1
|
Sun ED, Zhou OY, Hauptschein M, Rappoport N, Xu L, Navarro Negredo P, Liu L, Rando TA, Zou J, Brunet A. Spatiotemporal transcriptomic profiling and modeling of mouse brain at single-cell resolution reveals cell proximity effects of aging and rejuvenation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.16.603809. [PMID: 39071282 PMCID: PMC11275735 DOI: 10.1101/2024.07.16.603809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Old age is associated with a decline in cognitive function and an increase in neurodegenerative disease risk1. Brain aging is complex and accompanied by many cellular changes2-20. However, the influence that aged cells have on neighboring cells and how this contributes to tissue decline is unknown. More generally, the tools to systematically address this question in aging tissues have not yet been developed. Here, we generate spatiotemporal data at single-cell resolution for the mouse brain across lifespan, and we develop the first machine learning models based on spatial transcriptomics ('spatial aging clocks') to reveal cell proximity effects during brain aging and rejuvenation. We collect a single-cell spatial transcriptomics brain atlas of 4.2 million cells from 20 distinct ages and across two rejuvenating interventions-exercise and partial reprogramming. We identify spatial and cell type-specific transcriptomic fingerprints of aging, rejuvenation, and disease, including for rare cell types. Using spatial aging clocks and deep learning models, we find that T cells, which infiltrate the brain with age, have a striking pro-aging proximity effect on neighboring cells. Surprisingly, neural stem cells have a strong pro-rejuvenating effect on neighboring cells. By developing computational tools to identify mediators of these proximity effects, we find that pro-aging T cells trigger a local inflammatory response likely via interferon-γ whereas pro-rejuvenating neural stem cells impact the metabolism of neighboring cells possibly via growth factors (e.g. vascular endothelial growth factor) and extracellular vesicles, and we experimentally validate some of these predictions. These results suggest that rare cells can have a drastic influence on their neighbors and could be targeted to counter tissue aging. We anticipate that these spatial aging clocks will not only allow scalable assessment of the efficacy of interventions for aging and disease but also represent a new tool for studying cell-cell interactions in many spatial contexts.
Collapse
Affiliation(s)
- Eric D. Sun
- Department of Biomedical Data Science, Stanford University, CA, USA
- Department of Genetics, Stanford University, CA, USA
| | - Olivia Y. Zhou
- Department of Genetics, Stanford University, CA, USA
- Stanford Biophysics Program, Stanford University, CA, USA
- Stanford Medical Scientist Training Program, Stanford University, CA, USA
| | | | | | - Lucy Xu
- Department of Genetics, Stanford University, CA, USA
- Department of Biology, Stanford University, CA, USA
| | | | - Ling Liu
- Department of Neurology, Stanford University, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - Thomas A. Rando
- Department of Neurology, Stanford University, CA, USA
- Department of Neurology, UCLA, Los Angeles, CA, USA
- Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Biology, UCLA, Los Angeles, CA, USA
| | - James Zou
- Department of Biomedical Data Science, Stanford University, CA, USA
- These authors contributed equally: James Zou, Anne Brunet
| | - Anne Brunet
- Department of Genetics, Stanford University, CA, USA
- Glenn Center for the Biology of Aging, Stanford University, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, CA, USA
- These authors contributed equally: James Zou, Anne Brunet
| |
Collapse
|
2
|
Nannini G, Di Gloria L, Russo E, Sterrantino G, Kiros ST, Coppi M, Niccolai E, Baldi S, Ramazzotti M, Di Pilato V, Lagi F, Bartolucci G, Rossolini GM, Bartoloni A, Amedei A. Oral microbiota signatures associated with viremia and CD4 recovery in treatment-naïve HIV-1-infected patients. Microbes Infect 2024; 26:105339. [PMID: 38636822 DOI: 10.1016/j.micinf.2024.105339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/11/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
PURPOSE Few reports focused on the role of oral microbiome diversity in HIV infection. We characterized the microbiota-immunity axis in a cohort of treatment-naïve HIV-1-infected patients undergoing antiretroviral therapy (ART) focusing on the oral microbiome (OM) and immunological responsivity. METHODS The sequencing of 16S rRNA V3-V4 hypervariable region was performed on salivary samples of 15 healthy control (HC) and 12 HIV + patients before starting ART and after reaching virological suppression. Then, we correlated the OM composition with serum cytokines and the Short Chain Fatty acids (SCFAs). RESULTS The comparison between HIV patients and HC oral microbiota showed differences in the bacterial α-diversity and richness. We documented a negative correlation between oral Prevotella and intestinal valeric acid at before starting ART and a positive correlation between oral Veillonella and gut acetic acid after reaching virological suppression. Finally, an increase in the phylum Proteobacteria was observed comparing saliva samples of immunological responders (IRs) patients against immunological non-responders (INRs). CONCLUSIONS For the first time, we described an increase in the oral pro-inflammatory Proteobacteria phylum in INRs compared to IRs. We provided more evidence that saliva could be a non-invasive and less expensive approach for research involving the oral cavity microbiome in HIV patients.
Collapse
Affiliation(s)
- Giulia Nannini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Leandro Di Gloria
- Department of Biomedical, Experimental and Clinical "Mario Serio", University of Florence, Florence 50134, Italy
| | - Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Gaetana Sterrantino
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Seble Tekle Kiros
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Clinical Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Marco Coppi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Elena Niccolai
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Matteo Ramazzotti
- Department of Biomedical, Experimental and Clinical "Mario Serio", University of Florence, Florence 50134, Italy
| | - Vincenzo Di Pilato
- Department of Surgical Sciences and Integrated Diagnostics, University of Genoa, Genoa, Italy
| | - Filippo Lagi
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy
| | - Gianluca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence 50019, Italy
| | - Gian Maria Rossolini
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Clinical Microbiology and Virology Unit, Careggi University Hospital, Florence, Italy
| | - Alessandro Bartoloni
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy; Infectious and Tropical Diseases Unit, Careggi University Hospital, Florence, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence 50134, Italy.
| |
Collapse
|
3
|
Daud M, Dasari P, Adelfinger M, Langenhorst D, Lother J, Slavkovic-Lukic D, Berges C, Kruhm M, Galler A, Schleussner C, Luther CH, Alberter K, Althammer A, Shaikh H, Pallmann N, Bodem J, El-Mowafy M, Beilhack A, Dittrich M, Topp MS, Zipfel PF, Beyersdorf N. Enolase 1 of Candida albicans binds human CD4 + T cells and modulates naïve and memory responses. Eur J Immunol 2023; 53:e2250284. [PMID: 37503840 DOI: 10.1002/eji.202250284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 06/05/2023] [Accepted: 07/27/2023] [Indexed: 07/29/2023]
Abstract
To obtain a better understanding of the biology behind life-threatening fungal infections caused by Candida albicans, we recently conducted an in silico screening for fungal and host protein interaction partners. We report here that the extracellular domain of human CD4 binds to the moonlighting protein enolase 1 (Eno1) of C. albicans as predicted bioinformatically. By using different anti-CD4 monoclonal antibodies, we determined that C. albicans Eno1 (CaEno1) primarily binds to the extracellular domain 3 of CD4. Functionally, we observed that CaEno1 binding to CD4 activated lymphocyte-specific protein tyrosine kinase (LCK), which was also the case for anti-CD4 monoclonal antibodies tested in parallel. CaEno1 binding to naïve human CD4+ T cells skewed cytokine secretion toward a Th2 profile indicative of poor fungal control. Moreover, CaEno1 inhibited human memory CD4+ T-cell recall responses. Therapeutically, CD4+ T cells transduced with a p41/Crf1-specific T-cell receptor developed for adoptive T-cell therapy were not inhibited by CaEno1 in vitro. Together, the interaction of human CD4+ T cells with CaEno1 modulated host CD4+ T-cell responses in favor of the fungus. Thus, CaEno1 mediates not only immune evasion through its interference with complement regulators but also through the direct modulation of CD4+ T-cell responses.
Collapse
Affiliation(s)
- Muhammad Daud
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Prasad Dasari
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
| | - Marion Adelfinger
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Daniela Langenhorst
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Jasmin Lother
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Dragana Slavkovic-Lukic
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Carsten Berges
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Michaela Kruhm
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | | | | | | | - Karl Alberter
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Anton Althammer
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Haroon Shaikh
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Niklas Pallmann
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Jochen Bodem
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Mohammed El-Mowafy
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
- Faculty of Pharmacy, Department of Microbiology & Immunology, Mansoura University, Mansoura, Egypt
| | - Andreas Beilhack
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Marcus Dittrich
- Chair of Bioinformatics, University of Würzburg, Würzburg, Germany
- Institute of Human Genetics, University of Würzburg, Würzburg, Germany
| | - Max S Topp
- Department of Internal Medicine II, Division of Hematology, University Hospital Würzburg, Würzburg, Germany
| | - Peter F Zipfel
- Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute, Jena, Germany
- Friedrich Schiller University, Jena, Germany
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Klaver D, Thurnher M. P2Y 11/IL-1 receptor crosstalk controls macrophage inflammation: a novel target for anti-inflammatory strategies? Purinergic Signal 2023; 19:501-511. [PMID: 37016172 PMCID: PMC10073626 DOI: 10.1007/s11302-023-09932-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/06/2023] [Indexed: 04/06/2023] Open
Abstract
Although first cloning of the human ATP receptor P2Y11 was successful 25 years ago, the exact downstream signaling pathways of P2Y11 receptor, which can couple to Gq and Gs proteins, have remained unclear. Especially the lack of rodent models as well as the limited availability of antibodies and pharmacological tools have hampered examination of P2Y11 expression and function. Many meaningful observations related to P2Y11 have been made in primary immune cells, indicating that P2Y11 receptors are important regulators of inflammation and cell migration, also by controlling mitochondrial activity. Our recent studies have shown that P2Y11 is upregulated during macrophage development and activates signaling through IL-1 receptor, which is well known for its ability to direct inflammatory and migratory processes. This review summarizes the results of the first transcriptomic and secretomic analyses of both, ectopic and native P2Y11 receptors, and discusses how P2Y11 crosstalk with the IL-1 receptor may govern anti-inflammatory and pro-angiogenic processes in human M2 macrophages.
Collapse
Affiliation(s)
- Dominik Klaver
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innrain 66a, 6020 Innsbruck, Austria
| | - Martin Thurnher
- Immunotherapy Unit, Department of Urology, Medical University of Innsbruck, Innrain 66a, 6020 Innsbruck, Austria
| |
Collapse
|
5
|
Pojero F, Gervasi F, Fiore SD, Aiello A, Bonacci S, Caldarella R, Attanzio A, Candore G, Caruso C, Ligotti ME, Procopio A, Restivo I, Tesoriere L, Allegra M, Accardi G. Anti-Inflammatory Effects of Nutritionally Relevant Concentrations of Oleuropein and Hydroxytyrosol on Peripheral Blood Mononuclear Cells: An Age-Related Analysis. Int J Mol Sci 2023; 24:11029. [PMID: 37446206 DOI: 10.3390/ijms241311029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/26/2023] [Accepted: 06/30/2023] [Indexed: 07/15/2023] Open
Abstract
Immunosenescence and inflammaging facilitate the insurgence of chronic diseases. The Mediterranean diet is a non-invasive intervention to improve the chronic low-grade inflammatory status associated with aging. Olive oil oleuropein (OLE) and hydroxytyrosol (HT) demonstrated a controversial modulatory action on inflammation in vitro when tested at concentrations exceeding those detectable in human plasma. We studied the potential anti-inflammatory effects of OLE and HT at nutritionally relevant concentrations on peripheral blood mononuclear cells (PBMCs) as regards cell viability, frequency of leukocyte subsets, and cytokine release, performing an age-focused analysis on two groups of subjects: Adult (age 18-64 years) and Senior (age ≥ 65 years). OLE and HT were used alone or as a pre-treatment before challenging PBMCs with lipopolysaccharide (LPS). Both polyphenols had no effect on cell viability irrespective of LPS, but 5 µM HT had an LPS-like effect on monocytes, reducing the intermediate subset in Adult subjects. OLE and HT had no effect on LPS-triggered release of TNF-α, IL-6 and IL-8, but 5 µM HT reduced IL-10 secretion by PBMCs from Adult vs. Senior group. In summary, nutritionally relevant concentrations of OLE and HT elicit no anti-inflammatory effect and influence the frequency of immune cell subsets with age-related different outcomes.
Collapse
Affiliation(s)
- Fanny Pojero
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Francesco Gervasi
- Specialistic Oncology Laboratory Unit, ARNAS Hospitals Civico Di Cristina e Benfratelli, 90127 Palermo, Italy
| | - Salvatore Davide Fiore
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Anna Aiello
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Sonia Bonacci
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Rosalia Caldarella
- Department of Laboratory Medicine, "P. Giaccone" University Hospital, 90127 Palermo, Italy
| | - Alessandro Attanzio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Giuseppina Candore
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Mattia Emanuela Ligotti
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| | - Antonio Procopio
- Department of Health Sciences, University Magna Graecia of Catanzaro, 88100 Catanzaro, Italy
| | - Ignazio Restivo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Luisa Tesoriere
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Mario Allegra
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123 Palermo, Italy
| | - Giulia Accardi
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Technologies, University of Palermo, 90133 Palermo, Italy
| |
Collapse
|
6
|
Elsaghir A, El-Sabaa EMW, Ahmed AK, Abdelwahab SF, Sayed IM, El-Mokhtar MA. The Role of Cluster of Differentiation 39 (CD39) and Purinergic Signaling Pathway in Viral Infections. Pathogens 2023; 12:279. [PMID: 36839551 PMCID: PMC9967413 DOI: 10.3390/pathogens12020279] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 02/11/2023] Open
Abstract
CD39 is a marker of immune cells such as lymphocytes and monocytes. The CD39/CD73 pathway hydrolyzes ATP into adenosine, which has a potent immunosuppressive effect. CD39 regulates the function of a variety of immunologic cells through the purinergic signaling pathways. CD39+ T cells have been implicated in viral infections, including Human Immunodeficiency Virus (HIV), Cytomegalovirus (CMV), viral hepatitis, and Corona Virus Disease 2019 (COVID-19) infections. The expression of CD39 is an indicator of lymphocyte exhaustion, which develops during chronicity. During RNA viral infections, the CD39 marker can profile the populations of CD4+ T lymphocytes into two populations, T-effector lymphocytes, and T-regulatory lymphocytes, where CD39 is predominantly expressed on the T-regulatory cells. The level of CD39 in T lymphocytes can predict the disease progression, antiviral immune responses, and the response to antiviral drugs. Besides, the percentage of CD39 and CD73 in B lymphocytes and monocytes can affect the status of viral infections. In this review, we investigate the impact of CD39 and CD39-expressing cells on viral infections and how the frequency and percentage of CD39+ immunologic cells determine disease prognosis.
Collapse
Affiliation(s)
- Alaa Elsaghir
- Department of Microbiology & Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | - Ehsan M. W. El-Sabaa
- Department of Microbiology & Immunology, Faculty of Pharmacy, Assiut University, Assiut 71515, Egypt
| | | | - Sayed F. Abdelwahab
- Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
| | - Ibrahim M. Sayed
- Department of Biomedical and Nutritional Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| | - Mohamed A. El-Mokhtar
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt
| |
Collapse
|
7
|
Gopinath A, Mackie PM, Phan LT, Mirabel R, Smith AR, Miller E, Franks S, Syed O, Riaz T, Law BK, Urs N, Khoshbouei H. Who Knew? Dopamine Transporter Activity Is Critical in Innate and Adaptive Immune Responses. Cells 2023; 12:cells12020269. [PMID: 36672204 PMCID: PMC9857305 DOI: 10.3390/cells12020269] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/22/2022] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
The dopamine transporter (DAT) regulates the dimension and duration of dopamine transmission. DAT expression, its trafficking, protein-protein interactions, and its activity are conventionally studied in the CNS and within the context of neurological diseases such as Parkinson's Diseases and neuropsychiatric diseases such as drug addiction, attention deficit hyperactivity and autism. However, DAT is also expressed at the plasma membrane of peripheral immune cells such as monocytes, macrophages, T-cells, and B-cells. DAT activity via an autocrine/paracrine signaling loop regulates macrophage responses to immune stimulation. In a recent study, we identified an immunosuppressive function for DAT, where blockade of DAT activity enhanced LPS-mediated production of IL-6, TNF-α, and mitochondrial superoxide levels, demonstrating that DAT activity regulates macrophage immune responses. In the current study, we tested the hypothesis that in the DAT knockout mice, innate and adaptive immunity are perturbed. We found that genetic deletion of DAT (DAT-/-) results in an exaggerated baseline inflammatory phenotype in peripheral circulating myeloid cells. In peritoneal macrophages obtained from DAT-/- mice, we identified increased MHC-II expression and exaggerated phagocytic response to LPS-induced immune stimulation, suppressed T-cell populations at baseline and following systemic endotoxemia and exaggerated memory B cell expansion. In DAT-/- mice, norepinephrine and dopamine levels are increased in spleen and thymus, but not in circulating serum. These findings in conjunction with spleen hypoplasia, increased splenic myeloid cells, and elevated MHC-II expression, in DAT-/- mice further support a critical role for DAT activity in peripheral immunity. While the current study is only focused on identifying the role of DAT in peripheral immunity, our data point to a much broader implication of DAT activity than previously thought. This study is dedicated to the memory of Dr. Marc Caron who has left an indelible mark in the dopamine transporter field.
Collapse
Affiliation(s)
- Adithya Gopinath
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (A.G.); (H.K.)
| | - Phillip M. Mackie
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Leah T. Phan
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Rosa Mirabel
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Aidan R. Smith
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Emily Miller
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Stephen Franks
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Ohee Syed
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Tabish Riaz
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
| | - Brian K. Law
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Nikhil Urs
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32611, USA
| | - Habibeh Khoshbouei
- Department of Neuroscience, University of Florida, Gainesville, FL 32611, USA
- Correspondence: (A.G.); (H.K.)
| |
Collapse
|
8
|
Molnar K, Riedel H, Schwarz J, Dietz S, Spring B, Haag L, Poets CF, Gille C, Köstlin-Gille N. Group B streptococci infection model shows decreased regulatory capacity of cord blood cells. Pediatr Res 2022; 92:1407-1416. [PMID: 35165359 PMCID: PMC9700511 DOI: 10.1038/s41390-021-01880-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/17/2021] [Accepted: 07/27/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND Sepsis is one of the leading causes of morbidity and mortality in the neonatal period. Compared to adults, neonates are more susceptible to infections, especially to systemic infections with Group B Streptococcus (GBS). Furthermore, neonates show defects in terminating inflammation. The immunological causes for the increased susceptibility to infection and the prolonged inflammatory response are still incompletely understood. METHODS In the present study, we aimed to investigate the reaction of cord blood mononuclear cells (MNC) to stimulation with GBS in comparison to that of MNC from adult blood with focus on the proliferative response in an in vitro infection model with heat-inactivated GBS. RESULTS We demonstrate that after stimulation with GBS the proliferation of T cells from adult blood strongly decreased, while the proliferation of cord blood T cells remained unchanged. This effect could be traced back to a transformation of adult monocytes, but not cord blood monocytes, to a suppressive phenotype with increased expression of the co-inhibitory molecule programmed death ligand 1 (PD-L1). CONCLUSIONS These results point towards an increased inflammatory capacity of neonatal MNC after stimulation with GBS. Targeting the prolonged inflammatory response of neonatal immune cells may be a strategy to prevent complications of neonatal infections. IMPACT Neonatal sepsis often leads to post-inflammatory complications. Causes for sustained inflammation in neonates are incompletely understood. We show that cord blood T cells exhibited increased proliferative capacity after stimulation with group B streptococci (GBS) in comparison to adult T cells. Adult monocytes but not cord blood monocytes acquired suppressive activity and expressed increased levels of PD-L1 after GBS stimulation. Increased proliferative capacity of neonatal T cells and decreased suppressive activity of neonatal monocytes during GBS infection may contribute to prolonged inflammation and development of post-inflammatory diseases in newborns.
Collapse
Affiliation(s)
- Kriszta Molnar
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Hannah Riedel
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Julian Schwarz
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Stefanie Dietz
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Bärbel Spring
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Laura Haag
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Christian F Poets
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| | - Christian Gille
- Department of Neonatology, Heidelberg University Children's Hospital, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany.
| | - Natascha Köstlin-Gille
- Department of Neonatology, Tübingen University Children's Hospital, Calwerstrasse 7, 72076, Tübingen, Germany
| |
Collapse
|
9
|
Li C, Yu X, Zhang L, Peng Y, Zhang T, Li Y, Luan Y, Yin C. The potential role and regulatory mechanism of IL-33/ST2 axis on T lymphocytes during lipopolysaccharide stimulation or perinatal Listeria infection. Int Immunopharmacol 2022; 108:108742. [DOI: 10.1016/j.intimp.2022.108742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/20/2022] [Accepted: 03/29/2022] [Indexed: 11/05/2022]
|
10
|
Kustermann M, Dasari P, Knape I, Keltsch E, Liu J, Pflüger S, Osen W, Holzmann K, Huber-Lang M, Debatin KM, Strauss G. Adoptively Transferred in vitro-Generated Myeloid-Derived Suppressor Cells Improve T-Cell Function and Antigen-Specific Immunity after Traumatic Lung Injury. J Innate Immun 2022; 15:78-95. [PMID: 35691281 PMCID: PMC10643914 DOI: 10.1159/000525088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 05/07/2022] [Indexed: 11/19/2022] Open
Abstract
Immune reactions after trauma are characterized by immediate activation of innate immunity and simultaneously downregulation of adaptive immunity leading to a misbalanced immunohomeostasis and immunosuppression of the injured host. Therefore, the susceptibility to secondary infections is strongly increased after trauma. Immune responses are regulated by a network of immune cells influencing each other and at the same time modifying their functions dependent on the inflammatory environment. Although myeloid-derived suppressor cells (MDSCs) are initially described as T-cell suppressors, their immunomodulatory capacity after trauma is mostly undefined. Therefore, in vitro-generated MDSCs were adoptively transferred into mice after blunt chest trauma (TxT). A single MDSC treatment-induced splenic T-cell expansion decreased apoptosis sensitivity and improved proliferation in the absence of T-cell exhaustion until 2 weeks after trauma. MDSC treatment had a long-lasting effect on the genomic landscape of CD4+ T cells by upregulating primarily Th2-associated genes. Remarkably, immune-activating functions of MDSCs supported the ability of TxT mice to respond to post-traumatic secondary antigen challenge. Secondary insults were mimicked by immunizing MDSC-treated TxT mice with ovalbumin (OVA), followed by OVA restimulation in vitro. MDSC treatment significantly increased the frequency of OVA-specific T cells, enhanced their Th1/Th2 cytokine expression, and induced upregulation of cytolytic molecules finally improving OVA-specific cytotoxicity. Overall, we could show that therapeutic MDSC treatment after TxT improves post-traumatic T-cell functions, which might enable the traumatic host to counterbalance trauma-induced immunoparalysis.
Collapse
Affiliation(s)
- Monika Kustermann
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Prasad Dasari
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Ingrid Knape
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Emma Keltsch
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Jianing Liu
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Silvia Pflüger
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Wolfram Osen
- GMP & T Cell Therapy, German Cancer Research Center, Heidelberg, Germany
| | | | - Markus Huber-Lang
- Institute of Experimental Trauma-Immunology, University Medical Center Ulm, Ulm, Germany
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
11
|
Savio LEB. P2X receptors in the balance between inflammation and pathogen control in sepsis. Purinergic Signal 2022; 18:241-243. [PMID: 35612709 DOI: 10.1007/s11302-022-09870-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 05/15/2022] [Indexed: 11/24/2022] Open
Affiliation(s)
- Luiz Eduardo Baggio Savio
- Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro (UFRJ), Edifício Do Centro de Ciências da Saúde, Bloco G. Av. Carlos Chagas Filho, 373. Cidade Universitária, Ilha Do Fundão, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
12
|
Xie Y, Li J, Qin H, Wang Q, Chen Z, Liu C, Zheng L, Wang J. Paramylon from Euglena gracilis Prevents Lipopolysaccharide-Induced Acute Liver Injury. Front Immunol 2022; 12:797096. [PMID: 35126359 PMCID: PMC8812190 DOI: 10.3389/fimmu.2021.797096] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/15/2021] [Indexed: 12/14/2022] Open
Abstract
Acute liver injury (ALI) is a life-threatening syndrome with high mortality and lacks effective therapies. Rodents under LPS (lipopolysaccharide)/D-Gal (D-galactosamine) stress mimic ALI by presenting dramatically increased inflammation and cell death in the liver. Euglena gracilis, functioning like dietary fiber, is commonly used as a paramylon (Pa)-rich nutritional supplement that has various biological effects such as regulating immune system, anti-obesity, and anti-tumor. Here, we found that Pa or sonicated and alkalized paramylon (SA-Pa) alleviated the LPS/D-Gal-induced hepatic histopathological abnormalities in mice. Compared with Pa, SA-Pa had lower molecular weights/sizes and showed better efficacy in alleviating injury-induced hepatic functions, as well as the transcriptional levels of inflammatory cytokines. Moreover, SA-Pa treatment promoted M2 macrophage activation that enhanced the anti-inflammatory function in the liver, and downregulated STAT3 target genes, such as Fos, Jun, and Socs3 upon the injury. Meanwhile, SA-Pa treatment also alleviated apoptosis and necroptosis caused by the injury. Our results demonstrated that SA-Pa efficiently protected the liver from LPS/D-Gal-induced ALI by alleviating inflammation and cell death.
Collapse
Affiliation(s)
- Yunhao Xie
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jin Li
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China.,College of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, China
| | - Huan Qin
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Qing Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zixi Chen
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| | - Chengyu Liu
- Department of Transfusion Medicine, Wuhan Hospital of Traditional Chinese and Western Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jiangxin Wang
- Shenzhen Key Laboratory of Marine Bioresource and Eco-Environmental Science, Shenzhen Engineering Laboratory for Marine Algal Biotechnology, Guangdong Provincial Key Laboratory for Plant Epigenetics, College of Life Sciences and Oceanography, Shenzhen University, Shenzhen, China
| |
Collapse
|
13
|
Allard D, Allard B, Stagg J. On the mechanism of anti-CD39 immune checkpoint therapy. J Immunother Cancer 2021; 8:jitc-2019-000186. [PMID: 32098829 PMCID: PMC7057429 DOI: 10.1136/jitc-2019-000186] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/28/2019] [Indexed: 12/26/2022] Open
Abstract
With the coming of age of cancer immunotherapy, the search for new therapeutic targets has led to the identification of immunosuppressive adenosine as an important regulator of antitumor immunity. This resulted in the development of selective inhibitors targeting various components of the adenosinergic pathway, including small molecules antagonists targeting the high affinity A2A adenosine receptor and low affinity A2B receptor, therapeutic monoclonal antibodies (mAbs) and small molecules targeting CD73 and therapeutic mAbs targeting CD39. As each regulator of the adenosinergic pathway present non-overlapping biologic functions, a better understanding of the mechanisms of action of each targeted approach should accelerate clinical translation and improve rational design of combination treatments. In this review, we discuss the potential mechanisms-of-action of anti-CD39 cancer therapy and potential toxicities that may emerge from sustained CD39 inhibition. Caution should be taken, however, in extrapolating data from gene-targeted mice to patients treated with blocking anti-CD39 agents. As phase I clinical trials are now underway, further insights into the mechanism of action and potential adverse events associated with anti-CD39 therapy are anticipated in coming years.
Collapse
Affiliation(s)
- David Allard
- Faculty of Pharmacy, Centre Hospitalier de L'Universite de Montreal, Montreal, Quebec, Canada
| | - Bertrand Allard
- Institut du Cancer de Montreal, Centre Hospitalier de L'Universite de Montreal, Montreal, Quebec, Canada
| | - John Stagg
- Faculty of Pharmacy, Centre Hospitalier de L'Universite de Montreal, Montreal, Quebec, Canada .,Institut du Cancer de Montreal, Centre Hospitalier de L'Universite de Montreal, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Xue J, Xie L, Liu B, Zhou L, Hu Y, Ajuwon KM, Fang R. Dietary Supplementation of EGF Ameliorates the Negatively Effects of LPS on Early-Weaning Piglets: From Views of Growth Performance, Nutrient Digestibility, Microelement Absorption and Possible Mechanisms. Animals (Basel) 2021; 11:ani11061598. [PMID: 34071588 PMCID: PMC8227379 DOI: 10.3390/ani11061598] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/22/2021] [Accepted: 05/26/2021] [Indexed: 12/13/2022] Open
Abstract
Simple Summary This study aims to investigate how epidermal growth factor (EGF) attenuates the effect of lipopolysaccharide (LPS) on the growth performance, nutrient digestibility, microelement absorption of early-weaned pigs. A total of 48 early weaned piglets were randomly distributed to four groups consisting of a 2 × 2 factorial design. The main factors were the level of LPS (HLPS = high LPS: 100 μg/kg body weight; ZLPS = low LPS: 0 μg/kg body weight) and EGF (HEGF = high EGF: 2 mg/kg diet; ZEGF = low EGF: 0 mg/kg diet). Each group had four replicates and each replicate consisted of three piglets. The results showed that HLPS level decreased the growth performance and the apparent digestibility of crude fat, while HEGF level increased the average daily feed intake. The concentration of most microelements in the gastrointestinal tract chyme and feces were increased by HLPS level and decreased by HEGF level. The expression levels of most microelement transport-relative genes in the mucosa of gastrointestinal tissues were decreased by HLPS level and increased by HEGF level. In conclusion, dietary EGF could attenuate the negative effect of LPS exposure on the apparent digestibility of crude fat and microelement absorption through changing the expression levels of microelement transport-relative genes. EGF can be used as an additive to increase the essential trace elements absorption in the early weaning piglets. Abstract Epidermal growth factor (EGF) plays an important role in nutrients absorption. However, whether it can be an effective additive to improve the growth performance and nutrients absorption in lipopolysaccharide (LPS) challenged early weaning piglets is still unknown. A 14-days trial was conducted to investigate how EGF attenuates the effect of LPS on the growth performance, nutrient digestibility, microelement absorption of early-weaned pigs, and study the underlying mechanism. A total of 48 early weaned piglets, aged 25 days, were randomly distributed to four groups (control, EGF, LPS and EGF + LPS groups) consisting of a 2 × 2 factorial design. The main factors were the level of LPS (HLPS = high LPS: 100 μg/kg body weight; ZLPS = low LPS: 0 μg/kg body weight) and EGF (HEGF = high EGF: 2 mg/kg diet; ZEGF = low EGF: 0 mg/kg diet). Each group had four replicates and each replicate consisted of three piglets. The results showed that piglets injected with HLPS level significantly decreased the average daily gain (ADG), and significantly increased the feed conversion ratio (FCR) compared with the piglets injected with ZLPS level, while piglets fed HEGF level significantly increased the average daily feed intake (ADFI) compared with the piglets fed ZEGF level (p < 0.05). Piglets injected with HLPS level significantly decreased the apparent digestibility of crude fat compared with the piglets injected with ZLPS level (p < 0.05). Piglets injected with HLPS level significantly increased the concentration of most microelements in the gastrointestinal tract chyme and feces, and significantly decreased the expression levels of most microelement transport-relative genes in the mucosa of gastrointestinal tissues compared with the piglets injected with ZLPS level (p < 0.05). Piglets fed HEGF level significantly decreased the concentration of microelement in the gastrointestinal tract chyme and feces, and significantly increased the expression levels of the microelement transport-relative genes in the mucosa of gastrointestinal tissues compared with the piglets fed ZEGF level (p < 0.05). In conclusion, dietary EGF could attenuate the negative effect of LPS exposure on the apparent digestibility of crude fat and microelement absorption of early-weaning piglets. EGF and LPS influenced the absorption of essential trace element through changing the expression levels of microelement transport-relative genes in the mucosa of gastrointestinal tissues. In the early weaning piglets, EGF can be used as an additive to increase the essential trace elements absorption.
Collapse
Affiliation(s)
- Junjing Xue
- College of Animal Science and Technology, Hunan Agricultural University, No. 1 Nongda Road, Furong District, Changsha 410128, China; (J.X.); (L.X.); (B.L.); (L.Z.); (Y.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Liang Xie
- College of Animal Science and Technology, Hunan Agricultural University, No. 1 Nongda Road, Furong District, Changsha 410128, China; (J.X.); (L.X.); (B.L.); (L.Z.); (Y.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Bo Liu
- College of Animal Science and Technology, Hunan Agricultural University, No. 1 Nongda Road, Furong District, Changsha 410128, China; (J.X.); (L.X.); (B.L.); (L.Z.); (Y.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Liyuan Zhou
- College of Animal Science and Technology, Hunan Agricultural University, No. 1 Nongda Road, Furong District, Changsha 410128, China; (J.X.); (L.X.); (B.L.); (L.Z.); (Y.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Yajun Hu
- College of Animal Science and Technology, Hunan Agricultural University, No. 1 Nongda Road, Furong District, Changsha 410128, China; (J.X.); (L.X.); (B.L.); (L.Z.); (Y.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
| | - Kolapo Matthew Ajuwon
- Department of Animal Sciences, Purdue University, West Lafayette, IN 47907-2054, USA;
| | - Rejun Fang
- College of Animal Science and Technology, Hunan Agricultural University, No. 1 Nongda Road, Furong District, Changsha 410128, China; (J.X.); (L.X.); (B.L.); (L.Z.); (Y.H.)
- Hunan Co-Innovation Center of Animal Production Safety, Changsha 410128, China
- Correspondence: ; Tel.: +86-(0)731-8618177
| |
Collapse
|
15
|
Leite-Aguiar R, Alves VS, Savio LEB, Coutinho-Silva R. Targeting Purinergic Signaling in the Dynamics of Disease Progression in Sepsis. Front Pharmacol 2021; 11:626484. [PMID: 33519492 PMCID: PMC7840482 DOI: 10.3389/fphar.2020.626484] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 12/03/2020] [Indexed: 12/29/2022] Open
Affiliation(s)
- Raíssa Leite-Aguiar
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vinícius Santos Alves
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luiz Eduardo Baggio Savio
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
16
|
Ledderose C, Junger WG. Mitochondria Synergize With P2 Receptors to Regulate Human T Cell Function. Front Immunol 2020; 11:549889. [PMID: 33133068 PMCID: PMC7550529 DOI: 10.3389/fimmu.2020.549889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Intracellular ATP is the universal energy carrier that fuels many cellular processes. However, immune cells can also release a portion of their ATP into the extracellular space. There, ATP activates purinergic receptors that mediate autocrine and paracrine signaling events needed for the initiation, modulation, and termination of cell functions. Mitochondria contribute to these processes by producing ATP that is released. Here, we summarize the synergistic interplay between mitochondria and purinergic signaling that regulates T cell functions. Specifically, we discuss how mitochondria interact with P2X1, P2X4, and P2Y11 receptors to regulate T cell metabolism, cell migration, and antigen recognition. These mitochondrial and purinergic signaling mechanisms are indispensable for host immune defense. However, they also represent an Achilles heel that can render the host susceptible to infections and inflammatory disorders. Hypoxia and mitochondrial dysfunction deflate the purinergic signaling mechanisms that regulate T cells, while inflammation and tissue damage generate excessive systemic ATP levels that distort autocrine purinergic signaling and impair T cell function. An improved understanding of the metabolic and purinergic signaling mechanisms that regulate T cells may lead to novel strategies for the diagnosis and treatment of infectious and inflammatory diseases.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| |
Collapse
|
17
|
Ledderose C, Bromberger S, Slubowski CJ, Sueyoshi K, Aytan D, Shen Y, Junger WG. The purinergic receptor P2Y11 choreographs the polarization, mitochondrial metabolism, and migration of T lymphocytes. Sci Signal 2020; 13:13/651/eaba3300. [PMID: 32994212 DOI: 10.1126/scisignal.aba3300] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
T cells must migrate to encounter antigen-presenting cells and perform their roles in host defense. Here, we found that autocrine stimulation of the purinergic receptor P2Y11 regulates the migration of human CD4 T cells. P2Y11 receptors redistributed from the front to the back of polarized cells where they triggered intracellular cAMP/PKA signals that attenuated mitochondrial metabolism at the back. The absence of P2Y11 receptors at the front of cells resulted in hotspots of mitochondrial metabolism and localized ATP production that stimulated P2X4 receptors, Ca2+ influx, and pseudopod protrusion at the front. This regulatory function of P2Y11 receptors depended on their subcellular redistribution and autocrine stimulation by cellular ATP release and was perturbed by indiscriminate global stimulation. We conclude that excessive extracellular ATP-such as in response to inflammation, sepsis, and cancer-disrupts this autocrine feedback mechanism, which results in defective T cell migration, impaired T cell function, and loss of host immune defense.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Sophie Bromberger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Christian J Slubowski
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Koichiro Sueyoshi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dilan Aytan
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yong Shen
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
18
|
Pulido-Méndez MM, Azuaje E, Rodríguez-Acosta A. Immunotoxicological effects triggered by the rattlesnake Crotalus durissus cumanensis, mapanare ( Bothrops colombiensis) venoms and its purified fractions on spleen and lymph nodes cells. Immunopharmacol Immunotoxicol 2020; 42:484-492. [PMID: 32806962 DOI: 10.1080/08923973.2020.1810272] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Purpose: The snakes in Venezuela vary in their different venom composition amid the species. In this sense, studies have been carried out elucidating mechanisms related to their immunostimulatory and/or immunosuppressive effects in vitro, measuring inhibition or stimulation on the mice spleen and lymph nodes lymphocytes under the rattlesnake (Crotalus durissus cumanensis) (Cdc) and mapanare (Bothrops colombiensis) crude venoms actions, and also its purified fraction crotoxin (CTX) (Cdc) and a semi-purified fraction (SPF) (Bc) activities. Material and methods: The stimulation of lymphocyte proliferation was carried out in the presence or absence of Concanavalin A (ConA) and lipopolysaccharides (LPS). Results: The lymphocyte response was measured by the Alamar Blue® (Resazurin) assay, observing that the Crotalus crude venom increased basal proliferation in the spleen and lymph nodes, being also increased with ConA and LPS. CTX slightly decreased the proliferative response in the presence of mitogens. Both Bc venom and its SPF fraction had no significant effect on basal proliferation in the spleen and lymph nodes, but a decrease in the response with ConA was observed. These results suggest that CTX has an inhibitory action on lymphocyte proliferation, while Cdc crude venom has a stimulatory action on T and B cell populations. Bothrops colombiensis venom had no effect on these two types of cell populations. As it is known, lymphocytes are cells of enormous flexibility and can operate in diverse aspects, warranting that the correct immune response persists controlled. Conclusions: These results suggested that these different toxins can modulate lymphocyte functional activation toward an inhibitory or stimulatory state.
Collapse
Affiliation(s)
- María M Pulido-Méndez
- Laboratory of Immunology, Experimental Medicine Institute, Universidad Central de Venezuela, Caracas, Bolivarian Republic of Venezuela
| | - Elvia Azuaje
- Laboratory of Immunology, Experimental Medicine Institute, Universidad Central de Venezuela, Caracas, Bolivarian Republic of Venezuela
| | - Alexis Rodríguez-Acosta
- Immunochemistry and Ultrastructural Laboratory, Anatomical Institute, Universidad Central de Venezuela, Caracas, Bolivarian Republic of Venezuela
| |
Collapse
|
19
|
de la Rosa G, Gómez AI, Baños MC, Pelegrín P. Signaling Through Purinergic Receptor P2Y 2 Enhances Macrophage IL-1β Production. Int J Mol Sci 2020; 21:ijms21134686. [PMID: 32630144 PMCID: PMC7370188 DOI: 10.3390/ijms21134686] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/19/2020] [Accepted: 06/27/2020] [Indexed: 12/14/2022] Open
Abstract
The release of nucleotides during necrosis or apoptosis has been described to have both proinflammatory and anti-inflammatory effect on the surrounding cells. Here we describe how low concentrations of UTP and ATP applied during macrophage priming enhance IL-1β production when subsequently the NLRP3 inflammasome is activated in murine resident peritoneal macrophages. Deficiency or pharmacological inhibition of the purinergic receptor P2Y2 reverted the increase of IL-1β release induced by nucleotides. IL-1β increase was found dependent on the expression of Il1b gene and probably involving JNK activity. On the contrary, nucleotides decreased the production of a different proinflammatory cytokines such as TNF-α. These results suggest that nucleotides could shape the response of macrophages to obtain a unique proinflammatory signature that might be relevant in unrevealing specific inflammatory conditions.
Collapse
|
20
|
Ledderose C, Bromberger S, Slubowski CJ, Sueyoshi K, Junger WG. Frontline Science: P2Y11 receptors support T cell activation by directing mitochondrial trafficking to the immune synapse. J Leukoc Biol 2020; 109:497-508. [PMID: 32531829 DOI: 10.1002/jlb.2hi0520-191r] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 05/08/2020] [Accepted: 05/23/2020] [Indexed: 12/21/2022] Open
Abstract
T cells form an immune synapse (IS) with antigen-presenting cells (APCs) to detect antigens that match their TCR. Mitochondria, pannexin-1 (panx1) channels, and P2X4 receptors congregate at the IS where mitochondria produce the ATP that panx1 channels release in order to stimulate P2X4 receptors. P2X4 receptor stimulation causes cellular Ca2+ influx that up-regulates mitochondrial metabolism and localized ATP production at the IS. Here we show that P2Y11 receptors are essential players that sustain these T cell activation mechanisms. We found that P2Y11 receptors retract from the IS toward the back of cells where their stimulation by extracellular ATP induces cAMP/PKA signaling that redirects mitochondrial trafficking to the IS. P2Y11 receptors thus reinforce IS signaling by promoting the aggregation of mitochondria with panx1 ATP release channels and P2X4 receptors at the IS. This dual purinergic signaling mechanism involving P2X4 and P2Y11 receptors focuses mitochondrial metabolism to the IS where localized ATP production sustains synaptic activity in order to allow successful completion of T cell activation responses. Our findings have practical implications because rodents lack P2Y11 receptors, raising concerns as to the validity of rodent models to study treatment of infections and inflammatory conditions.
Collapse
Affiliation(s)
- Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Sophie Bromberger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian J Slubowski
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Koichiro Sueyoshi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Zyma M, Pawliczak R. Characteristics and the role of purinergic receptors in pathophysiology with focus on immune response. Int Rev Immunol 2020; 39:97-117. [PMID: 32037918 DOI: 10.1080/08830185.2020.1723582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The nucleotide adenosine-5'-triphosphate (ATP) is mostly thought to be energy carrier, but evidence presented in multiple studies proves ATP involvement into variety of processes, due to its neuromodulatory capabilities. ATP and its metabolite-adenosine, bind to the purinergic receptors, which are divided into two types: adenosine binding P1 receptor and ADP/ATP binding P2 receptor. These receptors are expressed in different tissues and organs. Recent studies report their immunomodulatory characteristics, connected with varying immunological processes, such as immunological response or antigen presentation. Besides, they seem to play an important role in medical conditions such as bronchial asthma or variety of cancers. In this article, we would like to review recent discoveries on the field of purinergic receptors research focusing on their role in immunological system, and shed a new light upon the importance of these receptors in modern medicine development.
Collapse
Affiliation(s)
- Marharyta Zyma
- Department of Immunopathology, Division of Biomedical Science, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Division of Biomedical Science, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
22
|
Denning NL, Aziz M, Gurien SD, Wang P. DAMPs and NETs in Sepsis. Front Immunol 2019; 10:2536. [PMID: 31736963 PMCID: PMC6831555 DOI: 10.3389/fimmu.2019.02536] [Citation(s) in RCA: 376] [Impact Index Per Article: 62.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 10/11/2019] [Indexed: 12/21/2022] Open
Abstract
Sepsis is a deadly inflammatory syndrome caused by an exaggerated immune response to infection. Much has been focused on host response to pathogens mediated through the interaction of pathogen-associated molecular patterns (PAMPs) and pattern recognition receptors (PRRs). PRRs are also activated by host nuclear, mitochondrial, and cytosolic proteins, known as damage-associated molecular patterns (DAMPs) that are released from cells during sepsis. Some well described members of the DAMP family are extracellular cold-inducible RNA-binding protein (eCIRP), high mobility group box 1 (HMGB1), histones, and adenosine triphosphate (ATP). DAMPs are released from the cell through inflammasome activation or passively following cell death. Similarly, neutrophil extracellular traps (NETs) are released from neutrophils during inflammation. NETs are webs of extracellular DNA decorated with histones, myeloperoxidase, and elastase. Although NETs contribute to pathogen clearance, excessive NET formation promotes inflammation and tissue damage in sepsis. Here, we review DAMPs and NETs and their crosstalk in sepsis with respect to their sources, activation, release, and function. A clear grasp of DAMPs, NETs and their interaction is crucial for the understanding of the pathophysiology of sepsis and for the development of novel sepsis therapeutics.
Collapse
Affiliation(s)
- Naomi-Liza Denning
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Monowar Aziz
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States
| | - Steven D Gurien
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, United States.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Ping Wang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Manhasset, NY, United States.,Elmezzi Graduate School of Molecular Medicine, Manhasset, NY, United States.,Department of Surgery, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States.,Department of Molecular Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
23
|
Karmakar M, Dubyak GR. Microbes mediate mitochondrial misinformation to misguide neutrophils. J Leukoc Biol 2019; 106:1197-1200. [PMID: 31633233 DOI: 10.1002/jlb.3ce0819-263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Discussion on LPS disruption of mitochondrial localization and autocrine purinergic signaling in neutrophil chemotaxis for control of E. coli infection.
Collapse
Affiliation(s)
- Mausita Karmakar
- Department of Physiology & Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| | - George R Dubyak
- Department of Physiology & Biophysics, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
24
|
Kondo Y, Ledderose C, Slubowski CJ, Fakhari M, Sumi Y, Sueyoshi K, Bezler AK, Aytan D, Arbab M, Junger WG. Frontline Science: Escherichia coli use LPS as decoy to impair neutrophil chemotaxis and defeat antimicrobial host defense. J Leukoc Biol 2019; 106:1211-1219. [PMID: 31392789 DOI: 10.1002/jlb.4hi0319-109r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 06/14/2019] [Accepted: 07/24/2019] [Indexed: 12/17/2022] Open
Abstract
Bacterial infections and sepsis are leading causes of morbidity and mortality in critically ill patients. Currently, there are no effective treatments available to improve clinical outcome in sepsis. Here, we elucidated a mechanism by which Escherichia coli (E. coli) bacteria impair neutrophil (PMN) chemotaxis and we studied whether this mechanism can be therapeutically targeted to improve chemotaxis and antimicrobial host defense. PMNs detect bacteria with formyl peptide receptors (FPR). FPR stimulation triggers mitochondrial ATP production and release. Autocrine stimulation of purinergic receptors exerts excitatory and inhibitory downstream signals that induce cell polarization and cell shape changes needed for chemotaxis. Here we show that the bacterial cell wall product LPS dose-dependently impairs PMN chemotaxis. Exposure of human PMNs to LPS triggered excessive mitochondrial ATP production and disorganized intracellular trafficking of mitochondria, resulting in global ATP release that disrupted purinergic signaling, cell polarization, and chemotaxis. In mice infected i.p. with E. coli, LPS treatment increased the spread of bacteria at the infection site and throughout the systemic circulation. Removal of excessive systemic ATP with apyrase improved chemotaxis of LPS-treated human PMNs in vitro and enhanced the clearance of E. coli in infected and LPS-treated mice. We conclude that systemic ATP accumulation in response to LPS is a potential therapeutic target to restore PMN chemotaxis and to boost the antimicrobial host immune defense in sepsis.
Collapse
Affiliation(s)
- Yutaka Kondo
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Carola Ledderose
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Christian J Slubowski
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mahtab Fakhari
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Yuka Sumi
- Department of Emergency and Critical Care Medicine, Juntendo University, Urayasu Hospital, Chiba, Japan
| | - Koichiro Sueyoshi
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ann-Katrin Bezler
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Dilan Aytan
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Mona Arbab
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Wolfgang G Junger
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Ludwig Boltzmann Institute for Traumatology, Vienna, Austria
| |
Collapse
|