1
|
Leifer VP, Fang F, Song L, Kim J, Papanikolaou JF, Smeeton J, Thomopoulos S. Single-cell RNA-sequencing analysis of immune and mesenchymal cell crosstalk in the developing enthesis. Sci Rep 2024; 14:26839. [PMID: 39500962 PMCID: PMC11538517 DOI: 10.1038/s41598-024-77958-7] [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: 06/29/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Autoimmunity underlies many painful disorders, such as enthesopathies, which localize to the enthesis. From infiltration of the synovium and axial skeleton by B cells, to disturbances in the ratio of M1/M2 enthesis macrophages, to CD8 + T cell mediated inflammation, autoimmune dysregulation is becoming increasingly well characterized in enthesopathies. Tissue resident B cells, macrophages, neutrophils, and T cells have also been localized in healthy human entheses. However, the potential developmental origins, presence, and role of immune cells (ICs) in enthesis development is not known. Here, we use single-cell RNA-sequencing analysis to describe IC subtypes present in the enthesis before, during, and after mineralization, and to infer regulatory interactions between ICs and mesenchymal cells (MCs). We report the presence of nine phenotypically distinct IC subtypes, including B cells, macrophages, neutrophils, and T cells. We find that specific IC subtypes may promote MC-proliferation and differentiation, and that MCs may regulate IC phenotype and autoimmunity. Our findings suggest that bidirectional regulatory interactions between ICs and MCs may be important to enthesis mineralization, and suggest that progenitor MCs have a unique ability to limit autoimmunity during development.
Collapse
Affiliation(s)
- Valia P Leifer
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Fei Fang
- Department Orthopedics, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Lee Song
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Jieon Kim
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - John F Papanikolaou
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA
| | - Joanna Smeeton
- Department of Rehabilitation and Regenerative Medicine, Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
- Department of Genetics and Development, Columbia Stem Cell Initiative, Columbia University, New York, NY, 10032, USA
| | - Stavros Thomopoulos
- Department of Orthopedic Surgery, Columbia University, New York, NY, 10032, USA.
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA.
| |
Collapse
|
2
|
Fernández OL, Belew AT, Rosales-Chilama M, Sánchez-Hidalgo A, Colmenares M, Saravia NG, El-Sayed NM. Interplay of human macrophage response and natural resistance of L. ( V .) panamensis to pentavalent antimony. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.29.620724. [PMID: 39553936 PMCID: PMC11566005 DOI: 10.1101/2024.10.29.620724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Macrophages are the principal host cells of Leishmania spp . in human infection and play a critical role in controlling infection and enabling parasite survival and persistence. Nevertheless, understanding of drug resistance in leishmaniasis has primarily focused on the parasite. This investigation provides evidence of the significant differential macrophage response to ex vivo infection with clinical strains of L. (V . ) panamensis naturally resistant (zymodeme 2.3/zym 2.3) or sensitive (zymodeme 2.2/zym 2.2) to antimonial drug, and the distinct effect of this drug on the activation of macrophages. Transcriptome analysis of infected monocyte-derived macrophages from healthy donors revealed significant interferon responses and cytokine signaling by zym 2.3 strains compared to zym 2.2 strains. Furthermore, in the presence of antimony, macrophages infected with zym 2.3 strains, but not with zym 2.2 strains, significantly increased the expression of genes associated with M-CSF-generated macrophages (M-MØ, anti-inflammatory). Notably, macrophages infected with zym 2.3 strains exhibited elevated expression of genes associated with homeostasis and microbicidal regulation, such as the IDO1/IL4I1-Kyn-AHR pathways and superoxide dismutase, and downregulation of transporters like ABC and AQP , compared to macrophages infected with zym 2.2 strains. Remarkably, the majority of these pathways remained upregulated even in the presence of the strong modulatory effect of antimonial drug. Together, these findings demonstrate that the initial and specific parasite-host interaction influences the ex vivo macrophage response to antimony. Identification of key pathways in macrophage responses associated with natural resistance to this antileishmanial, enhances understanding of host-response mechanisms in the outcome of Leishmania infection and response to treatment. IMPORTANCE Drug resistance and treatment failure are increasingly recognized in human leishmaniasis. Investigation of resistance has predominantly focused on parasite-mediated mechanisms. This study examines the role of host macrophages in natural resistance to antimonial drug. Our findings reveal distinct responses by macrophages infected with Leishmania (Viannia) panamensis strains that are naturally resistant to antimonial drug versus sensitive strains, both in the presence and absence of the drug. Distinctively, resistant parasites induced regulatory pathways that modulate inflammatory responses and alter host cell transporter expression, potentially contributing to parasite survival under antimony exposure. The host cell-parasite interaction in the context of drug resistant intracellular infections presents opportunities for innovative therapeutic strategies targeting host cell responses. Graphical abstract Clinical strains of L. (V . ) panamensis naturally resistant or sensitive to antimonial drug (SbV) induce different profiles of human macrophage activation. L. (V . ) panamensis strains with natural SbV resistance induce a significant interferon response in macrophages, accompanied by overexpression of the IDO1 / IL4I1 - Kyn - AHR pathways and SOD2 , associated with immune homeostasis and regulation of microbicidal activity. Homeostatic regulation by the IDO1 / IL4I1 - Kyn - AHR pathways, induced in macrophages by infection with naturally SbV-resistant strains of L. (V . ) panamensis , prevails despite significant modulation of macrophage activation by antimony exposure. Antimony treatment promotes a more anti-inflammatory (M-MØ) profile in macrophages infected with naturally resistant L. (V . ) panamensis strains, while macrophages infected with sensitive strains maintain a more proinflammatory profile (GM-MØ). Infection with the zym 2.3 strains in the presence of SbV leads to downregulation of specific macrophage transporter genes, supporting the capacity of these naturally SbV-resistant parasites to elicit macrophage responses that enable antimony resistance.
Collapse
|
3
|
Sánchez-García S, Castrillo A, Boscá L, Prieto P. Potential Beneficial Role of Nitric Oxide in SARS-CoV-2 Infection: Beyond Spike-Binding Inhibition. Antioxidants (Basel) 2024; 13:1301. [PMID: 39594443 PMCID: PMC11591382 DOI: 10.3390/antiox13111301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/28/2024] Open
Abstract
SARS-CoV-2, the causative virus for the COVID-19 disease, uses its spike glycoprotein to bind to human ACE2 as a first step for viral entry into the cell. For this reason, great efforts have been made to find mechanisms that disrupt this interaction, avoiding the infection. Nitric oxide (NO) is a soluble endogenous gas with known antiviral and immunomodulatory properties. In this study, we aimed to test whether NO could inhibit the binding of the viral spike to ACE2 in human cells and its effects on ACE2 enzymatic activity. Our results show that ACE2 activity was decreased by the NO donors DETA-NONOate and GSNO and by the NO byproduct peroxynitrite. Furthermore, we found that DETA-NONOate could break the spike-ACE2 interaction using the spike from two different variants (Alpha and Gamma) and in two different human cell types. Moreover, the same result was obtained when using NO-producing murine macrophages, while no significant changes were observed in ACE2 expression or distribution within the cell. These results support that it is worth considering NO as a therapeutic agent for COVID-19, as previous reports have suggested.
Collapse
Affiliation(s)
- Sergio Sánchez-García
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
| | - Antonio Castrillo
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
- Unidad de Biomedicina (Unidad Asociada al CSIC), Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM, Arturo Duperier 4, 28029 Madrid, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, 28029 Madrid, Spain
| | - Patricia Prieto
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Av. Monforte de Lemos 3-5, P-11, 28029 Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Plaza Ramón y Cajal, 28040 Madrid, Spain
| |
Collapse
|
4
|
Gonzalez-Menendez P, Sainz RM, Evelson P. Editorial: Oxidative metabolism in inflammation. Front Immunol 2024; 15:1507700. [PMID: 39497826 PMCID: PMC11532071 DOI: 10.3389/fimmu.2024.1507700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/07/2024] Open
Affiliation(s)
- Pedro Gonzalez-Menendez
- Departamento de Morfología y Biología Celular, School of Medicine, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Rosa M. Sainz
- Departamento de Morfología y Biología Celular, School of Medicine, Oviedo, Spain
- Instituto Universitario de Oncología del Principado de Asturias (IUOPA), University of Oviedo, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Hospital Universitario Central de Asturias (HUCA), Oviedo, Spain
| | - Pablo Evelson
- Facultad de Farmacia y Bioquímica, Departamento de Ciencias Químicas, Cátedra de Química General e Inorgánica, Universidad de Buenos Aires, Buenos Aires, Argentina
- CONICET, Instituto de Bioquímica y Medicina Molecular (IBIMOL), Universidad de Buenos Aires, Buenos Aires, Argentina
| |
Collapse
|
5
|
Lv X, Min J, Huang J, Wang H, Wei S, Huang C, Dai J, Chen Z, Zhou H, Xu Y, Zhao H, Liu Z, Wang J. Simultaneously Controlling Inflammation and Infection by Smart Nanomedicine Responding to the Inflammatory Microenvironment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403934. [PMID: 39225387 PMCID: PMC11497003 DOI: 10.1002/advs.202403934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/12/2024] [Indexed: 09/04/2024]
Abstract
The overactivated immune cells in the infectious lesion may lead to irreversible organ damages under severe infections. However, clinically used immunosuppressive anti-inflammatory drugs will usually disturb immune homeostasis and conversely increase the risk of infections. Regulating the balance between anti-inflammation and anti-infection is thus critical in treating certain infectious diseases. Herein, considering that hydrogen peroxide (H2O2), myeloperoxidase (MPO), and neutrophils are upregulated in the inflammatory microenvironment and closely related to the severity of appendectomy patients, an inflammatory-microenvironment-responsive nanomedicine is designed by using poly(lactic-co-glycolic) acid (PLGA) nanoparticles to load chlorine E6 (Ce6), a photosensitizer, and luminal (Lum), a chemiluminescent agent. The obtained Lum/Ce6@PLGA nanoparticles, being non-toxic within normal physiological environment, can generate cytotoxic single oxygen via bioluminescence resonance energy transfer (BRET) in the inflammatory microenvironment with upregulated H2O2 and MPO, simultaneously killing pathogens and excessive inflammatory immune cells in the lesion, without disturbing immune homeostasis. As evidenced in various clinically relevant bacterial infection models and virus-induced pneumonia, Lum/Ce6@PLGA nanoparticles appeared to be rather effective in controlling both infection and inflammation, resulting in significantly improved animal survival. Therefore, the BRET-based nanoparticles by simultaneously controlling infections and inflammation may be promising nano-therapeutics for treatment of severe infectious diseases.
Collapse
Affiliation(s)
- Xinjing Lv
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - Jie Min
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - Jie Huang
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - Hairong Wang
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - Song Wei
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - Chenxiao Huang
- Institutes of Biology and Medical SciencesJiangsu Key Laboratory of Infection and ImmunitySoochow UniversitySuzhouJiangsu215123China
| | - Jianfeng Dai
- Institutes of Biology and Medical SciencesJiangsu Key Laboratory of Infection and ImmunitySoochow UniversitySuzhouJiangsu215123China
| | - Zhengrong Chen
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - Huiting Zhou
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - Yunyun Xu
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - He Zhao
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon‐Based Functional Materials & DevicesSoochow UniversitySuzhouJiangsu215123China
| | - Jian Wang
- Children's Hospital of Soochow UniversityPediatric Research Institute of Soochow UniversitySuzhouJiangsu215123China
| |
Collapse
|
6
|
Matsui M, Kajikuri J, Kito H, Elboray EE, Suzuki T, Ohya S. Downregulation of IL-8 and IL-10 by LRRC8A Inhibition through the NOX2-Nrf2-CEBPB Transcriptional Axis in THP-1-Derived M 2 Macrophages. Int J Mol Sci 2024; 25:9612. [PMID: 39273558 PMCID: PMC11395230 DOI: 10.3390/ijms25179612] [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: 08/07/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
M2-polarized, tumor-associated macrophages (TAMs) produce pro-tumorigenic and angiogenic mediators, such as interleukin-8 (IL-8) and IL-10. Leucine-rich repeat-containing protein 8 members (LRRC8s) form volume-regulated anion channels and play an important role in macrophage functions by regulating cytokine and chemokine production. We herein examined the role of LRRC8A in IL-8 and IL-10 expression in THP-1-differentiated M2-like macrophages (M2-MACs), which are a useful tool for investigating TAMs. In M2-MACs, the pharmacological inhibition of LRRC8A led to hyperpolarizing responses after a transient depolarization phase, followed by a slight elevation in the intracellular concentration of Ca2+. Both the small interfering RNA-mediated and pharmacological inhibition of LRRC8A repressed the transcriptional expression of IL-8 and IL-10, resulting in a significant reduction in their secretion. The inhibition of LRRC8A decreased the nuclear translocation of phosphorylated nuclear factor-erythroid 2-related factor 2 (Nrf2), while the activation of Nrf2 reversed the LRRC8A inhibition-induced transcriptional repression of IL-8 and IL-10 in M2-MACs. We identified the CCAAT/enhancer-binding protein isoform B, CEBPB, as a downstream target of Nrf2 signaling in M2-MACs. Moreover, among several upstream candidates, the inhibition of nicotinamide adenine dinucleotide phosphate (NADPH) oxidase 2 (NOX2) suppressed the Nrf2-CEBPB transcriptional axis in M2-MACs. Collectively, the present results indicate that the inhibition of LRRC8A repressed IL-8 and IL-10 transcription in M2-MACs through the NOX2-Nrf2-CEBPB axis and suggest that LRRC8A inhibitors suppress the IL-10-mediated evasion of tumor immune surveillance and IL-8-mediated metastasis and neovascularization in TAMs.
Collapse
Affiliation(s)
- Miki Matsui
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan; (M.M.); (J.K.); (H.K.)
| | - Junko Kajikuri
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan; (M.M.); (J.K.); (H.K.)
| | - Hiroaki Kito
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan; (M.M.); (J.K.); (H.K.)
| | - Elghareeb E. Elboray
- Department of Complex Molecular Chemistry, SANKEN, Osaka University, Osaka 560-0043, Japan; (E.E.E.); (T.S.)
- Department of Chemistry, Faculty of Science, South Valley University, Qena 83523, Egypt
| | - Takayoshi Suzuki
- Department of Complex Molecular Chemistry, SANKEN, Osaka University, Osaka 560-0043, Japan; (E.E.E.); (T.S.)
| | - Susumu Ohya
- Department of Pharmacology, Graduate School of Medical Sciences, Nagoya City University, Nagoya 467-8601, Japan; (M.M.); (J.K.); (H.K.)
| |
Collapse
|
7
|
Diwan B, Yadav R, Goyal R, Sharma R. Sustained exposure to high glucose induces differential expression of cellular senescence markers in murine macrophages but impairs immunosurveillance response to senescent cells secretome. Biogerontology 2024; 25:627-647. [PMID: 38240923 DOI: 10.1007/s10522-024-10092-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 01/02/2024] [Indexed: 07/02/2024]
Abstract
The influence of chronic diseases on various facets of macrophage cellular senescence is poorly understood. This study evaluated the impact of chronic hyperglycemia on the induction of cellular senescence and subsequent immunosurveillance functions in RAW264.7 macrophages. Macrophages were cultured under normal glucose (NG; 5 mM), high glucose (HG; 20 mM), and very high glucose (VHG; 40 mM) conditions and assessed for markers of cellular senescence. Hyperglycemia induced strong upregulation of SA-β-gal activity, and loss of PCNA and Lamin B1 gene expression while markers of cell cycle arrest generally decreased. Non-significant changes in SASP-related proteins were observed while ROS levels slightly decreased and mitochondrial membrane potential increased. Protein concentration on the exosome membrane surface and their stability appeared to increase under hyperglycemic conditions. However, when macrophages were exposed to the secretory media (SM) of senescent preadipocytes, a dramatic increase in the levels of all inflammatory proteins was recorded especially in the VHG group that was also accompanied by upregulation of NF-κB and NLRP3 gene expression. SM treatment to hyperglycemic macrophages activated the TLR-2/Myd88 pathway but decreased the expression of scavenger receptors RAGE, CD36, and Olr-1 while CD44 and CXCL16 expression increased. On exposure to LPS, a strong upregulation in NO, ROS, and inflammatory cytokines was observed. Together, these results suggest that primary markers of cellular senescence are aberrantly expressed under chronic hyperglycemic conditions in macrophages with no significant SASP activation. Nonetheless, hyperglycemia strongly deregulates macrophage functions leading to impaired immunosurveillance of senescent cells and aggravation of inflamm-aging. This work provides novel insights into how hyperglycemia-induced dysfunctions can impact the potency of macrophages to manage senescent cell burden in aging tissues.
Collapse
Affiliation(s)
- Bhawna Diwan
- Nutrigerontology Laboratory, Faculty of Applied Sciences & Biotechnology, Shoolini University, Solan, 173229, India
| | - Rahul Yadav
- Nutrigerontology Laboratory, Faculty of Applied Sciences & Biotechnology, Shoolini University, Solan, 173229, India
| | - Rohit Goyal
- School of Pharmaceutical Sciences, Shoolini University, Solan, 173229, India
| | - Rohit Sharma
- Nutrigerontology Laboratory, Faculty of Applied Sciences & Biotechnology, Shoolini University, Solan, 173229, India.
| |
Collapse
|
8
|
Salti T, Braunstein I, Haimovich Y, Ziv T, Benhar M. Widespread S-persulfidation in activated macrophages as a protective mechanism against oxidative-inflammatory stress. Redox Biol 2024; 72:103125. [PMID: 38574432 PMCID: PMC11000178 DOI: 10.1016/j.redox.2024.103125] [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: 01/29/2024] [Revised: 03/10/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024] Open
Abstract
Acute inflammatory responses often involve the production of reactive oxygen and nitrogen species by innate immune cells, particularly macrophages. How activated macrophages protect themselves in the face of oxidative-inflammatory stress remains a long-standing question. Recent evidence implicates reactive sulfur species (RSS) in inflammatory responses; however, how endogenous RSS affect macrophage function and response to oxidative and inflammatory insults remains poorly understood. In this study, we investigated the endogenous pathways of RSS biogenesis and clearance in macrophages, with a particular focus on exploring how hydrogen sulfide (H2S)-mediated S-persulfidation influences macrophage responses to oxidative-inflammatory stress. We show that classical activation of mouse or human macrophages using lipopolysaccharide and interferon-γ (LPS/IFN-γ) triggers substantial production of H2S/RSS, leading to widespread protein persulfidation. Biochemical and proteomic analyses revealed that this surge in cellular S-persulfidation engaged ∼2% of total thiols and modified over 800 functionally diverse proteins. S-persulfidation was found to be largely dependent on the cystine importer xCT and the H2S-generating enzyme cystathionine γ-lyase and was independent of changes in the global proteome. We further investigated the role of the sulfide-oxidizing enzyme sulfide quinone oxidoreductase (SQOR), and found that it acts as a negative regulator of S-persulfidation. Elevated S-persulfidation following LPS/IFN-γ stimulation or SQOR inhibition was associated with increased resistance to oxidative stress. Upregulation of persulfides also inhibited the activation of the macrophage NLRP3 inflammasome and provided protection against inflammatory cell death. Collectively, our findings shed light on the metabolism and effects of RSS in macrophages and highlight the crucial role of persulfides in enabling macrophages to withstand and alleviate oxidative-inflammatory stress.
Collapse
Affiliation(s)
- Talal Salti
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ilana Braunstein
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yael Haimovich
- Smoler Proteomics Center and Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tamar Ziv
- Smoler Proteomics Center and Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
9
|
Kazek M, Chodáková L, Lehr K, Strych L, Nedbalová P, McMullen E, Bajgar A, Opekar S, Šimek P, Moos M, Doležal T. Glucose and trehalose metabolism through the cyclic pentose phosphate pathway shapes pathogen resistance and host protection in Drosophila. PLoS Biol 2024; 22:e3002299. [PMID: 38713712 PMCID: PMC11101078 DOI: 10.1371/journal.pbio.3002299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 05/17/2024] [Accepted: 04/12/2024] [Indexed: 05/09/2024] Open
Abstract
Activation of immune cells requires the remodeling of cell metabolism in order to support immune function. We study these metabolic changes through the infection of Drosophila larvae by parasitoid wasp. The parasitoid egg is neutralized by differentiating lamellocytes, which encapsulate the egg. A melanization cascade is initiated, producing toxic molecules to destroy the egg while the capsule also protects the host from the toxic reaction. We combined transcriptomics and metabolomics, including 13C-labeled glucose and trehalose tracing, as well as genetic manipulation of sugar metabolism to study changes in metabolism, specifically in Drosophila hemocytes. We found that hemocytes increase the expression of several carbohydrate transporters and accordingly uptake more sugar during infection. These carbohydrates are metabolized by increased glycolysis, associated with lactate production, and cyclic pentose phosphate pathway (PPP), in which glucose-6-phosphate is re-oxidized to maximize NADPH yield. Oxidative PPP is required for lamellocyte differentiation and resistance, as is systemic trehalose metabolism. In addition, fully differentiated lamellocytes use a cytoplasmic form of trehalase to cleave trehalose to glucose and fuel cyclic PPP. Intracellular trehalose metabolism is not required for lamellocyte differentiation, but its down-regulation elevates levels of reactive oxygen species, associated with increased resistance and reduced fitness. Our results suggest that sugar metabolism, and specifically cyclic PPP, within immune cells is important not only to fight infection but also to protect the host from its own immune response and for ensuring fitness of the survivor.
Collapse
Affiliation(s)
- Michalina Kazek
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Lenka Chodáková
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Katharina Lehr
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Lukáš Strych
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Pavla Nedbalová
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Ellen McMullen
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Adam Bajgar
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Stanislav Opekar
- Laboratory of Analytical Biochemistry and Metabolomics, Institute of Entomology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Petr Šimek
- Laboratory of Analytical Biochemistry and Metabolomics, Institute of Entomology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Martin Moos
- Laboratory of Analytical Biochemistry and Metabolomics, Institute of Entomology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
| | - Tomáš Doležal
- Department of molecular biology and genetics, Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| |
Collapse
|
10
|
Mohammad-Rafiei F, Negahdari S, Tahershamsi Z, Gheibihayat SM. Interface between Resolvins and Efferocytosis in Health and Disease. Cell Biochem Biophys 2024; 82:53-65. [PMID: 37794303 DOI: 10.1007/s12013-023-01187-4] [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: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 10/06/2023]
Abstract
Acute inflammation resolution acts as a vital process for active host response, tissue support, and homeostasis maintenance, during which resolvin D (RvD) and E (RvE) as mediators derived from omega-3 polyunsaturated fatty acids display specific and stereoselective anti-inflammations like restricting neutrophil infiltration and pro-resolving activities. On the other side of the coin, potent macrophage-mediated apoptotic cell clearance, namely efferocytosis, is essential for successful inflammation resolution. Further studies mentioned a linkage between efferocytosis and resolvins. For instance, resolvin D1 (RvD1), which is endogenously formed from docosahexaenoic acid within the inflammation resolution, thereby provoking efferocytosis. There is still limited information regarding the mechanism of action of RvD1-related efferocytosis enhancement at the molecular level. The current review article was conducted to explore recent data on how the efferocytosis process and resolvins relate to each other during the inflammation resolution in illness and health. Understanding different aspects of this connection sheds light on new curative approaches for medical conditions caused by defective efferocytosis and disrupted inflammation resolution.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Samira Negahdari
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany.
| |
Collapse
|
11
|
Majumder N, Roy S, Sharma A, Arora S, Vaishya R, Bandyopadhyay A, Ghosh S. Assessing the advantages of 3D bioprinting and 3D spheroids in deciphering the osteoarthritis healing mechanism using human chondrocytes and polarized macrophages. Biomed Mater 2024; 19:025005. [PMID: 38198731 DOI: 10.1088/1748-605x/ad1d18] [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: 07/03/2023] [Accepted: 01/10/2024] [Indexed: 01/12/2024]
Abstract
The molecular niche of an osteoarthritic microenvironment comprises the native chondrocytes, the circulatory immune cells, and their respective inflammatory mediators. Although M2 macrophages infiltrate the joint tissue during osteoarthritis (OA) to initiate cartilage repair, the mechanistic crosstalk that dwells underneath is still unknown. Our study established a co-culture system of human OA chondrocytes and M2 macrophages in 3D spheroids and 3D bioprinted silk-gelatin constructs. It is already well established that Silk fibroin-gelatin bioink supports chondrogenic differentiation due to upregulation of the Wnt/β-catenin pathway. Additionally, the presence of anti-inflammatory M2 macrophages significantly upregulated the expression of chondrogenic biomarkers (COL-II, ACAN) with an attenuated expression of the chondrocyte hypertrophy (COL-X), chondrocyte dedifferentiation (COL-I) and matrix catabolism (MMP-1 and MMP-13) genes even in the absence of the interleukins. Furthermore, the 3D bioprinted co-culture model displayed an upper hand in stimulating cartilage regeneration and OA inhibition than the spheroid model, underlining the role of silk fibroin-gelatin in encouraging chondrogenesis. Additionally, the 3D bioprinted silk-gelatin constructs further supported the maintenance of stable anti-inflammatory phenotype of M2 macrophage. Thus, the direct interaction between the primary OAC and M2 macrophages in the 3D context, along with the release of the soluble anti-inflammatory factors by the M2 cells, significantly contributed to a better understanding of the molecular mechanisms responsible for immune cell-mediated OA healing.
Collapse
Affiliation(s)
- Nilotpal Majumder
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Subhadeep Roy
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Aarushi Sharma
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi 110016, India
| | - Shuchi Arora
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Raju Vaishya
- Indraprastha Apollo Hospitals Delhi, New Delhi 110076, India
| | - Amitabha Bandyopadhyay
- Department of Biological Sciences & Bioengineering, Indian Institute of Technology, Kanpur 208016, India
| | - Sourabh Ghosh
- Regenerative Engineering Laboratory, Department of Textile and Fiber Engineering, Indian Institute of Technology, New Delhi 110016, India
| |
Collapse
|
12
|
Zhang J, Berk BC, Hsu CG. A Natural Metabolite and Inhibitor of the NLRP3 Inflammasome: 4-hydroxynonenal. JOURNAL OF CELLULAR IMMUNOLOGY 2024; 6:76-81. [PMID: 38873034 PMCID: PMC11174152 DOI: 10.33696/immunology.6.192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
The NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome, crucial in the innate immune response, is linked to various human diseases. However, the effect of endogenous metabolites, like 4-hydroxynonenal (HNE), on NLRP3 inflammasome activity remains underexplored. Recent research highlights HNE's inhibitory role in NLRP3 inflammasome activation, shedding light on its potential as an endogenous regulator of inflammatory responses. Studies demonstrate that HNE blocks NLRP3 inflammasome-mediated pyroptosis and IL-1β secretion. Additionally, covalent targeting emerges as a common mechanism for inhibiting NLRP3 inflammasome assembly, offering promising avenues for therapeutic intervention. Further investigation is needed to understand the impact of endogenous HNE on NLRP3 inflammasome activation, especially in settings where lipid peroxidation byproducts like HNE are produced. Understanding the intricate interplay between HNE and the NLRP3 inflammasome holds significant potential for unraveling novel therapeutic strategies for inflammatory disorders.
Collapse
Affiliation(s)
- Jinmin Zhang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Bradford C. Berk
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
- Department of Physical Medicine and Rehabilitation, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Chia George Hsu
- Department of Kinesiology, The University of Texas at San Antonio, San Antonio, TX, USA
| |
Collapse
|
13
|
Li Z, Yan M, Wang Z, An Y, Wei X, Li T, Xu M, Xia Y, Wang L, Gao C. Ferroptosis of Endothelial Cells Triggered by Erythrophagocytosis Contributes to Thrombogenesis in Uremia. Thromb Haemost 2023; 123:1116-1128. [PMID: 37364609 PMCID: PMC10686749 DOI: 10.1055/a-2117-7890] [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: 09/28/2022] [Accepted: 06/12/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND Although thrombosis events are the leading complication of uremia, their mechanism is largely unknown. The interaction between endothelial cells (ECs) and red blood cells (RBCs) in uremic solutes and its prothrombotic role need to be investigated. METHODS AND RESULTS Here, we established an in vitro co-incubation model of uremic RBC and EC as well as a uremic rat model induced by adenine. Using flow cytometry, confocal microscopy, and electron microscopy, we found increased erythrophagocytosis by EC accompanied by increased reactive oxygen species, lipid peroxidation, and impairment of mitochondria, indicating that ECs undergo ferroptosis. Further investigations showed increased proteins' expression of heme oxygenase-1 and ferritin and labile iron pool accumulation in EC, which could be suppressed by deferoxamine (DFO). The ferroptosis-negative regulators glutathione peroxidase 4 and SLC7A11 were decreased in our erythrophagocytosis model and could be enhanced by ferrostatin-1 or DFO. In vivo, we observed that vascular EC phagocytosed RBC and underwent ferroptosis in the kidney of the uremic rat, which could be inhibited by blocking the phagocytic pathway or inhibiting ferroptosis. Next, we found that the high tendency of thrombus formation was accompanied by erythrophagocytosis-induced ferroptosis in vitro and in vivo. Importantly, we further revealed that upregulated TMEM16F expression mediated phosphatidylserine externalization on ferroptotic EC, which contributed to a uremia-associated hypercoagulable state. CONCLUSION Our results indicate that erythrophagocytosis-triggered ferroptosis followed by phosphatidylserine exposure of EC may play a key role in uremic thrombotic complications, which may be a promising target to prevent thrombogenesis of uremia.
Collapse
Affiliation(s)
- Zhanni Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Meishan Yan
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Zelong Wang
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Yao An
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Xinyu Wei
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Tingting Li
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Minghui Xu
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Yanshi Xia
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| | - Liqiu Wang
- Department of Clinical Laboratory, The Fifth Hospital, Harbin Medical University, Daqing, China
| | - Chunyan Gao
- Department of Medical Laboratory Science and Technology, Harbin Medical University-Daqing, Daqing, China
| |
Collapse
|
14
|
Kweon B, Kim DU, Oh JY, Bae GS, Park SJ. Guggulsterone protects against lipopolysaccharide-induced inflammation and lethal endotoxemia via heme oxygenase-1. Int Immunopharmacol 2023; 124:111073. [PMID: 37844468 DOI: 10.1016/j.intimp.2023.111073] [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: 08/14/2023] [Revised: 10/10/2023] [Accepted: 10/10/2023] [Indexed: 10/18/2023]
Abstract
Guggulsterone (GS) is a phytosterol used to treat inflammatory diseases. Although many studies have examined the anti-inflammatory activities of GS, the detailed mechanisms of GS in lipopolysaccharide (LPS)-induced inflammation and endotoxemia have not yet been examined. Therefore, we investigated the anti-inflammatory effects of GS on LPS-induced inflammation. In murine peritoneal macrophages, the anti-inflammatory activity of GS was primarily mediated by heme oxygenase-1 (HO-1) induction. HO-1 induction by GS was mediated by GSH depletion and reactive oxygen species (ROS) production. The ROS generated by GS caused the phosphorylation of GSK3β (ser9/21) and p38, leading to the translocation of nuclear factor erythroid-related factor 2 (Nrf2), which ultimately induced HO-1. In addition, GS pretreatment significantly inhibited inducible nitric oxide synthase (iNOS), iNOS-derived NO, and COX-2 protein and mRNA expression, and production of COX-derived prostaglandin PGE2, interleukin (IL)-1β, IL-6, and tumor necrosis factor-α (TNF-α). In a mouse model of endotoxemia, GS treatment prolonged survival and inhibited the expression of inflammatory mediators, including IL-1β, IL-6, and TNF-α. GS treatment also inhibited LPS-induced liver injury. These results suggest that GS-induced HO-1 could exert anti-inflammatory effects via ROS-dependent GSK (ser21/9)-p38 phosphorylation and nuclear translocation of Nrf2.
Collapse
Affiliation(s)
- Bitna Kweon
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea; Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea
| | - Dong-Uk Kim
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea; Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea
| | - Jin-Young Oh
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea; Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea
| | - Gi-Sang Bae
- Department of Pharmacology, School of Korean Medicine, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea; Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea.
| | - Sung-Joo Park
- Hanbang Cardio-Renal Syndrome Research Center, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea; Department of Herbology, School of Korean Medicine, Wonkwang University, Iksan, 54538 Jeonbuk, South Korea.
| |
Collapse
|
15
|
Shen X, Chen H, Zhang H, Luo L, Wen T, Liu L, Hu Q, Wang L. A natural sesquiterpene lactone isolinderalactone attenuates lipopolysaccharide-induced inflammatory response and acute lung injury through inhibition of NF-κB pathway and activation Nrf2 pathway in macrophages. Int Immunopharmacol 2023; 124:110965. [PMID: 37741124 DOI: 10.1016/j.intimp.2023.110965] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/07/2023] [Accepted: 09/17/2023] [Indexed: 09/25/2023]
Abstract
Isolinderalactone is the main sesquiterpene lactone isolated from Lindera aggregata, a traditional Chinese medicine widely used to treat pain and inflammation. Although isolinderalactone has been demonstrated to possess anti-cancer effect, its anti-inflammatory activity and underlying mechanism has not been well characterized. Herein, isolinderalactone was able to significantly inhibit the production of NO and PGE2 by reducing the expressions of iNOS and COX2 in LPS-stimulated RAW264.7 macrophages and BMDMs, and decreased the mRNA levels of IL-1β, IL-6, and TNF-α in LPS-induced RAW264.7 cells. In vivo, isolinderalactone effectively alleviated LPS-induced acute lung injury (ALI), which manifested as reduction in pulmonary inflammatory infiltration, myeloperoxidase activity, and production of PGE2, IL-1β, IL-6, TNF-α, and malondialdehyde. Furthermore, isolinderalactone inhibited phosphorylation of IKKα/β, phosphorylation and degradation of IκBα, and nuclear translocation of NF-κB p65, thereby blocking NF-κB pro-inflammatory pathway. Meanwhile, isolinderalactone reduced the intracellular ROS through promoting the activation of Nrf2-HMOX1 antioxidant axis. By using drug affinity responsive target stability assay and molecular docking, isolinderalactone was found to covalently interact with IKKα/β and Keap1, which may contribute to its anti-inflammatory action. Additionally, a thiol donor β-mercaptoethanol significantly abolished isolinderalactone-mediated anti-inflammatory action in vitro, indicating the crucial role of the unsaturated lactone of isolinderalactone on its anti-inflammatory effects. Taken together, isolinderalactone protected against LPS-induced ALI in mice, which may be associated with its inhibition of NF-κB pathway and activation of Nrf2 signaling in macrophages.
Collapse
Affiliation(s)
- Xiaofei Shen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hongqing Chen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hai Zhang
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Liuling Luo
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tian Wen
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Lu Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiongying Hu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Lun Wang
- Center for Natural Products Research, Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China.
| |
Collapse
|
16
|
Chun J, Mah SY, Kim YS. Anti-Inflammatory Effect of Ebractenoid F, a Major Active Compound of Euphorbia ebracteolata Hayata, through Inhibition of Nuclear Factor-κB Activation. PLANTS (BASEL, SWITZERLAND) 2023; 12:2845. [PMID: 37570999 PMCID: PMC10421244 DOI: 10.3390/plants12152845] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/13/2023] [Accepted: 07/31/2023] [Indexed: 08/13/2023]
Abstract
Euphorbia ebracteolata Hayata (Euphorbiaceae family) is a perennial plant that is widely distributed in Korea, Japan, and China. Its roots contain bioactive diterpenes that have anti-inflammatory properties. However, the anti-inflammatory mechanisms are not yet fully understood. This study aimed to identify the most active anti-inflammatory compound from the roots of E. ebracteolata Hayata, using bioassay-guided fractionation and a combinative method of high-speed countercurrent chromatography (HSCCC) and preparative high-performance liquid chromatography (HPLC). Then, we investigated its anti-inflammatory mechanism in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages. Ebractenoid F was identified as the most potent bioactive compound of E. ebracteolata Hayata. Ebractenoid F significantly decreased nitric oxide (NO) production and nuclear factor-κB (NF-κB) activation induced by LPS in RAW 264.7 macrophages. Moreover, ebractenoid F decreased the degradation of inhibitory κB-α, the nuclear translocation of the p65 and p50 subunits of NF-κB, and the expression of NF-κB downstream genes. Furthermore, ebractenoid F inhibited the phosphorylation of Akt and mitogen-activated protein kinases (MAPKs), such as extracellular signal-regulated kinase (ERK) and c-Jun NH2 terminal kinase (JNK), in LPS-stimulated RAW 264.7 cells. In conclusion, ebractenoid F exerts the most potent anti-inflammatory effect by suppressing NF-κB-mediated NO production in LPS-stimulated RAW 264.7 cells. Ebractenoid F may be a useful therapeutic compound for the prevention or treatment of inflammation-associated diseases.
Collapse
Affiliation(s)
- Jaemoo Chun
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Sang Yeon Mah
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Yeong Shik Kim
- Natural Products Research Institute, College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
17
|
Sezgin-Bayindir Z, Losada-Barreiro S, Fernández-Bravo S, Bravo-Díaz C. Innovative Delivery and Release Systems for Antioxidants and Other Active Substances in the Treatment of Cancer. Pharmaceuticals (Basel) 2023; 16:1038. [PMID: 37513948 PMCID: PMC10383431 DOI: 10.3390/ph16071038] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023] Open
Abstract
Cancer is one of the major diseases leading to death worldwide, and the fight against the disease is still challenging. Cancer diseases are usually associated with increased oxidative stress and the accumulation of reactive oxygen and nitrogen species as a result of metabolic alterations or signaling aberrations. While numerous antioxidants exhibit potential therapeutic properties, their clinical efficiency against cancer is limited and even unproven. Conventional anticancer antioxidants and drugs have, among others, the great disadvantage of low bioavailability, poor targeting efficiency, and serious side effects, constraining their use in the fight against diseases. Here, we review the rationale for and recent advances in potential delivery systems that could eventually be employed in clinical research on antioxidant therapy in cancer. We also review some of the various strategies aimed at enhancing the solubility of poorly water-soluble active drugs, including engineered delivery systems such as lipid-based, polymeric, and inorganic formulations. The use of cyclodextrins, micro- and nanoemulsions, and thermosensitive smart liposomes as useful systems for the delivery and release of poorly aqueous-soluble drugs, improving their bioactivity and stability, is also addressed. We also provide some details on their formulation processes and their use in a variety of medical applications. Finally, we briefly cover a case study specifically focused on the use of delivery systems to minimize oral cancer and associated dental problems.
Collapse
Affiliation(s)
- Zerrin Sezgin-Bayindir
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University, Ankara 06560, Turkey
| | - Sonia Losada-Barreiro
- Departamento de Química-Física, Facultade de Química, Universidade de Vigo, 36200 Vigo, Spain
| | - Sofía Fernández-Bravo
- Odontology Department, Primary Health Care Unit, Galician Health Service (SERGAS), Camiño do Lodairo s/n, 15570 Narón, Spain
| | - Carlos Bravo-Díaz
- Departamento de Química-Física, Facultade de Química, Universidade de Vigo, 36200 Vigo, Spain
| |
Collapse
|
18
|
Asaro RJ, Profumo E, Buttari B, Cabrales P. The Double-Edged Sword of Erythrocytes in Health and Disease via Their Adhesiveness. Int J Mol Sci 2023; 24:10382. [PMID: 37373527 DOI: 10.3390/ijms241210382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
Their widespread presence throughout the vasculature, coupled with their reactivity, and thereby to their potential to release reactive oxidative species, or to utilize their anti-oxidative capacities, has promoted much discussion of the role(s) of red blood cells (RBCs) in the progression of health or, alternatively, a wide range of disease states. Moreover, these role(s) have been linked to the development of adhesiveness and, in fact, thereby to the essential pathway to their eventual clearance, e.g., by macrophages in the spleen. These disparate roles coupled with the mechanisms involved are reviewed and given. Following an analysis, novel perspectives are provided; these perspectives can lead to novel assays for identifying the potential for RBC adhesiveness as suggested herein. We describe this paradigm, that involves RBC adhesiveness, hemolysis, and ghost formation, with examples including, inter alia, the progression of atherosclerosis and the suppression of tumor growth along with other disease states.
Collapse
Affiliation(s)
- Robert J Asaro
- Department of Structural Engineering, University of California, La Jolla, CA 92093-0085, USA
| | - Elisabetta Profumo
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Brigitta Buttari
- Department of Cardiovascular and Endocrine-Metabolic Diseases, and Aging, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Pedro Cabrales
- Department of Bioengineering, University of California, La Jolla, CA 92093-0085, USA
| |
Collapse
|
19
|
Fang J, Chen W, Hou P, Liu Z, Zuo M, Liu S, Feng C, Han Y, Li P, Shi Y, Shao C. NAD + metabolism-based immunoregulation and therapeutic potential. Cell Biosci 2023; 13:81. [PMID: 37165408 PMCID: PMC10171153 DOI: 10.1186/s13578-023-01031-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/14/2023] [Indexed: 05/12/2023] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) is a critical metabolite that acts as a cofactor in energy metabolism, and serves as a cosubstrate for non-redox NAD+-dependent enzymes, including sirtuins, CD38 and poly(ADP-ribose) polymerases. NAD+ metabolism can regulate functionality attributes of innate and adaptive immune cells and contribute to inflammatory responses. Thus, the manipulation of NAD+ bioavailability can reshape the courses of immunological diseases. Here, we review the basics of NAD+ biochemistry and its roles in the immune response, and discuss current challenges and the future translational potential of NAD+ research in the development of therapeutics for inflammatory diseases, such as COVID-19.
Collapse
Affiliation(s)
- Jiankai Fang
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Wangwang Chen
- Laboratory Animal Center, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Pengbo Hou
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Zhanhong Liu
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Muqiu Zuo
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Shisong Liu
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
| | - Chao Feng
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Yuyi Han
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China
- Department of Experimental Medicine and Biochemical Sciences, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Peishan Li
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| | - Yufang Shi
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
- Shanghai Institute of Nutrition and Health, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Changshun Shao
- Institutes for Translational Medicine, State Key Laboratory of Radiation Medicine and Protection, The First Affiliated Hospital of Soochow University, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
20
|
Pereira MMR, de Oliveira FM, da Costa AC, Junqueira-Kipnis AP, Kipnis A. Ferritin from Mycobacterium abscessus is involved in resistance to antibiotics and oxidative stress. Appl Microbiol Biotechnol 2023; 107:2577-2595. [PMID: 36862179 DOI: 10.1007/s00253-023-12420-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/22/2023] [Accepted: 01/30/2023] [Indexed: 03/03/2023]
Abstract
Mycobacterium abscessus subsp. massiliense (Mycma) is a rapidly growing Mycobacterium belonging to the M. abscessus complex that is often associated with lung and soft tissue infection outbreaks. Mycma is resistant to many antimicrobials, including those used for treating tuberculosis. Therefore, Mycma infections are difficult to treat and may lead to high infectious complication rates. Iron is essential for bacterial growth and establishment of infection. During infection, the host reduces iron concentrations as a defense mechanism. To counteract the host-induced iron deficiency, Mycma produces siderophores to capture iron. Mycma has two ferritins (encoded by mycma_0076 and mycma_0077) modulated by different iron concentrations, which allow the survival of this pathogen during iron scarcity. In this study, we constructed knockout (Mycma 0076KO) and complemented (Mycma 0076KOc) gene strains for mycma_0076 to understand the function of 0076 ferritin. Deletion of mycma_0076 in Mycma led to the transition in colony morphology from smooth to rough, alteration of the glycopeptidolipids spectra, increased permeability of the envelope, reduction in biofilm formation, increased susceptibility to antimicrobials and hydrogen peroxide-induced oxidative stress, and decreased internalization by macrophages. This study shows that Mycma_0076 ferritin in Mycma is involved in resistance to oxidative stress and antimicrobials, and alteration of cell envelope architecture. KEY POINTS: • Deletion of the mycma_0076 gene altered colony morphology to rough; • Mycma 0076KO changed GPL profile; • Absence of Mycma_0076 ferritin results in increased susceptibility to antimicrobials and oxidative stress in Mycma. Legend: a In wild-type M. abscessus subsp. massiliense strain, iron is captured from the environment by carboxymycobactins and mycobactins (1). Iron-dependent regulator (IdeR) proteins bind to ferrous iron (Fe+2) in the bacterial cytoplasm leading to the activation of the IdeR-Fe+2 complex (2). The activated complex binds to the promoter regions of iron-dependent genes, called iron box, which in turn help in the recruitment of RNA polymerase to promote transcription of genes such as mycma_0076 and mycma_0077 ferritin genes (3). Mycma_0076 and Mycma_0077 ferritins bind to excess iron in the medium and promote Fe2+ oxidation into ferric iron (Fe3+) and store iron molecules to be released under iron scarcity conditions. (4) Genes related to biosynthesis and transport of glycopeptidolipids (GPL) are expressed normally and the cell envelope is composed of different GPL species (colored squares represented on the cell surface (GPLs). Consequently, WT Mycma present smooth colony phenotype (5). b In Mycma 0076KO strain, the lack of ferritin 0076 causes overexpression of mycma_0077 (6), but does not restore wild-type iron homeostasis and thus may result in free intracellular iron, even in the presence of miniferritins (MaDps). The excess iron potentiates oxidative stress (7) by generating hydroxyl radicals through Fenton Reaction. During this process, through an unknown mechanism, that could involve Lsr2 (8), the expression of GPL synthesis locus is regulated positively and/or negatively, resulting in alteration of GPL composition in the membrane (as represented by different colors of squares on the cell surface), resulting in a rough colony phenotype (9). The changes of GPL can increase cell wall permeability, contributing to antimicrobial susceptibility (10).
Collapse
Affiliation(s)
- Maria Micaella Rodrigues Pereira
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
- Tropical Medicine and Public Health Graduate Program at Federal, University of Goiás, Goiânia, GO, Brazil
| | - Fábio Muniz de Oliveira
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil
- Tropical Medicine and Public Health Graduate Program at Federal, University of Goiás, Goiânia, GO, Brazil
- Indiana Center for Regenerative Medicine and Engineering, School of Medicine, Indiana University, Indianapolis, IN, USA
| | | | | | - André Kipnis
- Institute of Tropical Pathology and Public Health, Federal University of Goiás, Goiânia, GO, Brazil.
| |
Collapse
|
21
|
Peraza DA, Povo-Retana A, Mojena M, García-Redondo AB, Avilés P, Boscá L, Valenzuela C. Trabectedin modulates macrophage polarization in the tumor-microenvironment. Role of K V1.3 and K V1.5 channels. Biomed Pharmacother 2023; 161:114548. [PMID: 36940615 DOI: 10.1016/j.biopha.2023.114548] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 03/03/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023] Open
Abstract
Immune cells have an important role in the tumor-microenvironment. Macrophages may tune the immune response toward inflammatory or tolerance pathways. Tumor-associated macrophages (TAM) have a string of immunosuppressive functions and they are considered a therapeutic target in cancer. This study aimed to analyze the effects of trabectedin, an antitumor agent, on the tumor-microenvironment through the characterization of the electrophysiological and molecular phenotype of macrophages. Experiments were performed using the whole-cell configuration of the patch-clamp technique in resident peritoneal mouse macrophages. Trabectedin does not directly interact with KV1.5 and KV1.3 channels, but their treatment (16 h) with sub-cytotoxic concentrations of trabectedin increased their KV current due to an upregulation of KV1.3 channels. In vitro generated TAM (TAMiv) exhibited an M2-like phenotype. TAMiv generated a small KV current and express high levels of M2 markers. K+ current from TAMs isolated from tumors generated in mice is a mixture of KV and KCa, and in TAM isolated from tumors generated in trabectedin-treated mice, the current is mostly driven by KCa. We conclude that the antitumor capacity of trabectedin is not only due to its effects on tumor cells, but also to the modulation of the tumor microenvironment, due, at least in part, to the modulation of the expression of different macrophage ion channels.
Collapse
Affiliation(s)
- Diego A Peraza
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain.
| | - Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain
| | - Marina Mojena
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain
| | - Ana B García-Redondo
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; Hospital La Paz Institute for Health Research (IdiPAZ), 28046 Madrid, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Pablo Avilés
- Departamento de Toxicología y Farmacología Preclínica, PharmaMar S.A., 28770 Colmenar Viejo, Madrid, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), 28029 Madrid, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain.
| |
Collapse
|
22
|
Murani E, Hadlich F. Exploration of genotype-by-environment interactions affecting gene expression responses in porcine immune cells. Front Genet 2023; 14:1157267. [PMID: 37007953 PMCID: PMC10061014 DOI: 10.3389/fgene.2023.1157267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
As one of the keys to healthy performance, robustness of farm animals is gaining importance, and with this comes increasing interest in genetic dissection of genotype-by-environment interactions (G×E). Changes in gene expression are among the most sensitive responses conveying adaptation to environmental stimuli. Environmentally responsive regulatory variation thus likely plays a central role in G×E. In the present study, we set out to detect action of environmentally responsive cis-regulatory variation by the analysis of condition-dependent allele specific expression (cd-ASE) in porcine immune cells. For this, we harnessed mRNA-sequencing data of peripheral blood mononuclear cells (PBMCs) stimulated in vitro with lipopolysaccharide, dexamethasone, or their combination. These treatments mimic common challenges such as bacterial infection or stress, and induce vast transcriptome changes. About two thirds of the examined loci showed significant ASE in at least one treatment, and out of those about ten percent exhibited cd-ASE. Most of the ASE variants were not yet reported in the PigGTEx Atlas. Genes showing cd-ASE were enriched in cytokine signaling in immune system and include several key candidates for animal health. In contrast, genes showing no ASE featured cell-cycle related functions. We confirmed LPS-dependent ASE for one of the top candidates, SOD2, which ranks among the major response genes in LPS-stimulated monocytes. The results of the present study demonstrate the potential of in vitro cell models coupled with cd-ASE analysis for the investigation of G×E in farm animals. The identified loci may benefit efforts to unravel the genetic basis of robustness and improvement of health and welfare in pigs.
Collapse
Affiliation(s)
- Eduard Murani
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | | |
Collapse
|
23
|
BDE-47 Induces Immunotoxicity in RAW264.7 Macrophages through the Reactive Oxygen Species-Mediated Mitochondrial Apoptotic Pathway. Molecules 2023; 28:molecules28052036. [PMID: 36903282 PMCID: PMC10004313 DOI: 10.3390/molecules28052036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Polybrominated diphenyl ethers (PBDEs) are classic and emerging pollutants that are potentially harmful to the human immune system. Research on their immunotoxicity and mechanisms suggests that they play an important role in the resulting pernicious effects of PBDEs. 2,2',4,4'-Tetrabrominated biphenyl ether (BDE-47) is the most biotoxic PBDE congener, and, in this study, we evaluated its toxicity toward RAW264.7 cells of mouse macrophages. The results show that exposure to BDE-47 led to a significant decrease in cell viability and a prominent increase in apoptosis. A decrease in mitochondrial membrane potential (MMP) and an increase in cytochrome C release and caspase cascade activation thus demonstrate that cell apoptosis induced by BDE-47 occurs via the mitochondrial pathway. In addition, BDE-47 inhibits phagocytosis in RAW264.7 cells, changes the related immune factor index, and causes immune function damage. Furthermore, we discovered a significant increase in the level of cellular reactive oxygen species (ROS), and the regulation of genes linked to oxidative stress was also demonstrated using transcriptome sequencing. The degree of apoptosis and immune function impairment caused by BDE-47 could be reversed after treatment with the antioxidant NAC and, conversely, exacerbated by treatment with the ROS-inducer BSO. These findings indicate that oxidative damage caused by BDE-47 is a critical event that leads to mitochondrial apoptosis in RAW264.7 macrophages, ultimately resulting in the suppression of immune function.
Collapse
|
24
|
Förster S, Niu Y, Eggers B, Nokhbehsaim M, Kramer FJ, Bekeschus S, Mustea A, Stope MB. Modulation of the Tumor-Associated Immuno-Environment by Non-Invasive Physical Plasma. Cancers (Basel) 2023; 15:cancers15041073. [PMID: 36831415 PMCID: PMC9953794 DOI: 10.3390/cancers15041073] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
Over the past 15 years, investigating the efficacy of non-invasive physical plasma (NIPP) in cancer treatment as a safe oxidative stress inducer has become an active area of research. So far, most studies focused on the NIPP-induced apoptotic death of tumor cells. However, whether NIPP plays a role in the anti-tumor immune responses need to be deciphered in detail. In this review, we summarized the current knowledge of the potential effects of NIPP on immune cells, tumor-immune interactions, and the immunosuppressive tumor microenvironment. In general, relying on their inherent anti-oxidative defense systems, immune cells show a more resistant character than cancer cells in the NIPP-induced apoptosis, which is an important reason why NIPP is considered promising in cancer management. Moreover, NIPP treatment induces immunogenic cell death of cancer cells, leading to maturation of dendritic cells and activation of cytotoxic CD8+ T cells to further eliminate the cancer cells. Some studies also suggest that NIPP treatment may promote anti-tumor immune responses via other mechanisms such as inhibiting tumor angiogenesis and the desmoplasia of tumor stroma. Though more evidence is required, we expect a bright future for applying NIPP in clinical cancer management.
Collapse
Affiliation(s)
- Sarah Förster
- Department of Pathology, University Hospital Bonn, 35127 Bonn, Germany
| | - Yuequn Niu
- Department of Pathology, University Hospital Bonn, 35127 Bonn, Germany
| | - Benedikt Eggers
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, 53111 Bonn, Germany
| | - Marjan Nokhbehsaim
- Section of Experimental Dento-Maxillo-Facial Medicine, University Hospital Bonn, 53111 Bonn, Germany
| | - Franz-Josef Kramer
- Department of Oral, Maxillofacial and Plastic Surgery, University Hospital Bonn, 53111 Bonn, Germany
| | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), 17489 Greifswald, Germany
| | - Alexander Mustea
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, 53127 Bonn, Germany
| | - Matthias B. Stope
- Department of Gynecology and Gynecological Oncology, University Hospital Bonn, 53127 Bonn, Germany
- Correspondence: ; Tel.: +49-228-287-11361
| |
Collapse
|
25
|
Balas M, Iconaru SL, Dinischiotu A, Buton N, Predoi D. Response of the Endogenous Antioxidant Defense System Induced in RAW 264.7 Macrophages upon Exposure to Dextran-Coated Iron Oxide Nanoparticles. Pharmaceutics 2023; 15:552. [PMID: 36839874 PMCID: PMC9967892 DOI: 10.3390/pharmaceutics15020552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Presently, iron oxide nanoparticles are the only ones approved for clinical use as contrast agents in magnetic resonance imaging (MRI). Even though there is a high demand for these types of nanoparticles both for clinical use as well as for research, there are difficulties in obtaining stable nanoparticles with reproducible properties. In this context, in this study, we report the obtaining by an adapted coprecipitation method of dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs). The morphology and structure of the dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs) were determined using scanning electron microscopy (SEM) and transmission electron microscopy (TEM). The TEM and SEM micrographs highlighted the obtaining of particles of nanometric size and spherical shape morphology. Furthermore, the high-resolution transmission electron microscopy (HRTEM), as well as selected area diffraction (SAED), revealed that the obtained samples presented the structure of cubic maghemite. In this study, we also explored the effects of the co-precipitation synthesized dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs) on the redox status of macrophages. For cytotoxicity evaluation of these NPs, murine macrophages (RAW 264.7 cell line) were exposed to different concentrations of dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs) corresponding to 0-500 μg Fe3+/mL and incubated for 24, 48, and 72 h. Intracellular iron uptake, changes in the oxidative stress parameters (reactive oxygen species production and malondialdehyde level), and the activity of antioxidant enzymes, as well as GSH concentration in cells, were evaluated after incubation with a lower (50 μg Fe3+/mL) and higher (500 μg Fe3+/mL) dose of NPs. The results indicated a significant decrease in RAW 264.7 cell viability after 72 h in the presence of NPs at concentrations above 25 μg Fe3+/mL. An important accumulation of NPs, dependent on dose and exposure time, was detected in macrophages, but it induced only a limited raise in the oxidative status. We showed here that the antioxidant capacity of RAW 264.7 macrophages was efficient in counteracting dextran-coated maghemite nanoparticles (ɤ-Fe2O3 NPs) toxicity even at higher doses.
Collapse
Affiliation(s)
- Mihaela Balas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| | | | - Anca Dinischiotu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 91-95 Splaiul Independentei, 050095 Bucharest, Romania
| | - Nicolas Buton
- National Institute of Materials Physics, 405A Atomistilor Street, 077125 Magurele, Romania
| | - Daniela Predoi
- HORIBA Jobin Yvon S.A.S., 6-18, Rue du Canal, CEDEX, 91165 Longjumeau, France
| |
Collapse
|
26
|
Hsu CG, Li W, Sowden M, Chávez CL, Berk BC. Pnpt1 mediates NLRP3 inflammasome activation by MAVS and metabolic reprogramming in macrophages. Cell Mol Immunol 2023; 20:131-142. [PMID: 36596874 PMCID: PMC9886977 DOI: 10.1038/s41423-022-00962-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 11/13/2022] [Indexed: 01/05/2023] Open
Abstract
Polyribonucleotide nucleotidyltransferase 1 (Pnpt1) plays critical roles in mitochondrial homeostasis by controlling mitochondrial RNA (mt-RNA) processing, trafficking and degradation. Pnpt1 deficiency results in mitochondrial dysfunction that triggers a type I interferon response, suggesting a role in inflammation. However, the role of Pnpt1 in inflammasome activation remains largely unknown. In this study, we generated myeloid-specific Pnpt1-knockout mice and demonstrated that Pnpt1 depletion enhanced interleukin-1 beta (IL-1β) and interleukin-18 (IL-18) secretion in a mouse sepsis model. Using cultured peritoneal and bone marrow-derived macrophages, we demonstrated that Pnpt1 regulated NLRP3 inflammasome-dependent IL-1β release in response to lipopolysaccharide (LPS), followed by nigericin, ATP or poly (I:C) treatment. Pnpt1 deficiency in macrophages increased glycolysis after LPS administration and mt-reactive oxygen species (mt-ROS) after NLRP3 inflammasome activation. Pnpt1 activation of the inflammasome was dependent on increased glycolysis and the expression of mitochondrial antiviral-signaling protein (MAVS) but not NF-κB signaling. Collectively, these data suggest that Pnpt1 is an important mediator of inflammation, as shown by activation of the NLRP3 inflammasome in murine sepsis and cultured macrophages.
Collapse
Affiliation(s)
- Chia George Hsu
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, USA
| | - Wenjia Li
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, USA
- Department of Cardiovascular Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Mark Sowden
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, USA
| | - Camila Lage Chávez
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, USA
| | - Bradford C Berk
- Department of Medicine, Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, USA.
| |
Collapse
|
27
|
USP13 reduces septic mediated cardiomyocyte oxidative stress and inflammation by inducing Nrf2. Allergol Immunopathol (Madr) 2023; 51:160-167. [PMID: 36916102 DOI: 10.15586/aei.v51i1.813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/12/2023] [Indexed: 03/16/2023]
Abstract
BACKGROUND Sepsis is a common cardiovascular complication that can cause heart damage. The regulatory role of ubiquitin-specific peptidase 13 (USP13) on erythroid 2-related factor 2 (Nrf2) has been reported, but its regulatory role in septic cardiomyopathy remains unclear. METHODS The Sprague Dawley (SD) rat model of septic myocardial injury was constructed by lipopolysaccharides (LPS). The serum lactate dehydrogenase (LDH) and creatine kinase (CK) levels were detected, the mRNA and protein expression levels of Nrf2 and USP13 in tissues were detected by real-time quantitative reverse transcription PCR (qRT-PCR) and western blot (WB), and the expression of USP13 at the treatment time of 3 h, 6 h, and 12 h was also detected. The cell viability and USP13, Nrf-2 and heme oxygenase-1 (HO-1) expression levels of H9C2-treated cells by LPS and the oxidative stress level and inflammatory response of H9C2 cells were detected by enzyme-linked immunosorbent assay (ELISA) and WB. RESULTS The results showed that USP13 was downregulated in septic myocardial injury tissues, and the Nrf2 level was increased in vitro after the cells were treated with LPS. Overexpression of USP13 further induced Nrf2 to reduce apoptosis, oxidative stress, and expression of inflammatory factors. CONCLUSION In conclusion, this study demonstrated that USP13 was downregulated in septic myocardial injury tissues, and USP13 overexpression increased Nrf2 levels and reduced apoptosis. Further studies showed that USP13 reduced LPS-induced oxidative stress and inflammation by inducing Nrf2.
Collapse
|
28
|
Thakur M, Muniyappa K. Macrophage activation highlight an important role for NER proteins in the survival, latency and multiplication of Mycobacterium tuberculosis. Tuberculosis (Edinb) 2023; 138:102284. [PMID: 36459831 DOI: 10.1016/j.tube.2022.102284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 11/14/2022] [Accepted: 11/20/2022] [Indexed: 11/27/2022]
Abstract
Nucleotide excision repair (NER) is one of the most extensively studied DNA repair processes in both prokaryotes and eukaryotes. The NER pathway is a highly conserved, ATP-dependent multi-step process involving several proteins/enzymes that function in a concerted manner to recognize and excise a wide spectrum of helix-distorting DNA lesions and bulky adducts by nuclease cleavage on either side of the damaged bases. As such, the NER pathway of Mycobacterium tuberculosis (Mtb) is essential for its survival within the hostile environment of macrophages and disease progression. This review focuses on present published knowledge about the crucial roles of Mtb NER proteins in the survival and multiplication of the pathogen within the macrophages and as potential targets for drug discovery.
Collapse
Affiliation(s)
- Manoj Thakur
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India.
| | - K Muniyappa
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
29
|
Alves-Soares R, de Oliveira HD, Campos DDCO, Gomes-Vasconcelos YDA, Ferreira-da-Silva FW, Silva-Alves KS, Coelho-de-Souza LN, Diniz LRL, Leal-Cardoso JH, Coelho-de-Souza AN. The Essential Oil of Hyptis crenata Inhibits the Increase in Secretion of Inflammatory Mediators. PLANTS (BASEL, SWITZERLAND) 2022; 11:3048. [PMID: 36432777 PMCID: PMC9695298 DOI: 10.3390/plants11223048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/03/2022] [Accepted: 11/03/2022] [Indexed: 06/16/2023]
Abstract
Background: Hyptis crenata is a plant of great ethnopharmacological importance widely distributed in South American countries. In Northeast Brazil, teas or infusions of its aerial parts are used in folk medicine to treat several acute and chronic inflammatory diseases. In a previous work we have demonstrated that the essential oil of H. crenata (EOHc) has an antiedematogenic effect. The aim of this work was to evaluate the effect of EOHc on cytokines secretion and cellular infiltration. Methods: Peritonitis and paw edema models induced by carrageenan were used to determine leucocyte count, myeloperoxidase (MPO) activity, nitrite, and cytokines secretion. Results: EOHc (10−300 mg/kg) significantly inhibited leucocyte migration and reduced the neutrophil count (control: 1.46 × 103 ± 0.031 × 103/mL) of the total leucocytes population in extracellular exudate (control: 2.14 × 103 ± 0.149 × 103/mL) by 15.00%, 43.29%, 65.52%, and 72.83% for the doses of 10, 30, 100, and 300 mg/kg EOHc, respectively (EC50: 24.15 mg/kg). EOHc (100 mg/kg) inhibited the increase in myeloperoxidase activity and completely blocked the increase in nitrite concentration induced by carrageenan. EOHc markedly reduced the pro-inflammatory cytokines (IL-6, MCP-1, IFN-γ, TNF-α, and IL-12p70) and increased IL-10, an anti-inflammatory cytokine (compared to control group, p < 0.05). Conclusions: This study demonstrates that EOHc has a long-lasting anti-inflammatory effect mediated through interference on MPO activity, and nitrite, and cytokines secretion. This effect, coupled with low EOHc toxicity, as far as results obtained in mice could be translated to humans, suggests that EOHc has great potentiality as a therapeutic agent.
Collapse
Affiliation(s)
- Rutyleia Alves-Soares
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Hermógenes David de Oliveira
- Postgraduate Program in Biochemistry, Department of Biochemistry and Molecular Biology, Campus do Pici, Federal University of Ceara (UFC), Av. Humberto Monte, s/n Bloco 907, Fortaleza CEP 60440-990, Ceara, Brazil
| | - Dyély de Carvalho Oliveira Campos
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Yuri de Abreu Gomes-Vasconcelos
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Francisco Walber Ferreira-da-Silva
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Kerly Shamyra Silva-Alves
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Lianna Noronha Coelho-de-Souza
- Graduate Program of Medicine, Center for Health Sciences (CCS), Fortaleza University (UNIFOR), Av. Washington Soares 1321, Edson Queiroz, Fortaleza CEP 60811-905, Ceara, Brazil
| | - Lúcio Ricardo Leite Diniz
- Researcher at the National Institute of the Semiarid Region (INSA), Av. Francisco Lopes de Almeida, s/n Serrotão, Campina Grande CEP 58434-70, Paraiba, Brazil
| | - José Henrique Leal-Cardoso
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| | - Andrelina Noronha Coelho-de-Souza
- Postgraduate Program in Physiological Sciences, Superior Institute of Biomedical Sciences (ISCB), Campus do Itaperi, State University of Ceara (UECE), Av. Silas Munguba 1700, Fortaleza CEP 60.714.903, Ceara, Brazil
| |
Collapse
|
30
|
Winning S, Fandrey J. Oxygen Sensing in Innate Immune Cells: How Inflammation Broadens Classical Hypoxia-Inducible Factor Regulation in Myeloid Cells. Antioxid Redox Signal 2022; 37:956-971. [PMID: 35088604 DOI: 10.1089/ars.2022.0004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Significance: Oxygen deprivation (hypoxia) is a common feature at sites of inflammation. Immune cells and all other cells present at the inflamed site have to adapt to these conditions. They do so by stabilization and activation of hypoxia-inducible factor subunit α (HIF-1α and HIF-2α, respectively), enabling constant generation of adenosine triphosphate (ATP) under these austere conditions by the induction of, for example, glycolytic pathways. Recent Advances: During recent years, it has become evident that HIFs play a far more important role than initially believed because they shape the inflammatory phenotype of immune cells. They are indispensable for migration, phagocytosis, and the induction of inflammatory cytokines by innate immune cells and thereby enable a crosstalk between innate and adaptive immunity. In short, they ensure the survival and function of immune cells under critical conditions. Critical Issues: Up to now, there are still open questions regarding the individual roles of HIF-1 and HIF-2 for the different cell types. In particular, the loss of both HIF-1 and HIF-2 in myeloid cells led to unexpected and contradictory results in the mouse models analyzed so far. Similarly, the role of HIF-1 in dendritic cell maturation is unclear due to inconsistent results from in vitro experiments. Future Directions: The HIFs are indispensable for immune cell survival and action under inflammatory conditions, but they might also trigger over-activation of immune cells. Therefore, they might be excellent setscrews to adjust the inflammatory response by pharmaceuticals. China and Japan and very recently (August 2021) Europe have approved prolyl hydroxylase inhibitors (PHIs) to stabilize HIF such as roxadustat for clinical use to treat anemia by increasing the production of erythropoietin, the classical HIF target gene. Nonetheless, we need further work regarding the use of PHIs under inflammatory conditions, because HIFs show specific activation and distinct expression patterns in innate immune cells. The extent to which HIF-1 or HIF-2 as a transcription factor regulates the adaptation of immune cells to inflammatory hypoxia differs not only by the cell type but also with the inflammatory challenge and the surrounding tissue. Therefore, we urgently need isoform- and cell type-specific modulators of the HIF pathway. Antioxid. Redox Signal. 37, 956-971.
Collapse
Affiliation(s)
- Sandra Winning
- Institut für Physiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| | - Joachim Fandrey
- Institut für Physiologie, Universitätsklinikum Essen, Universität Duisburg-Essen, Essen, Germany
| |
Collapse
|
31
|
Sphingosine-1-Phosphate Alleviates Irradiation Induced Salivary Gland Hypofunction through Preserving Endothelial Cells and Resident Macrophages. Antioxidants (Basel) 2022; 11:antiox11102050. [PMID: 36290773 PMCID: PMC9598384 DOI: 10.3390/antiox11102050] [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: 07/31/2022] [Revised: 10/08/2022] [Accepted: 10/13/2022] [Indexed: 11/25/2022] Open
Abstract
Radiotherapy for head-and-neck cancers frequently causes long-term hypofunction of salivary glands that severely compromises quality of life and is difficult to treat. Here, we studied effects and mechanisms of Sphingosine-1-phosphate (S1P), a versatile signaling sphingolipid, in preventing irreversible dry mouth caused by radiotherapy. Mouse submandibular glands (SMGs) were irradiated with or without intra-SMG S1P pretreatment. The saliva flow rate was measured following pilocarpine stimulation. The expression of genes related to S1P signaling and radiation damage was examined by flow cytometry, immunohistochemistry, quantitative RT-PCR, Western blotting, and/or single-cell RNA-sequencing. S1P pretreatment ameliorated irradiation-induced salivary dysfunction in mice through a decrease in irradiation-induced oxidative stress and consequent apoptosis and cellular senescence, which is related to the enhancement of Nrf2-regulated anti-oxidative response. In mouse SMGs, endothelial cells and resident macrophages are the major cells capable of producing S1P and expressing the pro-regenerative S1P receptor S1pr1. Both mouse SMGs and human endothelial cells are protected from irradiation damage by S1P pretreatment, likely through the S1pr1/Akt/eNOS axis. Moreover, intra-SMG-injected S1P did not affect the growth and radiosensitivity of head-and-neck cancer in a mouse model. These data indicate that S1P signaling pathway is a promising target for alleviating irradiation-induced salivary gland hypofunction.
Collapse
|
32
|
Campo-Sabariz J, García-Vara A, Moral-Anter D, Briens M, Hachemi MA, Pinloche E, Ferrer R, Martín-Venegas R. Hydroxy-Selenomethionine, an Organic Selenium Source, Increases Selenoprotein Expression and Positively Modulates the Inflammatory Response of LPS-Stimulated Macrophages. Antioxidants (Basel) 2022; 11:antiox11101876. [PMID: 36290599 PMCID: PMC9598155 DOI: 10.3390/antiox11101876] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/17/2022] [Accepted: 09/17/2022] [Indexed: 11/16/2022] Open
Abstract
The role of 2-hydroxy-(4-methylseleno)butanoic acid (OH-SeMet), a form of organic selenium (Se), in selenoprotein synthesis and inflammatory response of THP1-derived macrophages stimulated with lipopolysaccharide (LPS) has been investigated. Glutathione peroxidase (GPX) activity, GPX1 gene expression, selenoprotein P (SELENOP) protein and gene expression, and reactive oxygen species (ROS) production were studied in Se-deprived conditions (6 and 24 h). Then, macrophages were supplemented with OH-SeMet for 72 h and GPX1 and SELENOP gene expression were determined. The protective effect of OH-SeMet against oxidative stress was studied in H2O2-stimulated macrophages, as well as the effect on GPX1 gene expression, oxidative stress, cytokine production (TNFα, IL-1β and IL-10), and phagocytic and killing capacities after LPS stimulation. Se deprivation induced a reduction in GPX activity, GPX1 gene expression, and SELENOP protein and gene expression at 24 h. OH-SeMet upregulated GPX1 and SELENOP gene expression and decreased ROS production after H2O2 treatment. In LPS-stimulated macrophages, OH-SeMet upregulated GPX1 gene expression, enhanced phagocytic and killing capacities, and reduced ROS and cytokine production. Therefore, OH-SeMet supplementation supports selenoprotein expression and controls oxidative burst and cytokine production while enhancing phagocytic and killing capacities, modulating the inflammatory response, and avoiding the potentially toxic insult produced by highly activated macrophages.
Collapse
Affiliation(s)
- Joan Campo-Sabariz
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), 08028 Barcelona, Spain
| | - Adriana García-Vara
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), 08028 Barcelona, Spain
| | - David Moral-Anter
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), 08028 Barcelona, Spain
| | | | | | | | - Ruth Ferrer
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), 08028 Barcelona, Spain
| | - Raquel Martín-Venegas
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l’Alimentació, Universitat de Barcelona (UB), Institut de Recerca en Nutrició i Seguretat Alimentària (INSA-UB), 08028 Barcelona, Spain
- Correspondence:
| |
Collapse
|
33
|
Effect of Nrf2 on Phenotype Changes of Macrophages in the Anterior Vaginal Wall of Women With Pelvic Organ Prolapse. Female Pelvic Med Reconstr Surg 2022; 28:616-623. [PMID: 35703292 DOI: 10.1097/spv.0000000000001212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
OBJECTIVE The aim of this study was to observe the effect of nuclear factor-erythroid 2-related factor 2 (Nrf2) on the phenotype changes of macrophages in the anterior vaginal wall of patients with pelvic organ prolapse (POP). METHODS The tissues of the anterior vaginal wall of the control group (n = 30) and POP groups (n = 60) were collected during operation. The expressions of Nrf2, iNOS (representative factor of M1 macrophages), and CD206 (representative factor of M2 macrophages) were determined by immunohistochemical staining and Western blot. Morphological changes and collagen distribution of the anterior vaginal wall were observed by hematoxylin-eosin staining and Masson trichrome staining. RESULTS Compared with the control group, the expression levels of Nrf2 and CD206 protein in the anterior vaginal wall tissues of the POP groups were significantly decreased ( P < 0.05), and were negatively proportional to the degree of prolapse ( P < 0.05). The expression of iNOS was significantly increased and was directly proportional to the degree of prolapse ( P < 0.05). Hematoxylin-eosin staining and Masson trichrome staining showed that the collagen fibers are more sparsely arranged and disordered in the POP group than the control. CONCLUSIONS In patients with POP, the expression of antioxidant factor Nrf2 is reduced in the vaginal anterior wall tissues and the antioxidant capacity is weakened, leading to the blocked polarization of macrophages and the accumulation of a large number of M1 macrophages in the tissue, affecting the occurrence and development of POP.
Collapse
|
34
|
Basova LV, Vien W, Bortell N, Najera JA, Marcondes MCG. Methamphetamine signals transcription of IL1β and TNFα in a reactive oxygen species-dependent manner and interacts with HIV-1 Tat to decrease antioxidant defense mechanisms. Front Cell Neurosci 2022; 16:911060. [PMID: 36060276 PMCID: PMC9434488 DOI: 10.3389/fncel.2022.911060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Methamphetamine (Meth) abuse is a common HIV co-morbidity that is linked to aggravated Central Nervous System (CNS) inflammation, which accentuates HIV- associated neurological disorders, triggered both directly or indirectly by the drug. We used the well-established human innate immune macrophage cell line system (THP1) to demonstrate that Reactive Oxygen Species (ROS) immediately induced by Meth play a role in the increased transcription of inflammatory genes, in interaction with HIV-1 Tat peptide. Meth and Tat, alone and together, affect early events of transcriptional activity, as indicated by changes in RNA polymerase (RNAPol) recruitment patterns throughout the genome, via ROS-dependent and -independent mechanisms. IL1β (IL1β) and TNF α (TNFα), two genes with defining roles in the inflammatory response, were both activated in a ROS-dependent manner. We found that this effect occurred via the activation of the activator protein 1 (AP-1) comprising cFOS and cJUN transcription factors and regulated by the SRC kinase. HIV-1 Tat, which was also able to induce the production of ROS, did not further impact the effects of ROS in the context of Meth, but promoted gene activity independently from ROS, via additional transcription factors. For instance, HIV-1 Tat increased NFkB activation and activated gene clusters regulated by Tata box binding peptide, ING4 and IRF2. Importantly, HIV-1 Tat decreased the expression of anti-oxidant genes, where its suppression of the detoxifying machinery may contribute to the aggravation of oxidative stress induced by ROS in the context of Meth. Our results provide evidence of effects of Meth via ROS and interactions with HIV Tat that promote the transcription of inflammatory genes such as IL1β and TNFα.
Collapse
Affiliation(s)
- Liana V. Basova
- San Diego Biomedical Research Institute, San Diego, CA, United States
- The Scripps Research Institute, La Jolla, CA, United States
| | - Whitney Vien
- The Scripps Research Institute, La Jolla, CA, United States
- University of California San Diego, La Jolla, CA, United States
| | - Nikki Bortell
- The Scripps Research Institute, La Jolla, CA, United States
| | | | - Maria Cecilia Garibaldi Marcondes
- San Diego Biomedical Research Institute, San Diego, CA, United States
- The Scripps Research Institute, La Jolla, CA, United States
- *Correspondence: Maria Cecilia Garibaldi Marcondes,
| |
Collapse
|
35
|
Intrinsic ROS Drive Hair Follicle Cycle Progression by Modulating DNA Damage and Repair and Subsequently Hair Follicle Apoptosis and Macrophage Polarization. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8279269. [PMID: 35903712 PMCID: PMC9315455 DOI: 10.1155/2022/8279269] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 01/29/2022] [Accepted: 02/14/2022] [Indexed: 11/17/2022]
Abstract
Hair follicles (HFs) maintain homeostasis through the hair cycles; therefore, disrupting the hair cycle may lead to hair loss. Our previous study showed that apoptosis-inducing factor (AIF) nuclear translocation and poly [ADP-ribose] polymerase 1 (PARP1) upregulation induced apoptosis in mouse hair follicles during the hair cycle transition from anagen to catagen. However, the mechanism underlying this phenomenon remains unclear. In this study, we found that intrinsic ROS levels increased during the hair follicle cycle transition from anagen to catagen, followed by abrupt DNA breaks and activation of homologous recombinant and nonhomologous end joining DNA repair, along with the enhancement of apoptosis. Mice in different stages of the hair cycle were sacrificed, and the dorsal skins were collected. The results of western blot and histological staining indicated that AIF-PARP1 plays a key role in HF apoptosis, but their role in the regulation of the HF cycle is not clear. Mice were treated with inhibitors from anagen to catagen: treatment with BMN 673, a PARP1 inhibitor, increased DNA breaks and activated the cytochrome c/caspase-3-mediated apoptotic pathway, accelerating HF regression. Ac-DEVD-CHO (Ac), a caspase-3 inhibitor, attenuated HF degeneration by upregulating PARP1 expression, suggesting a seesaw relationship between cytochrome c-caspase-3- and AIF-PARP1-mediated apoptosis, wherein PARP1 may be the fulcrum. In addition, macrophages were involved in regulating the hair cycle, and the rate of M1 macrophages around HFs increased during catagen, while more M2 macrophages were found during anagen and telogen. Our results indicate that intrinsic ROS drive HF cycle progression through DNA damage and repair, followed by apoptosis. Intrinsic ROS drive hair follicle cycle progression by modulating DNA damage and repair, and consecutively, hair follicle apoptosis and macrophage polarization work together to promote the hair follicle cycle.
Collapse
|
36
|
Guo S, Song Z, Ji DK, Reina G, Fauny JD, Nishina Y, Ménard-Moyon C, Bianco A. Combined Photothermal and Photodynamic Therapy for Cancer Treatment Using a Multifunctional Graphene Oxide. Pharmaceutics 2022; 14:1365. [PMID: 35890259 PMCID: PMC9318106 DOI: 10.3390/pharmaceutics14071365] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 06/13/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
Graphene oxide (GO) is one of the most studied nanomaterials in many fields, including the biomedical field. Most of the nanomaterials developed for drug delivery and phototherapies are based on noncovalent approaches that lead to an unspecific release of physisorbed molecules in complex biological environments. Therefore, preparing covalently functionalized GO using straightforward and versatile methods is highly valuable. Phototherapies, including photothermal therapy (PTT) and photodynamic therapy (PDT), have shown great potential as effective therapeutic approaches against cancer. To overcome the limits of a single method, the combination of PTT and PDT can lead to a combined effect with a higher therapeutic efficiency. In this work, we prepare a folic acid (FA) and chlorin e6 (Ce6) double-functionalized GO for combined targeted PTT/PDT. This conjugate can penetrate rapidly into cancer cells and macrophages. A combined effect of PTT and PDT is observed, leading to a higher killing efficiency toward different types of cells involved in cancer and other diseases. Our work provides a simple protocol to prepare multifunctional platforms for the treatment of various diseases.
Collapse
Affiliation(s)
- Shi Guo
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, 67000 Strasbourg, France; (S.G.); (Z.S.); (D.-K.J.); (G.R.); (J.-D.F.); (C.M.-M.)
| | - Zhengmei Song
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, 67000 Strasbourg, France; (S.G.); (Z.S.); (D.-K.J.); (G.R.); (J.-D.F.); (C.M.-M.)
| | - Ding-Kun Ji
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, 67000 Strasbourg, France; (S.G.); (Z.S.); (D.-K.J.); (G.R.); (J.-D.F.); (C.M.-M.)
| | - Giacomo Reina
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, 67000 Strasbourg, France; (S.G.); (Z.S.); (D.-K.J.); (G.R.); (J.-D.F.); (C.M.-M.)
| | - Jean-Daniel Fauny
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, 67000 Strasbourg, France; (S.G.); (Z.S.); (D.-K.J.); (G.R.); (J.-D.F.); (C.M.-M.)
| | - Yuta Nishina
- Graduate School of Natural Science and Technology, Okayama University, Tsushimanaka, Kita-ku, Okayama 700-8530, Japan;
- Research Core for Interdisciplinary Sciences, Okayama University, Tsushimanaka, Kita-ku, Okayama 700-8530, Japan
| | - Cécilia Ménard-Moyon
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, 67000 Strasbourg, France; (S.G.); (Z.S.); (D.-K.J.); (G.R.); (J.-D.F.); (C.M.-M.)
| | - Alberto Bianco
- CNRS, Immunology, Immunopathology and Therapeutic Chemistry, UPR3572, University of Strasbourg, ISIS, 67000 Strasbourg, France; (S.G.); (Z.S.); (D.-K.J.); (G.R.); (J.-D.F.); (C.M.-M.)
| |
Collapse
|
37
|
Antioxidant Therapy in Cancer: Rationale and Progress. Antioxidants (Basel) 2022; 11:antiox11061128. [PMID: 35740025 PMCID: PMC9220137 DOI: 10.3390/antiox11061128] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 02/05/2023] Open
Abstract
Cancer is characterized by increased oxidative stress, an imbalance between reactive oxygen species (ROS) and antioxidants. Enhanced ROS accumulation, as a result of metabolic disturbances and signaling aberrations, can promote carcinogenesis and malignant progression by inducing gene mutations and activating pro-oncogenic signaling, providing a possible rationale for targeting oxidative stress in cancer treatment. While numerous antioxidants have demonstrated therapeutic potential, their clinical efficacy in cancer remains unproven. Here, we review the rationale for, and recent advances in, pre-clinical and clinical research on antioxidant therapy in cancer, including targeting ROS with nonenzymatic antioxidants, such as NRF2 activators, vitamins, N-acetylcysteine and GSH esters, or targeting ROS with enzymatic antioxidants, such as NOX inhibitors and SOD mimics. In addition, we will offer insights into prospective therapeutic options for improving the effectiveness of antioxidant therapy, which may expand its applications in clinical cancer treatment.
Collapse
|
38
|
Msheik Z, El Massry M, Rovini A, Billet F, Desmoulière A. The macrophage: a key player in the pathophysiology of peripheral neuropathies. J Neuroinflammation 2022; 19:97. [PMID: 35429971 PMCID: PMC9013246 DOI: 10.1186/s12974-022-02454-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/29/2022] [Indexed: 12/22/2022] Open
Abstract
Macrophages are present in all mammalian tissues and coexist with various cell types in order to respond to different environmental cues. However, the role of these cells has been underestimated in the context of peripheral nerve damage. More importantly, macrophages display divergent characteristics, associated with their origin, and in response to the modulatory effects of their microenvironment. Interestingly, the advent of new techniques such as fate mapping and single-cell transcriptomics and their synergistic use has helped characterize in detail the origin and fate of tissue-resident macrophages in the peripheral nervous system (PNS). Furthermore, these techniques have allowed a better understanding of their functions from simple homeostatic supervisors to chief regulators in peripheral neuropathies. In this review, we summarize the latest knowledge about macrophage ontogeny, function and tissue identity, with a particular focus on PNS-associated cells, as well as their interaction with reactive oxygen species under physiological and pathological conditions. We then revisit the process of Wallerian degeneration, describing the events accompanying axon degeneration, Schwann cell activation and most importantly, macrophage recruitment to the site of injury. Finally, we review these processes in light of internal and external insults to peripheral nerves leading to peripheral neuropathies, the involvement of macrophages and the potential benefit of the targeting of specific macrophages for the alleviation of functional defects in the PNS.
Collapse
|
39
|
The Effects of Vitamins and Micronutrients on Helicobacter pylori Pathogenicity, Survival, and Eradication: A Crosstalk between Micronutrients and Immune System. J Immunol Res 2022; 2022:4713684. [PMID: 35340586 PMCID: PMC8942682 DOI: 10.1155/2022/4713684] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 02/19/2022] [Accepted: 02/25/2022] [Indexed: 12/11/2022] Open
Abstract
Helicobacter pylori as a class I carcinogen is correlated with a variety of severe gastroduodenal diseases; therefore, H. pylori eradication has become a priority to prevent gastric carcinogenesis. However, due to the emergence and spread of multidrug and single drug resistance mechanisms in H. pylori, as well as serious side effects of currently used antibiotic interventions, achieving successful H. pylori eradication has become exceedingly difficult. Recent studies expressed the intention of seeking novel strategies to improve H. pylori management and reduce the risk of H. pylori-associated intestinal and extragastrointestinal disorders. For which, vitamin supplementation has been demonstrated in many studies to have a tight interaction with H. pylori infection, either directly through the regulation of the host inflammatory pathways or indirectly by promoting the host immune response. On the other hand, H. pylori infection is reported to result in micronutrient malabsorption or deficiency. Furthermore, serum levels of particular micronutrients, especially vitamin D, are inversely correlated to the risk of H. pylori infection and eradication failure. Accordingly, vitamin supplementation might increase the efficiency of H. pylori eradication and reduce the risk of drug-related adverse effects. Therefore, this review aims at highlighting the regulatory role of micronutrients in H. pylori-induced host immune response and their potential capacity, as intrinsic antioxidants, for reducing oxidative stress and inflammation. We also discuss the uncovered mechanisms underlying the molecular and serological interactions between micronutrients and H. pylori infection to present a perspective for innovative in vitro investigations, as well as novel clinical implications.
Collapse
|
40
|
Liu J, Zhu Z, Leung GKK. Erythrophagocytosis by Microglia/Macrophage in Intracerebral Hemorrhage: From Mechanisms to Translation. Front Cell Neurosci 2022; 16:818602. [PMID: 35237132 PMCID: PMC8882619 DOI: 10.3389/fncel.2022.818602] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/10/2022] [Indexed: 12/17/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating condition characterized by hematoma related mass effect. Microglia/macrophage (M φ) are rapidly recruited in order to remove the red blood cells through erythrophagocytosis. Efficient erythrophagocytosis can detoxify hemolytic products and facilitate neurological recovery after ICH. The underlying mechanisms include modulation of inflammatory response and oxidative stress, among others. It is a dynamic process mediated by a cascade of signal transduction, including “find-me” signals, “eat-me” signals and a set of phagocytotic receptors-ligand pairs that may be exploited as therapeutic targets. This review summarizes mechanistic signaling pathways of erythrophagocytosis and highlights the potential of harnessing M φ-mediated phagocytosis for ICH treatment.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
| | - Zhiyuan Zhu
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- Department of Functional Neurosurgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Guangzhou, China
- Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gilberto Ka-Kit Leung
- Department of Surgery, LKS Faculty of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, Hong Kong SAR, China
- *Correspondence: Gilberto Ka-Kit Leung,
| |
Collapse
|
41
|
Jofre BL, Eliçabe RJ, Silva JE, Pérez Sáez JM, Paez MD, Callegari E, Mariño KV, Di Genaro MS, Rabinovich GA, Davicino RC. Galectin-1 Cooperates with Yersinia Outer Protein (Yop) P to Thwart Protective Immunity by Repressing Nitric Oxide Production. Biomolecules 2021; 11:1636. [PMID: 34827634 PMCID: PMC8615707 DOI: 10.3390/biom11111636] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/26/2021] [Accepted: 10/29/2021] [Indexed: 11/18/2022] Open
Abstract
Yersinia enterocolitica (Ye) inserts outer proteins (Yops) into cytoplasm to infect host cells. However, in spite of considerable progress, the mechanisms implicated in this process, including the association of Yops with host proteins, remain unclear. Here, we evaluated the functional role of Galectin-1 (Gal1), an endogenous β-galactoside-binding protein, in modulating Yop interactions with host cells. Our results showed that Gal1 binds to Yops in a carbohydrate-dependent manner. Interestingly, Gal1 binding to Yops protects these virulence factors from trypsin digestion. Given that early control of Ye infection involves activation of macrophages, we evaluated the role of Gal1 and YopP in the modulation of macrophage function. Although Gal1 and YopP did not influence production of superoxide anion and/or TNF by Ye-infected macrophages, they coordinately inhibited nitric oxide (NO) production. Notably, recombinant Gal1 (rGal1) did not rescue NO increase observed in Lgals1-/- macrophages infected with the YopP mutant Ye ∆yopP. Whereas NO induced apoptosis in macrophages, no significant differences in cell death were detected between Gal1-deficient macrophages infected with Ye ∆yopP, and WT macrophages infected with Ye wt. Strikingly, increased NO production was found in WT macrophages treated with MAPK inhibitors and infected with Ye wt. Finally, rGal1 administration did not reverse the protective effect in Peyer Patches (PPs) of Lgals1-/- mice infected with Ye ∆yopP. Our study reveals a cooperative role of YopP and endogenous Gal1 during Ye infection.
Collapse
Affiliation(s)
- Brenda Lucila Jofre
- División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis CP5700, Argentina; (B.L.J.); (R.J.E.); (J.E.S.); (M.S.D.G.)
- Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis C5700, Argentina
| | - Ricardo Javier Eliçabe
- División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis CP5700, Argentina; (B.L.J.); (R.J.E.); (J.E.S.); (M.S.D.G.)
- Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis C5700, Argentina
| | - Juan Eduardo Silva
- División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis CP5700, Argentina; (B.L.J.); (R.J.E.); (J.E.S.); (M.S.D.G.)
- Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis C5700, Argentina
| | - Juan Manuel Pérez Sáez
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (IBYME-CONICET), Buenos Aires C1428ADN, Argentina; (J.M.P.S.); (G.A.R.)
| | - Maria Daniela Paez
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 66544, USA; (M.D.P.); (E.C.)
| | - Eduardo Callegari
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South Dakota, Vermillion, SD 66544, USA; (M.D.P.); (E.C.)
| | - Karina Valeria Mariño
- Laboratorio de Glicómica Funcional y Molecular, Instituto de Biología y Medicina Experimental, Consejo Nacional de Investigaciones Científicas y Técnicas (IBYME-CONICET), Buenos Aires C1428ADN, Argentina;
| | - María Silvia Di Genaro
- División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis CP5700, Argentina; (B.L.J.); (R.J.E.); (J.E.S.); (M.S.D.G.)
- Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis C5700, Argentina
| | - Gabriel Adrián Rabinovich
- Laboratorio de Glicomedicina, Instituto de Biología y Medicina Experimental (IBYME), Consejo Nacional de Investigaciones Científicas y Técnicas (IBYME-CONICET), Buenos Aires C1428ADN, Argentina; (J.M.P.S.); (G.A.R.)
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1428, Argentina
| | - Roberto Carlos Davicino
- División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, San Luis CP5700, Argentina; (B.L.J.); (R.J.E.); (J.E.S.); (M.S.D.G.)
- Instituto Multidisciplinario de Investigaciones Biológicas (IMIBIO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), San Luis C5700, Argentina
- Roberto Davicino, División de Inmunología, Facultad de Química, Bioquímica y Farmacia, Universidad Nacional de San Luis, Ejercito de los Andes 950, San Luis CP5700, Argentina
| |
Collapse
|
42
|
Povo-Retana A, Mojena M, Boscá A, Pedrós J, Peraza DA, Valenzuela C, Laparra JM, Calle F, Boscá L. Graphene Particles Interfere with Pro-Inflammatory Polarization of Human Macrophages: Functional and Electrophysiological Evidence. Adv Biol (Weinh) 2021; 5:e2100882. [PMID: 34590442 DOI: 10.1002/adbi.202100882] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/09/2021] [Indexed: 02/05/2023]
Abstract
The interaction of two types of fragmented graphene particles (30-160 nm) with human macrophages is studied. Since macrophages have significant phagocytic activity, the incorporation of graphene particles into cells has an effect on the response to functional polarization stimuli, favoring an anti-inflammatory profile. Incubation of macrophages with graphene foam particles, prepared by chemical vapor deposition, and commercially available graphene nanoplatelet particles does not affect cell viability when added at concentrations up to 100 µg mL-1 ; macrophages exhibit differential quantitative responses to each type of graphene particles. Although both materials elicit similar increases in the release of reactive oxygen species, the impact on the transcriptional regulation associated with the polarization profile is different; graphene nanoplatelets significantly modify this transcriptomic profile. Moreover, these graphene particles differentially affect the motility and phagocytosis of macrophages. After the incorporation of both graphene types into the macrophages, they exhibit specific responses in terms of the mitochondrial oxygen consumption and electrophysiological potassium currents at the cell plasma membrane. These data support the view that the physical structure of the graphene particles has an impact on human macrophage responses, paving the way for the development of new mechanisms to modulate the activity of the immune system.
Collapse
Affiliation(s)
- Adrián Povo-Retana
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
| | - Marina Mojena
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
| | - Alberto Boscá
- Instituto de Sistemas Optoelectrónicos y Microtecnología (ISOM) and Departamento de Ingeniería Electrónica, E.T.S.I. de Telecomunicación, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - Jorge Pedrós
- Instituto de Sistemas Optoelectrónicos y Microtecnología (ISOM) and Departamento de Ingeniería Electrónica, E.T.S.I. de Telecomunicación, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - Diego Alberto Peraza
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
| | - Carmen Valenzuela
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares, CIBERCV, Melchor Fernández Almagro, Madrid, 28029, Spain
| | - José Moisés Laparra
- J. M. Laparra, Madrid Institute for Advanced studies in Food (IMDEA Food), Ctra. Cantoblanco 8, Madrid, 28049, Spain
| | - Fernando Calle
- Instituto de Sistemas Optoelectrónicos y Microtecnología (ISOM) and Departamento de Ingeniería Electrónica, E.T.S.I. de Telecomunicación, Universidad Politécnica de Madrid, Madrid, 28040, Spain
| | - Lisardo Boscá
- Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), Arturo Duperier 4, Madrid, 28029, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares, CIBERCV, Melchor Fernández Almagro, Madrid, 28029, Spain
| |
Collapse
|
43
|
Jaccard A, Li X, Ho PC. Glutamine gluttony of efferocytes. Nat Metab 2021; 3:1280-1281. [PMID: 34650274 DOI: 10.1038/s42255-021-00462-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Alison Jaccard
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| | - Xiaoyun Li
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Ping-Chih Ho
- Department of Fundamental Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
44
|
Han B, García‐Mendoza D, van den Berg H, van den Brink NW. Modulatory Effects of Mercury (II) Chloride (HgCl 2 ) on Chicken Macrophage and B-Lymphocyte Cell Lines with Viral-Like Challenges In Vitro. ENVIRONMENTAL TOXICOLOGY AND CHEMISTRY 2021; 40:2813-2824. [PMID: 34288095 PMCID: PMC9291928 DOI: 10.1002/etc.5169] [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] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 06/28/2021] [Accepted: 07/14/2021] [Indexed: 06/13/2023]
Abstract
Mercury (Hg) is a toxic trace metal ubiquitously distributed in the environment. Inorganic mercury (as HgCl2 ) can cause immunotoxicity in birds, but the mechanisms of action are still not fully resolved, especially with respect to responses to viral infections. To investigate the potential immunomodulatory effects of Hg2+ on specific cell types of the avian immune system, chicken macrophage (HD-11) and B-lymphocyte (DT40) cell lines were applied as in vitro models for the innate and adaptive immune systems, respectively. The cells were stimulated with synthetic double-stranded RNA, which can be recognized by toll-like receptor-3 to mimic a viral infection. The Hg2+ showed concentration-dependent cytotoxicity in both cell lines, with similar median effect concentrations at 30 µM. The cytotoxicity of Hg2+ was closely related to glutathione (GSH) depletion and reactive oxygen species induction, whereas the de novo synthesis of GSH acted as a primary protective strategy. Nitric oxide produced by activated macrophages was strongly inhibited by Hg2+ , and was also influenced by cellular GSH levels. Cell proliferation, gene expression of microRNA-155, and cellular IgM levels in B cells were decreased at noncytotoxic Hg2+ concentrations. The secretion of antiviral interferon-α was induced by Hg2+ in both cell lines. Overall, our results suggest that Hg2+ exposure can cause immunomodulatory effects in birds by disrupting immune cell proliferation and cytokine production, and might result in disorders of the avian immune system. Environ Toxicol Chem 2021;40:2813-2824. © 2021 The Authors. Environmental Toxicology and Chemistry published by Wiley Periodicals LLC on behalf of SETAC.
Collapse
Affiliation(s)
- Biyao Han
- Division of Toxicology, Wageningen University and ResearchWageningenThe Netherlands
| | - Diego García‐Mendoza
- Division of Toxicology, Wageningen University and ResearchWageningenThe Netherlands
| | - Hans van den Berg
- Division of Toxicology, Wageningen University and ResearchWageningenThe Netherlands
| | | |
Collapse
|
45
|
Li L, Zhang Q, Zhang X, Xu X, Wang X, Huang X, Wang T, Jiang Z, Xiao L, Zhang L, Sun L. Protective effects of Nrf2 against sepsis-induced hepatic injury. Life Sci 2021; 282:119807. [PMID: 34245771 DOI: 10.1016/j.lfs.2021.119807] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 06/09/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023]
Abstract
AIM This study was designed to investigate the changes of liver injury and Nrf2 signaling pathway in the process of sepsis. We also aimed to examine the role of Nrf2 in resisting oxidative stress and relieving inflammation in sepsis-induced hepatic injury. MAIN METHODS By operating cecal ligation and puncture (CLP) on Nrf2-/- mice and wild type mice, a sepsis-induced hepatic injury model was established. We compared and contrasted the wild type mice with the Nrf2-/- mice during sepsis-induced hepatic injury, and evaluated the liver damage by biochemical analyses and staining hematoxylin-eosin (HE). Western blot or real-time PCR was performed to detect Nrf2 and its regulated genes NQO-1, GCLM and HO-1. Additionally, we detected the expressions and secretion of pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), Interleukin-6 (IL-6), IL-1β and anti-inflammatory cytokines IL-10. We assessed the oxidative stress through the levels of MDA and NO. KEY FINDINGS The results showed that Nrf2 expressions at mRNA and protein levels were increased 1 day after CLP, namely the early stage of sepsis. Compared with wild type mice after CLP, Nrf2-/- mice showed more severe liver injury, accompanied by higher expression of inflammatory cytokines and oxidative stress. Notably, Nrf2-regulated genes GCLM and NQO-1, were strongly downregulated in Nrf2-/- mice. SIGNIFICANCE Nrf2 was probably implicated in decreasing inflammatory cytokine levels and counteracting oxidative stress to alleviate sepsis-induced hepatic injury, mainly through regulating GCLM and NQO-1 in the early stage after CLP.
Collapse
Affiliation(s)
- Liping Li
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Qianwen Zhang
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xi Zhang
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoting Xu
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xinzhi Wang
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Xin Huang
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Tao Wang
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Zhenzhou Jiang
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China
| | - Li Xiao
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou 313000, China
| | - Luyong Zhang
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lixin Sun
- Jiangsu Center Pharmacodynamic Research and Evaluation, China Pharmaceutical University, Nanjing 210009, China; Laboratory of Drug Quality Control and Pharmacovigilance (China Pharmaceutical University), Ministry of Education, Nanjing 210009, China.
| |
Collapse
|
46
|
Wang X, Nie Y, Si B, Wang T, Hei TK, Du H, Zhao G, Chen S, Xu A, Liu Y. Silver nanoparticles protect against arsenic induced genotoxicity via attenuating arsenic bioaccumulation and elevating antioxidation in mammalian cells. JOURNAL OF HAZARDOUS MATERIALS 2021; 413:125287. [PMID: 33930940 DOI: 10.1016/j.jhazmat.2021.125287] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/05/2021] [Accepted: 01/27/2021] [Indexed: 06/12/2023]
Abstract
Arsenic (As) and its compounds have been classified as Group I carcinogenic agents by the International Agency for Research on Cancer (IARC); however, there is few specific and efficient antidotes used for As detoxification. The present study aimed to investigate the protective effects of silver nanoparticles (AgNPs) at non-toxic concentrations on As(Ⅲ) induced genotoxicity and the underlying mechanism. Our data showed that AgNPs pretreatment significantly inhibited the generation of phosphorylated histone H2AX (γ-H2AX, marker of nuclear DNA double strand breaks) and the mutation frequencies induced by As(Ⅲ) exposure. Atomic fluorescence spectrometer (AFS) and laser ablation-inductively coupled plasma-mass spectrometry (LA-ICP-MS) analysis revealed that the intracellular accumulation of As(Ⅲ) in human-hamster hybrid AL cells was declined by AgNPs via suppressing the expression of specific As(Ⅲ)-binding protein (Gal-1). Moreover, the activities of antioxidant enzymes were greatly up-regulated by AgNPs, which eventually inhibited the generation of reactive oxygen species (ROS) induced by As(Ⅲ) and the downstream stress-activated protein kinases/c-Jun amino-terminal kinases (SAPK/JNK) signaling pathway. These results provided clear evidence that AgNPs dramatically suppressed the genotoxic response of As(Ⅲ) in mammalian cells via decreasing As(Ⅲ) bioaccumulation and elevating intracellular antioxidation, which might provide a new clue for AgNPs applications in As(Ⅲ) detoxification.
Collapse
Affiliation(s)
- Xue Wang
- School of Environmental Science and Optoelectronic Technology, University of Science and Technology of China, Hefei, Anhui 230026, PR China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China
| | - Yaguang Nie
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, PR China
| | - Bo Si
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China
| | - Tong Wang
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China
| | - Tom K Hei
- Center for Radiological Research, Department of Radiation Oncology, College of Physicians and Surgeons, Columbia University, New York, NY 10032, United States
| | - Hua Du
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China
| | - Guoping Zhao
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China
| | - Shaopeng Chen
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China
| | - An Xu
- School of Environmental Science and Optoelectronic Technology, University of Science and Technology of China, Hefei, Anhui 230026, PR China; Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China; Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, PR China.
| | - Yun Liu
- Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Chinese Academy of Sciences; Anhui Province Key Laboratory of Environmental Toxicology and Pollution Control Technology, High Magnetic Field Laboratory, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, Anhui 230031, PR China.
| |
Collapse
|
47
|
Fransen LFH, Leonard MO. CD34+ derived macrophage and dendritic cells display differential responses to paraquat. Toxicol In Vitro 2021; 75:105198. [PMID: 34097952 PMCID: PMC8444090 DOI: 10.1016/j.tiv.2021.105198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/28/2021] [Accepted: 06/02/2021] [Indexed: 01/23/2023]
Abstract
Paraquat (PQ) is a redox cycling herbicide known for its acute toxicity in humans. Airway parenchymal cells have been identified as primary sites for PQ accumulation, tissue inflammation and cellular injury. However, the role of immune cells in PQ induced tissue injury is largely unknown. To explore this further, primary cultures of human CD34+ stem cell derived macrophages (MCcd34) and dendritic cells (DCcd34) were established and characterised using RNA-Seq profiling. The impact of PQ on DCcd34 and MCcd34 cytotoxicity revealed increased effect within DCcd34 cultures. PQ toxicity mechanisms were examined using sub-cytotoxic concentrations and TempO-seq transcriptomic assays. Comparable increases for several stress response pathway (NFE2L2, NF-kB and HSF) dependent genes were observed across both cell types. Interestingly, PQ induced unfolded protein response (UPR), p53, Irf and DC maturation genes in DCcd34 but not in MCcd34. Further exploration of the immune modifying potential of PQ was performed using the common allergen house dust mite (HD). Co-treatment of PQ and HD resulted in enhanced inflammatory responses within MCcd34 but not DCcd34. These results demonstrate immune cell type differential responses to PQ, that may underlie aspects of acute toxicity and susceptibility to inflammatory disease. Paraquat induces inflammatory and oxidative events in immune cells. Paraquat prompts selective induction of several pathways in dendritic cells. Paraquat and dust mite co-exposure enhances inflammatory response in macrophages. These results provide insight into paraquat mechanisms of toxicity.
Collapse
Affiliation(s)
- Leonie F H Fransen
- Toxicology Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Harwell Campus, OX11 0RQ, UK.
| | - Martin O Leonard
- Toxicology Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Harwell Campus, OX11 0RQ, UK.
| |
Collapse
|
48
|
Luque-Campos N, Bustamante-Barrientos FA, Pradenas C, García C, Araya MJ, Bohaud C, Contreras-López R, Elizondo-Vega R, Djouad F, Luz-Crawford P, Vega-Letter AM. The Macrophage Response Is Driven by Mesenchymal Stem Cell-Mediated Metabolic Reprogramming. Front Immunol 2021; 12:624746. [PMID: 34149687 PMCID: PMC8213396 DOI: 10.3389/fimmu.2021.624746] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 05/13/2021] [Indexed: 12/17/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent adult stromal cells widely studied for their regenerative and immunomodulatory properties. They are capable of modulating macrophage plasticity depending on various microenvironmental signals. Current studies have shown that metabolic changes can also affect macrophage fate and function. Indeed, changes in the environment prompt phenotype change. Therefore, in this review, we will discuss how MSCs orchestrate macrophage’s metabolic plasticity and the impact on their function. An improved understanding of the crosstalk between macrophages and MSCs will improve our knowledge of MSC’s therapeutic potential in the context of inflammatory diseases, cancer, and tissue repair processes in which macrophages are pivotal.
Collapse
Affiliation(s)
- Noymar Luque-Campos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Programa de Doctorado en Biomedicina, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| | - Felipe A Bustamante-Barrientos
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Carolina Pradenas
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Cynthia García
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - María Jesús Araya
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Escuela de Biotecnología, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | | | | | - Roberto Elizondo-Vega
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | | | - Patricia Luz-Crawford
- Laboratorio de Inmunología Celular y Molecular, Facultad de Medicina, Universidad de los Andes, Santiago, Chile.,Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile
| | - Ana María Vega-Letter
- Centro de Investigación e Innovación Biomédica, Universidad de los Andes, Santiago, Chile.,Cells for Cells, Regenero, Las Condes, Santiago, Chile.,Laboratory of Nano-Regenerative Medicine, Facultad de Medicina, Universidad de los Andes, Santiago, Chile
| |
Collapse
|
49
|
Zhang W, Cheng C, Sha Z, Chen C, Yu C, Lv N, Ji P, Wu X, Ma T, Cheng H, Shi L. Rosmarinic acid prevents refractory bacterial pneumonia through regulating Keap1/Nrf2-mediated autophagic pathway and mitochondrial oxidative stress. Free Radic Biol Med 2021; 168:247-257. [PMID: 33812997 DOI: 10.1016/j.freeradbiomed.2021.03.038] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/18/2021] [Accepted: 03/28/2021] [Indexed: 12/30/2022]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) is the leading cause of bacterial pneumonia, featured with exuberant inflammatory cytokine production, extensive oxidative stress and tissue injury. The Keap1/Nrf2 system is the major apparatus essential for host defense against oxidative and electrophilic stresses of both exogenous and endogenous origins, representing a logical target for host-directed strategy to treat severe inflammatory diseases including MRSA-induced pneumonia. In an effort to search therapeutics for bacterial pneumonia, we identify rosmarinic acid (RA) as a covalent modifier of Keap1 and hence an activator of Nrf2. Specifically, RA forms a covalent bond with the cysteine 151 of Keap1 in BTB domain, and blocks its association with Nrf2 for proteasome-mediated degradation. Consequently, RA treatment caused the increased Nrf2 nuclear translocation to initiate antioxidant and mitochondrial biogenic programs, as well as macrophage bactericidal activity through inducing autophagic pathway, which eventually led to expedited bacterial eradication, inflammation resolution, and disease recovery. Collectively, our findings establish RA as a specific inducer of Nrf2 and show its potential to prevent MRSA pneumonia.
Collapse
Affiliation(s)
- Wei Zhang
- School of Medicine, Nanjing University of Chinese Medicine, 210046, Nanjing, China
| | - Cheng Cheng
- The First School of Clinical Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, 210029, Nanjing, China
| | - Zhou Sha
- School of Medicine, Nanjing University of Chinese Medicine, 210046, Nanjing, China
| | - Changmai Chen
- School of Pharmacy, Fujian Medical University, Fuzhou, 350122, China; State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 100191, Beijing, China
| | - Chengtao Yu
- Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, 210023, Nanjing, China
| | - Nianyin Lv
- School of Medicine, Nanjing University of Chinese Medicine, 210046, Nanjing, China
| | - Peng Ji
- School of Medicine, Nanjing University of Chinese Medicine, 210046, Nanjing, China
| | - Xiaohui Wu
- School of Medicine, Nanjing University of Chinese Medicine, 210046, Nanjing, China
| | - Tonghui Ma
- School of Medicine, Nanjing University of Chinese Medicine, 210046, Nanjing, China
| | - Haibo Cheng
- The First School of Clinical Medicine, The Affiliated Hospital of Nanjing University of Chinese Medicine, 210029, Nanjing, China; Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, 210023, Nanjing, China.
| | - Liyun Shi
- School of Medicine, Nanjing University of Chinese Medicine, 210046, Nanjing, China; Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, 210023, Nanjing, China; International Medical College, Zhejiang Shuren University, 310022, Hangzhou, China.
| |
Collapse
|
50
|
Differential Effect of Non-Thermal Plasma RONS on Two Human Leukemic Cell Populations. Cancers (Basel) 2021; 13:cancers13102437. [PMID: 34069922 PMCID: PMC8157554 DOI: 10.3390/cancers13102437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary As the number of investigations into the use of non-thermal plasma (NTP) for cancer treatment expands, it is becoming apparent that susceptibility of different cancer cells to NTP varies. We hypothesized that such differences could be attributed to the cell type-dependent interactions between NTP-generated reactive oxygen and nitrogen species (RONS) and the target cells. To test this hypothesis, we examined how two different human leukemic cell lines—Jurkat T lymphocytes and THP-1 monocytes—influence hydrogen peroxide and nitrite content in media after NTP exposure. We also assessed the potential of NTP to enhance immunogenicity in these cells and assayed phagocytosis of NTP-exposed leukemic cells by macrophages. Our results highlight the significance of target-mediated modulation of plasma chemical species in the development and clinical use of protocols involving plasma sources for use in cancer therapeutic application. Abstract Non-thermal plasma application to cancer cells is known to induce oxidative stress, cytotoxicity and indirect immunostimulatory effects on antigen presenting cells (APCs). The purpose of this study was to evaluate the responses of two leukemic cell lines—Jurkat T lymphocytes and THP-1 monocytes—to NTP-generated reactive oxygen and nitrogen species (RONS). Both cell types depleted hydrogen peroxide, but THP-1 cells neutralized it almost immediately. Jurkat cells transiently blunted the frequency-dependent increase in nitrite concentrations in contrast to THP-1 cells, which exhibited no immediate effect. A direct relationship between frequency-dependent cytotoxicity and mitochondrial superoxide was observed only in Jurkat cells. Jurkat cells were very responsive to NTP in their display of calreticulin and heat shock proteins 70 and 90. In contrast, THP-1 cells were minimally responsive or unresponsive. Despite no NTP-dependent decrease in cell surface display of CD47 in either cell line, both cell types induced migration of and phagocytosis by APCs. Our results demonstrate that cells modulate the RONS-mediated changes in liquid chemistry, and, importantly, the resultant immunomodulatory effects of NTP can be independent of NTP-induced cytotoxicity.
Collapse
|