1
|
Sánchez M, Mercader Ruiz J, Marijuán Pinel D, Sánchez P, Fiz N, Guadilla J, Azofra J, Beitia M, Delgado D. Increasing the concentration of plasma molecules improves the biological activity of platelet-rich plasma for tissue regeneration. Sci Rep 2025; 15:4523. [PMID: 39915642 PMCID: PMC11802898 DOI: 10.1038/s41598-025-88918-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Platelet-rich plasma (PRP) has emerged as a promising therapy in a variety of medical fields. However, it is crucial to go beyond simple platelet concentration and examine the complex molecular composition both inside and outside platelets. The present work studies the effectiveness of a novel type of PRP named 'balanced protein-concentrate plasma' (BPCP). Different growth factor (GF) levels were measured using Enzyme Linked Immunosorbent Assay (ELISA), and in addition to the increase in intra-platelet GFs found in standard PRP (sPRP), BPCP also showed a higher concentration of plasmatic protein. Furthermore, extracellular vesicle (EV) concentration was significantly higher in BPCP. Cell proliferation was higher in cells incubated with lysates derived from BPCP compared to those cultured with sPRP. Regarding cell migration capacity, it was found that the process is platelet-dependent. Finally, the anti-inflammatory effect of BPCP was evaluated by inducing an inflammatory environment in M1-type macrophages. Cytokine levels were measured by ELISA following BPCP administration, showing a significant decrease in pro-inflammatory IL-1β, IL-6 and TNF-α. In summary, although further preclinical and clinical studies are needed in order to determine the therapeutic potential of BPCP, this PRP with unique characteristics demonstrates encouraging in vitro results that could potentially enhance tissue regeneration capacity.
Collapse
Affiliation(s)
- Mikel Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain.
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain.
| | - Jon Mercader Ruiz
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Daniel Marijuán Pinel
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Pello Sánchez
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Nicolás Fiz
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Jorge Guadilla
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Juan Azofra
- Arthroscopic Surgery Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Maider Beitia
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| | - Diego Delgado
- Advanced Biological Therapy Unit, Hospital Vithas Vitoria, 01008, Vitoria-Gasteiz, Spain
| |
Collapse
|
2
|
Campo-Grande GC, D'Agostin JB, de Santana Filho AP, da Silva Mello G, Benedetti PR, Dos Santos WF, Simas FF, Cipriani TR. Structural characterization of an arabinogalactan rich fraction from Bauhinia forficata Link leaves and evaluation of its effect on THP-1 macrophages. Int J Biol Macromol 2024; 283:137731. [PMID: 39551310 DOI: 10.1016/j.ijbiomac.2024.137731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 10/22/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Bauhinia forficata is a medicinal plant known as cow's paw, used for many purposes. Although there are studies that aimed to elucidate compounds from the plant leaves, there is no information about its polysaccharides. This study intended to obtain a polysaccharide rich fraction from its leaves, structurally characterize the water-soluble polysaccharides, as well as evaluate their effect on THP-1 cells. From the aqueous extract, followed by purification processes, a polysaccharide fraction (TCA-S) was obtained, constituted mainly of arabinose and galactose. Bidimensional NMR (13C/1H, HSQC) and methylation analyses identified type I and type II arabinogalactans, arabinan and starch as the major polysaccharides of the fraction. TCA-S was then submitted to starch removal process and renamed as TCA-Sα. TCA-Sα (2 to 500 μg/mL) was not cytotoxic to THP-1-cells and exhibited an immunostimulatory effect by increasing the secretion of nitric oxide and both pro-inflammatory cytokine IL-1β and anti-inflammatory cytokine IL-10. Immunomodulatory effect on IL-6 secretion was observed when macrophages were treated with TCA-Sα at 500 μg/mL. Additionally, the ratio between the concentrations of pro and anti-inflammatory cytokines produced by LPS-treated cells was higher than that produced by LPS plus TCA-Sα treated ones, suggesting that the polysaccharide fraction could modulate the LPS inflammation effects.
Collapse
Affiliation(s)
| | - Jessica Boschini D'Agostin
- Laboratory of Inflammatory and Neoplasic Cells, Cell Biology Department, Federal University of Paraná, CEP 81531-980 Curitiba, PR, Brazil
| | | | - Genilza da Silva Mello
- Biochemistry and Molecular Biology Department, Federal University of Paraná, CEP 81531-980 Curitiba, PR, Brazil
| | | | | | - Fernanda Fogagnoli Simas
- Laboratory of Inflammatory and Neoplasic Cells, Cell Biology Department, Federal University of Paraná, CEP 81531-980 Curitiba, PR, Brazil
| | - Thales Ricardo Cipriani
- Biochemistry and Molecular Biology Department, Federal University of Paraná, CEP 81531-980 Curitiba, PR, Brazil.
| |
Collapse
|
3
|
Wang X, Lin R, Fu C, Yang C, Dong D, Wu X, Chen X, Wang L, Hou J. Echinococcus granulosus cyst fluid inhibits inflammatory responses through inducing histone demethylase KDM5B in macrophages. Parasit Vectors 2023; 16:321. [PMID: 37689671 PMCID: PMC10492338 DOI: 10.1186/s13071-023-05948-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 08/26/2023] [Indexed: 09/11/2023] Open
Abstract
BACKGROUND Echinococcus granulosus cyst fluid (EgCF) weakens macrophage inflammatory responses, thereby enabling the parasite to evade the immune system. However, the role of histone modification in this process remains to be explored. METHODS The levels of IL-6, TNF-α, IL-10, H3K4me3, and KDM5B were detected using quantitative real-time PCR, ELISA, and Western blotting. The enrichment of H3K4me3 and KDM5B at the promoter of inflammatory factors was detected by chromatin immunoprecipitation. RESULTS Based on EgCF-stimulated macrophage models, we found that EgCF significantly inhibited mRNA expression and protein secretion of IL-6 and TNF-α and upregulated mRNA expression of IL-10 under the influence of TLR4. EgCF lowered the level of H3K4me3 and promoted the transcription and protein stability of histone demethylase KDM5B. Chromatin immunoprecipitation analysis revealed that EgCF suppressed the enrichment of H3K4me3 modification at the promoters of TNF-α and IL-6 and downregulated their expression in macrophages. Additionally, the inhibition of KDM5B activity by CPI-455 weakened the anti-inflammatory effect of EgCF. CONCLUSIONS Our findings demonstrate a novel mechanism through which EgCF promotes KDM5B expression and inhibits the enrichment of H3K4me3 at the promoters of inflammatory cytokines to suppress the inflammatory response.
Collapse
Affiliation(s)
- Xiaopeng Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Ruolin Lin
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chunxue Fu
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Chun Yang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Dan Dong
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xiangwei Wu
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Xueling Chen
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China
| | - Lianghai Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| | - Jun Hou
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases, The First Affiliated Hospital, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
- Key Laboratory of Xinjiang Endemic and Ethnic Diseases, Shihezi University School of Medicine, Shihezi, Xinjiang, China.
| |
Collapse
|
4
|
Portilla Y, Mulens-Arias V, Daviu N, Paradela A, Pérez-Yagüe S, Barber DF. Interaction of Iron Oxide Nanoparticles with Macrophages Is Influenced Distinctly by "Self" and "Non-Self" Biological Identities. ACS APPLIED MATERIALS & INTERFACES 2023. [PMID: 37478159 PMCID: PMC10401511 DOI: 10.1021/acsami.3c05555] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/23/2023]
Abstract
Upon contact with biological fluids like serum, a protein corona (PC) complex forms on iron oxide nanoparticles (IONPs) in physiological environments and the proteins it contains influence how IONPs act in biological systems. Although the biological identity of PC-IONP complexes has often been studied in vitro and in vivo, there have been inconsistent results due to the differences in the animal of origin, the type of biological fluid, and the physicochemical properties of the IONPs. Here, we identified differences in the PC composition when it was derived from the sera of three species (bovine, murine, or human) and deposited on IONPs with similar core diameters but with different coatings [dimercaptosuccinic acid (DMSA), dextran (DEX), or 3-aminopropyl triethoxysilane (APS)], and we assessed how these differences influenced their effects on macrophages. We performed a comparative proteomic analysis to identify common proteins from the three sera that adsorb to each IONP coating and the 10 most strongly represented proteins in PCs. We demonstrated that the PC composition is dependent on the origin of the serum rather than the nature of the coating. The PC composition critically affects the interaction of IONPs with macrophages in self- or non-self identity models, influencing the activation and polarization of macrophages. However, such effects were more consistent for DMSA-IONPs. As such, a self biological identity of IONPs promotes the activation and M2 polarization of murine macrophages, while a non-self biological identity favors M1 polarization, producing larger quantities of ROS. In a human context, we observed the opposite effect, whereby a self biological identity of DMSA-IONPs promotes a mixed M1/M2 polarization with an increase in ROS production. Conversely, a non-self biological identity of IONPs provides nanoparticles with a stealthy character as no clear effects on human macrophages were evident. Thus, the biological identity of IONPs profoundly affects their interaction with macrophages, ultimately defining their biological impact on the immune system.
Collapse
Affiliation(s)
- Yadileiny Portilla
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Vladimir Mulens-Arias
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Neus Daviu
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Alberto Paradela
- Proteomics Facility, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Sonia Pérez-Yagüe
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| | - Domingo F Barber
- Department of Immunology and Oncology and Nanobiomedicine Initiative, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
5
|
Manoharan RR, Sedlářová M, Pospíšil P, Prasad A. Detection and characterization of free oxygen radicals induced protein adduct formation in differentiating macrophages. Biochim Biophys Acta Gen Subj 2023; 1867:130324. [PMID: 36775000 DOI: 10.1016/j.bbagen.2023.130324] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/24/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023]
Abstract
Reactive oxygen species play a key role in cellular homeostasis and redox signaling at physiological levels, where excessive production affects the function and integrity of macromolecules, specifically proteins. Therefore, it is important to define radical-mediated proteotoxic stress in macrophages and identify target protein to prevent tissue dysfunction. A well employed, THP-1 cell line was utilized as in vitro model to study immune response and herein we employ immuno-spin trapping technique to investigate radical-mediated protein oxidation in macrophages. Hydroxyl radical formation along macrophage differentiation was confirmed by electron paramagnetic resonance along with confocal laser scanning microscopy using hydroxyphenyl fluorescein. Lipid peroxidation product, malondialdehyde, generated under experimental conditions as detected using swallow-tailed perylene derivative fluorescence observed by confocal laser scanning microscopy and high-performance liquid chromatography, respectively. The results obtained from this study warrant further corroboration and study of specific proteins involved in the macrophage activation and their role in inflammations.
Collapse
Affiliation(s)
- Renuka Ramalingam Manoharan
- Department of Biophysics, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Michaela Sedlářová
- Department of Botany, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Pavel Pospíšil
- Department of Biophysics, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic
| | - Ankush Prasad
- Department of Biophysics, Faculty of Science, Palacký University, Šlechtitelů 27, 783 71 Olomouc, Czech Republic.
| |
Collapse
|
6
|
Zhang F, Ke C, Zhou Z, Xu K, Wang Y, Liu Y, Tu J. Scutellaria baicalensis Pith-decayed Root Inhibits Macrophage-related Inflammation Through the NF-κB/NLRP3 Pathway to Alleviate LPS-induced Acute Lung Injury. PLANTA MEDICA 2023; 89:493-507. [PMID: 35716667 DOI: 10.1055/a-1878-5704] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Acute lung injury (ALI) is one of the representative "lung heat syndromes" in traditional Chinese medicine (TCM). Scutellaria baicalensis is an herbal medicine used in TCM for treating lung diseases, due to its remarkable anti-inflammatory and antiviral effects. When used in TCM, S. baicalensis root is divided into two categories: S. baicalensis pith-not-decayed root (SN) and S. baicalensis pith-decayed root (SD). Compared to SN, SD has a better effect on lung diseases. We constructed a lipopolysaccharide (LPS)-induced acute lung injury (ALI) mouse model to study the pharmacodynamic mechanism of SD. The ethanolic extract of Scutellaria baicalensis pith-decayed root (EESD) significantly affected LPS-induced ALI by reducing alveolar interstitial thickening, pulmonary edema, and other pathological symptoms, decreasing the infiltration of inflammatory cells, especially macrophages, and inhibiting IL-1β, TNF-α, and IL-6 transcription and translation. Furthermore, in the THP-1 macrophage model induced by LPS, EESD inhibited the expression of phosphorylated nuclear factor inhibitory protein alpha (p-IκBα), phosphorylated nuclear factor-κB P65 (p-p65), cleaved-caspase-1, cleaved-IL-1β protein, and the release of inflammatory factors in the NF-κB/NLRP3 pathway, inhibiting macrophage function. In vivo experiments yielded similar results. Therefore, the present study clarified the potential of EESD in the treatment of ALI and revealed its potential pharmacodynamic mechanism by inhibiting the NF-κB/NLRP3 inflammasome pathway and suppressing the pro-inflammatory phenotype activation of lung tissue macrophages.
Collapse
Affiliation(s)
- Fanglei Zhang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, P. R. China
| | - Chang Ke
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, P. R. China
| | - Zhongshi Zhou
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, P. R. China
| | - Kang Xu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, P. R. China
- Hubei Research Center of Chinese Materia Medica Processing Engineering and Technology, Hubei University of Chinese Medicine, Wuhan, P. R. China
| | - Yan Wang
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, P. R. China
| | - Yanju Liu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, P. R. China
- Hubei Research Center of Chinese Materia Medica Processing Engineering and Technology, Hubei University of Chinese Medicine, Wuhan, P. R. China
| | - Jiyuan Tu
- School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, P. R. China
- Hubei Research Center of Chinese Materia Medica Processing Engineering and Technology, Hubei University of Chinese Medicine, Wuhan, P. R. China
| |
Collapse
|
7
|
Röhrl J, Piqué-Borràs MR, Jaklin M, Werner M, Werz O, Josef H, Hölz H, Ammendola A, Künstle G. Anti-Inflammatory Activities of Arnica montana Planta Tota versus Flower Extracts: Analytical, In Vitro and In Vivo Mouse Paw Oedema Model Studies. PLANTS (BASEL, SWITZERLAND) 2023; 12:1348. [PMID: 36987036 PMCID: PMC10053944 DOI: 10.3390/plants12061348] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 02/22/2023] [Accepted: 03/08/2023] [Indexed: 06/19/2023]
Abstract
Arnica montana is well known for its anti-inflammatory properties. While the anti-inflammatory activity of Arnica flowers (Arnicae flos) has been extensively studied, that of the whole plant (Arnicae planta tota) is less characterized. We compared the ability of Arnicae planta tota and Arnicae flos extracts to inhibit the pro-inflammatory NF-κB-eicosanoid pathway, using several in vitro and in vivo assays. We showed that Arnicae planta tota inhibited NF-κB reporter activation, with an IC50 of 15.4 μg/mL (vs. 52.5 μg/mL for Arnicae flos). Arnicae planta tota also inhibited LPS-induced expression of ALOX5 and PTGS2 genes in human differentiated macrophages. ALOX5 and PTGS2 encode the 5-lipoxygenase (5-LO) and cyclooxygenase-2 (COX-2) enzymes that initialize the conversion of arachidonic acid into leukotrienes and prostaglandins, respectively. Arnicae planta tota inhibited 5-LO and COX-2 enzymatic activity in vitro and in human primary peripheral blood cells, with lower IC50 compared to Arnicae flos. Finally, Arnicae planta tota applied topically reduced carrageenan-induced mouse paw oedema more efficiently than Arnicae flos. Altogether, Arnicae planta tota displayed a superior anti-inflammatory activity compared to Arnicae flos, suggesting that Arnicae-planta-tota-containing products might be more effective in alleviating the manifestations of acute inflammation than those based on Arnicae flos alone.
Collapse
Affiliation(s)
- Johann Röhrl
- Preclinical Development, Weleda AG, 4144 Arlesheim, Switzerland
| | | | - Manuela Jaklin
- Preclinical Development, Weleda AG, 4144 Arlesheim, Switzerland
| | - Markus Werner
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Heinke Josef
- Quality Control, Weleda AG, 73525 Schwäbisch Gmünd, Germany
| | - Hubert Hölz
- Quality Control, Weleda AG, 73525 Schwäbisch Gmünd, Germany
| | - Aldo Ammendola
- Research and Development, Weleda AG, 4144 Arlesheim, Switzerland
| | - Gerald Künstle
- Preclinical Development, Weleda AG, 4144 Arlesheim, Switzerland
| |
Collapse
|
8
|
Sousa A, Rufino AT, Fernandes R, Malheiro A, Carvalho F, Fernandes E, Freitas M. Silver nanoparticles exert toxic effects in human monocytes and macrophages associated with the disruption of Δψm and release of pro-inflammatory cytokines. Arch Toxicol 2023; 97:405-420. [PMID: 36424514 DOI: 10.1007/s00204-022-03415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 11/03/2022] [Indexed: 11/27/2022]
Abstract
Silver nanoparticles (AgNP) are the most widely produced type of nanoparticles due to their antimicrobial and preservative properties. However, their systemic bioavailability may be considered a potential hazard. When AgNP reach the bloodstream, they interact with the immune cells, contributing to the onset and development of an inflammatory response. Monocytes and macrophages play a pivotal role in our defense system, but the interaction of AgNP with these cells is still not clear. Therefore, the main objective of this work was to assess the cytotoxic and pro-inflammatory effects induced by 5, 10, and 50 nm AgNP coated with polyvinylpyrrolidone (PVP) and citrate, in concentrations that could be attained in vivo (0-25 μg/mL), in human monocytes isolated from human blood and human macrophages derived from a monocytic cell line (THP-1). The effects of PVP and citrate-coated AgNP on cell viability, mitochondrial membrane potential, and cytokines release were evaluated. The results evidenced that AgNP exert strong harmful effects in both monocytes and macrophages, through the establishment of a strong pro-inflammatory response that culminates in cell death. The observed effects were dependent on the AgNP concentration, size and coating, being observed more pronounced cytotoxic effects with smaller PVP coated AgNP. The results showed that human monocytes seem to be more sensitive to AgNP exposure than human macrophages. Considering the increased daily use of AgNP, it is imperative to further explore the adverse outcomes and mechanistic pathways leading to AgNP-induced pro-inflammatory effects to deep insight into the molecular mechanism involved in this effect.
Collapse
Affiliation(s)
- Adelaide Sousa
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira No. 228, 4050-313, Porto, Portugal
| | - Ana T Rufino
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira No. 228, 4050-313, Porto, Portugal
| | - Rui Fernandes
- Histology and Electron Microscopy (HEMS), Instituto de Investigação e Inovação em Saúde i3S, Instituto de Biologia Molecular e Celular BMC, Universidade Do Porto, Porto, Portugal
| | - Ana Malheiro
- Histology and Electron Microscopy (HEMS), Instituto de Investigação e Inovação em Saúde i3S, Instituto de Biologia Molecular e Celular BMC, Universidade Do Porto, Porto, Portugal
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, 4050‑313, Porto, Portugal
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Porto, Portugal
| | - Eduarda Fernandes
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira No. 228, 4050-313, Porto, Portugal.
| | - Marisa Freitas
- LAQV, REQUIMTE, Laboratory of Applied Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira No. 228, 4050-313, Porto, Portugal.
| |
Collapse
|
9
|
Oliveira MM, Bonturi CR, Salu BR, Oliva MLV, Mortara RA, Orikaza CM. Modulation of STAT-1, STAT-3, and STAT-6 activities in THP-1 derived macrophages infected with two Trypanosoma cruzi strains. Front Immunol 2022; 13:1038332. [PMID: 36389843 PMCID: PMC9643828 DOI: 10.3389/fimmu.2022.1038332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 10/04/2022] [Indexed: 12/03/2022] Open
Abstract
Trypanosoma cruzi is the causative protozoan of Chagas' Disease, a neglected tropical disease that affects 6-7 million people worldwide. Interaction of the parasite with the host immune system is a key factor in disease progression and chronic symptoms. Although the human immune system is capable of controlling the disease, the parasite has numerous evasion mechanisms that aim to maintain intracellular persistence and survival. Due to the pronounced genetic variability of T. cruzi, co-infections or mixed infections with more than one parasite strain have been reported in the literature. The intermodulation in such cases is unclear. This study aimed to evaluate the co-infection of T. cruzi strains G and CL compared to their individual infections in human macrophages derived from THP-1 cells activated by classical or alternative pathways. Flow cytometry analysis demonstrated that trypomastigotes were more infective than extracellular amastigotes (EAs) and that strain G could infect more macrophages than strain CL. Classically activated macrophages showed lower number of infected cells and IL-4-stimulated cells displayed increased CL-infected macrophages. However, co-infection was a rare event. CL EAs decreased the production of reactive oxygen species (ROS), whereas G trypomastigotes displayed increased ROS detection in classically activated cells. Co-infection did not affect ROS production. Monoinfection by strain G or CL mainly induced an anti-inflammatory cytokine profile by decreasing inflammatory cytokines (IFN-γ, TNF-α, IL-1β) and/or increasing IL-4, IL-10, and TGF-β. Co-infection led to a predominant inflammatory milieu, with reduced IL-10 and TGF-β, and/or promotion of IFN-γ and IL-1β release. Infection by strain G reduced activation of intracellular signal transducer and activator of transcription (STAT) factors. In EAs, monoinfections impaired STAT-1 activity and promoted phosphorylation of STAT-3, both changes may prolong cell survival. Coinfected macrophages displayed pronounced activation of all STATs examined. These activations likely promoted parasite persistence and survival of infected cells. The collective results demonstrate that although macrophages respond to both strains, T. cruzi can modulate the intracellular environment, inducing different responses depending on the strain, parasite infective form, and co-infection or monoinfection. The modulation influences parasite persistence and survival of infected cells.
Collapse
Affiliation(s)
- Melissa Martins Oliveira
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Camila Ramalho Bonturi
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Bruno Ramos Salu
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Maria Luiza Vilela Oliva
- ²Biochemistry Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Renato Arruda Mortara
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| | - Cristina Mary Orikaza
- ¹Microbiology, Immunology and Parasitology Department, Escola Paulista de Medicina, Federal University of São Paulo - UNIFESP, São Paulo, Brazil
| |
Collapse
|
10
|
Hao D, Lu L, Song H, Duan Y, Chen J, Carney R, Li JJ, Zhou P, Nolta J, Lam KS, Leach JK, Farmer DL, Panitch A, Wang A. Engineered extracellular vesicles with high collagen-binding affinity present superior in situ retention and therapeutic efficacy in tissue repair. Theranostics 2022; 12:6021-6037. [PMID: 35966577 PMCID: PMC9373818 DOI: 10.7150/thno.70448] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 07/24/2022] [Indexed: 01/26/2023] Open
Abstract
Although stem cell-derived extracellular vesicles (EVs) have remarkable therapeutic potential for various diseases, the therapeutic efficacy of EVs is limited due to their degradation and rapid diffusion after administration, hindering their translational applications. Here, we developed a new generation of collagen-binding EVs, by chemically conjugating a collagen-binding peptide SILY to EVs (SILY-EVs), which were designed to bind to collagen in the extracellular matrix (ECM) and form an EV-ECM complex to improve EVs' in situ retention and therapeutic efficacy after transplantation. Methods: SILY was conjugated to the surface of mesenchymal stem/stromal cell (MSC)-derived EVs by using click chemistry to construct SILY-EVs. Nanoparticle tracking analysis (NTA), ExoView analysis, cryogenic electron microscopy (cryo-EM) and western-blot analysis were used to characterize the SILY-EVs. Fluorescence imaging (FLI), MTS assay, ELISA and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to evaluate the collagen binding and biological functions of SILY-EVs in vitro. In a mouse hind limb ischemia model, the in vivo imaging system (IVIS), laser doppler perfusion imaging (LDPI), micro-CT, FLI and RT-qPCR were used to determine the SILY-EV retention, inflammatory response, blood perfusion, gene expression, and tissue regeneration. Results:In vitro, the SILY conjugation significantly enhanced EV adhesion to the collagen surface and did not alter the EVs' biological functions. In the mouse hind limb ischemia model, SILY-EVs presented longer in situ retention, suppressed inflammatory responses, and significantly augmented muscle regeneration and vascularization, compared to the unmodified EVs. Conclusion: With the broad distribution of collagen in various tissues and organs, SILY-EVs hold promise to improve the therapeutic efficacy of EV-mediated treatment in a wide range of diseases and disorders. Moreover, SILY-EVs possess the potential to functionalize collagen-based biomaterials and deliver therapeutic agents for regenerative medicine applications.
Collapse
Affiliation(s)
- Dake Hao
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Lu Lu
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Hengyue Song
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Yixin Duan
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Jianing Chen
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
| | - Randy Carney
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Jian Jian Li
- Department of Radiation Oncology, University of California Davis, Sacramento, CA 95817, USA
| | - Ping Zhou
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Jan Nolta
- Stem Cell Program, Department of Internal Medicine, University of California Davis Medical Center, Sacramento, CA 95817, USA
| | - Kit S. Lam
- Department of Biochemistry and Molecular Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - J. Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, University of California Davis, Sacramento, CA 95817, USA
| | - Diana L Farmer
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
| | - Alyssa Panitch
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| | - Aijun Wang
- Department of Surgery, University of California Davis, Sacramento, CA 95817, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children Northern California, Sacramento, CA 95817, USA
- Department of Biomedical Engineering, University of California Davis, Davis, CA 95616, USA
| |
Collapse
|
11
|
Chantarasakha K, Asawapanumas T, Suntivich R, Panya A, Phonsatta N, Thiennimitr P, Laoteng K, Tepaamorndech S. Hatakabb, a herbal extract, contains pyrogallol as the novel mediator inhibiting LPS-induced TNF-α production by NF-κB inactivation and HMOX-1 upregulation. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.104992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
12
|
Immunomodulatory and Antioxidant Potential of Biogenic Functionalized Polymeric Nutmeg Oil/Polyurethane/ZnO Bionanocomposite. Pharmaceutics 2021; 13:pharmaceutics13122197. [PMID: 34959478 PMCID: PMC8703756 DOI: 10.3390/pharmaceutics13122197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 12/04/2022] Open
Abstract
The current study is focused on the biosynthesis of nutmeg oil/ polyurethane/ZnONPs bionanocomposite film for immunomodulatory and antioxidant activities. The fabricated film was prepared by using naturally extracted nutmeg oil functionalized with ZnONPs in the presence of polyutherane (PU) medium. The bionanocomposite film was obtained by incorporating dropwise 10 % (w/v) of nutmeg oil to the PU solution/ZnONPs blend. The active constituents of nutmeg oil were determined by gas chromatography coupled with mass spectrometry (GC-MS). The morphological characteristics of the resulting bionanocomposite film were confirmed using various microscopic and spectroscopic methods. Immunomodulatory potential of bionanocomposite was evaluated for RAW 264.7 macrophages. The results exhibited an excellent reduction in inflammatory cytokines (IL-6, IL-10, and TNFα) secretions after the treatment with bionanocomposite. The bionanocomposite exerted the highest inhibitory effects on certain cell signaling constituents that influence the initiation of expression of proinflammatory cytokines. The bionanocomposite was also tested for DPPH and ABTS free radicals scavenging assays and showed excellent antioxidant potential with IC50 values (0.28 ± 0.22 and 0.49 ± 0.36), respectively. The outcomes suggested promising immunomodulatory and antioxidant potentials for the biogenic synthesized nutmeg oil/PU/ZnONPs polymeric bionanocomposite.
Collapse
|
13
|
McCarrick S, Midander K, Krausová M, Carlander U, Karlsson HL. Gold Nanoparticles Dissolve Extracellularly in the Presence of Human Macrophages. Int J Nanomedicine 2021; 16:5895-5908. [PMID: 34475755 PMCID: PMC8405836 DOI: 10.2147/ijn.s314643] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 06/19/2021] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION Gold nanoparticles (AuNPs) have the potential to be used in various biomedical applications, partly due to the inertness and stability of gold. Upon intravenous injection, the NPs interact with the mononuclear phagocyte system, first with monocytes in the blood and then with macrophages in tissue. The NP-macrophage interaction will likely affect the stability of the AuNPs, but this is seldom analyzed. This study aimed to elucidate the role of macrophages in the biodissolution of AuNPs and underlying mechanisms. METHODS With an in vitro dissolution assay, we used inductively coupled plasma mass spectrometry to quantitatively compare the dissolution of 5 and 20 nm AuNPs coated with citrate or PEG in cell medium alone or in the presence of THP1-derived macrophages at 24 hours. In addition, we analyzed the cell dose, compared extra- and intracellular dissolution, and explored the possible role of reactive nitrogen species. RESULTS The results showed a higher cellular dose of the citrate-coated AuNPs, but dissolution was mainly evident for those sized 5 nm, irrespective of coating. The macrophages clearly assisted the dissolution, which was approximately fivefold higher in the presence of macrophages. The dissolution, however, appeared to take place mainly extracellularly. Acellular experiments demonstrated that peroxynitrite can initiate oxidation of gold, but a ligand is required to keep the gold ions in solution. CONCLUSION This study suggests extracellular dissolution of AuNPs in the presence of macrophages, likely with the contribution of the release of reactive nitrogen species, and provides new insight into the fate of AuNPs in the body.
Collapse
Affiliation(s)
- Sarah McCarrick
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, SE-171 77, Sweden
| | - Klara Midander
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, SE-171 77, Sweden
| | - Magdaléna Krausová
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, SE-171 77, Sweden
| | - Ulrika Carlander
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, SE-171 77, Sweden
| | - Hanna L Karlsson
- Institute of Environmental Medicine, Karolinska Institute, Stockholm, SE-171 77, Sweden
| |
Collapse
|
14
|
Güngör T, Ozleyen A, Yılmaz YB, Siyah P, Ay M, Durdağı S, Tumer TB. New nimesulide derivatives with amide/sulfonamide moieties: Selective COX-2 inhibition and antitumor effects. Eur J Med Chem 2021; 221:113566. [PMID: 34077833 DOI: 10.1016/j.ejmech.2021.113566] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Revised: 05/15/2021] [Accepted: 05/18/2021] [Indexed: 12/16/2022]
Abstract
Seventeen new amide/sulfonamide containing nimesulide derivatives were synthesized and characterized by several spectroscopic techniques and primarily investigated for their inhibitory potential on COX enzymes and other pro-inflammatory factors. Experimental analyses showed that among seventeen compounds, N8 and N10 have remarkable potency and selectivity for the COX-2 enzyme over COX-1 at very low doses as compared to nimesulide. Moreover, both N8 and N10 selectively reduced the Lipopolysaccharide (LPS)-stimulated COX-2 mRNA expression level while the COX-1 level remained stable. Both PGE2 release and nitric oxide production in macrophage cells were significantly suppressed by the N8 and N10 treatment groups. In silico ADME/Tox, molecular docking and molecular dynamics (MD) simulations were also conducted. Additionally, all compounds were also screened in a panel of cancer cell lines for their antiproliferative properties by MTT and SRB assays. Compound N17 exhibited a considerable antiproliferative effect on the colon (IC50: 9.24 μM) and breast (IC50: 11.35 μM) cancer cell lines. N17 exposure for 48 h decreased expression of anti-apoptotic protein BCL-2 and increased the expression of apoptogenic BAX. Besides, the BAX/BCL-2 ratio was increased with visible ultrastructural changes and apoptotic bodies under scanning electron microscopy. In order to investigate the structural and dynamical properties of selected hits on the target structures, multiscale molecular modeling studies are also conducted. Our combined in silico and in vitro results suggest that N8 and N10 could be further developed as potential nonsteroidal anti-inflammatory drugs (NSAIDs), while cytotoxic N17 might be studied as a potential lead compound that could be developed as an anticancer agent.
Collapse
Affiliation(s)
- Tuğba Güngör
- Department of Chemistry, Faculty of Sciences and Arts, Natural Products and Drug Research Laboratory, Çanakkale Onsekiz Mart University, Çanakkale, 17020, Turkey.
| | - Adem Ozleyen
- Graduate Program of Biomolecular Sciences, School of Graduate Studies, Canakkale Onsekiz Mart University, 17020, Çanakkale, Turkey; School of Chemistry, University of Leicester, LE1 7RH, Leicester, United Kingdom
| | - Yakup Berkay Yılmaz
- Graduate Program of Biomolecular Sciences, School of Graduate Studies, Canakkale Onsekiz Mart University, 17020, Çanakkale, Turkey; Department of Molecular Biology and Genetics, Faculty of Arts and Science, Çanakkale Onsekiz Mart University, 17020, Çanakkale, Turkey
| | - Pinar Siyah
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University, 34353, Istanbul, Turkey
| | - Mehmet Ay
- Department of Chemistry, Faculty of Sciences and Arts, Natural Products and Drug Research Laboratory, Çanakkale Onsekiz Mart University, Çanakkale, 17020, Turkey
| | - Serdar Durdağı
- Computational Biology and Molecular Simulations Laboratory, Department of Biophysics, School of Medicine, Bahçeşehir University, 34353, Istanbul, Turkey
| | - Tugba Boyunegmez Tumer
- Department of Molecular Biology and Genetics, Faculty of Arts and Science, Çanakkale Onsekiz Mart University, 17020, Çanakkale, Turkey.
| |
Collapse
|