1
|
Chakraborty S, Haider S, Mukherjee G, Chakrabarty A, Chowdhury G. O6-Alkylguanine-DNA Alkyltransferase Maintains Genome Integrity by Forming DNA-Protein Cross-Links during Inflammation-Associated Peroxynitrite-Mediated DNA Damage. Chem Res Toxicol 2024; 37:1952-1964. [PMID: 39431584 DOI: 10.1021/acs.chemrestox.4c00296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Inflammation is an early immune response against invading pathogens and damaged tissue. Although beneficial, uncontrolled inflammation leads to various diseases including cancer in a chronic setting. Peroxynitrite (PN) is a major reactive nitrogen species generated during inflammation. It produces various DNA lesions including 8-nitro-guanine which spontaneously converts into abasic sites, resulting in DNA strand breakage, and is suspected to be mutagenic. Here, we report the discovery of a previously unrecognized function of the human repair protein O6-alkylguanine-DNA alkyltransferase (hAGT or MGMT). We showed that hAGT through its active site nucleophilic Cys145 thiolate spontaneously reacts with 8-nitro-guanine in DNA to form a stable DNA-protein cross-link (DPC). Interestingly, the process of DPC formation provided protection from PN-mediated genome instability, growth arrest, and apoptosis. The Cys145 mutant of hAGT failed to form a DPC and was unable to protect cells from inflammation-associated PN-mediated cytotoxicity. Gel-shift, dot blot, and UV-vis assays showed formation of a covalent linkage between PN-damaged DNA and hAGT through its active site Cys145. Finally, expression of hAGT was found to be significantly increased by induced macrophages and PN. The data presented here clearly demonstrated hAGT as a dual-function protein that along with DNA repair is capable of maintaining genomic integrity and providing protection from the toxicity caused by PN-mediated DNA damage. Although DPCs are known to be detrimental to the cell, recently, multiple pathways have been identified in normal cells for their repair.
Collapse
Affiliation(s)
- Shayantani Chakraborty
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence (DTU), Greater Noida, Uttar Pradesh 201314, India
| | - Shaista Haider
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence (DTU), Greater Noida, Uttar Pradesh 201314, India
| | - Gargi Mukherjee
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence (DTU), Greater Noida, Uttar Pradesh 201314, India
| | - Anindita Chakrabarty
- Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institution of Eminence (DTU), Greater Noida, Uttar Pradesh 201314, India
| | - Goutam Chowdhury
- Independent Researcher, Greater Noida, Uttar Pradesh 201308, India
| |
Collapse
|
2
|
Kobayashi H, Tohyama S, Ichimura H, Ohashi N, Chino S, Soma Y, Tani H, Tanaka Y, Yang X, Shiba N, Kadota S, Haga K, Moriwaki T, Morita-Umei Y, Umei TC, Sekine O, Kishino Y, Kanazawa H, Kawagishi H, Yamada M, Narita K, Naito T, Seto T, Kuwahara K, Shiba Y, Fukuda K. Regeneration of Nonhuman Primate Hearts With Human Induced Pluripotent Stem Cell-Derived Cardiac Spheroids. Circulation 2024; 150:611-621. [PMID: 38666382 DOI: 10.1161/circulationaha.123.064876] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 03/21/2024] [Indexed: 08/21/2024]
Abstract
BACKGROUND The clinical application of human induced pluripotent stem cell-derived cardiomyocytes (CMs) for cardiac repair commenced with the epicardial delivery of engineered cardiac tissue; however, the feasibility of the direct delivery of human induced pluripotent stem cell-derived CMs into the cardiac muscle layer, which has reportedly induced electrical integration, is unclear because of concerns about poor engraftment of CMs and posttransplant arrhythmias. Thus, in this study, we prepared purified human induced pluripotent stem cell-derived cardiac spheroids (hiPSC-CSs) and investigated whether their direct injection could regenerate infarcted nonhuman primate hearts. METHODS We performed 2 separate experiments to explore the appropriate number of human induced pluripotent stem cell-derived CMs. In the first experiment, 10 cynomolgus monkeys were subjected to myocardial infarction 2 weeks before transplantation and were designated as recipients of hiPSC-CSs containing 2×107 CMs or the vehicle. The animals were euthanized 12 weeks after transplantation for histological analysis, and cardiac function and arrhythmia were monitored during the observational period. In the second study, we repeated the equivalent transplantation study using more CMs (6×107 CMs). RESULTS Recipients of hiPSC-CSs containing 2×107 CMs showed limited CM grafts and transient increases in fractional shortening compared with those of the vehicle (fractional shortening at 4 weeks after transplantation [mean ± SD]: 26.2±2.1%; 19.3±1.8%; P<0.05), with a low incidence of posttransplant arrhythmia. Transplantation of increased dose of CMs resulted in significantly greater engraftment and long-term contractile benefits (fractional shortening at 12 weeks after transplantation: 22.5±1.0%; 16.6±1.1%; P<0.01, left ventricular ejection fraction at 12 weeks after transplantation: 49.0±1.4%; 36.3±2.9%; P<0.01). The incidence of posttransplant arrhythmia slightly increased in recipients of hiPSC-CSs containing 6×107 CMs. CONCLUSIONS We demonstrated that direct injection of hiPSC-CSs restores the contractile functions of injured primate hearts with an acceptable risk of posttransplant arrhythmia. Although the mechanism for the functional benefits is not fully elucidated, these findings provide a strong rationale for conducting clinical trials using the equivalent CM products.
Collapse
Affiliation(s)
- Hideki Kobayashi
- Department of Cardiovascular Medicine (H. Kobayashi, K.K.), Shinshu University, Matsumoto, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Hajime Ichimura
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Noburo Ohashi
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
| | - Shuji Chino
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
| | - Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Yuki Tanaka
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Xiao Yang
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Naoko Shiba
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Shin Kadota
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
- Institute for Biomedical Sciences (S.K., H. Kawagishi, K.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Kotaro Haga
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Taijun Moriwaki
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Yuika Morita-Umei
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
- Kanagawa Institute of Industrial Science and Technology, Japan (Y.M.-U.)
| | - Tomohiko C Umei
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Otoya Sekine
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Hiroyuki Kawagishi
- Department of Molecular Pharmacology (H. Kawagishi, M.Y.), Shinshu University, Matsumoto, Japan
- Institute for Biomedical Sciences (S.K., H. Kawagishi, K.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Mitsuhiko Yamada
- Department of Molecular Pharmacology (H. Kawagishi, M.Y.), Shinshu University, Matsumoto, Japan
| | - Kazumasa Narita
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine (K.N., T.N.), Shinshu University, Matsumoto, Japan
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan (K.N., T.N.)
| | - Takafumi Naito
- Department of Clinical Pharmacology and Therapeutics, Graduate School of Medicine (K.N., T.N.), Shinshu University, Matsumoto, Japan
- Department of Pharmacy, Shinshu University Hospital, Matsumoto, Japan (K.N., T.N.)
| | - Tatsuichiro Seto
- Division of Cardiovascular Surgery, Department of Surgery (H.I., N.O., S.C., Y.T., T.S.), Shinshu University, Matsumoto, Japan
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| | - Koichiro Kuwahara
- Department of Cardiovascular Medicine (H. Kobayashi, K.K.), Shinshu University, Matsumoto, Japan
- Institute for Biomedical Sciences (S.K., H. Kawagishi, K.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Yuji Shiba
- School of Medicine, Department of Regenerative Science and Medicine (H.I., Y.T., X.Y., N.S., S.K., Y. Shiba), Shinshu University, Matsumoto, Japan
- Institute for Biomedical Sciences (S.K., H. Kawagishi, K.K., Y. Shiba), Shinshu University, Matsumoto, Japan
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (S.T., Y. Soma, H.T., K.H., T.M., Y.M.-U., T.C.U., O.S., Y.K., H. Kanazawa, K.F.)
| |
Collapse
|
3
|
Elkhamary A, Gerner I, Bileck A, Oreff GL, Gerner C, Jenner F. Comparative proteomic profiling of the ovine and human PBMC inflammatory response. Sci Rep 2024; 14:14939. [PMID: 38942936 PMCID: PMC11213919 DOI: 10.1038/s41598-024-66059-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/26/2024] [Indexed: 06/30/2024] Open
Abstract
Understanding the cellular and molecular mechanisms of inflammation requires robust animal models. Sheep are commonly used in immune-related studies, yet the validity of sheep as animal models for immune and inflammatory diseases remains to be established. This cross-species comparative study analyzed the in vitro inflammatory response of ovine (oPBMCs) and human PBMCs (hPBMCs) using mass spectrometry, profiling the proteome of the secretome and whole cell lysate. Of the entire cell lysate proteome (oPBMCs: 4217, hPBMCs: 4574 proteins) 47.8% and in the secretome proteome (oPBMCs: 1913, hPBMCs: 1375 proteins) 32.8% were orthologous between species, among them 32 orthologous CD antigens, indicating the presence of six immune cell subsets. Following inflammatory stimulation, 71 proteins in oPBMCs and 176 in hPBMCs showed differential abundance, with only 7 overlapping. Network and Gene Ontology analyses identified 16 shared inflammatory-related terms and 17 canonical pathways with similar activation/inhibition patterns in both species, demonstrating significant conservation in specific immune and inflammatory responses. However, ovine PMBCs also contained a unique WC1+γδ T-cell subset, not detected in hPBMCs. Furthermore, differences in the activation/inhibition trends of seven canonical pathways and the sets of DAPs between sheep and humans, emphasize the need to consider interspecies differences in translational studies and inflammation research.
Collapse
Affiliation(s)
- A Elkhamary
- Department for Companion Animals and Horses, Veterm, University Equine Hospital, Vetmeduni Vienna, Vienna, Austria
- Department for Surgery, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - I Gerner
- Department for Companion Animals and Horses, Veterm, University Equine Hospital, Vetmeduni Vienna, Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - A Bileck
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - G L Oreff
- Department for Companion Animals and Horses, Veterm, University Equine Hospital, Vetmeduni Vienna, Vienna, Austria
| | - C Gerner
- Department of Analytical Chemistry, Faculty of Chemistry, University of Vienna, Vienna, Austria
| | - F Jenner
- Department for Companion Animals and Horses, Veterm, University Equine Hospital, Vetmeduni Vienna, Vienna, Austria.
- Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
4
|
Srinontong P, Aengwanich W, Somphon S, Khonwai S, Nitsinsakul T, Wu Z, Chalalai T, Saraphol B, Srisanyong W. Comparison of lipopolysaccharide-mediated peripheral blood mononuclear cell activation between Brahman and Brahman × Thai native crossbreed cattle. Vet World 2024; 17:804-810. [PMID: 38798282 PMCID: PMC11111707 DOI: 10.14202/vetworld.2024.804-810] [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: 12/07/2023] [Accepted: 03/21/2024] [Indexed: 05/29/2024] Open
Abstract
Background and Aims Lipopolysaccharide (LPS) is a robust endotoxin known to activate the immune system in cattle. The objective of this study was to investigate the effect of LPS on the morphology, cell viability, malondialdehyde (MDA), nitric oxide (NO), and total antioxidant capacity (TAC) of peripheral blood mononuclear cells (PBMCs) in Brahman and Brahman × Thai native crossbreed cattle. Materials and Methods PBMCs were isolated from Brahman and Brahman × Thai native crossbreed cattle and treated with 0, 0.1, 1, and 10 μg/mL Escherichia coli LPS, respectively. Morphological changes in PBMCs were assessed at 24 and 48 h. In addition, we measured PBMC cell viability, MDA, NO, and TAC. Results LPS stimulation caused cell deformation and partial PBMC area enlargement, but there were no differences between Brahman and Brahman × Thai native crossbreed cattle. Stimulation at all levels did not affect the viability of PBMCs (p > 0.05). MDA and NO levels were significantly higher in Brahman cattle than in Brahman Thai native crossbred cattle (p < 0.05). TAC was significantly higher in Brahman × Thai native crossbred cattle than in Brahman cattle (p < 0.05). Conclusion Immune cells of crossbreed cattle have a higher activation response to LPS than those of purebred cattle, and native crossbreed beef cattle have a higher antioxidant capacity than purebred beef cattle. This result may explain why hybrid cattle of indigenous breeds are more resistant to disease than purebred cattle.
Collapse
Affiliation(s)
- Piyarat Srinontong
- Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
- Bioveterinary Research Unit, Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Worapol Aengwanich
- Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
- Stress and Oxidative Stress in Animal Research Unit, Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Sattabongkod Somphon
- Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Siriyakorn Khonwai
- Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Thanasorn Nitsinsakul
- Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Zhiliang Wu
- Department of Parasitology and Infectious Diseases, Gifu University Graduate School of Medicine, Gifu 5011194, Japan
| | - Thanyakorn Chalalai
- Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Bhuripit Saraphol
- Faculty of Veterinary Sciences, Mahasarakham University, Mahasarakham 44000, Thailand
| | - Wilasinee Srisanyong
- Department of Veterinary Technology, Faculty of Agriculture Technology, Kalasin University, Kalasin 46000, Thailand
| |
Collapse
|
5
|
Herzig MC, Christy BA, Montgomery RK, Cantu-Garza C, Barrera GD, Lee JH, Mucha N, Talackine JR, Abaasah IA, Bynum JA, Cap AP. Short-term assays for mesenchymal stromal cell immunosuppression of T-lymphocytes. Front Immunol 2023; 14:1225047. [PMID: 37822938 PMCID: PMC10562633 DOI: 10.3389/fimmu.2023.1225047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/31/2023] [Indexed: 10/13/2023] Open
Abstract
Introduction Trauma patients are susceptible to coagulopathy and dysfunctional immune responses. Mesenchymal stromal cells (MSCs) are at the forefront of the cellular therapy revolution with profound immunomodulatory, regenerative, and therapeutic potential. Routine assays to assess immunomodulation activity examine MSC effects on proliferation of peripheral blood mononuclear cells (PBMCs) and take 3-7 days. Assays that could be done in a shorter period of time would be beneficial to allow more rapid comparison of different MSC donors. The studies presented here focused on assays for MSC suppression of mitogen-stimulated PBMC activation in time frames of 24 h or less. Methods Three potential assays were examined-assays of apoptosis focusing on caspase activation, assays of phosphatidyl serine externalization (PS+) on PBMCs, and measurement of tumor necrosis factor alpha (TNFα) levels using rapid ELISA methods. All assays used the same initial experimental conditions: cryopreserved PBMCs from 8 to 10 pooled donors, co-culture with and without MSCs in 96-well plates, and PBMC stimulation with mitogen for 2-72 h. Results Suppression of caspase activity in activated PBMCs by incubation with MSCs was not robust and was only significant at times after 24 h. Monitoring PS+ of live CD3+ or live CD4+/CD3+ mitogen-activated PBMCs was dose dependent, reproducible, robust, and evident at the earliest time point taken, 2 h, although no increase in the percentage of PS+ cells was seen with time. The ability of MSC in co-culture to suppress PBMC PS+ externalization compared favorably to two concomitant assays for MSC co-culture suppression of PBMC proliferation, at 72 h by ATP assay, or at 96 h by fluorescently labeled protein signal dilution. TNFα release by mitogen-activated PBMCs was dose dependent, reproducible, robust, and evident at the earliest time point taken, with accumulating signal over time. However, suppression levels with MSC co-culture was reliably seen only after 24 h. Discussion Takeaways from these studies are as follows: (1) while early measures of PBMC activation is evident at 2-6 h, immunosuppression was only reliably detected at 24 h; (2) PS externalization at 24 h is a surrogate assay for MSC immunomodulation; and (3) rapid ELISA assay detection of TNFα release by PBMCs is a robust and sensitive assay for MSC immunomodulation at 24 h.
Collapse
Affiliation(s)
- Maryanne C. Herzig
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Barbara A. Christy
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Robbie K. Montgomery
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Carolina Cantu-Garza
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Gema D. Barrera
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Ji H. Lee
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Nicholas Mucha
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Jennifer R. Talackine
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - Isaac A. Abaasah
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| | - James A. Bynum
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
- Department of Surgery, University of Texas, Health Science Center, San Antonio, TX, United States
| | - Andrew P. Cap
- Blood and Shock Research, US Army Institute of Surgical Research, Fort Sam Houston, TX, United States
| |
Collapse
|
6
|
Pojero F, Gervasi F, Fiore SD, Aiello A, Bonacci S, Caldarella R, Attanzio A, Candore G, Caruso C, Ligotti ME, Procopio A, Restivo I, Tesoriere L, Allegra M, Accardi G. Anti-Inflammatory Effects of Nutritionally Relevant Concentrations of Oleuropein and Hydroxytyrosol on Peripheral Blood Mononuclear Cells: An Age-Related Analysis. Int J Mol Sci 2023; 24:11029. [PMID: 37446206 DOI: 10.3390/ijms241311029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Immunosenescence and inflammaging facilitate the insurgence of chronic diseases. The Mediterranean diet is a non-invasive intervention to improve the chronic low-grade inflammatory status associated with aging. Olive oil oleuropein (OLE) and hydroxytyrosol (HT) demonstrated a controversial modulatory action on inflammation in vitro when tested at concentrations exceeding those detectable in human plasma. We studied the potential anti-inflammatory effects of OLE and HT at nutritionally relevant concentrations on peripheral blood mononuclear cells (PBMCs) as regards cell viability, frequency of leukocyte subsets, and cytokine release, performing an age-focused analysis on two groups of subjects: Adult (age 18-64 years) and Senior (age ≥ 65 years). OLE and HT were used alone or as a pre-treatment before challenging PBMCs with lipopolysaccharide (LPS). Both polyphenols had no effect on cell viability irrespective of LPS, but 5 µM HT had an LPS-like effect on monocytes, reducing the intermediate subset in Adult subjects. OLE and HT had no effect on LPS-triggered release of TNF-α, IL-6 and IL-8, but 5 µM HT reduced IL-10 secretion by PBMCs from Adult vs. Senior group. In summary, nutritionally relevant concentrations of OLE and HT elicit no anti-inflammatory effect and influence the frequency of immune cell subsets with age-related different outcomes.
Collapse
Affiliation(s)
- Fanny Pojero
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Francesco Gervasi
- Specialistic Oncology Laboratory Unit, ARNAS Hospitals Civico Di Cristina e Benfratelli, 90127 Palermo, Italy
| | - Salvatore Davide Fiore
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Sonia Bonacci
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Rosalia Caldarella
- Department of Laboratory Medicine, "P. Giaccone" University Hospital, 90127 Palermo, Italy
| | - Alessandro Attanzio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Antonio Procopio
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Mario Allegra
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| |
Collapse
|
7
|
Preglej T, Brinkmann M, Steiner G, Aletaha D, Göschl L, Bonelli M. Advanced immunophenotyping: A powerful tool for immune profiling, drug screening, and a personalized treatment approach. Front Immunol 2023; 14:1096096. [PMID: 37033944 PMCID: PMC10080106 DOI: 10.3389/fimmu.2023.1096096] [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: 11/11/2022] [Accepted: 03/13/2023] [Indexed: 04/11/2023] Open
Abstract
Various autoimmune diseases are characterized by distinct cell subset distributions and activation profiles of peripheral blood mononuclear cells (PBMCs). PBMCs can therefore serve as an ideal biomarker material, which is easily accessible and allows for screening of multiple cell types. A detailed understanding of the immune landscape is critical for the diagnosis of patients with autoimmune diseases, as well as for a personalized treatment approach. In our study, we investigate the potential of multi-parameter spectral flow cytometry for the identification of patients suffering from autoimmune diseases and its power as an evaluation tool for in vitro drug screening approaches (advanced immunophenotyping). We designed a combination of two 22-color immunophenotyping panels for profiling cell subset distribution and cell activation. Downstream bioinformatics analyses included percentages of individual cell populations and median fluorescent intensity of defined markers which were then visualized as heatmaps and in dimensionality reduction approaches. In vitro testing of epigenetic immunomodulatory drugs revealed an altered activation status upon treatment, which supports the use of spectral flow cytometry as a high-throughput drug screening tool. Advanced immunophenotyping might support the exploration of novel therapeutic drugs and contribute to future personalized treatment approaches in autoimmune diseases and beyond.
Collapse
Affiliation(s)
| | | | | | | | - Lisa Göschl
- *Correspondence: Lisa Göschl, ; Michael Bonelli,
| | | |
Collapse
|
8
|
Harwood OE, Balgeman AJ, Weaver AJ, Ellis-Connell AL, Weiler AM, Erickson KN, Matschke LM, Golfinos AE, Vezys V, Skinner PJ, Safrit JT, Edlefsen PT, Reynolds MR, Friedrich TC, O’Connor SL. Transient T Cell Expansion, Activation, and Proliferation in Therapeutically Vaccinated Simian Immunodeficiency Virus-Positive Macaques Treated with N-803. J Virol 2022; 96:e0142422. [PMID: 36377872 PMCID: PMC9749465 DOI: 10.1128/jvi.01424-22] [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: 09/15/2022] [Accepted: 10/25/2022] [Indexed: 11/16/2022] Open
Abstract
Vaccine strategies aimed at eliciting human immunodeficiency virus (HIV)-specific CD8+ T cells are one major target of interest in HIV functional cure strategies. We hypothesized that CD8+ T cells elicited by therapeutic vaccination during antiretroviral therapy (ART) would be recalled and boosted by treatment with the interleukin 15 (IL-15) superagonist N-803 after ART discontinuation. We intravenously immunized four simian immunodeficiency virus-positive (SIV+) Mauritian cynomolgus macaques receiving ART with vesicular stomatitis virus (VSV), modified vaccinia virus Ankara strain (MVA), and recombinant adenovirus serotype 5 (rAd-5) vectors all expressing SIVmac239 Gag. Immediately after ART cessation, these animals received three doses of N-803. Four control animals received no vaccines or N-803. The vaccine regimen generated a high-magnitude response involving Gag-specific CD8+ T cells that were proliferative and biased toward an effector memory phenotype. We then compared cells elicited by vaccination (Gag specific) to cells elicited by SIV infection and unaffected by vaccination (Nef specific). We found that N-803 treatment enhanced the frequencies of both bulk and proliferating antigen-specific CD8+ T cells elicited by vaccination and the antigen-specific CD8+ T cells elicited by SIV infection. In sum, we demonstrate that a therapeutic heterologous prime-boost-boost (HPBB) vaccine can elicit antigen-specific effector memory CD8+ T cells that are boosted by N-803. IMPORTANCE While antiretroviral therapy (ART) can suppress HIV replication, it is not a cure. It is therefore essential to develop therapeutic strategies to enhance the immune system to better become activated and recognize virus-infected cells. Here, we evaluated a novel therapeutic vaccination strategy delivered to SIV+ Mauritian cynomolgus macaques receiving ART. ART was then discontinued and we delivered an immunotherapeutic agent (N-803) after ART withdrawal with the goal of eliciting and boosting anti-SIV cellular immunity. Immunologic and virologic analysis of peripheral blood and lymph nodes collected from these animals revealed transient boosts in the frequency, activation, proliferation, and memory phenotype of CD4+ and CD8+ T cells following each intervention. Overall, these results are important in educating the field of the transient nature of the immunological responses to this particular therapeutic regimen and the similar effects of N-803 on boosting T cells elicited by vaccination or elicited naturally by infection.
Collapse
Affiliation(s)
- Olivia E. Harwood
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Alexis J. Balgeman
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Abigail J. Weaver
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Amy L. Ellis-Connell
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Andrea M. Weiler
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | | | - Lea M. Matschke
- Department of Pathobiological Sciences, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Athena E. Golfinos
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Vaiva Vezys
- Center for Immunology, Department of Microbiology and Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Pamela J. Skinner
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Paul T. Edlefsen
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Matthew R. Reynolds
- Department of Pathobiological Sciences, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Thomas C. Friedrich
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
- Department of Pathobiological Sciences, University of Wisconsin—Madison, Madison, Wisconsin, USA
| | - Shelby L. O’Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin—Madison, Madison, Wisconsin, USA
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| |
Collapse
|
9
|
Jacques C, Floris I. Special Focus on the Cellular Anti-Inflammatory Effects of Several Micro-Immunotherapy Formulations: Considerations Regarding Intestinal-, Immune-Axis-Related- and Neuronal-Inflammation Contexts. J Inflamm Res 2022; 15:6695-6717. [PMID: 36536643 PMCID: PMC9759027 DOI: 10.2147/jir.s389614] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 11/24/2022] [Indexed: 04/11/2024] Open
Abstract
INTRODUCTION Chronic inflammation is a pernicious underlying status, well-known for its contribution to the progressive development of various diseases. In this regard, Micro-immunotherapy (MI) might be a promising therapeutic strategy. MI employs low doses (LD) and ultra-low doses (ULD) of immune regulators in their formulations. In particular, as both IL-1β and TNF-α are often used at ULD in MI medicines (MIM), a special emphasis has been made on formulations that include these factors in their compositions. METHODS Several in vitro models have been employed in order to assess the effects of two unitary MIM consisting of ULD of IL-1β and TNF-α (u-MIM-1 and u-MIM-2, respectively), and four complex MIM (c-MIM-1, -2, -3 and -4) characterized by the presence of ULD of IL-1β and TNF-α amongst other factors. Thus, we first investigated the anti-inflammatory effects of u-MIM-1 and u-MIM-2 in a model of inflamed colon carcinoma cells. In addition, the anti-inflammatory potential of c-MIM-1, -2, -3 and -4, was assessed in in vitro models of intestinal and neuronal inflammation. RESULTS The results revealed that u-MIM-1 and u-MIM-2 both induced a slight decrease in the levels of IL-1β and TNF-α transcripts. Regarding the c-MIMs' effects, c-MIM-1 displayed the capability to restore the altered transepithelial electrical resistance in inflamed-HCoEpiC cells. Moreover, c-MIM-1 also slightly increased the expression of the junction-related protein claudin-1, both at the mRNA and protein levels. In addition, our in vitro investigations on c-MIM-2 and c-MIM-3 revealed their immune-modulatory effects in LPS-inflamed human monocytes, macrophages, and granulocytes, on the secretion of cytokines such as TNF-α, PGE2, and IL-6. Finally, c-MIM-4 restored the cell viability of LPS/IFN-γ-inflamed rat cortical neurons, while reducing the secretion of TNF-α in rat glial cells. DISCUSSION Our results shed the light on the potential role of these MIM formulations in managing several chronic inflammation-related conditions.
Collapse
Affiliation(s)
- Camille Jacques
- Preclinical Research Department, Labo’Life France, Nantes, France
| | - Ilaria Floris
- Preclinical Research Department, Labo’Life France, Nantes, France
| |
Collapse
|
10
|
Kolotilin I. Plant-produced recombinant cytokines IL-37b and IL-38 modulate inflammatory response from stimulated human PBMCs. Sci Rep 2022; 12:19450. [PMID: 36376518 PMCID: PMC9663505 DOI: 10.1038/s41598-022-23828-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022] Open
Abstract
Affordable therapeutics are vitally needed for humans worldwide. Plant-based production of recombinant proteins can potentially enhance, back-up, or even substitute for the manufacturing capacity of the conventional, fermenter-based technologies. We plastome-engineered a tobacco cultivar to express high levels of two "plantakines" - recombinant human cytokines, interleukins IL-37b and IL-38, and confirmed their native conformation and folding. Assessment of their biological functionality was performed ex vivo by analyzing the effects exerted by the plantakines on levels of 11 cytokines secreted from human peripheral blood mononuclear cells (PBMCs) challenged with an inflammatory agent. Application of the plant-produced IL-37b and IL-38 in PBMCs stimulated with Lipopolysaccharide or Phytohaemagglutinin resulted in significant, and in particular cases-dose-dependent modulation of pro-inflammatory cytokines secretion, showing attenuation in two-thirds of significant level modulations observed. Plantakine treatments that increased inflammatory responses were associated with the higher dosage. Our results demonstrate feasibility of manufacturing functional recombinant human proteins using scalable, cost-effective and eco-friendly plant-based bioreactors.
Collapse
|
11
|
Domínguez-Mozo MI, García-Frontini Nieto MC, Gómez-Calcerrada MI, Pérez-Pérez S, García-Martínez MÁ, Villar LM, Villarrubia N, Costa-Frossard L, Arroyo R, Alvarez-Lafuente R. Mitochondrial Impairments in Peripheral Blood Mononuclear Cells of Multiple Sclerosis Patients. BIOLOGY 2022; 11:1633. [PMID: 36358334 PMCID: PMC9687791 DOI: 10.3390/biology11111633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 11/09/2022]
Abstract
Although impaired mitochondrial function has been proposed as a hallmark of multiple sclerosis (MS) disease, few studies focus on the mitochondria of immune cells. We aimed to compare the mitochondrial function of the peripheral blood mononuclear cells (PBMCs) from MS patients with (M+) and without (M-) lipid-specific oligoclonal immunoglobulin M bands (LS-OCMB), and healthydonors (HD). We conducted an exploratory cross-sectional study with 19 untreated MS patients (M+ = 9 and M- = 10) and 17 HDs. Mitochondrial superoxide anion production and mitochondrial mass in PBMCs were assessed without and with phytohemagglutinin by flow cytometry. The PBMCs' mitochondrial function was analyzed using Seahorse technology. Superoxide anion production corrected by the mitochondrial mass was higher in MS patients compared with HDs (p = 0.011). Mitochondrial function from M+ patients showed some impairments compared with M- patients. Without stimulus, we observed higher proton leak (p = 0.041) but lower coupling efficiency (p = 0.041) in M+ patients; and under stimulation, lower metabolic potential ECAR (p = 0.011), and lower stressed OCR/ECAR in the same patients. Exclusively among M+ patients, we described a higher mitochondrial dysfunction in the oldest ones. The mitochondrial impairments found in the PBMCs from MS patients, specifically in M+ patients, could help to better understand the disease's physiopathology.
Collapse
Affiliation(s)
- María Inmaculada Domínguez-Mozo
- Environmental Factors in Degenerative Diseases Research Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - María Celeste García-Frontini Nieto
- Environmental Factors in Degenerative Diseases Research Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - María Isabel Gómez-Calcerrada
- Environmental Factors in Degenerative Diseases Research Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Silvia Pérez-Pérez
- Environmental Factors in Degenerative Diseases Research Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - María Ángel García-Martínez
- Environmental Factors in Degenerative Diseases Research Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| | - Luisa María Villar
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | - Noelia Villarrubia
- Department of Immunology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), 28040 Madrid, Spain
| | | | - Rafael Arroyo
- Department of Neurology, Hospital Universitario Quironsalud Madrid, 28223 Madrid, Spain
| | - Roberto Alvarez-Lafuente
- Environmental Factors in Degenerative Diseases Research Group, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), 28040 Madrid, Spain
| |
Collapse
|
12
|
Structure modifications of 2-phenylquinoline by Aspergillus genera produce novel derivatives with potent leishmanicidal and anti-inflammatory properties. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
13
|
Yan J, Zhu J, Zhu X, Liu H, Chen G. Circ_0092012 knockdown restrains non-small cell lung cancer progression by inhibiting cell malignant phenotype and immune escape through microRNA-635/programmed death ligand 1 axis. Bioengineered 2022; 13:13929-13943. [PMID: 35723188 PMCID: PMC9276036 DOI: 10.1080/21655979.2022.2080386] [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] [Indexed: 11/03/2022] Open
Abstract
Circular RNAs have been reported to play roles in non-small cell lung cancer (NSCLC) progression. Herein, this work aimed to investigate the potential value of circ_0092012 in NSCLC progression. Levels of genes and proteins were detected using quantitative reverse transcription-polymerase chain reaction and Western blot, respectively. The growth, malignant phenotypes and immune escape in NSCLC were investigated. The binding between microRNA (miR)-635 and circ_0092012 or programmed death ligand 1 (PDL1) was verified. Circ_0092012 was highly expressed in NSCLC. Circ_0092012 deficiency suppressed NSCLC cell proliferation, invasion and migration, moreover, as well as was able to inhibit the apoptosis of CD8 + T cells and induce higher interferon-γ and tumor necrosis factor-α levels when co-cultured with peripheral blood mononuclear cells. Mechanistically, circ_0092012 sponged miR-635, which targeted PDL1. Further rescue experiments suggested that the anticancer effects of circ_0092012 knockdown were reversed by miR-635 inhibition. Additionally, miR-635 re-expression suppressed NSCLC cell malignant phenotypes mentioned above and immune escape, which were attenuated by PDL1 overexpression. Moreover, circ_0092012 deletion retarded NSCLC growth in vivo. In all, circ_0092012 knockdown suppressed NSCLC cell oncogenic phenotypes and immune escape by miR-635/PDL1 axis.
Collapse
Affiliation(s)
- Jin Yan
- Department of Respiratory and Critical Care Medicine, Binhai County People's Hospital, Yancheng, Jiangsu, China
| | - Jian Zhu
- Department of Respiratory and Critical Care Medicine, Binhai County People's Hospital, Yancheng, Jiangsu, China
| | - Xiaoli Zhu
- Department of Respiratory and Critical Care Medicine, Zhongda Hospital Southeast University, Nanjing, Jiangsu, China
| | - Hailing Liu
- Department of Radiology, Binhai County People's Hospital, Yancheng, Jiangsu, China
| | - Guoping Chen
- Department of Respiratory and Critical Care Medicine, Binhai County People's Hospital, Yancheng, Jiangsu, China
| |
Collapse
|
14
|
Bogéa GMR, Silva-Carvalho AÉ, Filiú-Braga LDDC, Neves FDAR, Saldanha-Araujo F. The Inflammatory Status of Soluble Microenvironment Influences the Capacity of Melanoma Cells to Control T-Cell Responses. Front Oncol 2022; 12:858425. [PMID: 35419291 PMCID: PMC8996246 DOI: 10.3389/fonc.2022.858425] [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: 01/19/2022] [Accepted: 02/28/2022] [Indexed: 11/20/2022] Open
Abstract
The development of immunotherapeutic approaches for the treatment of melanoma requires a better understanding of immunoescape mechanisms of tumor cells and how they interact with other tumor-resident cell types. Here, we evaluated how the conditioned media of resting (rCM) and immune-activated PBMCs (iCM) influence the ability of a metastatic melanoma cell line (MeWo) to control T-cells function. MeWo cells were expanded in RPMI, rCM, or iCM and the secretome generated after cell expansion was identified as MeSec (RPMI), niSec (non-inflammatory), or iSec (inflammatory secretome), respectively. Then, the immunomodulatory potential of such secretomes was tested in PHA-activated PBMCs. iCM induced higher levels of IFN-γ and IL-10 in treated melanoma cells compared to rCM, as well as higher IDO and PD-L1 expression. The iSec was able to inhibit T-cell activation and proliferation. Interestingly, PBMCs treated with iSec presented a reduced expression of the regulators of Th1 and Th2 responses T-BET and GATA-3, as well as low expression of IFN-γ, and co-stimulatory molecules TIM-3 and LAG-3. Importantly, our findings show that melanoma may benefit from an inflammatory microenvironment to enhance its ability to control the T-cell response. Interestingly, such an immunomodulatory effect involves the inhibition of the checkpoint molecules LAG-3 and TIM-3, which are currently investigated as important therapeutic targets for melanoma treatment. Further studies are needed to better understand how checkpoint molecules are modulated by paracrine and cell contact-dependent interaction between melanoma and immune cells. Such advances are fundamental for the development of new therapeutic approaches focused on melanoma immunotherapy.
Collapse
Affiliation(s)
- Gabriela Muller Reche Bogéa
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
| | - Amandda Évelin Silva-Carvalho
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
| | - Luma Dayane de Carvalho Filiú-Braga
- Laboratório de Farmacologia Molecular, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
| | | | - Felipe Saldanha-Araujo
- Laboratório de Hematologia e Células-Tronco, Faculdade de Ciências da Saúde, Universidade de Brasília, Brasília, Brazil
| |
Collapse
|