1
|
Barbosa AI, Yousef I, Costa Lima SA, Reis S. Design and characterization of κ-Carrageenan:PVA hydrogels to repurpose the topical delivery of betamethasone. Int J Pharm 2025; 671:125305. [PMID: 39894090 DOI: 10.1016/j.ijpharm.2025.125305] [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: 09/25/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Atopic dermatitis (AD) is a severe inflammatory skin disorder, affecting children and adults worldwide, and despite the several existing treatments, it is necessary to find new alternative topical therapies. Hydrogels may represent a good tool to treat AD due to their high water content, making them excellent candidates for drug delivery vehicles in skin research. This work aimed to develop and characterize hybrid hydrogels composed of gel-forming polymers (k-carrageenan and polyvinyl alcohol) for cutaneous delivery of betamethasone (up to 0.2 mg mL-1) widely used to manage AD, with high skin retention. Bergamot oil and menthol essential oils were also incorporated into the hydrogels to study their effects on penetration and retention of the corticosteroid. Rheological properties revealed the pseudoplastic behavior of the hydrogels, a favorable characteristic for skin application. Cytocompatibility towards fibroblasts and keratinocytes was determined, revealing safe usage of the hydrogel blends up to 100 mg mL-1, corresponding to 20 µg mL-1 in betamethasone, but was compromised by the presence of the essential oils in the higher hydrogel tested concentrations (50 and 100 mg mL-1). The ex vivo pig ear skin permeation assay showed that hydrogels promote betamethasone retention up to 20 % of the added dose (c.a. 10 µg) even after 24 h of permeation, independently of the use of essential oils' use in the composition, showing that they might be a good strategy to treat AD skin.
Collapse
Affiliation(s)
- Ana Isabel Barbosa
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto, Portugal
| | - Ibraheem Yousef
- ALBA Synchrotron, Carrer de la Llum 2-26, Cerdanyola del Vallès 08290 Barcelona, Spain
| | - Sofia A Costa Lima
- LAQV, REQUIMTE, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto, Portugal.
| | - Salette Reis
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 228 4050-313 Porto, Portugal
| |
Collapse
|
2
|
Kealy DJ, Wilson JC, Jaconelli T, Hogg K, Coop R, Forshaw G, Todd N, Yates D, Signoret N. Blood immune profiles reveal a CXCR3/CCR5 axis of dysregulation in early sepsis. J Leukoc Biol 2025; 117:qiae204. [PMID: 39312202 DOI: 10.1093/jleuko/qiae204] [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: 07/26/2024] [Accepted: 09/20/2024] [Indexed: 03/06/2025] Open
Abstract
We report on a pilot study exploring whether blood immune signatures can reveal early specific indicator profiles for patients meeting sepsis criteria upon hospital admission. We analyzed samples of sepsis-suspected patients (n = 20) and age-spanning healthy controls (n = 12) using flow cytometry-based assays. We measured inflammatory markers from plasma fractions and immunophenotyped freshly isolated unfixed peripheral blood mononucleated cells for leukocyte subset representation and expression of activation markers, including chemokine receptors. We found that besides IL-6 and sCD14, CXCR3 ligands (CXCL9 and CXCL10) separated sepsis-suspected patients from healthy controls. The abundance of CD4+ T cells was significantly reduced in patients, while they displayed substantial losses of CCR5-expressing monocytes and CXCR3/CCR5 double-positive T cells. Post hoc subgrouping of patients according to their sepsis diagnosis on discharge identified CXCR3/CCR5 double expression on T cells as a separating characteristic for confirmed cases. This work suggests a potential novel axis of dysregulation affecting CXCR3 and CCR5 in early sepsis.
Collapse
Affiliation(s)
- David J Kealy
- Hull York Medical School, Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| | - Julie C Wilson
- Department of Mathematics, James College, University of York, York YO10 5DD, UK
| | - Tom Jaconelli
- York & Scarborough Teaching Hospital NHS Foundation Trust, Wigginton Rd, Clifton, York YO31 8HE, UK
| | - Karen Hogg
- Bioscience Technology Facility, Departement of Biology, University of York, York YO10 5DD, UK
| | - Rebecca Coop
- York & Scarborough Teaching Hospital NHS Foundation Trust, Wigginton Rd, Clifton, York YO31 8HE, UK
| | - Greg Forshaw
- York & Scarborough Teaching Hospital NHS Foundation Trust, Wigginton Rd, Clifton, York YO31 8HE, UK
| | - Neil Todd
- York & Scarborough Teaching Hospital NHS Foundation Trust, Wigginton Rd, Clifton, York YO31 8HE, UK
| | - David Yates
- York & Scarborough Teaching Hospital NHS Foundation Trust, Wigginton Rd, Clifton, York YO31 8HE, UK
| | - Nathalie Signoret
- Hull York Medical School, Department of Biology, University of York, Wentworth Way, York YO10 5DD, UK
| |
Collapse
|
3
|
Patel S, Ellis K, Scipione CA, Fish JE, Howe KL. Epigallocatechin gallate (EGCG) modulates senescent endothelial cell-monocyte communication in age-related vascular inflammation. Front Cardiovasc Med 2025; 11:1506360. [PMID: 39906338 PMCID: PMC11790594 DOI: 10.3389/fcvm.2024.1506360] [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: 10/05/2024] [Accepted: 12/26/2024] [Indexed: 02/06/2025] Open
Abstract
Aging significantly affects intercellular communication between vascular endothelial cells (ECs) and hematopoietic cells, leading to vascular inflammation and age-associated diseases. This study determined how senescent ECs communicate with monocytes, whether extracellular vesicles (EVs) released from senescent ECs affect monocyte functions, and investigated the potential for epigallocatechin-3-gallate (EGCG), a flavonoid in green tea, to reverse these effects. Human umbilical vein endothelial cells (HUVECs) were treated with Etoposide (10 µM, 24 h) to induce senescence, followed by EGCG (100 µM, 24 h) treatment to evaluate its potential as a senotherapeutic agent. The interaction between ECs and monocytes was analyzed using a co-culture system and direct treatment of monocytes with EC-derived EVs. EGCG reduced senescence-associated phenotypes in ECs, as evidenced by decreased senescence-associated (SA)-β-Gal activity and reversal of Etoposide-induced senescence markers. Monocytes co-cultured with EGCG-treated senescent ECs showed decreased pro-inflammatory responses compared to those co-cultured with untreated senescent ECs. Additionally, senescent ECs produced more EVs than non-senescent ECs. EVs from senescent ECs enhanced lipopolysaccharide (LPS)-induced pro-inflammatory activation of monocytes, whereas EVs from EGCG-treated senescent ECs mitigated this activation, maintaining monocyte activation at normal levels. Our findings reveal that EGCG confers anti-senescent effects via modulation of the senescent EC secretome (including EVs) with the capacity to modify monocyte activation. These findings suggest that EGCG could act as a senotherapeutic agent to reduce vascular inflammation related to aging.
Collapse
Affiliation(s)
- Sarvatit Patel
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Kai Ellis
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
- Department of Genetics and Genome Biology, SickKids Research Institute, Toronto, ON, Canada
| | - Corey A. Scipione
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
Stafford LS, Plummer CE, Smith WC, Gibson DJ, Sharma J, Vicuna V, Diakite S, Larkin J. A peptide mimic of SOCS1 modulates equine peripheral immune cells in vitro and ocular effector functions in vivo: implications for recurrent uveitis. Front Immunol 2025; 15:1513157. [PMID: 39867889 PMCID: PMC11757128 DOI: 10.3389/fimmu.2024.1513157] [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: 10/17/2024] [Accepted: 12/17/2024] [Indexed: 01/28/2025] Open
Abstract
Introduction Recurrent uveitis (RU), an autoimmune disease, is a leading cause of ocular detriment in humans and horses. Equine and human RU share many similarities including spontaneous disease and aberrant cytokine signaling. Reduced levels of SOCS1, a critical regulator of cytokine signaling, is associated with several autoimmune diseases. Topical administration of SOCS1-KIR, a peptide mimic of SOCS1, was previously correlated to reduced ocular pathologies within ERU patients. Methods To further assess the translational potential of a SOCS1 mimetic to treat RU, we assessed peptide-mediated modulation of immune functions in vitro, using equine peripheral blood mononuclear cells (PBMC), and in vivo through topical administration of SOCS1-KIR into the eyes of experimental (non-uveitic) horses. Equine PBMCs from non-uveitic control and ERU horses were cultured with or without SOCS1-KIR pretreatment, followed by 72 hours of mitogen stimulation. Proliferation was assessed using MTT, and cytokine production within cell supernatants was assessed by Luminex. SOCS1-KIR or carrier eye-drops were topically applied to experimental horse eyes twice daily for 21 days, followed by enucleation and isolation of ocular aqueous and vitreous humor. Histology was used to assess peptide treatment safety and localization within treated equine eyes. Cytokine secretion within aqueous humor and vitreous, isolated from experimental equine eyes, was measured by Luminex. Results Following SOCS1-KIR pretreatment, cell proliferation significantly decreased in control, but not ERU-derived PBMCs. Despite differential regulation of cellular proliferation, SOCS1-KIR significantly reduced TNFα and IL-10 secretion in PHA-stimulated control and ERU equine PBMC. SOCS1-KIR increased PBMC secretion of IL-8. Topically administered SOCS1-KIR was well tolerated. Although SOCS1-KIR was undetectable within the eye, topically treated equine eyes had significant reductions in TNFα and IL-10. Interestingly, we found that while SOCS1-KIR treatment reduced TNFα and IL-10 production in healthy and ERU PBMC, SOCS1-KIR differentially modulated proliferation, IP-10 production, and RANTES within these two groups suggesting possible differences in cell types or activation status. Discussion Topical administration of a SOCS1 peptide mimic is safe to the equine eye and reduces ERU associated cytokines IL-10 and TNFα serving as potential biomarkers of drug efficacy in a future clinical trial.
Collapse
Affiliation(s)
- Lauren Stewart Stafford
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| | - Caryn E. Plummer
- Departments of Large and Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL, United States
| | - W. Clay Smith
- Department of Ophthalmology, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Daniel J. Gibson
- Capstone College of Nursing, University of Alabama, Tuscaloosa, AL, United States
| | - Jatin Sharma
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| | - Valeria Vicuna
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| | - Sisse Diakite
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| | - Joseph Larkin
- Microbiology and Cell Science, Institute of Food and Agricultural Science, University of Florida, Gainesville, FL, United States
| |
Collapse
|
5
|
Zhu X, Tian C, Yao D, Li S, Lv J, Chen Y, Huang X. Anti-inflammatory properties of ophioglonin derived from the fern Ophioglossum vulgatum L. via inactivating NF-κB and MAPK signaling pathways. FEBS Open Bio 2025; 15:122-139. [PMID: 39455284 PMCID: PMC11705509 DOI: 10.1002/2211-5463.13914] [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: 06/08/2024] [Revised: 09/24/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Medicinal plants contain bioactive compounds that have therapeutic effects on human health. Ophioglossum vulgatum L. is a representative species of the fern genus Ophioglossum that has anti-inflammatory properties as recognized in folk medicine. Herein, we performed a nitric oxide (NO) assay-guided screening in RAW264.7 cells to investigate the active components of the plant. We found that ophioglonin (OPN), a characteristic homoflavonoid of the genus Ophioglossum, is one of the bioactive components. Therefore, we performed a comparative analysis of the isolated compounds and found that OPN has effects similar to those of isolated dihydroquercetin and luteolin at the concentrations tested. The antioxidant and anti-inflammatory activities of OPN were extensively validated using lipopolysaccharide -stimulated RAW264.7 cells, mouse bone marrow-derived macrophages (BMDMs), and peritoneal exudate macrophages (PEMs). In vivo experiments with a carrageenan-induced mouse paw edema model further confirmed the anti-inflammatory effect of OPN. Additionally, we found that OPN and Ophioglossum vulgatum extracts inhibit the activation of signal transducers, NF-ĸB p65, IĸBα, ERK, p38, and JNK, consistent with the findings of pathway enrichment analysis. This work reinforces the anti-inflammatory properties of Ophioglossum vulgatum and indicates that OPN is a promising therapeutic agent for inflammation-associated disorders. Further clinical evaluations, including clinical trials, would be beneficial to validate the anti-inflammatory properties of OPN.
Collapse
Affiliation(s)
- Xiaoqing Zhu
- Science and Technology Industry Development Center, Chongqing Medical and Pharmaceutical CollegeChongqingChina
- Institute of Immunology, People's Liberation ArmyThird Military Medical UniversityChongqingChina
| | - Cheng Tian
- Science and Technology Industry Development Center, Chongqing Medical and Pharmaceutical CollegeChongqingChina
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery SystemChongqing Medical and Pharmaceutical CollegeChongqingChina
| | - Dan Yao
- Science and Technology Industry Development Center, Chongqing Medical and Pharmaceutical CollegeChongqingChina
| | - Siqi Li
- Institute of Immunology, People's Liberation ArmyThird Military Medical UniversityChongqingChina
| | - Junjiang Lv
- Science and Technology Industry Development Center, Chongqing Medical and Pharmaceutical CollegeChongqingChina
| | - Yongwen Chen
- Institute of Immunology, People's Liberation ArmyThird Military Medical UniversityChongqingChina
| | - Xiaoyong Huang
- Institute of Immunology, People's Liberation ArmyThird Military Medical UniversityChongqingChina
| |
Collapse
|
6
|
Ismaya WT, Karsono AH, Tandrasasmita OM, Tjandrawinata RR, Rachmawati H. Agaricus bisporus Mannose-Binding Protein Stimulates the Innate Immune Cells. Adv Pharm Bull 2024; 14:944-950. [PMID: 40190680 PMCID: PMC11970490 DOI: 10.34172/apb.43767] [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: 09/23/2024] [Revised: 12/03/2024] [Accepted: 12/03/2024] [Indexed: 04/09/2025] Open
Abstract
Purpose A lectin-like protein from the mushroom Agaricus bisporus has been shown to slightly increase the proliferation of RAW 264.7 cells. Following its identification as a mannose-binding lectin, henceforth called A. bisporus mannose-binding protein (Abmb), the protein is hypothesized to stimulate the innate immune cells response. The present work was aimed to substantiate that hypothesis. Furthermore, this study complements Abmb exploration as a potential agent for anti-breast cancer, which its treatment is hampered with compromised immunity of patient receiving chemotherapy. Methods Abmb's effect on the phagocytic activity of the macrophage was measured with FACS. Nitric oxide (NO) production was checked using Griess test while expression of the cytokines in the RAW 264.7 cells was analysed at gene and protein level using polymerase chain reaction (PCR) and FACS, respectively. Abmb's effect on the expression of surface markers of the human immune cells in the peripheral blood mononuclear cells (PBMCs) was checked with specific antibodies for targeted cluster differentiation (CD) and analysed using FACS. Results Abmb increased the phagocytic activity of the macrophage and NO production. Abmb increased the expression of cytokines i.e. tumour necrosis factor (TNF)-α, interleukin (IL)-6, and IL-10. With the PBMCs, Abmb activated dendritic and natural killer (NK) cells, but not the B- or T-cells. Conclusion Abmb increased the activity of the macrophage cells and activated the immune cells that are related to the innate immune system, particularly the cellular immunity.
Collapse
Affiliation(s)
- Wangsa Tirta Ismaya
- Dexa Laboratories of Biomolecular Sciences, Industri Selatan V Blok PP-7, Jababeka II Industrial Estate, Cikarang 17559, West Java, Indonesia
| | - Agung Heru Karsono
- Dexa Laboratories of Biomolecular Sciences, Industri Selatan V Blok PP-7, Jababeka II Industrial Estate, Cikarang 17559, West Java, Indonesia
| | - Olivia Mayasari Tandrasasmita
- Dexa Laboratories of Biomolecular Sciences, Industri Selatan V Blok PP-7, Jababeka II Industrial Estate, Cikarang 17559, West Java, Indonesia
| | - Raymond Rubianto Tjandrawinata
- Dexa Laboratories of Biomolecular Sciences, Industri Selatan V Blok PP-7, Jababeka II Industrial Estate, Cikarang 17559, West Java, Indonesia
- Faculty of Biotechnology, Atma Jaya Catholic University of Indonesia, Jl. Raya Cisauk-Lapan 10, Tangerang 15345, Banten, Indonesia
| | - Heni Rachmawati
- Research Group of Pharmaceutics, School of Pharmacy, Bandung Institute of Technology, Ganesa 10, Bandung 40132, West Java, Indonesia
- Research Centre for Nanosciences and Nanotechnology, Bandung Institute of Technology, Ganesa 10, Bandung 40132, West Java, Indonesia
| |
Collapse
|
7
|
Owokoniran OH, Honda A, Ichinose T, Ishikawa R, Nagao M, Miyasaka N, Wang Z, Takai S, Omori I, Zhang K, Liu W, Higaki Y, Kameda T, Matsuda T, Fujiwara T, Okuda T, Takano H. Co-exposure of ferruginous components of subway particles with lipopolysaccharide impairs vascular function: A comparative study with ambient particulate matter. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 288:117356. [PMID: 39579445 DOI: 10.1016/j.ecoenv.2024.117356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/05/2024] [Accepted: 11/14/2024] [Indexed: 11/25/2024]
Abstract
Several empirical studies have linked subway and ambient particle exposure to toxicity, pro-inflammatory responses, and vascular dysfunction. However, the health effects of pollutants generated from varying sources, particularly when combined with lipopolysaccharide (LPS), are still unexplored. Therefore, the aim of this study was to investigate the characteristic health effects of iron oxide particles (the main components of subway particles) in comparison with urban aerosols (UA) and vehicle exhaust particles (VEP), alone and in combination with LPS. This study revealed that iron oxides caused a more significant reduction in human umbilical vein endothelial cell viability, increased lactate dehydrogenase release, and decreased the production of plasminogen activator inhibitor-1, a fibrinolytic modulator, and endothelin-1, a vasoconstrictor, compared to those by VEP and UA at marginally toxic and toxic concentrations. While VEP and UA induced an increase in interleukin (IL)-6 production, iron oxides, particularly Fe3O4, increased IL-8 production at slightly toxic and non-cytotoxic concentrations. In addition, co-exposure of all particles and LPS at non-cytotoxic concentrations promoted pro-inflammatory cytokine (IL-6 and IL-8) production relative to exposure to the particles alone. Interestingly, the tendency towards either coagulation or fibrinolytic conditions was dependent on the concentration of exposed particles at the same LPS concentration. Furthermore, increases in inflammation, neutrophil and lymphocyte recruitment around blood vessels, and edema were observed in murine lungs exposed to a combination of iron oxides and LPS compared to those in mice exposed to iron oxide alone. Thus, iron oxide-rich subway particulate poses more health risks than outdoor ambient particles since they can significantly impair endothelial function, particularly through gross cellular and vascular homeostatic protein damage, and induce exacerbated inflammatory responses during co-exposure. These findings provide novel empirical evidence for epidemiological studies seeking mechanisms responsible for the observed health impact of transport- and occupational-related exposures on vascular dysfunction.
Collapse
Affiliation(s)
| | - Akiko Honda
- Environmental Health Division, Graduate School of Engineering, Kyoto University, Kyoto, Japan.
| | - Takamichi Ichinose
- Environmental Health Division, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Raga Ishikawa
- Environmental Health Division, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Megumi Nagao
- Environmental Health Division, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Natsuko Miyasaka
- Environmental Health Division, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Zaoshi Wang
- Environmental Health Division, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Satsuki Takai
- Environmental Health Division, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Issei Omori
- Environmental Health Division, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Kerui Zhang
- Environmental Health Division, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Wei Liu
- Environmental Health Division, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Yuya Higaki
- Environmental Health Division, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Takayuki Kameda
- Department of Socio-Environmental Energy Science, Graduate School of Energy Science, Kyoto University, Kyoto, Japan
| | - Tomonari Matsuda
- Environmental Health Division, Graduate School of Engineering, Kyoto University, Kyoto, Japan
| | - Taku Fujiwara
- Environmental Health Division, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan
| | - Tomoaki Okuda
- Department of Applied Chemistry, Faculty of Science and Technology, Keio University, Kanagawa, Japan
| | - Hirohisa Takano
- Environmental Health Division, Graduate School of Global Environmental Studies, Kyoto University, Kyoto, Japan; Institute for International Academic Research, Kyoto University of Advanced Science, Kyoto, Japan; Research Institute for Coexistence and Health Science, Kyoto University of Advanced Science, Kyoto, Japan
| |
Collapse
|
8
|
Ng CYJ, Zhong L, Ng HS, Goh KS, Zhao Y. Managing Type 2 Diabetes Mellitus via the Regulation of Gut Microbiota: A Chinese Medicine Perspective. Nutrients 2024; 16:3935. [PMID: 39599721 PMCID: PMC11597546 DOI: 10.3390/nu16223935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Type 2 Diabetes Mellitus (T2DM) is a metabolic disorder characterized by insulin resistance and inadequate insulin production. Given the increased frequency of T2DM and the health issues it can cause, there is an increasing need to develop alternative T2DM management strategies. One such approach is Chinese Medicine (CM), a complementary therapy widely used in T2DM treatment. Given the emphasis on gut microbiota in current research, studying CM in the treatment of T2DM via gut microbiota modulation could be beneficial. Scope and approach: The use of various CM methods for managing T2DM via gut microbiota modulation is highlighted in this review. Following an introduction of the gut microbiota and its role in T2DM pathogenesis, we will review the potential interactions between gut microbiota and T2DM. Thereafter, we will review various CM treatment modalities that modulate gut microbiota and provide perspectives for future research. Key findings and discussion: In T2DM, Akkermansia, Bifidobacterium, and Firmicutes are examples of gut microbiota commonly imbalanced. Studies have shown that CM therapies can modulate gut microbiota, leading to beneficial effects such as reduced inflammation, improved metabolism, and improved immunity. Among these treatment modalities, Chinese Herbal Medicine and acupuncture are the most well-studied, and several in vivo studies have demonstrated their potential in managing T2DM by modulating gut microbiota. However, the underlying biomolecular mechanisms of actions are not well elucidated, which is a key area for future research. Future studies could also investigate alternate CM therapies such as moxibustion and CM exercises and conduct large-scale clinical trials to validate their effectiveness in treatment.
Collapse
Affiliation(s)
- Chester Yan Jie Ng
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Linda Zhong
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Han Seong Ng
- Singapore General Hospital, Outram Rd., Singapore 169608, Singapore
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
| | - Kia Seng Goh
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
- Singapore College of Traditional Chinese Medicine, 640 Lor 4 Toa Payoh, Singapore 319522, Singapore
| | - Yan Zhao
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Academy of Chinese Medicine Singapore, 705 Serangoon Road, Singapore 328127, Singapore
| |
Collapse
|
9
|
Klimina KM, Dyachkova MS, Veselovsky VA, Zakharevich NV, Strokach AA, Selezneva OV, Shitikov EA, Bespiatykh DA, Yunes RA, Poluektova EU, Odorskaya MV, Ostroukhova PS, Bruskin SA, Danilenko VN, Olekhnovich EI. Transcriptional Responses of Lacticaseibacillus rhamnosus to TNFα, IL-6, IL-8, and IL-10 Cytokines. BIOLOGY 2024; 13:931. [PMID: 39596886 PMCID: PMC11591797 DOI: 10.3390/biology13110931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/29/2024]
Abstract
The interaction between gut microbiota and the host immune system is a complex and understudied field, with cytokines like TNFα, IL-6, IL-8, and IL-10 playing pivotal roles. Commensal bacteria, including lactobacilli, respond to these cytokines through adaptive mechanisms that support their survival and function within the gut. While the influence of cytokines on pathogenic bacteria is well documented, their impact on commensal bacteria, particularly lactobacilli, remains underexplored. This study investigates the transcriptional responses of Lacticaseibacillus rhamnosus strains K32 and R19-3 to various cytokines using next-generation RNA sequencing (RNA-seq). Our findings reveal that cytokines, especially IL-8 and IL-10, significantly alter the L. rhamnosus transcriptome, affecting genes involved in carbohydrate metabolism, stress response, and transcriptional regulation. Notably, IL-8 and IL-10 induce a significant downregulation of genes related to the phosphotransferase system, suggesting a reduction in metabolic activity in response to inflammatory signals. This study unveils a previously unexplored aspect of L. rhamnosus adaptation, highlighting its intricate response to cytokine signals. By modulating gene expression, L. rhamnosus may mitigate the adverse effects of inflammation and promote gut health. These insights could inform the development of targeted probiotic therapies for inflammatory bowel disease (IBD) and other conditions with altered cytokine levels. Our results suggest that co-evolution between a host and gut microbiota enables bacteria to respond to specific cytokines through gene expression changes, revealing a unique and underexplored facet of the interaction between commensal bacteria and the host organism.
Collapse
Affiliation(s)
- Ksenia M. Klimina
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya Str. 1a, Moscow 119435, Russia
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Marina S. Dyachkova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Vladimir A. Veselovsky
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya Str. 1a, Moscow 119435, Russia
| | - Natalia V. Zakharevich
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya Str. 1a, Moscow 119435, Russia
| | - Aleksandra A. Strokach
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya Str. 1a, Moscow 119435, Russia
| | - Oksana V. Selezneva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya Str. 1a, Moscow 119435, Russia
| | - Egor A. Shitikov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya Str. 1a, Moscow 119435, Russia
| | - Dmitry A. Bespiatykh
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya Str. 1a, Moscow 119435, Russia
| | - Roman A. Yunes
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Elena U. Poluektova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Maya V. Odorskaya
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Polina S. Ostroukhova
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Sergey A. Bruskin
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Valeriy N. Danilenko
- Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow 119991, Russia
| | - Evgenii I. Olekhnovich
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Malaya Pirogovskaya Str. 1a, Moscow 119435, Russia
| |
Collapse
|
10
|
Appel K, Rose T, Zimmermann C, Günnewich N. In Vitro Anti-inflammatory Effects of Larch Turpentine, Turpentine Oil, Eucalyptus Oil, and Their Mixture as Contained in a Marketed Ointment. PLANTA MEDICA 2024; 90:1023-1029. [PMID: 39260387 PMCID: PMC11614573 DOI: 10.1055/a-2388-7527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 08/01/2024] [Indexed: 09/13/2024]
Abstract
An ointment containing larch turpentine, turpentine oil, and eucalyptus oil has been used for almost a century for the symptomatic treatment of mild, localized, purulent inflammations of the skin. Its clinical efficacy in the treatment of skin infections has been shown in clinical trials, but the mode of action of the active ingredients on inflammation is not known. We studied the anti-inflammatory properties of the active ingredients of the ointment and their mixture in a human monocyte cell model, in which the cells were stimulated with lipopolysaccharide and incubated with the test substances. The cytotoxic threshold of each test substance and the mixture was identified using the alamarBlue assay, and their anti-inflammatory activity was assessed by measuring the release of interleukins IL-1β, IL-6, IL-8, monocyte chemoattractant protein-1, prostaglandin E2, and TNF-α. Cell toxicity was observed at a mixture concentration of 10 µg/mL. All immunological assays were carried out at nontoxic concentrations. Larch turpentine decreased IL-1β, monocyte chemoattractant protein-1, and prostaglandin E2 release at a concentration of 3.9 µg/mL and TNF-α at concentrations > 1.95 µg/mL, whereas eucalyptus oil and turpentine oil had no relevant inhibitory effects. The mixture dose-dependently inhibited IL-1β, IL-6, monocyte chemoattractant protein-1, prostaglandin E2, and TNF-α release at concentrations > 1 µg/mL. IL-8 release was only marginally affected. The anti-inflammatory activity of the herbal ingredients and their mixture was confirmed in this model. This effect seems to be mediated mainly by larch turpentine, with turpentine oil and eucalyptus oil exerting an additive or possibly synergistic function.
Collapse
Affiliation(s)
- Kurt Appel
- VivaCell Biotechnology GmbH, Denzlingen, Germany
| | | | | | | |
Collapse
|
11
|
Nossol C, Landgraf P, Oster M, Kahlert S, Barta-Böszörmenyi A, Kluess J, Wimmers K, Isermann B, Stork O, Dieterich DC, Dänicke S, Rothkötter HJ. Deoxynivalenol triggers the expression of IL-8-related signaling cascades and decreases protein biosynthesis in primary monocyte-derived cells. Mycotoxin Res 2024; 40:279-293. [PMID: 38498144 PMCID: PMC11043135 DOI: 10.1007/s12550-024-00528-3] [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: 12/05/2023] [Revised: 03/01/2024] [Accepted: 03/09/2024] [Indexed: 03/20/2024]
Abstract
Humans and their immune system are confronted with mold-contaminated food and/or mold-contaminated air in daily life and indoor activities. This results in metabolic stress and unspecific disease symptoms. Other studies provided evidence that exposure to mold is associated with the etiology of allergies. Deoxynivalenol (DON) is of great concern due to its frequent occurrence in toxically relevant concentrations. The exposure to this toxin is a permanent health risk for both humans and farm animals because DON cannot be significantly removed during standard milling and processing procedures. However, the direct effect on immunity or hematology is poorly defined because most investigations could not separate the effect of DON-contaminated feed intake. Due to the widespread distribution of DON after rapid absorption, it is not surprising that DON is known to affect the immune system. The immune system of the organism has one important function, to defend against the invasion of unknown substances/organisms. This study shows for the first time a synergistic effect of both-low physiological DON-doses in combination with low LPS-doses with the focus on the IL-8 expression on protein and RNA level. Both doses were found in vivo. IL-8 together with other anorectic cytokines like IL-1β can affect the food intake and anorexia. We could also show that a calcium-response is not involved in the increased IL-8 production after acute DON stimulation with high or low concentrations.
Collapse
Affiliation(s)
- Constanze Nossol
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany.
| | - P Landgraf
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - M Oster
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - S Kahlert
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - A Barta-Böszörmenyi
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - J Kluess
- Institute of Animal Nutrition, Friedrich-Loeffler-Institute (FLI), Federal Research Institute for Animal Health, Bundesallee 50, 38116, Braunschweig, Germany
| | - K Wimmers
- Research Institute for Farm Animal Biology (FBN), Wilhelm-Stahl-Allee 2, 18196, Dummerstorf, Germany
| | - B Isermann
- Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostics, Medical Faculty, Paul-List-Str. 13-15, 04103, Leipzig, Germany
| | - O Stork
- Institute of Biology, Faculty of Natural Science, Otto-von-Guericke University Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - D C Dieterich
- Institute for Pharmacology and Toxicology, Medical Faculty, Otto-von-Guericke University Magdeburg, Leipziger Straße 44, 39120, Magdeburg, Germany
| | - S Dänicke
- Institute of Animal Nutrition, Friedrich-Loeffler-Institute (FLI), Federal Research Institute for Animal Health, Bundesallee 50, 38116, Braunschweig, Germany
| | - H-J Rothkötter
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke University Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany
| |
Collapse
|
12
|
Zeng M, Hodges JK, Pokala A, Khalafi M, Sasaki GY, Pierson J, Cao S, Brock G, Yu Z, Zhu J, Vodovotz Y, Bruno RS. A green tea extract confection decreases circulating endotoxin and fasting glucose by improving gut barrier function but without affecting systemic inflammation: A double-blind, placebo-controlled randomized trial in healthy adults and adults with metabolic syndrome. Nutr Res 2024; 124:94-110. [PMID: 38430822 DOI: 10.1016/j.nutres.2024.02.001] [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: 11/28/2023] [Revised: 01/11/2024] [Accepted: 02/01/2024] [Indexed: 03/05/2024]
Abstract
Anti-inflammatory activities of catechin-rich green tea extract (GTE) in obese rodents protect against metabolic endotoxemia by decreasing intestinal permeability and absorption of gut-derived endotoxin. However, translation to human health has not been established. We hypothesized that GTE would reduce endotoxemia by decreasing gut permeability and intestinal and systemic inflammation in persons with metabolic syndrome (MetS) compared with healthy persons. A randomized, double-blind, placebo-controlled, crossover trial in healthy adults (n = 19, 34 ± 2 years) and adults with MetS (n = 21, 40 ± 3 years) examined 4-week administration of a decaffeinated GTE confection (890 mg/d total catechins) on serum endotoxin, intestinal permeability, gut and systemic inflammation, and cardiometabolic parameters. Compared with the placebo, the GTE confection decreased serum endotoxin (P = .023) in both healthy persons and those with MetS, while increasing concentrations of circulating catechins (P < .0001) and γ-valerolactones (P = .0001). Fecal calprotectin (P = .029) and myeloperoxidase (P = .048) concentrations were decreased by GTE regardless of health status. Following the ingestion of gut permeability probes, urinary lactose/mannitol (P = .043) but not sucralose/erythritol (P > .05) was decreased by GTE regardless of health status. No between-treatment differences (P > .05) were observed for plasma aminotransferases, blood pressure, plasma lipids, or body mass nor were plasma tumor necrosis factor-α, interleukin-6, or the ratio of lipopolysaccharide-binding protein/soluble cluster of differentiation-14 affected. However, fasting glucose in both study groups was decreased (P = .029) by the GTE confection compared with within-treatment arm baseline concentrations. These findings demonstrate that catechin-rich GTE is effective to decrease circulating endotoxin and improve glycemic control in healthy adults and those with MetS, likely by reducing gut inflammation and small intestinal permeability but without affecting systemic inflammation.
Collapse
Affiliation(s)
- Min Zeng
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Joanna K Hodges
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA; Department of Nutritional Sciences, The Pennsylvania State University, State College, PA, 16801, USA
| | - Avinash Pokala
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Mona Khalafi
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Geoffrey Y Sasaki
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Jillian Pierson
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Sisi Cao
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Guy Brock
- Department of Biomedical Informatics, The Ohio State University, Columbus, OH, 43210, USA
| | - Zhongtang Yu
- Department of Animal Sciences, The Ohio State University, Columbus, OH, 43210, USA
| | - Jiangjiang Zhu
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Yael Vodovotz
- Department of Food Science and Technology, The Ohio State University, Columbus, OH, 43210, USA
| | - Richard S Bruno
- Human Nutrition Program, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
13
|
Seipel K, Mandhair H, Bacher U, Pabst T. FLT3 and IRAK4 Inhibitor Emavusertib in Combination with BH3-Mimetics in the Treatment of Acute Myeloid Leukemia. Curr Issues Mol Biol 2024; 46:2946-2960. [PMID: 38666914 PMCID: PMC11049208 DOI: 10.3390/cimb46040184] [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: 02/27/2024] [Revised: 03/22/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
Targeting the FLT3 receptor and the IL-1R associated kinase 4 as well as the anti-apoptotic proteins MCL1 and BCL2 may be a promising novel approach in the treatment of acute myeloid leukemia (AML). The FLT3 and IRAK4 inhibitor emavusertib (CA4948), the MCL1 inhibitor S63845, the BCL2 inhibitor venetoclax, and the HSP90 inhibitor PU-H71 were assessed as single agents and in combination for their ability to induce apoptosis and cell death in leukemic cells in vitro. AML cells represented all major morphologic and molecular subtypes, including FLT3-ITD and NPM1 mutant AML cell lines and a variety of patient-derived AML cells. Emavusertib in combination with MCL1 inhibitor S63845 or BCL2 inhibitor venetoclax induced cell cycle arrest and apoptosis in MOLM-13 cells. In primary AML cells, the response to emavusertib was associated with the presence of the FLT3 gene mutation with an allelic ratio >0.5 and the presence of NPM1 gene mutations. S63845 was effective in all tested AML cell lines and primary AML samples. Blast cell percentage was positively associated with the response to CA4948, S63845, and venetoclax, with elevated susceptibility of primary AML with blast cell fraction >80%. Biomarkers of the response to venetoclax included the blast cell percentage and bone marrow infiltration rate, as well as the expression levels of CD11b, CD64, and CD117. Elevated susceptibility to CA4948 combination treatments with S63845 or PU-H71 was associated with FLT3-mutated AML and CD34 < 30%. The combination of CA4948 and BH3-mimetics may be effective in the treatment in FLT3-mutated AML with differential target specificity for MCL1 and BCL2 inhibitors. Moreover, the combination of CA4948 and PU-H71 may be a candidate combination treatment in FLT3-mutated AML.
Collapse
Affiliation(s)
- Katja Seipel
- Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Harpreet Mandhair
- Department for Biomedical Research, University of Bern, 3008 Bern, Switzerland;
| | - Ulrike Bacher
- Department of Hematology, University Hospital Bern, 3010 Bern, Switzerland;
| | - Thomas Pabst
- Department of Medical Oncology, University Hospital Bern, 3010 Bern, Switzerland
| |
Collapse
|
14
|
Zhang Z, Scanlan A, Koneru R, Morrell CR, Reece MD, Edwards E, Roa S, Gavegnano C, Bimonte-Nelson H, Arbiser J, Tyor W. Honokiol hexafluoro confers reversal of neuropathological markers of HIV infection in a murine SCID model. Neurotherapeutics 2024; 21:e00329. [PMID: 38388224 PMCID: PMC10943487 DOI: 10.1016/j.neurot.2024.e00329] [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: 10/19/2023] [Revised: 01/27/2024] [Accepted: 01/28/2024] [Indexed: 02/24/2024] Open
Abstract
Cognitive impairment remains a persistent challenge in people living with HIV (PWLH) despite antiretroviral therapy (ART) due to ART's inability to eliminate brain HIV. HIV-induced cognitive dysfunction results from immune dysregulation, ongoing neuroinflammation, and the continuous virus presence, collectively contributing to cognitive deficits. Therefore, adjunctive therapies are needed to reduce cerebral HIV reservoirs, mitigate neuroinflammation, and impede cognitive dysfunction progression. Our study focused on Honokiol, known for its anti-inflammatory and neuroprotective properties, in an experimental mouse model simulating HIV-induced cognitive dysfunction. Using Honokiol Hexafluoro (HH), a synthetic analogue, we comprehensively evaluated its potential to ameliorate cognitive dysfunction and cerebral pathology in HIV-associated cognitive dysfunction. Our findings showed that HH treatment effectively reversed HIV-induced cognitive dysfunction, concurrently suppressing astrocyte activation, restoring neuronal dendritic arborization, and reducing microglial activation. Furthermore, HH remodeled the metabolic profile of HIV-infected human monocyte-derived macrophages, resulting in decreased activation and the promotion of a quiescent state in vitro.
Collapse
Affiliation(s)
- Zhan Zhang
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Aaron Scanlan
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Rajeth Koneru
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Chelsea Richardson Morrell
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA
| | - Monica D Reece
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Emily Edwards
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Sebastian Roa
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Christina Gavegnano
- Atlanta VA Medical Center, Decatur, GA, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA; Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, USA; Center for the Study of Human Health, Emory College, Atlanta, GA, USA; Harvard Medical School, Center for Bioethics, Boston, MA, USA
| | | | - Jack Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, USA; Metroderm/United Derm Partners, Atlanta, GA, USA
| | - William Tyor
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA; Atlanta VA Medical Center, Decatur, GA, USA.
| |
Collapse
|
15
|
Constantinescu S, Niculescu AG, Hudiță A, Grumezescu V, Rădulescu D, Bîrcă AC, Dorcioman G, Gherasim O, Holban AM, Gălățeanu B, Vasile BȘ, Grumezescu AM, Bolocan A, Rădulescu R. Nanostructured Coatings Based on Graphene Oxide for the Management of Periprosthetic Infections. Int J Mol Sci 2024; 25:2389. [PMID: 38397066 PMCID: PMC10889398 DOI: 10.3390/ijms25042389] [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: 01/17/2024] [Revised: 02/12/2024] [Accepted: 02/13/2024] [Indexed: 02/25/2024] Open
Abstract
To modulate the bioactivity and boost the therapeutic outcome of implantable metallic devices, biodegradable coatings based on polylactide (PLA) and graphene oxide nanosheets (nGOs) loaded with Zinforo™ (Zin) have been proposed in this study as innovative alternatives for the local management of biofilm-associated periprosthetic infections. Using a modified Hummers protocol, high-purity and ultra-thin nGOs have been obtained, as evidenced by X-ray diffraction (XRD) and transmission electron microscopy (TEM) investigations. The matrix-assisted pulsed laser evaporation (MAPLE) technique has been successfully employed to obtain the PLA-nGO-Zin coatings. The stoichiometric and uniform transfer was revealed by infrared microscopy (IRM) and scanning electron microscopy (SEM) studies. In vitro evaluation, performed on fresh blood samples, has shown the excellent hemocompatibility of PLA-nGO-Zin-coated samples (with a hemolytic index of 1.15%), together with their anti-inflammatory ability. Moreover, the PLA-nGO-Zin coatings significantly inhibited the development of mature bacterial biofilms, inducing important anti-biofilm efficiency in the as-coated samples. The herein-reported results evidence the promising potential of PLA-nGO-Zin coatings to be used for the biocompatible and antimicrobial surface modification of metallic implants.
Collapse
Affiliation(s)
- Sorin Constantinescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (S.C.); (D.R.); (A.B.); (R.R.)
| | - Adelina-Gabriela Niculescu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (A.-G.N.); (A.H.); (A.M.H.)
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (A.C.B.); (B.Ș.V.)
| | - Ariana Hudiță
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (A.-G.N.); (A.H.); (A.M.H.)
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania;
| | - Valentina Grumezescu
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania; (V.G.); (G.D.); (O.G.)
| | - Dragoș Rădulescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (S.C.); (D.R.); (A.B.); (R.R.)
| | - Alexandra Cătălina Bîrcă
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (A.C.B.); (B.Ș.V.)
| | - Gabriela Dorcioman
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania; (V.G.); (G.D.); (O.G.)
| | - Oana Gherasim
- Lasers Department, National Institute for Lasers, Plasma and Radiation Physics, 409 Atomistilor Street, 077125 Magurele, Romania; (V.G.); (G.D.); (O.G.)
| | - Alina Maria Holban
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (A.-G.N.); (A.H.); (A.M.H.)
- Microbiology and Immunology Department, Faculty of Biology, University of Bucharest, 1-3 Portocalelor Lane, 77206 Bucharest, Romania
| | - Bianca Gălățeanu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 91-95 Splaiul Independentei Street, 050095 Bucharest, Romania;
| | - Bogdan Ștefan Vasile
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (A.C.B.); (B.Ș.V.)
| | - Alexandru Mihai Grumezescu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 90-92 Panduri, 050663 Bucharest, Romania; (A.-G.N.); (A.H.); (A.M.H.)
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 1-7 Gh. Polizu Street, 060042 Bucharest, Romania; (A.C.B.); (B.Ș.V.)
| | - Alexandra Bolocan
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (S.C.); (D.R.); (A.B.); (R.R.)
| | - Radu Rădulescu
- Faculty of Medicine, Carol Davila University of Medicine and Pharmacy, 8 Eroii Sanitari Street, 050474 Bucharest, Romania; (S.C.); (D.R.); (A.B.); (R.R.)
| |
Collapse
|
16
|
Wang M, Feng J, Zhou D, Wang J. Bacterial lipopolysaccharide-induced endothelial activation and dysfunction: a new predictive and therapeutic paradigm for sepsis. Eur J Med Res 2023; 28:339. [PMID: 37700349 PMCID: PMC10498524 DOI: 10.1186/s40001-023-01301-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 08/18/2023] [Indexed: 09/14/2023] Open
Abstract
BACKGROUND Lipopolysaccharide, a highly potent endotoxin responsible for severe sepsis, is the major constituent of the outer membrane of gram-negative bacteria. Endothelial cells participate in both innate and adaptive immune responses as the first cell types to detect lipopolysaccharide or other foreign debris in the bloodstream. Endothelial cells are able to recognize the presence of LPS and recruit specific adaptor proteins to the membrane domains of TLR4, thereby initiating an intracellular signaling cascade. However, lipopolysaccharide binding to endothelial cells induces endothelial activation and even damage, manifested by the expression of proinflammatory cytokines and adhesion molecules that lead to sepsis. MAIN FINDINGS LPS is involved in both local and systemic inflammation, activating both innate and adaptive immunity. Translocation of lipopolysaccharide into the circulation causes endotoxemia. Endothelial dysfunction, including exaggerated inflammation, coagulopathy and vascular leakage, may play a central role in the dysregulated host response and pathogenesis of sepsis. By discussing the many strategies used to treat sepsis, this review attempts to provide an overview of how lipopolysaccharide induces the ever more complex syndrome of sepsis and the potential for the development of novel sepsis therapeutics. CONCLUSIONS To reduce patient morbidity and mortality, preservation of endothelial function would be central to the management of sepsis.
Collapse
Affiliation(s)
- Min Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Jun Feng
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China
| | - Daixing Zhou
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
| | - Junshuai Wang
- Department of Emergency Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
- Department of Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 JieFang Avenue, Wuhan, 430030, Hubei, People's Republic of China.
| |
Collapse
|
17
|
Shalchi-Amirkhiz P, Bensch T, Proschmann U, Stock AK, Ziemssen T, Akgün K. Pilot study on the influence of acute alcohol exposure on biophysical parameters of leukocytes. Front Mol Biosci 2023; 10:1243155. [PMID: 37614440 PMCID: PMC10442941 DOI: 10.3389/fmolb.2023.1243155] [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: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 08/25/2023] Open
Abstract
Objective: This pilot study explores the influence of acute alcohol exposure on cell mechanical properties of steady-state and activated leukocytes conducted with real-time deformability cytometry. Methods: Nineteen healthy male volunteers were enrolled to investigate the effect of binge drinking on biophysical properties and cell counts of peripheral blood leukocytes. Each participant consumed an individualized amount of alcohol to achieve a blood alcohol concentration of 1.2 ‰ as a mean peak. In addition, we also incubated whole blood samples from healthy donors with various ethanol concentrations and performed stimulation experiments using lipopolysaccharide and CytoStim™ in the presence of ethanol. Results: Our findings indicate that the biophysical properties of steady-state leukocytes are not significantly affected by a single episode of binge drinking within the first two hours. However, we observed significant alterations in relative cell counts and a shift toward a memory T cell phenotype. Moreover, exposure to ethanol during stimulation appears to inhibit the cytoskeleton reorganization of monocytes, as evidenced by a hindered increase in cell deformability. Conclusion: Our observations indicate the promising potential of cell mechanical analysis in understanding the influence of ethanol on immune cell functions. Nevertheless, additional investigations in this field are warranted to validate biophysical properties as biomarkers or prognostic indicators for alcohol-related changes in the immune system.
Collapse
Affiliation(s)
- Puya Shalchi-Amirkhiz
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Tristan Bensch
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Undine Proschmann
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Ann-Kathrin Stock
- Cognitive Neurophysiology, Department of Child and Adolescent Psychiatry, Faculty of Medicine of the TU Dresden, Dresden, Germany
- Biopsychology, Department of Psychology, School of Science, TU Dresden, Dresden, Germany
| | - Tjalf Ziemssen
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Katja Akgün
- Multiple Sclerosis Center, Center of Clinical Neuroscience, Department of Neurology, University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| |
Collapse
|
18
|
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
|
19
|
Hulme J. COVID-19 and Diarylamidines: The Parasitic Connection. Int J Mol Sci 2023; 24:6583. [PMID: 37047556 PMCID: PMC10094973 DOI: 10.3390/ijms24076583] [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: 03/15/2023] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/05/2023] Open
Abstract
As emerging severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants (Omicron) continue to outpace and negate combinatorial vaccines and monoclonal antibody therapies targeting the spike protein (S) receptor binding domain (RBD), the appetite for developing similar COVID-19 treatments has significantly diminished, with the attention of the scientific community switching to long COVID treatments. However, treatments that reduce the risk of "post-COVID-19 syndrome" and associated sequelae remain in their infancy, particularly as no established criteria for diagnosis currently exist. Thus, alternative therapies that reduce infection and prevent the broad range of symptoms associated with 'post-COVID-19 syndrome' require investigation. This review begins with an overview of the parasitic-diarylamidine connection, followed by the renin-angiotensin system (RAS) and associated angiotensin-converting enzyme 2 (ACE2) and transmembrane serine protease 2 (TMPRSSR2) involved in SARS-CoV-2 infection. Subsequently, the ability of diarylamidines to inhibit S-protein binding and various membrane serine proteases associated with SARS-CoV-2 and parasitic infections are discussed. Finally, the roles of diarylamidines (primarily DIZE) in vaccine efficacy, epigenetics, and the potential amelioration of long COVID sequelae are highlighted.
Collapse
Affiliation(s)
- John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Sujung-gu, Seongnam-si 461-701, Republic of Korea
| |
Collapse
|
20
|
Sittisak T, Guntawang T, Srivorakul S, Photichai K, Boonprasert K, Khammesri S, Chuammitri P, Thitaram C, Hsu WL, Thanawongnuwech R, Pringproa K. Response of elephant peripheral blood mononuclear cells when stimulated with elephant endotheliotropic herpesvirus glycoprotein B (EEHV-gB). Vet Immunol Immunopathol 2023; 258:110577. [PMID: 36867998 DOI: 10.1016/j.vetimm.2023.110577] [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: 12/20/2022] [Revised: 02/10/2023] [Accepted: 02/26/2023] [Indexed: 03/05/2023]
Abstract
Elephant endotheliotropic herpesvirus-hemorrhagic disease (EEHV-HD) is the most highly fatal infectious disease among young Asian elephants. Despite the fact that antiviral therapy has been widely used, its therapeutic outcomes remain uncertain. Additionally, the virus has yet to be successfully cultivated in vitro in the process of develop viral envelope glycoproteins for vaccine design. The present study aims to investigate and evaluate EEHV1A glycoprotein B (gB) antigenic epitopes as potential candidates for further vaccine development. Epitopes of EEHV1A-gB were employed in in silico predictions and designed by using online antigenic predicting tools. Candidate genes were then constructed, transformed and expressed in the E. coli vectors prior to examine their potential for acceleration elephant immune responses in vitro. Elephant peripheral blood mononuclear cells (PBMCs) isolated from 16 healthy juvenile Asian elephants were investigated for their proliferative capability and cytokine responses after being stimulated with EEHV1A-gB epitopes. Exposure of elephant PBMCs to 20 µg/mL of gB for 72 h resulted in a significant proliferation of CD3 + cells when compared with the control group. Furthermore, proliferation of CD3 + cells was associated with a marked up-regulation of cytokine mRNA expression, involving IL-1β, IL-8, IL-12 and IFN-γ. It remains to be determined whether these candidate EEHV1A-gB epitopes could activate immune responses in animal models or elephants in vivo. Our potentially promising results demonstrate a degree of feasibility for the use of these gB epitopes in expanding EEHV vaccine development.
Collapse
Affiliation(s)
- Tidaratt Sittisak
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Thunyamas Guntawang
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Saralee Srivorakul
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Kornravee Photichai
- Veterinary Diagnostic Center, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Khajohnpat Boonprasert
- Center of Excellence in Elephant and Wildlife Research, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Siripat Khammesri
- Center of Excellence in Elephant and Wildlife Research, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Phongsakorn Chuammitri
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Chatchote Thitaram
- Center of Excellence in Elephant and Wildlife Research, Chiang Mai University, Chiang Mai 50100, Thailand; Department of Companion Animals and Wildlife Clinics, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand
| | - Wei-Li Hsu
- Graduate Institute of Microbiology and Public Health, College of Veterinary Medicine, National Chung Hsing University, Taichung 402, Taiwan
| | - Roongroje Thanawongnuwech
- Department of Pathology, Faculty of Veterinary Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kidsadagon Pringproa
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai 50100, Thailand; Center of Excellence in Elephant and Wildlife Research, Chiang Mai University, Chiang Mai 50100, Thailand.
| |
Collapse
|
21
|
Zamorina S, Timganova V, Bochkova M, Shardina K, Uzhviyuk S, Khramtsov P, Usanina D, Rayev M. The Effect of PEGylated Graphene Oxide Nanoparticles on the Th17-Polarization of Activated T Helpers. MATERIALS (BASEL, SWITZERLAND) 2023; 16:877. [PMID: 36676614 PMCID: PMC9865146 DOI: 10.3390/ma16020877] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 06/17/2023]
Abstract
We investigated the direct effect of PEGylated graphene oxide (P-GO) nanoparticles on the differentiation, viability, and cytokine profile of activated T helper type 17 (Th17) in vitro. The subject of the study were cultures of "naive" T-helpers (CD4+) isolated by immunomagnetic separation and polarized into the Th17 phenotype with a TCR activator and cytokines. It was found that P-GO at low concentrations (5 µg/mL) had no effect on the parameters studied. The presence of high concentrations of P-GO in T-helper cultures (25 μg/mL) did not affect the number and viability of these cells. However, the percentage of proliferating T-helpers in these cultures was reduced. GO nanoparticles modified with linear polyethylene glycol (PEG) significantly increased the percentage of Th17/22 cells in cultures of Th17-polarized T helpers and the production of IFN-γ, whereas those modified with branched PEG suppressed the synthesis of IL-17. Thus, a low concentration of PEGylated GO nanoparticles (5 μg/mL), in contrast to a concentration of 25 μg/mL, has no effect on the Th17-polarization of T helpers, allowing their further use for in-depth studies of the functions of T lymphocytes and other immune cells. Overall, we have studied for the first time the direct effect of P-GO nanoparticles on the conversion of T helper cells to the Th17 phenotype.
Collapse
Affiliation(s)
- Svetlana Zamorina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Valeria Timganova
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Maria Bochkova
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Kseniya Shardina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Sofya Uzhviyuk
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
| | - Pavel Khramtsov
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Darya Usanina
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| | - Mikhail Rayev
- Branch of the Perm Federal Research Center, Ural Branch of the Russian Academy of Sciences, Institute of Ecology and Genetics of Microorganisms, Goleva st., 13, Perm 614081, Russia
- Department of Microbiology and Immunology, Faculty of Biology, Perm State National Research University, Bukireva st., 15, Perm 614990, Russia
| |
Collapse
|
22
|
Gurram PC, Satarker S, Nassar A, Mudgal J, Nampoothiri M. Virtual structure-based docking and molecular dynamics of FDA-approved drugs for the identification of potential IKKB inhibitors possessing dopaminergic activity in Alzheimer’s disease. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-022-02598-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
AbstractIn Alzheimer's disease (AD), neuroinflammation is detrimental in causing neurodegeneration. In the central nervous system, inhibitor of nuclear factor kappa B kinase subunit beta (IKK2/IKKβ/IKKB/IKBKB) signaling is linked to neuroinflammation-mediated learning and memory deficits through canonical pathway, while dopamine agonists have been known to reverse such effects. Our in silico analysis predicted if dopaminergic agonists could have IKKB inhibitory actions, to ameliorate neuroinflammation-associated learning and memory deficits. Here, the FDA-approved Zinc 15 database was screened with IKKB (PDB ID 4KIK). Potential molecules with IKKB inhibition were identified through docking, which also possessed dopaminergic activity. Molecular mechanics—generalized Born and surface area (MMGBSA), induced fit docking (IFD) and molecular dynamic (MD) studies of 100 ns simulation time were done. Apomorphine and rotigotine showed greater non-bonding and bonding interactions with amino acids of IKKB as compared to Aripiprazole in docking studies. The IFD studies predicted improved interactions with IKKB. MMGBSA scores indicated that the complex binding free energies were favorable, and MD studies showed an acceptable root mean square deviation between protein and ligands. The protein–ligand interactions showed hydrogen bonds, water and salt bridges necessary for IKKB inhibition, as well as solvent system stability. On the protein–ligand contact map, the varying color band intensities represented the ligand’s ability to bind with amino acids. Dopamine agonists apomorphine, rotigotine, and aripiprazole were predicted to bind and inhibit IKKB in in silico system.
Graphical Abstract
Collapse
|