1
|
Le Moli R, Naselli A, Piticchio T, Tumminia A, Pallotti F, Belfiore A, Frasca F. The monocyte/HDLc ratio and LDLc are two independent predictors of the response of Graves' ophthalmopathy patients to parenteral glucocorticoids. Endocrine 2025:10.1007/s12020-025-04185-z. [PMID: 39939523 DOI: 10.1007/s12020-025-04185-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Accepted: 01/29/2025] [Indexed: 02/14/2025]
Abstract
PURPOSE Graves' ophthalmopathy (GO) is an inflammatory-autoimmune disease and parenteral glucocorticoids (IvGCs) are the first-line therapy in the moderate to severe forms. Oxidative stress (OX) and cholesterol have been related to severe forms and to the clinical outcome of GO. Recently some new biomarkers have been proposed as predictors of the clinical outcome in some cardiovascular and autoimmune diseases. We hypothesized that the monocyte-to-high-density lipoprotein cholesterol (HDL) ratio (MHR) could be a useful biomarker in GO management and aimed to evaluate the possible role of the MHR as a predictor of the clinical outcome in patients with active, moderate to severe GO treated with IvGCs. METHODS We retrospectively studied 115 patients, 86 females and 29 males, with active, moderate to severe GO who were treated with IvGCs for 12 weeks at our institution. GO severity was evaluated according to EUGOGO suggestions, GO clinical activity and the clinical outcome of GO to IvGCs were evaluated by the seven-point Clinical Activity Score (CAS). RESULTS The baseline low density lipoproteins cholesterol (LDLc) and MHR were negatively and independently related to the improvement of GO at 12 weeks (p = 0.024 and p = 0.012, respectively). The value of the MHR = 0.0095 was identified as the best cut off by ROC curve and appeared to be a potentially useful tool to help identify patients with a poor response to IvGCs. CONCLUSIONS The MHR might be an useful tool to manage the immunosuppressant therapy in GO patients; our study confirms the role of LDLc as a predictor of GO outcome after IvGCs treatment.
Collapse
Affiliation(s)
- Rosario Le Moli
- Department of Clinical and Experimental Medicine, University of Catania, Endocrinology Unit, Catania, Italy.
- Department of Medicine and Surgery, University of Enna "Kore", Enna, Italy.
| | - Adriano Naselli
- Department of Clinical and Experimental Medicine, University of Catania, Endocrinology Unit, Catania, Italy
| | - Tommaso Piticchio
- Department of Clinical and Experimental Medicine, University of Catania, Endocrinology Unit, Catania, Italy
- Department of Medicine and Surgery, University of Enna "Kore", Enna, Italy
| | - Andrea Tumminia
- Department of Clinical and Experimental Medicine, University of Catania, Endocrinology Unit, Catania, Italy
| | - Francesco Pallotti
- Department of Medicine and Surgery, University of Enna "Kore", Enna, Italy
| | - Antonino Belfiore
- Department of Clinical and Experimental Medicine, University of Catania, Endocrinology Unit, Catania, Italy
| | - Francesco Frasca
- Department of Clinical and Experimental Medicine, University of Catania, Endocrinology Unit, Catania, Italy
| |
Collapse
|
2
|
Moreno RJ, Azzam YW, Eng S, Rose D, Ashwood P. Altered Monocyte Populations and Activation Marker Expression in Children with Autism and Co-Occurring Gastrointestinal Symptoms. Biomolecules 2025; 15:207. [PMID: 40001509 PMCID: PMC11853397 DOI: 10.3390/biom15020207] [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: 12/12/2024] [Revised: 01/17/2025] [Accepted: 01/19/2025] [Indexed: 02/27/2025] Open
Abstract
Autism spectrum disorder (ASD) is an early-onset neurodevelopmental condition that now impacts 1 in 36 children in the United States and is characterized by deficits in social communication, repetitive behaviors, and restricted interests. Children with ASD also frequently experience co-morbidities including anxiety and ADHD, and up to 80% experience gastrointestinal (GI) symptoms such as constipation, diarrhea, and/or abdominal pain. Systemic immune activation and dysregulation, including increased pro-inflammatory cytokines, are frequently observed in ASD. Evidence has shown that the innate immune system may be impacted in ASD, as altered monocyte gene expression profiles and cytokine responses to pattern recognition ligands have been observed compared to typically developing (TD) children. In humans, circulating monocytes are often categorized into three subpopulations-classical, transitional (or "intermediate"), and nonclassical monocytes, which can vary in functions, including archetypal inflammatory and/or reparative functions, as well as their effector locations. The potential for monocytes to contribute to immune dysregulation in ASD and its comorbidities has so far not been extensively studied. This study aims to determine whether these monocyte subsets differ in frequency in children with ASD and if the presence of GI symptoms alters subset distribution, as has been seen for T cell subsets. Whole blood from ASD children with (ASD+GI+) and without gastrointestinal symptoms (ASD+GI-) and their TD counterparts was collected from children enrolled in the Childhood Autism Risk from Genetics and Environment (CHARGE) study. Peripheral blood mononuclear cells were isolated and stained for commonly used subset identifiers CD14 and CD16 as well as activation state markers CCR2, HLA-DR, PD-1, and PD-L1 for flow cytometry analysis. We identified changes in monocyte subpopulations and their expression of surface markers in children with ASD compared to TD children. These differences in ASD appear to be dependent on the presence or absence of GI symptoms. We found that the ASD+GI+ group have a different monocyte composition, evident in their classical, transitional, and nonclassical populations, compared to the ASD+GI- and TD groups. Both the ASD+GI+ and ASD+GI- groups exhibited greater frequencies of classical monocytes compared to the TD group. However, the ASD+GI+ group demonstrated lower frequencies of transitional and nonclassical monocytes than their ASD+GI- and TD counterparts. CCR2+ classical monocyte frequencies were highest in the ASD+GI- group. HLA-DR+ classical, transitional, and nonclassical monocytes were statistically comparable between groups, however, HLA-DR- nonclassical monocyte frequencies were lower in both ASD groups compared to TD. The frequency of classical monocytes displaying exhaustion markers PD-1 and PD-L1 were increased in the ASD+GI+ group compared to ASD+GI- and TD, suggesting potentially impaired ability for clearance of foreign pathogens or debris, typically associated with worsened inflammation. Taken together, the findings of differential proportions of the monocyte subpopulations and altered surface markers may explain some of the characteristics of immune dysregulation, such as in the gastrointestinal tract, observed in ASD.
Collapse
Affiliation(s)
- Rachel J. Moreno
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95616, USA
- MIND Institute, University of California, Sacramento, CA 95817, USA
| | - Yasmin W. Azzam
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95616, USA
- MIND Institute, University of California, Sacramento, CA 95817, USA
| | - Serena Eng
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95616, USA
- MIND Institute, University of California, Sacramento, CA 95817, USA
| | - Destanie Rose
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95616, USA
- MIND Institute, University of California, Sacramento, CA 95817, USA
| | - Paul Ashwood
- Department of Medical Microbiology and Immunology, University of California, Davis, CA 95616, USA
- MIND Institute, University of California, Sacramento, CA 95817, USA
| |
Collapse
|
3
|
Kondratyeva LG, Rakitina OA, Pleshkan VV, Kuzmich AI, Linge IA, Kondratieva SA, Snezhkov EV, Alekseenko IV, Sverdlov ED. The Cellular and Transcriptomic Early Innate Immune Response to BCG Vaccination in Mice. Cells 2024; 13:2043. [PMID: 39768135 PMCID: PMC11674076 DOI: 10.3390/cells13242043] [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: 10/31/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
It is established that BCG vaccination results in the development of both a specific immune response to mycobacterial infections and a nonspecific (heterologous) immune response, designated as trained immunity (TRIM), to other pathogens. We hypothesized that local BCG immunization may induce an early immune response in bone marrow and spleen innate immunity cells. The early transcriptomic response of various populations of innate immune cells, including monocytes, neutrophils, and natural killer (NK) cells, to BCG vaccination was examined. To this end, C57Bl/6J mice were subcutaneously immunized with 106 CFU of BCG. Three days following BCG administration, the three cell populations were collected from the control and BCG-vaccinated groups using FACS. All cell populations obtained were utilized for the preparation and sequencing of RNA-seq libraries. The analysis of FACS data revealed an increase in the proportion of splenic NK cells and monocytes 3 days post-vaccination. Transcriptomic analysis revealed the deregulation of genes associated with the regulation of immune response (according to Gene Ontology terms) in NK cells, monocytes, and unsorted bone marrow cells. Two NK cell-specific immune ligands (Tnfsf14 and S100a8) and two bone marrow-specific immune receptors (C5ar1 and Csf2rb) were identified among differentially expressed genes. No alterations were identified in neutrophils in either their percentage or at the transcriptomic level. Thus, in this study, we demonstrated that BCG vaccination provides an early increase in the proportion of murine bone marrow and spleen immune cell populations, as well as transcriptomic alterations in monocytes, NK cells, and non-sorted bone marrow cells. This early innate immune response may be beneficial for enhancing TRIM.
Collapse
Affiliation(s)
- Liya G. Kondratyeva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (O.A.R.); (V.V.P.); (A.I.K.); (S.A.K.); (E.V.S.); (I.V.A.)
- National Research Center “Kurchatov Institute”, Moscow 123182, Russia;
| | - Olga A. Rakitina
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (O.A.R.); (V.V.P.); (A.I.K.); (S.A.K.); (E.V.S.); (I.V.A.)
| | - Victor V. Pleshkan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (O.A.R.); (V.V.P.); (A.I.K.); (S.A.K.); (E.V.S.); (I.V.A.)
- National Research Center “Kurchatov Institute”, Moscow 123182, Russia;
| | - Alexey I. Kuzmich
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (O.A.R.); (V.V.P.); (A.I.K.); (S.A.K.); (E.V.S.); (I.V.A.)
| | - Irina A. Linge
- Central Tuberculosis Research Institute, Moscow 107564, Russia;
| | - Sofia A. Kondratieva
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (O.A.R.); (V.V.P.); (A.I.K.); (S.A.K.); (E.V.S.); (I.V.A.)
| | - Eugene V. Snezhkov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (O.A.R.); (V.V.P.); (A.I.K.); (S.A.K.); (E.V.S.); (I.V.A.)
| | - Irina V. Alekseenko
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow 117997, Russia; (O.A.R.); (V.V.P.); (A.I.K.); (S.A.K.); (E.V.S.); (I.V.A.)
- National Research Center “Kurchatov Institute”, Moscow 123182, Russia;
| | | |
Collapse
|
4
|
You M, Huang Y, Chen Y, Li D, Tang Y, Du YK, Yang H, Liang A, Hu G, Chen Y. ZnO nanoparticles induce melanoma-like lesions via recruiting dermal dendritic cells in barrier-damaged skin in mice. Food Chem Toxicol 2024; 193:114948. [PMID: 39197528 DOI: 10.1016/j.fct.2024.114948] [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: 05/04/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024]
Abstract
ZnO nanoparticles (NPs) are used in skin treatments and cosmetics, the toxicity of long-term and continuous exposure to ZnO NPs is unknown. Mice with epidermal barrier dysfunction revealed melanoma-like lesions after continuous exposure to ZnO NPs. However, the effects of metallic NPs on the skin microenvironment and immune system remain poorly understood. Mice with epidermal barrier failure were given continuous exposure to ZnO NPs for 7 weeks. The malignant transformation of melanocytes was induced with ZnO NPs 2.5 μg/ml for 72 h exposure. The supernatant of the culture medium from dendritic cells after being exposed to 10 μg/ml ZnO NPs for 24 h was applied to melanocytes to explore the effect of recruitment of DCs. The expressure of ZnO NPs resulted in a tendency of malignant transformation of melanocytes, the recruitment of DCs induces this process by produce inflammatory factors such as TNF-α. These DC-produced inflammatory factors, which were induced by ZnO NP exposure, increased the production of matrix metalloproteinases in melanocytes and expedited the malignant transformation process. Our findings revealed that the disrupted cutaneous microenvironment by ZnO NPs penetrated directly promoted the malignant transformation of melanocytes, which process also indirectly enhanced by the TNF-αsecreted from the recruited DCs.
Collapse
Affiliation(s)
- Menghan You
- Dongguan People's Hospital Biobank, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Guangzhou, Guangdong, 510515, China
| | - Yingying Huang
- Department of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, No. 1838 Guangzhou Avenue, Guangzhou, Guangdong, 510515, China
| | - Yeda Chen
- Dongguan People's Hospital Biobank, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Dan Li
- Department of Dermatology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China; Dongguan Key Laboratory of Translational Medicine in Skin and Immune Diseases, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Yaping Tang
- Department of Dermatology, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China; Dongguan Key Laboratory of Translational Medicine in Skin and Immune Diseases, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Yi Kuan Du
- Dongguan People's Hospital Central Laboratory, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Hong Yang
- Department of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Anfa Liang
- Dongguan People's Hospital Biobank, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China
| | - Guodong Hu
- Department of Respiratory and Critical Care Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China.
| | - Yinghua Chen
- Dongguan People's Hospital Biobank, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China; Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Guangzhou, Guangdong, 510515, China; Dongguan Key Laboratory of Translational Medicine in Skin and Immune Diseases, The Tenth Affiliated Hospital of Southern Medical University, Dongguan, Guangdong, 523059, China.
| |
Collapse
|
5
|
Sánchez V, Baumann A, Kromm F, Yergaliyev T, Brandt A, Scholda J, Kopp F, Camarinha-Silva A, Bergheim I. Oral supplementation of choline attenuates the development of alcohol-related liver disease (ALD). Mol Med 2024; 30:181. [PMID: 39425011 PMCID: PMC11488139 DOI: 10.1186/s10020-024-00950-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/04/2024] [Indexed: 10/21/2024] Open
Abstract
BACKGROUND Chronic alcohol intake is associated with alterations of choline metabolism in various tissues. Here, we assessed if an oral choline supplementation attenuated the development of alcohol-related liver disease (ALD) in mice. METHODS Female C57BL/6 J mice (n = 8/group) were either pair-fed a liquid control diet, or a Lieber DeCarli liquid diet (5% ethanol) ± 2.7 g choline/kg diet for 29 days. Liver damage, markers of intestinal permeability and intestinal microbiota composition were determined. Moreover, the effects of choline on ethanol-induced intestinal permeability were assessed in an ex vivo model. RESULTS ALD development as determined by liver histology and assessing markers of inflammation (e.g., nitric oxide, interleukin 6 and 4-hydroxynonenal protein adducts) was attenuated by the supplementation of choline. Intestinal permeability in small intestine being significantly higher in ethanol-fed mice was at the level of controls in ethanol-fed mice receiving choline. In contrast, no effects of the choline supplementation were found on intestinal microbiota composition. Choline also significantly attenuated the ethanol-induced intestinal barrier dysfunction in small intestinal tissue ex vivo, an effect almost entirely abolished by the choline oxidase inhibitor dimbunol. CONCLUSION Our results suggest that an oral choline supplementation attenuates the development of ALD in mice and is related to a protection from intestinal barrier dysfunction.
Collapse
Affiliation(s)
- Victor Sánchez
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), A-1090, Vienna, Austria
| | - Anja Baumann
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), A-1090, Vienna, Austria
| | - Franziska Kromm
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), A-1090, Vienna, Austria
| | - Timur Yergaliyev
- Livestock Microbial Ecology Department, Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | - Annette Brandt
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), A-1090, Vienna, Austria
| | - Julia Scholda
- Department of Pharmaceutical Sciences, Clinical Pharmacy Group, University of Vienna, Vienna, Austria
| | - Florian Kopp
- Department of Pharmaceutical Sciences, Clinical Pharmacy Group, University of Vienna, Vienna, Austria
| | - Amélia Camarinha-Silva
- Livestock Microbial Ecology Department, Institute of Animal Science, University of Hohenheim, Stuttgart, Germany
| | - Ina Bergheim
- Department of Nutritional Sciences, Molecular Nutritional Science, University of Vienna, Josef-Holaubek-Platz 2 (UZA II), A-1090, Vienna, Austria.
| |
Collapse
|
6
|
Hourani T, Sharma A, Luwor RB, Achuthan AA. Transforming growth factor-β in tumor microenvironment: Understanding its impact on monocytes and macrophages for its targeting. Int Rev Immunol 2024; 44:82-97. [PMID: 39377520 DOI: 10.1080/08830185.2024.2411998] [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: 05/29/2024] [Revised: 08/28/2024] [Accepted: 09/25/2024] [Indexed: 10/09/2024]
Abstract
TGF-β is a pivotal cytokine that orchestrates various aspects of cancer progression, including tumor growth, metastasis, and immune evasion. In this review, we present a comprehensive overview of the multifaceted role of transforming growth factor β (TGF-β) in cancer biology, focusing on its intricate interactions with monocytes and macrophages within the tumor microenvironment (TME). We specifically discuss how TGF-β modulates monocyte and macrophage activities, leading to immunosuppression and tumor progression. We conclude with the current translational and clinical efforts targeting TGF-β, recognizing the promising role of this strategy in immunooncology.
Collapse
Affiliation(s)
- Tetiana Hourani
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia
| | - Amit Sharma
- Department of Integrated Oncology, Center for Integrated Oncology (CIO) Bonn, University Hospital Bonn, Bonn, Germany
- Department of Neurosurgery, University Hospital Bonn, Bonn, Germany
| | - Rodney B Luwor
- Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia
- Fiona Elsey Cancer Research Institute, Ballarat, Australia
- Federation University, Ballarat, Australia
| | - Adrian A Achuthan
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, Australia
| |
Collapse
|
7
|
Alves PT, de Souza AG, Bastos VAF, Miguel EL, Ramos ACS, Cameron LC, Goulart LR, Cunha TM. The Modulation of Septic Shock: A Proteomic Approach. Int J Mol Sci 2024; 25:10641. [PMID: 39408970 PMCID: PMC11476436 DOI: 10.3390/ijms251910641] [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: 08/28/2024] [Revised: 09/15/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Sepsis poses a significant challenge due its lethality, involving multiple organ dysfunction and impaired immune responses. Among several factors affecting sepsis, monocytes play a crucial role; however, their phenotype, proteomic profile, and function in septic shock remain unclear. Our aim was to fully characterize the subpopulations and proteomic profiles of monocytes seen in septic shock cases and discuss their possible impact on the disease. Peripheral blood monocyte subpopulations were phenotype based on CD14/CD16 expression by flow cytometry, and proteins were extracted from the monocytes of individuals with septic shock and healthy controls to identify changes in the global protein expression in these cells. Analysis using 2D-nanoUPLC-UDMSE identified 67 differentially expressed proteins in shock patients compared to controls, in which 44 were upregulated and 23 downregulated. These proteins are involved in monocyte reprogramming, immune dysfunction, severe hypotension, hypo-responsiveness to vasoconstrictors, vasodilation, endothelial dysfunction, vascular injury, and blood clotting, elucidating the disease severity and therapeutic challenges of septic shock. This study identified critical biological targets in monocytes that could serve as potential biomarkers for the diagnosis, prognosis, and treatment of septic shock, providing new insights into the pathophysiology of the disease.
Collapse
Affiliation(s)
- Patrícia Terra Alves
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
| | - Aline Gomes de Souza
- Department of Medical Imaging, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirao Preto 14040-900, SP, Brazil;
| | - Victor Alexandre F. Bastos
- Laboratory of Biochemistry, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil;
| | - Eduarda L. Miguel
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| | - Augusto César S. Ramos
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| | - L. C. Cameron
- Arthritis Program, Schroeder Arthritis Institute, Krembil Research Institute, University Health Network, Toronto, ON M5T 0S8, Canada;
- Lorraine Protein Biochemistry Group, Graduate Program in Neurology, Gaffrée e Guinle University Hospital, Rio de Janeiro 20270-004, RJ, Brazil
| | - Luiz Ricardo Goulart
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
| | - Thúlio M. Cunha
- Laboratory of Nanobiotechnology, Institute of Biotechnology, Federal University of Uberlândia, Uberlândia 38402-022, MG, Brazil (T.M.C.)
- School of Medicine, Federal University of Uberlândia, Uberlândia 38408-100, MG, Brazil; (E.L.M.); (A.C.S.R.)
| |
Collapse
|
8
|
Hill KB, Mullen GP, Nagareddy PR, Zimmerman KA, Rudolph MC. Key questions and gaps in understanding adipose tissue macrophages and early-life metabolic programming. Am J Physiol Endocrinol Metab 2024; 327:E478-E497. [PMID: 39171752 PMCID: PMC11482221 DOI: 10.1152/ajpendo.00140.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
The global obesity epidemic, with its associated comorbidities and increased risk of early mortality, underscores the urgent need for enhancing our understanding of the origins of this complex disease. It is increasingly clear that metabolism is programmed early in life and that metabolic programming can have life-long health consequences. As a critical metabolic organ sensitive to early-life stimuli, proper development of adipose tissue (AT) is crucial for life-long energy homeostasis. Early-life nutrients, especially fatty acids (FAs), significantly influence the programming of AT and shape its function and metabolism. Of growing interest are the dynamic responses during pre- and postnatal development to proinflammatory omega-6 (n6) and anti-inflammatory omega-3 (n3) FA exposures in AT. In the US maternal diet, the ratio of "pro-inflammatory" n6- to "anti-inflammatory" n3-FAs has grown dramatically due to the greater prevalence of n6-FAs. Notably, AT macrophages (ATMs) form a significant population within adipose stromal cells, playing not only an instrumental role in AT formation and maintenance but also acting as key mediators of cell-to-cell lipid and cytokine signaling. Despite rapid advances in ATM and immunometabolism fields, research has focused on responses to obesogenic diets and during adulthood. Consequently, there is a significant gap in identifying the mechanisms contributing metabolic health, especially regarding lipid exposures during the establishment of ATM physiology. Our review highlights the current understanding of ATM diversity, their critical role in AT, their potential role in early-life metabolic programming, and the broader implications for metabolism and health.
Collapse
Affiliation(s)
- Kaitlyn B Hill
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Gregory P Mullen
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Cardiovascular Section, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Kurt A Zimmerman
- Department of Internal Medicine, Division of Nephrology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Michael C Rudolph
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
9
|
Larson EL, DeMeo DP, Young AB, Margevicius S, Rutter J, Davies AL, Rohan CA, Korman NJ, Travers JB, McCormick TS, Cooper KD. Circulating Monocytes Are Predictive and Responsive in Moderate-to-Severe Plaque Psoriasis Subjects Treated with Apremilast. J Invest Dermatol 2024; 144:1963-1974.e13. [PMID: 38431222 PMCID: PMC11344695 DOI: 10.1016/j.jid.2024.01.034] [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: 03/17/2023] [Revised: 12/19/2023] [Accepted: 01/08/2024] [Indexed: 03/05/2024]
Abstract
Monocytes play a critical role in the inflammation associated with psoriasis, and their abnormalities have been reported as biomarkers of cardiovascular event risk, a psoriasis comorbidity. Monocytic cells in chronic inflammatory disorders express elevated levels of cAMP phosphodiesterase. Restoring cAMP levels using the oral cAMP phosphodiesterase-4 inhibitor, apremilast, improves clinical outcomes for a subset of patients with psoriasis. We asked whether aberrant monocyte subsets or transcriptomic pathways can function as biomarkers of psoriasis endotypes that can predict enhanced clinical responses to cAMP phosphodiesterase inhibition. A 16-week open-label study of 22 patients with monocyte flow cytometric and transcriptomic analysis was performed. Subjects with elevated hyperadhesive monocyte doublets at baseline were more likely to be responders to apremilast (P < .0001); 82% of subjects with elevated hyperadhesive monocyte doublets achieved 50% reduction in PASI compared with 46% in those without elevated doublets. We observed a significant reduction in hyperadhesive monocyte-containing doublets and monocyte-platelet aggregates, suggesting an effect of apremilast on the adhesiveness of blood monocytes during chronic inflammation. Monocyte differentially expressed gene transcripts predictive of clinical response uncovered pharmacoendotypes with distinct patterns of nucleotide metabolism, energetics, and differentiation. Further study to understand the basis of drug responsiveness and to develop an apremilast psoriasis treatment algorithm using monocyte-refined gene expression is required to validate and become practical in clinical use, offering patients a test that personalizes their likelihood of clinical response.
Collapse
Affiliation(s)
- Emma L. Larson
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Dustin P. DeMeo
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Andrew B. Young
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
| | - Seunghee Margevicius
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Joseph Rutter
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Amanda L. Davies
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Craig A. Rohan
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine at Wright State University, Dayton Ohio, OH 45435, USA
| | - Neil J. Korman
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Jeffrey B. Travers
- Department of Pharmacology & Toxicology, Boonshoft School of Medicine at Wright State University, Dayton Ohio, OH 45435, USA
| | - Thomas S. McCormick
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Kevin D. Cooper
- Department of Dermatology, Case Western Reserve University, Cleveland, OH, USA
- Department of Dermatology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| |
Collapse
|
10
|
Dimitriadis K, Pyrpyris N, Theofilis P, Mantzouranis E, Beneki E, Kostakis P, Koutsopoulos G, Aznaouridis K, Aggeli K, Tsioufis K. Computed Tomography Angiography Identified High-Risk Coronary Plaques: From Diagnosis to Prognosis and Future Management. Diagnostics (Basel) 2024; 14:1671. [PMID: 39125547 PMCID: PMC11311283 DOI: 10.3390/diagnostics14151671] [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: 07/07/2024] [Revised: 07/29/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
CT angiography has become, in recent years, a main evaluating modality for patients with coronary artery disease (CAD). Recent advancements in the field have allowed us to identity not only the presence of obstructive disease but also the characteristics of identified lesions. High-risk coronary atherosclerotic plaques are identified in CT angiographies via a number of specific characteristics and may provide prognostic and therapeutic implications, aiming to prevent future ischemic events via optimizing medical treatment or providing coronary interventions. In light of new evidence evaluating the safety and efficacy of intervening in high-risk plaques, even in non-flow-limiting disease, we aim to provide a comprehensive review of the diagnostic algorithms and implications of plaque vulnerability in CT angiography, identify any differences with invasive imaging, analyze prognostic factors and potential future therapeutic options in such patients, as well as discuss new frontiers, including intervening in non-flow-limiting stenoses and the role of CT angiography in patient stratification.
Collapse
Affiliation(s)
- Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, Hippokration General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (N.P.); (P.T.); (E.M.); (E.B.); (P.K.); (G.K.); (K.A.); (K.A.); (K.T.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Larbi A. From Genesis to Old Age: Exploring the Immune System One Cell at a Time with Flow Cytometry. Biomedicines 2024; 12:1469. [PMID: 39062042 PMCID: PMC11275137 DOI: 10.3390/biomedicines12071469] [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: 05/12/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The immune system is a highly complex and tightly regulated system that plays a crucial role in protecting the body against external threats, such as pathogens, and internal abnormalities, like cancer cells. It undergoes development during fetal stages and continuously learns from each encounter with pathogens, allowing it to develop immunological memory and provide a wide range of immune protection. Over time, after numerous encounters and years of functioning, the immune system can begin to show signs of erosion, which is commonly named immunosenescence. In this review, we aim to explore how the immune system responds to initial encounters with antigens and how it handles persistent stimulations throughout a person's lifetime. Our understanding of the immune system has greatly benefited from advanced technologies like flow cytometry. In this context, we will discuss the valuable contribution of flow cytometry in enhancing our knowledge of the immune system behavior in aging, with a specific focus on T-cells. Moreover, we will expand our discussion to the flow cytometry-based assessment of extracellular vesicles, a recently discovered communication channel in biology, and their implications for immune system functioning.
Collapse
Affiliation(s)
- Anis Larbi
- Medical and Scientific Affairs, Beckman Coulter Life Sciences, 22 Avenue des Nations, 93420 Villepinte, France;
- Department of Medicine, Division of Geriatrics, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| |
Collapse
|
12
|
Tylek T, Wong J, Vaughan AE, Spiller KL. Biomaterial-mediated intracellular control of macrophages for cell therapy in pro-inflammatory and pro-fibrotic conditions. Biomaterials 2024; 308:122545. [PMID: 38547831 PMCID: PMC11264195 DOI: 10.1016/j.biomaterials.2024.122545] [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: 11/29/2023] [Revised: 03/07/2024] [Accepted: 03/19/2024] [Indexed: 05/03/2024]
Abstract
Macrophages are key modulators of all inflammatory diseases and essential for their resolution, making macrophage cell therapy a promising strategy for regenerative medicine. However, since macrophages change rapidly in response to microenvironmental cues, their phenotype must be controlled post-administration. We present a tunable biomaterial-based strategy to control macrophages intracellularly via small molecule-releasing microparticles. Poly(lactic-co-glycolic acid) microparticles encapsulating the anti-inflammatory and anti-fibrotic drug dexamethasone were administered to macrophages in vitro, with uptake rates controlled by different loading regimes. Microparticle dose and dexamethasone content directly affected macrophage phenotype and phagocytic capacity, independent of particle content per cell, leading to an overall pro-reparative, anti-inflammatory, anti-fibrotic phenotype with increased phagocytic and ECM degrading functionality. Intracellularly controlled macrophages partially maintained this phenotype in vivo in a murine pulmonary fibrosis model, with more prominent effects in a pro-fibrotic environment compared to pro-inflammatory. These results suggest that intracellular control using biomaterials has the potential to control macrophage phenotype post-administration, which is essential for successful macrophage cell therapy.
Collapse
Affiliation(s)
- Tina Tylek
- Drexel University, School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA 19104, USA
| | - Joanna Wong
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Andrew E Vaughan
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA 19104, USA
| | - Kara L Spiller
- Drexel University, School of Biomedical Engineering, Science and Health Systems, Philadelphia, PA 19104, USA.
| |
Collapse
|
13
|
Pêgo AC, Lima IS, Martins AC, Sá-Pereira I, Martins G, Gozzelino R. Infection vs. Reinfection: The Immunomodulation of Erythropoiesis. Int J Mol Sci 2024; 25:6153. [PMID: 38892340 PMCID: PMC11172545 DOI: 10.3390/ijms25116153] [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: 05/04/2024] [Revised: 05/28/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Severe malarial anemia (SMA) increases the morbidity and mortality of Plasmodium, the causative agent of malaria. SMA is mainly developed by children and pregnant women in response to the infection. It is characterized by ineffective erythropoiesis caused by impaired erythropoietin (EPO) signaling. To gain new insights into the pathogenesis of SMA, we investigated the relationship between the immune system and erythropoiesis, conducting comparative analyses in a mouse model of malaria. Red blood cell (RBC) production was evaluated in infected and reinfected animals to mimic endemic occurrences. Higher levels of circulating EPO were observed in response to (re)infection. Despite no major differences in bone marrow erythropoiesis, compensatory mechanisms of splenic RBC production were significantly reduced in reinfected mice. Concomitantly, a pronounced immune response activation was observed in erythropoietic organs of reinfected animals in relation to single-infected mice. Aged mice were also used to mimic the occurrence of malaria in the elderly. The increase in symptom severity was correlated with the enhanced activation of the immune system, which significantly impaired erythropoiesis. Immunocompromised mice further support the existence of an immune-shaping regulation of RBC production. Overall, our data reveal the strict correlation between erythropoiesis and immune cells, which ultimately dictates the severity of SMA.
Collapse
Affiliation(s)
| | | | | | | | | | - Raffaella Gozzelino
- NOVA Medical School Research, NOVA University of Lisbon, 1150-082 Lisbon, Portugal
| |
Collapse
|
14
|
Tajani A, Sadeghi M, Omidkhoda N, Mohammadpour AH, Samadi S, Jomehzadeh V. The association between C-reactive protein and coronary artery calcification: a systematic review and meta-analysis. BMC Cardiovasc Disord 2024; 24:204. [PMID: 38600488 PMCID: PMC11007925 DOI: 10.1186/s12872-024-03856-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Accepted: 03/25/2024] [Indexed: 04/12/2024] Open
Abstract
BACKGROUND While coronary artery calcification (CAC) is recognized as a reliable marker for coronary atherosclerosis, the relationship between the concentration of C-reactive protein (CRP) and the incidence and progression of CAC remains controversial. METHOD PubMed, Embase, Web of Science, and Scopus were systematically searched to identify relevant observational studies until October 2023. The methodological quality of the included studies was evaluated using the Newcastle-Ottawa Scale (NOS). A random-effects meta-analysis was employed to calculate pooled odd ratios (OR) and corresponding 95% confidence intervals, considering heterogeneity among the studies. RESULTS Out of the 2545 records, 42 cross-sectional and 9 cohort studies were included in the systematic review. The meta-analysis on 12 eligible cross-sectional studies revealed no significant association between CAC and CRP [pooled OR: 1.03 (1.00, 1.06)]. Additionally, an insignificant association was found between CAC and CRP through meta-analysis on three eligible cohort studies [pooled OR: 1.05 (0.95, 1.15)] with no considerable heterogeneity across studies. Sensitivity analyses indicated that the meta-analysis models were robust. There was no evidence of publication bias. CONCLUSION Based on the meta-analysis findings, elevated levels of CRP did not emerge as a valuable prognostic maker for CAC incidence and progression prediction.
Collapse
Affiliation(s)
- Amirhossein Tajani
- Department of Clinical Pharmacy, School of Pharmacy, Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Masoumeh Sadeghi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Navid Omidkhoda
- Department of Clinical Pharmacy, School of Pharmacy, Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Hooshang Mohammadpour
- Department of Clinical Pharmacy, School of Pharmacy, Pharmaceutical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sara Samadi
- Department of Internal Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Vahid Jomehzadeh
- Department of Surgery, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
15
|
Zhang T, Chien RC, Budachetri K, Lin M, Boyaka P, Huang W, Rikihisa Y. Ehrlichia effector TRP120 manipulates bacteremia to facilitate tick acquisition. mBio 2024; 15:e0047624. [PMID: 38501870 PMCID: PMC11005420 DOI: 10.1128/mbio.00476-24] [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: 02/15/2024] [Accepted: 03/03/2024] [Indexed: 03/20/2024] Open
Abstract
Ehrlichia species are obligatory intracellular bacteria that cause a potentially fatal disease, human ehrlichiosis. The biomolecular mechanisms of tick acquisition of Ehrlichia and transmission between ticks and mammals are poorly understood. Ehrlichia japonica infection of mice recapitulates the full spectrum of human ehrlichiosis. We compared the pathogenicity and host acquisition of wild-type E. japonica with an isogenic transposon mutant of E. japonica that lacks tandem repeat protein 120 (TRP120) (ΔTRP120). Both wild-type and ΔTRP120 E. japonica proliferated similarly in cultures of mammalian and tick cells. Upon inoculation into mice, both wild-type and ΔTRP120 E. japonica multiplied to high levels in various tissues, with similar clinical chemistry and hematologic changes, proinflammatory cytokine induction, and fatal disease. However, the blood levels of ΔTRP120 E. japonica were almost undetectable within 24 h, whereas the levels of the wild type increased exponentially. Greater than 90% of TRP120 was released from infected cells into the culture medium. Mouse blood monocytes exposed to native TRP120 from culture supernatants showed significantly reduced cell surface expression of the transmigration-related markers Ly6C and CD11b. Larval ticks attached to mice infected with either wild-type or ΔTRP120 E. japonica imbibed similar amounts of blood and subsequently molted to nymphs at similar rates. However, unlike wild-type E. japonica, the ΔTRP120 mutant was minimally acquired by larval ticks and subsequent molted nymphs and, thus, failed to transmit to naïve mice. Thus, TRP120 is required for bacteremia but not disease. These findings suggest a novel mechanism whereby an obligatory intracellular bacterium manipulates infected blood monocytes to sustain the tick-mammal transmission cycle. IMPORTANCE Effective prevention of tick-borne diseases such as human ehrlichiosis requires an understanding of how disease-causing organisms are acquired. Ehrlichia species are intracellular bacteria that require infection of both mammals and ticks, involving cycles of transmission between them. Mouse models of ehrlichiosis and tick-mouse transmission can advance our fundamental understanding of the pathogenesis and prevention of ehrlichiosis. Herein, a mutant of Ehrlichia japonica was used to investigate the role of a single Ehrlichia factor, named tandem repeat protein 120 (TRP120), in infection of mammalian and tick cells in culture, infection and disease progression in mice, and tick acquisition of E. japonica from infected mice. Our results suggest that TRP120 is necessary only for Ehrlichia proliferation in circulating mouse blood and ongoing bacteremia to permit Ehrlichia acquisition by ticks. This study provides new insights into the importance of bacterial factors in regulating bacteremia, which may facilitate tick acquisition of pathogens.
Collapse
Affiliation(s)
- Tsian Zhang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Rory C. Chien
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Khemraj Budachetri
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Mingqun Lin
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Prosper Boyaka
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Weiyan Huang
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
16
|
Reus P, Guthmann H, Uhlig N, Agbaria M, Issmail L, Eberlein V, Nordling-David MM, Jbara-Agbaria D, Ciesek S, Bojkova D, Cinatl J, Burger-Kentischer A, Rupp S, Zaliani A, Grunwald T, Gribbon P, Kannt A, Golomb G. Drug repurposing for the treatment of COVID-19: Targeting nafamostat to the lungs by a liposomal delivery system. J Control Release 2023; 364:654-671. [PMID: 37939853 DOI: 10.1016/j.jconrel.2023.10.050] [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: 07/13/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Despite tremendous global efforts since the beginning of the COVID-19 pandemic, still only a limited number of prophylactic and therapeutic options are available. Although vaccination is the most effective measure in preventing morbidity and mortality, there is a need for safe and effective post-infection treatment medication. In this study, we explored a pipeline of 21 potential candidates, examined in the Calu-3 cell line for their antiviral efficacy, for drug repurposing. Ralimetinib and nafamostat, clinically used drugs, have emerged as attractive candidates. Due to the inherent limitations of the selected drugs, we formulated targeted liposomes suitable for both systemic and intranasal administration. Non-targeted and targeted nafamostat liposomes (LipNaf) decorated with an Apolipoprotein B peptide (ApoB-P) as a specific lung-targeting ligand were successfully developed. The developed liposomal formulations of nafamostat were found to possess favorable physicochemical properties including nano size (119-147 nm), long-term stability of the normally rapidly degrading compound in aqueous solution, negligible leakage from the liposomes upon storage, and a neutral surface charge with low polydispersity index (PDI). Both nafamostat and ralimetinib liposomes showed good cellular uptake and lack of cytotoxicity, and non-targeted LipNaf demonstrated enhanced accumulation in the lungs following intranasal (IN) administration in non-infected mice. LipNaf retained its anti-SARS-CoV 2 activity in Calu 3 cells with only a modest decrease, exhibiting complete inhibition at concentrations >100 nM. IN, but not intraperitoneal (IP) treatment with targeted LipNaf resulted in a trend to reduced viral load in the lungs of K18-hACE2 mice compared to targeted empty Lip. Nevertheless, upon removal of outlier data, a statistically significant 1.9-fold reduction in viral load was achieved. This observation further highlights the importance of a targeted delivery into the respiratory tract. In summary, we were able to demonstrate a proof-of-concept of drug repurposing by liposomal formulations with anti-SARS-CoV-2 activity. The biodistribution and bioactivity studies with LipNaf suggest an IN or inhalation route of administration for optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Philipp Reus
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany; Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Paul-Ehrlich-Straße 40, 60596 Frankfurt am Main, Germany
| | - Hadar Guthmann
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy und Immunology IZI, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Majd Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy und Immunology IZI, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy und Immunology IZI, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Mirjam M Nordling-David
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Doaa Jbara-Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Sandra Ciesek
- Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Paul-Ehrlich-Straße 40, 60596 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Denisa Bojkova
- Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Paul-Ehrlich-Straße 40, 60596 Frankfurt am Main, Germany
| | - Jindrich Cinatl
- Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Paul-Ehrlich-Straße 40, 60596 Frankfurt am Main, Germany
| | - Anke Burger-Kentischer
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Steffen Rupp
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy und Immunology IZI, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Aimo Kannt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Fraunhofer Innovation Center TheraNova, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Institute for Clinical Pharmacology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
17
|
Corewyn LC, Kelaita MA, Nollman J, Hagnauer I, Blanco-Peña K, Lessnau RG, Clayton JB, Shields-Cutler R, Stoos KB. Hematology and blood biochemistry in a declining population of mantled howler monkeys (Alouatta palliata palliata) at La Pacifica, Costa Rica. J Med Primatol 2023; 52:353-360. [PMID: 37655719 PMCID: PMC10841258 DOI: 10.1111/jmp.12669] [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: 02/13/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Alouatta palliata palliata are an ecologically flexible howler monkey subspecies that has recently been relisted as Endangered. Populations are declining through much of the subspecies' range, including at our study site at La Pacifica, Costa Rica. Our objectives were to screen blood hematology and biochemistry samples collected from this wild population to elucidate their baseline health. METHODS We collected blood samples from 38 adult individuals from across the study site and analyzed 13 hematology and 14 biochemistry parameters. RESULTS Most hematology and blood biochemistry parameter values were similar between males and females. However, mean hemoglobin was significantly lower, and mean white blood cell count was significantly higher in females; and mean calcium and mean creatinine were significantly lower in females compared to males. CONCLUSIONS Overall, the La Pacifica population appeared healthy based on the blood parameters analyzed from sampled individuals. Our results were also largely consistent with published data available from other populations of A. p. palliata, and with reference values for captive Alouatta caraya.
Collapse
Affiliation(s)
- Lisa C Corewyn
- Department of Biology, Ithaca College, Ithaca, New York, USA
| | - Mary A Kelaita
- Department of Natural Sciences, St. Philip's College, San Antonio, Texas, USA
| | - Jenny Nollman
- The Cincinnati Zoo & Botanical Garden, Cincinnati, Ohio, USA
| | | | - Kinndle Blanco-Peña
- Instituto Regional de Estudios en Sustancias Tóxicas (IRET), Universidad Nacional, Costa Rica, Heredia, Costa Rica
| | | | - Jonathan B Clayton
- Department of Biology, University of Nebraska Omaha, Nebraska, Omaha, USA
- Nebraska Food for Health Center, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
- Primate Microbiome Project, University of Nebraska-Lincoln, Lincoln, Nebraska, USA
| | | | - Kari Brossard Stoos
- Department of Health Sciences & Public Health, Ithaca College, Ithaca, New York, USA
| |
Collapse
|
18
|
Kare AJ, Nichols L, Zermeno R, Raie MN, Tumbale SK, Ferrara KW. OMIP-095: 40-Color spectral flow cytometry delineates all major leukocyte populations in murine lymphoid tissues. Cytometry A 2023; 103:839-850. [PMID: 37768325 PMCID: PMC10843696 DOI: 10.1002/cyto.a.24788] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 07/26/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023]
Abstract
High-dimensional immunoprofiling is essential for studying host response to immunotherapy, infection, and disease in murine model systems. However, the difficulty of multiparameter panel design combined with a lack of existing murine tools has prevented the comprehensive study of all major leukocyte phenotypes in a single assay. Herein, we present a 40-color flow cytometry panel for deep immunophenotyping of murine lymphoid tissues, including the spleen, blood, Peyer's patches, inguinal lymph nodes, bone marrow, and thymus. This panel uses a robust set of surface markers capable of differentiating leukocyte subsets without the use of intracellular staining, thus allowing for the use of cells in downstream functional experiments or multiomic analyses. Our panel classifies T cells, B cells, natural killer cells, innate lymphoid cells, monocytes, macrophages, dendritic cells, basophils, neutrophils, eosinophils, progenitors, and their functional subsets by using a series of co-stimulatory, checkpoint, activation, migration, and maturation markers. This tool has a multitude of systems immunology applications ranging from serial monitoring of circulating blood signatures to complex endpoint analysis, especially in pre-clinical settings where treatments can modulate leukocyte abundance and/or function. Ultimately, this 40-color panel resolves a diverse array of immune cells on the axes of time, tissue, and treatment, filling the niche for a modern tool dedicated to murine immunophenotyping.
Collapse
Affiliation(s)
- Aris J. Kare
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Lisa Nichols
- Stanford Shared FACS Facility, Stanford University, Stanford, CA 94305, USA
| | - Ricardo Zermeno
- Stanford Shared FACS Facility, Stanford University, Stanford, CA 94305, USA
| | - Marina N. Raie
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | | | | |
Collapse
|
19
|
Hernández-Barrientos D, Pelayo R, Mayani H. The hematopoietic microenvironment: a network of niches for the development of all blood cell lineages. J Leukoc Biol 2023; 114:404-420. [PMID: 37386890 DOI: 10.1093/jleuko/qiad075] [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: 03/07/2023] [Revised: 05/25/2023] [Accepted: 06/15/2023] [Indexed: 07/01/2023] Open
Abstract
Blood cell formation (hematopoiesis) takes place mainly in the bone marrow, within the hematopoietic microenvironment, composed of a number of different cell types and their molecular products that together shape spatially organized and highly specialized microstructures called hematopoietic niches. From the earliest developmental stages and throughout the myeloid and lymphoid lineage differentiation pathways, hematopoietic niches play a crucial role in the preservation of cellular integrity and the regulation of proliferation and differentiation rates. Current evidence suggests that each blood cell lineage develops under specific, discrete niches that support committed progenitor and precursor cells and potentially cooperate with transcriptional programs determining the gradual lineage commitment and specification. This review aims to discuss recent advances on the cellular identity and structural organization of lymphoid, granulocytic, monocytic, megakaryocytic, and erythroid niches throughout the hematopoietic microenvironment and the mechanisms by which they interconnect and regulate viability, maintenance, maturation, and function of the developing blood cells.
Collapse
Affiliation(s)
- Daniel Hernández-Barrientos
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Av. Cuauhtemoc 330. Mexico City, 06720, Mexico
| | - Rosana Pelayo
- Onco-Immunology Laboratory, Eastern Biomedical Research Center, IMSS, Km 4.5 Atlixco-Metepec, 74360, Puebla, Mexico
| | - Hector Mayani
- Hematopoietic Stem Cells Laboratory, Oncology Research Unit, Oncology Hospital, National Medical Center, IMSS, Av. Cuauhtemoc 330. Mexico City, 06720, Mexico
| |
Collapse
|
20
|
Glorion M, Pascale F, Huriet M, Estephan J, Gouin C, Urien C, Bourge M, Egidy G, Richard C, Gelin V, De Wolf J, Le Guen M, Magnan A, Roux A, Devillier P, Schwartz-Cornil I, Sage E. Differential early response of monocyte/macrophage subsets to intra-operative corticosteroid administration in lung transplantation. Front Immunol 2023; 14:1281546. [PMID: 37942330 PMCID: PMC10628533 DOI: 10.3389/fimmu.2023.1281546] [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: 08/22/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction Lung transplantation often results in primary and/or chronic dysfunctions that are related to early perioperative innate allo-responses where myeloid subsets play a major role. Corticosteroids are administered upon surgery as a standard-of-care but their action on the different myeloid cell subsets in that context is not known. Methods To address this issue, we used a cross-circulatory platform perfusing an extracorporeal lung coupled to cell mapping in the pig model, that enabled us to study the recruited cells in the allogeneic lung over 10 hours. Results Myeloid cells, i.e. granulocytes and monocytic cells including classical CD14pos and non-classical/intermediate CD16pos cells, were the dominantly recruited subsets, with the latter upregulating the membrane expression of MHC class II and CD80/86 molecules. Whereas corticosteroids did not reduce the different cell subset recruitment, they potently dampened the MHC class II and CD80/86 expression on monocytic cells and not on alveolar macrophages. Besides, corticosteroids induced a temporary and partial anti-inflammatory gene profile depending on cytokines and monocyte/macrophage subsets. Discussion This work documents the baseline effects of the standard-of-care corticosteroid treatment for early innate allo-responses. These insights will enable further optimization and improvement of lung transplantation outcomes.
Collapse
Affiliation(s)
- Matthieu Glorion
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, Suresnes, France
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Florentina Pascale
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, Suresnes, France
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Maxime Huriet
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Jérôme Estephan
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Carla Gouin
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Céline Urien
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| | - Mickael Bourge
- Cytometry/Electronic Microscopy/Light Microcopy Facility, Imagerie-Gif, Université Paris-Saclay, CEA, CNRS, Institute for Integrative Biology of the Cell (I2BC), Gif-sur-Yvette, France
| | - Giorgia Egidy
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, Jouy-en-Josas, France
| | | | - Valérie Gelin
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
| | - Julien De Wolf
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, Suresnes, France
| | - Morgan Le Guen
- Department of Anesthesiology, Foch Hospital, Suresnes, France
| | - Antoine Magnan
- Department of Pulmonology, Foch Hospital, Suresnes, France
| | - Antoine Roux
- Department of Pulmonology, Foch Hospital, Suresnes, France
| | - Philippe Devillier
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
- Respiratory Pharmacology Research Unit - Exhalomics, Foch Hospital, Suresnes, France
| | | | - Edouard Sage
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, Suresnes, France
- Université Paris-Saclay, INRAE, UVSQ, VIM, Jouy-en-Josas, France
| |
Collapse
|
21
|
Lewis SA, Doratt BM, Qiao Q, Blanton M, Grant KA, Messaoudi I. Integrated single cell analysis shows chronic alcohol drinking disrupts monocyte differentiation in the bone marrow. Stem Cell Reports 2023; 18:1884-1897. [PMID: 37657446 PMCID: PMC10545484 DOI: 10.1016/j.stemcr.2023.08.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/01/2023] [Accepted: 08/01/2023] [Indexed: 09/03/2023] Open
Abstract
Chronic heavy alcohol drinking (CHD) rewires monocytes and macrophages toward heightened inflammatory states with compromised antimicrobial defenses that persist after 1-month abstinence. To determine whether these changes are mediated through alterations in the bone marrow niche, we profiled monocytes and hematopoietic stem cell progenitors (HSCPs) from CHD rhesus macaques using a combination of functional assays and single cell genomics. CHD resulted in transcriptional profiles consistent with increased activation and inflammation within bone marrow resident monocytes and macrophages. Furthermore, CHD resulted in transcriptional signatures associated with increased oxidative and cellular stress in HSCP. Differentiation of HSCP in vitro revealed skewing toward monocytes expressing "neutrophil-like" markers with greater inflammatory responses to bacterial agonists. Further analyses of HSCPs showed broad epigenetic changes that were in line with exacerbated inflammatory responses within monocytes and their progenitors. In summary, CHD alters HSCPs in the bone marrow leading to the production of monocytes poised to generate dysregulated hyper-inflammatory responses.
Collapse
Affiliation(s)
- Sloan A Lewis
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA
| | - Brianna M Doratt
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Qi Qiao
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Madison Blanton
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Kathleen A Grant
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine, Irvine, CA 92697, USA; Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
22
|
Ruder AV, Wetzels SMW, Temmerman L, Biessen EAL, Goossens P. Monocyte heterogeneity in cardiovascular disease. Cardiovasc Res 2023; 119:2033-2045. [PMID: 37161473 PMCID: PMC10478755 DOI: 10.1093/cvr/cvad069] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 02/07/2023] [Accepted: 02/21/2023] [Indexed: 05/11/2023] Open
Abstract
Monocytes circulate the vasculature at steady state and are recruited to sites of inflammation where they differentiate into macrophages (MФ) to replenish tissue-resident MФ populations and engage in the development of cardiovascular disease (CVD). Monocytes display considerable heterogeneity, currently reflected by a nomenclature based on their expression of cluster of differentiation (CD) 14 and CD16, distinguishing CD14++CD16- classical (cMo), CD14++CD16+ intermediate (intMo) and CD14+CD16++ non-classical (ncMo) monocytes. Several reports point to shifted subset distributions in the context of CVD, with significant association of intMo numbers with atherosclerosis, myocardial infarction, and heart failure. However, clear indications of their causal involvement as well as their predictive value for CVD are lacking. As recent high-parameter cytometry and single-cell RNA sequencing (scRNA-Seq) studies suggest an even higher degree of heterogeneity, better understanding of the functionalities of these subsets is pivotal. Considering their high heterogeneity, surprisingly little is known about functional differences between MФ originating from monocytes belonging to different subsets, and implications thereof for CVD pathogenesis. This paper provides an overview of recent findings on monocyte heterogeneity in the context of homeostasis and disease as well as functional differences between the subsets and their potential to differentiate into MФ, focusing on their role in vessels and the heart. The emerging paradigm of monocyte heterogeneity transcending the current tripartite subset division argues for an updated nomenclature and functional studies to substantiate marker-based subdivision and to clarify subset-specific implications for CVD.
Collapse
Affiliation(s)
- Adele V Ruder
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Suzan M W Wetzels
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Lieve Temmerman
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| | - Erik A L Biessen
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
- Institute for Molecular Cardiovascular Research, RWTH Aachen University, Pauwelsstraße 30, 52074 Aachen, Germany
| | - Pieter Goossens
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC+), P. Debyelaan 25, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
23
|
Grandoni F, Hussen J, Signorelli F, Napolitano F, Scatà MC, De Donato I, Cappelli G, Galiero G, Grassi C, De Carlo E, Petrini S, De Matteis G, Martucciello A. Evaluation of Hematological Profiles and Monocyte Subpopulations in Water Buffalo Calves after Immunization with Two Different IBR Marker Vaccines and Subsequent Infection with Bubaline alphaherpesvirus-1. Vaccines (Basel) 2023; 11:1405. [PMID: 37766082 PMCID: PMC10537172 DOI: 10.3390/vaccines11091405] [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: 06/29/2023] [Revised: 08/16/2023] [Accepted: 08/18/2023] [Indexed: 09/29/2023] Open
Abstract
Bubaline alphaherpesvirus-1 (BuAHV-1) and Bovine alphaherpesvirus-1 (BoAHV-1) are respiratory viruses that can cause an infection known as "Infectious Bovine Rhinotracheitis" (IBR) in both water buffalo and bovine species. As the main disease control strategy, vaccination can protect animals from clinical disease through the development of specific humoral and cell-mediated immune responses. In the present study, the time-related circulatory kinetics of hematological profile and bubaline monocyte subsets have been investigated in vaccinated buffalo calves after challenge infections with BuAHV-1. Thirteen buffalo calves were selected and grouped into the VAX-1 group, which received an IBR-live-attenuated gE-/tk-deleted marker vaccine; the VAX-2 group, which received an IBR-inactivated gE-deleted marker vaccine; the CNT group, which remained an unvaccinated control. Fifty-five days after the first vaccination, the animals were infected with 5 × 105.00 TCID50/mL of wild-type BuAHV-1 strain via the intranasal route. Whole blood samples were collected at 0, 2, 4, 7, 10, 15, 30, and 63 days post-challenge (PCDs) for the analysis of hematological profiles and the enumeration of monocyte subsets via flow cytometry. The analysis of leukocyte compositions revealed that neutrophils were the main leukocyte population, with a relative increase during the acute infection. On the other hand, a general decrease in the proportion of lymphocytes was observed early in the post-infection, both for the VAX-1 and VAX-2 groups, while in the CNT group, the decrease was observed later at +30 and +63 PCDs. An overall infection-induced increase in blood total monocytes was observed in all groups. The rise was especially marked in the animals vaccinated with an IBR-live-attenuated gE-/tK-deleted marker vaccine (VAX-1 group). A multicolor flow cytometry panel was used to identify the bubaline monocyte subpopulations (classical = cM; intermediate = intM; and non-classical = ncM) and to investigate their variations during BuAHV-1 infection. Our results showed an early increase in cMs followed by a second wave of intMs. This increase was observed mainly after stimulation with live-attenuated viruses in the VAX-1 group compared with the animals vaccinated with the inactivated vaccine or the non-vaccinated animal group. In summary, the present study characterized, for the first time, the hematological profile and distribution of blood monocyte subsets in vaccinated and non-vaccinated water buffalo in response to experimental infection with BuAHV-1. Although not experimentally proven, our results support the hypothesis of a linear developmental relationship between monocyte subsets.
Collapse
Affiliation(s)
- Francesco Grandoni
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Jamal Hussen
- Department of Microbiology, College of Veterinary Medicine, King Faisal University, Al-Ahsa 36362, Saudi Arabia
| | - Federica Signorelli
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Francesco Napolitano
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Maria Carmela Scatà
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Immacolata De Donato
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Giovanna Cappelli
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Giorgio Galiero
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Carlo Grassi
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Esterina De Carlo
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| | - Stefano Petrini
- National Reference Centre for Infectious Bovine Rhinotracheitis (IBR), Istituto Zooprofilattico Sperimentale Umbria-Marche, “Togo Rosati”, 06126 Perugia, Italy
| | - Giovanna De Matteis
- Research Centre for Animal Production and Aquaculture, Consiglio per la Ricerca in Agricoltura e l’Analisi dell’Economia Agraria (CREA), 00015 Monterotondo, Italy
| | - Alessandra Martucciello
- National Reference Centre for Hygiene and Technologies of Water Buffalo Farming and Productions, Istituto Zooprofilattico Sperimentale del Mezzogiorno, 84132 Salerno, Italy
| |
Collapse
|
24
|
Kim Y, Kamada N. The role of the microbiota in myelopoiesis during homeostasis and inflammation. Int Immunol 2023; 35:267-274. [PMID: 36694400 PMCID: PMC10199171 DOI: 10.1093/intimm/dxad002] [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: 10/10/2022] [Accepted: 01/23/2023] [Indexed: 01/26/2023] Open
Abstract
The microbiota engages in the development and maintenance of the host immune system. The microbiota affects not only mucosal tissues where it localizes but also the distal organs. Myeloid cells are essential for host defense as first responders of the host immune system. Their generation, called myelopoiesis, is regulated by environmental signals, including commensal microbiota. Hematopoietic stem and progenitor cells in bone marrow can directly or indirectly sense microbiota-derived signals, thereby giving rise to myeloid cell lineages at steady-state and during inflammation. In this review, we discuss the role of commensal microorganisms in the homeostatic regulation of myelopoiesis in the bone marrow. We also outline the effects of microbial signals on myelopoiesis during inflammation and infection, with a particular focus on the development of innate immune memory. Studying the relationship between the microbiota and myelopoiesis will help us understand how the microbiota regulates immune responses at a systemic level beyond the local mucosa.
Collapse
Affiliation(s)
- Yeji Kim
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nobuhiko Kamada
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Laboratory of Microbiology and Immunology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
25
|
Álvarez D, Morales-Prieto DM, Cadavid ÁP. Interaction between endothelial cell-derived extracellular vesicles and monocytes: A potential link between vascular thrombosis and pregnancy-related morbidity in antiphospholipid syndrome. Autoimmun Rev 2023; 22:103274. [PMID: 36649876 DOI: 10.1016/j.autrev.2023.103274] [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: 12/23/2022] [Accepted: 01/12/2023] [Indexed: 01/15/2023]
Abstract
Antiphospholipid syndrome (APS) is an autoimmune disease driven by a wide group of autoantibodies primarily directed against phospholipid-binding proteins (antiphospholipid antibodies). APS is defined by two main kinds of clinical manifestations: vascular thrombosis and pregnancy-related morbidity. In recent years, in vitro and in vivo assays, as well as the study of large groups of patients with APS, have led some authors to suggest that obstetric and vascular manifestations of the disease are probably the result of different pathogenic mechanisms. According to this hypothesis, the disease could be differentiated into two parallel entities: Vascular APS and obstetric APS. Thus, vascular APS is understood as an acquired thrombophilia in which a generalised phenomenon of endothelial activation and dysfunction (coupled with a triggering factor) causes thrombosis at any location. In contrast, obstetric APS seems to be due to an inflammatory phenomenon accompanied by trophoblast cell dysfunction. The recent approach to APS raises new issues; for instance, the mechanisms by which a single set of autoantibodies can lead to two different clinical entities are unclear. This review will address the monocyte, a cell with well-known roles in haemostasis and pregnancy, as a potential participant in vascular thrombosis and pregnancy-related morbidity in APS. We will discuss how in a steady state the monocyte-endothelial interaction occurs via extracellular vesicles (EVs), and how antiphospholipid antibodies, by inducing endothelial activation and dysfunction, may disturb this interaction to promote the release of monocyte-targeted procoagulant and inflammatory messages.
Collapse
Affiliation(s)
- Daniel Álvarez
- Grupo Reproducción, Departamento Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia.
| | - Diana M Morales-Prieto
- Placenta Lab, Department of Obstetrics, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Ángela P Cadavid
- Grupo Reproducción, Departamento Microbiología y Parasitología, Facultad de Medicina, Universidad de Antioquia UdeA, Calle 70 No. 52-21, Medellín, Colombia; Grupo de Investigación en Trombosis, Departamento Medicina Interna, Facultad de Medicina, Universidad de Antioquia UdeA, Medellín, Colombia.
| |
Collapse
|
26
|
Lewis SA, Doratt BM, Qiao Q, Blanton MB, Grant KA, Messaoudi I. Integrated single cell analysis shows chronic alcohol drinking disrupts monocyte differentiation in the bone marrow niche. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.29.534727. [PMID: 37034734 PMCID: PMC10081177 DOI: 10.1101/2023.03.29.534727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Chronic alcohol drinking rewires circulating monocytes and tissue-resident macrophages towards heightened inflammatory states with compromised anti-microbial defenses. As these effects remain consistent in short-lived monocytes after a 1-month abstinence period it is unclear whether these changes are restricted to the periphery or mediated through alterations in the progenitor niche. To test this hypothesis, we profiled monocytes/macrophages and hematopoietic stem cell progenitors (HSCP) of the bone marrow compartment from rhesus macaques after 12 months of ethanol consumption using a combination of functional assays and single cell genomics. Bone marrow-resident monocytes/macrophages from ethanol-consuming animals exhibited heightened inflammation. Differentiation of HSCP in vitro revealed skewing towards monocytes expressing neutrophil-like markers with heightened inflammatory responses to bacterial agonists. Single cell transcriptional analysis of HSCPs showed reduced proliferation but increased inflammatory markers in mature myeloid progenitors. We observed transcriptional signatures associated with increased oxidative and cellular stress as well as oxidative phosphorylation in immature and mature myeloid progenitors. Single cell analysis of the chromatin landscape showed altered drivers of differentiation in monocytes and progenitors. Collectively, these data indicate that chronic ethanol drinking results in remodeling of the transcriptional and epigenetic landscapes of the bone marrow compartment leading to altered functions in the periphery.
Collapse
Affiliation(s)
- Sloan A. Lewis
- Department of Molecular Biology and Biochemistry, University of California, Irvine CA 92697, USA
| | - Brianna M Doratt
- Department of Molecular Biology and Biochemistry, University of California, Irvine CA 92697, USA
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Qi Qiao
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Madison B. Blanton
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536
| | - Kathleen A. Grant
- Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Ilhem Messaoudi
- Department of Molecular Biology and Biochemistry, University of California, Irvine CA 92697, USA
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY 40536
| |
Collapse
|
27
|
Hanč P, Messou MA, Wang Y, von Andrian UH. Control of myeloid cell functions by nociceptors. Front Immunol 2023; 14:1127571. [PMID: 37006298 PMCID: PMC10064072 DOI: 10.3389/fimmu.2023.1127571] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 02/23/2023] [Indexed: 03/19/2023] Open
Abstract
The immune system has evolved to protect the host from infectious agents, parasites, and tumor growth, and to ensure the maintenance of homeostasis. Similarly, the primary function of the somatosensory branch of the peripheral nervous system is to collect and interpret sensory information about the environment, allowing the organism to react to or avoid situations that could otherwise have deleterious effects. Consequently, a teleological argument can be made that it is of advantage for the two systems to cooperate and form an “integrated defense system” that benefits from the unique strengths of both subsystems. Indeed, nociceptors, sensory neurons that detect noxious stimuli and elicit the sensation of pain or itch, exhibit potent immunomodulatory capabilities. Depending on the context and the cellular identity of their communication partners, nociceptors can play both pro- or anti-inflammatory roles, promote tissue repair or aggravate inflammatory damage, improve resistance to pathogens or impair their clearance. In light of such variability, it is not surprising that the full extent of interactions between nociceptors and the immune system remains to be established. Nonetheless, the field of peripheral neuroimmunology is advancing at a rapid pace, and general rules that appear to govern the outcomes of such neuroimmune interactions are beginning to emerge. Thus, in this review, we summarize our current understanding of the interaction between nociceptors and, specifically, the myeloid cells of the innate immune system, while pointing out some of the outstanding questions and unresolved controversies in the field. We focus on such interactions within the densely innervated barrier tissues, which can serve as points of entry for infectious agents and, where known, highlight the molecular mechanisms underlying these interactions.
Collapse
Affiliation(s)
- Pavel Hanč
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- *Correspondence: Pavel Hanč, ; Ulrich H. von Andrian,
| | - Marie-Angèle Messou
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Yidi Wang
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
| | - Ulrich H. von Andrian
- Department of Immunology, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of Massachusetts General Hospital (MGH), Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, United States
- *Correspondence: Pavel Hanč, ; Ulrich H. von Andrian,
| |
Collapse
|
28
|
Shao L, Wu B, Liu C, Chong W. VALPROIC ACID INHIBITS CLASSICAL MONOCYTE-DERIVED TISSUE FACTOR AND ALLEVIATES HEMORRHAGIC SHOCK-INDUCED ACUTE LUNG INJURY IN RATS. Shock 2023; 59:449-459. [PMID: 36443067 PMCID: PMC9997640 DOI: 10.1097/shk.0000000000002064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 09/27/2022] [Accepted: 11/22/2022] [Indexed: 11/30/2022]
Abstract
ABSTRACT Background: Monocytes and monocyte-derived tissue factor (TF) promote the development of sepsis-induced acute lung injury (ALI). Classical monocytes (C-Mcs) can be induced to express TF. Valproic acid (VPA) alleviates hemorrhagic shock (HS)-induced ALI (HS/ALI) and inhibits TF expression in monocytes. We hypothesized that C-Mcs and C-Mc-derived TF promoted HS/ALI and that VPA could inhibit C-Mc-derived TF expression and attenuate HS/ALI. Methods: Wistar rats and THP-1 cells were used to evaluate our hypothesis. Monocyte subtypes were analyzed by flow cytometry; mRNA expression was measured by fluorescence quantitative polymerase chain reaction; protein expression was measured by Western blotting, immunofluorescence, or immunohistology; inflammatory cytokines levels were measured by enzyme-linked immunosorbent assay; and ALI scores were used to determine the degree of ALI. Results: The blood %C-Mcs and C-Mcs/non-C-Mcs ratios, monocyte TF levels, serum and/or lung inflammatory cytokine levels, and ALI scores of HS rats were significantly increased ( P < 0.05). After monocyte depletion and thrombin inhibition, the inflammatory cytokine levels and ALI scores were significantly decreased ( P < 0.05). VPA reduced the %C-Mcs and C-Mc/non-C-Mc ratios, TF expression, inflammatory cytokine levels, and ALI scores during HS ( P < 0.05) and inhibited HS-induced monocyte Egr-1 and p-ERK1/2 expression ( P < 0.05). VPA inhibited hypoxia-induced TF expression in THP-1 cells by regulating the p-ERK1/2-Egr-1 axis. Conclusion: C-Mcs and C-Mc-derived TF accelerate the development of HS/ALI by increasing thrombin production. VPA inhibits HS-induced C-Mc production of TF by regulating the p-ERK1/2-Egr-1 axis and alleviates HS/ALI.
Collapse
Affiliation(s)
- Lina Shao
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
- Intensive Care Unit, Liaoning Cancer Hospital & Institute, Shenyang, Liaoning Province, China
- Intensive Care Unit, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province, China
- Intensive Care Unit, Cancer Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Bing Wu
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Chang Liu
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| | - Wei Chong
- Emergency Department, the First Hospital of China Medical University, Shenyang, Liaoning Province, China
| |
Collapse
|
29
|
Guglietta S, Krieg C. Phenotypic and functional heterogeneity of monocytes in health and cancer in the era of high dimensional technologies. Blood Rev 2023; 58:101012. [PMID: 36114066 DOI: 10.1016/j.blre.2022.101012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/23/2022]
Abstract
Monocytes have been traditionally classified in three discrete subsets, which can participate in the immune responses as effector cells or as precursors of myeloid-derived cells in circulation and tissues. However, recent advances in single-cell omics have revealed unprecedented phenotypic and functional heterogeneity that goes well beyond the three conventional monocytic subsets and propose a more fluid differentiation model. This novel concept does not only apply to the monocytes in circulation but also at the tissue site. Consequently, the binary model proposed for differentiating monocyte into M1 and M2 macrophages has been recently challenged by a spectrum model that more realistically mirrors the heterogeneous cues in inflammatory conditions. This review describes the latest results on the high dimensional characterization of monocytes and monocyte-derived myeloid cells in steady state and cancer. We discuss how environmental cues and monocyte-intrinsic properties may affect their differentiation toward specific functional and phenotypic subsets, the causes of monocyte expansion and reduction in cancer, their metabolic requirements, and the potential effect on tumor immunity.
Collapse
Affiliation(s)
- Silvia Guglietta
- Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA; Department of Regenerative Medicine & Cell Biology, Medical University of South Carolina (MUSC), 173 Ashley Avenue, CRI609, Charleston, SC 29425, USA.
| | - Carsten Krieg
- Department of Pathology & Laboratory Medicine, Medical University of South Carolina (MUSC), 68 President Street, BE415, Charleston, SC 29425, USA; Hollings Cancer Center, 86 Jonathan Lucas Street, Charleston, SC 29425, USA.
| |
Collapse
|
30
|
Reynoso GV, Gordon DN, Kalia A, Aguilar CC, Malo CS, Aleshnick M, Dowd KA, Cherry CR, Shannon JP, Vrba SM, Holmes AC, Alippe Y, Maciejewski S, Asano K, Diamond MS, Pierson TC, Hickman HD. Zika virus spreads through infection of lymph node-resident macrophages. Cell Rep 2023; 42:112126. [PMID: 36795561 PMCID: PMC10425566 DOI: 10.1016/j.celrep.2023.112126] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 01/03/2023] [Accepted: 02/01/2023] [Indexed: 02/17/2023] Open
Abstract
To disseminate through the body, Zika virus (ZIKV) is thought to exploit the mobility of myeloid cells, in particular monocytes and dendritic cells. However, the timing and mechanisms underlying shuttling of the virus by immune cells remains unclear. To understand the early steps in ZIKV transit from the skin, at different time points, we spatially mapped ZIKV infection in lymph nodes (LNs), an intermediary site en route to the blood. Contrary to prevailing hypotheses, migratory immune cells are not required for the virus to reach the LNs or blood. Instead, ZIKV rapidly infects a subset of sessile CD169+ macrophages in the LNs, which release the virus to infect downstream LNs. Infection of CD169+ macrophages alone is sufficient to initiate viremia. Overall, our experiments indicate that macrophages that reside in the LNs contribute to initial ZIKV spread. These studies enhance our understanding of ZIKV dissemination and identify another anatomical site for potential antiviral intervention.
Collapse
Affiliation(s)
- Glennys V Reynoso
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - David N Gordon
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Anurag Kalia
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Cynthia C Aguilar
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Courtney S Malo
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Maya Aleshnick
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Kimberly A Dowd
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Christian R Cherry
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - John P Shannon
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Sophia M Vrba
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Autumn C Holmes
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Yael Alippe
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Sonia Maciejewski
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Kenichi Asano
- Laboratory of Immune Regulation, School of Life Science, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Michael S Diamond
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA; The Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Theodore C Pierson
- Viral Pathogenesis Section, Laboratory of Viral Diseases (LVD), NIAID, NIH, Bethesda, MD, USA
| | - Heather D Hickman
- Viral Immunity and Pathogenesis Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
31
|
Analysis of Monocyte Recruitment During Inflammation by Intravital Imaging. Methods Mol Biol 2023; 2608:451-467. [PMID: 36653722 DOI: 10.1007/978-1-0716-2887-4_25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Monocytes play essential roles in the inflammatory and anti-inflammatory processes that take place during an immune response, acting both within the vascular network and interstitially. Monocytes are activated, mobilized, and recruited in response to an inflammatory stimulus or different forms of tissue injury. The recruitment of circulating monocytes to the inflamed tissue is essential to resolving the injury.Monocyte recruitment is a multistep process that begins with a decrease in rolling velocity, is followed by adhesion to the endothelium and crawling over the luminal vessel surface, and culminates in monocyte transmigration into the surrounding tissue. Intravital microscopy is a powerful visualization tool for the study of leukocyte behavior and function, intercellular interactions, cell trafficking, and recruitment in pathological and physiological conditions. This modality is therefore widely used for the detailed analysis of the immune response to multiple insults and the molecular mechanisms underlying monocyte interactions within the vascular system in vivo. This chapter describes a protocol for the use of intravital microscopy to analyze monocyte recruitment from the blood vessel to the inflammatory site.
Collapse
|
32
|
NGF and Its Role in Immunoendocrine Communication during Metabolic Syndrome. Int J Mol Sci 2023; 24:ijms24031957. [PMID: 36768281 PMCID: PMC9916855 DOI: 10.3390/ijms24031957] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 12/18/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Nerve growth factor (NGF) was the first neurotrophin described. This neurotrophin contributes to organogenesis by promoting sensory innervation and angiogenesis in the endocrine and immune systems. Neuronal and non-neuronal cells produce and secrete NGF, and several cell types throughout the body express the high-affinity neurotrophin receptor TrkA and the low-affinity receptor p75NTR. NGF is essential for glucose-stimulated insulin secretion and the complete development of pancreatic islets. Plus, this factor is involved in regulating lipolysis and thermogenesis in adipose tissue. Immune cells produce and respond to NGF, modulating their inflammatory phenotype and the secretion of cytokines, contributing to insulin resistance and metabolic homeostasis. This neurotrophin regulates the synthesis of gonadal steroid hormones, which ultimately participate in the metabolic homeostasis of other tissues. Therefore, we propose that this neurotrophin's imbalance in concentrations and signaling during metabolic syndrome contribute to its pathophysiology. In the present work, we describe the multiple roles of NGF in immunoendocrine organs that are important in metabolic homeostasis and related to the pathophysiology of metabolic syndrome.
Collapse
|
33
|
Abstract
Macrophages have been recognized as the primary mediators of innate immunity starting from embryonic/fetal development. Macrophage-mediated defenses may not be as antigen-specific as adaptive immunity, but increasing information suggests that these responses do strengthen with repeated immunological triggers. The concept of innate memory in macrophages has been described as "trained immunity" or "innate immune memory (IIM)." As currently understood, this cellular memory is rooted in epigenetic and metabolic reprogramming. The recognition of IIM may be particularly important in the fetus and the young neonate who are yet to develop protective levels of adaptive immunity, and could even be of preventive/therapeutic importance in many disorders. There may also be a possibility of therapeutic enhancement with targeted vaccination. This article presents a review of the properties, mechanisms, and possible clinical significance of macrophage-mediated IIM.
Collapse
Affiliation(s)
- Akhil Maheshwari
- Founding Chairman, Global Newborn Society, Clarksville, Maryland, United States of America
| |
Collapse
|
34
|
Ahmadi J, Hosseini E, Kargar F, Ghasemzadeh M. Stable CAD patients show higher levels of platelet-borne TGF-β1 associated with a superior pro-inflammatory state than the pro-aggregatory status; Evidence highlighting the importance of platelet-derived TGF-β1 in atherosclerosis. J Thromb Thrombolysis 2023; 55:102-115. [PMID: 36352058 DOI: 10.1007/s11239-022-02729-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/28/2022] [Indexed: 11/10/2022]
Abstract
Activated platelets are involved in the atherogenic stage of atherosclerosis, while they can also progress it to atherothrombosis which may cause an ischemic state and organ failure. In general, coronary artery disease (CAD) is considered as common and severe clinical consequence of atherosclerosis, manifesting as a chronic inflammatory condition with the release of platelet mediators, among which the importance of platelet-borne TGF-β1 is not yet well understood. Hence, for the first time, this study aimed to examine platelet level of TGF-β1 (latent/mature) in CAD-patients and its association with the expression of platelet pro-inflammatory molecules. Platelet from stable CAD-patients candidate for CABG and healthy controls were subjected to flowcytometry analysis to evaluate P-selectin and CD40L expressions and PAC-1 binding. Platelet-borne and soluble TGF-β1, both mature/active and latent forms were also examined with western blotting. Higher expression levels of P-selectin and CD40L in patients with CAD than in controls were associated with comparable levels of PAC-1 binding in both groups. Platelet TGF-β1 levels were also significantly higher in patients, while their platelets showed clear bands of mature TGF-β1 that were barely visible in healthy individuals. Soluble TGF-β1 was also higher in patients. Significant correlations between mature/active TGF-β1 and platelet pro-inflammatory markers (P-selectin and CD40L) as well as common indicators of inflammation (CRP and ESR) were observed in CAD patients. In this study, given the insignificant changes in pro-aggregatory potentials in stable CAD, the pro-inflammatory state of platelets may be more involved in disease development and progression. Direct correlations between active platelet-borne TGF-β1 and pro-inflammatory markers with its presence in CAD-patients, which was almost absent in the platelets of healthy individuals, may also underscore the significant contribution of platelet-borne TGF-β1 to the pathogenesis of the disease.
Collapse
Affiliation(s)
- Javad Ahmadi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Ehteramolsadat Hosseini
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Faranak Kargar
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Science, Tehran, Iran
| | - Mehran Ghasemzadeh
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran. .,Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Iranian Blood Transfusion Organization Building, Next to the Milad Tower, Hemmat Exp. Way, P.O.Box:14665-1157, Tehran, Iran.
| |
Collapse
|
35
|
Cruz CJ, Dewberry LS, Otto KJ, Allen KD. Neuromodulation as a Potential Disease-Modifying Therapy for Osteoarthritis. Curr Rheumatol Rep 2023; 25:1-11. [PMID: 36435890 PMCID: PMC11438129 DOI: 10.1007/s11926-022-01094-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2022] [Indexed: 11/28/2022]
Abstract
PURPOSE OF REVIEW The following review discusses the therapeutic potential of targeting the autonomic nervous system (ANS) for osteoarthritis (OA) treatment and encourages the field to consider the candidacy of bioelectronic medicine as a novel OA treatment strategy. RECENT FINDINGS The study of OA pathogenesis has focused on changes occurring at the joint level. As such, treatments for OA have been aimed at the local joint environment, intending to resolve local inflammation and decrease pain. However, OA pathogenesis has shown to be more than joint wear and tear. Specifically, OA-related peripheral and central sensitization can prompt neuroplastic changes in the nervous system beyond the articular joint. These neuroplastic changes may alter physiologic systems, like the neuroimmune axis. In this way, OA and related comorbidities may share roots in the form of altered neuroimmune communication and autonomic dysfunction. ANS modulation may be able to modify OA pathogenesis or reduce the impact of OA comorbidities. Moreover, blocking chronic nociceptive drive from the joint may help to prevent maladaptive nervous system plasticity in OA.
Collapse
Affiliation(s)
- Carlos J Cruz
- J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
- Pain Research and Intervention Center of Excellence, Gainesville, FL, USA
| | - L Savannah Dewberry
- J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
| | - Kevin J Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA
- Department of Materials Science and Engineering, University of Florida, Gainesville, FL, USA
- Department of Neurology, University of Florida, Gainesville, FL, USA
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL, USA
- Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Kyle D Allen
- J. Crayton Pruitt Family Department of Biomedical Engineering, Biomedical Sciences Building, University of Florida, 1275 Center Drive, Gainesville, FL, 32611, USA.
- Pain Research and Intervention Center of Excellence, Gainesville, FL, USA.
- Department of Orthopaedics and Rehabilitation, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
36
|
Abstract
Monocytes/macrophages are key components of the body's innate ability to restore tissue function after injury. In most tissues, both embryo-derived tissue-resident macrophages and recruited blood monocyte-derived macrophages contribute to the injury response. The developmental origin of injury-associated macrophages has a major impact on the outcome of the healing process. Macrophages are abundant at all stages of repair and coordinate the progression through the different phases of healing. They are highly plastic cells that continuously adapt to their environment and acquire phase-specific activation phenotypes. Advanced omics methodologies have revealed a vast heterogeneity of macrophage activation phenotypes and metabolic status at injury sites in different organs. In this review, we highlight the role of the developmental origin, the link between the wound phase-specific activation state and metabolic reprogramming as well as the fate of macrophages during the resolution of the wounding response.
Collapse
Affiliation(s)
| | - Louise Injarabian
- Department of Dermatology, University of Cologne, 50937 Cologne, Germany
| | - Sabine A Eming
- Department of Dermatology, University of Cologne, 50937 Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
- Institute of Zoology, Developmental Biology Unit, University of Cologne, 50674 Cologne, Germany
| |
Collapse
|
37
|
Olwenyi OA, Johnson SD, Bidokhti M, Thakur V, Pandey K, Thurman M, Acharya A, Uppada S, Callen S, Giavedoni L, Ranga U, Buch SJ, Byrareddy SN. Systems biology analyses reveal enhanced chronic morphine distortion of gut-brain interrelationships in simian human immunodeficiency virus infected rhesus macaques. Front Neurosci 2022; 16:1001544. [PMID: 36312033 PMCID: PMC9613112 DOI: 10.3389/fnins.2022.1001544] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/09/2022] [Indexed: 11/13/2022] Open
Abstract
Background Commonly used opioids, such as morphine have been implicated in augmented SIV/HIV persistence within the central nervous system (CNS). However, the extent of myeloid cell polarization and viral persistence in different brain regions remains unclear. Additionally, the additive effects of morphine on SIV/HIV dysregulation of gut-brain crosstalk remain underexplored. Therefore, studies focused on understanding how drugs of abuse such as morphine affect immune dynamics, viral persistence and gut-brain interrelationships are warranted. Materials and methods For a total of 9 weeks, rhesus macaques were ramped-up, and twice daily injections of either morphine (n = 4) or saline (n = 4) administered. This was later followed with infection with SHIVAD8EO variants. At necropsy, mononuclear cells were isolated from diverse brain [frontal lobe, cerebellum, medulla, putamen, hippocampus (HIP) and subventricular zone (SVZ)] and gut [lamina propria (LP) and muscularis (MUSC) of ascending colon, duodenum, and ileum] regions. Multiparametric flow cytometry was used to were profile for myeloid cell polarity/activation and results corroborated with indirect immunofluorescence assays. Simian human immunodeficiency virus (SHIV) DNA levels were measured with aid of the digital droplet polymerase chain reaction (PCR) assay. Luminex assays were then used to evaluate soluble plasma/CSF biomarker levels. Finally, changes in the fecal microbiome were evaluated using 16S rRNA on the Illumina NovaSeq platform. Results Flow Cytometry-based semi-supervised analysis revealed that morphine exposure led to exacerbated M1 (CD14/CD16)/M2 (CD163/CD206) polarization in activated microglia that spanned across diverse brain regions. This was accompanied by elevated SHIV DNA within the sites of neurogenesis-HIP and SVZ. HIP/SVZ CD16+ activated microglia positively correlated with SHIV DNA levels in the brain (r = 0.548, p = 0.042). Simultaneously, morphine dependence depleted butyrate-producing bacteria, including Ruminococcus (p = 0.05), Lachnospira (p = 0.068) genera and Roseburia_sp_831b (p = 0.068). Finally, morphine also altered the regulation of CNS inflammation by reducing the levels of IL1 Receptor antagonist (IL1Ra). Conclusion These findings are suggestive that morphine promotes CNS inflammation by altering receptor modulation, increasing myeloid brain activation, distorting gut-brain crosstalk, and causing selective enhancement of SHIV persistence in sites of neurogenesis.
Collapse
Affiliation(s)
- Omalla A. Olwenyi
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Samuel D. Johnson
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Mehdi Bidokhti
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Vandana Thakur
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Kabita Pandey
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, United States
| | - Michellie Thurman
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Arpan Acharya
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Srijayaprakash Uppada
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Shannon Callen
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Luis Giavedoni
- Department of Biology, Trinity University, San Antonio, TX, United States
| | - Udaykumar Ranga
- Molecular Biology and Genetics Unit, Jawaharlal Nehru Centre for Advanced Scientific Research, Bangalore, India
| | - Shilpa J. Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Siddappa N. Byrareddy
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Genetics, Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE, United States
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE, United States
| |
Collapse
|
38
|
Rigamonti A, Castagna A, Viatore M, Colombo FS, Terzoli S, Peano C, Marchesi F, Locati M. Distinct responses of newly identified monocyte subsets to advanced gastrointestinal cancer and COVID-19. Front Immunol 2022; 13:967737. [PMID: 36263038 PMCID: PMC9576306 DOI: 10.3389/fimmu.2022.967737] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/16/2022] [Indexed: 11/25/2022] Open
Abstract
Monocytes are critical cells of the immune system but their role as effectors is relatively poorly understood, as they have long been considered only as precursors of tissue macrophages or dendritic cells. Moreover, it is known that this cell type is heterogeneous, but our understanding of this aspect is limited to the broad classification in classical/intermediate/non-classical monocytes, commonly based on their expression of only two markers, i.e. CD14 and CD16. We deeply dissected the heterogeneity of human circulating monocytes in healthy donors by transcriptomic analysis at single-cell level and identified 9 distinct monocyte populations characterized each by a profile suggestive of specialized functions. The classical monocyte subset in fact included five distinct populations, each enriched for transcriptomic gene sets related to either inflammatory, neutrophil-like, interferon-related, and platelet-related pathways. Non-classical monocytes included two distinct populations, one of which marked specifically by elevated expression levels of complement components. Intermediate monocytes were not further divided in our analysis and were characterized by high levels of human leukocyte antigen (HLA) genes. Finally, we identified one cluster included in both classical and non-classical monocytes, characterized by a strong cytotoxic signature. These findings provided the rationale to exploit the relevance of newly identified monocyte populations in disease evolution. A machine learning approach was developed and applied to two single-cell transcriptome public datasets, from gastrointestinal cancer and Coronavirus disease 2019 (COVID-19) patients. The dissection of these datasets through our classification revealed that patients with advanced cancers showed a selective increase in monocytes enriched in platelet-related pathways. Of note, the signature associated with this population correlated with worse prognosis in gastric cancer patients. Conversely, after immunotherapy, the most activated population was composed of interferon-related monocytes, consistent with an upregulation in interferon-related genes in responder patients compared to non-responders. In COVID-19 patients we confirmed a global activated phenotype of the entire monocyte compartment, but our classification revealed that only cytotoxic monocytes are expanded during the disease progression. Collectively, this study unravels an unexpected complexity among human circulating monocytes and highlights the existence of specialized populations differently engaged depending on the pathological context.
Collapse
Affiliation(s)
- Alessandra Rigamonti
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandra Castagna
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Marika Viatore
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
| | | | - Sara Terzoli
- Laboratory of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, Milan, Italy
| | - Clelia Peano
- Genomic Unit, IRCCS Humanitas Research Hospital, Milan, Italy
- Institute of Genetic and Biomedical Research, UoS of Milan, National Research Council, Milan, Italy
| | - Federica Marchesi
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Massimo Locati
- Department of Immunology and Inflammation, IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
- *Correspondence: Massimo Locati,
| |
Collapse
|
39
|
Shih CP, Tang X, Kuo CW, Chueh DY, Chen P. Design principles of bioinspired interfaces for biomedical applications in therapeutics and imaging. Front Chem 2022; 10:990171. [PMID: 36405322 PMCID: PMC9673126 DOI: 10.3389/fchem.2022.990171] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 09/08/2022] [Indexed: 09/29/2023] Open
Abstract
In the past two decades, we have witnessed rapid developments in nanotechnology, especially in biomedical applications such as drug delivery, biosensing, and bioimaging. The most commonly used nanomaterials in biomedical applications are nanoparticles, which serve as carriers for various therapeutic and contrast reagents. Since nanomaterials are in direct contact with biological samples, biocompatibility is one of the most important issues for the fabrication and synthesis of nanomaterials for biomedical applications. To achieve specific recognition of biomolecules for targeted delivery and biomolecular sensing, it is common practice to engineer the surfaces of nanomaterials with recognition moieties. This mini-review summarizes different approaches for engineering the interfaces of nanomaterials to improve their biocompatibility and specific recognition properties. We also focus on design strategies that mimic biological systems such as cell membranes of red blood cells, leukocytes, platelets, cancer cells, and bacteria.
Collapse
Affiliation(s)
- Chun-Pei Shih
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Xiaofang Tang
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Chiung Wen Kuo
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Di-Yen Chueh
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
| | - Peilin Chen
- Research Center for Applied Sciences, Academia Sinica, Taipei, Taiwan
- Institute of Physics, Academia Sinica, Taipei, Taiwan
| |
Collapse
|
40
|
Vishnyakova P, Kuznetsova M, Poltavets A, Fomina M, Kiseleva V, Muminova K, Potapova A, Khodzhaeva Z, Pyregov A, Trofimov D, Elchaninov A, Sukhikh G, Fatkhudinov T. Distinct gene expression patterns for CD14++ and CD16++ monocytes in preeclampsia. Sci Rep 2022; 12:15469. [PMID: 36104441 PMCID: PMC9474473 DOI: 10.1038/s41598-022-19847-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 09/05/2022] [Indexed: 11/19/2022] Open
Abstract
Preeclampsia (PE) is a serious gestational complication affecting the life of a mother and child. The immunophenotype and gene expression profile of isolated blood monocyte subpopulations of pregnant women with PE have not been studied before. In this work, we assessed changes in CD14++ and CD16++ monocyte subpopulations in PE and physiological pregnancy (n = 33). Immunophenotyping, immunomagnetic sorting of monocytes and analysis of the transcriptional profile of their genes were carried out. The percentage of classical monocytes was significantly lower, while the intermediate fraction of monocytes was significantly higher in late-onset PE compared to control. Transcriptome analysis of late-onset PE classical CD14++ monocytes revealed significant activation of inflammation mediated by chemokine and cytokine signalling pathways; apoptosis; regulation of transcription from RNA polymerase II promoter in response to stress and others. The most suppressed signalling pathways were associated with T cell activation and selection. In CD16++ monocytes of late-onset PE cases, positive regulation of cell-cell adhesion, integrin signalling pathway, blood coagulation cascade were the most activated ones. The inflammation mediated by chemokine and cytokine signalling pathway and p53 pathway were the most down-regulated in CD16++ monocytes. The obtained results indicate profound changes occurring to two most polar monocyte subpopulations in PE and their different roles in the pathogenesis of this disease.
Collapse
Affiliation(s)
- Polina Vishnyakova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia.
- Peoples' Friendship University of Russia, Moscow, Russia.
| | - Maria Kuznetsova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Anastasiya Poltavets
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Mariia Fomina
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Viktoriia Kiseleva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Kamilla Muminova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Alena Potapova
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Zulfiya Khodzhaeva
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Alexey Pyregov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Dmitry Trofimov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Andrey Elchaninov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Gennady Sukhikh
- National Medical Research Center for Obstetrics, Gynecology and Perinatology Named After Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, Moscow, Russia
| | - Timur Fatkhudinov
- Peoples' Friendship University of Russia, Moscow, Russia
- A.P. Avtsyn Research Institute of Human Morphology, Moscow, Russia
| |
Collapse
|
41
|
Elchaninov A, Vishnyakova P, Menyailo E, Sukhikh G, Fatkhudinov T. An Eye on Kupffer Cells: Development, Phenotype and the Macrophage Niche. Int J Mol Sci 2022; 23:9868. [PMID: 36077265 PMCID: PMC9456487 DOI: 10.3390/ijms23179868] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/14/2022] [Accepted: 08/29/2022] [Indexed: 11/16/2022] Open
Abstract
Macrophages are key participants in the maintenance of tissue homeostasis under normal and pathological conditions, and implement a rich diversity of functions. The largest population of resident tissue macrophages is found in the liver. Hepatic macrophages, termed Kupffer cells, are involved in the regulation of multiple liver functionalities. Specific differentiation profiles and functional activities of tissue macrophages have been attributed to the shaping role of the so-called tissue niche microenvironments. The fundamental macrophage niche concept was lately shaken by a flood of new data, leading to a revision and substantial update of the concept, which constitutes the main focus of this review. The macrophage community discusses contemporary evidence on the developmental origins of resident macrophages, notably Kupffer cells and the issues of heterogeneity of the hepatic macrophage populations, as well as the roles of proliferation, cell death and migration processes in the maintenance of macrophage populations of the liver. Special consideration is given to interactions of Kupffer cells with other local cell lineages, including Ito cells, sinusoidal endothelium and hepatocytes, which participate in the maintenance of their phenotypical and functional identity.
Collapse
Affiliation(s)
- Andrey Elchaninov
- Laboratory of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia
- Histology Department, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| | - Polina Vishnyakova
- Laboratory of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia
- Histology Department, Medical Institute, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
| | - Egor Menyailo
- Laboratory of Growth and Development, Avtsyn Research Institute of Human Morphology of FSBI “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| | - Gennady Sukhikh
- Laboratory of Regenerative Medicine, National Medical Research Center for Obstetrics, Gynecology and Perinatology Named after Academician V.I. Kulakov of Ministry of Healthcare of Russian Federation, 117997 Moscow, Russia
| | - Timur Fatkhudinov
- Histology Department, Medical Institute, Peoples’ Friendship University of Russia, 117198 Moscow, Russia
- Laboratory of Growth and Development, Avtsyn Research Institute of Human Morphology of FSBI “Petrovsky National Research Centre of Surgery”, 117418 Moscow, Russia
| |
Collapse
|
42
|
Fink EE, Sona S, Tran U, Desprez PE, Bradley M, Qiu H, Eltemamy M, Wee A, Wolkov M, Nicolas M, Min B, Haber GP, Wessely O, Lee BH, Ting AH. Single-cell and spatial mapping Identify cell types and signaling Networks in the human ureter. Dev Cell 2022; 57:1899-1916.e6. [PMID: 35914526 PMCID: PMC9381170 DOI: 10.1016/j.devcel.2022.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 03/18/2022] [Accepted: 07/05/2022] [Indexed: 01/16/2023]
Abstract
Tissue engineering offers a promising treatment strategy for ureteral strictures, but its success requires an in-depth understanding of the architecture, cellular heterogeneity, and signaling pathways underlying tissue regeneration. Here, we define and spatially map cell populations within the human ureter using single-cell RNA sequencing, spatial gene expression, and immunofluorescence approaches. We focus on the stromal and urothelial cell populations to enumerate the distinct cell types composing the human ureter and infer potential cell-cell communication networks underpinning the bi-directional crosstalk between these compartments. Furthermore, we analyze and experimentally validate the importance of the sonic hedgehog (SHH) signaling pathway in adult progenitor cell maintenance. The SHH-expressing basal cells support organoid generation in vitro and accurately predict the differentiation trajectory from basal progenitor cells to terminally differentiated umbrella cells. Our results highlight the essential processes involved in adult ureter tissue homeostasis and provide a blueprint for guiding ureter tissue engineering.
Collapse
Affiliation(s)
- Emily E Fink
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Surbhi Sona
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Nutrition, Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Uyen Tran
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Pierre-Emmanuel Desprez
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Department of Urology, CHU Lille, Claude Huriez Hospital, Université Lille, 59000 Lille, France
| | - Matthew Bradley
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Hong Qiu
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Mohamed Eltemamy
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Alvin Wee
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Madison Wolkov
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Marlo Nicolas
- Pathology & Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Booki Min
- Department of Microbiology and Immunology, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Georges-Pascal Haber
- Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Oliver Wessely
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Byron H Lee
- Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA; Glickman Urological & Kidney Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Angela H Ting
- Genomic Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
43
|
Weber A, Schwiebs A, Solhaug H, Stenvik J, Nilsen AM, Wagner M, Relja B, Radeke HH. Nanoplastics affect the inflammatory cytokine release by primary human monocytes and dendritic cells. ENVIRONMENT INTERNATIONAL 2022; 163:107173. [PMID: 35303527 DOI: 10.1016/j.envint.2022.107173] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/13/2022] [Accepted: 03/03/2022] [Indexed: 06/14/2023]
Abstract
So far, the human health impacts of nano- and microplastics are poorly understood. Thus, we investigated whether nanoplastics exposure induces inflammatory processes in primary human monocytes and monocyte-derived dendritic cells. We exposed these cells in vitro to nanoplastics of different shapes (irregular vs. spherical), sizes (50-310 nm and polydisperse mixtures) and polymer types (polystyrene; polymethyl methacrylate; polyvinyl chloride, PVC) using concentrations of 30-300 particles cell-1. Our results show that irregular PVC particles induce the strongest cytokine release of these nanoplastics. Irregular polystyrene triggered a significantly higher pro-inflammatory response compared to spherical nanoplastics. The contribution of chemicals leaching from the particles was minor. The effects were concentration-dependent but varied markedly between cell donors. We conclude that nanoplastics exposure can provoke human immune cells to secrete cytokines as key initiators of inflammation. This response is specific to certain polymers (PVC) and particle shapes (fragments). Accordingly, nanoplastics cannot be considered one homogenous entity when assessing their health implications and the use of spherical polystyrene nanoplastics may underestimate their inflammatory effects.
Collapse
Affiliation(s)
- Annkatrin Weber
- Goethe University, Department of Aquatic Ecotoxicology, Faculty of Biological Sciences, Max-von-Laue-Straße 13, 60438 Frankfurt am Main, Germany
| | - Anja Schwiebs
- Goethe University Hospital, Institute of General Pharmacology and Toxicology, pharmazentrum frankfurt, Theodor-Stern-Kai 7/75, 60596 Frankfurt am Main, Germany
| | - Helene Solhaug
- Norwegian University of Science and Technology, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Erling Skjalgssons gate 1, Trondheim, Norway
| | - Jørgen Stenvik
- Norwegian University of Science and Technology, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Erling Skjalgssons gate 1, Trondheim, Norway; Norwegian University of Science and Technology, Centre of Molecular Inflammation Research, Olav Kyrres gate 17, Trondheim, Norway
| | - Asbjørn M Nilsen
- Norwegian University of Science and Technology, Department of Clinical and Molecular Medicine, Faculty of Medicine and Health Sciences, Erling Skjalgssons gate 1, Trondheim, Norway
| | - Martin Wagner
- Norwegian University of Science and Technology, Department of Biology, Høgskoleringen 5, Realfagbygget, 7491 Trondheim, Norway.
| | - Borna Relja
- Otto-von-Guericke University, Department of Radiology and Nuclear Medicine, Experimental Radiology, Leipziger Str. 44, 39120 Magdeburg, Germany
| | - Heinfried H Radeke
- Goethe University Hospital, Institute of General Pharmacology and Toxicology, pharmazentrum frankfurt, Theodor-Stern-Kai 7/75, 60596 Frankfurt am Main, Germany
| |
Collapse
|
44
|
True H, Blanton M, Sureshchandra S, Messaoudi I. Monocytes and macrophages in pregnancy: The good, the bad, and the ugly. Immunol Rev 2022; 308:77-92. [PMID: 35451089 DOI: 10.1111/imr.13080] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/08/2022] [Indexed: 12/12/2022]
Abstract
A successful human pregnancy requires precisely timed adaptations by the maternal immune system to support fetal growth while simultaneously protecting mother and fetus against microbial challenges. The first trimester of pregnancy is characterized by a robust increase in innate immune activity that promotes successful implantation of the blastocyst and placental development. Moreover, early pregnancy is also a state of increased vulnerability to vertically transmitted pathogens notably, human immunodeficiency virus (HIV), Zika virus (ZIKV), SARS-CoV-2, and Listeria monocytogenes. As gestation progresses, the second trimester is marked by the establishment of an immunosuppressive environment that promotes fetal tolerance and growth while preventing preterm birth, spontaneous abortion, and other gestational complications. Finally, the period leading up to labor and parturition is characterized by the reinstatement of an inflammatory milieu triggering childbirth. These dynamic waves of carefully orchestrated changes have been dubbed the "immune clock of pregnancy." Monocytes in maternal circulation and tissue-resident macrophages at the maternal-fetal interface play a critical role in this delicate balance. This review will summarize the current data describing the longitudinal changes in the phenotype and function of monocyte and macrophage populations in healthy and complicated pregnancies.
Collapse
Affiliation(s)
- Heather True
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA.,Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | - Madison Blanton
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA.,Department of Pharmaceutical Sciences, University of Kentucky College of Pharmacy, Lexington, Kentucky, USA
| | | | - Ilhem Messaoudi
- Department of Microbiology, Immunology, and Molecular Genetics, University of Kentucky College of Medicine, Lexington, Kentucky, USA
| |
Collapse
|
45
|
Jebari-Benslaiman S, Galicia-García U, Larrea-Sebal A, Olaetxea JR, Alloza I, Vandenbroeck K, Benito-Vicente A, Martín C. Pathophysiology of Atherosclerosis. Int J Mol Sci 2022; 23:ijms23063346. [PMID: 35328769 PMCID: PMC8954705 DOI: 10.3390/ijms23063346] [Citation(s) in RCA: 376] [Impact Index Per Article: 125.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/12/2022] [Accepted: 03/18/2022] [Indexed: 11/26/2022] Open
Abstract
Atherosclerosis is the main risk factor for cardiovascular disease (CVD), which is the leading cause of mortality worldwide. Atherosclerosis is initiated by endothelium activation and, followed by a cascade of events (accumulation of lipids, fibrous elements, and calcification), triggers the vessel narrowing and activation of inflammatory pathways. The resultant atheroma plaque, along with these processes, results in cardiovascular complications. This review focuses on the different stages of atherosclerosis development, ranging from endothelial dysfunction to plaque rupture. In addition, the post-transcriptional regulation and modulation of atheroma plaque by microRNAs and lncRNAs, the role of microbiota, and the importance of sex as a crucial risk factor in atherosclerosis are covered here in order to provide a global view of the disease.
Collapse
Affiliation(s)
- Shifa Jebari-Benslaiman
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
| | - Unai Galicia-García
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | - Asier Larrea-Sebal
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Fundación Biofisika Bizkaia, Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain
| | | | - Iraide Alloza
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
| | - Koen Vandenbroeck
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Inflammation & Biomarkers Group, Biocruces Bizkaia Health Research Institute, 48903 Barakaldo, Bizkaia, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Bizkaia, Spain
| | - Asier Benito-Vicente
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| | - César Martín
- Department of Biochemistry and Molecular Biology, Universidad del País Vasco UPV/EHU, 48940 Leioa, Bizkaia, Spain; (S.J.-B.); (I.A.); (K.V.)
- Biofisika Institute (UPV/EHU, CSIC), Barrio Sarriena s/n., 48940 Leioa, Bizkaia, Spain; (U.G.-G.); (A.L.-S.)
- Correspondence: (A.B.-V.); (C.M.); Tel.: +34-946-01-2741 (C.M.)
| |
Collapse
|
46
|
Pimkova Polidarova M, Brehova P, Dejmek M, Birkus G, Brazdova A. STING Agonist-Mediated Cytokine Secretion Is Accompanied by Monocyte Apoptosis. ACS Infect Dis 2022; 8:463-471. [PMID: 35132859 DOI: 10.1021/acsinfecdis.1c00554] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The cGAS-STING (cyclic GMP-AMP synthase-stimulator of interferon genes) pathway plays a crucial role in inducing an antiviral and antitumor immune response. We studied the effects of synthetic STING agonists on several immune populations and related cytokine production. In comparison with the toll-like receptor 7 (TLR7) agonist, STING agonists induced secretion of a broader proinflammatory cytokine spectrum. Unlike the TLR7 agonist, the structurally diverse STING agonists partially depleted B and NK cells and completely depleted CD14+ monocytes via induction of apoptosis. The TANK-binding kinase 1 inhibitor efficiently prevented interferon alpha (IFNα) secretion and cell depletion, suggesting their possible dependence on the cGAS-STING pathway activation. Finally, IFNα, tumor necrosis factor alpha, interleukin 6, and interleukin 1 beta secretion and CD14+ monocyte apoptosis were primary responses to STING agonists, whereas IFNγ was secreted secondarily. These findings bring new insights into the cGAS-STING pathway immunomodulation that is of future therapeutic importance.
Collapse
Affiliation(s)
- Marketa Pimkova Polidarova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
- Faculty of Science, Charles University, Albertov 6, Prague 12800, Czech Republic
| | - Petra Brehova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| | - Milan Dejmek
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| | - Gabriel Birkus
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| | - Andrea Brazdova
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo namesti 2, Prague 16000, Czech Republic
| |
Collapse
|
47
|
Schwäbe FV, Happonen L, Ekestubbe S, Neumann A. Host Defense Peptides LL-37 and Lactoferrin Trigger ET Release from Blood-Derived Circulating Monocytes. Biomedicines 2022; 10:biomedicines10020469. [PMID: 35203676 PMCID: PMC8962388 DOI: 10.3390/biomedicines10020469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
Neutrophils are commonly regarded as the first line of immune response during infection or in tissue injury-induced inflammation. The rapid influx of these cells results in the release of host defense proteins (HDPs) or formation of neutrophil extracellular traps (NETs). As a second wave during inflammation or infection, circulating monocytes arrive at the site. Earlier studies showed that HDPs LL-37 and Lactoferrin (LTF) activate monocytes while neutrophil elastase facilitates the formation of extracellular traps (ETs) in monocytes. However, the knowledge about the impact of HDPs on monocytes remains sparse. In the present study, we investigated the effect of LL-37 and LTF on blood-derived CD14+ monocytes. Both HDPs triggered a significant release of TNFα, nucleosomes, and monocyte ETs. Microscopic analysis indicated that ET formation by LL-37 depends on storage-operated calcium entry (SOCE), mitogen-activated protein kinase (MAPK), and ERK1/2, whereas the LTF-mediated ET release is not affected by any of the here used inhibitors. Quantitative proteomics mass spectrometry analysis of the neutrophil granular content (NGC) revealed a high abundance of Lactoferrin. The stimulation of CD14+ monocytes with NGC resulted in a significant secretion of TNFα and nucleosomes, and the formation of monocyte ETs. The findings of this study provide new insight into the complex interaction of HDPs, neutrophils, and monocytes during inflammation.
Collapse
|
48
|
Abo-Zaid MA, Hamdi AA. Evaluation of Immune Response and Haematological Parameters in Infected Male Albino Rats by Giardiasis. Parasite Immunol 2022; 44:e12908. [PMID: 35104007 DOI: 10.1111/pim.12908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 11/27/2022]
Abstract
The present work aimed to study the effects of G. lamblia infection on immunological, haematological studies and to evaluate immunoglobulins and some cytokines. Fifty male albino rats were divided into six groups. The control group including 20 rats and the infected group includes 30 rats. All the estimations were checked all over five checkpoints (CP) (7, 14, 21, 28, and 35 days post-infection). Serum levels of IgA, IgG, IgM and IgE. Cytokines INF-γ, TNF-alpha, IL-4, IL-10, and haematological parameters were determined. Cyst and trophozoite were counted. A considerable increase in the level of immunoglobulins and cytokines in all infected groups compared to the control group was documented. Furthermore, a significant decrease in red blood corpuscles, haemoglobin, and mean corpuscular haemoglobin concentration levels, whereas substantial increases in mean corpuscular volume, mean corpuscular haemoglobin and platelets were observed. Moreover, infected rats had a substantial rise in WBCs, lymphocytes, and eosinophil counts compared to the control group, whereas neutrophils and monocytes had a significant decrease. Number of trophozoites and cysts were significantly increased in infected groups before diminishing after day 28. The current results showed that Th1 and Th2 immune responses, which are characterized by the production of TNF-α, IFN-γ, IL-4 and IL-10, are important for protection against Giardia infections and also verified the balance between these cytokines and the timing of their production was crucial in G. lamblia immune response. Giardia lamblia, Immunity, Antibodies, cytokines, eosinophil.
Collapse
Affiliation(s)
- Mabrouk A Abo-Zaid
- Biology department faculty of science, Jazan University, Jazan, Saudi Arabia
| | - Aishah Ali Hamdi
- Biology department faculty of science, Jazan University, Jazan, Saudi Arabia
| |
Collapse
|
49
|
Malherbe DC, Messaoudi I. Transcriptional and Epigenetic Regulation of Monocyte and Macrophage Dysfunction by Chronic Alcohol Consumption. Front Immunol 2022; 13:911951. [PMID: 35844518 PMCID: PMC9277054 DOI: 10.3389/fimmu.2022.911951] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 05/27/2022] [Indexed: 02/05/2023] Open
Abstract
Drinking alcohol, even in moderation, can affect the immune system. Studies have shown disproportionate effects of alcohol on circulating and tissue-resident myeloid cells (granulocytes, monocytes, macrophages, dendritic cells). These cells orchestrate the body's first line of defense against microbial challenges as well as maintain tissue homeostasis and repair. Alcohol's effects on these cells are dependent on exposure pattern, with acute drinking dampening but chronic drinking enhancing production of inflammatory mediators. Although chronic drinking is associated with heightened systemic inflammation, studies on tissue resident macrophage populations in several organs including the spleen, liver, brain, and lung have also shown compromised functional and metabolic capacities of these cells. Many of these effects are thought to be mediated by oxidative stress caused by alcohol and its metabolites which can directly impact the cellular epigenetic landscapes. In addition, since myeloid cells are relatively short-lived in circulation and are under constant repopulation from the bone marrow compartment, alcohol's effects on bone marrow progenitors and hematopoiesis are important for understanding the impact of alcohol systemically on these myeloid populations. Alcohol-induced disruption of progenitor, circulating, and tissue resident myeloid populations contribute to the increased susceptibility of patients with alcohol use disorders to viral and bacterial infections. In this review, we provide an overview of the impact of chronic alcohol consumption on the function of monocytes and macrophages in host defense, tissue repair and inflammation. We then summarize our current understanding of the mechanisms underlying alcohol-induced disruption and examine changes in transcriptome and epigenome of monocytes and mcrophages. Overall, chronic alcohol consumption leads to hyper-inflammation concomitant with decreased microbial and wound healing responses by monocytes/macrophages due to a rewiring of the epigentic and transcriptional landscape. However, in advanced alcoholic liver disease, myeloid cells become immunosuppressed as a response to the surrounding hyper-inflammatory milieu. Therefore, the effect of chronic alcohol on the inflammatory response depends on disease state and the immune cell population.
Collapse
|
50
|
Fabry LAR, Triantafyllopoulou A. [The role of the response to DNA damage in granulomatous diseases]. Z Rheumatol 2022; 81:881-887. [PMID: 36006470 PMCID: PMC9732071 DOI: 10.1007/s00393-022-01260-y] [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] [Accepted: 07/20/2022] [Indexed: 12/14/2022]
Abstract
Granulomas are organized aggregates of immune cells, which are formed in response to a persistent stimulus and are found in various rheumatic diseases, including sarcoidosis, rheumatoid arthritis and granulomatosis with polyangiitis. The core of granulomas contains a multitude of different macrophage subtypes, including multinucleated macrophages and foam cells. The mechanisms which induce the formation of granulomas are not well understood; however, recent data show that the DNA damage response regulates granuloma macrophage differentiation.
Collapse
Affiliation(s)
- Lea A R Fabry
- Medizinische Klinik m.S. Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland.
- Deutsches Rheuma Forschungszentrum, ein Institut der Leibniz Gemeinschaft, Berlin, Deutschland.
| | - Antigoni Triantafyllopoulou
- Medizinische Klinik m.S. Rheumatologie und Klinische Immunologie, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland.
- Deutsches Rheuma Forschungszentrum, ein Institut der Leibniz Gemeinschaft, Berlin, Deutschland.
| |
Collapse
|