101
|
Nascimento MT, Viana DL, Peixoto FC, Arruda SM, Carvalho EM, Carvalho LP. Prostaglandin E2 contributes to L. braziliensis survival and therapeutic failure in cutaneous leishmaniasis. Emerg Microbes Infect 2023; 12:2261565. [PMID: 37729084 PMCID: PMC10540647 DOI: 10.1080/22221751.2023.2261565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023]
Abstract
Patients with cutaneous leishmaniasis (CL) present an exacerbated inflammatory response associated with tissue damage and ulcer development. In recent years, higher rates of failure to pentavalent antimoniate therapy have been observed, yet the underlying reason remains poorly understood. We hypothesize that the eicosanoid PGE2 favours the establishment of infection by L. braziliensis, which contributes to therapeutic failure. The aim of the present study was to investigate the influence of PGE2 on the survival of L. braziliensis in macrophages and rates of therapeutic failure in CL patients. PGE2, an eicosanoid derived from the metabolism of arachidonic acid by the COX-2 enzyme, plays several roles in immune response. We found that increased PGE2 decreases the microbicidal function of macrophages and is associated with disease severity and therapeutic failure. Additionally, the neutralization of COX-2 by NS398, a selective NSAID, increases the ability of macrophages to kill L. braziliensis and protects against the pathological inflammatory response. Our data suggest that NS398 may serve as an adjunct treatment for CL patients.
Collapse
Affiliation(s)
- Maurício T. Nascimento
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
- Serviço de Imunologia, SIM, Complexo Universitário Professor Edgar Santos, COM-HUPES, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, PPgCS, Universidade Federal da Bahia, Salvador, Brazil
| | - Débora L. Viana
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
| | - Fábio C. Peixoto
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, PPgCS, Universidade Federal da Bahia, Salvador, Brazil
| | - Sérgio M. Arruda
- Laboratório Avançado de Saúde Pública, LASP, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
| | - Edgar M. Carvalho
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
- Serviço de Imunologia, SIM, Complexo Universitário Professor Edgar Santos, COM-HUPES, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, PPgCS, Universidade Federal da Bahia, Salvador, Brazil
- Instituto de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador, Brazil
| | - Lucas P. Carvalho
- Laboratório de Pesquisas Clínicas, LAPEC, Instituto Gonçalo Moniz – Fiocruz, Salvador, Brazil
- Serviço de Imunologia, SIM, Complexo Universitário Professor Edgar Santos, COM-HUPES, Salvador, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, PPgCS, Universidade Federal da Bahia, Salvador, Brazil
- Instituto de Ciência e Tecnologia em Doenças Tropicais, INCT-DT, Salvador, Brazil
| |
Collapse
|
102
|
Sun Y, Lian T, Huang Q, Chang Y, Li Y, Guo X, Kong W, Yang Y, Zhang K, Wang P, Wang X. Nanomedicine-mediated regulated cell death in cancer immunotherapy. J Control Release 2023; 364:174-194. [PMID: 37871752 DOI: 10.1016/j.jconrel.2023.10.032] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 10/25/2023]
Abstract
Immunotherapy has attracted widespread attention in cancer treatment and has achieved considerable success in the clinical treatment of some tumors, but it has a low response rate in most tumors. To achieve sufficient activation of the immune response, significant efforts using nanotechnology have been made to enhance cancer immune response. In recent years, the induction of various regulated cell death (RCD) has emerged as a potential antitumor immuno-strategy, including processes related to apoptosis, autophagy, necroptosis, pyroptosis, ferroptosis, and cuproptosis. In particular, damage-associated molecular patterns (DAMPs) released from the damaged membrane of dying cells act as in situ adjuvants to trigger antigen-specific immune responses by the exposure of an increased antigenicity. Thus, RCD-based immunotherapy offers a new approach for enhancing cancer treatment efficacy. Furthermore, incorporation with multimodal auxiliary therapies in cell death-based immunotherapy can trigger stronger immune responses, resulting in more efficient therapeutic outcome. This review discusses different RCD modalities and summarizes recent nanotechnology-mediated RCDs in cancer immunotherapy.
Collapse
Affiliation(s)
- Yue Sun
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China; The Xi'an key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Ting Lian
- Research Center for Prevention and Treatment of Respiratory Disease, School of Clinical Medicine, Xi'an Medical University, Xi'an, Shaanxi 710021, China
| | - Qichao Huang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yawei Chang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yuan Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Xiaoyu Guo
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Weirong Kong
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Yifang Yang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Kun Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Pan Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Xiaobing Wang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
103
|
Biagini D, Oliveri P, Baj A, Gasperina DD, Ferrante FD, Lomonaco T, Ghimenti S, Lenzi A, Bonini A, Vivaldi F, Oger C, Galano JM, Balas L, Durand T, Maggi F, Di Francesco F. The effect of SARS-CoV-2 variants on the plasma oxylipins and PUFAs of COVID-19 patients. Prostaglandins Other Lipid Mediat 2023; 169:106770. [PMID: 37633481 DOI: 10.1016/j.prostaglandins.2023.106770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 08/16/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Oxylipins are important signalling compounds that are significantly involved in the regulation of the immune system and the resolution of inflammation. Lipid metabolism is strongly activated upon SARS-CoV-2 infection, however the modulating effects of oxylipins induced by different variants remain unexplored. Here, we compare the plasma profiles of thirty-seven oxylipins and four PUFAs in subjects infected with Wild-type, Alpha (B.1.1.7), Delta (B.1.617.2), and Omicron (B.1.1.529) variants. The results suggest that oxidative stress and inflammation resulting from COVID-19 were highly dependent on the SARS-CoV-2 variant, and that the Wild-type elicited the strongest inflammatory storm. The Alpha and Delta variants induced a comparable lipid profile alteration upon infection, which differed significantly from Omicron. The latter variant increased the levels of pro-inflammatory mediators and decreased the levels of omega-3 PUFA in infected patients. We speculate that changes in therapeutics, vaccination, and prior infections may have a role in the alteration of the oxylipin profile besides viral mutations. The results shed new light on the evolution of the inflammatory response in COVID-19.
Collapse
Affiliation(s)
- Denise Biagini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy.
| | | | - Andreina Baj
- Department of Medicine and Technological Innovation, University of Insubria, Varese, Italy
| | | | | | - Tommaso Lomonaco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Silvia Ghimenti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Alessio Lenzi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Andrea Bonini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Federico Vivaldi
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy
| | - Camille Oger
- Institut des Biomolécules Max Mousseron (IBMM), Pôle Chimie Balard Recherche, University of Montpellier, ENSCN, UMR 5247 CNRS, France
| | - Jean-Marie Galano
- Institut des Biomolécules Max Mousseron (IBMM), Pôle Chimie Balard Recherche, University of Montpellier, ENSCN, UMR 5247 CNRS, France
| | - Laurence Balas
- Institut des Biomolécules Max Mousseron (IBMM), Pôle Chimie Balard Recherche, University of Montpellier, ENSCN, UMR 5247 CNRS, France
| | - Thierry Durand
- Institut des Biomolécules Max Mousseron (IBMM), Pôle Chimie Balard Recherche, University of Montpellier, ENSCN, UMR 5247 CNRS, France
| | - Fabrizio Maggi
- Laboratory of Virology, National Institute for Infectious Diseases "Lazzaro Spallanzani" - IRCCS, Rome, Italy
| | - Fabio Di Francesco
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa, Italy.
| |
Collapse
|
104
|
Andersen C, Jacobsen S, Uvebrant K, Griffin JF, Vonk LA, Walters M, Berg LC, Lundgren-Åkerlund E, Lindegaard C. Integrin α10β1-Selected Mesenchymal Stem Cells Reduce Pain and Cartilage Degradation and Increase Immunomodulation in an Equine Osteoarthritis Model. Cartilage 2023:19476035231209402. [PMID: 37990503 DOI: 10.1177/19476035231209402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2023] Open
Abstract
OBJECTIVE Integrin α10β1-selected mesenchymal stem cells (integrin α10-MSCs) have previously shown potential in treating cartilage damage and osteoarthritis (OA) in vitro and in animal models in vivo. The aim of this study was to further investigate disease-modifying effects of integrin α10-MSCs. DESIGN OA was surgically induced in 17 horses. Eighteen days after surgery, horses received 2 × 107 integrin α10-MSCs intra-articularly or were left untreated. Lameness and response to carpal flexion was assessed weekly along with synovial fluid (SF) analysis. On day 52 after treatment, horses were euthanized, and carpi were evaluated by computed tomography (CT), MRI, histology, and for macroscopic pathology and integrin α10-MSCs were traced in the joint tissues. RESULTS Lameness and response to carpal flexion significantly improved over time following integrin α10-MSC treatment. Treated horses had milder macroscopic cartilage pathology and lower cartilage histology scores than the untreated group. Prostaglandin E2 and interleukin-10 increased in the SF after integrin α10-MSC injection. Integrin α10-MSCs were found in SF from treated horses up to day 17 after treatment, and in the articular cartilage and subchondral bone from 5 of 8 treated horses after euthanasia at 52 days after treatment. The integrin α10-MSC injection did not cause joint flare. CONCLUSION This study demonstrates that intra-articular (IA) injection of integrin α10-MSCs appears to be safe, alleviate pathological changes in the joint, and improve joint function in an equine post-traumatic osteoarthritis (PTOA) model. The results suggest that integrin α10-MSCs hold promise as a disease-modifying osteoarthritis drug (DMOAD).
Collapse
Affiliation(s)
- Camilla Andersen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
- Xintela AB, Lund, Sweden
| | - Stine Jacobsen
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | | | - John F Griffin
- Department of Large Animal Clinical Sciences, Texas A&M University, College Station, TX, USA
| | | | - Marie Walters
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | - Lise Charlotte Berg
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| | | | - Casper Lindegaard
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Taastrup, Denmark
| |
Collapse
|
105
|
Maghsoudi H, Sheikhnia F, Sitarek P, Hajmalek N, Hassani S, Rashidi V, Khodagholi S, Mir SM, Malekinejad F, Kheradmand F, Ghorbanpour M, Ghasemzadeh N, Kowalczyk T. The Potential Preventive and Therapeutic Roles of NSAIDs in Prostate Cancer. Cancers (Basel) 2023; 15:5435. [PMID: 38001694 PMCID: PMC10670652 DOI: 10.3390/cancers15225435] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 11/26/2023] Open
Abstract
Prostate cancer (PC) is the second most common type of cancer and the leading cause of death among men worldwide. Preventing the progression of cancer after treatments such as radical prostatectomy, radiation therapy, and hormone therapy is a major concern faced by prostate cancer patients. Inflammation, which can be caused by various factors such as infections, the microbiome, obesity and a high-fat diet, is considered to be the main cause of PC. Inflammatory cells are believed to play a crucial role in tumor progression. Therefore, nonsteroidal anti-inflammatory drugs along with their effects on the treatment of inflammation-related diseases, can prevent cancer and its progression by suppressing various inflammatory pathways. Recent evidence shows that nonsteroidal anti-inflammatory drugs are effective in the prevention and treatment of prostate cancer. In this review, we discuss the different pathways through which these drugs exert their potential preventive and therapeutic effects on prostate cancer.
Collapse
Affiliation(s)
- Hossein Maghsoudi
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Farhad Sheikhnia
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Przemysław Sitarek
- Department of Medical Biology, Medical University of Lodz, 90-151 Lodz, Poland
| | - Nooshin Hajmalek
- Department of Clinical Biochemistry, School of Medicine, Babol University of Medical Sciences, Babol 47176-47754, Iran;
| | - Sepideh Hassani
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Vahid Rashidi
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
| | - Sadaf Khodagholi
- School of Kinesiology and Health Science, York University, Toronto, ON M3J 1P3, Canada;
| | - Seyed Mostafa Mir
- Metabolic Disorders Research Center, Department of Biochemistry and Biophysics, Gorgan Faculty of Medicine, Golestan University of Medical Sciences, Gorgan 49189-36316, Iran;
| | - Faezeh Malekinejad
- Student Research Committee, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (H.M.); (F.S.); (V.R.); (F.M.)
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Fatemeh Kheradmand
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
- Cellular and Molecular Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57147-83734, Iran
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia 57147-83734, Iran
| | - Mansour Ghorbanpour
- Department of Medicinal Plants, Faculty of Agriculture and Natural Resources, Arak University, Arak 38156-88349, Iran;
| | - Navid Ghasemzadeh
- Department of Clinical Biochemistry, School of Medicine, Urmia University of Medical Sciences, Urmia 57147-83734, Iran; (S.H.); (F.K.); (N.G.)
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-237 Lodz, Poland;
| |
Collapse
|
106
|
Aguirre GA, Goulart MR, Dalli J, Kocher HM. Arachidonate 15-lipoxygenase-mediated production of Resolvin D5 n-3 DPA abrogates pancreatic stellate cell-induced cancer cell invasion. Front Immunol 2023; 14:1248547. [PMID: 38035115 PMCID: PMC10687150 DOI: 10.3389/fimmu.2023.1248547] [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: 06/28/2023] [Accepted: 10/26/2023] [Indexed: 12/02/2023] Open
Abstract
Activation of pancreatic stellate cells (PSCs) to cancer-associated fibroblasts (CAFs) is responsible for the extensive desmoplastic reaction observed in PDAC stroma: a key driver of pancreatic ductal adenocarcinoma (PDAC) chemoresistance leading to poor prognosis. Specialized pro-resolving mediators (SPMs) are prime modulators of inflammation and its resolution, traditionally thought to be produced by immune cells. Using liquid chromatography-tandem mass spectrometry (LC-MS/MS)-based lipid mediator profiling PSCs as well as primary human CAFs express enzymes and receptors to produce and respond to SPMs. Human PSC/CAF SPM secretion profile can be modulated by rendering these cells activated [transforming growth factor beta (TGF-β)] or quiescent [all-trans retinoic acid (ATRA)]. ATRA-induced nuclear translocation of arachidonate-15-lipoxygenase (ALOX15) was linked to increased production of n-3 docosapentaenoic acid-derived Resolvin D5 (RvD5n-3 DPA), among other SPMs. Inhibition of RvD5n-3 DPA formation increases cancer cell invasion, whereas addback of this molecule reduced activated PSC-mediated cancer cell invasion. We also observed that circulating concentrations of RvD5n-3 DPA levels were decreased in peripheral blood of metastatic PDAC patients when compared with those measured in plasma of non-metastatic PDAC patients. Together, these findings indicate that RvD5n-3 DPA may regulate cancer-stroma cross-talk and invasion.
Collapse
Affiliation(s)
- Gabriel A. Aguirre
- Centre for Tumour Biology, Barts Cancer Institute, London, United Kingdom
| | | | | | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, John Vane Science Centre, Queen Mary University of London, London, United Kingdom
| | - Hemant M. Kocher
- Centre for Tumour Biology, Barts Cancer Institute, London, United Kingdom
| |
Collapse
|
107
|
Hog P, Kuntschar S, Rappl P, Huard A, Weigert A, Brüne B, Schmid T. Prostaglandin E 2 Boosts the Hyaluronan-Mediated Increase in Inflammatory Response to Lipopolysaccharide by Enhancing Lyve1 Expression. BIOLOGY 2023; 12:1441. [PMID: 37998039 PMCID: PMC10669677 DOI: 10.3390/biology12111441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/09/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023]
Abstract
Macrophages are a highly versatile and heterogenic group of immune cells, known for their involvement in inflammatory reactions. However, our knowledge about distinct subpopulations of macrophages and their specific contribution to the resolution of inflammation remains incomplete. We have previously shown, in an in vivo peritonitis model, that inhibition of the synthesis of the pro-inflammatory lipid mediator prostaglandin E2 (PGE2) attenuates efficient resolution of inflammation. PGE2 levels during later stages of the inflammatory process further correlate with expression of the hyaluronan (HA) receptor Lyve1 in peritoneal macrophages. In the present study, we therefore aimed to understand if PGE2 might contribute to the regulation of Lyve1 and how this might impact inflammatory responses. In line with our in vivo findings, PGE2 synergized with dexamethasone to enhance Lyve1 expression in bone marrow-derived macrophages, while expression of the predominant hyaluronan receptor CD44 remained unaltered. PGE2-mediated Lyve1 upregulation was strictly dependent on PGE2 receptor EP2 signaling. While PGE2/dexamethasone-treated macrophages, despite their enhanced Lyve1 expression, did not show inflammatory responses upon stimulation with low (LMW) or high-molecular-weight hyaluronan (HMW)-HA, they were sensitized towards LMW-HA-dependent augmentation of lipopolysaccharide (LPS)-induced inflammatory responses. Thus, Lyve1-expressing macrophages emerged as a subpopulation of macrophages integrating inflammatory stimuli with extracellular matrix-derived signals.
Collapse
Affiliation(s)
- Pauline Hog
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Silvia Kuntschar
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Peter Rappl
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Arnaud Huard
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe University Frankfurt, 60596 Frankfurt, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology, 60596 Frankfurt, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe University Frankfurt, 60590 Frankfurt, Germany
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
| |
Collapse
|
108
|
Liu Q, Li L, Qin W, Chao T. Repurposing drugs for solid tumor treatment: focus on immune checkpoint inhibitors. Cancer Biol Med 2023; 20:j.issn.2095-3941.2023.0281. [PMID: 37929901 PMCID: PMC10690875 DOI: 10.20892/j.issn.2095-3941.2023.0281] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023] Open
Abstract
Cancer remains a significant global health challenge with limited treatment options beyond systemic therapies, such as chemotherapy, radiotherapy, and molecular targeted therapy. Immunotherapy has emerged as a promising therapeutic modality but the efficacy has plateaued, which therefore provides limited benefits to patients with cancer. Identification of more effective approaches to improve patient outcomes and extend survival are urgently needed. Drug repurposing has emerged as an attractive strategy for drug development and has recently garnered considerable interest. This review comprehensively analyses the efficacy of various repurposed drugs, such as transforming growth factor-beta (TGF-β) inhibitors, metformin, receptor activator of nuclear factor-κB ligand (RANKL) inhibitors, granulocyte macrophage colony-stimulating factor (GM-CSF), thymosin α1 (Tα1), aspirin, and bisphosphonate, in tumorigenesis with a specific focus on their impact on tumor immunology and immunotherapy. Additionally, we present a concise overview of the current preclinical and clinical studies investigating the potential therapeutic synergies achieved by combining these agents with immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Qingxu Liu
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Long Li
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Wan Qin
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Tengfei Chao
- Department of Oncology, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
109
|
Krangvichian P, Techawiwattanaboon T, Palaga T, Ritprajak P, Kueanjinda P, Kaewraemruaen C, Patarakul K. Impaired functions of human monocyte-derived dendritic cells and induction of regulatory T cells by pathogenic Leptospira. PLoS Negl Trop Dis 2023; 17:e0011781. [PMID: 37983293 PMCID: PMC10695387 DOI: 10.1371/journal.pntd.0011781] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 12/04/2023] [Accepted: 11/08/2023] [Indexed: 11/22/2023] Open
Abstract
Leptospirosis is a global zoonosis caused by pathogenic Leptospira. The disease outcome is influenced by the interplay between innate and adaptive immune responses. Dendritic cells (DCs) play a crucial role in shaping the adaptive immune response. A recent study revealed that pathogenic Leptospira limited the activation of human monocyte-derived dendritic cells (MoDCs) compared to non-pathogenic Leptospira, but their impact on T-cell responses has not been investigated. Our study is the first to explore how viable pathogenic and non-pathogenic Leptospira affect the interaction between human MoDCs and T cells. We found that MoDCs infected with pathogenic leptospires (L. interrogans serovar Pomona and a clinical isolate, MoDCs-P) exhibited lower levels of CD80 and CD83 expression, suggesting partially impaired MoDC maturation, induced regulatory T cells (Tregs) while failing to induce CD4+ T cell proliferation, compared to MoDCs infected with non-pathogenic leptospires (L. biflexa serovar Patoc and L. meyeri serovar Ranarum, MoDCs-NP). In contrast, non-pathogenic leptospires enhanced MoDC maturation and induced higher T cell proliferation including IFN-γ-producing CD4+ T cells, indicative of a Th1-type response. Furthermore, pathogenic leptospires induced higher MoDC apoptosis through a cysteine aspartic acid-specific protease-3 (caspase-3)-dependent pathway and upregulated expression of the prostaglandin-endoperoxide synthase 2 (PTGS2) gene. Notably, prostaglandin E2 (PGE2), a product of the PTGS2 pathway, was found at higher levels in the sera of patients with acute leptospirosis and in the supernatant of MoDCs-P, possibly contributing to Treg induction, compared to those of healthy donors and MoDCs-NP, respectively. In conclusion, this study reveals a novel immunosuppressive strategy employed by pathogenic Leptospira to evade host immunity by partially impairing MoDC maturation and inducing Tregs. These findings deepen our understanding of leptospirosis pathogenesis in humans and may provide a novel strategy to modulate DCs for the prevention and treatment of the disease.
Collapse
Affiliation(s)
- Pratomporn Krangvichian
- Medical Microbiology, Interdisciplinary Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
| | - Teerasit Techawiwattanaboon
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
| | - Tanapat Palaga
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Patcharee Ritprajak
- Research Unit in Integrative Immuno-Microbial Biochemistry and Bioresponsive Nanomaterials, Department of Microbiology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Patipark Kueanjinda
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Chamraj Kaewraemruaen
- Department of Science and Bioinnovation, Faculty of Liberal Arts and Science, Kasetsart University, Kamphaeng Saen Campus, Nakhon Pathom, Thailand
| | - Kanitha Patarakul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Bangkok, Thailand
- Chula Vaccine Research Center (Chula VRC), Center of Excellence in Vaccine Research and Development, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
110
|
Ruiz-Cantero MC, Huerta MÁ, Tejada MÁ, Santos-Caballero M, Fernández-Segura E, Cañizares FJ, Entrena JM, Baeyens JM, Cobos EJ. Sigma-1 receptor agonism exacerbates immune-driven nociception: Role of TRPV1 + nociceptors. Biomed Pharmacother 2023; 167:115534. [PMID: 37729726 DOI: 10.1016/j.biopha.2023.115534] [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: 06/21/2023] [Revised: 09/14/2023] [Accepted: 09/17/2023] [Indexed: 09/22/2023] Open
Abstract
The analgesic effects of sigma-1 antagonists are undisputed, but the effects of sigma-1 agonists on pain are not well studied. Here, we used a mouse model to show that the administration of the sigma-1 agonists dextromethorphan (a widely used antitussive drug), PRE-084 (a standard sigma-1 ligand), and pridopidine (a selective drug being investigated in clinical trials for the treatment of neurodegenerative diseases) enhances PGE2-induced mechanical hyperalgesia. Superficial plantar incision induced transient weight-bearing asymmetry at early time points, but the mice appeared to recover at 24 h, despite noticeable edema and infiltration of neutrophils (a well-known cellular source of PGE2) at the injured site. Sigma-1 agonists induced a relapse of weight bearing asymmetry in a manner dependent on the presence of neutrophils. The effects of sigma-1 agonists were all reversed by administration of the sigma-1 antagonist BD-1063 in wild-type mice, and were absent in sigma-1 knockout mice, supporting the selectivity of the effects observed. The proalgesic effects of sigma-1 agonism were also abolished by the TRP antagonist ruthenium red and by in vivo resiniferatoxin ablation of TRPV1 + peripheral sensory neurons. Therefore, sigma-1 agonism exacerbates pain-like responses in mice with a mild inflammatory state through the action of TRPV1 + nociceptors. We also show that sigma-1 receptors are present in most (if not all) mouse and human DRG neurons. If our findings translate to humans, further studies will be needed to investigate potential proalgesic effects induced by sigma-1 agonism in patients treated with sigma-1 agonists.
Collapse
Affiliation(s)
- M Carmen Ruiz-Cantero
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miguel Á Huerta
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miguel Á Tejada
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Miriam Santos-Caballero
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Eduardo Fernández-Segura
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Histology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - Francisco J Cañizares
- Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Department of Histology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
| | - José M Entrena
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - José M Baeyens
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain
| | - Enrique J Cobos
- Department of Pharmacology, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Institute of Neuroscience, Biomedical Research Center, University of Granada, Armilla, 18100 Granada, Spain; Biosanitary Research Institute ibs.GRANADA, 18012 Granada, Spain; Teófilo Hernando Institute for Drug Discovery, 28029 Madrid, Spain.
| |
Collapse
|
111
|
Boni C, Rossi M, Montali I, Tiezzi C, Vecchi A, Penna A, Doselli S, Reverberi V, Ceccatelli Berti C, Montali A, Schivazappa S, Laccabue D, Missale G, Fisicaro P. What Is the Current Status of Hepatitis B Virus Viro-Immunology? Clin Liver Dis 2023; 27:819-836. [PMID: 37778772 DOI: 10.1016/j.cld.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2023]
Abstract
The natural history of hepatitis B virus (HBV) infection is closely dependent on the dynamic interplay between the host immune response and viral replication. Spontaneous HBV clearance in acute self-limited infection is the result of an adequate and efficient antiviral immune response. Instead, it is widely recognized that in chronic HBV infection, immunologic dysfunction contributes to viral persistence. Long-lasting exposure to high viral antigens, upregulation of multiple co-inhibitory receptors, dysfunctional intracellular signaling pathways and metabolic alterations, and intrahepatic regulatory mechanisms have been described as features ultimately leading to a hierarchical loss of effector functions up to full T-cell exhaustion.
Collapse
Affiliation(s)
- Carolina Boni
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy.
| | - Marzia Rossi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Ilaria Montali
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Camilla Tiezzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Andrea Vecchi
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Amalia Penna
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Sara Doselli
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Valentina Reverberi
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | | | - Anna Montali
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Simona Schivazappa
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy
| | - Diletta Laccabue
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Gabriele Missale
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy; Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Paola Fisicaro
- Unit of Infectious Diseases and Hepatology, Azienda Ospedaliero-Universitaria di Parma, Parma, Italy.
| |
Collapse
|
112
|
Geurts F, Xue L, Kramers BJ, Zietse R, Gansevoort RT, Fenton RA, Meijer E, Salih M, Hoorn EJ. Prostaglandin E2, Osmoregulation, and Disease Progression in Autosomal Dominant Polycystic Kidney Disease. Clin J Am Soc Nephrol 2023; 18:1426-1434. [PMID: 37574650 PMCID: PMC10637469 DOI: 10.2215/cjn.0000000000000269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 08/06/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND Prostaglandin E2 (PGE2) plays a physiological role in osmoregulation, a process that is affected early in autosomal dominant polycystic kidney disease (ADPKD). PGE2 has also been implicated in the pathogenesis of ADPKD in preclinical models, but human data are limited. Here, we hypothesized that urinary PGE2 excretion is associated with impaired osmoregulation, disease severity, and disease progression in human ADPKD. METHODS Urinary excretions of PGE2 and its metabolite (PGEM) were measured in a prospective cohort of patients with ADPKD. The associations between urinary PGE2 and PGEM excretions, markers of osmoregulation, eGFR and height-adjusted total kidney volume were assessed using linear regression models. Cox regression and linear mixed models were used for the longitudinal analysis of the associations between urinary PGE2 and PGEM excretions and disease progression defined as 40% eGFR loss or kidney failure, and change in eGFR over time. In two intervention studies, we quantified the effect of starting tolvaptan and adding hydrochlorothiazide to tolvaptan on urinary PGE2 and PGEM excretions. RESULTS In 562 patients with ADPKD (61% female, eGFR 63±28 ml/min per 1.73 m 2 ), higher urinary PGE2 or PGEM excretions were independently associated with higher plasma copeptin, lower urine osmolality, lower eGFR, and greater total kidney volume. Participants with higher baseline urinary PGE2 and PGEM excretions had a higher risk of 40% eGFR loss or kidney failure (hazard ratio, 1.28; 95% confidence interval [CI], 1.13 to 1.46 and hazard ratio, 1.50; 95% CI, 1.26 to 1.80 per two-fold higher urinary PGE2 or PGEM excretions) and a faster change in eGFR over time (-0.39 [95% CI, -0.59 to -0.20] and -0.53 [95% CI, -0.75 to -0.31] ml/min per 1.73 m 2 per year). In the intervention studies, urinary PGEM excretion was higher after starting tolvaptan, while urinary PGE2 excretion was higher after adding hydrochlorothiazide to tolvaptan. CONCLUSIONS Higher urinary PGE2 and PGEM excretions in patients with ADPKD are associated with impaired osmoregulation, disease severity, and progression.
Collapse
Affiliation(s)
- Frank Geurts
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Laixi Xue
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Bart J. Kramers
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Robert Zietse
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ron T. Gansevoort
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | | | - Esther Meijer
- Department of Internal Medicine, Division of Nephrology, University Medical Center Groningen, Groningen, The Netherlands
| | - Mahdi Salih
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Ewout J. Hoorn
- Department of Internal Medicine, Division of Nephrology and Transplantation, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| |
Collapse
|
113
|
Marseglia GL, Ciprandi G. Clinical use of ketoprofen lysine salt: a reappraisal in adolescents with acute respiratory infections. Allergol Immunopathol (Madr) 2023; 51:76-82. [PMID: 37937499 DOI: 10.15586/aei.v51i6.918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/23/2023] [Indexed: 11/09/2023]
Abstract
Upper respiratory infections are widespread, and they are mainly of viral etiology. It has to be remarked that every infection is always associated with an inflammatory response. Inflammation implicates a cascade of bothersome symptoms, including fever, pain (headache, myalgia, and arthralgia), malaise, and respiratory complaints. As a result, anti-inflammatory medications could be beneficial as they act on different pathogenetic pathways. The ketoprofen lysine salt (KLS) has a potent anti-inflammatory activity associated with effective analgesic and antipyretic effects and has a valuable safety profile. However, adolescents present peculiar psychological characteristics that determine their difficulty to be managed. In this regard, an adolescent with a respiratory infection requires a prompt and adequate cure. KLS, thanks to its pharmacologic profile, could be favorably used in this regard. A recent primary-care experience outlined its effectiveness in this issue.
Collapse
Affiliation(s)
- Gian Luigi Marseglia
- Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy;
| | | |
Collapse
|
114
|
Hou CH, Chen PC, Liu JF. CXCL1 enhances COX-II expression in rheumatoid arthritis synovial fibroblasts by CXCR2, PLC, PKC, and NF-κB signal pathway. Int Immunopharmacol 2023; 124:110909. [PMID: 37722260 DOI: 10.1016/j.intimp.2023.110909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/26/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023]
Abstract
Rheumatoid arthritis (RA) is the most common autoimmune disease, affecting the joints of the hands and feet. Several chemokines and their receptors are crucial in RA pathogenesis through immune cell recruitment. C-X-C Motif Chemokine Ligand 1 (CXCL1), a chemokine for the recruitment of various immune cells, can be upregulated in patients with RA. However, the discussion on the role of CXCL1 in RA pathogenesis is insufficient. Here, we found that CXCL1 promoted cyclooxygenase-2 (COX-II) expression in a dose- and time-dependent manner in rheumatoid arthritis synovial fibroblasts (RASFs). CXCL1 overexpression in RASFs led to a significant increase in COX-II expression, while the transfection of RASFs with the shRNA plasmid resulted in a noticeable decrease in COX-II expression. Next, we delineated the molecular mechanism underlying CXCL1-promoted COX-II expression and noted that CXC chemokine receptor 2 (CXCR2), phospholipase C (PLC), and protein kinase C (PKC) signal transduction were responsible for COX-II expression after CXCL1 incubation for RASFs. Finally, we confirmed the transcriptional activation of nuclear factor κB (NF-κB) in RASFs after incubation with CXCL1. In conclusion, the current study provided a novel insight into the role of CXCL1 in RA pathogenesis.
Collapse
Affiliation(s)
- Chun-Han Hou
- Department of Orthopedic Surgery, National Taiwan University Hospital, Taipei City, Taiwan
| | - Po-Chun Chen
- School of Life Science, National Taiwan Normal University, Taipei, Taiwan; Translational Medicine Center, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei City, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan.
| | - Ju-Fang Liu
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan; School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
115
|
Schlicher L, Green LG, Romagnani A, Renner F. Small molecule inhibitors for cancer immunotherapy and associated biomarkers - the current status. Front Immunol 2023; 14:1297175. [PMID: 38022587 PMCID: PMC10644399 DOI: 10.3389/fimmu.2023.1297175] [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: 09/19/2023] [Accepted: 10/17/2023] [Indexed: 12/01/2023] Open
Abstract
Following the success of cancer immunotherapy using large molecules against immune checkpoint inhibitors, the concept of using small molecules to interfere with intracellular negative regulators of anti-tumor immune responses has emerged in recent years. The main targets for small molecule drugs currently include enzymes of negative feedback loops in signaling pathways of immune cells and proteins that promote immunosuppressive signals within the tumor microenvironment. In the adaptive immune system, negative regulators of T cell receptor signaling (MAP4K1, DGKα/ζ, CBL-B, PTPN2, PTPN22, SHP1), co-receptor signaling (CBL-B) and cytokine signaling (PTPN2) have been preclinically validated as promising targets and initial clinical trials with small molecule inhibitors are underway. To enhance innate anti-tumor immune responses, inhibitory immunomodulation of cGAS/STING has been in the focus, and inhibitors of ENPP1 and TREX1 have reached the clinic. In addition, immunosuppressive signals via adenosine can be counteracted by CD39 and CD73 inhibition, while suppression via intratumoral immunosuppressive prostaglandin E can be targeted by EP2/EP4 antagonists. Here, we present the status of the most promising small molecule drug candidates for cancer immunotherapy, all residing relatively early in development, and the potential of relevant biomarkers.
Collapse
Affiliation(s)
- Lisa Schlicher
- Cancer Cell Targeted Therapy, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Luke G. Green
- Therapeutic Modalities, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Andrea Romagnani
- Cancer Cell Targeted Therapy, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Florian Renner
- Cancer Cell Targeted Therapy, Roche Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| |
Collapse
|
116
|
Hossain F, Ucar DA, Monticone G, Ran Y, Majumder S, Larter K, Luu H, Wyczechowska D, Heidari S, Xu K, Shanthalingam S, Matossian M, Xi Y, Burow M, Collins-Burow B, Del Valle L, Hicks C, Zabaleta J, Golde T, Osborne B, Miele L. Sulindac sulfide as a non-immune suppressive γ-secretase modulator to target triple-negative breast cancer. Front Immunol 2023; 14:1244159. [PMID: 37901240 PMCID: PMC10612326 DOI: 10.3389/fimmu.2023.1244159] [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: 06/21/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Triple-negative breast cancer (TNBC) comprises a heterogeneous group of clinically aggressive tumors with high risk of recurrence and metastasis. Current pharmacological treatment options remain largely limited to chemotherapy. Despite promising results, the efficacy of immunotherapy and chemo-immunotherapy in TNBC remains limited. There is strong evidence supporting the involvement of Notch signaling in TNBC progression. Expression of Notch1 and its ligand Jagged1 correlate with poor prognosis. Notch inhibitors, including g-secretase inhibitors (GSIs), are quite effective in preclinical models of TNBC. However, the success of GSIs in clinical trials has been limited by their intestinal toxicity and potential for adverse immunological effects, since Notch plays key roles in T-cell activation, including CD8 T-cells in tumors. Our overarching goal is to replace GSIs with agents that lack their systemic toxicity and ideally, do not affect tumor immunity. We identified sulindac sulfide (SS), the active metabolite of FDA-approved NSAID sulindac, as a potential candidate to replace GSIs. Methods We investigated the pharmacological and immunotherapeutic properties of SS in TNBC models in vitro, ex-vivo and in vivo. Results We confirmed that SS, a known γ-secretase modulator (GSM), inhibits Notch1 cleavage in TNBC cells. SS significantly inhibited mammosphere growth in all human and murine TNBC models tested. In a transplantable mouse TNBC tumor model (C0321), SS had remarkable single-agent anti-tumor activity and eliminated Notch1 protein expression in tumors. Importantly, SS did not inhibit Notch cleavage in T- cells, and the anti-tumor effects of SS were significantly enhanced when combined with a-PD1 immunotherapy in our TNBC organoids and in vivo. Discussion Our data support further investigation of SS for the treatment of TNBC, in conjunction with chemo- or -chemo-immunotherapy. Repurposing an FDA-approved, safe agent for the treatment of TNBC may be a cost-effective, rapidly deployable therapeutic option for a patient population in need of more effective therapies.
Collapse
Affiliation(s)
- Fokhrul Hossain
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Deniz A. Ucar
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Giulia Monticone
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Yong Ran
- Department of Pharmacological and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Samarpan Majumder
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Kristina Larter
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Hanh Luu
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Dorota Wyczechowska
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
| | - Soroor Heidari
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Keli Xu
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS, United States
| | - Sudarvili Shanthalingam
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | | | - Yaguang Xi
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Matthew Burow
- School of Medicine, Tulane University, New Orleans, LA, United States
| | | | - Luis Del Valle
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
- Department of Pathology, Louisiana State University Health Sciences Center - New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Chindo Hicks
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology, LSUHSC-NO, New Orleans, LA, United States
| | - Todd Golde
- Department of Pharmacological and Chemical Biology, Emory University, Atlanta, GA, United States
| | - Barbara Osborne
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Lucio Miele
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans (LSUHSC-NO), New Orleans, LA, United States
| |
Collapse
|
117
|
Zheng Y, Sun L, Guo J, Ma J. The crosstalk between ferroptosis and anti-tumor immunity in the tumor microenvironment: molecular mechanisms and therapeutic controversy. Cancer Commun (Lond) 2023; 43:1071-1096. [PMID: 37718480 PMCID: PMC10565387 DOI: 10.1002/cac2.12487] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 08/13/2023] [Accepted: 09/11/2023] [Indexed: 09/19/2023] Open
Abstract
The advent of immunotherapy has significantly reshaped the landscape of cancer treatment, greatly enhancing therapeutic outcomes for multiple types of cancer. However, only a small subset of individuals respond to it, underscoring the urgent need for new methods to improve its response rate. Ferroptosis, a recently discovered form of programmed cell death, has emerged as a promising approach for anti-tumor therapy, with targeting ferroptosis to kill tumors seen as a potentially effective strategy. Numerous studies suggest that inducing ferroptosis can synergistically enhance the effects of immunotherapy, paving the way for a promising combined treatment method in the future. Nevertheless, recent research has raised concerns about the potential negative impacts on anti-tumor immunity as a consequence of inducing ferroptosis, leading to conflicting views within the scientific community about the interplay between ferroptosis and anti-tumor immunity, thereby underscoring the necessity of a comprehensive review of the existing literature on this relationship. Previous reviews on ferroptosis have touched on related content, many focusing primarily on the promoting role of ferroptosis on anti-tumor immunity while overlooking recent evidence on the inhibitory effects of ferroptosis on immunity. Others have concentrated solely on discussing related content either from the perspective of cancer cells and ferroptosis or from immune cells and ferroptosis. Given that both cancer cells and immune cells exist in the tumor microenvironment, a one-sided discussion cannot comprehensively summarize this topic. Therefore, from the perspectives of both tumor cells and tumor-infiltrating immune cells, we systematically summarize the current conflicting views on the interplay between ferroptosis and anti-tumor immunity, intending to provide potential explanations and identify the work needed to establish a translational basis for combined ferroptosis-targeted therapy and immunotherapy in treating tumors.
Collapse
Affiliation(s)
- Yichen Zheng
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Lingqi Sun
- Department of NeurologyAir Force Hospital of the Western Theater of the Chinese People's Liberation ArmyChengduSichuanP. R. China
| | - Jiamin Guo
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| | - Ji Ma
- Division of Abdominal Tumor Multimodality TreatmentCancer CenterWest China HospitalSichuan UniversityChengduSichuanP. R. China
| |
Collapse
|
118
|
Zhang B, Zhu G, Liu J, Zhang C, Yao K, Huang X, Cen X, Zhao Z. Single-cell transcriptional profiling reveals immunomodulatory properties of stromal and epithelial cells in periodontal immune milieu with diabetes in rats. Int Immunopharmacol 2023; 123:110715. [PMID: 37562294 DOI: 10.1016/j.intimp.2023.110715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 07/01/2023] [Accepted: 07/24/2023] [Indexed: 08/12/2023]
Abstract
Periodontitis is the sixth major complication of diabetes. Gingiva, as an important component of periodontal tissues, serves as the first defense barrier against infectious stimuli. However, relatively little is known about cellular heterogeneity and cell-specific changes in gingiva in response to diabetes-associated periodontitis. To characterize molecular changes linking diabetes with periodontitis, we profiled single-cell transcriptome analyses of a total of 45,259 cells from rat gingiva with periodontitis under normoglycemic and diabetic condition. The single-cell profiling revealed that stromal and epithelial cells of gingiva contained inflammation-related subclusters enriched in functions of immune cell recruitment. Compared to normoglycemic condition, diabetes led to a reduction in epithelial basal cells, fibroblasts and smooth muscle cells in gingiva with periodontitis. Analysis of differentially expressed genes indicated that stromal and epithelial populations were reprogrammed towards pro-inflammatory phenotypes promoting immune cell recruitment in diabetes-related periodontitis. In aspect of immune cells, diabetes prominently enhanced neutrophil and M1 macrophage infiltration in periodontitis lesions. Cell-cell communications revealed enhanced crosstalk between stromal/epithelial cells and immune cells mediating by chemokine/chemokine receptor interplay in diabetes-associated periodontitis. Our findings deconvolved cellular heterogeneity of rat gingiva associated with periodontitis and diabetes, uncovered altered immune milieu caused by the disease, and revealed immunomodulatory functions of stromal and epithelial cells in gingival immune niche. The present study improves the understanding of the link between the diabetes and periodontitis and helps in formulating precise therapeutic strategies for diabetes-enhanced periodontitis.
Collapse
Affiliation(s)
- Bo Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Guanyin Zhu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Junqi Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chenghao Zhang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Ke Yao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xinqi Huang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xiao Cen
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Zhihe Zhao
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
119
|
Hou X, Shi H, Jiang Y, Li X, Chen K, Li Q, Liu R. Transcriptome analysis reveals the neuroactive receptor genes response to Streptococcus agalactiae infection in tilapia, Oreochromis niloticus. FISH & SHELLFISH IMMUNOLOGY 2023; 141:109090. [PMID: 37722443 DOI: 10.1016/j.fsi.2023.109090] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 09/13/2023] [Accepted: 09/16/2023] [Indexed: 09/20/2023]
Abstract
The detailed crosstalk between the neuroendocrine and immune systems in Oreochromis niloticus, an economically important fish, in response to pathogenic infections, remains unclear. This study revealed the head kidney transcriptional profiles of O. niloticus upon infections with Streptococcus agalactiae, a prevalent pathogen known to cause severe meningitis. Twelve cDNA libraries of O. niloticus head kidney, representing four treatment time points (0, 6, 24, and 48 h), were constructed and a total of 2,528 differentially expressed genes were identified based on pairwise comparisons. KEGG pathway analysis revealed a significant enrichment of the 'neuroactive ligand-receptor interaction' pathway (ko04080), with 13 genes exhibiting differential expression during S. agalactiae infection. Among these, six neuroactive receptor genes (lepr, nr3c1, ptger4, thrb, tspo, and β2-ar) were selected, cloned, and characterized. Although these genes are ubiquitously expressed, and in head kidney leukocytes, their expression was mainly observed in T cells, Mo/Mφ, and NCCs, which are characterized by antimicrobial responses. Furthermore, we examined the response patterns of these six neuroactive receptor genes to gram-positive (S. agalactiae) and gram-negative (Aeromonas hydrophila) bacteria in four different tissues. Notably, lepr, ptger4, tspo, and β2-ar were upregulated in all selected tissues in response to S. agalactiae and A. hydrophila infections. However, nr3c1 and thrb were downregulated in response to S. agalactiae infection in the head kidney and spleen, whereas nr3c1 was upregulated, and thrb was unresponsive to A. hydrophila infection. Our findings provide a theoretical foundation for understanding new links between the neuroendocrine and immune systems during bacterial infection in teleost fish.
Collapse
Affiliation(s)
- Xitan Hou
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China.
| | - Haokai Shi
- College of Medical Engineering, Jining Medical University, Jining, China
| | - Yan Jiang
- Shandong Freshwater Fisheries Research Institute, Jinan, China
| | - Xiaoke Li
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
| | - Kaiqi Chen
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
| | - Qi Li
- Fisheries College, Jimei University, Xiamen, China.
| | - Ruonan Liu
- College of Medical Engineering, Jining Medical University, Jining, China.
| |
Collapse
|
120
|
Huh G, Oh Y, Jeon Y, Kang KS, Kim SN, Jung SH, Kim SH, Kim YJ. Insampaedok-San Extract Exerts an Immune-Enhancing Effect through NF- κB p65 Pathway Activation. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5458504. [PMID: 37780486 PMCID: PMC10541303 DOI: 10.1155/2023/5458504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/09/2023] [Accepted: 09/01/2023] [Indexed: 10/03/2023]
Abstract
Insampaedok-san (IS) has traditionally been prescribed as a medication for cold-related symptoms in Northeast Asia, including Korea and China. In this study, we focused on elucidating the molecular mechanism underlying the immunomodulatory activity of IS water extract (ISE) in macrophages. ISE significantly enhanced the levels of nitric oxide (NO) and prostaglandin E2 (PGE2) by increasing the expression of inducible NO synthase and cyclooxygenase-2 (COX-2) in a dose-dependent manner. ISE, which consists of many herbs, contains a large number of active compounds whose pharmacological targets and mechanisms are complicated. Therefore, network pharmacology analysis was used to predict the potential key components, targets, and mechanisms of ISE as immunomodulators. Subsequently, the network pharmacology results were validated experimentally. Seven key components were identified through HPLC-QTOF-MS. As predicted by the network pharmacology analysis, ISE increased the mRNA expression of Tnf and Il6. Furthermore, ISE increased the phosphorylation, nuclear translocation, and transcriptional activity of the p65 subunit of the nuclear factor-κB (NF-κB) signaling pathway. In contrast, rapamycin, an NF-κB inhibitor, suppressed the ISE-induced mRNA expression of Tnf and Il6. In conclusion, ISE is an immune activator that can elevate the production of NO, PGE2, and proinflammatory cytokines mediated by NF-κB signaling.
Collapse
Affiliation(s)
- Gyuwon Huh
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Youngse Oh
- College of Pharmacy, Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - Youngsic Jeon
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Ki Sung Kang
- College of Korean Medicine, Gachon University, Seongnam 13120, Republic of Korea
| | - Su Nam Kim
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| | - Sang Hoon Jung
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Seung Hyun Kim
- College of Pharmacy, Yonsei Institute of Pharmaceutical Science, Yonsei University, Incheon 21983, Republic of Korea
| | - Young-Joo Kim
- Natural Product Research Center, Korea Institute of Science and Technology, Gangneung 25451, Republic of Korea
| |
Collapse
|
121
|
Zhou Q, Fan M, Wang Y, Ma Y, Si H, Dai G. Association between Dietary Vitamin E Intake and Human Papillomavirus Infection among US Adults: A Cross-Sectional Study from National Health and Nutrition Examination Survey. Nutrients 2023; 15:3825. [PMID: 37686857 PMCID: PMC10490162 DOI: 10.3390/nu15173825] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 08/27/2023] [Accepted: 08/30/2023] [Indexed: 09/10/2023] Open
Abstract
Persistent high-risk human papillomavirus (HPV) infection is responsible for most genital, anal, and oropharyngeal cancers, in which men contribute significantly to infection and subsequent tumorigenesis in women. Vitamin E has been shown to be associated with vaginal HPV infection and cervical cancer. However, the association of vitamin E consumption with HPV infection among the overall population remains unclear. We investigate the association between vitamin E consumption and genital and oral HPV infection in both men and women. We used cross-sectional data from the National Health and Nutrition Examination Survey between 2013 and 2016 to collect details on their dietary vitamin E intake, genital and oral HPV infection status, and other essential variables. In total, 5809 participants aged 18-59 years were identified, with overall prevalence of high-risk and low-risk HPV infection of 23.7% and 21.1%, respectively. Compared with the lowest vitamin E group Q1 (<5.18 mg/day), the adjusted OR for vitamin E consumption and overall high-risk HPV infection in Q2 (5.18-7.54 mg/day), Q3 (7.55-10.82 mg/day), and Q4 (>10.82 mg/day) were 0.91 (95% CI: 0.81-1.03, p = 0.134), 0.77 (95% CI: 0.69-0.87, p < 0.001), and 0.72 (95% CI: 0.65-0.80, p < 0.001), respectively. Restricted cubic spline regression showed a linear relationship between vitamin E consumption and overall high-risk HPV infection. This linear relationship also existed for vitamin E consumption and overall low-risk HPV infection. After being stratified by gender and site, vitamin E consumption was inversely related to vaginal low- and high-risk HPV infection, penile high-risk HPV infection, and male oral low-risk HPV infection. In conclusion, we identified inverse linear relationships between dietary vitamin E intake and overall high- and low-risk HPV infection. Future well-designed longitudinal studies are still required to validate the impact of vitamin E on HPV carcinogenesis.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Oncology, Medical School of Chinese People’s Liberation Army, Beijing 100853, China; (Q.Z.); (M.F.); (Y.W.); (Y.M.)
- The Fifth Medical Center, Department of Medical Oncology, Chinese People’s Liberation Army General Hospital, Beijing 100853, China;
| | - Mengjiao Fan
- Department of Oncology, Medical School of Chinese People’s Liberation Army, Beijing 100853, China; (Q.Z.); (M.F.); (Y.W.); (Y.M.)
- The Fifth Medical Center, Department of Medical Oncology, Chinese People’s Liberation Army General Hospital, Beijing 100853, China;
| | - Yanrong Wang
- Department of Oncology, Medical School of Chinese People’s Liberation Army, Beijing 100853, China; (Q.Z.); (M.F.); (Y.W.); (Y.M.)
- The Fifth Medical Center, Department of Medical Oncology, Chinese People’s Liberation Army General Hospital, Beijing 100853, China;
| | - Yue Ma
- Department of Oncology, Medical School of Chinese People’s Liberation Army, Beijing 100853, China; (Q.Z.); (M.F.); (Y.W.); (Y.M.)
- The Fifth Medical Center, Department of Medical Oncology, Chinese People’s Liberation Army General Hospital, Beijing 100853, China;
| | - Haiyan Si
- The Fifth Medical Center, Department of Medical Oncology, Chinese People’s Liberation Army General Hospital, Beijing 100853, China;
| | - Guanghai Dai
- The Fifth Medical Center, Department of Medical Oncology, Chinese People’s Liberation Army General Hospital, Beijing 100853, China;
| |
Collapse
|
122
|
Villain E, Chanson A, Mainka M, Kampschulte N, Le Faouder P, Bertrand-Michel J, Brandolini-Bulon M, Charbit B, Musvosvi M, Bilek N, Scriba TJ, Quintana-Murci L, Schebb NH, Duffy D, Gladine C. Integrated analysis of whole blood oxylipin and cytokine responses after bacterial, viral, and T cell stimulation reveals new immune networks. iScience 2023; 26:107422. [PMID: 37575177 PMCID: PMC10415927 DOI: 10.1016/j.isci.2023.107422] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/24/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Oxylipins are major immunomodulating mediators, yet studies of inflammation focus mainly on cytokines. Here, using a standardized whole-blood stimulation system, we characterized the oxylipin-driven inflammatory responses to various stimuli and their relationships with cytokine responses. We performed a pilot study in 25 healthy individuals using 6 different stimuli: 2 bacterial stimuli (LPS and live BCG), 2 viral stimuli (vaccine-grade poly I:C and live H1N1 attenuated influenza), an enterotoxin superantigen and a Null control. All stimuli induced a strong production of oxylipins but most importantly, bacterial, viral, and T cell immune responses show distinct oxylipin signatures. Integration of the oxylipin and cytokine responses for each condition revealed new immune networks improving our understanding of inflammation regulation. Finally, the oxylipin responses and oxylipin-cytokine networks were compared in patients with active tuberculosis or with latent infection. This revealed different responses to BCG but not LPS stimulation highlighting new regulatory pathways for further investigations.
Collapse
Affiliation(s)
- Etienne Villain
- Institut Pasteur, Université Paris Cité, Translational Immunology Unit, Paris, France
| | - Aurélie Chanson
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| | - Malwina Mainka
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Nadja Kampschulte
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Pauline Le Faouder
- MetaToul, MetaboHUB, Inserm/UPS UMR 1048-I2MC, Institut des Maladies Métaboliques et Cardiovasculaires, 31400 Toulouse, France
| | - Justine Bertrand-Michel
- MetaToul, MetaboHUB, Inserm/UPS UMR 1048-I2MC, Institut des Maladies Métaboliques et Cardiovasculaires, 31400 Toulouse, France
| | - Marion Brandolini-Bulon
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
- Université Clermont Auvergne, INRAE, UNH, Plateforme D’Exploration Du Métabolisme, MetaboHUB Clermont, Clermont-Ferrand, France
| | - Bruno Charbit
- Institut Pasteur, Université Paris Cité, CBUTechS, Paris, France
| | - Munyaradzi Musvosvi
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Nicole Bilek
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Thomas J. Scriba
- South African Tuberculosis Vaccine Initiative (SATVI), Division of Immunology, Department of Pathology and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Lluis Quintana-Murci
- Institut Pasteur, Université Paris Cité, CNRS UMR2000, Human Evolutionary Genetics Unit, Paris, France
- Collège de France, 75005 Paris, France
| | - Nils Helge Schebb
- Chair of Food Chemistry, Faculty of Mathematics and Natural Sciences, University of Wuppertal, Wuppertal, Germany
| | - Darragh Duffy
- Institut Pasteur, Université Paris Cité, Translational Immunology Unit, Paris, France
- Institut Pasteur, Université Paris Cité, CBUTechS, Paris, France
| | - Cécile Gladine
- Université Clermont Auvergne, INRAE, UNH, Clermont-Ferrand, France
| |
Collapse
|
123
|
Gutierrez-Hoffmann M, Fan J, O’Meally RN, Cole RN, Florea L, Antonescu C, Talbot CC, Tiniakou E, Darrah E, Soloski MJ. The Interaction of Borrelia burgdorferi with Human Dendritic Cells: Functional Implications. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:612-625. [PMID: 37405694 PMCID: PMC10527078 DOI: 10.4049/jimmunol.2300235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 06/01/2023] [Indexed: 07/06/2023]
Abstract
Dendritic cells bridge the innate and adaptive immune responses by serving as sensors of infection and as the primary APCs responsible for the initiation of the T cell response against invading pathogens. The naive T cell activation requires the following three key signals to be delivered from dendritic cells: engagement of the TCR by peptide Ags bound to MHC molecules (signal 1), engagement of costimulatory molecules on both cell types (signal 2), and expression of polarizing cytokines (signal 3). Initial interactions between Borrelia burgdorferi, the causative agent of Lyme disease, and dendritic cells remain largely unexplored. To address this gap in knowledge, we cultured live B. burgdorferi with monocyte-derived dendritic cells (mo-DCs) from healthy donors to examine the bacterial immunopeptidome associated with HLA-DR. In parallel, we examined changes in the expression of key costimulatory and regulatory molecules as well as profiled the cytokines released by dendritic cells when exposed to live spirochetes. RNA-sequencing studies on B. burgdorferi-pulsed dendritic cells show a unique gene expression signature associated with B. burgdorferi stimulation that differs from stimulation with lipoteichoic acid, a TLR2 agonist. These studies revealed that exposure of mo-DCs to live B. burgdorferi drives the expression of both pro- and anti-inflammatory cytokines as well as immunoregulatory molecules (e.g., PD-L1, IDO1, Tim3). Collectively, these studies indicate that the interaction of live B. burgdorferi with mo-DCs promotes a unique mature DC phenotype that likely impacts the nature of the adaptive T cell response generated in human Lyme disease.
Collapse
Affiliation(s)
- Maria Gutierrez-Hoffmann
- Lyme Disease Research Center, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| | - Jinshui Fan
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| | - Robert N. O’Meally
- Mass Spectrometry and Proteomics Facility,
Department of Biological Chemistry, Johns Hopkins University School of Medicine,
Baltimore, MD 21205, USA
| | - Robert N. Cole
- Mass Spectrometry and Proteomics Facility,
Department of Biological Chemistry, Johns Hopkins University School of Medicine,
Baltimore, MD 21205, USA
| | - Liliana Florea
- Department of Genetic Medicine, Johns Hopkins
University, School of Medicine, Baltimore, MD 21205, USA
| | - Corina Antonescu
- Department of Genetic Medicine, Johns Hopkins
University, School of Medicine, Baltimore, MD 21205, USA
| | - C. Conover Talbot
- Institute for Basic Biomedical Sciences, Johns
Hopkins University, School of Medicine, Baltimore, MD 21205, USA
| | - Eleni Tiniakou
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| | - Erika Darrah
- Lyme Disease Research Center, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| | - Mark J. Soloski
- Lyme Disease Research Center, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
- Division of Rheumatology, Johns Hopkins University,
School of Medicine, Baltimore, MD 21224, USA
| |
Collapse
|
124
|
Cornet-Gomez A, Moreira LR, Gomez-Samblás M, Osuna A. Extracellular vesicles of Trypanosoma cruzi and immune complexes they form with sialylated and non-sialylated IgGs increase small peritoneal macrophage subpopulation and elicit different cytokines profiles. Front Immunol 2023; 14:1215913. [PMID: 37600828 PMCID: PMC10434529 DOI: 10.3389/fimmu.2023.1215913] [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: 05/02/2023] [Accepted: 07/17/2023] [Indexed: 08/22/2023] Open
Abstract
American trypanosomiasis, or Chagas disease, is caused by the protozoan parasite Trypanosoma cruzi and is characterized by the presence of cardiac or gastrointestinal symptoms in a large number of patients during the chronic phase of the disease. Although the origin of the symptoms is not clear, several mechanisms have been described involving factors related to T. cruzi and the host immune response. In this sense, the extracellular vesicles (EVs) secreted by the parasite and the immune complexes (ICs) formed after their recognition by host IgGs (EVs-IgGs) may play an important role in the immune response during infection. The aim of the present work is to elucidate the modulation of the immune response exerted by EVs and the ICs they form by analyzing the variation in the subpopulations of small and large peritoneal macrophages after intraperitoneal inoculation in mice and to evaluate the role of the sialylation of the host IgGs in this immunomodulation. Both macrophage subpopulations were purified and subjected to cytokine expression analysis by RT-qPCR. The results showed an increase in the small peritoneal macrophage subpopulation after intraperitoneal injection of parasite EVs, but a greater increase in this subpopulation was observed when sialylated and non-sialylated ICs were injected, which was similar to inoculation with the trypomastigote stage of the parasite. The cytokine expression results showed the ability of both subpopulations to express inflammatory and non-inflammatory cytokines. These results suggest the role of free EVs in the acute phase of the disease and the possible role of immune complexes in the immune response in the chronic phase of the disease, when the levels of antibodies against the parasite allow the formation of immune complexes. The differential expression of interleukins showed after the inoculation of immune complexes formed with sialylated and non-sialylated IgGs and the interleukins expression induced by EVs, demonstrates that the IgG glycosilation is involved in the type of immune response that dominates in each of the phases of the Chagas disease.
Collapse
Affiliation(s)
- Alberto Cornet-Gomez
- Grupo de Bioquímica y Parasitología Molecular (CTS 183), Departamento de Parasitología, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| | - Lissette Retana Moreira
- Grupo de Bioquímica y Parasitología Molecular (CTS 183), Departamento de Parasitología, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
- Departamento de Parasitología, Facultad de Microbiología, Universidad de Costa Rica, San José, Costa Rica
- Centro de Investigación en Enfermedades Tropicales (CIET), Universidad de Costa Rica, San José, Costa Rica
| | - Mercedes Gomez-Samblás
- Grupo de Bioquímica y Parasitología Molecular (CTS 183), Departamento de Parasitología, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| | - Antonio Osuna
- Grupo de Bioquímica y Parasitología Molecular (CTS 183), Departamento de Parasitología, Instituto de Biotecnología, Universidad de Granada, Granada, Spain
| |
Collapse
|
125
|
Wanjari UR, Gopalakrishnan AV. A review on immunological aspects in male reproduction: An immune cells and cytokines. J Reprod Immunol 2023; 158:103984. [PMID: 37390629 DOI: 10.1016/j.jri.2023.103984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/17/2023] [Accepted: 06/25/2023] [Indexed: 07/02/2023]
Abstract
The male reproductive system, particularly the male gamete, offers a unique barrier to the immune system. The growing germ cells in the testis need to be shielded from autoimmune damage. Hence the testis has to establish and sustain an immune-privileged milieu. Sertoli cells create this safe space, protected by the blood-testis barrier. Cytokines are a type of immune reaction that can positively and negatively affect male reproductive health. Inflammation, disease, and obesity are just a few physiological conditions for which cytokines mediate signals. They interact with steroidogenesis, shaping the adrenals and testes to produce the hormones needed for survival. In particular pathological condition, including autoimmune disorders, contains high levels of the same cytokines in semen that play an essential role in the immunomodulation of the male gonad. This review focuses on understanding the immunological role of cytokines in the control and development of male reproduction. Also, in maintaining male reproductive health and diseases linked with their aberrant function in the testis.
Collapse
Affiliation(s)
- Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Bio-Sciences and Technology, Vellore Institute of Technology (VIT), Vellore, Tamil Nadu 632014, India.
| |
Collapse
|
126
|
Francica BJ, Holtz A, Lopez J, Freund D, Chen A, Wang D, Powell D, Kipper F, Panigrahy D, Dubois RN, Whiting CC, Prasit P, Dubensky TW. Dual Blockade of EP2 and EP4 Signaling is Required for Optimal Immune Activation and Antitumor Activity Against Prostaglandin-Expressing Tumors. CANCER RESEARCH COMMUNICATIONS 2023; 3:1486-1500. [PMID: 37559947 PMCID: PMC10408683 DOI: 10.1158/2767-9764.crc-23-0249] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/08/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023]
Abstract
While the role of prostaglandin E2 (PGE2) in promoting malignant progression is well established, how to optimally block the activity of PGE2 signaling remains to be demonstrated. Clinical trials with prostaglandin pathway targeted agents have shown activity but without sufficient significance or dose-limiting toxicities that have prevented approval. PGE2 signals through four receptors (EP1-4) to modulate tumor progression. EP2 and EP4 signaling exacerbates tumor pathology and is immunosuppressive through potentiating cAMP production. EP1 and EP3 signaling has the opposite effect through increasing IP3 and decreasing cAMP. Using available small-molecule antagonists of single EP receptors, the cyclooxygenase-2 (COX-2) inhibitor celecoxib, or a novel dual EP2/EP4 antagonist generated in this investigation, we tested which approach to block PGE2 signaling optimally restored immunologic activity in mouse and human immune cells and antitumor activity in syngeneic, spontaneous, and xenograft tumor models. We found that dual antagonism of EP2 and EP4 together significantly enhanced the activation of PGE2-suppressed mouse and human monocytes and CD8+ T cells in vitro as compared with single EP antagonists. CD8+ T-cell activation was dampened by single EP1 and EP3 antagonists. Dual EP2/EP4 PGE2 receptor antagonists increased tumor microenvironment lymphocyte infiltration and significantly reduced disease burden in multiple tumor models, including in the adenomatous polyposis coli (APC)min+/- spontaneous colorectal tumor model, compared with celecoxib. These results support a hypothesis that redundancy of EP2 and EP4 receptor signaling necessitates a therapeutic strategy of dual blockade of EP2 and EP4. Here we describe TPST-1495, a first-in-class orally available small-molecule dual EP2/EP4 antagonist. Significance Prostaglandin (PGE2) drives tumor progression but the pathway has not been effectively drugged. We demonstrate significantly enhanced immunologic potency and antitumor activity through blockade of EP2 and EP4 PGE2 receptor signaling together with a single molecule.
Collapse
Affiliation(s)
| | - Anja Holtz
- Tempest Therapeutics, Brisbane, California
| | | | | | | | - Dingzhi Wang
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | | | - Franciele Kipper
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Dipak Panigrahy
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Raymond N. Dubois
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, South Carolina
| | | | | | | |
Collapse
|
127
|
Zhu P, Wu K, Zhang C, Batool SS, Li A, Yu Z, Huang J. Advances in new target molecules against schistosomiasis: A comprehensive discussion of physiological structure and nutrient intake. PLoS Pathog 2023; 19:e1011498. [PMID: 37498810 PMCID: PMC10374103 DOI: 10.1371/journal.ppat.1011498] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023] Open
Abstract
Schistosomiasis, a severe parasitic disease, is primarily caused by Schistosoma mansoni, Schistosoma japonicum, or Schistosoma haematobium. Currently, praziquantel is the only recommended drug for human schistosome infection. However, the lack of efficacy of praziquantel against juvenile worms and concerns about the emergence of drug resistance are driving forces behind the research for an alternative medication. Schistosomes are obligatory parasites that survive on nutrients obtained from their host. The ability of nutrient uptake depends on their physiological structure. In short, the formation and maintenance of the structure and nutrient supply are mutually reinforcing and interdependent. In this review, we focus on the structural features of the tegument, esophagus, and intestine of schistosomes and their roles in nutrient acquisition. Moreover, we introduce the significance and modes of glucose, lipids, proteins, and amino acids intake in schistosomes. We linked the schistosome structure and nutrient supply, introduced the currently emerging targets, and analyzed the current bottlenecks in the research and development of drugs and vaccines, in the hope of providing new strategies for the prevention and control of schistosomiasis.
Collapse
Affiliation(s)
- Peng Zhu
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Kaijuan Wu
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
| | - Chaobin Zhang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Syeda Sundas Batool
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Anqiao Li
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- XiangYa School of Medicine, Central South University, Changsha, Hunan, China
| | - Zheng Yu
- Department of Microbiology, School of Basic Medical Science, Central South University, Changsha, Hunan, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| | - Jing Huang
- Department of Parasitology, School of Basic Medical Science, Central South University, Changsha, China
- China-Africa Research Center of Infectious Diseases, Central South University, Changsha, China
| |
Collapse
|
128
|
Chen Y, Wang L, Hou WT, Zha Z, Xu K, Zhou CZ, Li Q, Chen Y. Structural insights into human ABCC4-mediated transport of platelet agonist and antagonist. NATURE CARDIOVASCULAR RESEARCH 2023; 2:693-701. [PMID: 39195918 DOI: 10.1038/s44161-023-00289-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 05/19/2023] [Indexed: 08/29/2024]
Abstract
Human platelets contribute to hemostasis and thrombosis, the imbalance of which can cause cardiovascular diseases. The activation and accumulation of platelets can be induced by agonists or inhibited by antagonists. Thus, the human ABC transporter ABCC4, which pumps out platelet agonists and antagonists, might become a promising target for preventing cardiovascular diseases. Here we define five structures of human ABCC4: the apo and three complexed forms in the inward-facing conformation, in addition to an outward-facing occluded conformation upon ATP binding. Combined with biochemical assays, we structurally prove that U46619, a synthetic analog of the unstable agonist TXA2, and the antagonist aspirin are substrates of ABCC4. In addition, we found that the platelet antagonist dipyridamole is a strong competitive inhibitor against ABCC4. These complex structures also enable us to identify a transmembrane pocket in ABCC4 that provides a defined space for the rational design of specific platelet antagonists.
Collapse
Affiliation(s)
- Yu Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Liang Wang
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Wen-Tao Hou
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Zhihui Zha
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Kang Xu
- School of Life Sciences, University of Science and Technology of China, Hefei, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China
| | - Cong-Zhao Zhou
- School of Life Sciences, University of Science and Technology of China, Hefei, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| | - Qiong Li
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- School of Life Sciences, University of Science and Technology of China, Hefei, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| | - Yuxing Chen
- Department of Endocrinology, Institute of Endocrine and Metabolic Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
- School of Life Sciences, University of Science and Technology of China, Hefei, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
129
|
Czajka-Francuz P, Prendes MJ, Mankan A, Quintana Á, Pabla S, Ramkissoon S, Jensen TJ, Peiró S, Severson EA, Achyut BR, Vidal L, Poelman M, Saini KS. Mechanisms of immune modulation in the tumor microenvironment and implications for targeted therapy. Front Oncol 2023; 13:1200646. [PMID: 37427115 PMCID: PMC10325690 DOI: 10.3389/fonc.2023.1200646] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 06/05/2023] [Indexed: 07/11/2023] Open
Abstract
The efficacy of cancer therapies is limited to a great extent by immunosuppressive mechanisms within the tumor microenvironment (TME). Numerous immune escape mechanisms have been identified. These include not only processes associated with tumor, immune or stromal cells, but also humoral, metabolic, genetic and epigenetic factors within the TME. The identification of immune escape mechanisms has enabled the development of small molecules, nanomedicines, immune checkpoint inhibitors, adoptive cell and epigenetic therapies that can reprogram the TME and shift the host immune response towards promoting an antitumor effect. These approaches have translated into series of breakthroughs in cancer therapies, some of which have already been implemented in clinical practice. In the present article the authors provide an overview of some of the most important mechanisms of immunosuppression within the TME and the implications for targeted therapies against different cancers.
Collapse
Affiliation(s)
| | | | | | - Ángela Quintana
- Breast Cancer Unit, Vall d'Hebrón Institute of Oncology, Barcelona, Spain
| | | | | | | | - Sandra Peiró
- Breast Cancer Unit, Vall d'Hebrón Institute of Oncology, Barcelona, Spain
| | | | | | | | | | - Kamal S. Saini
- Fortrea, Inc., Durham, NC, United States
- Addenbrooke’s Hospital, Cambridge University Hospitals National Health Service (NHS) Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
130
|
Bayerl F, Meiser P, Donakonda S, Hirschberger A, Lacher SB, Pedde AM, Hermann CD, Elewaut A, Knolle M, Ramsauer L, Rudolph TJ, Grassmann S, Öllinger R, Kirchhammer N, Trefny M, Anton M, Wohlleber D, Höchst B, Zaremba A, Krüger A, Rad R, Obenauf AC, Schadendorf D, Zippelius A, Buchholz VR, Schraml BU, Böttcher JP. Tumor-derived prostaglandin E2 programs cDC1 dysfunction to impair intratumoral orchestration of anti-cancer T cell responses. Immunity 2023; 56:1341-1358.e11. [PMID: 37315536 DOI: 10.1016/j.immuni.2023.05.011] [Citation(s) in RCA: 67] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 02/08/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023]
Abstract
Type 1 conventional dendritic cells (cDC1s) are critical for anti-cancer immunity. Protective anti-cancer immunity is thought to require cDC1s to sustain T cell responses within tumors, but it is poorly understood how this function is regulated and whether its subversion contributes to immune evasion. Here, we show that tumor-derived prostaglandin E2 (PGE2) programmed a dysfunctional state in intratumoral cDC1s, disabling their ability to locally orchestrate anti-cancer CD8+ T cell responses. Mechanistically, cAMP signaling downstream of the PGE2-receptors EP2 and EP4 was responsible for the programming of cDC1 dysfunction, which depended on the loss of the transcription factor IRF8. Blockade of the PGE2-EP2/EP4-cDC1 axis prevented cDC1 dysfunction in tumors, locally reinvigorated anti-cancer CD8+ T cell responses, and achieved cancer immune control. In human cDC1s, PGE2-induced dysfunction is conserved and associated with poor cancer patient prognosis. Our findings reveal a cDC1-dependent intratumoral checkpoint for anti-cancer immunity that is targeted by PGE2 for immune evasion.
Collapse
Affiliation(s)
- Felix Bayerl
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Philippa Meiser
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sainitin Donakonda
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany; German Center for Infection Research, Munich, Germany
| | - Anna Hirschberger
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Sebastian B Lacher
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Anna-Marie Pedde
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Chris D Hermann
- Institute of Experimental Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Anais Elewaut
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Moritz Knolle
- Institute for Artificial Intelligence in Medicine & Healthcare, School of Medicine, Technical University of Munich, Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Thomas J Rudolph
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Simon Grassmann
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, Munich, Germany
| | - Rupert Öllinger
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Nicole Kirchhammer
- Cancer Immunology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Marcel Trefny
- Cancer Immunology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Martina Anton
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Dirk Wohlleber
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Bastian Höchst
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Anne Zaremba
- Department for Dermatology, University Hospital Essen, Essen, Germany
| | - Achim Krüger
- Institute of Experimental Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Roland Rad
- Institute of Molecular Oncology and Functional Genomics, School of Medicine, Technical University of Munich, Munich, Germany
| | - Anna C Obenauf
- Research Institute of Molecular Pathology, Vienna Biocenter, Vienna, Austria
| | - Dirk Schadendorf
- Department for Dermatology, University Hospital Essen, Essen, Germany
| | - Alfred Zippelius
- Cancer Immunology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Veit R Buchholz
- Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, Munich, Germany
| | - Barbara U Schraml
- Walter-Brendel Center for Experimental Medicine, LMU Munich, Planegg-Martinsried, Germany; Biomedical Center, Institute for Cardiovascular Physiology and Pathophysiology, LMU Munich, Planegg-Martinsried, Germany
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany.
| |
Collapse
|
131
|
Wu J, Meng F, Ran D, Song Y, Dang Y, Lai F, Yang L, Deng M, Song Y, Zhu J. The Metabolism and Immune Environment in Diffuse Large B-Cell Lymphoma. Metabolites 2023; 13:734. [PMID: 37367892 DOI: 10.3390/metabo13060734] [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: 03/21/2023] [Revised: 06/01/2023] [Accepted: 06/06/2023] [Indexed: 06/28/2023] Open
Abstract
Cells utilize different metabolic processes to maintain their growth and differentiation. Tumor cells have made some metabolic changes to protect themselves from malnutrition. These metabolic alterations affect the tumor microenvironment and macroenvironment. Developing drugs targeting these metabolic alterations could be a good direction. In this review, we briefly introduce metabolic changes/regulations of the tumor macroenvironment and microenvironment and summarize potential drugs targeting the metabolism in diffuse large B-cell lymphoma.
Collapse
Affiliation(s)
- Jianbo Wu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing 100142, China
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing 100191, China
| | - Fuqing Meng
- School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100191, China
| | - Danyang Ran
- School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100191, China
| | - Yalong Song
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Yunkun Dang
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Fan Lai
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Longyan Yang
- Center for Endocrine Metabolism and Immune Diseases, Beijing Luhe Hospital, Capital Medical University, Beijing 101149, China
| | - Mi Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing 100142, China
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing 100191, China
- School of Basic Medical Sciences, Health Science Center, Peking University, Beijing 100191, China
| | - Yuqin Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jun Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Lymphoma, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
132
|
Adamczyk AM, Leicaj ML, Fabiano MP, Cabrerizo G, Bannoud N, Croci DO, Witwer KW, Remes Lenicov F, Ostrowski M, Pérez PS. Extracellular vesicles from human plasma dampen inflammation and promote tissue repair functions in macrophages. J Extracell Vesicles 2023; 12:e12331. [PMID: 37272889 PMCID: PMC10241174 DOI: 10.1002/jev2.12331] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 04/14/2023] [Accepted: 05/04/2023] [Indexed: 06/06/2023] Open
Abstract
Although inflammation is a vital defence response to infection, if left uncontrolled, it can lead to pathology. Macrophages are critical players both in driving the inflammatory response and in the subsequent events required for restoring tissue homeostasis. Extracellular vesicles (EVs) are membrane-enclosed structures released by cells that mediate intercellular communication and are present in all biological fluids, including blood. Herein, we show that extracellular vesicles from plasma (pEVs) play a relevant role in the control of inflammation by counteracting PAMP-induced macrophage activation. Indeed, pEV-treatment of macrophages simultaneously with or prior to PAMP exposure reduced the secretion of pro-inflammatory IL-6 and TNF-α and increased IL-10 response. This anti-inflammatory activity was associated with the promotion of tissue-repair functions in macrophages, characterized by augmented efferocytosis and pro-angiogenic capacity, and increased expression of VEGFa, CD300e, RGS2 and CD93, genes involved in cell growth and tissue remodelling. We also show that simultaneous stimulation of macrophages with a PAMP and pEVs promoted COX2 expression and CREB phosphorylation as well as the accumulation of higher concentrations of PGE2 in cell culture supernatants. Remarkably, the anti-inflammatory activity of pEVs was abolished if cells were treated with a pharmacological inhibitor of COX2, indicating that pEV-mediated induction of COX2 is critical for the pEV-mediated inhibition of inflammation. Finally, we show that pEVs added to monocytes prior to their M-CSF-induced differentiation to macrophages increased efferocytosis and diminished pro-inflammatory cytokine responses to PAMP stimulation. In conclusion, our results suggest that pEVs are endogenous homeostatic modulators of macrophages, activating the PGE2/CREB pathway, decreasing the production of inflammatory cytokines and promoting tissue repair functions.
Collapse
Affiliation(s)
- Alan M. Adamczyk
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - María Luz Leicaj
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Martina Paula Fabiano
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Gonzalo Cabrerizo
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Nadia Bannoud
- Laboratorio de Glicobiología y Biología VascularInstituto de Histología y Embriología de MendozaCONICET‐Universidad Nacional de CuyoMendozaArgentina
| | - Diego O. Croci
- Laboratorio de Glicobiología y Biología VascularInstituto de Histología y Embriología de MendozaCONICET‐Universidad Nacional de CuyoMendozaArgentina
| | - Kenneth W. Witwer
- Department of Molecular and Comparative PathobiologyThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of NeurologyThe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Federico Remes Lenicov
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Matías Ostrowski
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| | - Paula Soledad Pérez
- Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS)Universidad de Buenos Aires‐CONICETBuenos AiresArgentina
| |
Collapse
|
133
|
Kalkavan H, Rühl S, Shaw JJP, Green DR. Non-lethal outcomes of engaging regulated cell death pathways in cancer. NATURE CANCER 2023; 4:795-806. [PMID: 37277528 PMCID: PMC10416134 DOI: 10.1038/s43018-023-00571-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Accepted: 04/27/2023] [Indexed: 06/07/2023]
Abstract
Regulated cell death (RCD) is essential for successful systemic cancer therapy. Yet, the engagement of RCD pathways does not inevitably result in cell death. Instead, RCD pathways can take part in diverse biological processes if the cells survive. Consequently, these surviving cells, for which we propose the term 'flatliners', harbor important functions. These evolutionarily conserved responses can be exploited by cancer cells to promote their own survival and growth, with challenges and opportunities for cancer therapy.
Collapse
Affiliation(s)
- Halime Kalkavan
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
- West German Cancer Center, Department of Medical Oncology, University Hospital Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site University Hospital Essen, Essen, Germany
| | - Sebastian Rühl
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
- T3 Pharmaceuticals AG, Allschwil, Switzerland
| | - Jeremy J P Shaw
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA
| | - Douglas R Green
- Department of Immunology, St Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
134
|
Dave M, Dev A, Somoza RA, Zhao N, Viswanath S, Mina PR, Chirra P, Obmann VC, Mahabeleshwar GH, Menghini P, Johnson BD, Nolta J, Soto C, Osme A, Khuat LT, Murphy W, Caplan AI, Cominelli F. Mesenchymal stem cells ameliorate inflammation in an experimental model of Crohn's disease via the mesentery. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.22.541829. [PMID: 37292753 PMCID: PMC10245893 DOI: 10.1101/2023.05.22.541829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Objective Mesenchymal stem cells (MSCs) are novel therapeutics for treatment of Crohn's disease. However, their mechanism of action is unclear, especially in disease-relevant chronic models of inflammation. Thus, we used SAMP-1/YitFc, a chronic and spontaneous murine model of small intestinal inflammation, to study the therapeutic effect and mechanism of human bone marrow-derived MSCs (hMSC). Design hMSC immunosuppressive potential was evaluated through in vitro mixed lymphocyte reaction, ELISA, macrophage co-culture, and RT-qPCR. Therapeutic efficacy and mechanism in SAMP were studied by stereomicroscopy, histopathology, MRI radiomics, flow cytometry, RT-qPCR, small animal imaging, and single-cell RNA sequencing (Sc-RNAseq). Results hMSC dose-dependently inhibited naïve T lymphocyte proliferation in MLR via PGE 2 secretion and reprogrammed macrophages to an anti-inflammatory phenotype. hMSC promoted mucosal healing and immunologic response early after administration in SAMP model of chronic small intestinal inflammation when live hMSCs are present (until day 9) and resulted in complete response characterized by mucosal, histological, immunologic, and radiological healing by day 28 when no live hMSCs are present. hMSC mediate their effect via modulation of T cells and macrophages in the mesentery and mesenteric lymph nodes (mLN). Sc-RNAseq confirmed the anti-inflammatory phenotype of macrophages and identified macrophage efferocytosis of apoptotic hMSCs as a mechanism of action that explains their long-term efficacy. Conclusion hMSCs result in healing and tissue regeneration in a chronic model of small intestinal inflammation. Despite being short-lived, exert long-term effects via macrophage reprogramming to an anti-inflammatory phenotype. Data Transparency Statement Single-cell RNA transcriptome datasets are deposited in an online open access repository 'Figshare' (DOI: https://doi.org/10.6084/m9.figshare.21453936.v1 ).
Collapse
|
135
|
Amano H, Eshima K, Ito Y, Nakamura M, Kitasato H, Ogawa F, Hosono K, Iwabuchi K, Uematsu S, Akira S, Narumiya S, Majima M. The microsomal prostaglandin E synthase-1/prostaglandin E2 axis induces recovery from ischaemia via recruitment of regulatory T cells. Cardiovasc Res 2023; 119:1218-1233. [PMID: 35986688 PMCID: PMC10411941 DOI: 10.1093/cvr/cvac137] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 07/24/2022] [Accepted: 07/26/2022] [Indexed: 11/13/2022] Open
Abstract
AIMS Microsomal prostaglandin E synthase-1 (mPGES-1)/prostaglandin E2 (PGE2) induces angiogenesis through the prostaglandin E2 receptor (EP1-4). Among immune cells, regulatory T cells (Tregs), which inhibit immune responses, have been implicated in angiogenesis, and PGE2 is known to modulate the function and differentiation of Tregs. We hypothesized that mPGES-1/PGE2-EP signalling could contribute to recovery from ischaemic conditions by promoting the accumulation of Tregs. METHODS AND RESULTS Wild-type (WT), mPGES-1-deficient (mPges-1-/-), and EP4 receptor-deficient (Ep4-/-) male mice, 6-8 weeks old, were used. Hindlimb ischaemia was induced by femoral artery ligation. Recovery from ischaemia was suppressed in mPges-1-/- mice and compared with WT mice. The number of accumulated forkhead box protein P3 (FoxP3)+ cells in ischaemic muscle tissue was decreased in mPges-1-/- mice compared with that in WT mice. Expression levels of transforming growth factor-β (TGF-β) and stromal cell derived factor-1 (SDF-1) in ischaemic tissue were also suppressed in mPges-1-/- mice. The number of accumulated FoxP3+ cells and blood flow recovery were suppressed when Tregs were depleted by injecting antibody against folate receptor 4 in WT mice but not in mPges-1-/- mice. Recovery from ischaemia was significantly suppressed in Ep4-/- mice compared with that in WT mice. Furthermore, mRNA levels of Foxp3 and Tgf-β were suppressed in Ep4-/- mice. Moreover, the number of accumulated FoxP3+ cells in ischaemic tissue was diminished in Ep4-/- mice compared with that in Ep4+/+ mice. CONCLUSION These findings suggested that mPGES-1/PGE2 induced neovascularization from ischaemia via EP4 by promoting the accumulation of Tregs. Highly selective EP4 agonists could be useful for the treatment of peripheral artery disease.
Collapse
Affiliation(s)
- Hideki Amano
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Koji Eshima
- Department of Immunology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Yoshiya Ito
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Masaki Nakamura
- Department of Microbiology, Kitasato University School of Allied Health Science, Kanagawa, Japan
| | - Hidero Kitasato
- Department of Microbiology, Kitasato University School of Allied Health Science, Kanagawa, Japan
| | - Fumihiro Ogawa
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Kanako Hosono
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
| | - Kazuya Iwabuchi
- Department of Immunology, Kitasato University School of Medicine, Kanagawa, Japan
| | - Satoshi Uematsu
- Department of Immunology and Genomics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shizuo Akira
- Laboratory of Host Defense, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Shuh Narumiya
- Department of Drug Discovery Medicine, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Masataka Majima
- Department of Pharmacology, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara, Kanagawa 252-0373, Japan
- Department of Medical Therapeutics, Kanagawa Institute of Technology, Atsugi, Kanagawa, Japan
| |
Collapse
|
136
|
Bowen CM, Deng N, Reyes-Uribe L, Parra ER, Rocha P, Solis LM, Wistuba II, Sepeda VO, Vornik L, Perloff M, Szabo E, Umar A, Sinha KM, Brown PH, Vilar E. Naproxen chemoprevention induces proliferation of cytotoxic lymphocytes in Lynch Syndrome colorectal mucosa. Front Immunol 2023; 14:1162669. [PMID: 37207208 PMCID: PMC10189148 DOI: 10.3389/fimmu.2023.1162669] [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: 02/09/2023] [Accepted: 04/20/2023] [Indexed: 05/21/2023] Open
Abstract
Background Recent clinical trial data from Lynch Syndrome (LS) carriers demonstrated that naproxen administered for 6-months is a safe primary chemoprevention that promotes activation of different resident immune cell types without increasing lymphoid cellularity. While intriguing, the precise immune cell types enriched by naproxen remained unanswered. Here, we have utilized cutting-edge technology to elucidate the immune cell types activated by naproxen in mucosal tissue of LS patients. Methods Normal colorectal mucosa samples (pre- and post-treatment) from a subset of patients enrolled in the randomized and placebo-controlled 'Naproxen Study' were obtained and subjected to a tissue microarray for image mass cytometry (IMC) analysis. IMC data was processed using tissue segmentation and functional markers to ascertain cell type abundance. Computational outputs were then used to quantitatively compare immune cell abundance in pre- and post-naproxen specimens. Results Using data-driven exploration, unsupervised clustering identified four populations of immune cell types with statistically significant changes between treatment and control groups. These four populations collectively describe a unique cell population of proliferating lymphocytes within mucosal samples from LS patients exposed to naproxen. Conclusions Our findings show that daily exposure of naproxen promotes T-cell proliferation in the colonic mucosa, which paves way for developing combination of immunoprevention strategies including naproxen for LS patients.
Collapse
Affiliation(s)
- Charles M. Bowen
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nan Deng
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Laura Reyes-Uribe
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Edwin Roger Parra
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Pedro Rocha
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Luisa M. Solis
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Ignacio I. Wistuba
- Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Valerie O. Sepeda
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Lana Vornik
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Marjorie Perloff
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, United States
| | - Eva Szabo
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, United States
| | - Asad Umar
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, United States
| | - Krishna M. Sinha
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Powel H. Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Eduardo Vilar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Clinical Cancer Genetics Program, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
137
|
Li L, Tian Y. The role of metabolic reprogramming of tumor-associated macrophages in shaping the immunosuppressive tumor microenvironment. Biomed Pharmacother 2023; 161:114504. [PMID: 37002579 DOI: 10.1016/j.biopha.2023.114504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/04/2023] [Accepted: 03/07/2023] [Indexed: 03/15/2023] Open
Abstract
Macrophages are potent immune effector cells in innate immunity and exert dual-effects in the tumor microenvironment (TME). Tumor-associated macrophages (TAMs) make up a significant portion of TME immune cells. Similar to M1/M2 macrophages, TAMs are also highly plastic, and their functions are regulated by cytokines, chemokines and other factors in the TME. The metabolic changes in TAMs are significantly associated with polarization towards a protumour or antitumour phenotype. The metabolites generated via TAM metabolic reprogramming in turn promote tumor progression and immune tolerance. In this review, we explore the metabolic reprogramming of TAMs in terms of energy, amino acid and fatty acid metabolism and the potential roles of these changes in immune suppression.
Collapse
Affiliation(s)
- Lunxu Li
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yu Tian
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
138
|
Chakraborty P, Aravindhan V, Mukherjee S. Helminth-derived biomacromolecules as therapeutic agents for treating inflammatory and infectious diseases: What lessons do we get from recent findings? Int J Biol Macromol 2023; 241:124649. [PMID: 37119907 DOI: 10.1016/j.ijbiomac.2023.124649] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/24/2023] [Indexed: 05/01/2023]
Abstract
Despite the tremendous progress in healthcare sectors, a number of life-threatening infectious, inflammatory, and autoimmune diseases are continuously challenging mankind throughout the globe. In this context, recent successes in utilizing helminth parasite-derived bioactive macromolecules viz. glycoproteins, enzymes, polysaccharides, lipids/lipoproteins, nucleic acids/nucleotides, and small organic molecules for treating various disorders primarily resulted from inflammation. Among the several parasites that infect humans, helminths (cestodes, nematodes, and trematodes) are known as efficient immune manipulators owing to their explicit ability to modulate and modify the innate and adaptive immune responses of humans. These molecules selectively bind to immune receptors on innate and adaptive immune cells and trigger multiple signaling pathways to elicit anti-inflammatory cytokines, expansion of alternatively activated macrophages, T-helper 2, and immunoregulatory T regulatory cell types to induce an anti-inflammatory milieu. Reduction of pro-inflammatory responses and repair of tissue damage by these anti-inflammatory mediators have been exploited for treating a number of autoimmune, allergic, and metabolic diseases. Herein, the potential and promises of different helminths/helminth-derived products as therapeutic agents in ameliorating immunopathology of different human diseases and their mechanistic insights of function at cell and molecular level alongside the molecular signaling cross-talks have been reviewed by incorporating up-to-date findings achieved in the field.
Collapse
Affiliation(s)
- Pritha Chakraborty
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India
| | | | - Suprabhat Mukherjee
- Integrative Biochemistry & Immunology Laboratory, Department of Animal Science, Kazi Nazrul University, Asansol 713340, India.
| |
Collapse
|
139
|
Abstract
T cells and natural killer (NK) cells have complementary roles in tumor immunity, and dual T cell and NK cell attack thus offers opportunities to deepen the impact of immunotherapy. Recent work has also shown that NK cells play an important role in recruiting dendritic cells to tumors and thus enhance induction of CD8 T cell responses, while IL-2 secreted by T cells activates NK cells. Targeting of immune evasion mechanisms from the activating NKG2D receptor and its MICA and MICB ligands on tumor cells offers opportunities for therapeutic intervention. Interestingly, T cells and NK cells share several important inhibitory and activating receptors that can be targeted to enhance T cell- and NK cell-mediated immunity. These inhibitory receptor-ligand systems include CD161-CLEC2D, TIGIT-CD155, and NKG2A/CD94-HLA-E. We also discuss emerging therapeutic strategies based on inhibitory and activating cytokines that profoundly impact the function of both lymphocyte populations within tumors.
Collapse
Affiliation(s)
- Oleksandr Kyrysyuk
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA;
| | - Kai W Wucherpfennig
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA;
- Department of Neurology, Brigham & Women's Hospital, Boston, Massachusetts, USA
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
140
|
Yen NTH, Oh JH, Van Anh NT, Le QV, Park SM, Park YJ, Cho YS, Moon KS, Nguyen HT, Shin JG, Long NP, Kim DH. Systems-level multi-omics characterization provides novel molecular insights into indomethacin toxicity. Chem Biol Interact 2023; 375:110430. [PMID: 36868495 DOI: 10.1016/j.cbi.2023.110430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/25/2023] [Accepted: 02/28/2023] [Indexed: 03/05/2023]
Abstract
The mechanism of indomethacin toxicity at the systemic level is largely unknown. In this study, multi-specimen molecular characterization was conducted in rats treated with three doses of indomethacin (2.5, 5, and 10 mg/kg) for 1 week. Kidney, liver, urine, and serum samples were collected and analyzed using untargeted metabolomics. The kidney and liver transcriptomics data (10 mg indomethacin/kg and control) were subjected to a comprehensive omics-based analysis. Indomethacin exposure at 2.5 and 5 mg/kg doses did not cause significant metabolome changes, whereas considerable alterations in the metabolic profile compared to the control were induced by a dose of 10 mg/kg. Decreased levels of metabolites and an increased creatine level in the urine metabolome indicated injury to the kidney. The integrated omics analysis in both liver and kidney revealed an oxidant-antioxidant imbalance due to an excess of reactive oxygen species, likely originating from dysfunctional mitochondria. Specifically, indomethacin exposure induced changes in metabolites related to the citrate cycle, cell membrane composition, and DNA synthesis in the kidney. The dysregulation of genes related to ferroptosis and suppression of amino acid and fatty acid metabolism were evidence of indomethacin-induced nephrotoxicity. In conclusion, a multi-specimen omics investigation provided important insights into the mechanism of indomethacin toxicity. The identification of targets that ameliorate indomethacin toxicity will enhance the therapeutic utility of this drug.
Collapse
Affiliation(s)
- Nguyen Thi Hai Yen
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Jung-Hwa Oh
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Nguyen Thi Van Anh
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, 70000, Viet Nam
| | - Se-Myo Park
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Young Jin Park
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Yong-Soon Cho
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Kyoung-Sik Moon
- Department of Predictive Toxicology, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Huy Truong Nguyen
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, 70000, Viet Nam
| | - Jae-Gook Shin
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, 47392, Republic of Korea
| | - Nguyen Phuoc Long
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea; Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan, 47392, Republic of Korea.
| | - Dong Hyun Kim
- Department of Pharmacology and PharmacoGenomics Research Center, Inje University College of Medicine, Busan, 47392, Republic of Korea.
| |
Collapse
|
141
|
Saha R, Patkar S, Pillai MM, Tayalia P. Bilayered skin substitute incorporating rutin nanoparticles for antioxidant, anti-inflammatory, and anti-fibrotic effect. BIOMATERIALS ADVANCES 2023; 150:213432. [PMID: 37119696 DOI: 10.1016/j.bioadv.2023.213432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 03/20/2023] [Accepted: 04/17/2023] [Indexed: 05/01/2023]
Abstract
Hypertrophic scarring in large burns and delayed healing in chronic wounds are consequences of prolonged and aggravated inflammation, sustained infiltration of immune cells, free radical generation, and abundance of inflammatory mediators. Therefore, it is imperative to curb hyperinflammation to expedite wound healing. In this study, rutin nanoparticles (RNPs) were synthesized without an encapsulant and incorporated into eggshell membrane powder-crosslinked gelatin-chitosan cryogels to impart antioxidant and anti-inflammatory properties for treating hyperinflammation. The resultant nanoparticles were found to be 17.53 ± 4.03 nm in size and were stable at room temperature for a month with no visible sedimentation. RNPs were found to be non-cytotoxic and exhibited anti-inflammatory (by increasing IL-10 levels) and antioxidant properties (by controlling the generation of reactive oxygen species and enhancing catalase production in human macrophages). Additionally, RNPs were found to reduce α-SMA expression in fibroblasts, thereby demonstrating their anti-scarring effect. In vivo studies with a bilayered skin substitute constituting an RNP-incorporated cryogel proved that it is biocompatible, does not induce renal toxicity, aids wound healing, and induces better re-epithelialization than the control groups at the initial stages. Thus, RNP-incorporated cryogels containing bilayered skin substitutes are an advanced and novel alternative to commercial dermo-epidermal substitutes that lack anti-inflammatory or anti-scarring properties.
Collapse
Affiliation(s)
- Rituparna Saha
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Shivali Patkar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Mamatha M Pillai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Prakriti Tayalia
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India.
| |
Collapse
|
142
|
Murthy D, Attri KS. PTGES Expression Is Associated with Metabolic and Immune Reprogramming in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2023; 24:ijms24087304. [PMID: 37108468 PMCID: PMC10138618 DOI: 10.3390/ijms24087304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/18/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Metabolic reprogramming is an established hallmark of multiple cancers, including pancreatic cancer. Dysregulated metabolism is utilized by cancer cells for tumor progression, metastasis, immune microenvironment remodeling, and therapeutic resistance. Prostaglandin metabolites have been shown to be critical for inflammation and tumorigenesis. While the functional role of prostaglandin E2 metabolite has been extensively studied, there is a limited understanding of the PTGES enzyme in pancreatic cancer. Here, we investigated the relationship between expression of prostaglandin E synthase (PTGES) isoforms and the pathogenesis and regulation of pancreatic cancer. Our analysis identified higher expression of PTGES in pancreatic tumors compared to normal pancreatic tissues, suggesting an oncogenic function. Only PTGES1 expression was significantly correlated with worse prognosis of pancreatic cancer patients. Further, utilizing cancer genome atlas data, PTGES was found to be positively correlated with epithelial-mesenchymal transition, metabolic pathways, mucin oncogenic proteins, and immune pathways in cancer cells. PTGES expression was also correlated with higher mutational burden in key driver genes, such as TP53 and KRAS. Furthermore, our analysis indicated that the oncogenic pathway controlled by PTGES1 could be regulated via DNA methylation-dependent epigenetic mechanisms. Notably, the glycolysis pathway was positively correlated with PTGES and may fuel cancer cell growth. PTGES expression was also associated with downregulation of the MHC pathway and negatively correlated with CD8+ T cell activation markers. In summary, our study established an association of PTGES expression with pancreatic cancer metabolism and the immune microenvironment.
Collapse
Affiliation(s)
- Divya Murthy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Kuldeep S Attri
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
143
|
Nativel F, Smith A, Boulestreau J, Lépine C, Baron J, Marquis M, Vignes C, Le Guennec Y, Veziers J, Lesoeur J, Loll F, Halgand B, Renard D, Abadie J, Legoff B, Blanchard F, Gauthier O, Vinatier C, Rieux AD, Guicheux J, Le Visage C. Micromolding-based encapsulation of mesenchymal stromal cells in alginate for intraarticular injection in osteoarthritis. Mater Today Bio 2023; 19:100581. [PMID: 36896417 PMCID: PMC9988569 DOI: 10.1016/j.mtbio.2023.100581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/27/2023] [Accepted: 02/10/2023] [Indexed: 02/15/2023] Open
Abstract
Osteoarthritis (OA) is an inflammatory joint disease that affects cartilage, subchondral bone, and joint tissues. Undifferentiated Mesenchymal Stromal Cells are a promising therapeutic option for OA due to their ability to release anti-inflammatory, immuno-modulatory, and pro-regenerative factors. They can be embedded in hydrogels to prevent their tissue engraftment and subsequent differentiation. In this study, human adipose stromal cells are successfully encapsulated in alginate microgels via a micromolding method. Microencapsulated cells retain their in vitro metabolic activity and bioactivity and can sense and respond to inflammatory stimuli, including synovial fluids from OA patients. After intra-articular injection in a rabbit model of post-traumatic OA, a single dose of microencapsulated human cells exhibit properties matching those of non-encapsulated cells. At 6 and 12 weeks post-injection, we evidenced a tendency toward a decreased OA severity, an increased expression of aggrecan, and a reduced expression of aggrecanase-generated catabolic neoepitope. Thus, these findings establish the feasibility, safety, and efficacy of injecting cells encapsulated in microgels, opening the door to a long-term follow-up in canine OA patients.
Collapse
Affiliation(s)
- Fabien Nativel
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Audrey Smith
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France.,UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200, Bruxelles, Belgium
| | - Jeremy Boulestreau
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Charles Lépine
- Nantes Université, CHU Nantes, Department of Pathology, F-44000 Nantes, France
| | - Julie Baron
- Nantes Université, CHU Nantes, Department of Pathology, F-44000 Nantes, France
| | - Melanie Marquis
- UR1268 BIA (Biopolymères Interactions Assemblages), INRAE, F-44300 Nantes, France
| | - Caroline Vignes
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Yoan Le Guennec
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Joelle Veziers
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Julie Lesoeur
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - François Loll
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Boris Halgand
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Denis Renard
- UR1268 BIA (Biopolymères Interactions Assemblages), INRAE, F-44300 Nantes, France
| | - Jerome Abadie
- LabONIRIS, ONIRIS (Nantes Atlantic College of Veterinary Medicine, Food Science and Engineering), F-44300 Nantes, France
| | - Benoit Legoff
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Frederic Blanchard
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Olivier Gauthier
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France.,ONIRIS Nantes-Atlantic College of Veterinary Medicine, Centre de Recherche et D'investigation Préclinique (CRIP), F-44300 Nantes, France
| | - Claire Vinatier
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Anne des Rieux
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, 1200, Bruxelles, Belgium
| | - Jerome Guicheux
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| | - Catherine Le Visage
- Nantes Université, ONIRIS, Univ Angers, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, RMeS, UMR 1229, F-44000 Nantes, France
| |
Collapse
|
144
|
Sandgruber F, Höger AL, Kunze J, Schenz B, Griehl C, Kiehntopf M, Kipp K, Kühn J, Stangl GI, Lorkowski S, Dawczynski C. Impact of Regular Intake of Microalgae on Nutrient Supply and Cardiovascular Risk Factors: Results from the NovAL Intervention Study. Nutrients 2023; 15:nu15071645. [PMID: 37049486 PMCID: PMC10097350 DOI: 10.3390/nu15071645] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/25/2023] [Accepted: 03/26/2023] [Indexed: 03/30/2023] Open
Abstract
A 14-day randomized controlled study with a parallel design was conducted with 80 healthy participants. Intervention groups I (IG1) and II (IG2) received a defined background diet and consumed a smoothie enriched with either 15 g of Chlorella dry weight (d.w.) or 15 g of Microchloropsis d.w. daily. Control group II (CG2) received a defined background diet without the smoothie. Control group I (CG1) received neither. Blood samples and 24-h urine were collected at the beginning and the end of the study. Serum concentrations of 25-hydroxyvitamin D3, vitamin D3, selenium, iron, ferritin, transferrin saturation, total cholesterol, low-density lipoprotein (LDL) cholesterol, high-density lipoprotein (HDL) cholesterol, non-HDL cholesterol and the LDL-cholesterol/HDL cholesterol ratio decreased in IG1 (p < 0.05), while 25-hydroxyvitamin D2 increased (p < 0.05). In IG2, vitamin D3, 25-hydroxyvitamins D2 and D3 decreased (p < 0.05), while concentrations of fatty acids C20:5n3 and C22:5n3 increased. Serum and urine uric acid increased in IG1 and IG2 (p < 0.05). Microchloropsis is a valuable source of n3 fatty acids, as is Chlorella of vitamin D2. Regular consumption of Chlorella may affect the iron and selenium status negatively but may impact blood lipids positively. An elevated uric acid concentration in blood and urine following the regular consumption of microalgae poses potential risks for human health.
Collapse
Affiliation(s)
- Fabian Sandgruber
- Junior Research Group Nutritional Concepts, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 29, 07743 Jena, Germany; (F.S.); (J.K.); (B.S.)
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Dornburger Str. 25, 07743 Jena, Germany; (J.K.); (G.I.S.); (S.L.)
| | - Anna-Lena Höger
- Competence Center Algal Biotechnology, Anhalt University of Applied Science, Bernburger Str. 55, 06366 Köthen, Germany; (A.-L.H.); (C.G.)
| | - Julia Kunze
- Junior Research Group Nutritional Concepts, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 29, 07743 Jena, Germany; (F.S.); (J.K.); (B.S.)
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Dornburger Str. 25, 07743 Jena, Germany; (J.K.); (G.I.S.); (S.L.)
| | - Benjamin Schenz
- Junior Research Group Nutritional Concepts, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 29, 07743 Jena, Germany; (F.S.); (J.K.); (B.S.)
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Dornburger Str. 25, 07743 Jena, Germany; (J.K.); (G.I.S.); (S.L.)
| | - Carola Griehl
- Competence Center Algal Biotechnology, Anhalt University of Applied Science, Bernburger Str. 55, 06366 Köthen, Germany; (A.-L.H.); (C.G.)
| | - Michael Kiehntopf
- Institute of Clinical Chemistry and Laboratory Diagnostics, University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany;
| | - Kristin Kipp
- Department of Pediatrics and Adolescent Medicine, Sophien- and Hufeland Hospital, Henry-Van-De-Velde-Str. 1, 99425 Weimar, Germany;
| | - Julia Kühn
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Dornburger Str. 25, 07743 Jena, Germany; (J.K.); (G.I.S.); (S.L.)
- Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Von-Danckelmann-Platz 2, 06120 Halle, Germany
| | - Gabriele I. Stangl
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Dornburger Str. 25, 07743 Jena, Germany; (J.K.); (G.I.S.); (S.L.)
- Institute of Agricultural and Nutritional Science, Martin Luther University Halle-Wittenberg, Von-Danckelmann-Platz 2, 06120 Halle, Germany
| | - Stefan Lorkowski
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Dornburger Str. 25, 07743 Jena, Germany; (J.K.); (G.I.S.); (S.L.)
- Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 25, 07743 Jena, Germany
| | - Christine Dawczynski
- Junior Research Group Nutritional Concepts, Institute of Nutritional Sciences, Friedrich Schiller University Jena, Dornburger Str. 29, 07743 Jena, Germany; (F.S.); (J.K.); (B.S.)
- Competence Cluster for Nutrition and Cardiovascular Health (nutriCARD) Halle-Jena-Leipzig, Dornburger Str. 25, 07743 Jena, Germany; (J.K.); (G.I.S.); (S.L.)
- Correspondence:
| |
Collapse
|
145
|
Meunier S, Frontczak A, Balssa L, Blanc J, Benhmida S, Pernot M, Quivrin M, Martin E, Hammoud Y, Créhange G, Boustani J. Elevated Baseline Neutrophil Count Correlates with Worse Outcomes in Patients with Muscle-Invasive Bladder Cancer Treated with Chemoradiation. Cancers (Basel) 2023; 15:cancers15061886. [PMID: 36980771 PMCID: PMC10047214 DOI: 10.3390/cancers15061886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/08/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND The role of inflammation in the development and prognosis of bladder cancer (BC) is now established. We evaluated the significance of neutrophil-to-lymphocyte ratio (NLR) and neutrophil count (PNN) in patients with localized BC treated with chemoradiation. METHODS Clinical characteristics and baseline biological data were retrospectively collected. We tested the association between NLR, PNN, and overall survival (OS) and progression-free survival (PFS). RESULTS One hundred and ninety-four patients were included. Median PNN was 4000.0/mm3 [1500.0-16,858.0] and median NLR was 2.6 [0.6-19.2]. In patients with NLR > 2.6, median OS and PFS were lower (OS: 25.5 vs. 58.4 months, p = 0.02; PFS: 14.1 vs. 26.7 months, p = 0.07). Patients with PNN > 4000/mm3 had significantly lower OS (21.8 vs. 70.1 months, p < 0.001) and PFS (13.7 vs. 38.8 months, p < 0.001). Contrary to NLR, PNN > 4000/mm3 was associated with shorter OS and PFS in multivariate analysis. CONCLUSIONS Elevated PNN at baseline was associated with worse OS and PFS. NLR was not an independent prognostic factor.
Collapse
Affiliation(s)
- Sébastien Meunier
- Department of Radiation Oncology, Centre Georges François Leclerc, 21000 Dijon, France
| | - Alexandre Frontczak
- Department of Urology, University Hospital of Besançon, 25000 Besançon, France
| | - Loïc Balssa
- Department of Urology, University Hospital of Besançon, 25000 Besançon, France
| | - Julie Blanc
- Department of Biostatistics, Centre Georges François Leclerc, 21000 Dijon, France
| | - Salim Benhmida
- Department of Radiation Oncology, University Hospital of Besançon, 25000 Besançon, France
| | - Mandy Pernot
- Department of Radiation Oncology, University Hospital of Besançon, 25000 Besançon, France
| | - Magali Quivrin
- Department of Radiation Oncology, Centre Georges François Leclerc, 21000 Dijon, France
| | - Etienne Martin
- Department of Radiation Oncology, Centre Georges François Leclerc, 21000 Dijon, France
| | - Yasser Hammoud
- Department of Radiation Oncology, University Hospital of Besançon, 25000 Besançon, France
| | - Gilles Créhange
- Department of Radiation Oncology, Institut Curie, 92210 Saint-Cloud, France
| | - Jihane Boustani
- Department of Radiation Oncology, University Hospital of Besançon, 25000 Besançon, France
- INSERM, EFS BFC, UMR1098, RIGHT, Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, University of Bourgogne Franche-Comté, 25000 Besançon, France
| |
Collapse
|
146
|
Hajihashemi P, Feizi A, Heidari Z, Haghighatdoost F. Association of omega-6 polyunsaturated fatty acids with blood pressure: A systematic review and meta-analysis of observational studies. Crit Rev Food Sci Nutr 2023; 63:2247-2259. [PMID: 36939291 DOI: 10.1080/10408398.2021.1973364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
OBJECTIVES This systematic review and meta-analysis aimed at summarizing earlier findings on the association of n-6 PUFAs levels in diets or blood with blood pressure. METHODS PubMed/Medline, Scopus, and Web of Science were searched for observational studies. Publications with data on the risk of hypertension, or the correlation between n-6 PUFAs or mean values of serum n-6 PUFAs levels in normotensive and hypertensive were included. RESULTS Twenty-two studies (16 cross-sectional studies, 5 cohorts and one case-control) were eligible. Combining 14 extracted effect sizes showed that higher circulatory/dietary n-6 PUFAs tended to be associated with 10% lower risk of HTN (95% CI: 0.81, 1.00), whereas combining 23 effect sizes illustrated no difference in circulatory/dietary n-6 PUFAs mean levels between normotensive and hypertensive subjects. According to subgroup analysis based on fatty acid types, total n-6 PUFAs (OR = 0.82, 95% CI: 0.70, 0.97) and linoleic acid (OR = 0.56, 95% CI: 0.39, 0.82) were inversely related to the risk of HTN. Circulatory/dietary n-6 PUFAs were correlated neither with systolic nor with diastolic blood pressure. CONCLUSIONS Higher circulatory/dietary n-6 PUFAs tend to be associated with lower odds of HTN. Particularly, total n-6 PUFAs and linoleic acid were associated with lower risk of HTN.
Collapse
Affiliation(s)
- Parisa Hajihashemi
- Hypertension Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Awat Feizi
- Biostatistics and Epidemiology Department, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zahra Heidari
- Biostatistics and Epidemiology Department, School of Health, Isfahan University of Medical Sciences, Isfahan, Iran.,Cardiac Rehabilitation Research Centre, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Fahimeh Haghighatdoost
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
147
|
Wu T, Orschell CM. The delayed effects of acute radiation exposure (DEARE): characteristics, mechanisms, animal models, and promising medical countermeasures. Int J Radiat Biol 2023; 99:1066-1079. [PMID: 36862990 PMCID: PMC10330482 DOI: 10.1080/09553002.2023.2187479] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/25/2023] [Accepted: 02/20/2023] [Indexed: 03/04/2023]
Abstract
PURPOSE Terrorist use of nuclear weapons and radiation accidents put the human population at risk for exposure to life-threatening levels of radiation. Victims of lethal radiation exposure face potentially lethal acute injury, while survivors of the acute phase are plagued with chronic debilitating multi-organ injuries for years after exposure. Developing effective medical countermeasures (MCM) for the treatment of radiation exposure is an urgent need that relies heavily on studies conducted in reliable and well-characterized animal models according to the FDA Animal Rule. Although relevant animal models have been developed in several species and four MCM for treatment of the acute radiation syndrome are now FDA-approved, animal models for the delayed effects of acute radiation exposure (DEARE) have only recently been developed, and there are no licensed MCM for DEARE. Herein, we provide a review of the DEARE including key characteristics of the DEARE gleaned from human data as well as animal, mechanisms common to multi-organ DEARE, small and large animal models used to study the DEARE, and promising new or repurposed MCM under development for alleviation of the DEARE. CONCLUSIONS Intensification of research efforts and support focused on better understanding of mechanisms and natural history of DEARE are urgently needed. Such knowledge provides the necessary first steps toward the design and development of MCM that effectively alleviate the life-debilitating consequences of the DEARE for the benefit of humankind worldwide.
Collapse
Affiliation(s)
- Tong Wu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Christie M Orschell
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
148
|
Rees A, Edwards-I-Coll Z, Richards O, Raikes ME, Angelini R, Thornton CA. The dynamic inflammatory profile of pregnancy can be monitored using a novel lipid-based mass spectrometry technique. Mol Omics 2023; 19:340-350. [PMID: 36883215 PMCID: PMC10167726 DOI: 10.1039/d2mo00294a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
The lipid environment changes throughout pregnancy both physiologically with emergent insulin resistance and pathologically e.g., gestational diabetes mellitus (GDM). Novel mass spectrometry (MS) techniques applied to minimally processed blood might lend themselves to monitoring changing lipid profiles to inform care decisions across pregnancy. In this study we use an intact-sandwich, MALDI-ToF MS method to identify phosphatidylcholine (PC) and lysophosphatidylcholine (LPC) species and calculate their ratio as an indicator of inflammation. Plasma and sera were prepared from venous blood of non-pregnant women (aged 18-40) and pregnant women at 16 weeks, 28 weeks (including GDM-positive women), and 37+ weeks (term) of gestation alongside umbilical cord blood (UCB). Women with a normal menstrual cycle and age-matched men provided finger-prick derived capillary sera at 6 time-points over a month. Serum rather than plasma was preferable for PC/LPC measurement. As pregnancy progresses, an anti-inflammatory phenotype dominates the maternal circulation, evidenced by increasing PC/LPC ratio. In contrast, the PC/LPC ratio of UCB was aligned to that of non-pregnant donors. BMI had no significant effect on the PC/LPC ratio, but GDM-complicated pregnancies had significantly lower PC/LPC at 16 weeks of gestation. To further translate the use of the PC/LPC ratio clinically, the utility of finger-prick blood was evaluated; no significant difference between capillary versus venous serum was found and we revealed the PC/LPC ratio oscillates with the menstrual cycle. Overall, we show that the PC/LPC ratio can be measured simply in human serum and has the potential to be used as a time-efficient and less invasive biomarker of (mal)adaptative inflammation.
Collapse
Affiliation(s)
- April Rees
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK, SA2 8PP.
| | - Zoe Edwards-I-Coll
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK, SA2 8PP.
| | - Oliver Richards
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK, SA2 8PP.
| | - Molly E Raikes
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK, SA2 8PP.
| | - Roberto Angelini
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK, SA2 8PP.
| | - Catherine A Thornton
- Institute of Life Science, Swansea University Medical School, Swansea, Wales, UK, SA2 8PP.
| |
Collapse
|
149
|
Mehta H, Tasin I, Hackstein CP, Willberg C, Klenerman P. Prostaglandins differentially modulate mucosal-associated invariant T-cell activation and function according to stimulus. Immunol Cell Biol 2023; 101:262-272. [PMID: 36541521 PMCID: PMC10152717 DOI: 10.1111/imcb.12617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 05/29/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022]
Abstract
Mucosal-associated invariant T (MAIT) cells are an innate-like T-cell type conserved in many mammals and especially abundant in humans. Their semi-invariant T-cell receptor (TCR) recognizes the major histocompatibility complex-like molecule MR1 presenting riboflavin intermediates associated with microbial metabolism. Full MAIT cell triggering requires costimulation via cytokines, and the cells can also be effectively triggered in a TCR-independent manner by cytokines [e.g. interleukin (IL)-12 and IL-18 in combination]. Thus, triggering of MAIT cells is highly sensitive to local soluble mediators. Suppression of MAIT cell activation has not been well explored and could be very relevant to their roles in infection, inflammation and cancer. Prostaglandins (PG) are major local mediators of these microenvironments which can have regulatory roles for T cells. Here, we explored whether prostaglandins suppressed MAIT cell activation in response to TCR-dependent and TCR-independent signals. We found that protaglandin E2 (PGE2 ) and to a lesser extent protaglandin D2 (PGD2 ), but not leukotrienes, suppressed MAIT cell responses to Escherichia coli or TCR triggers. However, there was no impact on cytokine-induced triggering. The inhibition was blocked by targeting the signaling mediated via PG receptor 2 (PTGER2) and 4 (PTGER4) receptors in combination. These data indicate that prostaglandins can potentially modulate local MAIT cell functions in vivo and indicate distinct regulation of the TCR-dependent and TCR-independent pathways of MAIT cell activation.
Collapse
Affiliation(s)
- Hema Mehta
- The Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUK
| | - Irene Tasin
- The Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUK
| | | | - Christian Willberg
- The Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUK
| | - Paul Klenerman
- The Peter Medawar Building for Pathogen ResearchUniversity of OxfordOxfordUK
- NIHR Biomedical Research CentreUniversity of OxfordOxfordUK
| |
Collapse
|
150
|
Ahluwalia M, Mcmichael H, Kumar M, Espinosa MP, Bosomtwi A, Lu Y, Khodadadi H, Jarrahi A, Khan MB, Hess DC, Rahimi SY, Vender JR, Vale FL, Braun M, Baban B, Dhandapani KM, Vaibhav K. Altered endocannabinoid metabolism compromises the brain-CSF barrier and exacerbates chronic deficits after traumatic brain injury in mice. Exp Neurol 2023; 361:114320. [PMID: 36627040 PMCID: PMC9904276 DOI: 10.1016/j.expneurol.2023.114320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/07/2022] [Accepted: 01/06/2023] [Indexed: 01/09/2023]
Abstract
Endocannabinoids [2-arachidonoylglycerol (2-AG) and N-arachidonoylethanolamine (AEA)], endogenously produced arachidonate-based lipids, are anti-inflammatory physiological ligands for two known cannabinoid receptors, CB1 and CB2, yet the molecular and cellular mechanisms underlying their effects after brain injury are poorly defined. In the present study, we hypothesize that traumatic brain injury (TBI)-induced loss of endocannabinoids exaggerates neurovascular injury, compromises brain-cerebrospinal fluid (CSF) barriers (BCB) and causes behavioral dysfunction. Preliminary analysis in human CSF and plasma indicates changes in endocannabinoid levels. This encouraged us to investigate the levels of endocannabinoid-metabolizing enzymes in a mouse model of controlled cortical impact (CCI). Reductions in endocannabinoid (2-AG and AEA) levels in plasma were supported by higher expression of their respective metabolizing enzymes, monoacylglycerol lipase (MAGL), fatty acid amide hydrolase (FAAH), and cyclooxygenase 2 (Cox-2) in the post-TBI mouse brain. Following increased metabolism of endocannabinoids post-TBI, we observed increased expression of CB2, non-cannabinoid receptor Transient receptor potential vanilloid-1 (TRPV1), aquaporin 4 (AQP4), ionized calcium binding adaptor molecule 1 (IBA1), glial fibrillary acidic protein (GFAP), and acute reduction in cerebral blood flow (CBF). The BCB and pericontusional cortex showed altered endocannabinoid expressions and reduction in ventricular volume. Finally, loss of motor functions and induced anxiety behaviors were observed in these TBI mice. Taken together, our findings suggest endocannabinoids and their metabolizing enzymes play an important role in the brain and BCB integrity and highlight the need for more extensive studies on these mechanisms.
Collapse
Affiliation(s)
- Meenakshi Ahluwalia
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Hannah Mcmichael
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Manish Kumar
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Mario P Espinosa
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Asamoah Bosomtwi
- Georgia Cancer Center, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Yujiao Lu
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Hesam Khodadadi
- Department of Oral Biology and Diagnostic Sciences, Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Abbas Jarrahi
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Mohammad Badruzzaman Khan
- Department of Neurology, Neuroscience Center of Excellence, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - David C Hess
- Department of Neurology, Neuroscience Center of Excellence, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Scott Y Rahimi
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - John R Vender
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Fernando L Vale
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Molly Braun
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America; Department of Psychiatry and Behavioral Sciences, University of Washington School of Medicine, Seattle, WA, United States of America; VISN 20 Mental Illness Research, Education and Clinical Center (MIRECC), VA Puget Sound Health Care System, Seattle, WA, United States of America
| | - Babak Baban
- Department of Oral Biology and Diagnostic Sciences, Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, United States of America; Department of Neurology, Neuroscience Center of Excellence, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Krishnan M Dhandapani
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America
| | - Kumar Vaibhav
- Department of Neurosurgery, Medical College of Georgia, Augusta University, Augusta, GA, United States of America; Department of Oral Biology and Diagnostic Sciences, Center for Excellence in Research, Scholarship and Innovation, Dental College of Georgia, Augusta University, Augusta, GA, United States of America.
| |
Collapse
|