

PubMed Abstract | CrossRef Full Text | Google Scholar

Gao, W., Cai, L., Xu, X., Fan, J., Xue, X., Yan, X., et al. (2014). Anti-CTGF single-chain variable fragment dimers inhibit human airway smooth muscle (ASM) cell proliferation by down-regulating p-Akt and p-mTOR levels. PLoS ONE 9 (12), e113980. doi:10.1371/journal.pone.0113980

PubMed Abstract | CrossRef Full Text | Google Scholar

Gavino, A. C., Nahmod, K., Bharadwaj, U., Makedonas, G., and Tweardy, D. J. (2016). STAT3 inhibition prevents lung inflammation, remodeling, and accumulation of Th2 and Th17 cells in a murine asthma model. Allergy 71 (12), 1684–1692. doi:10.1111/all.12937

PubMed Abstract | CrossRef Full Text | Google Scholar

Girodet, P. O., Allard, B., Thumerel, M., Begueret, H., Dupin, I., Ousova, O., et al. (2016). Bronchial smooth muscle remodeling in nonsevere asthma. Am. J. Respir. Crit. Care Med. 193 (6), 627–633. doi:10.1164/rccm.201507-1404OC

PubMed Abstract | CrossRef Full Text | Google Scholar

Girodet, P. O., Dournes, G., Thumerel, M., Begueret, H., Santos, P. D., Ozier, A., et al. (2015). Calcium channel blocker reduces airway remodeling in severe asthma. A proof-of-concept study. Am. J. Respir. Crit. Care Med. 191 (8), 876–883. doi:10.1164/rccm.201410-1874OC

PubMed Abstract | CrossRef Full Text | Google Scholar

Kim, D. H., Phillips, J. F., and Lockey, R. F. (2011). Oral curcumin supplementation in patients with atopic asthma. Allergy Rhinol. 2 (2), e51–e53. doi:10.2500/ar.2011.2.0016

PubMed Abstract | CrossRef Full Text | Google Scholar

Lelli, D., Sahebkar, A., Johnston, T. P., and Pedone, C. (2017). Curcumin use in pulmonary diseases: state of the art and future perspectives. Pharmacol. Res. 115, 133–148. doi:10.1016/j.phrs.2016.11.017

PubMed Abstract | CrossRef Full Text | Google Scholar

Ma, Y., Ye, S., Sun, K., and Gu, Y. (2024). Effect of curcumin nanoparticles on proliferation and migration of mouse airway smooth muscle cells and airway inflammatory infiltration. Front. Pharmacol. 15, 1344333. doi:10.3389/fphar.2024.1344333

PubMed Abstract | CrossRef Full Text | Google Scholar

Ng, Z. Y., Wong, J. Y., Panneerselvam, J., Madheswaran, T., Kumar, P., Pillay, V., et al. (2018). Assessing the potential of liposomes loaded with curcumin as a therapeutic intervention in asthma. Colloids Surf. B Biointerfaces 172, 51–59. doi:10.1016/j.colsurfb.2018.08.027

PubMed Abstract | CrossRef Full Text | Google Scholar

Panknin, T. M., Howe, C. L., Hauer, M., Bucchireddigari, B., Rossi, A. M., and Funk, J. L. (2023). Curcumin supplementation and human disease: a scoping review of clinical trials. Int. J. Mol. Sci. 24 (5), 4476. doi:10.3390/ijms24054476

PubMed Abstract | CrossRef Full Text | Google Scholar

Park, J. Y., Chu, G. E., Park, S., Park, C., Aryal, S., Kang, W. J., et al. (2020). Therapeutic efficacy of curcumin enhanced by microscale discoidal polymeric particles in a murine asthma model. Pharmaceutics 12 (8), 739. doi:10.3390/pharmaceutics12080739

PubMed Abstract | CrossRef Full Text | Google Scholar

Roth, M., Johnson, P. R., Borger, P., Bihl, M. P., Rüdiger, J. J., King, G. G., et al. (2004). Dysfunctional interaction of C/EBPalpha and the glucocorticoid receptor in asthmatic bronchial smooth-muscle cells. New Englang J. Med. 351, 560–574. doi:10.1056/NEJMoa021660

PubMed Abstract | CrossRef Full Text | Google Scholar

Saunders, R., Kaul, H., Berair, R., Gonem, S., Singapuri, A., Sutcliffe, A. J., et al. (2019). DP2 antagonism reduces airway smooth muscle mass in asthma by decreasing eosinophilia and myofibroblast recruitment. Sci. Transl. Med. 11 (479), eaao6451. doi:10.1126/scitranslmed.aao6451

PubMed Abstract | CrossRef Full Text | Google Scholar

Trian, T., Allard, B., Dupin, I., Carvalho, G., Ousova, O., Maurat, E., et al. (2015). House dust mites induce proliferation of severe asthmatic smooth muscle cells via an epithelium-dependent pathway. Am. J. Respir. Crit. Care Med. 191 (5), 538–546. doi:10.1164/rccm.201409-1582OC

PubMed Abstract | CrossRef Full Text | Google Scholar

Trian, T., Benard, G., Begueret, H., Rossignol, R., Girodet, P. O., Ghosh, D., et al. (2007). Bronchial smooth muscle remodeling involves calcium-dependent enhanced mitochondrial biogenesis in asthma. J. Exp. Med. 204 (13), 3173–3181. doi:10.1084/jem.20070956

PubMed Abstract | CrossRef Full Text | Google Scholar

Varricchi, G., Ferri, S., Pepys, J., Poto, R., Spadaro, G., Nappi, E., et al. (2022). Biologics and airway remodeling in severe asthma. Allergy 77 (12), 3538–3552. doi:10.1111/all.15473

PubMed Abstract | CrossRef Full Text | Google Scholar

Wong, J. Y., Yin, Ng Z., Mehta, M., Shukla, S. D., Panneerselvam, J., Madheswaran, T., et al. (2020). Curcumin-loaded niosomes downregulate mRNA expression of pro-inflammatory markers involved in asthma: an in vitro study. Nanomed 15 (30), 2955–2970. doi:10.2217/nnm-2020-0260

CrossRef Full Text | Google Scholar

Keywords: asthma, bronchial remodeling, smooth muscle, proliferation, migration

Citation: Beaufils F and Berger P (2024) Commentary: Effect of curcumin nanoparticles on proliferation and migration of mouse airway smooth muscle cells and airway inflammatory infiltration. Front. Pharmacol. 15:1432397. doi: 10.3389/fphar.2024.1432397

Received: 14 May 2024; Accepted: 08 July 2024;
Published: 24 July 2024.

Edited by:

Onur Bender, Ankara University, Türkiye

Reviewed by:

Jane Elizabeth Bourke, Monash University, Australia
Reshed Abohalaka, University of Gothenburg, Sweden
Bianca Martins Estevão, Italian Institute of Technology (IIT), Italy

Copyright © 2024 Beaufils and Berger. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Fabien Beaufils, fabien.beaufils@u-bordeaux.fr

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.