1
|
Di Virgilio F, Vultaggio-Poma V, Tarantini M, Giuliani AL. Overview of the role of purinergic signaling and insights into its role in cancer therapy. Pharmacol Ther 2024; 262:108700. [PMID: 39111410 DOI: 10.1016/j.pharmthera.2024.108700] [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: 07/05/2024] [Accepted: 07/31/2024] [Indexed: 08/30/2024]
Abstract
Innovation of cancer therapy has received a dramatic acceleration over the last fifteen years thanks to the introduction of the novel immune checkpoint inhibitors (ICI). On the other hand, the conspicuous scientific knowledge accumulated in purinergic signaling since the early seventies is finally being transferred to the clinic. Several Phase I/II clinical trials are currently underway to investigate the effect of drugs interfering with purinergic signaling as stand-alone or combination therapy in cancer. This is supporting the novel concept of "purinergic immune checkpoint" (PIC) in cancer therapy. In the present review we will address a) the basic pharmacology and cell biology of the purinergic system; b) principles of its pathophysiology in human diseases; c) implications for cell death, cell proliferation and cancer; d) novel molecular tools to investigate nucleotide homeostasis in the extracellular environment; e) recent developments in the pharmacology of P1, P2 receptors and related ecto-enzymes; f) P1 and P2 ligands as novel diagnostic tools; g) current issues in PIC-based anti-cancer therapy. This review will provide an appraisal of the current status of purinergic signaling in cancer and will help identify future avenues of development.
Collapse
Affiliation(s)
| | | | - Mario Tarantini
- Department of Medical Sciences, University of Ferrara, Italy
| | | |
Collapse
|
2
|
Zhang H, Felthaus O, Eigenberger A, Klein S, Prantl L. Treg Cell Therapeutic Strategies for Breast Cancer: Holistic to Local Aspects. Cells 2024; 13:1526. [PMID: 39329710 PMCID: PMC11429654 DOI: 10.3390/cells13181526] [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: 07/30/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
Regulatory T cells (Tregs) play a key role in maintaining immune homeostasis and preventing autoimmunity through their immunosuppressive function. There have been numerous reports confirming that high levels of Tregs in the tumor microenvironment (TME) are associated with a poor prognosis, highlighting their role in promoting an immunosuppressive environment. In breast cancer (BC), Tregs interact with cancer cells, ultimately leading to the suppression of immune surveillance and promoting tumor progression. This review discusses the dual role of Tregs in breast cancer, and explores the controversies and therapeutic potential associated with targeting these cells. Researchers are investigating various strategies to deplete or inhibit Tregs, such as immune checkpoint inhibitors, cytokine antagonists, and metabolic inhibition. However, the heterogeneity of Tregs and the variable precision of treatments pose significant challenges. Understanding the functional diversity of Tregs and the latest advances in targeted therapies is critical for the development of effective therapies. This review highlights the latest approaches to Tregs for BC treatment that both attenuate Treg-mediated immunosuppression in tumors and maintain immune tolerance, and advocates precise combination therapy strategies to optimize breast cancer outcomes.
Collapse
Affiliation(s)
- Hanwen Zhang
- Department of Plastic, Hand and Reconstructive Surgery, University Hospital Regensburg, Franz-Josef-Strauss Allee 11, 93053 Regensburg, Germany (L.P.)
| | | | | | | | | |
Collapse
|
3
|
Kiaie SH, Hatami Z, Nasr MS, Pazooki P, Hemmati S, Baradaran B, Valizadeh H. Pharmacological interaction and immune response of purinergic receptors in therapeutic modulation. Purinergic Signal 2024; 20:321-343. [PMID: 37843749 PMCID: PMC11303644 DOI: 10.1007/s11302-023-09966-7] [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: 05/12/2023] [Accepted: 09/10/2023] [Indexed: 10/17/2023] Open
Abstract
Nucleosides and purine nucleotides serve as transmitter and modulator agents that extend their functions beyond the cell. In this context, purinergic signaling plays a crucial role in regulating energy homeostasis and modulating metabolic alterations in tumor cells. Therefore, it is essential to consider the pharmacological targeting of purinergic receptors (PUR), which encompass the expression and inhibition of P1 receptors (metabotropic adenosine receptors) as well as P2 receptors (extracellular ATP/ADP) comprising P2X and P2Y receptors. Thus, the pharmacological interaction between inhibitors (such as RNA, monoclonal antibodies, and small molecules) and PUR represents a key aspect in facilitating the development of therapeutic interventions. Moreover, this review explores recent advancements in pharmacological inhibitors and the regulation of innate and adaptive immunity of PUR, specifically in relation to immunological and inflammatory responses. These responses encompass the release of pro-inflammatory cytokines (PIC), the production of reactive oxygen and nitrogen species (ROS and RNS), the regulation of T cells, and the activation of inflammasomes in all human leukocytes.
Collapse
Affiliation(s)
- Seyed Hossein Kiaie
- Drug Applied Research Center, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Hatami
- Department of Immunology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Mohammad Sadegh Nasr
- Department of Computer Science and Engineering Multi-Interprofessional Center for Health Informatics (MICHI), The University of Texas at Arlington, Arlington, TX, USA
| | - Pouya Pazooki
- Cellular and Molecular Biology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Salar Hemmati
- Institute Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Hadi Valizadeh
- Drug Applied Research Center, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
4
|
Sossou D, Ezinmegnon S, Agbota G, Gbedande K, Accrombessi M, Massougbodji A, d’Almeida M, Alao JM, Dossou-Dagba I, Pachot A, Vachot L, Brengel-Pesce K, Cottrell G, Yessoufou A, Briand V, Tissières P, Fievet N. Regulatory T cell homing and activation is a signature of neonatal sepsis. Front Immunol 2024; 15:1420554. [PMID: 39072327 PMCID: PMC11272980 DOI: 10.3389/fimmu.2024.1420554] [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: 04/20/2024] [Accepted: 06/13/2024] [Indexed: 07/30/2024] Open
Abstract
Regulatory T cells (Treg) play a prominent role in utero tolerating non-inherited maternal antigens and in regulating immune responses against pathogens at birth. This study investigates Treg immunity in newborns in West Africa, where sepsis remains a major public health problem. Treg phenotypes on neonates subgroups with early-onset sepsis (EOS), presumed sepsis, and healthy newborn with and without prenatal risk factors were evaluated. Treg phenotypes varied according to prenatal conditions, with increase in Treg frequency and Foxp3 expression in healthy newborns with prenatal risk factors compared to those with none risk. Compared to healthy newborns with prenatal risk factors, EOS neonates had a significantly reduced frequency of Treg and Foxp3 expression. In the Treg pool, higher frequency of activated Treg was observed in EOS neonates, suggesting an in-utero activation upstream of the sepsis onset. Their migration to the infection site may explain the reduced frequency of circulating Integrin α4β1+ Treg suggestive of homing to the endothelial tissue. EOS neonates show increases expression of CTLA-4, PD-1 and CD39 on Treg, which negatively regulate the activation of effector T cells (Teff) corroborating by the lower frequency of Teff in EOS neonates. The higher frequency of CD39+ Treg and the lower frequency of integrinα4β1+ Treg in EOS non-survivor suggests that Treg exhaustement and endothelial homing are associated with outcome severity. Neonates developing EOS are born with an altered Treg phenotypic profile. Treg expression of CTLA-4, PD-1, CD39, and integrinα4β1 cell markers can be considered as early warning or diagnostic markers of EOS.
Collapse
Affiliation(s)
- Darius Sossou
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
- Faculty of Sciences and Technology (FAST), University of Abomey-Calavi, Institute of Applied Biomedical Sciences (ISBA), Laboratory of Cell Biology and Physiology, Cotonou, Benin
- Institut de Recherche Clinique du Bénin (IRCB), Calavi, Benin
| | - Sem Ezinmegnon
- Faculty of Sciences and Technology (FAST), University of Abomey-Calavi, Institute of Applied Biomedical Sciences (ISBA), Laboratory of Cell Biology and Physiology, Cotonou, Benin
- Fédérations Hospitalo-Universitaires (FHU) Sepsis, AP-HP/Université Paris Saclay/Inserm, Le Kremlin-Bicêtre, France
| | - Gino Agbota
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Calavi, Benin
| | - Komi Gbedande
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Calavi, Benin
| | - Manfred Accrombessi
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
- Institut de Recherche Clinique du Bénin (IRCB), Calavi, Benin
| | | | - Marceline d’Almeida
- Pediatric Department, National University Hospital Center (CNHU), Cotonou, Benin
| | - Jules M. Alao
- Pediatric Department, Mother and Child University and Hospital Center (CHUMEL), Cotonou, Benin
| | | | - Alexandre Pachot
- Medical Diagnostic Discovery Department, bioMérieux, Marcy l’Etoile, France
| | - Laurence Vachot
- Medical Diagnostic Discovery Department, bioMérieux, Marcy l’Etoile, France
| | | | - Gilles Cottrell
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
| | - Akadiri Yessoufou
- Faculty of Sciences and Technology (FAST), University of Abomey-Calavi, Institute of Applied Biomedical Sciences (ISBA), Laboratory of Cell Biology and Physiology, Cotonou, Benin
| | - Valérie Briand
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
| | - Pierre Tissières
- Fédérations Hospitalo-Universitaires (FHU) Sepsis, AP-HP/Université Paris Saclay/Inserm, Le Kremlin-Bicêtre, France
- Institute of Integrative Biology of the Cell (I2BC), Centre National de la Recherche Scientifique (CNRS), Commissariat à l'Energie Atomique et aux énergies alternatives (CEA), University Paris Saclay, Gif-sur-Yvette, France
- Pediatric Intensive Care and Neonatal Medicine, Assistance Publique - Hôpitaux de Paris (AP-HP) Paris Saclay University, Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Nadine Fievet
- Paris-City University, Mére et Enfants en Milieu Tropical: pathogénes, systéme de santé et transition épidémiologique (MERIT), Institute of Research for Development (IRD), Paris, France
| |
Collapse
|
5
|
Qin H, Teng Y, Dai R, Wang A, Liu J. Glycan-based scaffolds and nanoparticles as drug delivery system in cancer therapy. Front Immunol 2024; 15:1395187. [PMID: 38799466 PMCID: PMC11116596 DOI: 10.3389/fimmu.2024.1395187] [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: 03/03/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024] Open
Abstract
Glycan-based scaffolds are unique in their high specificity, versatility, low immunogenicity, and ability to mimic natural carbohydrates, making them attractive candidates for use in cancer treatment. These scaffolds are made up of glycans, which are biopolymers with well biocompatibility in the human body that can be used for drug delivery. The versatility of glycan-based scaffolds allows for the modulation of drug activity and targeted delivery to specific cells or tissues, which increases the potency of drugs and reduces side effects. Despite their promise, there are still technical challenges in the design and production of glycan-based scaffolds, as well as limitations in their therapeutic efficacy and specificity.
Collapse
Affiliation(s)
- Henan Qin
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yibin Teng
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Rui Dai
- Department of Pharmacy, Peking Union Medical University Hospital, Beijing, China
| | - Aman Wang
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Oncology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
6
|
Giannitelli SM, Peluzzi V, Raniolo S, Roscilli G, Trombetta M, Mozetic P, Rainer A. On-chip recapitulation of the tumor microenvironment: A decade of progress. Biomaterials 2024; 306:122482. [PMID: 38301325 DOI: 10.1016/j.biomaterials.2024.122482] [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: 08/29/2023] [Revised: 01/17/2024] [Accepted: 01/20/2024] [Indexed: 02/03/2024]
Abstract
One of the hurdles to the development of new anticancer therapies is the lack of in vitro models which faithfully reproduce the in vivo tumor microenvironment (TME). Understanding the dynamic relationships between the components of the TME in a controllable, scalable, and reliable setting would indeed support the discovery of biological targets impacting cancer diagnosis and therapy. Cancer research is increasingly shifting from traditional two-dimensional (2D) cell culture toward three-dimensional (3D) culture models, which have been demonstrated to increase the significance and predictive value of in vitro data. In this scenario, microphysiological systems (also known as organs-on-chip) have emerged as a relevant technological platform enabling more predictive investigation of cell-cell and cell-ECM interplay in cancer, attracting a significant research effort in the last years. This review illustrates one decade of progress in the field of tumor-microenvironment-on-chip (TMOC) approaches, exploiting either cell-laden microfluidic chambers or microfluidic confined tumor spheroids to model the TME. TMOCs have been designed to recapitulate several aspects of the TME, including tumor cells, the tumor-associated stroma, the immune system, and the vascular component. Significantly, the last aspect has emerged for its pivotal role in orchestrating cellular interactions and modulating drug pharmacokinetics on-chip. A further advancement has been represented by integration of TMOCs into multi-organ microphysiological systems, with the final aim to follow the metastatic cascade to target organs and to study the effects of chemotherapies at a systemic level. We highlight that the increased degree of complexity achieved by the most advanced TMOC models has enabled scientists to shed new light on the role of microenvironmental factors in tumor progression, metastatic cascade, and response to drugs.
Collapse
Affiliation(s)
- S M Giannitelli
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, via Álvaro del Portillo, 21, 00128, Rome, Italy.
| | - V Peluzzi
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128, Rome, Italy.
| | - S Raniolo
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, via Álvaro del Portillo, 21, 00128, Rome, Italy.
| | - G Roscilli
- Takis s.r.l., Via di Castel Romano 100, 00128, Rome, Italy.
| | - M Trombetta
- Department of Science and Technology for Sustainable Development and One Health, Università Campus Bio-Medico di Roma, via Álvaro del Portillo, 21, 00128, Rome, Italy.
| | - P Mozetic
- Institute of Nanotechnology (NANOTEC), National Research Council, via Monteroni, 73100, Lecce, Italy.
| | - A Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, via Álvaro del Portillo 21, 00128, Rome, Italy; Fondazione Policlinico Universitario Campus Bio-Medico di Roma, via Álvaro del Portillo 200, 00128, Rome, Italy.
| |
Collapse
|
7
|
Burk AC, Apostolova P. Metabolic instruction of the graft-versus-leukemia immunity. Front Immunol 2024; 15:1347492. [PMID: 38500877 PMCID: PMC10944922 DOI: 10.3389/fimmu.2024.1347492] [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: 11/30/2023] [Accepted: 02/05/2024] [Indexed: 03/20/2024] Open
Abstract
Allogeneic hematopoietic cell transplantation (allo-HCT) is frequently performed to cure hematological malignancies, such as acute myeloid leukemia (AML), through the graft-versus-leukemia (GVL) effect. In this immunological process, donor immune cells eliminate residual cancer cells in the patient and exert tumor control through immunosurveillance. However, GVL failure and subsequent leukemia relapse are frequent and associated with a dismal prognosis. A better understanding of the mechanisms underlying AML immune evasion is essential for developing novel therapeutic strategies to boost the GVL effect. Cellular metabolism has emerged as an essential regulator of survival and cell fate for both cancer and immune cells. Leukemia and T cells utilize specific metabolic programs, including the orchestrated use of glucose, amino acids, and fatty acids, to support their growth and function. Besides regulating cell-intrinsic processes, metabolism shapes the extracellular environment and plays an important role in cell-cell communication. This review focuses on recent advances in the understanding of how metabolism might affect the anti-leukemia immune response. First, we provide a general overview of the mechanisms of immune escape after allo-HCT and an introduction to leukemia and T cell metabolism. Further, we discuss how leukemia and myeloid cell metabolism contribute to an altered microenvironment that impairs T cell function. Next, we review the literature linking metabolic processes in AML cells with their inhibitory checkpoint ligand expression. Finally, we focus on recent findings concerning the role of systemic metabolism in sustained GVL efficacy. While the majority of evidence in the field still stems from basic and preclinical studies, we discuss translational findings and propose further avenues for bridging the gap between bench and bedside.
Collapse
Affiliation(s)
- Ann-Cathrin Burk
- German Cancer Consortium (DKTK), partner site Freiburg, a partnership between DKFZ and Medical Center - University of Freiburg, Freiburg, Germany
- Department of Medicine I, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Petya Apostolova
- Department of Biomedicine, University Hospital Basel and University of Basel, Basel, Switzerland
- Division of Hematology, University Hospital Basel, Basel, Switzerland
| |
Collapse
|
8
|
Villa M, Sanin DE, Apostolova P, Corrado M, Kabat AM, Cristinzio C, Regina A, Carrizo GE, Rana N, Stanczak MA, Baixauli F, Grzes KM, Cupovic J, Solagna F, Hackl A, Globig AM, Hässler F, Puleston DJ, Kelly B, Cabezas-Wallscheid N, Hasselblatt P, Bengsch B, Zeiser R, Sagar, Buescher JM, Pearce EJ, Pearce EL. Prostaglandin E 2 controls the metabolic adaptation of T cells to the intestinal microenvironment. Nat Commun 2024; 15:451. [PMID: 38200005 PMCID: PMC10781727 DOI: 10.1038/s41467-024-44689-2] [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: 03/14/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Immune cells must adapt to different environments during the course of an immune response. Here we study the adaptation of CD8+ T cells to the intestinal microenvironment and how this process shapes the establishment of the CD8+ T cell pool. CD8+ T cells progressively remodel their transcriptome and surface phenotype as they enter the gut wall, and downregulate expression of mitochondrial genes. Human and mouse intestinal CD8+ T cells have reduced mitochondrial mass, but maintain a viable energy balance to sustain their function. We find that the intestinal microenvironment is rich in prostaglandin E2 (PGE2), which drives mitochondrial depolarization in CD8+ T cells. Consequently, these cells engage autophagy to clear depolarized mitochondria, and enhance glutathione synthesis to scavenge reactive oxygen species (ROS) that result from mitochondrial depolarization. Impairing PGE2 sensing promotes CD8+ T cell accumulation in the gut, while tampering with autophagy and glutathione negatively impacts the T cell pool. Thus, a PGE2-autophagy-glutathione axis defines the metabolic adaptation of CD8+ T cells to the intestinal microenvironment, to ultimately influence the T cell pool.
Collapse
Affiliation(s)
- Matteo Villa
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany.
- Division of Rheumatology and Immunology, Department of Internal Medicine, Medical University of Graz, 8036, Graz, Austria.
| | - David E Sanin
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Petya Apostolova
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Medicine I (Hematology and Oncology), University Medical Center Freiburg, 79106, Freiburg, Germany
| | - Mauro Corrado
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Agnieszka M Kabat
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Carmine Cristinzio
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Department of Medical Biotechnology, University of Siena, Siena, Italy
| | - Annamaria Regina
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Department of Life Sciences, University of Trieste, 34128, Trieste, Italy
| | - Gustavo E Carrizo
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Nisha Rana
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Michal A Stanczak
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Francesc Baixauli
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Katarzyna M Grzes
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Jovana Cupovic
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Francesca Solagna
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Alexandra Hackl
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Anna-Maria Globig
- Department of Medicine II, University Medical Center Freiburg, 79106, Freiburg, Germany
| | - Fabian Hässler
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Daniel J Puleston
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Beth Kelly
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | | | - Peter Hasselblatt
- Department of Medicine II, University Medical Center Freiburg, 79106, Freiburg, Germany
| | - Bertram Bengsch
- Department of Medicine II, University Medical Center Freiburg, 79106, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, Freiburg, Germany
| | - Robert Zeiser
- Department of Medicine I (Hematology and Oncology), University Medical Center Freiburg, 79106, Freiburg, Germany
- CIBSS Centre for Integrative Biological Signalling Studies, Freiburg, Germany
| | - Sagar
- Department of Medicine II, University Medical Center Freiburg, 79106, Freiburg, Germany
| | - Joerg M Buescher
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
| | - Edward J Pearce
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- CIBSS Centre for Integrative Biological Signalling Studies, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - Erika L Pearce
- Max Planck Institute for Immunobiology and Epigenetics, 79108, Freiburg, Germany.
- Bloomberg-Kimmel Institute of Immunotherapy, Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- CIBSS Centre for Integrative Biological Signalling Studies, Freiburg, Germany.
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
9
|
Amer EI, Allam SR, Hassan AY, El-Fakharany EM, Agwa MM, Khattab SN, Sheta E, El-Faham MH. Can antibody conjugated nanomicelles alter the prospect of antibody targeted therapy against schistosomiasis mansoni? PLoS Negl Trop Dis 2023; 17:e0011776. [PMID: 38039267 PMCID: PMC10691730 DOI: 10.1371/journal.pntd.0011776] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 11/07/2023] [Indexed: 12/03/2023] Open
Abstract
BACKGROUND CLA (conjugated linoleic acid)-mediated activation of the schistosome tegument-associated sphingomyelinase and consequent disruption of the outer membrane might allow host antibodies to access the apical membrane antigens. Here, we investigated a novel approach to enhance specific antibody delivery to concealed surface membrane antigens of Schistosoma mansoni utilising antibody-conjugated-CLA nanomicelle technology. METHODOLOGY/PRINCIPAL FINDINGS We invented and characterised an amphiphilic CLA-loaded whey protein co-polymer (CLA-W) as an IV injectable protein nanocarrier. Rabbit anti-Schistosoma mansoni infection (anti-SmI) and anti-Schistosoma mansoni alkaline phosphatase specific IgG antibodies were purified from rabbit sera and conjugated to the surface of CLA-W co-polymer to form antibody-conjugated-CLA-W nanomicelles (Ab-CLA-W). We investigated the schistosomicidal effects of CLA-W and Ab-CLA-W in a mouse model of Schistosoma mansoni against early and late stages of infection. Results showed that conjugation of nanomicelles with antibodies, namely anti-SmI, significantly enhanced the micelles' schistosomicidal and anti-pathology activities at both the schistosomula and adult worm stages of the infection resulting in 64.6%-89.9% reductions in worm number; 72.5-94% and 66.4-85.2% reductions in hepatic eggs and granulomas, respectively. Treatment induced overall improvement in liver histopathology, reducing granuloma size and fibrosis and significantly affecting egg viability. Indirect immunofluorescence confirmed CLA-W-mediated antigen exposure on the worm surface. Electron microscopy revealed extensive ultrastructural damage in worm tegument induced by anti-SmI-CLA-W. CONCLUSION/SIGNIFICANCE The novel antibody-targeted nano-sized CLA delivery system offers great promise for treatment of Schistosoma mansoni infection and control of its transmission. Our in vivo observations confirm an immune-mediated enhanced effect of the schistosomicidal action of CLA and hints at the prospect of nanotechnology-based immunotherapy, not only for schistosomiasis, but also for other parasitic infections in which chemotherapy has been shown to be immune-dependent. The results propose that the immunodominant reactivity of the anti-SmI serum, Schistosoma mansoni fructose biphosphate aldolase, SmFBPA, merits serious attention as a therapeutic and vaccine candidate.
Collapse
Affiliation(s)
- Eglal I. Amer
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Sonia R. Allam
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Aceel Y. Hassan
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Esmail M. El-Fakharany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg EL-Arab, Alexandria, Egypt
| | - Mona M. Agwa
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, Dokki, Giza, Egypt
| | - Sherine N. Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Eman Sheta
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Marwa H. El-Faham
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
10
|
Moghaddam MZ, Mousavi MJ, Ghotloo S. Cell-based therapies for the treatment of rheumatoid arthritis. Immun Inflamm Dis 2023; 11:e1091. [PMID: 38018576 PMCID: PMC10664399 DOI: 10.1002/iid3.1091] [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: 04/17/2023] [Revised: 11/01/2023] [Accepted: 11/03/2023] [Indexed: 11/30/2023] Open
Abstract
Autoimmune diseases, including rheumatoid arthritis that is the most prevalent rheumatic autoimmune disorder, affect autologous connective tissues caused by the breakdown of the self-tolerance mechanisms of the immune system. During the last two decades, cell-based therapy, including stem cells and none-stem cells has been increasingly considered as a therapeutic option in various diseases. This is partly due to the unique properties of stem cells that divide and differentiate from the specialized cells in the damaged tissue. Moreover, stem cells and none-stem cells, impose immunomodulatory properties affecting the diseases caused by immunological abnormalities such as rheumatic autoimmune disorders. In the present review, the efficacy of cell-based therapy with four main types of stem cells, including mesenchymal stem cells, hematopoietic stem cells, embryonic stem cells, and human amniotic membrane cells, as well as none-stem cells, including regulatory T cells, chimeric antigen receptor T cells, and tolerogenic dendritic cells will be evaluated. Moreover, other related issues, including safety, changes in immunological parameters, suitable choice of stem cell and none-stem cell origin, conditioning regimen, limitations, and complications will be discussed.
Collapse
Affiliation(s)
| | - Mohammad Javad Mousavi
- Department of HematologyFaculty of Allied Medicine, Bushehr University of Medical SciencesBushehrIran
| | - Somayeh Ghotloo
- Autoimmune Diseases Research CenterKashan University of Medical SciencesKashanIran
- Department of Clinical Laboratory SciencesKashan University of Medical SciencesKashanIran
| |
Collapse
|
11
|
Hamed A, Ghareeb D, Mohamed TM, Hamed M, Nofal MS, Gaber M. Caffeine-folic acid-loaded-chitosan nanoparticles combined with methotrexate as a novel HepG2 immunotherapy targeting adenosine A2A receptor downstream cascade. BMC Complement Med Ther 2023; 23:384. [PMID: 37891562 PMCID: PMC10604858 DOI: 10.1186/s12906-023-04212-4] [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: 02/11/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND Methotrexate (MTX) is a common chemotherapeutic drug that inhibits DNA synthesis and induces apoptosis. Treatment with MTX increased CD73 expression, which leads to higher levels of extracellular adenosine. Adenosine levels are also high in the tumor microenvironment through Cancer cells metabolism. That promotes the survival of cancer cells and contributes to tumor immune evasion through the Adenosine 2a Receptor. A2A receptor antagonists are an emerging class of agents that treat cancers by enhancing immunotherapy, both as monotherapy and in combination with other therapeutic agents. Caffeine is an adenosine receptor antagonist. Herein, we demonstrate the ability of a novel well prepared and characterized nano formula CAF-FA-CS-NPs (D4) for A2aR blockade when combination with MTX to improve its antitumor efficacy by enhancing the immune system and eliminating immune suppression. METHODS CAF-FA-CS-NPs (D4) were prepared and characterized for particle size, loading efficiency, and release profile. Molecular docking was used to validate the binding affinity of caffeine and folic acid to A2A receptor. The effects of the nano formula were evaluated on human liver cancer cells (HepG2), breast cancer cells (MCF-7), and MDA-MB-231, as well as normal human cells (WI-38). Different combination ratios of MTX and D4 were studied to identify the optimal combination for further genetic studies. RESULTS Molecular docking results validated that caffeine and folic acid have binding affinity to A2A receptor. The CS-NPs were successfully prepared using ionic gelation method, with caffeine and folic acid being loaded and conjugated to the nanoparticles through electrostatic interactions. The CAF loading capacity in D4 was 77.9 ± 4.37% with an encapsulation efficiency of 98.5 ± 0.37. The particle size was optimized through ratio variations. The resulting nanoparticles were fully characterized. The results showed that (D4) had antioxidant activity and cytotoxicity against different cancer cells. The combination of D4 with MTX (IC50 D4 + 0.5 IC50 MTX) resulted in the downregulation of Bcl-2, FOXP3, CD39, and CD73 gene expression levels and upregulation of Bax and A2AR gene expression levels in HepG2 cells. CONCLUSIONS This study suggests that CAF-FA-CS-NPs (D4) in combination with MTX may be a promising candidate for cancer immunotherapy, by inhibiting A2aR signaling and leading to improved immune activation and anti-tumor activity of MTX.
Collapse
Affiliation(s)
- Alaa Hamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt.
| | - Doaa Ghareeb
- Bio-Screening and Preclinical Trial Lab, Biochemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Tarek M Mohamed
- Biochemistry Division, Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| | - Mahmoud Hamed
- Pharmaceutical Services Center, Faculty of Pharmacy, Tanta University, Tanta, 31111, Egypt
| | - Mohammed S Nofal
- Center of Excellency for Drug Preclinical Studies (CE-DPS), Pharmaceutical and Fermentation Industries Development Centre, City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab, Alexandria, Egypt
| | - M Gaber
- Chemistry Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
12
|
Liao L, Xu H, Zhao Y, Zheng X. Metabolic interventions combined with CTLA-4 and PD-1/PD-L1 blockade for the treatment of tumors: mechanisms and strategies. Front Med 2023; 17:805-822. [PMID: 37897562 DOI: 10.1007/s11684-023-1025-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/16/2023] [Indexed: 10/30/2023]
Abstract
Immunotherapies based on immune checkpoint blockade (ICB) have significantly improved patient outcomes and offered new approaches to cancer therapy over the past decade. To date, immune checkpoint inhibitors (ICIs) of CTLA-4 and PD-1/PD-L1 represent the main class of immunotherapy. Blockade of CTLA-4 and PD-1/PD-L1 has shown remarkable efficacy in several specific types of cancers, however, a large subset of refractory patients presents poor responsiveness to ICB therapy; and the underlying mechanism remains elusive. Recently, numerous studies have revealed that metabolic reprogramming of tumor cells restrains immune responses by remodeling the tumor microenvironment (TME) with various products of metabolism, and combination therapies involving metabolic inhibitors and ICIs provide new approaches to cancer therapy. Nevertheless, a systematic summary is lacking regarding the manner by which different targetable metabolic pathways regulate immune checkpoints to overcome ICI resistance. Here, we demonstrate the generalized mechanism of targeting cancer metabolism at three crucial immune checkpoints (CTLA-4, PD-1, and PD-L1) to influence ICB therapy and propose potential combined immunotherapeutic strategies co-targeting tumor metabolic pathways and immune checkpoints.
Collapse
Affiliation(s)
- Liming Liao
- State Key Laboratory of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Huilin Xu
- State Key Laboratory of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Yuhan Zhao
- State Key Laboratory of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, 100871, China
| | - Xiaofeng Zheng
- State Key Laboratory of Protein and Plant Gene Research, Department of Biochemistry and Molecular Biology, School of Life Sciences, Peking University, Beijing, 100871, China.
| |
Collapse
|
13
|
Shao H, Kaplan HJ, Sun D. The Role of Adenosine in γδ T-Cell Regulation of Th17 Responses in Experimental Autoimmune Uveitis. Biomolecules 2023; 13:1432. [PMID: 37892114 PMCID: PMC10604616 DOI: 10.3390/biom13101432] [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: 07/25/2023] [Revised: 09/06/2023] [Accepted: 09/20/2023] [Indexed: 10/29/2023] Open
Abstract
Autoimmune diseases caused by T cells can arise from either T-helper 1 (Th1) or T-helper 17 (Th17)-type pathogenic T cells. However, it is unclear whether these two T-cell subsets are influenced by distinct pathogenic factors and whether treatments that are effective for Th1 responses also work for Th17 responses. To compare these two pathogenic responses, we conducted a systematic analysis in a mouse model of experimental autoimmune uveitis (EAU) to identify the factors that promote or inhibit each response and to determine their responses to various treatments. Our study found that the two types of pathogenic responses differ significantly in their pathological progressions and susceptibility to treatments. Specifically, we observed that extracellular adenosine is a crucial pathogenic molecule involved in the pathogenicity of inflammation and T-cell reactivity and that reciprocal interaction between adenosine and gamma delta (γδ) T cells plays a significant role in amplifying Th17 responses in the development of autoimmune diseases. The potential effect of targeting adenosine or adenosine receptors is analyzed regarding whether such targeting constitutes an effective approach to modulating both γδ T-cell responses and the pathogenic Th17 responses in autoimmune diseases.
Collapse
Affiliation(s)
- Hui Shao
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY 40202, USA
| | - Henry J. Kaplan
- Department of Ophthalmology and Biochemistry & Molecular Biology, St. Louis University School of Medicine, Saint Louis, MO 63104, USA
| | - Deming Sun
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90033, USA
| |
Collapse
|
14
|
Xu Y, Wang Y, Ji X. Immune and inflammatory mechanism of remote ischemic conditioning: A narrative review. Brain Circ 2023; 9:77-87. [PMID: 37576576 PMCID: PMC10419737 DOI: 10.4103/bc.bc_57_22] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 02/06/2023] [Accepted: 02/17/2023] [Indexed: 08/15/2023] Open
Abstract
The benefits of remote ischemic conditioning (RIC) on multiple organs have been extensively investigated. According to existing research, suppressing the immune inflammatory response is an essential mechanism of RIC. Based on the extensive effects of RIC on cardiovascular and cerebrovascular diseases, this article reviews the immune and inflammatory mechanisms of RIC and summarizes the effects of RIC on immunity and inflammation from three perspectives: (1) the mechanisms of the impact of RIC on inflammation and immunity; (2) evidence of the effects of RIC on immune and inflammatory processes in ischaemic stroke; and (3) possible future applications of this effect, especially in systemic infectious diseases such as sepsis and sepsis-associated encephalopathy. This review explores the possibility of using RIC as a treatment in more inflammation-related diseases, which will provide new ideas for the treatment of this kind of disease.
Collapse
Affiliation(s)
- Yi Xu
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- China-America Institute of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yuan Wang
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Xunming Ji
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- China-America Institute of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
15
|
Villa M, Sanin DE, Apostolova P, Corrado M, Kabat AM, Cristinzio C, Regina A, Carrizo GE, Rana N, Stanczak MA, Baixauli F, Grzes KM, Cupovic J, Solagna F, Hackl A, Globig AM, Hässler F, Puleston DJ, Kelly B, Cabezas-Wallscheid N, Hasselblatt P, Bengsch B, Zeiser R, Sagar, Buescher JM, Pearce EJ, Pearce EL. Prostaglandin E 2 controls the metabolic adaptation of T cells to the intestinal microenvironment. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.13.532431. [PMID: 36993703 PMCID: PMC10054978 DOI: 10.1101/2023.03.13.532431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Immune cells must adapt to different environments during the course of an immune response. We studied the adaptation of CD8 + T cells to the intestinal microenvironment and how this process shapes their residency in the gut. CD8 + T cells progressively remodel their transcriptome and surface phenotype as they acquire gut residency, and downregulate expression of mitochondrial genes. Human and mouse gut-resident CD8 + T cells have reduced mitochondrial mass, but maintain a viable energy balance to sustain their function. We found that the intestinal microenvironment is rich in prostaglandin E 2 (PGE 2 ), which drives mitochondrial depolarization in CD8 + T cells. Consequently, these cells engage autophagy to clear depolarized mitochondria, and enhance glutathione synthesis to scavenge reactive oxygen species (ROS) that result from mitochondrial depolarization. Impairing PGE 2 sensing promotes CD8 + T cell accumulation in the gut, while tampering with autophagy and glutathione negatively impacts the T cell population. Thus, a PGE 2 -autophagy-glutathione axis defines the metabolic adaptation of CD8 + T cells to the intestinal microenvironment, to ultimately influence the T cell pool.
Collapse
|
16
|
Skelly PJ, Nation CS, Da'Dara AA. Schistosoma mansoni and the purinergic halo. Trends Parasitol 2022; 38:1080-1088. [PMID: 36182536 PMCID: PMC9669209 DOI: 10.1016/j.pt.2022.09.001] [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: 07/13/2022] [Revised: 08/25/2022] [Accepted: 09/02/2022] [Indexed: 01/13/2023]
Abstract
Intravascular schistosomes may control immune and hemostatic responses by regulating the nature and amount of selected host purinergic signaling molecules - such as adenosine triphosphate (ATP), adenosine diphosphate (ADP), and nicotinamide adenine dinucleotide (NAD) - surrounding them. Such metabolites are collectively known as the worm's 'purinergic halo'. Host-interactive, membrane-bound, tegumental ectonucleotidases, notably SmATPDase1, SmNPP5, SmAP and SmNACE, can degrade proinflammatory, prothrombotic and immunomodulatory purinergic metabolites like those listed. A common catabolic product is the anti-inflammatory metabolite adenosine that can additionally be taken in by the worms as food. We envision the tegumental ectonucleotidases as having a twofold role at the worm surface: first, they degrade potentially harmful host signaling molecules, and second, they generate vital nutrients around the worms from where these can be conveniently imported.
Collapse
Affiliation(s)
- Patrick J Skelly
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA.
| | - Catherine S Nation
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| | - Akram A Da'Dara
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536, USA
| |
Collapse
|
17
|
Trelford CB, Dagnino L, Di Guglielmo GM. Transforming growth factor-β in tumour development. Front Mol Biosci 2022; 9:991612. [PMID: 36267157 PMCID: PMC9577372 DOI: 10.3389/fmolb.2022.991612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Accepted: 09/15/2022] [Indexed: 11/14/2022] Open
Abstract
Transforming growth factor-β (TGFβ) is a ubiquitous cytokine essential for embryonic development and postnatal tissue homeostasis. TGFβ signalling regulates several biological processes including cell growth, proliferation, apoptosis, immune function, and tissue repair following injury. Aberrant TGFβ signalling has been implicated in tumour progression and metastasis. Tumour cells, in conjunction with their microenvironment, may augment tumourigenesis using TGFβ to induce epithelial-mesenchymal transition, angiogenesis, lymphangiogenesis, immune suppression, and autophagy. Therapies that target TGFβ synthesis, TGFβ-TGFβ receptor complexes or TGFβ receptor kinase activity have proven successful in tissue culture and in animal models, yet, due to limited understanding of TGFβ biology, the outcomes of clinical trials are poor. Here, we review TGFβ signalling pathways, the biology of TGFβ during tumourigenesis, and how protein quality control pathways contribute to the tumour-promoting outcomes of TGFβ signalling.
Collapse
Affiliation(s)
- Charles B. Trelford
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| | - Lina Dagnino
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
- Department of Oncology, Children’s Health Research Institute and Lawson Health Research Institute, London, ON, Canada
| | - Gianni M. Di Guglielmo
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, Western University, London, ON, Canada
| |
Collapse
|
18
|
Ahmadi P, Yan M, Bauche A, Smeets R, Müller CE, Koch-Nolte F, Haag F, Fliegert R, Kluwe L, Wiesch JSZ, Hartjen P. Human dental pulp cells modulate CD8+ T cell proliferation and efficiently degrade extracellular ATP to adenosine in vitro. Cell Immunol 2022; 380:104589. [DOI: 10.1016/j.cellimm.2022.104589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 07/27/2022] [Accepted: 08/10/2022] [Indexed: 11/03/2022]
|
19
|
Xu S, Gu R, Bian X, Xu X, Xia X, Liu Y, Jia C, Gu Y, Zhang H. Remote Conditioning by Rhythmic Compression of Limbs Ameliorated Myocardial Infarction by Downregulation of Inflammation via A2 Adenosine Receptors. Front Cardiovasc Med 2022; 8:723332. [PMID: 35498376 PMCID: PMC9040771 DOI: 10.3389/fcvm.2021.723332] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 12/23/2021] [Indexed: 01/07/2023] Open
Abstract
Background Remote ischemic conditioning (RIC) is a cardioprotective phenomenon, yet transient ischemia is not a requisite trigger for remote cardioprotection. In fact, RIC is a stimulus compound containing interruption of the blood vessel and tissue compression. In this study, we evaluate the effects of remote tissue compression on infarct size after myocardial infarction and explore its preliminary mechanisms. Methods and Results We used a murine model of myocardial infarction to assess ischemia injury and identified remote conditioning by rhythmic compression on forelimb as a novel cardioprotective intervention. We show that the cardioprotective signal transduction of remote conditioning from the trigger limb to the heart involves the release of adenosine. Our results demonstrate that A2a and A2b receptors are indispensable parts for cardioprotection of remote conditioning, which is linked to its anti-inflammatory properties by the subsequent activation of cAMP/PKA/NF-κB axis. Conclusion Our results establish a new connection between remote tissue compression and cardiovascular diseases, which enhances our cognition about the role of tissue compression on RIC cardioprotection.
Collapse
Affiliation(s)
- Senlei Xu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
- Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, China
| | - Renjun Gu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiangyu Bian
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xin Xu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xuefeng Xia
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yuchen Liu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chengjie Jia
- Wuxi Municipal Rehabilitation Hospital, Wuxi, China
| | - Yihuang Gu
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Yihuang Gu
| | - Hongru Zhang
- School of Acupuncture and Tuina, School of Regimen and Rehabilitation, Nanjing University of Chinese Medicine, Nanjing, China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
- Hongru Zhang
| |
Collapse
|
20
|
Halpin-Veszeleiova K, Hatfield SM. Therapeutic Targeting of Hypoxia-A2-Adenosinergic Pathway in COVID-19 Patients. Physiology (Bethesda) 2022; 37:46-52. [PMID: 34486395 PMCID: PMC8742736 DOI: 10.1152/physiol.00010.2021] [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] [Indexed: 01/08/2023] Open
Abstract
The hypoxia-hypoxia-inducible factor (HIF)-1α-A2-adenosinergic pathway protects tissues from inflammatory damage during antipathogen immune responses. The elimination of this physiological tissue-protecting mechanism by supplemental oxygenation may contribute to the high mortality of oxygen-ventilated COVID-19 patients by exacerbating inflammatory lung damage. Restoration of this pathway with hypoxia-adenosinergic drugs may improve outcomes in these patients.
Collapse
Affiliation(s)
- Katarina Halpin-Veszeleiova
- New England Inflammation and Tissue Protection Institute, Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts
| | - Stephen M Hatfield
- New England Inflammation and Tissue Protection Institute, Department of Pharmaceutical Sciences, Northeastern University, Boston, Massachusetts
| |
Collapse
|
21
|
Abstract
Schistosomes are long lived, intravascular parasitic platyhelminths that infect >200 million people globally. The molecular mechanisms used by these blood flukes to dampen host immune responses are described in this review. Adult worms express a collection of host-interactive tegumental ectoenzymes that can cleave host signaling molecules such as the "alarmin" ATP (cleaved by SmATPDase1), the platelet activator ADP (SmATPDase1, SmNPP5), and can convert AMP into the anti-inflammatory mediator adenosine (SmAP). SmAP can additionally cleave the lipid immunomodulator sphingosine-1-phosphate and the proinflammatory anionic polymer, polyP. In addition, the worms release a barrage of proteins (e.g., SmCB1, SjHSP70, cyclophilin A) that can impinge on immune cell function. Parasite eggs also release their own immunoregulatory proteins (e.g., IPSE/α1, omega1, SmCKBP) as do invasive cercariae (e.g., Sm16, Sj16). Some schistosome glycans (e.g., LNFPIII, LNnT) and lipids (e.g., Lyso-PS, LPC), produced by several life stages, likewise affect immune cell responses. The parasites not only produce eicosanoids (e.g., PGE2, PGD2-that can be anti-inflammatory) but can also induce host cells to release these metabolites. Finally, the worms release extracellular vesicles (EVs) containing microRNAs, and these too have been shown to skew host cell metabolism. Thus, schistosomes employ an array of biomolecules-protein, lipid, glycan, nucleic acid, and more, to bend host biochemistry to their liking. Many of the listed molecules have been individually shown capable of inducing aspects of the polarized Th2 response seen following infection (with the generation of regulatory T cells (Tregs), regulatory B cells (Bregs) and anti-inflammatory, alternatively activated (M2) macrophages). Precisely how host cells integrate the impact of these myriad parasite products following natural infection is not known. Several of the schistosome immunomodulators described here are in development as novel therapeutics against autoimmune, inflammatory, and other, nonparasitic, diseases.
Collapse
Affiliation(s)
- Sreemoyee Acharya
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Akram A. Da’dara
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
| | - Patrick J. Skelly
- Molecular Helminthology Laboratory, Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
22
|
Sun D, Ko M, Shao H, Kaplan HJ. Adenosine receptor ligation tips the uveitogenic Th1 and Th17 balance towards the latter in experimental autoimmune uveitis-induced mouse. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:93-103. [PMID: 34825178 PMCID: PMC8612466 DOI: 10.1016/j.crimmu.2021.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Various pathological conditions are accompanied by release of adenosine triphosphate (ATP) from the intracellular to the extracellular compartment, where it degrades into adenosine and modulates immune responses. Previous studies concluded that both ATP and its degradation product adenosine are important immune-regulatory molecules; ATP acted as a danger signal that promotes immune responses, but adenosine's effect was inhibitory. We show that adenosine receptor ligation plays an important role in balancing Th1 and Th17 pathogenic T cell responses in experimental autoimmune uveitis (EAU). While its effect on Th1 responses is inhibitory, its effect on Th17 responses is enhancing, thereby impacting the balance between Th1 and Th17 responses. Mechanistic studies showed that this effect is mediated via several immune cells, among which γδ T cell activation and dendritic cell differentiation are prominent; adenosine- and γδ-mediated immunoregulation synergistically impact each other's effect. Adenosine receptor ligation augments the activation of γδ T cells, which is an important promoter for Th17 responses and has a strong effect on dendritic cell (DC) differentiation, tipping the balance from generation of DCs that stimulate Th1 responses to those that stimulate Th17 responses. The knowledge acquired in this study should improve our understanding of the immune-regulatory effect of extracellular ATP-adenosine metabolism and improve treatment for autoimmune diseases caused by both Th1-and Th17-type pathogenic T cells.
Collapse
Affiliation(s)
- Deming Sun
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90033, United States
- Corresponding author. Department of Ophthalmology, University of California Los Angeles, Los Angeles, CA90033, USA.
| | - Minhee Ko
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90033, United States
| | - Hui Shao
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY, 40202, United States
| | - Henry J. Kaplan
- Saint Louis University (SLU) Eye Institute, SLU School of Medicine, Saint Louis, MO, 63104, United States
| |
Collapse
|
23
|
Antioxidant and Antiinflammatory Effects of Epilobium parviflorum, Melilotus officinalis and Cardiospermum halicacabum Plant Extracts in Macrophage and Microglial Cells. Cells 2021; 10:cells10102691. [PMID: 34685671 PMCID: PMC8534520 DOI: 10.3390/cells10102691] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 09/30/2021] [Accepted: 10/05/2021] [Indexed: 12/17/2022] Open
Abstract
Background: We investigated the phenolic content characterizing different plant extracts from Epilobium parviflorum, Cardiospermum halicacabum, and Melilotus officinalis, their antioxidant, antiinflammatory effects, and their mechanism of action. Methods: plant samples were macerated in 40% ethanol or hot/ cold glycerate and assessed for polyphenols content. The antioxidant activity was investigated by DPPH radical scavenging assay and H2DCFDA test in LPS-stimulated RAW264.7 macrophages and N9 microglial cells. MTS experiments and antiinflammatory properties verified cellular toxicity through NO assay. Interaction with A2A adenosine receptors was evaluated through binding assays using [3H]ZM241385 radioligand. Results: Polyphenols were present in 40% ethanol plant extract, which at 0.1–10 µg/µL achieved good antioxidant effects, with a DPPH radical scavenging rate of about 90%. In LPS-stimulated cells, these plant extracts, at 1μg/μL, did not affect cell vitality, displayed significant inhibition of H2DCFDA and NO production, and inhibited ZM 241385 binding in CHO cells transfected with A2A receptors. RAW 264.7 and N9 cells presented a density of them quantified in 60 ± 9 and 45 ± 5 fmol/mg of protein, respectively. Conclusion: Epilobium parviflorum, Cardiospermum halicacabum, and Melilotus officinalis extracts may be considered a source of agents for treating disorders related to oxidative stress and inflammation.
Collapse
|
24
|
78495111110.1152/physrev.00046.2020" />
Abstract
This medical review addresses the hypothesis that CD38/NADase is at the center of a functional axis (i.e., intracellular Ca2+ mobilization/IFNγ response/reactive oxygen species burst) driven by severe acute respiratory syndrome coronavirus 2 infection, as already verified in respiratory syncytial virus pathology and CD38 activity in other cellular settings. Key features of the hypothesis are that 1) the substrates of CD38 (e.g., NAD+ and NADP+) are depleted by viral-induced metabolic changes; 2) the products of the enzymatic activity of CD38 [e.g., cyclic adenosine diphosphate-ribose (ADPR)/ADPR/nicotinic acid adenine dinucleotide phosphate] and related enzymes [e.g., poly(ADP-ribose)polymerase, Sirtuins, and ADP-ribosyl hydrolase] are involved in the anti‐viral and proinflammatory response that favors the onset of lung immunopathology (e.g., cytokine storm and organ fibrosis); and 3) the pathological changes induced by this kinetic mechanism may be reduced by distinct modulators of the CD38/NAD+ axis (e.g., CD38 blockers, NAD+ suppliers, among others). This view is supported by arrays of associative basic and applied research data that are herein discussed and integrated with conclusions reported by others in the field of inflammatory, immune, tumor, and viral diseases.
Collapse
Affiliation(s)
- Alberto L. Horenstein
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Angelo C. Faini
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Fabio Malavasi
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| |
Collapse
|
25
|
Vautrot V, Bentayeb H, Causse S, Garrido C, Gobbo J. Tumor-Derived Exosomes: Hidden Players in PD-1/PD-L1 Resistance. Cancers (Basel) 2021; 13:cancers13184537. [PMID: 34572764 PMCID: PMC8467727 DOI: 10.3390/cancers13184537] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary Immunotherapies such as anti-PD-1/PD-L1 have garnered increasing importance in cancer therapy, leading to substantial improvements in patient care and survival. However, a certain proportion of patients present tumors that resist these treatments. Exosomes, small vesicles secreted by almost every cell, including tumor cells, have proven to be key actors in this resistance. In this review, we describe the involvement of immune checkpoints and immune modulators in tumor-derived exosomes (TEXs) in the context of cancer. We will focus on the most promising proteins under scrutiny for use in combination with PD-1 blockade therapy in a clinical setting: PD-L1, CTLA-4, TIM-3, CD73/39, LAG-3, and TIGIT. Finally, we will discuss how they can change the game in immunotherapy, notably through their role in immunoresistance and how they can guide therapeutic decisions, as well as the current obstacles in the field. Abstract Recently, immunotherapy has garnered increasing importance in cancer therapy, leading to substantial improvements in patient care and survival. By blocking the immune checkpoints—protein regulators of the immune system—immunotherapy prevents immune tolerance toward tumors and reactivates the immune system, prompting it to fight cancer cell growth and diffusion. A widespread strategy for this is the blockade of the interaction between PD-L1 and PD-1. However, while patients generally respond well to immunotherapy, a certain proportion of patients present tumors that resist these treatments. This portion can be very high in some cancers and hinders cancer curability. For this reason, current efforts are focusing on combining PD-1/PD-L1 immunotherapy with the targeting of other immune checkpoints to counter resistance and achieve better results. Exosomes, small vesicles secreted by almost any cell, including tumor cells, have proven to be key actors in this resistance. The exosomes released by tumor cells spread the immune-suppressive properties of the tumor throughout the tumor microenvironment and participate in establishing metastatic niches. In this review, we will describe immune checkpoints and immune modulators whose presence in tumor-derived exosomes (TEXs) has been established. We will focus on the most promising proteins under scrutiny for use in combination with PD-1 blockade therapy in a clinical setting, such as PD-L1, CTLA-4, TIM-3, CD73/39, LAG-3, and TIGIT. We will explore the immunosuppressive impact of these exosomal proteins on a variety of immune cells. Finally, we will discuss how they can change the game in immunotherapy and guide therapeutic decisions, as well as the current limits of this approach. Depending on the viewpoint, these exosomal proteins may either provide key missing information on tumor growth and resistance mechanisms or they may be the next big challenge to overcome in improving cancer treatment.
Collapse
Affiliation(s)
- Valentin Vautrot
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
- Centre Georges-François Leclerc, F-21079 Dijon, France
| | - Hafidha Bentayeb
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
- Centre Georges-François Leclerc, F-21079 Dijon, France
| | - Sébastien Causse
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
| | - Carmen Garrido
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
- Centre Georges-François Leclerc, F-21079 Dijon, France
| | - Jessica Gobbo
- Research Center UMR 1231, Label Ligue Nationale Contre le Cancer and LipSTIC, INSERM, F-21000 Dijon, France; (V.V.); (H.B.); (S.C.); (C.G.)
- Unité de Formation et de Recherches Sciences de la Santé, University of Bourgogne Franche-Comté, F-21000 Dijon, France
- Centre Georges-François Leclerc, F-21079 Dijon, France
- Centre Georges-François Leclerc, Early Phase Unit INCa CLIP², Department of Oncology, F-21079 Dijon, France
- Clinical Investigation Center CIC1432, Module Plurithématique, INSERM, F-21079 Dijon, France
- Correspondence:
| |
Collapse
|
26
|
Audrito V, Messana VG, Brandimarte L, Deaglio S. The Extracellular NADome Modulates Immune Responses. Front Immunol 2021; 12:704779. [PMID: 34421911 PMCID: PMC8371318 DOI: 10.3389/fimmu.2021.704779] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/21/2021] [Indexed: 12/30/2022] Open
Abstract
The term NADome refers to the intricate network of intracellular and extracellular enzymes that regulate the synthesis or degradation of nicotinamide adenine dinucleotide (NAD) and to the receptors that engage it. Traditionally, NAD was linked to intracellular energy production through shuffling electrons between oxidized and reduced forms. However, recent data indicate that NAD, along with its biosynthetic and degrading enzymes, has a life outside of cells, possibly linked to immuno-modulating non-enzymatic activities. Extracellular NAD can engage puriginergic receptors triggering an inflammatory response, similar - to a certain extent - to what described for adenosine triphosphate (ATP). Likewise, NAD biosynthetic and degrading enzymes have been amply reported in the extracellular space, where they possess both enzymatic and non-enzymatic functions. Modulation of these enzymes has been described in several acute and chronic conditions, including obesity, cancer, inflammatory bowel diseases and sepsis. In this review, the role of the extracellular NADome will be discussed, focusing on its proposed role in immunomodulation, together with the different strategies for its targeting and their potential therapeutic impact.
Collapse
Affiliation(s)
- Valentina Audrito
- Laboratory of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Vincenzo Gianluca Messana
- Laboratory of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Lorenzo Brandimarte
- Laboratory of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| | - Silvia Deaglio
- Laboratory of Cancer Immunogenetics, Department of Medical Sciences, University of Turin, Turin, Italy
| |
Collapse
|
27
|
Sun D, Ko MK, Shao H, Kaplan HJ. Augmented Th17-stimulating activity of BMDCs as a result of reciprocal interaction between γδ and dendritic cells. Mol Immunol 2021; 134:13-24. [PMID: 33689926 PMCID: PMC8629029 DOI: 10.1016/j.molimm.2021.02.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/08/2021] [Accepted: 02/23/2021] [Indexed: 12/17/2022]
Abstract
Our previous studies demonstrated that γδ T cells have a strong regulatory effect on Th17 autoimmune responses in experimental autoimmune uveitis (EAU). In the current study, we show that reciprocal interactions between mouse γδ T cells and dendritic cells (DCs) played a major role in γδ regulation of Th17 responses. Mouse bone marrow-derived dendritic cells (BMDCs) acquired an increased ability to enhance Th17 autoimmune responses after exposure to γδ T cells; meanwhile, after exposure, a significant portion of the BMDCs expressed CD73 - a molecule that is fundamental in the conversion of immunostimulatory ATP into immunosuppressive adenosine. Functional studies showed that CD73+ BMDCs were uniquely effective in stimulating the Th17 responses, as compared to CD73- BMDCs; and activated γδ T cells are much more effective than non-activated γδ T cells at inducing CD73+ BMDCs. As a result, activated γδ T cells acquired greater Th17-enhancing activity. Treatment of BMDCs with the CD73-specific antagonist APCP abolished the enhancing effect of the BMDCs. γδ T cells more effectively induced CD73+ BMDCs from the BMDCs that were pre-exposed to TLR ligands, and the response was further augmented by adenosine. Moreover, BMDCs acquired increased ability to stimulate γδ activation after pre-exposure to TLR ligands and adenosine. Our results demonstrated that both extra-cellular adenosine and TLR ligands are critical factors in augmented Th17 responses in this autoimmune disease, and the reciprocal interactions between γδ T cells and DCs play a major role in promoting Th17 responses.
Collapse
Affiliation(s)
- Deming Sun
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90033, United States.
| | - Minhee K Ko
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90033, United States
| | - Hui Shao
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, KY, 40202, United States
| | - Henry J Kaplan
- Saint Louis University (SLU) Eye Institute, SLU School of Medicine, Saint Louis, MO, 63104, United States
| |
Collapse
|
28
|
Jiang Q, Yang G, Liu Q, Wang S, Cui D. Function and Role of Regulatory T Cells in Rheumatoid Arthritis. Front Immunol 2021; 12:626193. [PMID: 33868244 PMCID: PMC8047316 DOI: 10.3389/fimmu.2021.626193] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid arthritis (RA) is a systemic and heterogeneous autoimmune disease with symmetrical polyarthritis as its critical clinical manifestation. The basic cause of autoimmune diseases is the loss of tolerance to self or harmless antigens. The loss or functional deficiency of key immune cells, regulatory T (Treg) cells, has been confirmed in human autoimmune diseases. The pathogenesis of RA is complex, and the dysfunction of Tregs is one of the proposed mechanisms underlying the breakdown of self-tolerance leading to the progression of RA. Treg cells are a vital component of peripheral immune tolerance, and the transcription factor Foxp3 plays a major immunosuppressive role. Clinical treatment for RA mainly utilizes drugs to alleviate the progression of disease and relieve disease activity, and the ideal treatment strategy should be to re-induce self-tolerance before obvious tissue injury. Treg cells are one of the ideal options. This review will introduce the classification, mechanism of action, and characteristics of Treg cells in RA, which provides insights into clinical RA treatment.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Guocan Yang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Qi Liu
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Shengjun Wang
- Department of Laboratory Medicine, The Affiliated People's Hospital, Jiangsu University, Zhenjiang, China.,Department of Immunology, Jiangsu Key Laboratory of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
29
|
Abstract
This medical review addresses the hypothesis that CD38/NADase is at the center of a functional axis (i.e., intracellular Ca2+ mobilization/IFNγ response/reactive oxygen species burst) driven by severe acute respiratory syndrome coronavirus 2 infection, as already verified in respiratory syncytial virus pathology and CD38 activity in other cellular settings. Key features of the hypothesis are that 1) the substrates of CD38 (e.g., NAD+ and NADP+) are depleted by viral-induced metabolic changes; 2) the products of the enzymatic activity of CD38 [e.g., cyclic adenosine diphosphate-ribose (ADPR)/ADPR/nicotinic acid adenine dinucleotide phosphate] and related enzymes [e.g., poly(ADP-ribose)polymerase, Sirtuins, and ADP-ribosyl hydrolase] are involved in the anti‐viral and proinflammatory response that favors the onset of lung immunopathology (e.g., cytokine storm and organ fibrosis); and 3) the pathological changes induced by this kinetic mechanism may be reduced by distinct modulators of the CD38/NAD+ axis (e.g., CD38 blockers, NAD+ suppliers, among others). This view is supported by arrays of associative basic and applied research data that are herein discussed and integrated with conclusions reported by others in the field of inflammatory, immune, tumor, and viral diseases.
Collapse
Affiliation(s)
- Alberto L Horenstein
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Angelo C Faini
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| | - Fabio Malavasi
- Department of Medical Science, University of Turin, Turin, Italy; and Centro Ricerca Medicina, Sperimentale (CeRMS) and Fondazione Ricerca Molinette Onlus, Turin, Italy
| |
Collapse
|
30
|
Lee JS, Chowdhury N, Roberts JS, Yilmaz Ö. Host surface ectonucleotidase-CD73 and the opportunistic pathogen, Porphyromonas gingivalis, cross-modulation underlies a new homeostatic mechanism for chronic bacterial survival in human epithelial cells. Virulence 2021; 11:414-429. [PMID: 32419582 PMCID: PMC7239027 DOI: 10.1080/21505594.2020.1763061] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Cell surface nucleotide-metabolizing enzyme, ectonucleotidase-CD73, has emerged as a central component of the cellular homeostatic-machinery that counterbalances the danger-molecule (extracellular-ATP)-driven proinflammatory response in immune cells. While the importance of CD73 in microbial host fitness and symbiosis is gradually being unraveled, there remains a significant gap in knowledge of CD73 and its putative role in epithelial cells. Here, we depict a novel host-pathogen adaptation mechanism where CD73 takes a center role in the intracellular persistence of Porphyromonas gingivalis, a major colonizer of oral mucosa, using human primary gingival epithelial cell (GEC) system. Temporal analyses revealed, upon invasion into the GECs, P. gingivalis can significantly elevate the host-surface CD73 activity and expression. The enhanced and active CD73 significantly increases P. gingivalis intracellular growth in the presence of substrate-AMP and simultaneously acts as a negative regulator of reactive oxygen species (ROS) generation upon eATP treatment. The inhibition of CD73 by siRNA or by a specific inhibitor markedly increases ROS production. Moreover, CD73 and P. gingivalis cross-signaling significantly modulates pro-inflammatory interleukin-6 (IL-6) in the GECs. Conversely, exogenous treatment of the infected GECs with IL-6 suppresses the intracellular bacteria via amplified ROS generation. However, the decreased bacterial levels can be restored by overexpressing functionally active CD73. Together, these findings illuminate how the local extracellular-purine-metabolism, in which CD73 serves as a core molecular switch, can alter intracellular microbial colonization resistance. Further, host-adaptive pathogens such as P. gingivalis can target host ectonucleotidases to disarm specific innate defenses for successful intracellular persistence in mucosal epithelia.
Collapse
Affiliation(s)
- Jaden S Lee
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Nityananda Chowdhury
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - JoAnn S Roberts
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Özlem Yilmaz
- Department of Oral Health Sciences, Medical University of South Carolina, Charleston, SC, USA.,Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
31
|
Mandapathil M, Szczepanski M, Harasymczuk M, Ren J, Cheng D, Jackson EK, Gorelik E, Johnson J, Lang S, Whiteside TL. CD26 expression and adenosine deaminase activity in regulatory T cells (Treg) and CD4(+) T effector cells in patients with head and neck squamous cell carcinoma. Oncoimmunology 2021; 1:659-669. [PMID: 22934258 PMCID: PMC3429570 DOI: 10.4161/onci.20387] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Adenosine deaminase (ADA) is responsible for the deamination of immunosuppressive adenosine to inosine. In human T lymphocytes, ADA is associated with dipeptidyl peptidase IV (CD26). ADA expression and activity were evaluated in regulatory T cells (Treg) and CD4(+) T effector cells (Teff) of patients with head and neck squamous cell cancer (HNSCC). CD4(+)CD39(+) and CD4(+)CD39(neg) T cells were isolated by single-cell sorting from the peripheral blood of 15 HNSCC patients and 15 healthy donors (NC). CD26/ADA expression in these cells was studied by multicolor flow cytometry, confocal microscopy, RT-PCR and immunohistochemistry in tumor tissues. ADA activity was evaluated by mass spectrometry, suppression of Teff proliferation in CFSE assays and cytokine production by Luminex. CD4(+)CD39(+) Treg had low and CD4(+)CD39(neg) Teff high CD26/ADA expression and ADA activity in NC or HNSCC. The frequency and suppressor activity of CD39(+)CD26(neg) Treg were elevated in patients relative to NC (p < 0.01). However, ADA activity in patients' CD4(+)CD39(neg) Teff was decreased (p < 0.05), resulting in extracellular adenosine accumulation. Also, patients' Teff were more sensitive to inhibitory signals delivered via adenosine receptors. IL-2, IL12 and INFγ upregulated ADA expression and activity in CD4(+)CD39(neg) Teff, whereas IL-10, PGE(2) and CADO downregulated it. The differentially expressed CD26/ADA can serve as surface markers for functionally-active CD39(+)CD26(neg) Treg.
Collapse
Affiliation(s)
- Magis Mandapathil
- University of Pittsburgh Cancer Institute; Pittsburgh, PA USA ; Department of Otorhinolaryngology; University of Duisburg-Essen; Essen, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Koh S, Dupuis N, Auvin S. Ketogenic diet and Neuroinflammation. Epilepsy Res 2020; 167:106454. [DOI: 10.1016/j.eplepsyres.2020.106454] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/26/2020] [Accepted: 09/01/2020] [Indexed: 12/13/2022]
|
33
|
Eisenstein A, Chitalia SV, Ravid K. Bone Marrow and Adipose Tissue Adenosine Receptors Effect on Osteogenesis and Adipogenesis. Int J Mol Sci 2020; 21:E7470. [PMID: 33050467 PMCID: PMC7589187 DOI: 10.3390/ijms21207470] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 10/01/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022] Open
Abstract
Adenosine is an extracellular signaling molecule that is particularly relevant in times of cellular stress, inflammation and metabolic disturbances when the levels of the purine increase. Adenosine acts on two G-protein-coupled stimulatory and on two G-protein-coupled inhibitory receptors, which have varying expression profiles in different tissues and conditions, and have different affinities for the endogenous ligand. Studies point to significant roles of adenosine and its receptors in metabolic disease and bone health, implicating the receptors as potential therapeutic targets. This review will highlight our current understanding of the dichotomous effects of adenosine and its receptors on adipogenesis versus osteogenesis within the bone marrow to maintain bone health, as well as its relationship to obesity. Therapeutic implications will also be reviewed.
Collapse
Affiliation(s)
- Anna Eisenstein
- Department of Dermatology, Yale School of Medicine, New Haven, CT 06520, USA
| | - Shlok V. Chitalia
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA; (S.V.C.); (K.R.)
| | - Katya Ravid
- Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA; (S.V.C.); (K.R.)
| |
Collapse
|
34
|
Steingold JM, Hatfield SM. Targeting Hypoxia-A2A Adenosinergic Immunosuppression of Antitumor T Cells During Cancer Immunotherapy. Front Immunol 2020; 11:570041. [PMID: 33117358 PMCID: PMC7553081 DOI: 10.3389/fimmu.2020.570041] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 08/26/2020] [Indexed: 12/25/2022] Open
Abstract
The blockade of immunological negative regulators offered a novel therapeutic approach that revolutionized the immunotherapy of cancer. Still, a significant portion of patients fail to respond to anti-PD-1/PD-L1 and/or anti-CTLA-4 therapy or experience significant adverse effects. We propose that one of the major reasons that many patients do not respond to this form of therapy is due to the powerful physiological suppression mediated by hypoxia-adenosinergic signaling. Indeed, both inflamed and cancerous tissues are hypoxic and rich in extracellular adenosine, in part due to stabilization of the transcription factor hypoxia-inducible factor 1 alpha (HIF-1α). Adenosine signals through adenosine A2A receptors (A2AR) to suppress anti-tumor and anti-pathogen immune responses. Several classes of anti-hypoxia-A2AR therapeutics have been offered to refractory cancer patients, with A2AR blockers, inhibitors of adenosine-generating enzymes such as CD39 and CD73, and hypoxia-targeting drugs now reaching the clinical stage. Clinical results have confirmed preclinical observations that blockade of the hypoxia-adenosine-A2AR axis synergizes with inhibitors of immune checkpoints to induce tumor rejection. Thus, A2AR blockers provide a new hope for the majority of patients who are nonresponsive to current immunotherapeutic approaches including checkpoint blockade. Here, we discuss the discoveries that firmly implicate the A2AR as a critical and non-redundant biochemical negative regulator of the immune response and highlight the importance of targeting the hypoxia-adenosine-A2AR axis to manipulate anti-pathogen and anti-tumor immune responses.
Collapse
Affiliation(s)
- Joseph M Steingold
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouvé College of Health Sciences, Northeastern University, Boston, MA, United States
| | - Stephen M Hatfield
- Department of Pharmaceutical Sciences, New England Inflammation and Tissue Protection Institute, Bouvé College of Health Sciences, Northeastern University, Boston, MA, United States
| |
Collapse
|
35
|
Magni G, Ceruti S. Adenosine Signaling in Autoimmune Disorders. Pharmaceuticals (Basel) 2020; 13:ph13090260. [PMID: 32971792 PMCID: PMC7558305 DOI: 10.3390/ph13090260] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
The molecular components of the purinergic system (i.e., receptors, metabolizing enzymes and membrane transporters) are widely expressed in the cells of the immune system. Additionally, high concentrations of adenosine are generated from the hydrolysis of ATP in any "danger" condition, when oxygen and energy availability dramatically drops. Therefore, adenosine acts as a retaliatory metabolite to counteract the nucleotide-mediated boost of the immune reaction. Based on this observation, it can be foreseen that the recruitment with selective agonists of the receptors involved in the immunomodulatory effect of adenosine might represent an innovative anti-inflammatory approach with potential exploitation in autoimmune disorders. Quite surprisingly, pro-inflammatory activity exerted by some adenosine receptors has been also identified, thus paving the way for the hypothesis that at least some autoimmune disorders may be caused by a derailment of adenosine signaling. In this review article, we provide a general overview of the roles played by adenosine on immune cells with a specific focus on the development of adenosine-based therapies for autoimmune disorders, as demonstrated by the exciting data from concluded and ongoing clinical trials.
Collapse
|
36
|
Helms RS, Powell JD. Rethinking the adenosine-A 2AR checkpoint: implications for enhancing anti-tumor immunotherapy. Curr Opin Pharmacol 2020; 53:77-83. [PMID: 32781414 DOI: 10.1016/j.coph.2020.07.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022]
Abstract
Adenosine signaling through A2AR serves as a negative regulator of the immune system. Unique to this suppressive pathway is its ability to impact numerous stromal and immune cells. Additionally, tumors exhibit elevated concentrations of adenosine further advancing the pathway's potential as a powerful target for activating anti-tumor immunity. The promise of this therapeutic strategy has been repeatedly demonstrated in mice, but has so far only yielded limited success in the clinic. Nonetheless, it is notable that many of these observed clinical responses have been in individuals resistant to prior immunotherapy. These observations suggest this pathway is indeed involved in tumor immune evasion. Thus, identifying the disparities between the translational and clinical implementation of this therapy becomes necessary. To this end, this review will revisit how and where adenosine-A2AR signaling regulates the immune system and anti-tumor immunity so as to reveal opportunities for improving the translational success of this immunotherapy.
Collapse
Affiliation(s)
- Rachel S Helms
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, 1650 Orleans Street, CRB-I Rm443, Baltimore, MD, 21231, USA
| | - Jonathan D Powell
- The Bloomberg-Kimmel Institute for Cancer Immunotherapy at Johns Hopkins, 1650 Orleans Street, CRB-I Rm443, Baltimore, MD, 21231, USA.
| |
Collapse
|
37
|
Blockade of Adenosine A 2A Receptor Protects Photoreceptors after Retinal Detachment by Inhibiting Inflammation and Oxidative Stress. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7649080. [PMID: 32714489 PMCID: PMC7354651 DOI: 10.1155/2020/7649080] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 05/19/2020] [Accepted: 06/03/2020] [Indexed: 12/20/2022]
Abstract
Purpose Adenosine A2A receptor (A2AR) signaling is neuroprotective in some retinal damage models, but its role in neuronal survival during retinal detachment (RD) is unclear. We tested the hypothesis that A2AR antagonist ZM241385 would prevent photoreceptor apoptosis by inhibiting retinal inflammation and oxidative stress after RD. Methods The A2AR antagonist ZM241385 was delivered daily to C57BL/6J mice for three days at a dose (3 mg/kg, i.p.) starting 2 hours prior to creating RD. A2AR expression, microglia proliferation and reactivity, glial fibrillary acidic protein (GFAP) accumulation, IL-1β expression, and reactive oxygen species (ROS) production were evaluated with immunofluorescence. Photoreceptor TUNEL was analyzed. Results A2AR expression obviously increased and accumulated in microglia and Müller cells in the retinas after RD. The A2AR antagonist ZM241385 effectively inhibited retinal microglia proliferation and reactivity, decreased GFAP upregulation and proinflammatory cytokine IL-1β expression of Müller cells, and suppressed ROS overproduction, resulting in attenuation of photoreceptor apoptosis after RD. Conclusions The A2AR antagonist ZM241385 is an effective suppressor of microglia proliferation and reactivity, gliosis, neuroinflammation, oxidative stress, and photoreceptor apoptosis in a mouse model of RD. This suggests that A2AR blockade may be an important therapeutic strategy to protect photoreceptors in RD and other CNS diseases that share a common etiology.
Collapse
|
38
|
Townsend MH, Tellez Freitas CM, Larsen D, Piccolo SR, Weber KS, Robison RA, O'Neill KL. Hypoxanthine Guanine Phosphoribosyltransferase expression is negatively correlated with immune activity through its regulation of purine synthesis. Immunobiology 2020; 225:151931. [PMID: 32291109 DOI: 10.1016/j.imbio.2020.151931] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 02/13/2020] [Accepted: 03/03/2020] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The purpose of this study was to examine the effects of elevated Hypoxanthine Guanine Phosphoribosyltransferase (HPRT) on the immune response in the tumor microenvironment. METHODOLOGY HPRT expression was evaluated in cancer patients and correlated with cytokine expression, survival, and immune cell infiltration. An HPRT knockdown cell line was created to evaluate HPRT impact on purine expression and subsequent purine treatment was administered to immune cells to determine their influence on cell activation. RESULTS HPRT expression was negatively correlated with the general expression of both pro-inflammatory and anti-inflammatory cytokines. Additionally, HPRT expression was also negatively correlated with the infiltration of immune cell subsets: B-cells, CD4 + T cells, macrophages, neutrophils, and dendritic cells (p < 0.001) and CD8 + T-cells (p < 0.01). When HPRT was knocked down in a Raji cell line, the levels of adenosine were reduced significantly compared to the wild type. When examining the level of Ca2+ influx of Raji compared to the HPRT Raji knockdown cell, there was a significant decrease in calcium influx in the knockdown cells when compared to the wild type cells. This demonstrates that HPRT had a significant impact on overall cell activation and the ability of the cells to properly influx calcium needed for their activation. CONCLUSIONS We conclude that purine levels significantly reduce immune cell activation in cancer and the upregulation of HPRT in malignant tissue is a contributing factors to the immunosuppressive microenvironment.
Collapse
Affiliation(s)
- Michelle H Townsend
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA.
| | - Claudia M Tellez Freitas
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA; College of Dental Medicine, Roseman University of Health Science, South Jordan, UT, USA
| | - Dallas Larsen
- Department of Biology, Brigham Young University, Provo, UT, USA
| | - Stephen R Piccolo
- Department of Biology, Brigham Young University, Provo, UT, USA; Department of Biomedical Informatics, University of Utah, Salt Lake City, UT, USA
| | - K Scott Weber
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Richard A Robison
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| | - Kim L O'Neill
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT, USA
| |
Collapse
|
39
|
Zhao R, Qiao J, Zhang X, Zhao Y, Meng X, Sun D, Peng X. Toll-Like Receptor-Mediated Activation of CD39 Internalization in BMDCs Leads to Extracellular ATP Accumulation and Facilitates P2X7 Receptor Activation. Front Immunol 2019; 10:2524. [PMID: 31736956 PMCID: PMC6834529 DOI: 10.3389/fimmu.2019.02524] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/10/2019] [Indexed: 12/19/2022] Open
Abstract
Toll-like receptors (TLRs) trigger innate immune responses through their recognition of conserved molecular ligands of either endogenous or microbial origin. Although activation, function, and signaling pathways of TLRs were already well-studied, their precise function in specific cell types, especially innate immune cells, needs to be further clarified. In this study, we showed that when significantly decreased amounts of membrane CD39, an adenosine triphosphate (ATP)-degrading enzyme, were detected in lipopolysaccharide (LPS)-treated bone marrow-derived dendritic cells (BMDCs), Cd39 mRNA expression, and whole-cell CD39 expression were at the same levels as those in untreated BMDCs. Further experiments demonstrated that the downregulation of membrane CD39 expression in LPS-treated BMDCs was mediated by endocytosis, leading to membrane-exposed CD39 downregulation, which was positively associated with decreased enzymatic activity in ATP metabolism and increased extracellular ATP accumulation. The accumulated ATP promoted intracellular calcium accumulation and IL-1β production in BMDCs through P2X7 signaling activation. Further research revealed that not only LPS but also other TLR ligands, excluding polyI:C, induced CD39 internalization in BMDCs and that the MyD88 pathway was critical in this process. The results suggested that the activation of CD39 internalization in DCs induced by a TLR ligand caused increased ATP accumulation, leading to P2X7 receptor activation that mediated a proinflammatory effect. Considering the strong modulatory effect of extracellular ATP accumulation on the immune response and inflammation, the manipulation of membrane CD39 expression on DCs may have implications on the regulation and treatment of inflammatory responses.
Collapse
Affiliation(s)
- Ronglan Zhao
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Weifang Medical University, Weifang, China
| | - Jinjuan Qiao
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Weifang Medical University, Weifang, China
| | - Xumei Zhang
- Department of Pathology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Yansong Zhao
- Department of Ophthalmology, Affiliated Hospital of Weifang Medical University, Weifang, China
| | - Xiangying Meng
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Weifang Medical University, Weifang, China
| | - Deming Sun
- Department of Ophthalmology, David Geffen School of Medicine at UCLA, Doheny Eye Institute, Los Angeles, CA, United States
| | - Xiaoxiang Peng
- Department of Laboratory Medicine, Weifang Medical University, Weifang, China.,Institutional Key Laboratory of Clinical Laboratory Diagnostics, 12th 5-Year Project of Shandong Province, Weifang Medical University, Weifang, China
| |
Collapse
|
40
|
Ohta A. Oxygen-dependent regulation of immune checkpoint mechanisms. Int Immunol 2019; 30:335-343. [PMID: 29846615 DOI: 10.1093/intimm/dxy038] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 05/27/2018] [Indexed: 12/21/2022] Open
Abstract
Immunotherapy of cancer has finally materialized following the success of immune checkpoint blockade. Since down-regulation of immune checkpoint mechanisms is beneficial in cancer treatment, it is important to ask why tumors are infamously filled with the immunosuppressive mechanisms. Indeed, immune checkpoints are physiological negative feedback mechanisms of immune activities, and the induction of such mechanisms is important in preventing excessive destruction of inflamed normal tissues. A condition commonly found in tumors and inflamed tissues is tissue hypoxia. Oxygen deprivation under hypoxic conditions by itself is immunosuppressive because proper oxygen supply could support bioenergetic demands of immune cells for optimal immune responses. However, importantly, hypoxia has been found to up-regulate a variety of immune checkpoints and to be able to drive a shift toward a more immunosuppressive environment. Moreover, extracellular adenosine, which accumulates due to tissue hypoxia, also contributes to the up-regulation of other immune checkpoints. Taken together, tissue oxygen is a key regulator of the immune response by directly affecting the energy status of immune effectors and by regulating the intensity of immunoregulatory activity in the environment. The regulators of various immune checkpoint mechanisms may represent the next focus to modulate the intensity of immune responses and to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Akio Ohta
- Department of Immunology, Foundation for Biomedical Research and Innovation at Kobe, Minatojima-Minamimachi, Kobe, Japan
| |
Collapse
|
41
|
Horenstein AL, Bracci C, Morandi F, Malavasi F. CD38 in Adenosinergic Pathways and Metabolic Re-programming in Human Multiple Myeloma Cells: In-tandem Insights From Basic Science to Therapy. Front Immunol 2019; 10:760. [PMID: 31068926 PMCID: PMC6491463 DOI: 10.3389/fimmu.2019.00760] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 03/21/2019] [Indexed: 01/10/2023] Open
Abstract
Tumor microenvironments are rich in extracellular nucleotides that can be metabolized by ectoenzymes to produce adenosine, a nucleoside involved in controlling immune responses. Multiple myeloma, a plasma cell malignancy developed within a bone marrow niche, exploits adenosinergic pathways to customize the immune homeostasis of the tumor. CD38, a multifunctional protein that acts as both receptor and ectoenzyme, is overexpressed at all stages of myeloma. At neutral and acidic pH, CD38 catalyzes the extracellular conversion of NAD+ to regulators of calcium signaling. The initial disassembly of NAD+ is also followed by adenosinergic activity, if CD38 is operating in the presence of CD203a and CD73 nucleotidases. cAMP extruded from tumor cells provides another substrate for metabolizing nucleotidases to signaling adenosine. These pathways flank or bypass the canonical adenosinergic pathway subjected to the conversion of ATP by CD39. All of the adenosinergic networks can be hijacked by the tumor, thus controlling the homeostatic reprogramming of the myeloma in the bone marrow. In this context, adenosine assumes the role of a local hormone: cell metabolism is adjusted via low- or high-affinity purinergic receptors expressed by immune and bone cells as well as by tumor cells. The result is immunosuppression, which contributes to the failure of immune surveillance in cancer. A similar metabolic strategy silences immune effectors during the progression of indolent gammopathies to symptomatic overt multiple myeloma disease. Plasma from myeloma aspirates contains elevated levels of adenosine resulting from interactions between myeloma and other cells lining the niche and adenosine concentrations are known to increase as the disease progresses. This is statistically reflected in the International Staging System for multiple myeloma. Along with the ability to deplete CD38+ malignant plasma cell populations which has led to their widespread therapeutic use, anti-CD38 antibodies are involved in the polarization and release of microvesicles characterized by the expression of multiple adenosine-producing molecules. These adenosinergic pathways provide new immune checkpoints for improving immunotherapy protocols by helping to restore the depressed immune response.
Collapse
Affiliation(s)
- Alberto L Horenstein
- Laboratory of Immunogenetics, Department of Medical Sciences, Turin, Italy.,CeRMS, University of Torino, Turin, Italy
| | - Cristiano Bracci
- Laboratory of Immunogenetics, Department of Medical Sciences, Turin, Italy.,CeRMS, University of Torino, Turin, Italy
| | - Fabio Morandi
- Stem Cell Laboratory and Cell Therapy Center, Istituto Giannina Gaslini, Genova, Italy
| | - Fabio Malavasi
- Laboratory of Immunogenetics, Department of Medical Sciences, Turin, Italy.,CeRMS, University of Torino, Turin, Italy
| |
Collapse
|
42
|
Vijayamahantesh, Vijayalaxmi. Tinkering with targeting nucleotide signaling for control of intracellular Leishmania parasites. Cytokine 2019; 119:129-143. [PMID: 30909149 DOI: 10.1016/j.cyto.2019.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/23/2022]
Abstract
Nucleotides are one of the most primitive extracellular signalling molecules across all phyla and regulate a multitude of responses. The biological effects of extracellular nucleotides/sides are mediated via the specific purinergic receptors present on the cell surface. In mammalian system, adenine nucleotides are the predominant nucleotides found in the extracellular milieu and mediate a constellation of physiological functions. In the context of host-pathogen interaction, extracellular ATP is recognized as a danger signal and potentiates the release of pro-inflammatory mediators from activated immune cells, on the other hand, its breakdown product adenosine exerts potential anti-inflammatory and immunosuppressive actions. Therefore, it is increasingly apparent that the interplay between extracellular ATP/adenosine ratios has a significant role in coordinating the regulation of the immune system in health and diseases. Several pathogens express ectonucleotidases on their surface and exploit the purinergic signalling as one of the mechanisms to modulate the host immune response. Leishmania pathogens are one of the most successful intracellular pathogens which survive within host macrophages and manipulate protective Th1 response into disease promoting Th2 response. In this review, we discuss the regulation of extracellular ATP and adenosine levels, the role of ATP/adenosine counter signalling in regulating the inflammation and immune responses during infection and how Leishmania parasites exploit the purinergic signalling to manipulate host response. We also discuss the challenges and opportunities in targeting purinergic signalling and the future prospects.
Collapse
Affiliation(s)
- Vijayamahantesh
- Department of Biochemistry, Indian Institute of Science (IISc), Bengaluru, Karnataka, India.
| | - Vijayalaxmi
- Department of Zoology, Karnatak University, Dharwad, Karnataka, India
| |
Collapse
|
43
|
Ando Y, Siegler E, Ta HP, Cinay GE, Zhou H, Gorrell KA, Au H, Jarvis BM, Wang P, Shen K. Evaluating CAR-T Cell Therapy in a Hypoxic 3D Tumor Model. Adv Healthc Mater 2019; 8:e1900001. [PMID: 30734529 PMCID: PMC6448565 DOI: 10.1002/adhm.201900001] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2019] [Revised: 01/18/2019] [Indexed: 12/31/2022]
Abstract
Despite its revolutionary success in hematological malignancies, chimeric antigen receptor T (CAR-T) cell therapy faces disappointing clinical results in solid tumors. The poor efficacy has been partially attributed to the lack of understanding in how CAR-T cells function in a solid tumor microenvironment. Hypoxia plays a critical role in cancer progression and immune editing, which potentially results in solid tumors escaping immunosurveillance and CAR-T cell-mediated cytotoxicity. Mechanistic studies of CAR-T cell biology in a physiological environment has been limited by the complexity of tumor-immune interactions in clinical and animal models, as well as by a lack of reliable in vitro models. A microdevice platform that recapitulates a 3D tumor section with a gradient of oxygen and integrates fluidic channels surrounding the tumor for CAR-T cell delivery is engineered. The design allows for the evaluation of CAR-T cell cytotoxicity and infiltration in the heterogeneous oxygen landscape of in vivo solid tumors at a previously unachievable scale in vitro.
Collapse
Affiliation(s)
- Yuta Ando
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Elizabeth Siegler
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Hoang P. Ta
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Gunce E. Cinay
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Hao Zhou
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Kimberly A. Gorrell
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Hannah Au
- Department of Immunology and Pathogenesis, College of Letters and Science, University of California, Berkeley, CA 94720
| | - Bethany M. Jarvis
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
| | - Pin Wang
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
- Mork Family Department of Chemical Engineering and Materials Science, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| | - Keyue Shen
- Department of Biomedical Engineering, Viterbi School of Engineering, University of Southern California, Los Angeles, CA 90089
- Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
- Department of Stem Cell Biology and Regenerative Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
44
|
Aires ID, Boia R, Rodrigues-Neves AC, Madeira MH, Marques C, Ambrósio AF, Santiago AR. Blockade of microglial adenosine A 2A receptor suppresses elevated pressure-induced inflammation, oxidative stress, and cell death in retinal cells. Glia 2019; 67:896-914. [PMID: 30667095 PMCID: PMC6590475 DOI: 10.1002/glia.23579] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 11/08/2018] [Accepted: 11/28/2018] [Indexed: 12/20/2022]
Abstract
Glaucoma is a retinal degenerative disease characterized by the loss of retinal ganglion cells and damage of the optic nerve. Recently, we demonstrated that antagonists of adenosine A2A receptor (A2A R) control retinal inflammation and afford protection to rat retinal cells in glaucoma models. However, the precise contribution of microglia to retinal injury was not addressed, as well as the effect of A2A R blockade directly in microglia. Here we show that blocking microglial A2A R prevents microglial cell response to elevated pressure and it is sufficient to protect retinal cells from elevated pressure-induced death. The A2A R antagonist SCH 58261 or the knockdown of A2A R expression with siRNA in microglial cells prevented the increase in microglia response to elevated hydrostatic pressure. Furthermore, in retinal neural cell cultures, the A2A R antagonist decreased microglia proliferation, as well as the expression and release of pro-inflammatory mediators. Microglia ablation prevented neural cell death triggered by elevated pressure. The A2A R blockade recapitulated the effects of microglia depletion, suggesting that blocking A2A R in microglia is able to control neurodegeneration in glaucoma-like conditions. Importantly, in human organotypic retinal cultures, A2A R blockade prevented the increase in reactive oxygen species and the morphological alterations in microglia triggered by elevated pressure. These findings place microglia as the main contributors for retinal cell death during elevated pressure and identify microglial A2A R as a therapeutic target to control retinal neuroinflammation and prevent neural apoptosis elicited by elevated pressure.
Collapse
Affiliation(s)
- Inês Dinis Aires
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Raquel Boia
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Ana Catarina Rodrigues-Neves
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Maria Helena Madeira
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Carla Marques
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - António Francisco Ambrósio
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal
| | - Ana Raquel Santiago
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,CNC.IBILI Consortium, University of Coimbra, Coimbra, Portugal.,Association for Innovation and Biomedical Research on Light and Image (AIBILI), Coimbra, Portugal
| |
Collapse
|
45
|
ATP as a Pathophysiologic Mediator of Bacteria-Host Crosstalk in the Gastrointestinal Tract. Int J Mol Sci 2018; 19:ijms19082371. [PMID: 30103545 PMCID: PMC6121306 DOI: 10.3390/ijms19082371] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 08/02/2018] [Accepted: 08/06/2018] [Indexed: 12/12/2022] Open
Abstract
Extracellular nucleotides, such as adenosine triphosphate (ATP), are released from host cells including nerve termini, immune cells, injured or dead cells, and the commensal bacteria that reside in the gut lumen. Extracellular ATP interacts with the host through purinergic receptors, and promotes intercellular and bacteria-host communication to maintain the tissue homeostasis. However, the release of massive concentrations of ATP into extracellular compartments initiates acute and chronic inflammatory responses through the activation of immunocompetent cells (e.g., T cells, macrophages, and mast cells). In this review, we focus on the functions of ATP as a pathophysiologic mediator that is required for the induction and resolution of inflammation and inter-species communication.
Collapse
|
46
|
Adenosine signaling and adenosine deaminase regulation of immune responses: impact on the immunopathogenesis of HIV infection. Purinergic Signal 2018; 14:309-320. [PMID: 30097807 DOI: 10.1007/s11302-018-9619-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Accepted: 07/03/2018] [Indexed: 02/07/2023] Open
Abstract
Infection by human immunodeficiency virus (HIV) causes the acquired immune deficiency syndrome (AIDS), which has devastating effects on the host immune system. HIV entry into host cells and subsequent viral replication induce a proinflammatory response, hyperactivating immune cells and leading them to death, disfunction, and exhaustion. Adenosine is an immunomodulatory molecule that suppresses immune cell function to protect tissue integrity. The anti-inflammatory properties of adenosine modulate the chronic inflammation and immune activation caused by HIV. Lack of adenosine contributes to pathogenic events in HIV infection. However, immunosuppression by adenosine has its shortcomings, such as impairing the immune response, hindering the elimination of the virus and control of viral replication. By attempting to control inflammation, adenosine feeds a pathogenic cycle affecting immune cells. Deamination of adenosine by ADA (adenosine deaminase) counteracts the negative effects of adenosine in immune cells, boosting the immune response. This review comprises the connection between adenosinergic system and HIV immunopathogenesis, exploring defects in immune cell function and the role of ADA in protecting these cells against damage.
Collapse
|
47
|
Leone RD, Sun IM, Oh MH, Sun IH, Wen J, Englert J, Powell JD. Inhibition of the adenosine A2a receptor modulates expression of T cell coinhibitory receptors and improves effector function for enhanced checkpoint blockade and ACT in murine cancer models. Cancer Immunol Immunother 2018; 67:1271-1284. [PMID: 29923026 PMCID: PMC11028354 DOI: 10.1007/s00262-018-2186-0] [Citation(s) in RCA: 116] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 06/08/2018] [Indexed: 01/08/2023]
Abstract
Adenosine signaling via the A2a receptor (A2aR) is emerging as an important checkpoint of immune responses. The presence of adenosine in the inflammatory milieu or generated by the CD39/CD73 axis on tissues or T regulatory cells serves to regulate immune responses. By nature of the specialized metabolism of cancer cells, adenosine levels are increased in the tumor microenvironment and contribute to tumor immune evasion. To this end, small molecule inhibitors of the A2aR are being pursued clinically to enhance immunotherapy. Herein, we demonstrate the ability of the novel A2aR antagonist, CPI-444, to dramatically enhance immunologic responses in models of checkpoint therapy and ACT in cancer. Furthermore, we demonstrate that A2aR blockade with CPI-444 decreases expression of multiple checkpoint pathways, including PD-1 and LAG-3, on both CD8+ effector T cells (Teff) and FoxP3+ CD4+ regulatory T cells (Tregs). Interestingly, our studies demonstrate that A2aR blockade likely has its most profound effects during Teff cell activation, significantly decreasing PD-1 and LAG-3 expression at the draining lymph nodes of tumor bearing mice. In contrast to previous reports using A2aR knockout models, pharmacologic blockade with CPI-444 did not impede CD8 T cell persistence or memory recall. Overall these findings not only redefine our understanding of the mechanisms by which adenosine inhibits immunity but also have important implications for the design of novel immunotherapy regimens.
Collapse
MESH Headings
- Adenosine A2 Receptor Antagonists/pharmacology
- Animals
- Antigens, CD/chemistry
- Antigens, CD/metabolism
- CD8-Positive T-Lymphocytes/immunology
- Colonic Neoplasms/drug therapy
- Colonic Neoplasms/immunology
- Colonic Neoplasms/metabolism
- Colonic Neoplasms/pathology
- Female
- Gene Expression Regulation, Neoplastic
- Immunotherapy
- Lymphocytes, Tumor-Infiltrating
- Male
- Melanoma, Experimental/drug therapy
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Programmed Cell Death 1 Receptor/metabolism
- Receptor, Adenosine A2A/chemistry
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes, Regulatory/immunology
- Tumor Cells, Cultured
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
- Lymphocyte Activation Gene 3 Protein
Collapse
Affiliation(s)
- Robert D Leone
- Department of Oncology, Sidney-Kimmel Comprehensive Cancer Research Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Im-Meng Sun
- Department of Oncology, Sidney-Kimmel Comprehensive Cancer Research Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Min-Hee Oh
- Department of Oncology, Sidney-Kimmel Comprehensive Cancer Research Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Im-Hong Sun
- Department of Oncology, Sidney-Kimmel Comprehensive Cancer Research Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jiayu Wen
- Department of Oncology, Sidney-Kimmel Comprehensive Cancer Research Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Judson Englert
- Department of Oncology, Sidney-Kimmel Comprehensive Cancer Research Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- MedImmune, LLC, Gaithersburg, MD, 20878, USA
| | - Jonathan D Powell
- Department of Oncology, Sidney-Kimmel Comprehensive Cancer Research Center, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, 1650 Orleans Street, CRB1 Room 453, Baltimore, MD, 21231, USA.
| |
Collapse
|
48
|
Jung D, Alshaikh A, Ratakonda S, Bashir M, Amin R, Jeon S, Stevens J, Sharma S, Ahmed W, Musch M, Hassan H. Adenosinergic signaling inhibits oxalate transport by human intestinal Caco2-BBE cells through the A 2B adenosine receptor. Am J Physiol Cell Physiol 2018; 315:C687-C698. [PMID: 30020825 DOI: 10.1152/ajpcell.00024.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Most kidney stones (KS) are composed of calcium oxalate, and small increases in urine oxalate affect the stone risk. Intestinal oxalate secretion mediated by anion exchanger SLC26A6 (PAT1) plays a crucial role in limiting net absorption of ingested oxalate, thereby preventing hyperoxaluria and related KS, reflecting the importance of understanding regulation of intestinal oxalate transport. We previously showed that ATP and UTP inhibit oxalate transport by human intestinal Caco2-BBE cells (C2). Since ATP is rapidly degraded to adenosine (ADO), we examined whether intestinal oxalate transport is regulated by ADO. We measured [14C]oxalate uptake in the presence of an outward Cl gradient as an assay of Cl-oxalate exchange activity, ≥49% of which is PAT1-mediated in C2 cells. We found that ADO significantly inhibited oxalate transport by C2 cells, an effect completely blocked by the nonselective ADO receptor antagonist 8- p-sulfophenyltheophylline. ADO also significantly inhibited oxalate efflux by C2 cells, which is important since PAT1 mediates oxalate efflux in vivo. Using pharmacological antagonists and A2B adenosine receptor (A2B AR) siRNA knockdown studies, we observed that ADO inhibits oxalate transport through the A2B AR, phospholipase C, and PKC. ADO inhibits oxalate transport by reducing PAT1 surface expression as shown by biotinylation studies. We conclude that ADO inhibits oxalate transport by lowering PAT1 surface expression in C2 cells through signaling pathways including the A2B AR, PKC, and phospholipase C. Given higher ADO levels and overexpression of the A2B AR in inflammatory bowel disease (IBD), our findings have potential relevance to pathophysiology of IBD-associated hyperoxaluria and related KS.
Collapse
Affiliation(s)
- Daniel Jung
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | - Altayeb Alshaikh
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | | | - Mohamed Bashir
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | - Ruhul Amin
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | - Sohee Jeon
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | - Jan Stevens
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | - Sapna Sharma
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | - Wahaj Ahmed
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | - Mark Musch
- Department of Medicine, The University of Chicago , Chicago, Illinois
| | - Hatim Hassan
- Department of Medicine, The University of Chicago , Chicago, Illinois
| |
Collapse
|
49
|
Liang D, Shao H, Born WK, O'Brien RL, Kaplan HJ, Sun D. High level expression of A2ARs is required for the enhancing function, but not for the inhibiting function, of γδ T cells in the autoimmune responses of EAU. PLoS One 2018; 13:e0199601. [PMID: 29928041 PMCID: PMC6013223 DOI: 10.1371/journal.pone.0199601] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022] Open
Abstract
We previously reported that activated γδ T cells greatly enhance autoimmune responses, particularly the Th17 response. To determine the mechanisms involved, we made a series of comparisons between activated and non-activated γδ T cells. Our results showed that activated γδ T cells expressed greatly increased levels of A2A adenosine receptor (A2AR) and decreased amounts of CD73, as well as increased amounts of T cell activation markers such as CD69, CD44 and CD25. We show that A2AR is a major functional molecule in the enhancing activity of γδ T cells. A2AR-/- γδ T cells (isolated from A2AR-/- mouse), lost their Th17-enhancing activity as did A2AR+/+ γδ T cells (isolated from wt-B6 mouse) after treatment with an A2AR antagonist. Since γδ T cells possess either an enhancing or an inhibiting effect, we also tested whether A2AR expression on γδ T cells is essential to their inhibiting effect. Our results showed that the inhibiting effect of A2AR-/- γδ T cells was as potent as that of A2AR+/+ γδ T cells. In a previous report we showed that the expression of different levels of CD73 molecule allowed γδ T cells to adjust their suppressive activity; in the current study, we show that expression of increased amounts of A2AR allows γδ T cells to more effectively exert their enhancing function.
Collapse
Affiliation(s)
- Dongchun Liang
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| | - Hui Shao
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Willi K. Born
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States of America
| | - Rebecca L. O'Brien
- Department of Biomedical Research, National Jewish Health, Denver, CO, United States of America
| | - Henry J. Kaplan
- Department of Ophthalmology and Visual Sciences, Kentucky Lions Eye Center, University of Louisville, Louisville, Kentucky, United States of America
| | - Deming Sun
- Doheny Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, United States of America
| |
Collapse
|
50
|
López-Cruz L, Salamone JD, Correa M. Caffeine and Selective Adenosine Receptor Antagonists as New Therapeutic Tools for the Motivational Symptoms of Depression. Front Pharmacol 2018; 9:526. [PMID: 29910727 PMCID: PMC5992708 DOI: 10.3389/fphar.2018.00526] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 05/01/2018] [Indexed: 01/06/2023] Open
Abstract
Major depressive disorder is one of the most common and debilitating psychiatric disorders. Some of the motivational symptoms of depression, such anergia (lack of self-reported energy) and fatigue are relatively resistant to traditional treatments such as serotonin uptake inhibitors. Thus, new pharmacological targets are being investigated. Epidemiological data suggest that caffeine consumption can have an impact on aspects of depressive symptomatology. Caffeine is a non-selective adenosine antagonist for A1/A2A receptors, and has been demonstrated to modulate behavior in classical animal models of depression. Moreover, selective adenosine receptor antagonists are being assessed for their antidepressant effects in animal studies. This review focuses on how caffeine and selective adenosine antagonists can improve different aspects of depression in humans, as well as in animal models. The effects on motivational symptoms of depression such as anergia, fatigue, and psychomotor slowing receive particular attention. Thus, the ability of adenosine receptor antagonists to reverse the anergia induced by dopamine antagonism or depletion is of special interest. In conclusion, although further studies are needed, it appears that caffeine and selective adenosine receptor antagonists could be therapeutic agents for the treatment of motivational dysfunction in depression.
Collapse
Affiliation(s)
- Laura López-Cruz
- Àrea de Psicobiologia, Universitat Jaume I, Castellón de la Plana, Spain
| | - John D. Salamone
- Behavioral Neuroscience Division, University of Connecticut, Storrs, CT, United States
| | - Mercè Correa
- Àrea de Psicobiologia, Universitat Jaume I, Castellón de la Plana, Spain
- Behavioral Neuroscience Division, University of Connecticut, Storrs, CT, United States
| |
Collapse
|