1
|
Cruz-Muñoz JR, Valdez-Morales EE, Barajas-Espinosa A, Barrios-García T, Liñán-Rico A, Guerrero-Alba R. Gene expression alterations of purinergic signaling components in obesity-associated intestinal low-grade inflammation in type 2 diabetes. Purinergic Signal 2024; 20:629-643. [PMID: 38587723 PMCID: PMC11555165 DOI: 10.1007/s11302-024-10006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 03/26/2024] [Indexed: 04/09/2024] Open
Abstract
Intestinal low-grade inflammation induced by a high-fat diet has been found to detonate chronic systemic inflammation, which is a hallmark of obesity, and precede the apparition of insulin resistance, a key factor for developing type 2 diabetes (T2D). Aberrant purinergic signaling pathways have been implicated in the pathogenesis of inflammatory bowel disease and other gastrointestinal diseases. However, their role in the gut inflammation associated with obesity and T2D remains unexplored. C57BL/6 J mice were fed a cafeteria diet for 21 weeks and received one injection of streptozotocin in their sixth week into the diet. The gene expression profile of purinergic signaling components in colon tissue was assessed by RT-qPCR. Compared to control mice, the treated group had a significant reduction in colonic length and mucosal and muscular layer thickness accompanied by increased NF-κB and IL-1β mRNA expression. Furthermore, colonic P2X2, P2X7, and A3R gene expression levels were lower, while the P2Y2, NT5E, and ADA expression levels increased. In conclusion, these data suggest that these purinergic signaling components possibly play a role in intestinal low-grade inflammation associated with obesity and T2D and thus could represent a novel therapeutic target for the treatment of the metabolic complications related to these diseases.
Collapse
Affiliation(s)
- José R Cruz-Muñoz
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Ags, México
| | - Eduardo E Valdez-Morales
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Ags, México
| | - Alma Barajas-Espinosa
- Escuela Superior de Huejutla, Universidad Autónoma del Estado de Hidalgo, Huejutla de Reyes, Hidalgo, México
| | - Tonatiuh Barrios-García
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Ags, México
| | - Andrómeda Liñán-Rico
- Centro Universitario de Investigaciones Biomédicas. Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT), Universidad de Colima, Colima, México.
| | - Raquel Guerrero-Alba
- Departamento de Fisiología y Farmacología, Centro de Ciencias Básicas, Universidad Autónoma de Aguascalientes, Aguascalientes, Ags, México.
| |
Collapse
|
2
|
Ma K, Miao L, Li B, Yu W, Liu F, Liu K, Li Y, Huang C, Yang Z. Mechanism of action of Nrf2 and its related natural regulators in rheumatoid arthritis. J Orthop Surg Res 2024; 19:759. [PMID: 39543632 DOI: 10.1186/s13018-024-05221-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by synovitis that can lead to joint deformities. To date, more than 18 million individuals worldwide have been diagnosed with RA, making it one of the most prevalent autoimmune diseases globally and posing a significant threat to public health and safety. Due to the complex pathogenesis of the disease, which involves autoimmunity, genetics, inflammation and oxidative stress in the body's tissues, the current drug therapy generally targets a single molecule, and effective and efficient drugs involving multiple levels and targets are lacking; thus, there is an urgent need for high-quality research and treatment in this field. Nuclear transcription factor erythroid 2-associated factor 2 (Nrf2) plays a crucial role in cellular resistance to oxidative stress and electrophilic attacks and is a potential pharmacological target for chronic disease treatment. While currently no drugs that target Nrf2 have been approved specifically for RA treatment, such an approach holds great significance. In recent years, the use of natural products to treat RA and other chronic conditions has become increasingly widespread because of their superior efficacy and minimal side effects. Therefore, this article provides a review of the mechanism of Nrf2 in RA and summarizes natural products that target Nrf2 and its associated pathways in the treatment of RA, aiming to offer new insights and strategies for the prevention and management of RA.
Collapse
Affiliation(s)
- Ke Ma
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Lili Miao
- Department of Experiment Center, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Bo Li
- Department of Orthopaedics, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Wenfei Yu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Fengzhao Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Kun Liu
- Department of Orthopaedics, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Yang Li
- Department of Orthopaedics, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Chengcheng Huang
- Department of Endocrinology, Shandong University of Traditional Chinese Medicine Affiliated Hospital, Jinan, 250014, Shandong, China
| | - Zhenguo Yang
- Department of Orthopaedics, Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China.
| |
Collapse
|
3
|
Zhang X, Chen X, Meng X, Wu Y, Gao J, Chen H, Li X. Extracellular adenosine triphosphate: A new gateway for food allergy mechanism research? Food Chem 2024; 464:141821. [PMID: 39486282 DOI: 10.1016/j.foodchem.2024.141821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/24/2024] [Accepted: 10/26/2024] [Indexed: 11/04/2024]
Abstract
Although various studies have been conducted, the detailed mechanisms of food allergy remain a topic of ongoing debate. Recently, researchers have reported that extracellular adenosine triphosphate (eATP), a member of damage-associated molecular patterns secreted by stressed cells, plays a critical role in the progression of asthma and atopic dermatitis. These studies suggest that dysregulated eATP significantly influences various aspects of disease progression, from bodily sensitization to the emergence of clinical manifestations. Given the shared pathogenic mechanisms among asthma, atopic dermatitis, and food allergies, we hypothesize that eATP may also serve as a crucial regulator in the development of food allergies. To elucidate this hypothesis, we first summarize the evidence and limitations of food allergy theories, then discuss the roles of eATP in allergic diseases. We conclude with speculative insights into the potential influence of eATP on food allergy development, aiming to inspire further investigation into the molecular mechanisms of food allergies.
Collapse
Affiliation(s)
- Xing Zhang
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; School of Food Science and Technology, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Xiao Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; School of Food Science and Technology, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Xuanyi Meng
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Yong Wu
- Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Jinyan Gao
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; School of Food Science and Technology, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Hongbing Chen
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; School of Food Science and Technology, Nanchang University, Nanchang 330047, PR China; Sino-German Joint Research Institute, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China
| | - Xin Li
- State Key Laboratory of Food Science and Resources, Nanchang University, Nanchang 330047, PR China; School of Food Science and Technology, Nanchang University, Nanchang 330047, PR China; Jiangxi Province Key Laboratory of Food Allergy, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
4
|
Czopik AK, McNamee EN, Vaughn V, Huang X, Bang IH, Clark T, Wang Y, Ruan W, Nguyen T, Masterson JC, Tak E, Frank S, Collins CB, Li H, Rodriguez-Aguayo C, Lopez-Berestein G, Gerich ME, Furuta GT, Yuan X, Sood AK, de Zoeten EF, Eltzschig HK. HIF-2α-dependent induction of miR-29a restrains T H1 activity during T cell dependent colitis. Nat Commun 2024; 15:8042. [PMID: 39271652 PMCID: PMC11399416 DOI: 10.1038/s41467-024-52113-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Metabolic imbalance leading to inflammatory hypoxia and stabilization of hypoxia-inducible transcription factors (HIFs) is a hallmark of inflammatory bowel diseases. We hypothesize that HIF could be stabilized in CD4+ T cells during intestinal inflammation and alter the functional responses of T cells via regulation of microRNAs. Our assays reveal markedly increased T cell-intrinsic hypoxia and stabilization of HIF protein during experimental colitis. microRNA screen in primary CD4+ T cells points us towards miR-29a and our subsequent studies identify a selective role for HIF-2α in CD4-cell-intrinsic induction of miR-29a during hypoxia. Mice with T cell-intrinsic HIF-2α deletion display elevated T-bet (target of miR-29a) levels and exacerbated intestinal inflammation. Mice with miR-29a deficiency in T cells show enhanced intestinal inflammation. T cell-intrinsic overexpression of HIF-2α or delivery of miR-29a mimetic dampen TH1-driven colitis. In this work, we show a previously unrecognized function for hypoxia-dependent induction of miR-29a in attenuating TH1-mediated inflammation.
Collapse
Affiliation(s)
- Agnieszka K Czopik
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Eóin N McNamee
- Mucosal Inflammation Program, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Digestive Health Institute, Children's Hospital Colorado, Aurora, CO, USA
| | - Victoria Vaughn
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xiangsheng Huang
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - In Hyuk Bang
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Trent Clark
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yanyu Wang
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Wei Ruan
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Tom Nguyen
- Mucosal Inflammation Program, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Digestive Health Institute, Children's Hospital Colorado, Aurora, CO, USA
| | - Joanne C Masterson
- Mucosal Inflammation Program, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Gastrointestinal Eosinophilic Disease Program University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Eunyoung Tak
- Digestive Health Institute, Children's Hospital Colorado, Aurora, CO, USA
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Sandra Frank
- Organ Protection Program, Department of Anesthesiology, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
- Department of Anaesthesiology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Colm B Collins
- Mucosal Inflammation Program, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Howard Li
- Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado - Anschutz Medical Campus, Aurora, CO, USA
| | - Cristian Rodriguez-Aguayo
- Departmental of Experimental Therapeutics and Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Lopez-Berestein
- Departmental of Experimental Therapeutics and Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark E Gerich
- Mucosal Inflammation Program, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Division of Gastroenterology & Hepatology, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Glenn T Furuta
- Mucosal Inflammation Program, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Gastrointestinal Eosinophilic Disease Program University of Colorado Anschutz School of Medicine, Aurora, CO, USA
- Department of Pediatrics, University of Colorado Anschutz School of Medicine, Aurora, CO, USA
| | - Xiaoyi Yuan
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Anil K Sood
- Departmental of Experimental Therapeutics and Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Edwin F de Zoeten
- Mucosal Inflammation Program, University of Colorado Anschutz School of Medicine, Aurora, CO, USA.
- Department of Pediatrics, University of Colorado Anschutz School of Medicine, Aurora, CO, USA.
| | - Holger K Eltzschig
- Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Outcomes Research, Department of Anesthesiology, Critical Care and Pain Medicine, McGovern Medical School, The University of Texas Health Science Center, Houston, TX, USA
| |
Collapse
|
5
|
Di Mattia M, Sallese M, Neri M, Lopetuso LR. Hypoxic Functional Regulation Pathways in the GI Tract: Focus on the HIF-1α and Microbiota's Crosstalk. Inflamm Bowel Dis 2024; 30:1406-1418. [PMID: 38484200 DOI: 10.1093/ibd/izae046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Indexed: 08/02/2024]
Abstract
Hypoxia is an essential gastrointestinal (GI) tract phenomenon that influences both physiologic and pathologic states. Hypoxia-inducible factors (HIFs), the primary drivers of cell adaptation to low-oxygen environments, have been identified as critical regulators of gut homeostasis: directly, through the induction of different proteins linked to intestinal barrier stabilization (ie, adherent proteins, tight junctions, mucins, integrins, intestinal trefoil factor, and adenosine); and indirectly, through the regulation of several immune cell types and the modulation of autophagy and inflammatory processes. Furthermore, hypoxia and HIF-related sensing pathways influence the delicate relationship existing between bacteria and mammalian host cells. In turn, gut commensals establish and maintain the physiologic hypoxia of the GI tract and HIF-α expression. Based on this premise, the goals of this review are to (1) highlight hypoxic molecular pathways in the GI tract, both in physiologic and pathophysiologic settings, such as inflammatory bowel disease; and (2) discuss a potential strategy for ameliorating gut-related disorders, by targeting HIF signaling, which can alleviate inflammatory processes, restore autophagy correct mechanisms, and benefit the host-microbiota equilibrium.
Collapse
Affiliation(s)
- Miriam Di Mattia
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Michele Sallese
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Matteo Neri
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
| | - Loris Riccardo Lopetuso
- Department of Medicine and Ageing Sciences, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, Gabriele d'Annunzio University of Chieti-Pescara, Chieti, Italy
- Medicina Interna e Gastroenterologia, CEMAD Centro Malattie dell'Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche, Fondazione Policlinico Universitario Gemelli IRCCS, Rome, Italy
| |
Collapse
|
6
|
Jonić N, Koprivica I, Chatzigiannis CM, Tsiailanis AD, Kyrkou SG, Tzakos EP, Pavić A, Dimitrijević M, Jovanović A, Jovanović MB, Marinho S, Castro-Almeida I, Otašević V, Moura-Alves P, Tzakos AG, Stojanović I. Development of FluoAHRL: A Novel Synthetic Fluorescent Compound That Activates AHR and Potentiates Anti-Inflammatory T Regulatory Cells. Molecules 2024; 29:2988. [PMID: 38998940 PMCID: PMC11243367 DOI: 10.3390/molecules29132988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/05/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Aryl Hydrocarbon Receptor (AHR) ligands, upon binding, induce distinct gene expression profiles orchestrated by the AHR, leading to a spectrum of pro- or anti-inflammatory effects. In this study, we designed, synthesized and evaluated three indole-containing potential AHR ligands (FluoAHRL: AGT-4, AGT-5 and AGT-6). All synthesized compounds were shown to emit fluorescence in the near-infrared. Their AHR agonist activity was first predicted using in silico docking studies, and then confirmed using AHR luciferase reporter cell lines. FluoAHRLs were tested in vitro using mouse peritoneal macrophages and T lymphocytes to assess their immunomodulatory properties. We then focused on AGT-5, as it illustrated the predominant anti-inflammatory effects. Notably, AGT-5 demonstrated the ability to foster anti-inflammatory regulatory T cells (Treg) while suppressing pro-inflammatory T helper (Th)17 cells in vitro. AGT-5 actively induced Treg differentiation from naïve CD4+ cells, and promoted Treg proliferation, cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) expression and interleukin-10 (IL-10) production. The increase in IL-10 correlated with an upregulation of Signal Transducer and Activator of Transcription 3 (STAT3) expression. Importantly, the Treg-inducing effect of AGT-5 was also observed in human tonsil cells in vitro. AGT-5 showed no toxicity when applied to zebrafish embryos and was therefore considered safe for animal studies. Following oral administration to C57BL/6 mice, AGT-5 significantly upregulated Treg while downregulating pro-inflammatory Th1 cells in the mesenteric lymph nodes. Due to its fluorescent properties, AGT-5 could be visualized both in vitro (during uptake by macrophages) and ex vivo (within the lamina propria of the small intestine). These findings make AGT-5 a promising candidate for further exploration in the treatment of inflammatory and autoimmune diseases.
Collapse
Affiliation(s)
- Natalija Jonić
- Department of Immunology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia; (N.J.); (I.K.); (M.D.)
| | - Ivan Koprivica
- Department of Immunology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia; (N.J.); (I.K.); (M.D.)
| | - Christos M. Chatzigiannis
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece; (C.M.C.); (A.D.T.); (S.G.K.)
| | - Antonis D. Tsiailanis
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece; (C.M.C.); (A.D.T.); (S.G.K.)
| | - Stavroula G. Kyrkou
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece; (C.M.C.); (A.D.T.); (S.G.K.)
| | | | - Aleksandar Pavić
- Laboratory for Microbial Molecular Genetics and Ecology, Institute for Molecular Genetics and Genetic Engineering, University of Belgrade, 11000 Belgrade, Serbia;
| | - Mirjana Dimitrijević
- Department of Immunology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia; (N.J.); (I.K.); (M.D.)
| | - Andjelina Jovanović
- Department of Otorhinolaryngology with Maxillofacial Surgery, Clinical Hospital Center “Zemun”, 11080 Belgrade, Serbia; (A.J.); (M.B.J.)
| | - Milan B. Jovanović
- Department of Otorhinolaryngology with Maxillofacial Surgery, Clinical Hospital Center “Zemun”, 11080 Belgrade, Serbia; (A.J.); (M.B.J.)
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Sérgio Marinho
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; (S.M.); (I.C.-A.)
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Inês Castro-Almeida
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; (S.M.); (I.C.-A.)
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Vesna Otašević
- Department of Molecular Biology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia;
| | - Pedro Moura-Alves
- Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; (S.M.); (I.C.-A.)
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Andreas G. Tzakos
- Section of Organic Chemistry & Biochemistry, Department of Chemistry, University of Ioannina, 45110 Ioannina, Greece; (C.M.C.); (A.D.T.); (S.G.K.)
- Institute of Materials Science and Computing, University Research Center of Ioannina (URCI), 45110 Ioannina, Greece
| | - Ivana Stojanović
- Department of Immunology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, 11108 Belgrade, Serbia; (N.J.); (I.K.); (M.D.)
| |
Collapse
|
7
|
Belyayev L, Kang J, Sadat M, Loh K, Patil D, Muralidaran V, Khan K, Kaufman S, Subramanian S, Gusev Y, Bhuvaneshwar K, Ressom H, Varghese R, Ekong U, Matsumoto CS, Robson SC, Fishbein TM, Kroemer A. Suppressor T helper type 17 cell responses in intestinal transplant recipients with allograft rejection. Hum Immunol 2024; 85:110773. [PMID: 38494386 DOI: 10.1016/j.humimm.2024.110773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
BACKGROUND Intestinal transplant (ITx) rejection is associated with memory T helper type 17 cell (Th17) infiltration of grafted tissues. Modulation of Th17 effector cell response is facilitated by T regulatory (Treg) cells, but a phenotypic characterization of this process is lacking in the context of allograft rejection. METHODS Flow cytometry was performed to examine the expression of surface receptors, cytokines, and transcription factors in Th17 and Treg cells in ITx control (n = 34) and rejection patients (n = 23). To elucidate key pathways guiding the rejection biology, we utilized RNA sequencing (RNAseq) and assessed epigenetic stability through pyrosequencing of the Treg-specific demethylated region (TSDR). RESULTS We found that intestinal allograft rejection is characterized by Treg cellular infiltrates, which are polarized toward Th17-type chemokine receptor, ROR-γt transcription factor expression, and cytokine production. These Treg cell subsets have maintained epigenetic stability, as defined by FoxP3-TSDR methylation status, but displayed upregulation of functional Treg and purinergic signaling genes by RNAseq analysis such as CD39, in keeping with suppressor Th17 properties. CONCLUSION We show that ITx rejection is associated with increased polarized cells that express a Th17-like phenotype concurrent with regulatory purinergic markers.
Collapse
Affiliation(s)
- Leonid Belyayev
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA; Walter Reed National Military Medical Center, 8901 Wisconsin Avenue, Bethesda, MD 20814, USA
| | - Jiman Kang
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA; Department of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC 20007, USA
| | - Mohammed Sadat
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Katrina Loh
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Digvijay Patil
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Vinona Muralidaran
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Khalid Khan
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Stuart Kaufman
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Sukanya Subramanian
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Yuriy Gusev
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, 2115 Wisconsin Ave NW, Suite 110, Washington, DC 20075, USA
| | - Krithika Bhuvaneshwar
- Innovation Center for Biomedical Informatics (ICBI), Georgetown University Medical Center, 2115 Wisconsin Ave NW, Suite 110, Washington, DC 20075, USA
| | - Habtom Ressom
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20008, USA
| | - Rency Varghese
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20008, USA
| | - Udeme Ekong
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Cal S Matsumoto
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Simon C Robson
- Center for Inflammation Research, Department of Anesthesiology, Beth Israel Deaconess Medical Center, Harvard Medical School, 25 Shattuck St, Boston, MA 02115, USA
| | - Thomas M Fishbein
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA
| | - Alexander Kroemer
- MedStar Georgetown Transplant Institute, MedStar Georgetown University Hospital and the Center for Translational Transplant Medicine, Georgetown University Medical Center, 3800 Reservoir Road NW, Washington, DC 20007, USA.
| |
Collapse
|
8
|
Wang H, Wei Y, Wang N. Purinergic pathways and their clinical use in the treatment of acute myeloid leukemia. Purinergic Signal 2024:10.1007/s11302-024-09997-8. [PMID: 38446337 DOI: 10.1007/s11302-024-09997-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/21/2024] [Indexed: 03/07/2024] Open
Abstract
Despite the use of various therapies such as hematopoietic stem cell transplantation and chimeric antigen receptor T cell therapy (CAR-T), the prognosis of patients with acute myeloid leukemia (AML) is still generally poor. However, immunotherapy is currently a hot topic in the treatment of hematological tumors. Extracellular adenosine triphosphate (ATP) can be converted to adenosine diphosphate (ADP) via CD39, and ADP can be converted to adenosine via CD73, which can bind to P1 and P2 receptors to exert immunomodulatory effects. Research on the mechanism of the purinergic signaling pathway can provide a new direction for the treatment of AML, and inhibitors of this signaling pathway have been discovered by several researchers and gradually applied in the clinic. In this paper, the mechanism of the purinergic signaling pathway and its clinical application are described, revealing a new target for the treatment of AML and subsequent improvement in patient prognosis.
Collapse
Affiliation(s)
- Huijuan Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Yujie Wei
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Na Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China.
| |
Collapse
|
9
|
Cao Y, Ibrahim KS, Li X, Wong A, Wu Y, Yu XD, Zhou X, Tan Z, He Z, Craft JA, Shu X. Chinese medicine, Qijudihuang pill, mediates cholesterol metabolism and regulates gut microbiota in high-fat diet-fed mice, implications for age-related macular degeneration. Front Immunol 2023; 14:1274401. [PMID: 37901244 PMCID: PMC10602650 DOI: 10.3389/fimmu.2023.1274401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/21/2023] [Indexed: 10/31/2023] Open
Abstract
Background Traditional Chinese Medicines have been used for thousands of years but without any sound empirical basis. One such preparation is the Qijudihuang pill (QP), a mixture of eight herbs, that has been used in China for the treatment of various conditions including age-related macular degeneration (AMD), the most common cause of blindness in the aged population. In order to explain the mechanism behind the effect of QP, we used an AMD model of high-fat diet (HFD) fed mice to investigate cholesterol homeostasis, oxidative stress, inflammation and gut microbiota. Methods Mice were randomly divided into three groups, one group was fed with control diet (CD), the other two groups were fed with high-fat-diet (HFD). One HFD group was treated with QP, both CD and the other HFD groups were treated with vehicles. Tissue samples were collected after the treatment. Cholesterol levels in retina, retinal pigment epithelium (RPE), liver and serum were determined using a commercial kit. The expression of enzymes involved in cholesterol metabolism, inflammation and oxidative stress was measured with qRT-PCR. Gut microbiota was analyzed using 16S rRNA sequencing. Results In the majority of the lipid determinations, analytes were elevated by HFD but this was reversed by QP. Cholesterol metabolism including the enzymes of bile acid (BA) formation was suppressed by HFD but again this was reversed by QP. BAs play a major role in signaling between host and microbiome and this is disrupted by HFD resulting in major changes in the composition of colonic bacterial communities. Associated with these changes are predictions of the metabolic pathway complexity and abundance of individual pathways. These concerned substrate breakdowns, energy production and the biosynthesis of pro-inflammatory factors but were changed back to control characteristics by QP. Conclusion We propose that the ability of QP to reverse these HFD-induced effects is related to mechanisms acting to lower cholesterol level, oxidative stress and inflammation, and to modulate gut microbiota.
Collapse
Affiliation(s)
- Yanqun Cao
- Pu Ai Medical School, Shaoyang University, Shaoyang, Hunan, China
| | - Khalid S. Ibrahim
- Department of Biological and Biomedical Sciences , Glasgow Caledonian University, Glasgow, United Kingdom
- Department of Biology, Faculty of Science, University of Zakho, Zakho, Iraq
| | - Xing Li
- Pu Ai Medical School, Shaoyang University, Shaoyang, Hunan, China
| | - Aileen Wong
- Department of Biological and Biomedical Sciences , Glasgow Caledonian University, Glasgow, United Kingdom
| | - Yi Wu
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xu-Dong Yu
- Pu Ai Medical School, Shaoyang University, Shaoyang, Hunan, China
| | - Xinzhi Zhou
- Department of Biological and Biomedical Sciences , Glasgow Caledonian University, Glasgow, United Kingdom
| | - Zhoujin Tan
- School of Traditional Chinese Medicine, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhiming He
- Pu Ai Medical School, Shaoyang University, Shaoyang, Hunan, China
| | - John A. Craft
- Department of Biological and Biomedical Sciences , Glasgow Caledonian University, Glasgow, United Kingdom
| | - Xinhua Shu
- Pu Ai Medical School, Shaoyang University, Shaoyang, Hunan, China
- Department of Biological and Biomedical Sciences , Glasgow Caledonian University, Glasgow, United Kingdom
- Department of Vision Science , Glasgow Caledonian University, Glasgow, United Kingdom
| |
Collapse
|
10
|
Lis-López L, Bauset C, Seco-Cervera M, Macias-Ceja D, Navarro F, Álvarez Á, Esplugues JV, Calatayud S, Barrachina MD, Ortiz-Masià D, Cosín-Roger J. P2X7 Receptor Regulates Collagen Expression in Human Intestinal Fibroblasts: Relevance in Intestinal Fibrosis. Int J Mol Sci 2023; 24:12936. [PMID: 37629116 PMCID: PMC10454509 DOI: 10.3390/ijms241612936] [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: 07/07/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Intestinal fibrosis is a common complication that affects more than 50% of Crohn´s Disease (CD) patients. There is no pharmacological treatment against this complication, with surgery being the only option. Due to the unknown role of P2X7 in intestinal fibrosis, we aim to analyze the relevance of this receptor in CD complications. Surgical resections from CD and non-Inflammatory Bowel Disease (IBD) patients were obtained. Intestinal fibrosis was induced with two different murine models: heterotopic transplant model and chronic-DSS colitis in wild-type and P2X7-/- mice. Human small intestine fibroblasts (HSIFs) were transfected with an siRNA against P2X7 and treated with TGF-β. A gene and protein expression of P2X7 receptor was significantly increased in CD compared to non-IBD patients. The lack of P2X7 in mice provoked an enhanced collagen deposition and increased expression of several profibrotic markers in both murine models of intestinal fibrosis. Furthermore, P2X7-/- mice exhibited a higher expression of proinflammatory cytokines and a lower expression of M2 macrophage markers. Moreover, the transient silencing of the P2X7 receptor in HSIFs significantly induced the expression of Col1a1 and potentiated the expression of Col4 and Col5a1 after TGF-β treatment. P2X7 regulates collagen expression in human intestinal fibroblasts, while the lack of this receptor aggravates intestinal fibrosis.
Collapse
Affiliation(s)
- Lluis Lis-López
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (L.L.-L.); (C.B.); (D.M.-C.); (Á.Á.); (J.V.E.); (S.C.); (M.D.B.)
| | - Cristina Bauset
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (L.L.-L.); (C.B.); (D.M.-C.); (Á.Á.); (J.V.E.); (S.C.); (M.D.B.)
| | - Marta Seco-Cervera
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica), Hospital Dr. Peset, 46017 Valencia, Spain;
| | - Dulce Macias-Ceja
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (L.L.-L.); (C.B.); (D.M.-C.); (Á.Á.); (J.V.E.); (S.C.); (M.D.B.)
| | - Francisco Navarro
- Servicio Cirugía y Coloproctología, Hospital de Manises, 46940 Valencia, Spain;
| | - Ángeles Álvarez
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (L.L.-L.); (C.B.); (D.M.-C.); (Á.Á.); (J.V.E.); (S.C.); (M.D.B.)
- CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), 28029 Madrid, Spain
| | - Juan Vicente Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (L.L.-L.); (C.B.); (D.M.-C.); (Á.Á.); (J.V.E.); (S.C.); (M.D.B.)
- CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), 28029 Madrid, Spain
| | - Sara Calatayud
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (L.L.-L.); (C.B.); (D.M.-C.); (Á.Á.); (J.V.E.); (S.C.); (M.D.B.)
- CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), 28029 Madrid, Spain
| | - Maria Dolores Barrachina
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (L.L.-L.); (C.B.); (D.M.-C.); (Á.Á.); (J.V.E.); (S.C.); (M.D.B.)
- CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), 28029 Madrid, Spain
| | - Dolores Ortiz-Masià
- CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), 28029 Madrid, Spain
- Departamento de Medicina, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain;
| | - Jesús Cosín-Roger
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, 46010 Valencia, Spain; (L.L.-L.); (C.B.); (D.M.-C.); (Á.Á.); (J.V.E.); (S.C.); (M.D.B.)
- CIBERehd (Centro de Investigaciones en Red Enfermedad Hepática y Digestiva), 28029 Madrid, Spain
| |
Collapse
|
11
|
Wang R, Wang Y, Wu C, Jin G, Zhu F, Yang Y, Wang Y, Zhou G. CD73 blockade alleviates intestinal inflammatory responses by regulating macrophage differentiation in ulcerative colitis. Exp Ther Med 2023; 25:272. [PMID: 37206543 PMCID: PMC10189750 DOI: 10.3892/etm.2023.11972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 03/17/2023] [Indexed: 05/21/2023] Open
Abstract
Ulcerative colitis (UC) is a type of inflammatory bowel disease characterized by excessive and persistent inflammation. Intestinal macrophages play a considerable role in regulating inflammatory immune reactions in the gut mucosa. It has previously been reported that CD73 is related to the pathogenesis of inflammatory or immune-related diseases; however, the roles of CD73 in UC remain unclear. In this study, CD73 expression in the inflamed mucosa of patients with UC was examined using reverse transcription-quantitative PCR (RT-qPCR), western blotting, and immunohistochemistry. Adenosine 5'-(α, β-methylene) diphosphate (APCP) was used to block the expression of CD73. Furthermore, the mRNA levels of proinflammatory mediators associated with macrophages following the blocking of CD73 were examined using RT-qPCR. Finally, the regulatory function of CD73 in intestinal inflammation was assessed by administering APCP in a mouse model of dextran sulfate sodium salt (DSS)-induced colitis. Notably, it was found that CD73 expression was significantly increased in the colonic mucosal tissues of patients with UC. Blockade of CD73 inhibited the expression of pro-inflammatory cytokines but promoted the production of anti-inflammatory cytokines in macrophages, while its promotion of M2 macrophage polarization was also verified. In vivo, CD73 blockade markedly alleviated DSS-induced colitis in mice, as characterized by reduced weight loss, reduction in the incidence of diarrhea, and reduced amount of bloody stool. Mechanistically, it was shown that CD73 regulated macrophage differentiation via the NF-κB and ERK signaling pathways. In conclusion, the findings of the present study indicate that CD73 may have a potential impact on the pathogenesis of UC by modulating the immune response of macrophage differentiation; thus, providing a novel pathway for modulating mucosal inflammation in UC.
Collapse
Affiliation(s)
- Ru Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
- Institute of Clinical Medicine, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Yan Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Chao Wu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
- Institute of Clinical Medicine, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Guiyuan Jin
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Fengqin Zhu
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Yonghong Yang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Yibo Wang
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
| | - Guangxi Zhou
- Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong 272000, P.R. China
- Correspondence to: Professor Guangxi Zhou, Department of Gastroenterology, Affiliated Hospital of Jining Medical University, Jining Medical University, 89 Guhuai Road, Jining, Shandong 272000, P.R. China
| |
Collapse
|
12
|
Chen Y, Wang Y, Fu Y, Yin Y, Xu K. Modulating AHR function offers exciting therapeutic potential in gut immunity and inflammation. Cell Biosci 2023; 13:85. [PMID: 37179416 PMCID: PMC10182712 DOI: 10.1186/s13578-023-01046-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 05/04/2023] [Indexed: 05/15/2023] Open
Abstract
Aryl hydrocarbon receptor (AHR) is a ligand-dependent transcription factor. 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) is a classical exogenous synthetic ligand of AHR that has significant immunotoxic effects. Activation of AHR has beneficial effects on intestinal immune responses, but inactivation or overactivation of AHR can lead to intestinal immune dysregulation and even intestinal diseases. Sustained potent activation of AHR by TCDD results in impairment of the intestinal epithelial barrier. However, currently, AHR research has been more focused on elucidating physiologic AHR function than on dioxin toxicity. The appropriate level of AHR activation plays a role in maintaining gut health and protecting against intestinal inflammation. Therefore, AHR offers a crucial target to modulate intestinal immunity and inflammation. Herein, we summarize our current understanding of the relationship between AHR and intestinal immunity, the ways in which AHR affects intestinal immunity and inflammation, the effects of AHR activity on intestinal immunity and inflammation, and the effect of dietary habits on intestinal health through AHR. Finally, we discuss the therapeutic role of AHR in maintaining gut homeostasis and relieving inflammation.
Collapse
Affiliation(s)
- Yue Chen
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Yadong Wang
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
| | - Yawei Fu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China
- College of Animal Science and Technology, Henan Agricultural University, Zhengzhou, 450000, China
| | - Kang Xu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, 410125, China.
| |
Collapse
|
13
|
Shao B, Ren SH, Wang ZB, Wang HD, Zhang JY, Qin H, Zhu YL, Sun CL, Xu YN, Li X, Wang H. CD73 mediated host purinergic metabolism in intestine contributes to the therapeutic efficacy of a novel mesenchymal-like endometrial regenerative cells against experimental colitis. Front Immunol 2023; 14:1155090. [PMID: 37180168 PMCID: PMC10167049 DOI: 10.3389/fimmu.2023.1155090] [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: 01/31/2023] [Accepted: 04/14/2023] [Indexed: 05/15/2023] Open
Abstract
Background The disruption of intestinal barrier functions and the dysregulation of mucosal immune responses, mediated by aberrant purinergic metabolism, are involved in the pathogenesis of inflammatory bowel diseases (IBD). A novel mesenchymal-like endometrial regenerative cells (ERCs) has demonstrated a significant therapeutic effect on colitis. As a phenotypic marker of ERCs, CD73 has been largely neglected for its immunosuppressive function in regulating purinergic metabolism. Here, we have investigated whether CD73 expression on ERCs is a potential molecular exerting its therapeutic effect against colitis. Methods ERCs either unmodified or with CD73 knockout (CD73-/-ERCs), were intraperitoneally administered to dextran sulfate sodium (DSS)-induced colitis mice. Histopathological analysis, colon barrier function, the proportion of T cells, and maturation of dendritic cells (DCs) were investigated. The immunomodulatory effect of CD73-expressing ERCs was evaluated by co-culture with bone marrow-derived DCs under LPS stimulation. FACS determined DCs maturation. The function of DCs was detected by ELISA and CD4+ cell proliferation assays. Furthermore, the role of the STAT3 pathway in CD73-expressing ERCs-induced DC inhibition was also elucidated. Results Compared with untreated and CD73-/-ERCs-treated groups, CD73-expressing ERCs effectively attenuated body weight loss, bloody stool, shortening of colon length, and pathological damage characterized by epithelial hyperplasia, goblet cell depletion, the focal loss of crypts and ulceration, and the infiltration of inflammatory cells. Knockout of CD73 impaired ERCs-mediated colon protection. Surprisingly, CD73-expressing ERCs significantly decreased the populations of Th1 and Th17 cells but increased the proportions of Tregs in mouse mesenteric lymph nodes. Furthermore, CD73-expressing ERCs markedly reduced the levels of pro-inflammatory cytokines (IL-6, IL-1β, TNF-α) and increased anti-inflammatory factors (IL-10) levels in the colon. CD73-expressing ERCs inhibited the antigen presentation and stimulatory function of DCs associated with the STAT-3 pathway, which exerted a potent therapeutic effect against colitis. Conclusions The knockout of CD73 dramatically abrogates the therapeutic ability of ERCs for intestinal barrier dysfunctions and the dysregulation of mucosal immune responses. This study highlights the significance of CD73 mediates purinergic metabolism contributing to the therapeutic effects of human ERCs against colitis in mice.
Collapse
Affiliation(s)
- Bo Shao
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Shao-hua Ren
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhao-bo Wang
- School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Hong-da Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing-yi Zhang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hong Qin
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang-lin Zhu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Cheng-lu Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yi-ni Xu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiang Li
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Hao Wang
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin General Surgery Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
14
|
Kong L, Pokatayev V, Lefkovith A, Carter GT, Creasey EA, Krishna C, Subramanian S, Kochar B, Ashenberg O, Lau H, Ananthakrishnan AN, Graham DB, Deguine J, Xavier RJ. The landscape of immune dysregulation in Crohn's disease revealed through single-cell transcriptomic profiling in the ileum and colon. Immunity 2023; 56:444-458.e5. [PMID: 36720220 PMCID: PMC9957882 DOI: 10.1016/j.immuni.2023.01.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 57.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 11/14/2022] [Accepted: 01/06/2023] [Indexed: 01/31/2023]
Abstract
Crohn's disease (CD) is a chronic gastrointestinal disease that is increasing in prevalence worldwide. CD is multifactorial, involving the complex interplay of genetic, immune, and environmental factors, necessitating a system-level understanding of its etiology. To characterize cell-type-specific transcriptional heterogeneity in active CD, we profiled 720,633 cells from the terminal ileum and colon of 71 donors with varying inflammation status. Our integrated datasets revealed organ- and compartment-specific responses to acute and chronic inflammation; most immune changes were in cell composition, whereas transcriptional changes dominated among epithelial and stromal cells. These changes correlated with endoscopic inflammation, but small and large intestines exhibited distinct responses, which were particularly apparent when focusing on IBD risk genes. Finally, we mapped markers of disease-associated myofibroblast activation and identified CHMP1A, TBX3, and RNF168 as regulators of fibrotic complications. Altogether, our results provide a roadmap for understanding cell-type- and organ-specific differences in CD and potential directions for therapeutic development.
Collapse
Affiliation(s)
- Lingjia Kong
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Vladislav Pokatayev
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Ariel Lefkovith
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Grace T Carter
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Elizabeth A Creasey
- Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Chirag Krishna
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Sathish Subramanian
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Bharati Kochar
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Orr Ashenberg
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Helena Lau
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ashwin N Ananthakrishnan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Daniel B Graham
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jacques Deguine
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Ramnik J Xavier
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Center for Computational and Integrative Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
15
|
Zysset-Burri DC, Morandi S, Herzog EL, Berger LE, Zinkernagel MS. The role of the gut microbiome in eye diseases. Prog Retin Eye Res 2023; 92:101117. [PMID: 36075807 DOI: 10.1016/j.preteyeres.2022.101117] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/19/2022] [Accepted: 08/22/2022] [Indexed: 02/01/2023]
Abstract
The gut microbiome is a complex ecosystem of microorganisms and their genetic entities colonizing the gastrointestinal tract. When in balanced composition, the gut microbiome is in symbiotic interaction with its host and maintains intestinal homeostasis. It is involved in essential functions such as nutrient metabolism, inhibition of pathogens and regulation of immune function. Through translocation of microbes and their metabolites along the epithelial barrier, microbial dysbiosis induces systemic inflammation that may lead to tissue destruction and promote the onset of various diseases. Using whole-metagenome shotgun sequencing, several studies have shown that the composition and associated functional capacities of the gut microbiome are associated with age-related macular degeneration, retinal artery occlusion, central serous chorioretinopathy and uveitis. In this review, we provide an overview of the current knowledge about the gut microbiome in eye diseases, with a focus on interactions between the microbiome, specific microbial-derived metabolites and the immune system. We explain how these interactions may be involved in the pathogenesis of age-related macular degeneration, retinal artery occlusion, central serous chorioretinopathy and uveitis and guide the development of new therapeutic approaches by microbiome-altering interventions for these diseases.
Collapse
Affiliation(s)
- Denise C Zysset-Burri
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland.
| | - Sophia Morandi
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland.
| | - Elio L Herzog
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Mittelstrasse 43, CH-3012, Bern, Switzerland.
| | - Lieselotte E Berger
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland.
| | - Martin S Zinkernagel
- Department of Ophthalmology, Inselspital, Bern University Hospital, University of Bern, Freiburgstrasse 15, CH-3010, Bern, Switzerland; Department for BioMedical Research, University of Bern, Murtenstrasse 24, CH-3008, Bern, Switzerland.
| |
Collapse
|
16
|
Franco R, Lillo A, Navarro G, Reyes-Resina I. The adenosine A 2A receptor is a therapeutic target in neurological, heart and oncogenic diseases. Expert Opin Ther Targets 2022; 26:791-800. [DOI: 10.1080/14728222.2022.2136570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Affiliation(s)
- Rafael Franco
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neurobiology laboratory, Department of Biochemistry and Molecular Biomedicine, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
| | - Alejandro Lillo
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neuropharmacology laboratory, Department of Biochemistry and Physiology. School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- Molecular Neuropharmacology laboratory, Department of Biochemistry and Physiology. School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| | - Irene Reyes-Resina
- CiberNed, Network Center for Neurodegenerative diseases, National Spanish Health Institute Carlos III, Madrid, Spain
- School of Chemistry, Universitat de Barcelona, Barcelona, Spain
- Molecular Neuropharmacology laboratory, Department of Biochemistry and Physiology. School of Pharmacy and Food Science, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
17
|
D'Antongiovanni V, Pellegrini C, Benvenuti L, Fornai M, Di Salvo C, Natale G, Ryskalin L, Bertani L, Lucarini E, Di Cesare Mannelli L, Ghelardini C, Nemeth ZH, Haskó G, Antonioli L. Anti-inflammatory Effects of Novel P2X4 Receptor Antagonists, NC-2600 and NP-1815-PX, in a Murine Model of Colitis. Inflammation 2022; 45:1829-1847. [PMID: 35338432 PMCID: PMC9197920 DOI: 10.1007/s10753-022-01663-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/14/2022] [Accepted: 03/14/2022] [Indexed: 11/21/2022]
Abstract
The pharmacological blockade of P2X4 receptors has shown potential benefits in the management of several immune/inflammatory diseases. However, data regarding the involvement of P2X4 receptors in the pathophysiological mechanisms of action in intestinal inflammation are not well defined. We aimed to evaluate the anti-inflammatory effects of two novel and selective P2X4 receptor antagonists, NC-2600 and NP-1815-PX, and characterize the molecular mechanisms of their action in a murine model of 2,4-dinitrobenzene sulfonic acid (DNBS)-induced colitis. These two drugs and dexamethasone (DEX) were administered orally for 6 days, immediately after the manifestation of DNBS. The body weight decrease, resulting from colitis, was attenuated by NC-2600 and NP-1815-PX, but not DEX. However, all three drugs attenuated the increase in spleen weight and ameliorated macroscopic and microscopic colonic tissue damage. Furthermore, all three compounds decreased tissue IL-1β levels and caspase-1 expression and activity. Colonic tissue increase of tumor necrosis factor was downregulated by DEX, while both NC-2600 and NP-1815-PX were ineffective. The reduction of occludin associated with colitis was ameliorated by NC-2600 and NP-1815-PX, but not DEX. In THP-1 cells, lipopolysaccharide and ATP upregulated IL-1β release and NLRP3, caspase-1, caspase-5, and caspase-8 activity, but not of caspase-4. These changes were prevented by NC-2600 and NP-1815-PX treatment. For the first time, the above findings show that the selective inhibition of P2X4 receptors represents a viable approach to manage bowel inflammation via the inhibition of NLRP3 inflammasome signaling pathways.
Collapse
Affiliation(s)
| | - Carolina Pellegrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Benvenuti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Matteo Fornai
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy.
- Unit of Pharmacology and Pharmacovigilance, Department of Clinical and Experimental Medicine, University of Pisa Via Roma 55, 56126 Pisa, Italy.
| | - Clelia Di Salvo
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Gianfranco Natale
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Larisa Ryskalin
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Lorenzo Bertani
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Elena Lucarini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Lorenzo Di Cesare Mannelli
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Carla Ghelardini
- Department of Neuroscience, Psychology, Drug Research and Child Health, Neurofarba, Pharmacology and Toxicology Section, University of Florence, Florence, Italy
| | - Zoltan H Nemeth
- Department of Surgery, Morristown Medical Center, Morristown, NJ, 07960, USA
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - György Haskó
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - Luca Antonioli
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
18
|
Dong LW, Ma ZC, Fu J, Huang BL, Liu FJ, Sun D, Lan C. Upregulated adenosine 2A receptor accelerates post-infectious irritable bowel syndrome by promoting CD4+ T cells’ T helper 17 polarization. World J Gastroenterol 2022; 28:2955-2967. [PMID: 35978875 PMCID: PMC9280732 DOI: 10.3748/wjg.v28.i25.2955] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/26/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Post-infectious irritable bowel syndrome (PI-IBS) is generally regarded as a functional disease. Several recent studies have reported the involvement of low-grade inflammation and immunological dysfunction in PI-IBS. T helper 17 (Th17) polarization occurs in IBS. Adenosine and its receptors participate in intestinal inflammation and immune regulation.
AIM To investigate the role of Th17 polarization of CD4+ T cells regulated by adenosine 2A receptor (A2AR) in PI-IBS.
METHODS A PI-IBS model was established by infecting mice with Trichinella spiralis. The intestinal A2AR and CD4+ T lymphocytes were detected by immunohistochemistry, and the inflammatory cytokines were detected by enzyme-linked immunoassay. CD4+ T lymphocytes present in the animal’s spleen were separated and cultured with or without A2AR agonist and antagonist. Western blotting and real-time quantitative polymerase chain reaction were performed to determine the effect of A2AR on the cells and intestinal tissue. Cytokine production was determined. The protein and mRNA levels of A2AR associated signaling pathway molecules were also evaluated. Furthermore, A2AR agonist and antagonist were injected into the mouse model and the clinical features were observed.
RESULTS The PI-IBS mouse model showed increased expression of ATP and A2AR (P < 0.05), and inhibition of A2AR improved the clinical features in PI-IBS, including the abdominal withdrawal reflex and colon transportation test (P < 0.05). The number of intestinal CD4+ T cells and interleukin-17 (IL-17) protein levels increased during PI-IBS, which was reversed by administration of the A2AR antagonist (P < 0.05). CD4+ T cells expressed A2AR and produced IL-17 in vitro, which was regulated by the A2AR agonist and antagonist. The A2AR antagonist increased the production of IL-17 by CD4+ T cells via the Janus kinase-signal transducer and activator of transcription-receptor-related orphan receptor γ signaling pathway.
CONCLUSION The results of the present study suggested that the upregulation of A2AR increases PI-IBS by promoting the Th17 polarization of CD4+ T cells.
Collapse
Affiliation(s)
- Li-Wei Dong
- Department of Gastroenterology, Hainan General Hospital, Affiliated Hainan Hospital, Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Zhi-Chao Ma
- Department of Gastroenterology, Hainan General Hospital, Affiliated Hainan Hospital, Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Jiao Fu
- Department of Gastroenterology, Hainan General Hospital, Affiliated Hainan Hospital, Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Bai-Li Huang
- Department of Gastroenterology, Hainan General Hospital, Affiliated Hainan Hospital, Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Fu-Jin Liu
- Department of Gastroenterology, Hainan General Hospital, Affiliated Hainan Hospital, Hainan Medical University, Haikou 570311, Hainan Province, China
| | - Deming Sun
- Doheny Eye Institute, Department of Ophthalmology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90033, United States
| | - Cheng Lan
- Department of Gastroenterology, Hainan General Hospital, Affiliated Hainan Hospital, Hainan Medical University, Haikou 570311, Hainan Province, China
| |
Collapse
|
19
|
Grubišić V, Bali V, Fried DE, Eltzschig HK, Robson SC, Mazei-Robison MS, Gulbransen BD. Enteric glial adenosine 2B receptor signaling mediates persistent epithelial barrier dysfunction following acute DSS colitis. Mucosal Immunol 2022; 15:964-976. [PMID: 35869148 PMCID: PMC9385475 DOI: 10.1038/s41385-022-00550-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 06/15/2022] [Accepted: 07/01/2022] [Indexed: 02/04/2023]
Abstract
Intestinal epithelial barrier function is compromised in inflammatory bowel disease and barrier dysfunction contributes to disease progression. Extracellular nucleotides/nucleosides generated in gut inflammation may regulate barrier function through actions on diverse cell types. Enteric glia modulate extracellular purinergic signaling and exert pathophysiological effects on mucosal permeability. These glia may regulate inflammation with paracrine responses, theoretically mediated via adenosine 2B receptor (A2BR) signaling. As the cell-specific roles of A2BRs in models of colitis and barrier dysfunction are unclear, we studied glial A2BRs in acute dextran sodium sulfate (DSS) colitis. We performed and validated conditional ablation of glial A2BRs in Sox10CreERT2+/-;Adora2bf/f mice. Overt intestinal disease activity indices in DSS-colitis were comparable between Sox10CreERT2+/-;Adora2bf/f mice and littermate controls. However, ablating glial A2BRs protected against barrier dysfunction following acute DSS-colitis. These benefits were associated with the normalization of tight junction protein expression and localization including claudin-1, claudin-8, and occludin. Glial A2BR signaling increased levels of proinflammatory mediators in the colon and cell-intrinsic regulation of genes including Csf3, Cxcl1, Cxcl10, and Il6. Our studies show that glial A2BR signaling exacerbates immune responses during DSS-colitis and that this adenosinergic cell-specific mechanism contributes to persistent gut epithelial barrier dysfunction.
Collapse
Affiliation(s)
- Vladimir Grubišić
- Department of Physiology and Neuroscience program, Michigan State University, East Lansing, MI, 48824, USA
- Department of Biomedical Sciences and Center for Biomedical Innovation, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, 11568, USA
| | - Vedrana Bali
- Department of Physiology and Neuroscience program, Michigan State University, East Lansing, MI, 48824, USA
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI, 48824, USA
| | - David E Fried
- Department of Physiology and Neuroscience program, Michigan State University, East Lansing, MI, 48824, USA
| | - Holger K Eltzschig
- McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Simon C Robson
- Division of Gastroenterology, Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, 02215, USA
| | - Michelle S Mazei-Robison
- Department of Physiology and Neuroscience program, Michigan State University, East Lansing, MI, 48824, USA
| | - Brian D Gulbransen
- Department of Physiology and Neuroscience program, Michigan State University, East Lansing, MI, 48824, USA.
| |
Collapse
|
20
|
Kolbe K, Wittner M, Hartjen P, Hüfner AD, Degen O, Ackermann C, Cords L, Stellbrink HJ, Haag F, Schulze zur Wiesch J. Inversed Ratio of CD39/CD73 Expression on γδ T Cells in HIV Versus Healthy Controls Correlates With Immune Activation and Disease Progression. Front Immunol 2022; 13:867167. [PMID: 35529864 PMCID: PMC9074873 DOI: 10.3389/fimmu.2022.867167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/15/2022] [Indexed: 12/16/2022] Open
Abstract
Background γδ T cells are unconventional T cells that have been demonstrated to be crucial for the pathogenesis and potentially for the cure of HIV-1 infection. The ectonucleotidase CD39 is part of the purinergic pathway that regulates immune responses by degradation of pro-inflammatory ATP in concert with CD73. Few studies on the expression of the ectoenzymes CD73 and CD39 on human γδ T cells in HIV have been performed to date. Methods PBMC of n=86 HIV-1-infected patients were compared to PBMC of n=26 healthy individuals using 16-color flow cytometry determining the surface expression of CD39 and CD73 on Vδ1 and Vδ2 T cells in association with differentiation (CD45RA, CD28, CD27), activation and exhaustion (TIGIT, PD-1, CD38, and HLA-DR), and assessing the intracellular production of pro- and anti-inflammatory cytokines (IL-2, TGF-ß, TNF-α, Granzyme B, IL-10, IFN-γ) after in vitro stimulation with PMA/ionomycin. Results CD39 and CD73 expression on γδ T cells were inversed in HIV infection which correlated with HIV disease progression and immune activation. CD39, but not CD73 expression on γδ T cells of ART-treated patients returned to levels comparable with those of healthy individuals. Only a small subset (<1%) of γδ T cells co-expressed CD39 and CD73 in healthy or HIV-infected individuals. There were significantly more exhausted and terminally differentiated CD39+ Vδ1 T cells regardless of the disease status. Functionally, IL-10 was only detectable in CD39+ γδ T cells after in vitro stimulation in all groups studied. Viremic HIV-infected patients showed the highest levels of IL-10 production. The highest percentage of IL-10+ cells was found in the small CD39/CD73 co-expressing γδ T-cell population, both in healthy and HIV-infected individuals. Also, CD39+ Vδ2 T cells produced IL-10 more frequently than their CD39+ Vδ1 counterparts in all individuals regardless of the HIV status. Conclusions Our results point towards a potential immunomodulatory role of CD39+ and CD73+ γδ T cells in the pathogenesis of chronic HIV infection that needs further investigation.
Collapse
Affiliation(s)
- Katharina Kolbe
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
| | - Melanie Wittner
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
- *Correspondence: Melanie Wittner,
| | - Philip Hartjen
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anja-Dorothee Hüfner
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Infectious Diseases Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olaf Degen
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Infectious Diseases Clinic, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christin Ackermann
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leon Cords
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schulze zur Wiesch
- First Department of Medicine, Section Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
| |
Collapse
|
21
|
Faraoni EY, Ju C, Robson SC, Eltzschig HK, Bailey-Lundberg JM. Purinergic and Adenosinergic Signaling in Pancreatobiliary Diseases. Front Physiol 2022; 13:849258. [PMID: 35360246 PMCID: PMC8964054 DOI: 10.3389/fphys.2022.849258] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022] Open
Abstract
Adenosine 5'-triphosphate (ATP), other nucleotides, and the nucleoside analogue, adenosine, all have the capacity to modulate cellular signaling pathways. The cellular processes linked to extracellular purinergic signaling are crucial in the initiation, evolution, and resolution of inflammation. Injured or dying cells in the pancreatobiliary tract secrete or release ATP, which results in sustained purinergic signaling mediated through ATP type-2 purinergic receptors (P2R). This process can result in chronic inflammation, fibrosis, and tumor development. In contrast, signaling via the extracellular nucleoside derivative adenosine via type-1 purinergic receptors (P1R) is largely anti-inflammatory, promoting healing. Failure to resolve inflammation, as in the context of primary sclerosing cholangitis or chronic pancreatitis, is a risk factor for parenchymal and end-organ scarring with the associated risk of pancreatobiliary malignancies. Emerging immunotherapeutic strategies suggest that targeting purinergic and adenosinergic signaling can impact the growth and invasive properties of cancer cells, potentiate anti-tumor immunity, and also block angiogenesis. In this review, we dissect out implications of disordered purinergic responses in scar formation, end-organ injury, and in tumor development. We conclude by addressing promising opportunities for modulation of purinergic/adenosinergic signaling in the prevention and treatment of pancreatobiliary diseases, inclusive of cancer.
Collapse
Affiliation(s)
- Erika Y. Faraoni
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Cynthia Ju
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Simon C. Robson
- Departments of Internal Medicine and Anesthesiology, Center for Inflammation Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Holger K. Eltzschig
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Jennifer M. Bailey-Lundberg
- Department of Anesthesiology, Center for Perioperative Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, United States
| |
Collapse
|
22
|
Juzenas S, Hübenthal M, Lindqvist CM, Kruse R, Steiert TA, Degenhardt F, Schulte D, Nikolaus S, Zeissig S, Bergemalm D, Almer S, Hjortswang H, Bresso F, Strüning N, Kupcinskas J, Keller A, Lieb W, Rosenstiel P, Schreiber S, D’Amato M, Halfvarson J, Hemmrich-Stanisak G, Franke A. Detailed Transcriptional Landscape of Peripheral Blood Points to Increased Neutrophil Activation in Treatment-Naïve Inflammatory Bowel Disease. J Crohns Colitis 2022; 16:1097-1109. [PMID: 35022690 PMCID: PMC9351981 DOI: 10.1093/ecco-jcc/jjac003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 12/13/2021] [Accepted: 01/08/2022] [Indexed: 12/24/2022]
Abstract
BACKGROUND AND AIMS Inflammatory bowel disease [IBD] is a chronic relapsing disorder of the gastrointestinal tract, which generally manifests as Crohn's disease [CD] or ulcerative colitis [UC]. These subtypes are heterogeneous in terms of disease location and histological features, while sharing common clinical presentation, genetic associations and, thus, common immune regulatory pathways. METHODS Using miRNA and mRNA coupled transcriptome profiling and systems biology approaches, we report a comprehensive analysis of blood transcriptomes from treatment-naïve [n = 110] and treatment-exposed [n = 177] IBD patients as well as symptomatic [n = 65] and healthy controls [n = 95]. RESULTS Broadly, the peripheral blood transcriptomes of CD and UC patients were similar. However, there was an extensive gene deregulation in the blood of IBD patients, while only a slight deregulation in symptomatic controls, when compared with healthy controls. The deregulated mRNAs and miRNAs are mainly involved in the innate immunity and are especially enriched in neutrophil activation-related pathways. Oxidative phosphorylation and neutrophil activation-related modules were found to be differentially co-expressed among treatment-naïve IBD as compared to healthy controls. In the deregulated neutrophil activation-related co-expression module, IL1B was identified as the central gene. Levels of co-expression among IL1B and chemosensing receptor [CXCR1/2 and FPR1/2] genes were reduced in the blood of IBD patients when compared with healthy controls. CONCLUSIONS Immune dysregulation seen in peripheral blood transcriptomes of treatment-naïve IBD patients is mainly driven by neutrophil activation.
Collapse
Affiliation(s)
- Simonas Juzenas
- Corresponding author: Simonas Juzenas, PhD, Institute of Clinical Molecular Biology (IKMB), Christian-Albrechts-University of Kiel (CAU), Rosalind-Franklin-Str. 12, D-24105 Kiel, Germany.
| | - Matthias Hübenthal
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany,Department of Dermatology, Quincke Research Center, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Carl Mårten Lindqvist
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany,School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Robert Kruse
- Department of Clinical Research Laboratory, Faculty of Medicine and Health, Örebro University, Örebro, Sweden,iRiSC – Inflammatory Response and Infection Susceptibility Centre, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Tim Alexander Steiert
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Frauke Degenhardt
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Dominik Schulte
- Division of Endocrinology, Diabetes and Clinical Nutrition, Department of Medicine I, University Hospital of Schleswig-Holstein, Kiel, Germany,Institute of Diabetes and Clinical Metabolic Research, Kiel University, Kiel, Germany
| | - Susanna Nikolaus
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sebastian Zeissig
- Medical Department 1, University Hospital Dresden, Technische Universität Dresden (TU Dresden), Dresden, Germany,Center for Regenerative Therapies Dresden (CRTD), Technische Universität (TU) Dresden, Dresden, Germany
| | - Daniel Bergemalm
- School of Medical Sciences, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Sven Almer
- Department of Medicine, Karolinska Institutet, Solna, and Division of Gastroenterology, Karolinska University Hospital, Stockholm, Sweden
| | - Henrik Hjortswang
- Department of Gastroenterology and Hepatology, Linköping University, Linköping, and Department of Health, Medicine, and Caring Sciences, Linköping University, Linköping, Sweden
| | - Francesca Bresso
- Department of Medicine, Karolinska Institutet, Solna, and Division of Gastroenterology, Karolinska University Hospital, Stockholm, Sweden
| | | | - Nina Strüning
- Department of Internal Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Juozas Kupcinskas
- Department of Gastroenterology and Institute for Digestive Research, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany,Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Wolfgang Lieb
- Institute of Epidemiology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany,Department of Internal Medicine I, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Mauro D’Amato
- Unit of Clinical Epidemiology, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden,Gastrointestinal Genetics Lab, CIC bioGUNE – BRTA, Derio, Spain,Ikerbasque, Basque Foundation for Science, Bilbao, Spain
| | | | | | | |
Collapse
|
23
|
Cheng N, Zhang L, Liu L. Understanding the Role of Purinergic P2X7 Receptors in the Gastrointestinal System: A Systematic Review. Front Pharmacol 2021; 12:786579. [PMID: 34987401 PMCID: PMC8721002 DOI: 10.3389/fphar.2021.786579] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/17/2021] [Indexed: 11/24/2022] Open
Abstract
Background: The role of purinergic P2X7 receptor (P2X7R) is of interest due to its involvement in inflammation and mediating immune cell responses. P2X7R is particularly implicated in the development of inflammatory bowel disease (IBD). However, the extent of the actions of P2X7R in the gastrointestinal (GI) system under physiological and pathophysiological conditions remains to be elucidated. This systematic review aimed to identify, summarize and evaluate the evidence for a critical role of P2X7R in the GI system. Methods: We searched PubMed, Embase and Scopus with search terms pertained to P2X7R in the GI system in disease or physiological state, including “P2X7 or P2X7 receptor or purinergic signaling” in combination with any of the terms “intestine or colon or gut or gastrointestinal,” “pathology or inflammation or disease or disorder,” and “physiology or expression.” Titles and abstracts were screened for potentially eligible full texts, and animal and human studies published in English were included in this study. Data were extracted from papers meeting inclusion criteria. Meta-analysis was not feasible given the study diversity. Results: There were 48 papers included in this review. We identified 14 experimental colitis models, three sepsis models and one ischemia-reperfusion injury model. Among them, 11 studies examined P2X7R in GI infections, six studies on immune cell regulation, four studies on GI inflammation, two studies on GI malignancies, three studies involving intestinal injury due to various causes, two studies on ATP-activated P2X7R in the GI system and two studies on metabolic regulation. Conclusion: Evidence supports P2X7R mediating inflammation and immune cell responses in GI inflammation, infections and injury due to IBD and other challenges to the intestinal wall. P2X7R inhibition by gene knockout or by application of P2X7R antagonists can reduce tissue damage by suppressing inflammation. P2X7R is also implicated in GI malignancies and glucose and lipid homeostasis. P2X7R blockade, however, did not always lead to beneficial outcomes in the various pathological models of study.
Collapse
Affiliation(s)
- Nathalie Cheng
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Li Zhang
- School of Biotechnology and Biomolecular Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Lu Liu
- School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
- *Correspondence: Lu Liu,
| |
Collapse
|
24
|
Zarei M, Sahebi Vaighan N, Ziai SA. Purinergic receptor ligands: the cytokine storm attenuators, potential therapeutic agents for the treatment of COVID-19. Immunopharmacol Immunotoxicol 2021; 43:633-643. [PMID: 34647511 PMCID: PMC8544669 DOI: 10.1080/08923973.2021.1988102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 09/25/2021] [Indexed: 12/13/2022]
Abstract
The coronavirus disease-19 (COVID-19), at first, was reported in Wuhan, China, and then rapidly became pandemic throughout the world. Cytokine storm syndrome (CSS) in COVID-19 patients is associated with high levels of cytokines and chemokines that cause multiple organ failure, systemic inflammation, and hemodynamic instabilities. Acute respiratory distress syndrome (ARDS), a common complication of COVID-19, is a consequence of cytokine storm. In this regard, several drugs have been being investigated to suppress this inflammatory condition. Purinergic signaling receptors comprising of P1 adenosine and P2 purinoceptors play a critical role in inflammation. Therefore, activation or inhibition of some subtypes of these kinds of receptors is most likely to be beneficial to attenuate cytokine storm. This article summarizes suggested therapeutic drugs with potential anti-inflammatory effects through purinergic receptors.
Collapse
Affiliation(s)
- Malek Zarei
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Navideh Sahebi Vaighan
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed Ali Ziai
- Department of Pharmacology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
25
|
Wang Z, Chen WD, Wang YD. Nuclear receptors: a bridge linking the gut microbiome and the host. Mol Med 2021; 27:144. [PMID: 34740314 PMCID: PMC8570027 DOI: 10.1186/s10020-021-00407-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 10/26/2021] [Indexed: 12/12/2022] Open
Abstract
Background The gut microbiome is the totality of microorganisms, bacteria, viruses, protozoa, and fungi within the gastrointestinal tract. The gut microbiome plays key roles in various physiological and pathological processes through regulating varieties of metabolic factors such as short-chain fatty acids, bile acids and amino acids. Nuclear receptors, as metabolic mediators, act as a series of intermediates between the microbiome and the host and help the microbiome regulate diverse processes in the host. Recently, nuclear receptors such as farnesoid X receptor, peroxisome proliferator-activated receptors, aryl hydrocarbon receptor and vitamin D receptor have been identified as key regulators of the microbiome-host crosstalk. These nuclear receptors regulate metabolic processes, immune activity, autophagy, non-alcoholic and alcoholic fatty liver disease, inflammatory bowel disease, cancer, obesity, and type-2 diabetes. Conclusion In this review, we have summarized the functions of the nuclear receptors in the gut microbiome-host axis in different physiological and pathological conditions, indicating that the nuclear receptors may be the good targets for treatment of different diseases through the crosstalk with the gut microbiome.
Collapse
Affiliation(s)
- Zixuan Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People's Republic of China
| | - Wei-Dong Chen
- Key Laboratory of Molecular Pathology, Key Laboratory of Receptors-Mediated Gene Regulation and Drug Discovery, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot, Inner Mongolia, People's Republic of China. .,School of Medicine, Key Laboratory of Receptors-Mediated Gene Regulation, The People' Hospital of Hebi, Henan University, Henan, People's Republic of China.
| | - Yan-Dong Wang
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, People's Republic of China.
| |
Collapse
|
26
|
Exosomes as a New Delivery Vehicle in Inflammatory Bowel Disease. Pharmaceutics 2021; 13:pharmaceutics13101644. [PMID: 34683937 PMCID: PMC8539337 DOI: 10.3390/pharmaceutics13101644] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a type of chronic relapsing inflammatory disease. The pathogenesis of IBD is still unclear, which may involve environmental factors, genetic factors, intestinal microbiota disorder, and abnormal immune responses. Exosomes (30–150 nm) are found in various body fluids, including blood, saliva, urine, and cerebrospinal fluid. Exosomes mediate intercellular communication and regulate cell biological activity by carrying non-coding RNAs, proteins, and lipids. There is evidence that exosomes are involved in the pathogenesis of IBD. In view of the important roles of exosomes in the pathogenesis of IBD, this work systematically reviews the latest research progress of exosomes in IBD, especially the roles of exosomes as non-coding RNA delivery systems in the pathogenesis of IBD, including a disordered immune response, barrier function, and intestinal microbiota. The review will help to clarify the pathogenesis of IBD and explore new diagnostic markers and therapeutic targets for patients with IBD.
Collapse
|
27
|
Schneider E, Winzer R, Rissiek A, Ricklefs I, Meyer-Schwesinger C, Ricklefs FL, Bauche A, Behrends J, Reimer R, Brenna S, Wasielewski H, Lauten M, Rissiek B, Puig B, Cortesi F, Magnus T, Fliegert R, Müller CE, Gagliani N, Tolosa E. CD73-mediated adenosine production by CD8 T cell-derived extracellular vesicles constitutes an intrinsic mechanism of immune suppression. Nat Commun 2021; 12:5911. [PMID: 34625545 PMCID: PMC8501027 DOI: 10.1038/s41467-021-26134-w] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 09/17/2021] [Indexed: 12/15/2022] Open
Abstract
Immune cells at sites of inflammation are continuously activated by local antigens and cytokines, and regulatory mechanisms must be enacted to control inflammation. The stepwise hydrolysis of extracellular ATP by ectonucleotidases CD39 and CD73 generates adenosine, a potent immune suppressor. Here we report that human effector CD8 T cells contribute to adenosine production by releasing CD73-containing extracellular vesicles upon activation. These extracellular vesicles have AMPase activity, and the resulting adenosine mediates immune suppression independently of regulatory T cells. In addition, we show that extracellular vesicles isolated from the synovial fluid of patients with juvenile idiopathic arthritis contribute to T cell suppression in a CD73-dependent manner. Our results suggest that the generation of adenosine upon T cell activation is an intrinsic mechanism of human effector T cells that complements regulatory T cell-mediated suppression in the inflamed tissue. Finally, our data underscore the role of immune cell-derived extracellular vesicles in the control of immune responses.
Collapse
Affiliation(s)
- Enja Schneider
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Riekje Winzer
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| | - Anne Rissiek
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Isabell Ricklefs
- Division of Pediatric Pneumology & Allergology, University Medical Center Schleswig-Holstein, 23538, Lübeck, Germany.,Airway Research Center North, Member of the German Center for Lung Research, Lübeck, Germany
| | - Catherine Meyer-Schwesinger
- Department of Cellular and Integrative Physiology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Andreas Bauche
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Jochen Behrends
- Core Facility Fluorescence Cytometry, Research Center Borstel, 23845, Borstel, Germany
| | - Rudolph Reimer
- Technology Platform Microscopy and Image Analysis, Heinrich Pette Institute/Leibniz Institute for Experimental Virology, 20251, Hamburg, Germany
| | - Santra Brenna
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Hauke Wasielewski
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Melchior Lauten
- Department of Pediatrics and Adolescent Medicine, University of Lübeck, 23538, Lübeck, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Berta Puig
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Filippo Cortesi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Ralf Fliegert
- Department of Biochemistry and Molecular Cell Biology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany
| | - Christa E Müller
- Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, 53121, Bonn, Germany
| | - Nicola Gagliani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.,Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Eva Tolosa
- Department of Immunology, University Medical Center Hamburg-Eppendorf, 20246, Hamburg, Germany.
| |
Collapse
|
28
|
Murtaza A, Afzal S, Zaman G, Saeed A, Pelletier J, Sévigny J, Iqbal J, Hassan A. Divergent synthesis and elaboration of structure activity relationship for quinoline derivatives as highly selective NTPDase inhibitor. Bioorg Chem 2021; 115:105240. [PMID: 34416508 DOI: 10.1016/j.bioorg.2021.105240] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 06/25/2021] [Accepted: 08/02/2021] [Indexed: 01/11/2023]
Abstract
Quinoline derivatives have interesting biological profile. In continuation for the comprehensive evaluations of substituted quinoline derivatives against human nucleoside triphosphate diphosphohydrolases (h-NTPDases) a series of substituted quinoline derivatives (2a-g, 3a-f, 4, 5a-c, 6) was synthesized. The inhibitory activities of the synthesized compounds were evaluated against four isoenzymes of human nucleoside triphosphate diphosphohydrolases (h-NTPDases). These quinoline derivatives had IC50 (µM) values in the range of 0.20-1.75, 0.77-2.20, 0.36-5.50 and 0.90-1.82 for NTPDase1, NTPDase2, NTPDase3 and NTPDase8, respectively. The derivative 3f was the most active compound against NTPDase1 (IC50, 0.20 ± 0.02 µM) that also possessed selectivity towards NTPDase1. Similarly, derivative 3b (IC50, 0.77 ± 0.06), 2h (IC50, 0.36 ± 0.01) and 2c (IC50, 0.90 ± 0.08) displayed excellent activity corresponding to NTPDase2, NTPDase3 and NTPdase8. The compound 5c emerged as a selective inhibitor of NTPDase8. The most active compounds were then investigated to determine their mode of inhibition and finally binding interactions of the active compounds were analyzed through molecular docking studies. The obtained results strongly support the quinoline scaffold's potential as potent and selective NTPDase inhibitor.
Collapse
Affiliation(s)
- Amna Murtaza
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Saira Afzal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Gohar Zaman
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan
| | - Julie Pelletier
- Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada
| | - Jean Sévigny
- Centre de recherche du CHU de Québec - Université Laval, Québec City, QC, Canada; Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, QC, Canada
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan.
| | - Abbas Hassan
- Department of Chemistry, Quaid-i-Azam University, Islamabad 45320, Pakistan.
| |
Collapse
|
29
|
Pasquini S, Contri C, Borea PA, Vincenzi F, Varani K. Adenosine and Inflammation: Here, There and Everywhere. Int J Mol Sci 2021; 22:7685. [PMID: 34299305 PMCID: PMC8304851 DOI: 10.3390/ijms22147685] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/16/2021] [Accepted: 07/16/2021] [Indexed: 02/06/2023] Open
Abstract
Adenosine is a ubiquitous endogenous modulator with the main function of maintaining cellular and tissue homeostasis in pathological and stress conditions. It exerts its effect through the interaction with four G protein-coupled receptor (GPCR) subtypes referred as A1, A2A, A2B, and A3 adenosine receptors (ARs), each of which has a unique pharmacological profile and tissue distribution. Adenosine is a potent modulator of inflammation, and for this reason the adenosinergic system represents an excellent pharmacological target for the myriad of diseases in which inflammation represents a cause, a pathogenetic mechanism, a consequence, a manifestation, or a protective factor. The omnipresence of ARs in every cell of the immune system as well as in almost all cells in the body represents both an opportunity and an obstacle to the clinical use of AR ligands. This review offers an overview of the cardinal role of adenosine in the modulation of inflammation, showing how the stimulation or blocking of its receptors or agents capable of regulating its extracellular concentration can represent promising therapeutic strategies for the treatment of chronic inflammatory pathologies, neurodegenerative diseases, and cancer.
Collapse
Affiliation(s)
- Silvia Pasquini
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.)
| | - Chiara Contri
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.)
| | | | - Fabrizio Vincenzi
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.)
| | - Katia Varani
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy; (S.P.); (C.C.); (K.V.)
| |
Collapse
|
30
|
Synthesis, In-vitro evaluation and molecular docking studies of oxoindolin phenylhydrazine carboxamides as potent and selective inhibitors of ectonucleoside triphosphate diphosphohydrolase (NTPDase). Bioorg Chem 2021; 112:104957. [PMID: 34020240 DOI: 10.1016/j.bioorg.2021.104957] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/13/2021] [Accepted: 04/28/2021] [Indexed: 12/13/2022]
Abstract
Members of the ectonucleoside triphosphate diphosphohydrolases (NTPDases) constitute the major family of enzymes responsible for the maintenance of extracellular levels of nucleotides and nucleosides by catalyzing the hydrolysis of nucleoside triphosphate (NTP) and nucleoside diphosphates (NDP) to nucleoside monophosphate (NMP). Although, NTPDase inhibitors can act as potential drug candidates for the treatment of various diseases, there is lack of potent as well as selective inhibitors of NTPDases. The current study describes the synthesis of a number of carboxamide derivatives that were tested on recombinant human (h) NTPDases. The most promising inhibitors were 2h (h-NTPDase1, IC50: 0.12 ± 0.03 µM), 2d (h-NTPDase2, IC50: 0.15 ± 0.01 µM) and 2a (h-NTPDase3, IC50: 0.30 ± 0.04 µM; h-NTPDase8, IC50: 0.16 ± 0.02 µM). Four compounds (2e, 2f, 2g and 2h) were associated with the selective inhibition of h-NTPDase1 while 2b was identified as a selective h-NTPDase3 inhibitor. Considering the importance of NTPDase3 in the regulation of insulin release, the NTPDase3 inhibitors were further investigated to elucidate their role in the insulin release. The obtained data suggested that compound 2a was actively participating in regulating the insulin release without producing any effect on NTPDase3 mRNA. Moreover, the most potent inhibitors were docked within the active site of respective enzyme and the observed interactions were in compliance with in vitro results. Hence, these compounds can be used as pharmacological tool to further investigate the role of NTPDase3 coupled to insulin release.
Collapse
|
31
|
Hyperinflammation and airway surface liquid dehydration in cystic fibrosis: purinergic system as therapeutic target. Inflamm Res 2021; 70:633-649. [PMID: 33904934 DOI: 10.1007/s00011-021-01464-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/06/2021] [Accepted: 04/19/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE AND DESIGN The exacerbate inflammatory response contributes to the progressive loss of lung function in cystic fibrosis (CF), a genetic disease that affects the osmotic balance of mucus and mucociliary clearance, resulting in a microenvironment that favors infection and inflammation. The purinergic system, an extracellular signaling pathway characterized by nucleotides, enzymes and receptors, may have a protective role in the disease, through its action in airway surface liquid (ASL) and anti-inflammatory response. MATERIALS AND METHODS To make up this review, studies covering topics of CF, inflammation, ASL and purinergic system were selected from the main medical databases, such as Pubmed and ScienceDirect. CONCLUSION We propose several ways to modulate the purinergic system as a potential therapy for CF, like inhibition of P2X7, activation of P2Y2, A2A and A2B receptors and blocking of adenosine deaminase. Among them, we postulate that the most suitable strategy is to block the action of adenosine deaminase, which culminates in the increase of Ado levels that presents anti-inflammatory actions and improves mucociliary clearance. Furthermore, it is possible to maintain the physiological levels of ATP to control the hydration of ASL. These therapies could correct the main mechanisms that contribute to the progression of CF.
Collapse
|
32
|
Franco R, Rivas-Santisteban R, Lillo J, Camps J, Navarro G, Reyes-Resina I. 5-Hydroxytryptamine, Glutamate, and ATP: Much More Than Neurotransmitters. Front Cell Dev Biol 2021; 9:667815. [PMID: 33937270 PMCID: PMC8083958 DOI: 10.3389/fcell.2021.667815] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 03/17/2021] [Indexed: 01/08/2023] Open
Abstract
5-hydroxytryptamine (5-HT) is derived from the essential amino acid L-tryptophan. Although the compound has been studied extensively for its neuronal handling and synaptic actions, serotonin 5-HT receptors can be found extra-synaptically and not only in neurons but in many types of mammalian cells, inside and outside the central nervous system (CNS). In sharp contrast, glutamate (Glu) and ATP are better known as metabolism-related molecules, but they also are neurotransmitters, and their receptors are expressed on almost any type of cell inside and outside the nervous system. Whereas 5-hydroxytryptamine and Glu are key regulators of the immune system, ATP actions are more general. 5-hydroxytryptamine, ATP and Glu act through both G protein-coupled receptors (GPCRs), and ionotropic receptors, i.e., ligand gated ion channels. These are the three examples of neurotransmitters whose actions as holistic regulatory molecules are briefly put into perspective here.
Collapse
Affiliation(s)
- Rafael Franco
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Madrid, Spain,*Correspondence: Rafael Franco, ;
| | - Rafael Rivas-Santisteban
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Jaume Lillo
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain,Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Madrid, Spain
| | - Jordi Camps
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain
| | - Gemma Navarro
- Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIberNed), Instituto de Salud Carlos III, Madrid, Spain,Department of Biochemistry and Physiology, School of Pharmacy, University of Barcelona, Barcelona, Spain
| | - Irene Reyes-Resina
- Department Biochemistry and Molecular Biomedicine, School of Biology, University of Barcelona, Barcelona, Spain,Irene Reyes-Resina,
| |
Collapse
|
33
|
Zimmermann H. Ectonucleoside triphosphate diphosphohydrolases and ecto-5'-nucleotidase in purinergic signaling: how the field developed and where we are now. Purinergic Signal 2021; 17:117-125. [PMID: 33336318 PMCID: PMC7954995 DOI: 10.1007/s11302-020-09755-6] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 11/12/2020] [Indexed: 12/17/2022] Open
Abstract
Geoffrey Burnstock will be remembered as the scientist who set up an entirely new field of intercellular communication, signaling via nucleotides. The signaling cascades involved in purinergic signaling include intracellular storage of nucleotides, nucleotide release, extracellular hydrolysis, and the effect of the released compounds or their hydrolysis products on target tissues via specific receptor systems. In this context ectonucleotidases play several roles. They inactivate released and physiologically active nucleotides, produce physiologically active hydrolysis products, and facilitate nucleoside recycling. This review briefly highlights the development of our knowledge of two types of enzymes involved in extracellular nucleotide hydrolysis and thus purinergic signaling, the ectonucleoside triphosphate diphosphohydrolases, and ecto-5'-nucleotidase.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Goethe University, Institute of Cell Biology and Neuroscience, Max-von-Laue-Str. 13, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
34
|
Zisimopoulos A, Klavdianou O, Theodossiadis P, Chatziralli I. The Role of the Microbiome in Age-Related Macular Degeneration: A Review of the Literature. Ophthalmologica 2021; 244:173-178. [PMID: 33550293 DOI: 10.1159/000515026] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 02/03/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND Age-related macular degeneration (AMD) is a progressive, multifactorial, degenerative disease and the leading cause of severe visual loss in the elderly population. The exact pathogenesis of AMD remains elusive, being the combination of genetic, environmental, metabolic, and functional processes. A better understanding of the disease's pathophysiology can lead to new treatment targets. The human microbiome seems to be a potential therapeutic pathway for AMD, as it has been recently proven to play a role in its pathogenesis. SUMMARY This review sheds light on the association between the microbiome and AMD. Key Messages: The current evidence based on the existing literature shows that there are differences in taxonomical and functional profiles in the human microbiome between patients with AMD and controls, suggesting that the microbiome is implicated in AMD onset and progression, being a link between AMD and nutrition/diet. Additionally, specific bacterial classes have been proposed as potential biomarkers for AMD diagnosis. Further randomized clinical studies with a large sample are needed to elucidate the role of the microbiome in AMD and to draw more solid conclusions.
Collapse
Affiliation(s)
| | - Olga Klavdianou
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Theodossiadis
- 2nd Department of Ophthalmology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Irini Chatziralli
- 2nd Department of Ophthalmology, Attikon University Hospital, National and Kapodistrian University of Athens, Athens, Greece,
| |
Collapse
|
35
|
Serrya MS, El-Sheakh AR, Makled MN. Evaluation of the therapeutic effects of mycophenolate mofetil targeting Nrf-2 and NLRP3 inflammasome in acetic acid induced ulcerative colitis in rats. Life Sci 2021; 271:119154. [PMID: 33539910 DOI: 10.1016/j.lfs.2021.119154] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 01/17/2021] [Accepted: 01/25/2021] [Indexed: 01/01/2023]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease that increases the risk of colorectal cancer. UC is highly associated with the disturbance of the immune system leading to oxidative stress and chronic inflammation of intestine. Therefore, the current study was conducted to investigate the potential anti-oxidant and anti-inflammatory effects of MMF against acetic acid-induced UC that might be associated with the regulation of Nrf-2 and NLRP3 inflammasome signaling. UC was induced in Sprague-Dawley rats by intracolonic instillation of acetic acid. Forty-eight hours post UC induction, MMF (50 mg/kg/day, orally) was given for 8 consecutive days. Then, colon tissues and blood samples were collected. Results showed that MMF significantly attenuated the acetic acid-induced functional, biochemical, and inflammatory injuries in colon. MMF significantly decreased oxidative stress as indicated by the decreased malondialdehyde concentration and the increased total antioxidant capacity, glutathione, catalase, and superoxide dismutase concentrations in colon tissues. MMF also significantly increased Nrf-2 and decreased NLRP3 inflammasome expressions. Moreover, MMF decreased expression of interferon-gamma and increased expression of interferon-alpha. MMF also significantly decreased expression of pro-inflammatory cytokines, interleukin (IL)-1β and IL-18. These results suggest that MMF has antioxidant and anti-inflammatory effects against acetic acid-induced UC through the upregulation of Nrf-2, and INF-α expression in addition to the suppression of NLRP3 inflammasome and subsequent release of IL1β, IL-18 and INF-γ.
Collapse
Affiliation(s)
- Marwa S Serrya
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed R El-Sheakh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Mirhan N Makled
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| |
Collapse
|
36
|
Serna E, Cespedes C, Vina J. Anti-Aging Physiological Roles of Aryl Hydrocarbon Receptor and Its Dietary Regulators. Int J Mol Sci 2020; 22:ijms22010374. [PMID: 33396477 PMCID: PMC7795126 DOI: 10.3390/ijms22010374] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 01/13/2023] Open
Abstract
The vast majority of the literature on the aryl hydrocarbon receptor is concerned with its functions in xenobiotic detoxification. However, in the course of evolution, this receptor had to have physiological (rather than toxicological) functions. Our aim was to review the aryl hydrocarbon receptor’s role in the physiological functions involved in aging. This study was performed by searching the MEDLINE and Google Academic databases. A total of 34 articles were selected that focused specifically on the aryl hydrocarbon receptor and aging, the aryl hydrocarbon receptor and physiological functions, and the combination of both. This receptor’s main physiological functions (mediated by the modulation of gene expression) were cell regeneration, the immune reaction, intestinal homeostasis, and cell proliferation. Furthermore, it was shown that the loss of this receptor led to premature aging. This process may be caused by the dysregulation of hematopoietic stem cells, loss of glucose and lipid homeostasis, increase in inflammation, and deterioration of the brain. We conclude that the aryl hydrocarbon receptor, apart from its well-established role in xenobiotic detoxication, plays an important role in physiological functions and in the aging process. Modulation of the signaling pathway of this receptor could be a therapeutic target of interest in aging.
Collapse
Affiliation(s)
- Eva Serna
- Correspondence: ; Tel.: +34-96-386-41-00 (ext. 83171)
| | | | | |
Collapse
|
37
|
Yegutkin GG. Adenosine metabolism in the vascular system. Biochem Pharmacol 2020; 187:114373. [PMID: 33340515 DOI: 10.1016/j.bcp.2020.114373] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/20/2022]
Abstract
The concept of extracellular purinergic signaling was first proposed by Geoffrey Burnstock in the early 1970s. Since then, extracellular ATP and its metabolites ADP and adenosine have attracted an enormous amount of attention in terms of their involvement in a wide range of immunomodulatory, thromboregulatory, angiogenic, vasoactive and other pathophysiological activities in different organs and tissues, including the vascular system. In addition to significant progress in understanding the properties of nucleotide- and adenosine-selective receptors, recent studies have begun to uncover the complexity of regulatory mechanisms governing the duration and magnitude of the purinergic signaling cascade. This knowledge has led to the development of new paradigms in understanding the entire purinome by taking into account the multitude of signaling and metabolic pathways involved in biological effects of ATP and adenosine and compartmentalization of the adenosine system. Along with the "canonical route" of ATP breakdown to adenosine via sequential ecto-nucleoside triphosphate diphosphohydrolase-1 (NTPDase1/CD39) and ecto-5'-nucleotidase/CD73 activities, it has now become clear that purine metabolism is the result of concerted effort between ATP release, its metabolism through redundant nucleotide-inactivating and counteracting ATP-regenerating ectoenzymatic pathways, as well as cellular nucleoside uptake and phosphorylation of adenosine to ATP through complex phosphotransfer reactions. In this review I provide an overview of key enzymes involved in adenosine metabolic network, with special emphasis on the emerging roles of purine-converting ectoenzymes as novel targets for cancer and vascular therapies.
Collapse
|
38
|
Equilibrative Nucleoside Transporter 2: Properties and Physiological Roles. BIOMED RESEARCH INTERNATIONAL 2020; 2020:5197626. [PMID: 33344638 PMCID: PMC7732376 DOI: 10.1155/2020/5197626] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Equilibrative nucleoside transporter 2 (ENT2) is a bidirectional transporter embedded in the biological membrane and is ubiquitously found in most tissue and cell types. ENT2 mediates the uptake of purine and pyrimidine nucleosides and nucleobase besides transporting a variety of nucleoside-derived drugs, mostly in anticancer therapy. Since high expression of ENT2 has been correlated with advanced stages of different types of cancers, consequently, this has gained significant interest in the role of ENT2 as a potential therapeutic target. Furthermore, ENT2 plays critical roles in signaling pathway and cell cycle progression. Therefore, elucidating the physiological roles of ENT2 and its properties may contribute to a better understanding of ENT2 roles beyond their transportation mechanism. This review is aimed at highlighting the main roles of ENT2 and at providing a brief update on the recent research.
Collapse
|
39
|
Afzal S, Al-Rashida M, Hameed A, Pelletier J, Sévigny J, Iqbal J. Functionalized Oxoindolin Hydrazine Carbothioamide Derivatives as Highly Potent Inhibitors of Nucleoside Triphosphate Diphosphohydrolases. Front Pharmacol 2020; 11:585876. [PMID: 33328992 PMCID: PMC7734281 DOI: 10.3389/fphar.2020.585876] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 10/13/2020] [Indexed: 12/13/2022] Open
Abstract
Ectonucleoside triphosphate diphosphohydrolases (NTPDases) are ectoenzymes that play an important role in the hydrolysis of nucleoside triphosphate and diphosphate to nucleoside monophosphate. NTPDase1, -2, -3 and -8 are the membrane bound members of this enzyme family that are responsible for regulating the levels of nucleotides in extracellular environment. However, the pathophysiological functions of these enzymes are not fully understood due to lack of potent and selective NTPDase inhibitors. Herein, a series of oxoindolin hydrazine carbothioamide derivatives is synthesized and screened for NTPDase inhibitory activity. Four compounds were identified as selective inhibitors of h-NTPDase1 having IC50 values in lower micromolar range, these include compounds 8b (IC50 = 0.29 ± 0.02 µM), 8e (IC50 = 0.15 ± 0.009 µM), 8f (IC50 = 0.24 ± 0.01 µM) and 8l (IC50 = 0.30 ± 0.03 µM). Similarly, compound 8k (IC50 = 0.16 ± 0.01 µM) was found to be a selective h-NTPDase2 inhibitor. In case of h-NTPDase3, most potent inhibitors were compounds 8c (IC50 = 0.19 ± 0.02 µM) and 8m (IC50 = 0.38 ± 0.03 µM). Since NTPDase3 has been reported to be associated with the regulation of insulin secretion, we evaluated our synthesized NTPDase3 inhibitors for their ability to stimulate insulin secretion in isolated mice islets. Promising results were obtained showing that compound 8m potently stimulated insulin secretion without affecting the NTPDase3 gene expression. Molecular docking studies of the most potent compounds were also carried out to rationalize binding site interactions. Hence, these compounds are useful tools to study the role of NTPDase3 in insulin secretion.
Collapse
Affiliation(s)
- Saira Afzal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Mariya Al-Rashida
- Department of Chemistry, Forman Christian College (A Chartered University), Lahore, Pakistan
| | - Abdul Hameed
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| | - Julie Pelletier
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Jean Sévigny
- Centre de Recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.,Département de Microbiologie-Infectiologie et d'Immunologie, Faculté de Médecine, Université Laval, Québec City, QC, Canada
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad, Pakistan
| |
Collapse
|
40
|
Zimmermann H. History of ectonucleotidases and their role in purinergic signaling. Biochem Pharmacol 2020; 187:114322. [PMID: 33161020 DOI: 10.1016/j.bcp.2020.114322] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022]
Abstract
Ectonucleotidases are key for purinergic signaling. They control the duration of activity of purinergic receptor agonists. At the same time, they produce hydrolysis products as additional ligands of purinergic receptors. Due to the considerable diversity of enzymes, purinergic receptor ligands and purinergic receptors, deciphering the impact of extracellular purinergic receptor control has become a challenge. The first group of enzymes described were the alkaline phosphatases - at the time not as nucleotide-metabolizing but as nonspecific phosphatases. Enzymes now referred to as nucleoside triphosphate diphosphohydrolases and ecto-5'-nucleotidase were the first and only nucleotide-specific ectonucleotidases identified. And they were the first group of enzymes related to purinergic signaling. Additional research brought to light a surprising number of ectoenzymes with broad substrate specificity, which can also hydrolyze nucleotides. This short overview traces the development of the field and briefly highlights important results and benefits for therapies of human diseases achieved within nearly a century of investigations.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Goethe University, Institute of Cell Biology and Neuroscience, Max-von-Laue-Str. 13, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
41
|
Libera J, Wittner M, Kantowski M, Woost R, Eberhard JM, de Heer J, Reher D, Huber S, Haag F, Schulze Zur Wiesch J. Decreased Frequency of Intestinal CD39 + γδ + T Cells With Tissue-Resident Memory Phenotype in Inflammatory Bowel Disease. Front Immunol 2020; 11:567472. [PMID: 33072107 PMCID: PMC7541837 DOI: 10.3389/fimmu.2020.567472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 08/13/2020] [Indexed: 12/22/2022] Open
Abstract
The ectoenzymes CD39 and CD73 play a major role in controlling tissue inflammation by regulating the balance between adenosine triphosphate (ATP) and adenosine. Still, little is known about the role of these two enzymes and ATP and its metabolites in the pathophysiology of inflammatory bowel disease (IBD). We isolated mononuclear cells from peripheral blood and lamina propria of the large intestine of patients diagnosed with IBD and of healthy volunteers. We then comprehensively analyzed the CD39 and CD73 expression patterns together with markers of activation (HLA-DR, CD38), differentiation (CCR7, CD45RA) and tissue-residency (CD69, CD103, CD49a) on CD4+, CD8+, γδ+ T cells and mucosa-associated invariant T cells using flow cytometry. CD39 expression levels of γδ+ and CD8+ T cells in lamina propria lymphocytes (LPL) were much higher compared to peripheral blood mononuclear cells. Moreover, the frequency of CD39+ CD4+ and CD8+, but not γδ+ LPL positively correlated with T-cell activation. The frequency of CD39+ cells among tissue-resident memory LPL (Trm) was higher compared to non-Trm for all subsets, confirming that CD39 is a marker for the tissue-resident memory phenotype. γδ+ Trm also showed a distinct cytokine profile upon stimulation – the frequency of IFN-γ+ and IL-17A+ cells was significantly lower in γδ+ Trm compared to non-Trm. Interestingly, we observed a decreased frequency of CD39+ γδ+ T cells in IBD patients compared to healthy controls (p = 0.0049). Prospective studies need to elucidate the exact role of this novel CD39+ γδ+ T-cell population with tissue-resident memory phenotype and its possible contribution to the pathogenesis of IBD and other inflammatory disorders.
Collapse
Affiliation(s)
- Jana Libera
- I. Department of Medicine, Infectious Disease Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Melanie Wittner
- I. Department of Medicine, Infectious Disease Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
| | - Marcus Kantowski
- Clinic and Polyclinic for Interdisciplinary Endoscopy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Robin Woost
- I. Department of Medicine, Infectious Disease Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
| | - Johanna M Eberhard
- I. Department of Medicine, Infectious Disease Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
| | - Jocelyn de Heer
- Clinic and Polyclinic for Interdisciplinary Endoscopy, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dominik Reher
- I. Department of Medicine, Infectious Disease Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, Infectious Disease Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Friedrich Haag
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Julian Schulze Zur Wiesch
- I. Department of Medicine, Infectious Disease Unit, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner Site Hamburg Lübeck Borstel Riems, Hamburg, Germany
| |
Collapse
|
42
|
Associations of the intestinal microbiome with the complement system in neovascular age-related macular degeneration. NPJ Genom Med 2020; 5:34. [PMID: 32922859 PMCID: PMC7463023 DOI: 10.1038/s41525-020-00141-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Age-related macular degeneration (AMD) is a leading cause of severe vision loss in the aged population. The etiology of AMD is multifactorial including nutritional factors, genetic variants mainly in the complement pathway, environmental risk factors and alterations in the intestinal microbiome. However, it remains unexplored whether there is an interdependency of these factors leading to the development of AMD. To investigate this issue, a shotgun metagenomics analysis of 57 neovascular AMD and 58 healthy controls as well as of 16 complement C3-deficient mice and 16 wildtypes was performed. Whereas the class Negativicutes was more abundant in patients, the genus Oscillibacter and species Bacteroides had a significantly higher prevalence in persons without AMD. Similar taxonomic features were identified that distinguished wildtype mice from C3-deficient mice. Moreover, several purine signaling pathways were associated with both, neovascular AMD and C3 deficiency. While SNPs within the complement factor B gene were more abundant in controls, SNPs within the high temperature requirement A serine peptidase 1 and complement factor H (CFH) genes were associated with neovascular AMD. Using a classification model, Negativicutes was identified as a potential biomarker for AMD and furthermore, it positively correlated with CFH. This study suggests an association between the intestinal microbiome and the complement system in neovascular AMD.
Collapse
|
43
|
Burster T, Knippschild U, Molnár F, Zhanapiya A. Cathepsin G and its Dichotomous Role in Modulating Levels of MHC Class I Molecules. Arch Immunol Ther Exp (Warsz) 2020; 68:25. [PMID: 32815043 DOI: 10.1007/s00005-020-00585-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 06/11/2020] [Indexed: 12/21/2022]
Abstract
Cathepsin G (CatG) is involved in controlling numerous processes of the innate and adaptive immune system. These features include the proteolytic activity of CatG and play a pivotal role in alteration of chemokines as well as cytokines, clearance of exogenous and internalized pathogens, platelet activation, apoptosis, and antigen processing. This is in contrast to the capability of CatG acting in a proteolytic-independent manner due to the net charge of arginine residues in the CatG sequence which interferes with bacteria. CatG is a double-edged sword; CatG is also responsible in pathophysiological conditions, such as autoimmunity, chronic pulmonary diseases, HIV infection, tumor progression and metastasis, photo-aged human skin, Papillon-Lefèvre syndrome, and chronic inflammatory pain. Here, we summarize the latest findings for functional responsibilities of CatG in immunity, including bivalent regulation of major histocompatibility complex class I molecules, which underscore an additional novel role of CatG within the immune system.
Collapse
Affiliation(s)
- Timo Burster
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Kabanbay Batyr Ave. 53, Nur-Sultan, 010000, Kazakhstan.
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Hospital, 89081, Ulm, Germany
| | - Ferdinand Molnár
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Kabanbay Batyr Ave. 53, Nur-Sultan, 010000, Kazakhstan
| | - Anuar Zhanapiya
- Department of Biology, School of Sciences and Humanities, Nazarbayev University, Kabanbay Batyr Ave. 53, Nur-Sultan, 010000, Kazakhstan
| |
Collapse
|
44
|
Vesicular Transport of Encapsulated microRNA between Glial and Neuronal Cells. Int J Mol Sci 2020; 21:ijms21145078. [PMID: 32708414 PMCID: PMC7404393 DOI: 10.3390/ijms21145078] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/09/2020] [Accepted: 07/15/2020] [Indexed: 02/06/2023] Open
Abstract
Exosomes (EXs) and extracellular microvesicles (EMVs) represent a diverse assortment of plasma membrane-derived nanovesicles, 30–1000 nm in diameter, released by all cell lineages of the central nervous system (CNS). They are examples of a very active and dynamic form of extracellular communication and the conveyance of biological information transfer essential to maintain homeostatic neurological functions and contain complex molecular cargoes representative of the cytoplasm of their cells of origin. These molecular cargoes include various mixtures of proteins, lipids, proteolipids, cytokines, chemokines, carbohydrates, microRNAs (miRNA) and messenger RNAs (mRNA) and other components, including end-stage neurotoxic and pathogenic metabolic products, such as amyloid beta (Aβ) peptides. Brain microglia, for example, respond to both acute CNS injuries and degenerative diseases with complex reactions via the induction of a pro-inflammatory phenotype, and secrete EXs and EMVs enriched in selective pathogenic microRNAs (miRNAs) such as miRNA-34a, miRNA-125b, miRNA-146a, miRNA-155, and others that are known to promote neuro-inflammation, induce complement activation, disrupt innate–immune signaling and deregulate the expression of neuron-specific phosphoproteins involved in neurotropism and synaptic signaling. This communication will review our current understanding of the trafficking of miRNA-containing EXs and EMVs from astrocytes and “activated pro-inflammatory” microglia to target neurons in neurodegenerative diseases with an emphasis on Alzheimer’s disease wherever possible.
Collapse
|
45
|
Conversion of extracellular ATP into adenosine: a master switch in renal health and disease. Nat Rev Nephrol 2020; 16:509-524. [PMID: 32641760 DOI: 10.1038/s41581-020-0304-7] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2020] [Indexed: 12/22/2022]
Abstract
ATP and its ultimate degradation product adenosine are potent extracellular signalling molecules that elicit a variety of pathophysiological functions in the kidney through the activation of P2 and P1 purinergic receptors, respectively. Extracellular purines can modulate immune responses, balancing inflammatory processes and immunosuppression; indeed, alterations in extracellular nucleotide and adenosine signalling determine outcomes of inflammation and healing processes. The functional activities of ectonucleotidases such as CD39 and CD73, which hydrolyse pro-inflammatory ATP to generate immunosuppressive adenosine, are therefore pivotal in acute inflammation. Protracted inflammation may result in aberrant adenosinergic signalling, which serves to sustain inflammasome activation and worsen fibrotic reactions. Alterations in the expression of ectonucleotidases on various immune cells, such as regulatory T cells and macrophages, as well as components of the renal vasculature, control purinergic receptor-mediated effects on target tissues within the kidney. The role of CD39 as a rheostat that can have an impact on purinergic signalling in both acute and chronic inflammation is increasingly supported by the literature, as detailed in this Review. Better understanding of these purinergic processes and development of novel drugs targeting these pathways could lead to effective therapies for the management of acute and chronic kidney disease.
Collapse
|
46
|
Jackson EK, Gillespie DG, Cheng D, Mi Z, Menshikova EV. Characterization of the N 6-etheno-bridge method to assess extracellular metabolism of adenine nucleotides: detection of a possible role for purine nucleoside phosphorylase in adenosine metabolism. Purinergic Signal 2020; 16:187-211. [PMID: 32367441 PMCID: PMC7367995 DOI: 10.1007/s11302-020-09699-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/08/2020] [Indexed: 12/11/2022] Open
Abstract
The goal of this study was to determine the validity of using N6-etheno-bridged adenine nucleotides to evaluate ecto-nucleotidase activity. We observed that the metabolism of N6-etheno-ATP versus ATP was quantitatively similar when incubated with recombinant CD39, ENTPD2, ENTPD3, or ENPP-1, and the quantitative metabolism of N6-etheno-AMP versus AMP was similar when incubated with recombinant CD73. This suggests that ecto-nucleotidases process N6-etheno-bridged adenine nucleotides similarly to endogenous adenine nucleotides. Four cell types rapidly (t1/2, 0.21 to 0.66 h) metabolized N6-etheno-ATP. Applied N6-etheno-ATP was recovered in the medium as N6-etheno-ADP, N6-etheno-AMP, N6-etheno-adenosine, and surprisingly N6-etheno-adenine; intracellular N6-etheno compounds were undetectable. This suggests minimal cellular uptake, intracellular metabolism, or deamination of these compounds. N6-etheno-ATP, N6-etheno-ADP, N6-etheno-AMP, N6-etheno-adenosine, and N6-etheno-adenine had little affinity for recombinant A1, A2A, or A2B receptors, for a subset of P2X receptors (3H-α,β-methylene-ATP binding to rat bladder membranes), or for a subset of P2Y receptors (35S-ATP-αS binding to rat brain membranes), suggesting minimal pharmacological activity. N6-etheno-adenosine was partially converted to N6-etheno-adenine in four different cell types; this was blocked by purine nucleoside phosphorylase (PNPase) inhibition. Intravenous N6-etheno-ATP was quickly metabolized, with N6-etheno-adenine being the main product in naïve rats, but not in rats pretreated with a PNPase inhibitor. PNPase inhibition reduced the urinary excretion of endogenous adenine and attenuated the conversion of exogenous adenosine to adenine in the renal cortex. The N6-etheno-bridge method is a valid technique to assess extracellular metabolism of adenine nucleotides by ecto-nucleotidases. Also, rats express an enzyme with PNPase-like activity that metabolizes N6-etheno-adenosine to N6-etheno-adenine.
Collapse
Affiliation(s)
- Edwin K. Jackson
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Delbert G. Gillespie
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Dongmei Cheng
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Zaichuan Mi
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| | - Elizabeth V. Menshikova
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, 100 Technology Drive, Room 514, Pittsburgh, PA 15219 USA
| |
Collapse
|
47
|
Abstract
Purinergic signaling was proposed in 1972, after it was demonstrated that adenosine 5'-triphosphate (ATP) was a transmitter in nonadrenergic, noncholinergic inhibitory nerves supplying the guinea-pig taenia coli. Later, ATP was identified as an excitatory cotransmitter in sympathetic and parasympathetic nerves, and it is now apparent that ATP acts as a cotransmitter in most, if not all, nerves in both the peripheral nervous system and central nervous system (CNS). ATP acts as a short-term signaling molecule in neurotransmission, neuromodulation, and neurosecretion. It also has potent, long-term (trophic) roles in cell proliferation, differentiation, and death in development and regeneration. Receptors to purines and pyrimidines have been cloned and characterized: P1 adenosine receptors (with four subtypes), P2X ionotropic nucleotide receptors (seven subtypes) and P2Y metabotropic nucleotide receptors (eight subtypes). ATP is released from different cell types by mechanical deformation, and after release, it is rapidly broken down by ectonucleotidases. Purinergic receptors were expressed early in evolution and are widely distributed on many different nonneuronal cell types as well as neurons. Purinergic signaling is involved in embryonic development and in the activities of stem cells. There is a growing understanding about the pathophysiology of purinergic signaling and there are therapeutic developments for a variety of diseases, including stroke and thrombosis, osteoporosis, pain, chronic cough, kidney failure, bladder incontinence, cystic fibrosis, dry eye, cancer, and disorders of the CNS, including Alzheimer's, Parkinson's. and Huntington's disease, multiple sclerosis, epilepsy, migraine, and neuropsychiatric and mood disorders.
Collapse
|
48
|
Savio LEB, de Andrade Mello P, Santos SACS, de Sousa JC, Oliveira SDS, Minshall RD, Kurtenbach E, Wu Y, Longhi MS, Robson SC, Coutinho-Silva R. P2X7 receptor activation increases expression of caveolin-1 and formation of macrophage lipid rafts, thereby boosting CD39 activity. J Cell Sci 2020; 133:jcs.237560. [PMID: 32005701 DOI: 10.1242/jcs.237560] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Macrophages are tissue-resident immune cells that are crucial for the initiation and maintenance of immune responses. Purinergic signaling modulates macrophage activity and impacts cellular plasticity. The ATP-activated purinergic receptor P2X7 (also known as P2RX7) has pro-inflammatory properties, which contribute to macrophage activation. P2X7 receptor signaling is, in turn, modulated by ectonucleotidases, such as CD39 (also known as ENTPD1), expressed in caveolae and lipid rafts. Here, we examined P2X7 receptor activity and determined impacts on ectonucleotidase localization and function in macrophages primed with lipopolysaccharide (LPS). First, we verified that ATP boosts CD39 activity and caveolin-1 protein expression in LPS-primed macrophages. Drugs that disrupt cholesterol-enriched domains - such as nystatin and methyl-β-cyclodextrin - decreased CD39 enzymatic activity in all circumstances. We noted that CD39 colocalized with lipid raft markers (flotillin-2 and caveolin-1) in macrophages that had been primed with LPS followed by treatment with ATP. P2X7 receptor inhibition blocked these ATP-mediated increases in caveolin-1 expression and inhibited the colocalization with CD39. Further, we found that STAT3 activation is significantly attenuated caveolin-1-deficient macrophages treated with LPS or LPS+BzATP. Taken together, our data suggest that P2X7 receptor triggers the initiation of lipid raft-dependent mechanisms that upregulates CD39 activity and could contribute to limit macrophage responses restoring homeostasis.
Collapse
Affiliation(s)
- Luiz Eduardo Baggio Savio
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Paola de Andrade Mello
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Stephanie Alexia Cristina Silva Santos
- Laboratory of Molecular Biology and Biochemistry of Proteins, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Júlia Costa de Sousa
- Laboratory of Molecular Biology and Biochemistry of Proteins, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Suellen D S Oliveira
- Departments of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Richard D Minshall
- Departments of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA.,Departments of Pharmacology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Eleonora Kurtenbach
- Laboratory of Molecular Biology and Biochemistry of Proteins, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| | - Yan Wu
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Maria Serena Longhi
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Simon C Robson
- Departments of Medicine and Anesthesia, Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard University, Boston, MA 02215, USA
| | - Robson Coutinho-Silva
- Laboratory of Immunophysiology, Biophysics Institute Carlos Chagas Filho, Federal University of Rio de Janeiro, 21941-902 Rio de Janeiro, Brazil
| |
Collapse
|
49
|
Fan H, Du J, Liu X, Zheng WW, Zhuang ZH, Wang CD, Gao R. Effects of pentasa-combined probiotics on the microflora structure and prognosis of patients with inflammatory bowel disease. TURKISH JOURNAL OF GASTROENTEROLOGY 2020; 30:680-685. [PMID: 31418411 DOI: 10.5152/tjg.2019.18426] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND/AIMS The aim of the present study was to investigate the effects of the combination treatment of pentasa and probiotics on the microflora composition and prognosis in patients with inflammatory bowel disease (IBD). MATERIALS AND METHODS A total of 40 patients with IBD (19 control group and 21 observation group) were randomized. Patients in the control group were given pentasa, and patients in the observation group were given probiotics along with pentasa. The microflora composition, biochemical indices, inflammatory markers, and activity scores of the two groups were analyzed. RESULTS After treatment, the number of enterobacteria, enterococci, saccharomyces, and bacteroides; the levels of fecal lactoferrin, 1-antitrypsin, β2-microglobulin, high-sensitivity C-reactive protein, and interleukin (IL)-6; activity scores; and recurrence rate in the observation group were significantly lower than those in the control group. Bifidobacterium and lactobacillus counts and IL-4 levels were significantly higher in the observation group than in the control group. CONCLUSION The combination of probiotics and pentasa can improve microflora composition in patients with IBD and reduce the level of inflammatory cytokines; therefore, it is worthy of further clinical validation.
Collapse
Affiliation(s)
- Hua Fan
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Juan Du
- Second Department of Gastroenterology, General Hospital of Yankuang Group, Zoucheng, Shandong Province, China
| | - Xia Liu
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Wei-Wei Zheng
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Ze-Hao Zhuang
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Cheng-Dang Wang
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| | - Rui Gao
- Department of Gastroenterology, The First Affiliated Hospital of Fujian Medical University, Fuzhou, Fujian Province, China
| |
Collapse
|
50
|
Ercan G, Yigitturk G, Erbas O. Therapeutic effect of adenosine on experimentally induced acute ulcerative colitis model in rats. Acta Cir Bras 2020; 34:e201901204. [PMID: 32074166 PMCID: PMC7025795 DOI: 10.1590/s0102-865020190120000004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 11/12/2019] [Indexed: 12/30/2022] Open
Abstract
Purpose To examine the therapeutic effect of external adenosine on an acetic acid-induced acute ulcerative colitis model in rats. Methods Thirty male mature rats were divided into three groups as control, acute colitis (AC) and AC+adenosine group (AC+AD). AC was induced by rectal administration of 4% acetic acid (AA). 5mg/kg/day adenosine was performed i.p for 4 weeks to AC+AD group. Rectum and colon were excised for microscopic and histopathological histopathologic evaluations, and immunohistochemical analysis of nuclear factor kappa B (NF-kB). Blood samples were collected for biochemical detection of TNF-α, Pentraxin-3 and malondialdehyde (MDA) levels. Results AC group had generalized hyperemia and hemorrhage with increased macroscopic and histopathological scores compared with control (P <0.0001) while adenosine treatment decreased these scores significantly (P <0.001), with reduced distribution of disrupted epithelium, leukocyte infiltrates, and focal hemorrhage. AC group showed significantly increased immunoexpression of NF-kB in rectum, plasma and tissue levels of TNF-α, plasma Pentraxin-3 and MDA levels (P <0.0001) while adenosine reduced these levels (P < 0.05). Conclusion Adenosine appears to promote healing of colon and rectum exposed to AA-induced AC, suggesting a boosting effect of adenosine on the intestinal immune system to cure ulcerative colitis.
Collapse
Affiliation(s)
- Gulcin Ercan
- University of Health Science Bagcilar Training and Research Hospital, Turkey
| | | | | |
Collapse
|