1
|
Kubaski Benevides AP, Marin AM, Wosniaki DK, Oliveira RN, Koerich GM, Kusma BN, Munhoz EC, Zanette DL, Aoki MN. Expression of HOTAIR and PTGS2 as potential biomarkers in chronic myeloid leukemia patients in Brazil. Front Oncol 2024; 14:1443346. [PMID: 39450252 PMCID: PMC11499243 DOI: 10.3389/fonc.2024.1443346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
Chronic myeloid leukemia (CML) is a clonal myeloproliferative neoplasm in which all the patients has the translocation (9;22) that generates de BCR::ABL1 tyrosine kinase. Despite this disease possessing a good biomarker (BCR::ABL1 transcripts level) for diagnosis and prognosis, many studies has been performed to investigate other molecules, such as the long noncoding RNAs (lncRNAs) and mRNAs, as potential biomarkers with the aim of predicting a change in BCR::ABL1 levels and as an associated biomarker. A RNAseq was performed comparing 6 CML patients with high BCR::ABL1 expression with 6 healthy control individuals, comprising the investigation cohort to investigate these molecules. To validate the results obtained by RNAseq, samples of 87 CML patients and 42 healthy controls were used in the validation cohort by RT-qPCR assays. The results showed lower expression of HOTAIR and PTGS2 in CML patients. The HOTAIR expression is inversely associated with BCR::ABL1 expression in imatinib-treated CML patients, and to PTGS2 showing that CML patients with high BCR::ABL1 expression showed reduced PTGS2 expression.
Collapse
Affiliation(s)
- Ana Paula Kubaski Benevides
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Anelis Maria Marin
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Denise K. Wosniaki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Rafaela Noga Oliveira
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Gabriela Marino Koerich
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Bianca Nichele Kusma
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | | | - Dalila Luciola Zanette
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| | - Mateus Nóbrega Aoki
- Laboratory for Applied Science and Technology in Health, Carlos Chagas Institute, Oswaldo Cruz Foundation (Fiocruz), Curitiba, Brazil
| |
Collapse
|
2
|
Alexander M, Cho E, Gliozheni E, Salem Y, Cheung J, Ichii H. Pathology of Diabetes-Induced Immune Dysfunction. Int J Mol Sci 2024; 25:7105. [PMID: 39000211 PMCID: PMC11241249 DOI: 10.3390/ijms25137105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/16/2024] Open
Abstract
Diabetes is associated with numerous comorbidities, one of which is increased vulnerability to infections. This review will focus on how diabetes mellitus (DM) affects the immune system and its various components, leading to the impaired proliferation of immune cells and the induction of senescence. We will explore how the pathology of diabetes-induced immune dysfunction may have similarities to the pathways of "inflammaging", a persistent low-grade inflammation common in the elderly. Inflammaging may increase the likelihood of conditions such as rheumatoid arthritis (RA) and periodontitis at a younger age. Diabetes affects bone marrow composition and cellular senescence, and in combination with advanced age also affects lymphopoiesis by increasing myeloid differentiation and reducing lymphoid differentiation. Consequently, this leads to a reduced immune system response in both the innate and adaptive phases, resulting in higher infection rates, reduced vaccine response, and increased immune cells' senescence in diabetics. We will also explore how some diabetes drugs induce immune senescence despite their benefits on glycemic control.
Collapse
Affiliation(s)
- Michael Alexander
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Eric Cho
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Eiger Gliozheni
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Yusuf Salem
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Joshua Cheung
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| | - Hirohito Ichii
- Division of Transplantation, Department of Surgery, University of California, Irvine, CA 92868, USA
| |
Collapse
|
3
|
Santiso A, Heinemann A, Kargl J. Prostaglandin E2 in the Tumor Microenvironment, a Convoluted Affair Mediated by EP Receptors 2 and 4. Pharmacol Rev 2024; 76:388-413. [PMID: 38697857 DOI: 10.1124/pharmrev.123.000901] [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: 04/13/2023] [Revised: 11/15/2023] [Accepted: 11/20/2023] [Indexed: 05/05/2024] Open
Abstract
The involvement of the prostaglandin E2 (PGE2) system in cancer progression has long been recognized. PGE2 functions as an autocrine and paracrine signaling molecule with pleiotropic effects in the human body. High levels of intratumoral PGE2 and overexpression of the key metabolic enzymes of PGE2 have been observed and suggested to contribute to tumor progression. This has been claimed for different types of solid tumors, including, but not limited to, lung, breast, and colon cancer. PGE2 has direct effects on tumor cells and angiogenesis that are known to promote tumor development. However, one of the main mechanisms behind PGE2 driving cancerogenesis is currently thought to be anchored in suppressed antitumor immunity, thus providing possible therapeutic targets to be used in cancer immunotherapies. EP2 and EP4, two receptors for PGE2, are emerging as being the most relevant for this purpose. This review aims to summarize the known roles of PGE2 in the immune system and its functions within the tumor microenvironment. SIGNIFICANCE STATEMENT: Prostaglandin E2 (PGE2) has long been known to be a signaling molecule in cancer. Its presence in tumors has been repeatedly associated with disease progression. Elucidation of its effects on immunological components of the tumor microenvironment has highlighted the potential of PGE2 receptor antagonists in cancer treatment, particularly in combination with immune checkpoint inhibitor therapeutics. Adjuvant treatment could increase the response rates and the efficacy of immune-based therapies.
Collapse
Affiliation(s)
- Ana Santiso
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Akos Heinemann
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| | - Julia Kargl
- Division of Pharmacology, Otto Loewi Research Center, Medical University of Graz, Graz, Austria
| |
Collapse
|
4
|
Gong R, Hu Y, Yu Q, Fang L, Ren H. Metabolic signatures in pancreatic ductal adenocarcinoma: diagnostic and therapeutic implications. JOURNAL OF PANCREATOLOGY 2023; 6:185-195. [DOI: 10.1097/jp9.0000000000000146] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is the prototypical aggressive cancer that develops in nutrient-deficient and hypoxic microenvironment. PDAC overcomes these restrictions by employing unconventional tactics for the procurement and usage of fuel sources. The substantial reprogramming of PDAC cell metabolism is driven by oncogene-mediated cell-autonomous pathways. PDAC cells use glucose, glutamine, and lipids for energy and depend on autophagy and macropinocytosis for survival and growth. They also interact metabolically with non-cancerous cells, aiding tumor progression. Many clinical trials focusing on altered metabolism are ongoing. Understanding the metabolic regulation of PDAC cells will not only help to increase understanding of the mechanisms of disease progression but also provide insights for the development of new diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Ruining Gong
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Yonglu Hu
- Department of Endocrinology, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - Qian Yu
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| | - Lin Fang
- Phase I Clinical Research Center, The Affiliated Hospital of Qingdao University, Qingdao, 266003, China
| | - He Ren
- Shandong Provincial Key Laboratory of Clinical Research for Pancreatic Diseases, Center for GI Cancer Diagnosis and Treatment, Tumor Immunology and Cytotherapy, Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao 266003, China
| |
Collapse
|
5
|
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
|
6
|
Noraldeen SAM, Rasulova I, Lalitha R, Hussin F, Alsaab HO, Alawadi AH, Alsaalamy A, Sayyid NH, Alkhafaji AT, Mustafa YF, Shayan SK. Involving stemness factors to improve CAR T-cell-based cancer immunotherapy. Med Oncol 2023; 40:313. [PMID: 37779152 DOI: 10.1007/s12032-023-02191-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/09/2023] [Indexed: 10/03/2023]
Abstract
Treatment with chimeric antigen receptor (CAR) T cells indicated remarkable clinical responses with liquid cancers such as hematological malignancies; however, their therapeutic efficacy faced with many challenges in solid tumors due to severe toxicities, antigen evasion, restricted and limited tumor tissue trafficking and infiltration, and, more importantly, immunosuppressive tumor microenvironment (TME) factors that impair the CAR T-cell function adds support survival of cancer stem cells (CSCs), responsible for tumor recurrence and resistance to current cancer therapies. Therefore, in-depth identification of TME and development of more potent CAR platform targeting CSCs may overcome the raised challenges, as presented in this review. We also discuss recent stemness-based innovations in CAR T-cell production and engineering to improve their efficacy in vivo, and finally, we propose solutions and strategies such as oncolytic virus-based therapy and combination therapy to revive the function of CAR T-cell therapy, especially in TME of solid tumors in future.
Collapse
Affiliation(s)
| | - Irodakhon Rasulova
- School of Humanities, Natural & Social Sciences, New Uzbekistan University, 54 Mustaqillik Ave., 100007, Tashkent, Uzbekistan
| | - Repudi Lalitha
- Department of Pharmaceutical Analysis, Chaitanya Deemed to be University, Hyderabad, Telangana, India.
| | - Farah Hussin
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Hashem O Alsaab
- Department of Pharmaceutics and Pharmaceutical Technology, Taif University, 21944, Taif, Saudi Arabia
| | - Ahmed Hussien Alawadi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| | - Ali Alsaalamy
- College of Technical Engineering, Imam Ja'afar Al-Sadiq University, Al-Muthanna, 66002, Iraq
| | - Nidhal Hassan Sayyid
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, 41001, Iraq
| | - Sepideh Karkon Shayan
- Student Research Committee, School of Medicine, Gonabad University of Medical Sciences, Gonabad, Iran.
| |
Collapse
|
7
|
Morin S, Tremblay A, Dumais E, Julien P, Flamand N, Pouliot R. Eicosapentaenoic Acid Influences the Lipid Profile of an In Vitro Psoriatic Skin Model Produced with T Cells. Biomolecules 2023; 13:1413. [PMID: 37759812 PMCID: PMC10526348 DOI: 10.3390/biom13091413] [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: 08/14/2023] [Revised: 09/07/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Psoriasis is a skin disease characterized by epidermal hyperplasia and an inappropriate activation of the adaptive immunity. A dysregulation of the skin's lipid mediators is reported in the disease with a predominance of the inflammatory cascade derived from n-6 polyunsaturated fatty acids (n-6 PUFAs). Bioactive lipid mediators derived from arachidonic acid (AA) are involved in the inflammatory functions of T cells in psoriasis, whereas n-3 PUFAs' derivatives are anti-inflammatory metabolites. Here, we sought to evaluate the influence of a supplementation of the culture media with eicosapentaenoic acid (EPA) on the lipid profile of a psoriatic skin model produced with polarized T cells. Healthy and psoriatic skin substitutes were produced following the auto-assembly technique. Psoriatic skin substitutes produced with or without T cells presented increased epidermal and dermal linolenic acid (LA) and AA levels. N-6 PUFA lipid mediators were strongly measured in psoriatic substitutes, namely, 13-hydroxyoctadecadienoic acid (13-HODE), prostaglandin E2 (PGE2) and 12-hydroxyeicosatetraenoic acid (12-HETE). The added EPA elevated the amounts of EPA, n-3 docosapentaenoic acid (DPA) and docosahexaenoic acid (DHA) in the epidermal and dermal phospholipids. The EPA supplementation balanced the production of epidermal lipid mediators, with an increase in prostaglandin E3 (PGE3), 12-hydroxyeicosapentaenoic acid (12-HEPE) and N-eicosapentaenoyl-ethanolamine (EPEA) levels. These findings show that EPA modulates the lipid composition of psoriatic skin substitutes by encouraging the return to a cutaneous homeostatic state.
Collapse
Affiliation(s)
- Sophie Morin
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, 1401 18e Rue, Québec, QC G1J 2Z4, Canada; (S.M.); (A.T.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Andréa Tremblay
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, 1401 18e Rue, Québec, QC G1J 2Z4, Canada; (S.M.); (A.T.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| | - Elizabeth Dumais
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (E.D.); (N.F.)
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC G1V 0A6, Canada
| | - Pierre Julien
- Centre de Recherche du CHU de Québec-Université Laval, Axe Endocrinologie et Néphrologie, Université Laval, Québec, QC G1V 4G2, Canada;
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Nicolas Flamand
- Centre de Recherche de l’Institut Universitaire de Cardiologie et de Pneumologie de Québec, Québec, QC G1V 4G5, Canada; (E.D.); (N.F.)
- Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health (CERC-MEND), Université Laval, Québec, QC G1V 0A6, Canada
- Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Roxane Pouliot
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Axe Médecine Régénératrice, Centre de Recherche du CHU de Québec-Université Laval, 1401 18e Rue, Québec, QC G1J 2Z4, Canada; (S.M.); (A.T.)
- Faculté de Pharmacie, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
8
|
Spanos F, Deleidi M. Glycolipids in Parkinson's disease: beyond neuronal function. FEBS Open Bio 2023; 13:1558-1579. [PMID: 37219461 PMCID: PMC10476577 DOI: 10.1002/2211-5463.13651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/10/2023] [Accepted: 05/22/2023] [Indexed: 05/24/2023] Open
Abstract
Glycolipid balance is key to normal body function, and its alteration can lead to a variety of diseases involving multiple organs and tissues. Glycolipid disturbances are also involved in Parkinson's disease (PD) pathogenesis and aging. Increasing evidence suggests that glycolipids affect cellular functions beyond the brain, including the peripheral immune system, intestinal barrier, and immunity. Hence, the interplay between aging, genetic predisposition, and environmental exposures could initiate systemic and local glycolipid changes that lead to inflammatory reactions and neuronal dysfunction. In this review, we discuss recent advances in the link between glycolipid metabolism and immune function and how these metabolic changes can exacerbate immunological contributions to neurodegenerative diseases, with a focus on PD. Further understanding of the cellular and molecular mechanisms that control glycolipid pathways and their impact on both peripheral tissues and the brain will help unravel how glycolipids shape immune and nervous system communication and the development of novel drugs to prevent PD and promote healthy aging.
Collapse
Affiliation(s)
- Fokion Spanos
- Institut Imagine, INSERM UMR1163Paris Cité UniversityFrance
- Aligning Science Across Parkinson's (ASAP) Collaborative Research NetworkChevy ChaseMDUSA
| | - Michela Deleidi
- Institut Imagine, INSERM UMR1163Paris Cité UniversityFrance
- Aligning Science Across Parkinson's (ASAP) Collaborative Research NetworkChevy ChaseMDUSA
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain ResearchUniversity of TübingenGermany
| |
Collapse
|
9
|
Nicola Candia AJ, Garcia Fallit M, Peña Agudelo JA, Pérez Küper M, Gonzalez N, Moreno Ayala MA, De Simone E, Giampaoli C, Casares N, Seilicovich A, Lasarte JJ, Zanetti FA, Candolfi M. Targeting FOXP3 Tumor-Intrinsic Effects Using Adenoviral Vectors in Experimental Breast Cancer. Viruses 2023; 15:1813. [PMID: 37766222 PMCID: PMC10537292 DOI: 10.3390/v15091813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/29/2023] Open
Abstract
The regulatory T cell master transcription factor, Forkhead box P3 (Foxp3), has been detected in cancer cells; however, its role in breast tumor pathogenesis remains controversial. Here we assessed Foxp3 tumor intrinsic effects in experimental breast cancer using a Foxp3 binder peptide (P60) that impairs Foxp3 nuclear translocation. Cisplatin upregulated Foxp3 expression in HER2+ and triple-negative breast cancer (TNBC) cells. Foxp3 inhibition with P60 enhanced chemosensitivity and reduced cell survival and migration in human and murine breast tumor cells. We also developed an adenoviral vector encoding P60 (Ad.P60) that efficiently transduced breast tumor cells, reduced cell viability and migration, and improved the cytotoxic response to cisplatin. Conditioned medium from transduced breast tumor cells contained lower levels of IL-10 and improved the activation of splenic lymphocytes. Intratumoral administration of Ad.P60 in breast-tumor-bearing mice significantly reduced tumor infiltration of Tregs, delayed tumor growth, and inhibited the development of spontaneous lung metastases. Our results suggest that Foxp3 exerts protumoral intrinsic effects in breast cancer cells and that gene-therapy-mediated blockade of Foxp3 could constitute a therapeutic strategy to improve the response of these tumors to standard treatment.
Collapse
Affiliation(s)
- Alejandro J. Nicola Candia
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina; (A.J.N.C.); (A.S.)
| | - Matías Garcia Fallit
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina; (A.J.N.C.); (A.S.)
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina
| | - Jorge A. Peña Agudelo
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina; (A.J.N.C.); (A.S.)
| | - Melanie Pérez Küper
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina; (A.J.N.C.); (A.S.)
| | - Nazareno Gonzalez
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina; (A.J.N.C.); (A.S.)
| | - Mariela A. Moreno Ayala
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina; (A.J.N.C.); (A.S.)
| | - Emilio De Simone
- Cátedra de Fisiología Animal, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires C1428BFA, Argentina
| | - Carla Giampaoli
- Cátedra de Fisiología Animal, Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires C1428BFA, Argentina
| | - Noelia Casares
- Program Immunology and Immunotherapy, Centro de Investigación Médica Aplicada (CIMA, CUN), 31008 Pamplona, Spain; (N.C.)
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain
| | - Adriana Seilicovich
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina; (A.J.N.C.); (A.S.)
| | - Juan José Lasarte
- Program Immunology and Immunotherapy, Centro de Investigación Médica Aplicada (CIMA, CUN), 31008 Pamplona, Spain; (N.C.)
- Instituto de Investigación Sanitaria de Navarra (IDISNA), 31008 Pamplona, Spain
| | - Flavia A. Zanetti
- Instituto de Ciencia y Tecnología “Dr. Cesar Milstein”, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Saladillo C1440FFX, Buenos Aires, Argentina;
| | - Marianela Candolfi
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires C1121A6B, Argentina; (A.J.N.C.); (A.S.)
| |
Collapse
|
10
|
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
|
11
|
Burkett JB, Doran AC, Gannon M. Harnessing prostaglandin E 2 signaling to ameliorate autoimmunity. Trends Immunol 2023; 44:162-171. [PMID: 36707339 PMCID: PMC9975049 DOI: 10.1016/j.it.2023.01.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 01/06/2023] [Accepted: 01/07/2023] [Indexed: 01/26/2023]
Abstract
The etiology of most autoimmune diseases remains unknown; however, shared among them is a disruption of immunoregulation. Prostaglandin lipid signaling molecules possess context-dependent immunoregulatory properties, making their role in autoimmunity difficult to decipher. For example, prostaglandin E2 (PGE2) can function as an immunosuppressive molecule as well as a proinflammatory mediator in different circumstances, contributing to the expansion and activation of T cell subsets associated with autoimmunity. Recently, PGE2 was shown to play important roles in the resolution and post-resolution phases of inflammation, promoting return to tissue homeostasis. We propose that PGE2 plays both proinflammatory and pro-resolutory roles in the etiology of autoimmunity, and that harnessing this signaling pathway during the resolution phase might help prevent autoimmune attack.
Collapse
Affiliation(s)
- Juliann B Burkett
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Amanda C Doran
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Maureen Gannon
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA; Department of Veterans Affairs Tennessee Valley, Nashville, TN, USA.
| |
Collapse
|
12
|
Wu B, Shi X, Jiang M, Liu H. Cross-talk between cancer stem cells and immune cells: potential therapeutic targets in the tumor immune microenvironment. Mol Cancer 2023; 22:38. [PMID: 36810098 PMCID: PMC9942413 DOI: 10.1186/s12943-023-01748-4] [Citation(s) in RCA: 91] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 02/15/2023] [Indexed: 02/24/2023] Open
Abstract
Ongoing research has revealed that the existence of cancer stem cells (CSCs) is one of the biggest obstacles in the current cancer therapy. CSCs make an influential function in tumor progression, recurrence and chemoresistance due to their typical stemness characteristics. CSCs are preferentially distributed in niches, and those niche sites exhibit characteristics typical of the tumor microenvironment (TME). The complex interactions between CSCs and TME illustrate these synergistic effects. The phenotypic heterogeneity within CSCs and the spatial interactions with the surrounding tumor microenvironment led to increased therapeutic challenges. CSCs interact with immune cells to protect themselves against immune clearance by exploiting the immunosuppressive function of multiple immune checkpoint molecules. CSCs also can protect themselves against immune surveillance by excreting extracellular vesicles (EVs), growth factors, metabolites and cytokines into the TME, thereby modulating the composition of the TME. Therefore, these interactions are also being considered for the therapeutic development of anti-tumor agents. We discuss here the immune molecular mechanisms of CSCs and comprehensively review the interplay between CSCs and the immune system. Thus, studies on this topic seem to provide novel ideas for reinvigorating therapeutic approaches to cancer.
Collapse
Affiliation(s)
- Bo Wu
- grid.459742.90000 0004 1798 5889Department of General Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Xiang Shi
- grid.459742.90000 0004 1798 5889Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042 China
| | - Meixi Jiang
- grid.412644.10000 0004 5909 0696Department of Neurology, The Fourth Affiliated Hospital, China Medical University, Shenyang, 110032 China
| | - Hongxu Liu
- Department of Thoracic Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, 110042, China.
| |
Collapse
|
13
|
VanLandingham NK, Nazarenko A, Grandis JR, Johnson DE. The mutational profiles and corresponding therapeutic implications of PI3K mutations in cancer. Adv Biol Regul 2023; 87:100934. [PMID: 36402737 PMCID: PMC9992323 DOI: 10.1016/j.jbior.2022.100934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/04/2022] [Accepted: 11/10/2022] [Indexed: 11/16/2022]
Abstract
Genetic alterations of the PIK3CA gene, encoding the p110α catalytic subunit of PI3Kα enzyme, are found in a broad spectrum of human cancers. Many cancer-associated PIK3CA mutations occur at 3 hotspot locations and are termed canonical mutations. Canonical mutations result in hyperactivation of PI3K and promote oncogenesis via the PI3K/AKT/mTOR and PI3K/COX-2/PGE2 signaling pathways. These mutations also may serve as predictive biomarkers of response to PI3K inhibitors, as well as NSAID therapy. A large number of non-canonical PIK3CA mutations have also been identified in human tumors, but their functional properties are poorly understood. Here we review the landscape of PIK3CA mutations in different cancers and efforts underway to define the functional properties of non-canonical PIK3CA mutations. In addition, we summarize what has been learned from clinical trials of PI3K inhibitors as well as current trials incorporating these molecular targeting agents.
Collapse
Affiliation(s)
- Nathan K VanLandingham
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, CA, USA
| | | | - Jennifer R Grandis
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, CA, USA
| | - Daniel E Johnson
- Department of Otolaryngology - Head and Neck Surgery, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
14
|
Wang Y, Wang Y, Ren Y, Zhang Q, Yi P, Cheng C. Metabolic modulation of immune checkpoints and novel therapeutic strategies in cancer. Semin Cancer Biol 2022; 86:542-565. [PMID: 35151845 DOI: 10.1016/j.semcancer.2022.02.010] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/08/2021] [Accepted: 02/05/2022] [Indexed: 02/07/2023]
Abstract
Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) or programmed death-1 (PD-1)/programmed death-ligand 1 (PD-L1)-based immune checkpoint inhibitors (ICIs) have led to significant improvements in the overall survival of patients with certain cancers and are expected to benefit patients by achieving complete, long-lasting remissions and cure. However, some patients who receive ICIs either fail treatment or eventually develop immunotherapy resistance. The existence of such patients necessitates a deeper understanding of cancer progression, specifically nutrient regulation in the tumor microenvironment (TME), which includes both metabolic cross-talk between metabolites and tumor cells, and intracellular metabolism in immune and cancer cells. Here we review the features and behaviors of the TME and discuss the recently identified major immune checkpoints. We comprehensively and systematically summarize the metabolic modulation of tumor immunity and immune checkpoints in the TME, including glycolysis, amino acid metabolism, lipid metabolism, and other metabolic pathways, and further discuss the potential metabolism-based therapeutic strategies tested in preclinical and clinical settings. These findings will help to determine the existence of a link or crosstalk between tumor metabolism and immunotherapy, which will provide an important insight into cancer treatment and cancer research.
Collapse
Affiliation(s)
- Yi Wang
- Health Management Center, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, 610072, China
| | - Yuya Wang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China
| | - Yifei Ren
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China; Department of Obstetrics and Gynecology, Daping Hospital, Army Medical Center, Chongqing, 400038, China
| | - Qi Zhang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, China
| | - Ping Yi
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, 401120, China.
| | - Chunming Cheng
- Department of Radiation Oncology, James Comprehensive Cancer Center and College of Medicine at The Ohio State University, Columbus, OH, 43221, United States.
| |
Collapse
|
15
|
Immunoregulatory signal networks and tumor immune evasion mechanisms: insights into therapeutic targets and agents in clinical development. Biochem J 2022; 479:2219-2260. [DOI: 10.1042/bcj20210233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/03/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022]
Abstract
Through activation of immune cells, the immune system is responsible for identifying and destroying infected or otherwise damaged cells including tumorigenic cells that can be recognized as foreign, thus maintaining homeostasis. However, tumor cells have evolved several mechanisms to avoid immune cell detection and killing, resulting in tumor growth and progression. In the tumor microenvironment, tumor infiltrating immune cells are inactivated by soluble factors or tumor promoting conditions and lose their effects on tumor cells. Analysis of signaling and crosstalk between immune cells and tumor cells have helped us to understand in more detail the mechanisms of tumor immune evasion and this forms basis for drug development strategies in the area of cancer immunotherapy. In this review, we will summarize the dominant signaling networks involved in immune escape and describe the status of development of therapeutic strategies to target tumor immune evasion mechanisms with focus on how the tumor microenvironment interacts with T cells.
Collapse
|
16
|
Saito RDF, Andrade LNDS, Bustos SO, Chammas R. Phosphatidylcholine-Derived Lipid Mediators: The Crosstalk Between Cancer Cells and Immune Cells. Front Immunol 2022; 13:768606. [PMID: 35250970 PMCID: PMC8889569 DOI: 10.3389/fimmu.2022.768606] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/13/2022] [Indexed: 01/16/2023] Open
Abstract
To become resistant, cancer cells need to activate and maintain molecular defense mechanisms that depend on an energy trade-off between resistance and essential functions. Metabolic reprogramming has been shown to fuel cell growth and contribute to cancer drug resistance. Recently, changes in lipid metabolism have emerged as an important driver of resistance to anticancer agents. In this review, we highlight the role of choline metabolism with a focus on the phosphatidylcholine cycle in the regulation of resistance to therapy. We analyze the contribution of phosphatidylcholine and its metabolites to intracellular processes of cancer cells, both as the major cell membrane constituents and source of energy. We further extended our discussion about the role of phosphatidylcholine-derived lipid mediators in cellular communication between cancer and immune cells within the tumor microenvironment, as well as their pivotal role in the immune regulation of therapeutic failure. Changes in phosphatidylcholine metabolism are part of an adaptive program activated in response to stress conditions that contribute to cancer therapy resistance and open therapeutic opportunities for treating drug-resistant cancers.
Collapse
Affiliation(s)
- Renata de Freitas Saito
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Luciana Nogueira de Sousa Andrade
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Silvina Odete Bustos
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| | - Roger Chammas
- Centro de Investigação Translacional em Oncologia (LIM24), Departamento de Radiologia e Oncologia, Faculdade de Medicina da Universidade de São Paulo and Instituto do Câncer do Estado de São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Lipid metabolism and neutrophil function. Cell Immunol 2022; 377:104546. [DOI: 10.1016/j.cellimm.2022.104546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/12/2022] [Accepted: 05/19/2022] [Indexed: 11/22/2022]
|
18
|
Dianat-Moghadam H, Mahari A, Salahlou R, Khalili M, Azizi M, Sadeghzadeh H. Immune evader cancer stem cells direct the perspective approaches to cancer immunotherapy. Stem Cell Res Ther 2022; 13:150. [PMID: 35395787 PMCID: PMC8994338 DOI: 10.1186/s13287-022-02829-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/22/2022] [Indexed: 12/12/2022] Open
Abstract
Exploration of tumor immunity leads to the development of immune checkpoint inhibitors and cell-based immunotherapies which improve the clinical outcomes in several tumor types. However, the poor clinical efficacy of these treatments observed for other tumors could be attributed to the inherent complex tumor microenvironment (TME), cellular heterogeneity, and stemness driven by cancer stem cells (CSCs). CSC-specific characteristics provide the bulk tumor surveillance and resistance to entire eradication upon conventional therapies. CSCs-immune cells crosstalk creates an immunosuppressive TME that reshapes the stemness in tumor cells, resulting in tumor formation and progression. Thus, identifying the immunological features of CSCs could introduce the therapeutic targets with powerful antitumor responses. In this review, we summarized the role of immune cells providing CSCs to evade tumor immunity, and then discussed the intrinsic mechanisms represented by CSCs to promote tumors' resistance to immunotherapies. Then, we outlined potent immunotherapeutic interventions followed by a perspective outlook on the use of nanomedicine-based drug delivery systems for controlled modulation of the immune system.
Collapse
Affiliation(s)
- Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amir Mahari
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - Reza Salahlou
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mostafa Khalili
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - Mehdi Azizi
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hadi Sadeghzadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
19
|
Wang C, Zhang X, Luo L, Luo Y, Yang X, Ding X, Wang L, Le H, Feldman LER, Men X, Yan C, Huang W, Feng Y, Liu F, Yang XO, Liu M. Adipocyte-derived PGE2 is required for intermittent fasting-induced Treg proliferation and improvement of insulin sensitivity. JCI Insight 2022; 7:153755. [PMID: 35260536 PMCID: PMC8983131 DOI: 10.1172/jci.insight.153755] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/26/2022] [Indexed: 02/01/2023] Open
Abstract
The intermittent fasting (IF) diet has profound benefits for diabetes prevention. However, the precise mechanisms underlying IF's beneficial effects remain poorly defined. Here, we show that the expression levels of cyclooxygenase-2 (COX-2), an enzyme that produces prostaglandins, are suppressed in white adipose tissue (WAT) of obese humans. In addition, the expression of COX-2 in WAT is markedly upregulated by IF in obese mice. Adipocyte-specific depletion of COX-2 led to reduced fractions of CD4+Foxp3+ Tregs and a substantial decrease in the frequency of CD206+ macrophages, an increase in the abundance of γδT cells in WAT under normal chow diet conditions, and attenuation of IF-induced antiinflammatory and insulin-sensitizing effects, despite a similar antiobesity effect in obese mice. Mechanistically, adipocyte-derived prostaglandin E2 (PGE2) promoted Treg proliferation through the CaMKII pathway in vitro and rescued Treg populations in adipose tissue in COX-2-deficient mice. Ultimately, inactivation of Tregs by neutralizing anti-CD25 diminished IF-elicited antiinflammatory and insulin-sensitizing effects, and PGE2 restored the beneficial effects of IF in COX-2-KO mice. Collectively, our study reveals that adipocyte COX-2 is a key regulator of Treg proliferation and that adipocyte-derived PGE2 is essential for IF-elicited type 2 immune response and metabolic benefits.
Collapse
Affiliation(s)
- Chunqing Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Xing Zhang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Liping Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Yan Luo
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Xin Yang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Xiaofeng Ding
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Lu Wang
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Huyen Le
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Lily Elizabeth R. Feldman
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Xuebo Men
- Baodi Clinical College of Tian Jin Medical University, Tianjin, China
| | - Cen Yan
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Wendong Huang
- Department of Diabetes Complications & Metabolism Research, City of Hope, Duarte, California, USA
| | - Yingmei Feng
- Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Feng Liu
- Metabolic Syndrome Research Center, Second Xiangya Hospital, Central South University, Changsha, China
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology and,Autophagy Inflammation and Metabolism Center for Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Meilian Liu
- Department of Biochemistry and Molecular Biology, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA.,Autophagy Inflammation and Metabolism Center for Biomedical Research Excellence, University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
20
|
Shiraki H, Tanaka S, Guo Y, Harada K, Hide I, Yasuda T, Sakai N. Potential role of inducible GPR3 expression under stimulated T cell conditions. J Pharmacol Sci 2022; 148:307-314. [PMID: 35177210 DOI: 10.1016/j.jphs.2022.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/01/2022] [Accepted: 01/11/2022] [Indexed: 02/06/2023] Open
Abstract
G protein-coupled receptor 3 (GPR3) constitutively activates Gαs proteins without any ligands and is predominantly expressed in neurons. Since the expression and physiological role of GPR3 in immune cells is still unknown, we examined the possible role of GPR3 in T lymphocytes. The expression of GPR3 was upregulated 2 h after phorbol 12-myristate 13-acetate (PMA)/ionomycin stimulation and was sustained in Jurkat cells, a human T lymphocyte cell line. In addition, the expression of nuclear receptor 4 group A member 2 (NR4A2) was highly modulated by GPR3 expression. Additionally, GPR3 expression was linked with the transcriptional promoter activity of NR4A in Jurkat cells. In mouse CD4+ T cells, transient GPR3 expression was induced immediately after the antigen receptor stimulation. However, the expression of NR4A2 was not modulated in CD4+ T cells from GPR3-knockout mice after stimulation, and the population of Treg cells in thymocytes and splenocytes was not affected by GPR3 knockout. By contrast, spontaneous effector activation in both CD4+ T cells and CD8+ T cells was observed in GPR3-knockout mice. In summary, GPR3 is immediately induced by T cell stimulation and play an important role in the suppression of effector T cell activation.
Collapse
Affiliation(s)
- Hiroko Shiraki
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Shigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan.
| | - Yun Guo
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Kana Harada
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Tomoharu Yasuda
- Department of Immunology, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
| |
Collapse
|
21
|
Han L, Shi H, Ma S, Luo Y, Sun W, Li S, Zhang N, Jiang X, Gao Y, Huang Z, Xie C, Gong Y. Agrin Promotes Non-Small Cell Lung Cancer Progression and Stimulates Regulatory T Cells via Increasing IL-6 Secretion Through PI3K/AKT Pathway. Front Oncol 2022; 11:804418. [PMID: 35111682 PMCID: PMC8801576 DOI: 10.3389/fonc.2021.804418] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/23/2021] [Indexed: 01/04/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) has high mortality rates worldwide. Agrin contributes to immune synapse information and is involved in tumor metastasis. However, its roles in NSCLC and tumor immune microenvironment remain unclear. This study examined the effects and the underlying mechanisms of Agrin in NSCLC and tumor-infiltrated immune cells. Clinical tissue samples were used to confirm the bioinformatic predictions. NSCLC cells were used to investigate the effects of Agrin on cell cycle and proliferation, as well as invasion and migration. Tumor xenograft mouse model was used to confirm the effects of Agrin on NSCLC growth and tumor-infiltrated regulatory T cells (Tregs) in vivo. Agrin levels in NSCLC cells were closely related to tumor progression and metastasis, and its function was enriched in the PI3K/AKT pathway. In vitro assays demonstrated that Agrin knockdown suppressed NSCLC cell proliferation and metastasis, while PI3K/AKT activators reversed the inhibitory effects of Agrin deficiency on NSCLC cell behaviors. Agrin expression was negatively associated with immunotherapy responses in NSCLC patients. Agrin knockdown suppressed Tregs, as well as interleukin (IL)-6 expression and secretion, while PI3K/AKT activators and exogenous IL-6 rescued the inhibitory effects. In the mouse model, Agrin downregulation alleviated NSCLC cell growth and Treg infiltration in vivo. Our results indicated that Agrin promotes tumor cell growth and Treg infiltration via increasing IL-6 expression and secretion through PI3K/AKT pathway in NSCLC. Our studies suggested Agrin as a therapeutically potential target to increase the efficacy of immunotherapy in NSCLC patients.
Collapse
Affiliation(s)
- Linzhi Han
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Hongjie Shi
- Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shijing Ma
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuan Luo
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wenjie Sun
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Shuying Li
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Nannan Zhang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Xueping Jiang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yanping Gao
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhengrong Huang
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Conghua Xie
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan, China.,Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yan Gong
- Department of Biological Repositories, Zhongnan Hospital of Wuhan University, Wuhan, China.,Tumor Precision Diagnosis and Treatment Technology and Translational Medicine, Hubei Engineering Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
22
|
Yuan Y, Li H, Pu W, Chen L, Guo D, Jiang H, He B, Qin S, Wang K, Li N, Feng J, Wen J, Cheng S, Zhang Y, Yang W, Ye D, Lu Z, Huang C, Mei J, Zhang HF, Gao P, Jiang P, Su S, Sun B, Zhao SM. Cancer metabolism and tumor microenvironment: fostering each other? SCIENCE CHINA. LIFE SCIENCES 2022; 65:236-279. [PMID: 34846643 DOI: 10.1007/s11427-021-1999-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 08/19/2021] [Indexed: 02/06/2023]
Abstract
The changes associated with malignancy are not only in cancer cells but also in environment in which cancer cells live. Metabolic reprogramming supports tumor cell high demand of biogenesis for their rapid proliferation, and helps tumor cell to survive under certain genetic or environmental stresses. Emerging evidence suggests that metabolic alteration is ultimately and tightly associated with genetic changes, in particular the dysregulation of key oncogenic and tumor suppressive signaling pathways. Cancer cells activate HIF signaling even in the presence of oxygen and in the absence of growth factor stimulation. This cancer metabolic phenotype, described firstly by German physiologist Otto Warburg, insures enhanced glycolytic metabolism for the biosynthesis of macromolecules. The conception of metabolite signaling, i.e., metabolites are regulators of cell signaling, provides novel insights into how reactive oxygen species (ROS) and other metabolites deregulation may regulate redox homeostasis, epigenetics, and proliferation of cancer cells. Moreover, the unveiling of noncanonical functions of metabolic enzymes, such as the moonlighting functions of phosphoglycerate kinase 1 (PGK1), reassures the importance of metabolism in cancer development. The metabolic, microRNAs, and ncRNAs alterations in cancer cells can be sorted and delivered either to intercellular matrix or to cancer adjacent cells to shape cancer microenvironment via media such as exosome. Among them, cancer microenvironmental cells are immune cells which exert profound effects on cancer cells. Understanding of all these processes is a prerequisite for the development of a more effective strategy to contain cancers.
Collapse
Affiliation(s)
- Yiyuan Yuan
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200438, China
| | - Huimin Li
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Wang Pu
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences and School of Life Sciences, Fudan University, Shanghai, 200032, China
| | - Leilei Chen
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences and School of Life Sciences, Fudan University, Shanghai, 200032, China
| | - Dong Guo
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Hongfei Jiang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China
| | - Bo He
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Siyuan Qin
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kui Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Na Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jingwei Feng
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jing Wen
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Shipeng Cheng
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Yaguang Zhang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China
| | - Weiwei Yang
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Dan Ye
- Molecular and Cell Biology Lab, Institutes of Biomedical Sciences and School of Life Sciences, Fudan University, Shanghai, 200032, China.
| | - Zhimin Lu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, 310029, China.
| | - Canhua Huang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Jun Mei
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hua-Feng Zhang
- CAS Centre for Excellence in Cell and Molecular Biology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
| | - Ping Gao
- School of Medicine, Institutes for Life Sciences, South China University of Technology, Guangzhou, 510006, China.
| | - Peng Jiang
- Tsinghua University School of Life Sciences, and Tsinghua-Peking Center for Life Sciences, Beijing, 100084, China.
| | - Shicheng Su
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Bing Sun
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, 200031, China. .,School of Life Science and Technology, ShanghaiTech University, Shanghai, 201210, China.
| | - Shi-Min Zhao
- Obstetrics & Gynecology Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, 200438, China.
| |
Collapse
|
23
|
Kojima F, Sekiya H, Hioki Y, Kashiwagi H, Kubo M, Nakamura M, Maehana S, Imamichi Y, Yuhki KI, Ushikubi F, Kitasato H, Ichikawa T. Facilitation of colonic T cell immune responses is associated with an exacerbation of dextran sodium sulfate-induced colitis in mice lacking microsomal prostaglandin E synthase-1. Inflamm Regen 2022; 42:1. [PMID: 34983695 PMCID: PMC8725565 DOI: 10.1186/s41232-021-00188-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/07/2021] [Indexed: 12/24/2022] Open
Abstract
Background Microsomal prostaglandin E synthase-1 (mPGES-1) is a key enzyme that acts downstream of cyclooxygenase and plays a major role in inflammation by converting prostaglandin (PG) H2 to PGE2. The present study investigated the effect of genetic deletion of mPGES-1 on the development of immunologic responses to experimental colitis induced by dextran sodium sulfate (DSS), a well-established model of inflammatory bowel disease (IBD). Methods Colitis was induced in mice lacking mPGES-1 (mPGES-1−/− mice) and wild-type (WT) mice by administering DSS for 7 days. Colitis was assessed by body weight loss, diarrhea, fecal bleeding, and histological features. The colonic expression of mPGES-1 was determined by real-time PCR, western blotting, and immunohistochemistry. The impact of mPGES-1 deficiency on T cell immunity was determined by flow cytometry and T cell depletion in vivo. Results After administration of DSS, mPGES-1−/− mice exhibited more severe weight loss, diarrhea, and fecal bleeding than WT mice. Histological analysis further showed significant exacerbation of colonic inflammation in mPGES-1−/− mice. In WT mice, the colonic expression of mPGES-1 was highly induced on both mRNA and protein levels and colonic PGE2 increased significantly after DSS administration. Additionally, mPGES-1 protein was localized in the colonic mucosal epithelium and infiltrated inflammatory cells in underlying connective tissues and the lamina propria. The abnormalities consistent with colitis in mPGES-1−/− mice were associated with higher expression of colonic T-helper (Th)17 and Th1 cytokines, including interleukin 17A and interferon-γ. Furthermore, lack of mPGES-1 increased the numbers of Th17 and Th1 cells in the lamina propria mononuclear cells within the colon, even though the number of suppressive regulatory T cells also increased. CD4+ T cell depletion effectively reduced symptoms of colitis as well as colonic expression of Th17 and Th1 cytokines in mPGES-1−/− mice, suggesting the requirement of CD4+ T cells in the exacerbation of DSS-induced colitis under mPGES-1 deficiency. Conclusions These results demonstrate that mPGES-1 is the main enzyme responsible for colonic PGE2 production and deficiency of mPGES-1 facilitates the development of colitis by affecting the development of colonic T cell–mediated immunity. mPGES-1 might therefore impact both the intestinal inflammation and T cell–mediated immunity associated with IBD. Supplementary Information The online version contains supplementary material available at 10.1186/s41232-021-00188-1.
Collapse
Affiliation(s)
- Fumiaki Kojima
- Department of Pharmacology, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan. .,Department of Regulation Biochemistry, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan. .,Regenerative Medicine and Cell Design Research Facility, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan.
| | - Hiroki Sekiya
- Department of Regulation Biochemistry, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Yuka Hioki
- Department of Pharmacology, Kitasato University School of Allied Health Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Hitoshi Kashiwagi
- Department of Pharmacology, Asahikawa Medical University, 2-1-1-1 Midorigaoka higashi, Asahikawa, 078-8510, Japan
| | - Makoto Kubo
- Regenerative Medicine and Cell Design Research Facility, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan.,Division of Clinical Immunology, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, 252-0373, Japan
| | - Masaki Nakamura
- Regenerative Medicine and Cell Design Research Facility, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan.,Department of Environmental Microbiology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Shotaro Maehana
- Regenerative Medicine and Cell Design Research Facility, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan.,Department of Environmental Microbiology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Yoshitaka Imamichi
- Department of Pharmacology, Asahikawa Medical University, 2-1-1-1 Midorigaoka higashi, Asahikawa, 078-8510, Japan
| | - Koh-Ichi Yuhki
- Department of Pharmacology, Asahikawa Medical University, 2-1-1-1 Midorigaoka higashi, Asahikawa, 078-8510, Japan
| | - Fumitaka Ushikubi
- Department of Pharmacology, Asahikawa Medical University, 2-1-1-1 Midorigaoka higashi, Asahikawa, 078-8510, Japan
| | - Hidero Kitasato
- Regenerative Medicine and Cell Design Research Facility, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan.,Department of Environmental Microbiology, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| | - Takafumi Ichikawa
- Department of Regulation Biochemistry, Kitasato University Graduate School of Medical Sciences, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan.,Regenerative Medicine and Cell Design Research Facility, 1-15-1 Kitasato, Sagamihara, 252-0373, Japan
| |
Collapse
|
24
|
Aaron N, Costa S, Rosen CJ, Qiang L. The Implications of Bone Marrow Adipose Tissue on Inflammaging. Front Endocrinol (Lausanne) 2022; 13:853765. [PMID: 35360075 PMCID: PMC8962663 DOI: 10.3389/fendo.2022.853765] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/16/2022] [Indexed: 12/30/2022] Open
Abstract
Once considered an inert filler of the bone cavity, bone marrow adipose tissue (BMAT) is now regarded as a metabolically active organ that plays versatile roles in endocrine function, hematopoiesis, bone homeostasis and metabolism, and, potentially, energy conservation. While the regulation of BMAT is inadequately understood, it is recognized as a unique and dynamic fat depot that is distinct from peripheral fat. As we age, bone marrow adipocytes (BMAds) accumulate throughout the bone marrow (BM) milieu to influence the microenvironment. This process is conceivably signaled by the secretion of adipocyte-derived factors including pro-inflammatory cytokines and adipokines. Adipokines participate in the development of a chronic state of low-grade systemic inflammation (inflammaging), which trigger changes in the immune system that are characterized by declining fidelity and efficiency and cause an imbalance between pro-inflammatory and anti-inflammatory networks. In this review, we discuss the local effects of BMAT on bone homeostasis and the hematopoietic niche, age-related inflammatory changes associated with BMAT accrual, and the downstream effect on endocrine function, energy expenditure, and metabolism. Furthermore, we address therapeutic strategies to prevent BMAT accumulation and associated dysfunction during aging. In sum, BMAT is emerging as a critical player in aging and its explicit characterization still requires further research.
Collapse
Affiliation(s)
- Nicole Aaron
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, United States
- Department of Pharmacology, Columbia University, New York, NY, United States
| | - Samantha Costa
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, ME, United States
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
| | - Clifford J. Rosen
- Center for Clinical and Translational Research, Maine Medical Center Research Institute, Scarborough, ME, United States
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME, United States
- *Correspondence: Clifford J. Rosen, ; Li Qiang,
| | - Li Qiang
- Naomi Berrie Diabetes Center, Columbia University, New York, NY, United States
- Department of Pathology, Columbia University, New York, NY, United States
- *Correspondence: Clifford J. Rosen, ; Li Qiang,
| |
Collapse
|
25
|
Marques RM, Gonzalez-Nunez M, Walker ME, Gomez EA, Colas RA, Montero-Melendez T, Perretti M, Dalli J. Loss of 15-lipoxygenase disrupts T reg differentiation altering their pro-resolving functions. Cell Death Differ 2021; 28:3140-3160. [PMID: 34040168 PMCID: PMC8563763 DOI: 10.1038/s41418-021-00807-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 05/08/2021] [Accepted: 05/11/2021] [Indexed: 02/04/2023] Open
Abstract
Regulatory T-cells (Tregs) are central in the maintenance of homeostasis and resolution of inflammation. However, the mechanisms that govern their differentiation and function are not completely understood. Herein, we demonstrate a central role for the lipid mediator biosynthetic enzyme 15-lipoxygenase (ALOX15) in regulating key aspects of Treg biology. Pharmacological inhibition or genetic deletion of ALOX15 in Tregs decreased FOXP3 expression, altered Treg transcriptional profile and shifted their metabolism. This was linked with an impaired ability of Alox15-deficient cells to exert their pro-resolving actions, including a decrease in their ability to upregulate macrophage efferocytosis and a downregulation of interferon gamma expression in Th1 cells. Incubation of Tregs with the ALOX15-derived specilized pro-resolving mediators (SPM)s Resolvin (Rv)D3 and RvD5n-3 DPA rescued FOXP3 expression in cells where ALOX15 activity was inhibited. In vivo, deletion of Alox15 led to increased vascular lipid load and expansion of Th1 cells in mice fed western diet, a phenomenon that was reversed when Alox15-deficient mice were reconstituted with wild type Tregs. Taken together these findings demonstrate a central role of pro-resolving lipid mediators in governing the differentiation of naive T-cells to Tregs.
Collapse
Affiliation(s)
- Raquel M Marques
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Maria Gonzalez-Nunez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Mary E Walker
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Esteban A Gomez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Romain A Colas
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
| | - Trinidad Montero-Melendez
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Mauro Perretti
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Jesmond Dalli
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK.
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK.
| |
Collapse
|
26
|
Lone AM, Giansanti P, Jørgensen MJ, Gjerga E, Dugourd A, Scholten A, Saez-Rodriguez J, Heck AJR, Taskén K. Systems approach reveals distinct and shared signaling networks of the four PGE 2 receptors in T cells. Sci Signal 2021; 14:eabc8579. [PMID: 34609894 DOI: 10.1126/scisignal.abc8579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anna M Lone
- Department of Cancer Immunology, Institute of Cancer Research, Oslo University Hospital, 0424 Oslo, Norway.,K.G. Jebsen Centre for Cancer Immunotherapy and K.G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, 0317 Oslo, Norway.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Piero Giansanti
- Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, University of Utrecht, 3584 CH Utrecht, Netherlands.,Chair of Proteomics and Bioanalytics, Technical University of Munich, Freising 85354, Germany
| | - Marthe Jøntvedt Jørgensen
- K.G. Jebsen Centre for Cancer Immunotherapy and K.G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, 0317 Oslo, Norway.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Enio Gjerga
- Joint Research Centre for Computational Biomedicine (JRC-Combine), RWTH-Aachen University Hospital, Faculty of Medicine, Aachen 52074, Germany.,Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University Hospital, Bioquant, Heidelberg University, Heidelberg 69120, Germany
| | - Aurelien Dugourd
- Joint Research Centre for Computational Biomedicine (JRC-Combine), RWTH-Aachen University Hospital, Faculty of Medicine, Aachen 52074, Germany.,Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University Hospital, Bioquant, Heidelberg University, Heidelberg 69120, Germany
| | - Arjen Scholten
- Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, University of Utrecht, 3584 CH Utrecht, Netherlands
| | - Julio Saez-Rodriguez
- Joint Research Centre for Computational Biomedicine (JRC-Combine), RWTH-Aachen University Hospital, Faculty of Medicine, Aachen 52074, Germany.,Faculty of Medicine, Institute for Computational Biomedicine, Heidelberg University Hospital, Bioquant, Heidelberg University, Heidelberg 69120, Germany
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Center for Biomolecular Research, University of Utrecht, 3584 CH Utrecht, Netherlands
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute of Cancer Research, Oslo University Hospital, 0424 Oslo, Norway.,K.G. Jebsen Centre for Cancer Immunotherapy and K.G. Jebsen Centre for B Cell Malignancies, Institute of Clinical Medicine, University of Oslo, 0317 Oslo, Norway.,Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| |
Collapse
|
27
|
Yaqub S, Bjørnbeth BA, Angelsen JH, Fristrup CW, Grønbech JE, Hemmingsson O, Isaksson B, Juel IS, Larsen PN, Lindell G, Mortensen FV, Mortensen KE, Rizell M, Sandström P, Sandvik OM, Sparrelid E, Taflin H, Taskén K. Aspirin as secondary prevention in colorectal cancer liver metastasis (ASAC trial): study protocol for a multicentre randomized placebo-controlled trial. Trials 2021; 22:642. [PMID: 34544470 PMCID: PMC8451095 DOI: 10.1186/s13063-021-05587-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 08/31/2021] [Indexed: 12/21/2022] Open
Abstract
Background Colorectal cancer is one the most common cancers in the western world with increasing incidence. Approximately 50% of the patients develop liver metastases. Resection of liver metastases is the treatment of choice although almost half of the resected patients get recurrence in the liver. Methods The ASAC trial is a Scandinavian, multicentre, double-blinded, randomized, placebo-controlled study to determine whether adjuvant treatment with low-dose aspirin (acetylsalicylic acid (ASA)) can improve disease-free survival in patients treated for colorectal cancer liver metastases (CRCLM). Up to 800 patients operated for CRCLM will be randomized to Arm#1 ASA 160 mg once daily or Arm#2 Placebo, for a period of 3 years or until disease recurrence. The patients will be recruited at all major hepatobiliary surgical units in Norway, Sweden and Denmark and have follow-up according to standard of care and the National Guidelines. Discussion The ASAC trial will be the first clinical interventional trial to assess the potential beneficial role of ASA in recurrence of CRCLM and survival. ASA is an inexpensive, well-tolerated and easily accessible drug that will be highly potential as adjuvant drug in secondary prevention of CRCLM if the study shows a beneficial effect. We will also determine the effect of ASA as adjuvant treatment on Health-Related Quality of Life and the cost-effectiveness. Trial registration ClinicalTrials.gov NCT03326791. Registered on 31 October 2017.
Collapse
Affiliation(s)
- Sheraz Yaqub
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway. .,Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| | - Bjørn Atle Bjørnbeth
- Department of Hepato-Pancreato-Biliary Surgery, Oslo University Hospital, Oslo, Norway
| | - Jon-Helge Angelsen
- Department of Acute and Digestive Surgery, Haukeland University Hospital, Bergen, Norway.,Departments of Clinical Medicine, University of Bergen, Bergen, Norway
| | | | - Jon Erik Grønbech
- Department of Gastrointestinal Surgery, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway.,Department of Clinical and Molecular Medicine, NTNU, Norwegian University of Science and Technology, Trondheim, Norway
| | - Oskar Hemmingsson
- Department of Surgical and Perioperative Sciences, Umeå University, Umea, Sweden
| | - Bengt Isaksson
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Ingebjørg Soterud Juel
- Department of Gastrointestinal Surgery, St. Olav's Hospital, Trondheim University Hospital, Trondheim, Norway
| | | | - Gert Lindell
- Department of Surgery, Skåne University Hospital, Lund, Sweden
| | | | - Kim Erlend Mortensen
- Department of Gastrointestinal Surgery, University Hospital of North, Tromsø, Norway
| | - Magnus Rizell
- Department of Transplantation, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Per Sandström
- Department of Surgery, County Council of Östergötland, Linköping, Sweden.,Department of Biomedical and Clinical Sciences, Division of Surgery, Orthopedics and Oncology, Linköping, Sweden
| | - Oddvar Mathias Sandvik
- Department of Gastrointestinal Surgery, Stavanger University Hospital, Stavanger, Norway
| | - Ernesto Sparrelid
- Division of Surgery, Department of Clinical Science, Intervention, and Technology (CLINTEC), Center for Digestive Diseases, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| | - Helena Taflin
- Department of Surgery, Institute of Clinical Sciences, University of Gothenburg, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Kjetil Taskén
- Institute for Cancer Research, Oslo University Hospital, and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | |
Collapse
|
28
|
Das UN. Molecular biochemical aspects of salt (sodium chloride) in inflammation and immune response with reference to hypertension and type 2 diabetes mellitus. Lipids Health Dis 2021; 20:83. [PMID: 34334139 PMCID: PMC8327432 DOI: 10.1186/s12944-021-01507-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 07/14/2021] [Indexed: 12/18/2022] Open
Abstract
Obesity, insulin resistance, type 2 diabetes mellitus (T2DM) and hypertension (HTN) are common that are associated with low-grade systemic inflammation. Diet, genetic factors, inflammation, and immunocytes and their cytokines play a role in their pathobiology. But the exact role of sodium, potassium, magnesium and other minerals, trace elements and vitamins in the pathogenesis of HTN and T2DM is not known. Recent studies showed that sodium and potassium can modulate oxidative stress, inflammation, alter the autonomic nervous system and induce dysfunction of the innate and adaptive immune responses in addition to their action on renin-angiotensin-aldosterone system. These actions of sodium, potassium and magnesium and other minerals, trace elements and vitamins are likely to be secondary to their action on pro-inflammatory cytokines IL-6, TNF-α and IL-17 and metabolism of essential fatty acids that may account for their involvement in the pathobiology of insulin resistance, T2DM, HTN and autoimmune diseases.
Collapse
Affiliation(s)
- Undurti N Das
- UND Life Sciences, 2221 NW 5th St, Battle Ground, WA, 98604, USA.
| |
Collapse
|
29
|
Schmid T, Brüne B. Prostanoids and Resolution of Inflammation - Beyond the Lipid-Mediator Class Switch. Front Immunol 2021; 12:714042. [PMID: 34322137 PMCID: PMC8312722 DOI: 10.3389/fimmu.2021.714042] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 06/30/2021] [Indexed: 12/19/2022] Open
Abstract
Bioactive lipid mediators play a major role in regulating inflammatory processes. Herein, early pro-inflammatory phases are characterized and regulated by prostanoids and leukotrienes, whereas specialized pro-resolving mediators (SPM), including lipoxins, resolvins, protectins, and maresins, dominate during the resolution phase. While pro-inflammatory properties of prostanoids have been studied extensively, their impact on later phases of the inflammatory process has been attributed mainly to their ability to initiate the lipid-mediator class switch towards SPM. Yet, there is accumulating evidence that prostanoids directly contribute to the resolution of inflammation and return to homeostasis. In this mini review, we summarize the current knowledge of the resolution-regulatory properties of prostanoids and discuss potential implications for anti-inflammatory, prostanoid-targeted therapeutic interventions.
Collapse
Affiliation(s)
- Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany.,German Cancer Consortium (DKTK) Partner Site Frankfurt, Frankfurt, Germany.,Frankfurt Cancer Institute, Goethe-University Frankfurt, Frankfurt, Germany.,Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt, Germany
| |
Collapse
|
30
|
Lone AM, Taskén K. Phosphoproteomics-Based Characterization of Prostaglandin E 2 Signaling in T Cells. Mol Pharmacol 2021; 99:370-382. [PMID: 33674363 DOI: 10.1124/molpharm.120.000170] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 03/01/2021] [Indexed: 12/24/2022] Open
Abstract
Prostaglandin E2 (PGE2) is a key lipid mediator in health and disease and serves as a crucial link between the immune response and cancer. With the advent of cancer therapies targeting PGE2 signaling pathways at different levels, there has been increased interest in mapping and understanding the complex and interconnected signaling pathways arising from the four distinct PGE2 receptors. Here, we review phosphoproteomics studies that have investigated different aspects of PGE2 signaling in T cells. These studies have elucidated PGE2's regulatory effect on T cell receptor signaling and T cell function, the key role of protein kinase A in many PGE2 signaling pathways, the temporal regulation of PGE2 signaling, differences in PGE2 signaling between different T cell subtypes, and finally, the crosstalk between PGE2 signaling pathways elicited by the four distinct PGE2 receptors present in T cells. SIGNIFICANCE STATEMENT: Through the reviewed studies, we now have a much better understanding of PGE2's signaling mechanisms and functional roles in T cells, as well as a solid platform for targeted and functional studies of specific PGE2-triggered pathways in T cells.
Collapse
Affiliation(s)
- Anna Mari Lone
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital (A.M.L., K.T.) and Institute for Clinical Medicine, University of Oslo, Oslo, Norway (K.T.)
| | - Kjetil Taskén
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital (A.M.L., K.T.) and Institute for Clinical Medicine, University of Oslo, Oslo, Norway (K.T.)
| |
Collapse
|
31
|
Tavares Pereira M, Nowaczyk R, Payan-Carreira R, Miranda S, Aslan S, Kaya D, Kowalewski MP. Selected Uterine Immune Events Associated With the Establishment of Pregnancy in the Dog. Front Vet Sci 2021; 7:625921. [PMID: 33634180 PMCID: PMC7900146 DOI: 10.3389/fvets.2020.625921] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/07/2020] [Indexed: 11/13/2022] Open
Abstract
In the dog, implantation takes place at approximately 17 days of embryonal life and, while exposed to relatively high circulating progesterone concentrations, embryos presence is required for the formation of decidua. Furthermore, a balance between pro- and anti-inflammatory responses in conceptus-maternal communication is crucial for the onset of pregnancy. Strikingly, the understanding of such immune mechanisms in canine reproduction is still elusive. Here, canine uterine samples from pre-implantation (day 10-12, E+) and corresponding non-pregnant controls (E-), implantation (day 17, Imp) and post-implantation (day 18-25, Post-Imp) stages of pregnancy were used to investigate the expression and localization of several immune-related factors. The most important findings indicate increased availability of CD4, MHCII, NCR1, IDO1, AIF1, CD25, CCR7, and IL6 in response to embryo presence (E+), while FoxP3 and CCL3 were more abundant in E- samples. Implantation was characterized by upregulated levels of FoxP3, IL12a, ENG, and CDH1, whereas CD4, CCR7, IL8, and -10 were less represented. Following implantation, decreased transcript levels of TNFR1, MHCII, NCR1, TLR4, CD206, FoxP3, and IL12a were observed concomitantly with the highest expression of IL6 and IL1β. MHCII, CD86, CD206, CD163, TNFα, IDO1, and AIF1 were immunolocalized in macrophages, CD4 and Nkp46 in lymphocytes, and some signals of IDO1, AIF1, and TNF-receptors could also be identified in endothelial cells and/or uterine glands. Cumulatively, new insights regarding uterine immunity in the peri-implantation period are provided, with apparent moderated pro-inflammatory signals prevailing during pre-implantation, while implantation and early trophoblast invasion appear to be associated with immunomodulatory and rather anti-inflammatory conditions.
Collapse
Affiliation(s)
- Miguel Tavares Pereira
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland
| | - Renata Nowaczyk
- Division of Histology and Embryology, Department of Biostructure and Animal Physiology, Faculty of Veterinary Medicine, Wroclaw University of Environmental and Life Sciences, Wroclaw, Poland
| | - Rita Payan-Carreira
- Mediterranean Institute for Agriculture, Environment (MED) and Department of Veterinary Medicine, University of Évora, Évora, Portugal
| | - Sonia Miranda
- Animal and Veterinary Research Center (CECAV), University of Trás-os-Montes and Alto Douro, Vila Real, Portugal
| | - Selim Aslan
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Near East University, Nicosia, Cyprus
| | - Duygu Kaya
- Department of Obstetrics and Gynecology, Faculty of Veterinary Medicine, Kafkas University, Kars, Turkey
| | - Mariusz P Kowalewski
- Institute of Veterinary Anatomy, Vetsuisse Faculty, University of Zurich (UZH), Zurich, Switzerland
| |
Collapse
|
32
|
Weng CY, Kao CX, Chang TS, Huang YH. Immuno-Metabolism: The Role of Cancer Niche in Immune Checkpoint Inhibitor Resistance. Int J Mol Sci 2021; 22:1258. [PMID: 33514004 PMCID: PMC7865434 DOI: 10.3390/ijms22031258] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
The use of immune checkpoint inhibitors (ICI) in treating cancer has revolutionized the approach to eradicate cancer cells by reactivating immune responses. However, only a subset of patients benefits from this treatment; the majority remains unresponsive or develops resistance to ICI therapy. Increasing evidence suggests that metabolic machinery in the tumor microenvironment (TME) plays a role in the development of ICI resistance. Within the TME, nutrients and oxygen are scarce, forcing immune cells to undergo metabolic reprogramming to adapt to harsh conditions. Cancer-induced metabolic deregulation in immune cells can attenuate their anti-cancer properties, but can also increase their immunosuppressive properties. Therefore, targeting metabolic pathways of immune cells in the TME may strengthen the efficacy of ICIs and prevent ICI resistance. In this review, we discuss the interactions of immune cells and metabolic alterations in the TME. We also discuss current therapies targeting cellular metabolism in combination with ICIs for the treatment of cancer, and provide possible mechanisms behind the cellular metabolic rewiring that may improve clinical outcomes.
Collapse
Affiliation(s)
- Chao-Yuan Weng
- School of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan;
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
| | - Cheng-Xiang Kao
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Te-Sheng Chang
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan 33382, Taiwan
- Division of Internal Medicine, Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Chiayi 61363, Taiwan
| | - Yen-Hua Huang
- Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan;
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cell Therapy and Regeneration Medicine, Taipei Medical University, Taipei 11031, Taiwan
- International Ph.D. Program for Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
- TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei 11031, Taiwan
- Center for Reproductive Medicine, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
- Comprehensive Cancer Center of Taipei Medical University, Taipei 11031, Taiwan
- PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
33
|
The Association of Gut Microbiota and Treg Dysfunction in Autoimmune Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:191-203. [PMID: 33523449 PMCID: PMC9290759 DOI: 10.1007/978-981-15-6407-9_10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Autoimmune conditions affect 23 million Americans or 7% of the US population. There are more than 100 autoimmune disorders, affecting every major organ system in humans. This chapter aims to further explain Treg dysfunction autoimmune disorders, including monogenic primary immune deficiency such as immune dysregulation polyendocrinopathy, enteropathy, X-linked inheritance (IPEX) syndrome, and polygenic autoimmune diseases with Treg dysfunction such as multiple sclerosis (MS), inflammatory bowel disease (IBD), and food allergy. These conditions are associated with an abnormal small intestinal and colonic microbiome. Some disorders clearly improve with therapies aimed at microbial modification, including probiotics and fecal microbiota transplantation (FMT). Approaches to prevent and treat these disorders will need to focus on the acquisition and maintenance of a healthy colonic microbiota, in addition to more focused approaches at immune suppression during acute disease exacerbations.
Collapse
|
34
|
Sajiki Y, Konnai S, Goto S, Okagawa T, Ohira K, Shimakura H, Maekawa N, Gondaira S, Higuchi H, Tajima M, Hirano Y, Kohara J, Murata S, Ohashi K. The Suppression of Th1 Response by Inducing TGF-β1 From Regulatory T Cells in Bovine Mycoplasmosis. Front Vet Sci 2020; 7:609443. [PMID: 33344537 PMCID: PMC7738317 DOI: 10.3389/fvets.2020.609443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/11/2020] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Tregs) regulate immune responses and maintain host immune homeostasis. Tregs contribute to the disease progression of several chronic infections by oversuppressing immune responses via the secretion of immunosuppressive cytokines, such as transforming growth factor (TGF)-β and interleukin-10. In the present study, we examined the association of Tregs with Mycoplasma bovis infection, in which immunosuppression is frequently observed. Compared with uninfected cattle, the percentage of Tregs, CD4+CD25highFoxp3+ T cells, was increased in M. bovis-infected cattle. Additionally, the plasma of M. bovis-infected cattle contained the high concentrations of TGF-β1, and M. bovis infection induced TGF-β1 production from bovine immune cells in in vitro cultures. Finally, we analyzed the immunosuppressive effects of TGF-β1 on bovine immune cells. Treatment with TGF-β1 significantly decreased the expression of CD69, an activation marker, in T cells, and Th1 cytokine production in vitro. These results suggest that the increase in Tregs and TGF-β1 secretion could be one of the immunosuppressive mechanisms and that lead to increased susceptibility to other infections in terms of exacerbation of disease during M. bovis infection.
Collapse
Affiliation(s)
- Yamato Sajiki
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Satoru Konnai
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Shinya Goto
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Tomohiro Okagawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kosuke Ohira
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Honami Shimakura
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Naoya Maekawa
- Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Satoshi Gondaira
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Hidetoshi Higuchi
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Motoshi Tajima
- School of Veterinary Medicine, Rakuno Gakuen University, Ebetsu, Japan
| | - Yuki Hirano
- Animal Research Center, Agriculture Research Department, Hokkaido Research Organization, Shintoku, Japan
| | - Junko Kohara
- Animal Research Center, Agriculture Research Department, Hokkaido Research Organization, Shintoku, Japan
| | - Shiro Murata
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kazuhiko Ohashi
- Department of Disease Control, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan.,Department of Advanced Pharmaceutics, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
35
|
Picado C, Roca-Ferrer J. Role of the Cyclooxygenase Pathway in the Association of Obstructive Sleep Apnea and Cancer. J Clin Med 2020; 9:E3237. [PMID: 33050416 PMCID: PMC7601393 DOI: 10.3390/jcm9103237] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 11/16/2022] Open
Abstract
The objective of this review is to examine the findings that link obstructive sleep apnea (OSA) with cancer and the role played by the cyclooxygenase (COX) pathway in this association. Epidemiological studies in humans suggest a link between OSA and increased cancer incidence and mortality. Studies carried out in animal models have shown that intermittent hypoxia (IH) induces changes in several signaling pathways involved in the regulation of host immunological surveillance that results in tumor establishment and invasion. IH induces the expression of cyclooxygenase 2 (COX-2) that results in an increased synthesis of prostaglandin E2 (PGE2). PGE2 modulates the function of multiple cells involved in immune responses including T lymphocytes, NK cells, dendritic cells, macrophages, and myeloid-derived suppressor cells. In a mouse model blockage of COX-2/PGE2 abrogated the pro-oncogenic effects of IH. Despite the fact that aspirin inhibits PGE2 production and prevents the development of cancer, none of the epidemiological studies that investigated the association of OSA and cancer included aspirin use in the analysis. Studies are needed to investigate the regulation of the COX-2/PGE2 pathway and PGE2 production in patients with OSA, to better define the role of this axis in the physiopathology of OSA and the potential role of aspirin in preventing the development of cancer.
Collapse
Affiliation(s)
- César Picado
- Hospital Clinic, Department of Medicine, Universitat de Barcelona, 08036 Barcelona, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto Carlos III, 28029 Madrid, Spain
| | - Jordi Roca-Ferrer
- Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBERES), Instituto Carlos III, 28029 Madrid, Spain
| |
Collapse
|
36
|
Liu Z, Wang J, Liu L, Yuan H, Bu Y, Feng J, Liu Y, Yang G, Zhao M, Yuan Y, Zhang H, Yun H, Zhang X. Chronic ethanol consumption and HBV induce abnormal lipid metabolism through HBx/SWELL1/arachidonic acid signaling and activate Tregs in HBV-Tg mice. Am J Cancer Res 2020; 10:9249-9267. [PMID: 32802190 PMCID: PMC7415795 DOI: 10.7150/thno.46005] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 07/09/2020] [Indexed: 02/07/2023] Open
Abstract
Rationale: Chronic ethanol consumption as a public health problem worldwide boosts the development of chronic liver diseases in hepatitis B virus (HBV)-infected patients. Arachidonic acid metabolite prostaglandin E2 (PGE2) activates regulatory T cells (Tregs) function. Here, we aim to investigate the underlying mechanism by which chronic ethanol consumption enriches the HBV-induced abnormal lipid metabolism and Tregs. Methods: The si-RNAs were used to weaken the expression of SWELL1 in HepG2, HepG2.2.15 and K180 cancer cell lines, followed by RNA sequencing from HepG2 cells. Arachidonic acid metabolite PGE2 and LTD4 were measured by ELISA assay in vivo and in vitro. Western blot analysis and RT-qPCR were used to examine HBx and SWELL1 and transcriptional factor Sp1 in clinical HCC samples and cell lines. The effect of chronic ethanol consumption on Tregs was tested by flow cytometry in HBV-Tg mice. The splenic Tregs were collected and analyzed by RNA sequencing. Results: The cooperative effect of ethanol and HBV in abnormal lipid metabolism was observed in vivo and in vitro. The depression of SWELL1 (or HBx) resulted in the reduction of lipid content and arachidonic acid metabolite, correlating with suppression of relative gene atlas. Ethanol and SWELL1 elevated the levels of PGE2 or LTD4 in the liver of mice and cell lines. Interestingly, the ethanol modulated abnormal lipid metabolism through activating HBx/Sp1/SWELL1/arachidonic acid signaling. Chronic ethanol consumption remarkably increased the population of PBL Tregs and splenic Tregs in HBV-Tg mice, consistently with the enhanced expression of PD-L1 in vivo and in vitro. Mechanically, RNA-seq data showed that multiple genes were altered in the transcriptomic atlas of Tregs sorting from ethanol-fed mice or HBV-Tg mice. Conclusion: The chronic ethanol intake enriches the HBV-enhanced abnormal lipid metabolism through HBx/SWELL1/arachidonic acid signaling and activates Tregs in mice.
Collapse
|
37
|
Cui G, Yuan A, Li Z, Goll R, Florholmen J. ST2 and regulatory T cells in the colorectal adenoma/carcinoma microenvironment: implications for diseases progression and prognosis. Sci Rep 2020; 10:5892. [PMID: 32246094 PMCID: PMC7125220 DOI: 10.1038/s41598-020-62502-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 03/13/2020] [Indexed: 02/07/2023] Open
Abstract
ST2 (also known as IL1RL1) is the critical functional receptor for interleukin (IL)-33 in stimulating regulatory T cell (Treg) expansion and function in physiological and pathological conditions. We examined the correlation between ST2 cell expression and FoxP3 positive Tregs in both colorectal adenoma and cancer (CRC) microenvironment by real-time PCR, immunohistochemistry (IHC) and double immunofluorescences. The clinicopathological and prognostic significance of cellular ST2-positive cells and FoxP3-positive Tregs in patients with adenoma and CRC were evaluated. Real-time PCR results revealed increased expression levels of ST2 and FoxP3 mRNAs in both adenoma and CRC tissues as compared with control tissues. IHC analysis confirmed increased densities of ST2-positive cells in both the adenoma/CRC epithelium and stroma, which show a close positive linear association with the densities of FoxP3-positive Tregs in respective compartments. Pathological feature analysis showed that densities of ST2-positive cells in the tumor stroma were notably associated with degree of dysplastic grading in patients with adenoma, and disease stages and lymph node metastasis in patients with CRC. Kaplan-Meier survival curves suggested that CRC patients with high densities of ST2-positive cells in the stroma tend to have a shorter overall survival. We therefore concluded that increased densities of ST2-postive cells relate to Treg accumulation within the adenoma/CRC microenvironment, suggesting the IL-33/ST2 pathway as a potential contributor for immunosuppressive milieu formation that impact disease stage and prognosis in patients with CRC.
Collapse
Affiliation(s)
- Guanglin Cui
- Department of Gastroenterology, the Second Affiliated Hospital of Zhengzhou University, Henan, China. .,Faculty of Health Science, Nord University at Campus Levanger, Levanger, Norway.
| | - Aping Yuan
- Department of Gastroenterology, the Second Affiliated Hospital of Zhengzhou University, Henan, China
| | - Zhenfeng Li
- Department of Gastroenterology, the Second Affiliated Hospital of Zhengzhou University, Henan, China
| | - Rasmus Goll
- Department of Gastroenterology & Nutrition, University Hospital of North Norway, Tromsø, University of Tromsø, Tromsø, Norway
| | - Jon Florholmen
- Department of Gastroenterology & Nutrition, University Hospital of North Norway, Tromsø, University of Tromsø, Tromsø, Norway
| |
Collapse
|
38
|
Naamani O, Riff R, Chaimovitz C, Mazar J, Douvdevani A. Pharmacological preconditioning with adenosine A 1 receptor agonist induces immunosuppression and improves graft survival in novel allogeneic transplantation models. Sci Rep 2020; 10:4464. [PMID: 32161272 PMCID: PMC7066190 DOI: 10.1038/s41598-020-60224-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 02/10/2020] [Indexed: 12/19/2022] Open
Abstract
Adenosine is widely known as a potent modulator of innate and acquired immunity. It is released during transplants, and acts on four subtype receptors. In previous studies, we demonstrated that pharmacological preconditioning (PPC), pre-administration of the selective A1 receptor (A1R) agonist led to A1R desensitization, is followed by upregulation of the adenosine A2A receptor. This immunosuppressive effect resulted in lymphopenia, and it reduced T-cell reactivity. The aim of the current study was to challenge the immunosuppressive effects of A1R-PPC in models of allogeneic grafts. PPC mice were treated by intraperitoneal injection using specific adenosine A1R agonist 24 h and 12 h before starting any procedure. We challenged our method in novel allogeneic muscle and skin grafts models. Mice and grafts were assessed by complete blood counts, MLR from PPC splenocytes, and pathological evaluation. We found a significant reduction in WBC and lymphocyte counts in PPC-treated mice. Two-way MLR with splenocytes from PPC grafted mice showed decreased proliferation and anergy. Histology of PPC allogeneic grafts revealed profoundly less infiltration and even less muscle necrosis compared to vehicle treated allografts. Similar results observed in PPC skin transplantation. To conclude, PPC moderated graft rejection in separate allogeneic challenges, and reduced lymphocytes infiltration and ischemic damage.
Collapse
Affiliation(s)
- Oshri Naamani
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel.
- Department of Science, Hemdat Hadarom, College of Education, Netivot, Israel.
| | - Reut Riff
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Cidio Chaimovitz
- Department of Nephrology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Julia Mazar
- Laboratory of Hematology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| | - Amos Douvdevani
- Department of Clinical Biochemistry and Pharmacology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
- Department of Nephrology, Faculty of Health Sciences, Ben-Gurion University of the Negev and Soroka University Medical Center, Beer-Sheva, Israel
| |
Collapse
|
39
|
Take Y, Koizumi S, Nagahisa A. Prostaglandin E Receptor 4 Antagonist in Cancer Immunotherapy: Mechanisms of Action. Front Immunol 2020; 11:324. [PMID: 32210957 PMCID: PMC7076081 DOI: 10.3389/fimmu.2020.00324] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 02/10/2020] [Indexed: 12/20/2022] Open
Abstract
A highly expressed prostaglandin E2 (PGE2) in tumor tissues suppresses antitumor immunity in the tumor microenvironment (TME) and causes tumor immune evasion leading to disease progression. In animal studies, selective inhibition of the prostaglandin E receptor 4 (EP4), one of four PGE2 receptors, suppresses tumor growth, restoring the tumor immune response toward an antitumorigenic condition. This review summarizes PGE2/EP4 signal inhibition in relation to the cancer-immunity cycle (C-IC), which describes fundamental tumor-immune interactions in cancer immunotherapy. PGE2 is suggested to slow down C-IC by inhibiting natural killer cell functions, suppressing the supply of conventional dendritic cell precursors to the TME. This is critical for the tumor-associated antigen priming of CD8+ T cells and their translocation to the tumor tissue from the tumor-draining lymph node. Furthermore, PGE2 activates several key immune-suppressive cells present in tumors and counteracts tumoricidal properties of the effector CD8+ T cells. These effects of PGE2 drive the tumors to non-T-cell-inflamed tumors and cause refractory conditions to cancer immunotherapies, e.g., immune checkpoint inhibitor (ICI) treatment. EP4 antagonist therapy is suggested to inhibit the immune-suppressive and tumorigenic roles of PGE2 in tumors, and it may sensitize the therapeutic effects of ICIs in patients with non-inflamed and C-IC-deficient tumors. This review provides insight into the mechanism of action of EP4 antagonists in cancer immunotherapy and suggests a C-IC modulating opportunity for EP4 antagonist therapy in combination with ICIs and/or other cancer therapies.
Collapse
|
40
|
COX-2 Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1277:87-104. [PMID: 33119867 DOI: 10.1007/978-3-030-50224-9_6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumorigenesis is a multistep, complicated process, and many studies have been completed over the last few decades to elucidate this process. Increasingly, many studies have shifted focus toward the critical role of the tumor microenvironment (TME), which consists of cellular players, cell-cell communications, and extracellular matrix (ECM). In the TME, cyclooxygenase-2 (COX-2) has been found to be a key molecule mediating the microenvironment changes. COX-2 is an inducible form of the enzyme that converts arachidonic acid into the signal transduction molecules (thromboxanes and prostaglandins). COX-2 is frequently expressed in many types of cancers and has been closely linked to its occurrence, progression, and prognosis. For example, COX-2 has been shown to (1) regulate tumor cell growth, (2) promote tissue invasion and metastasis, (3) inhibit apoptosis, (4) suppress antitumor immunity, and (5) promote sustainable angiogenesis. In this chapter, we summarize recent advances of studies that have evaluated COX-2 signaling in TME.
Collapse
|
41
|
Lai ZZ, Yang HL, Ha SY, Chang KK, Mei J, Zhou WJ, Qiu XM, Wang XQ, Zhu R, Li DJ, Li MQ. Cyclooxygenase-2 in Endometriosis. Int J Biol Sci 2019; 15:2783-2797. [PMID: 31853218 PMCID: PMC6909960 DOI: 10.7150/ijbs.35128] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/28/2019] [Indexed: 12/11/2022] Open
Abstract
Endometriosis (EMS) is the most common gynecological disease in women of reproductive age, and it is associated with chronic pelvic pain, dyspareunia and infertility. As a consequence of genetic, immune and environmental factors, endometriotic lesions have high cyclooxygenase (COX)-2 and COX-2-derived prostaglandin E2 (PGE2) biosynthesis compared with the normal endometrium. The transcription of the PTGS2 gene for COX-2 is associated with multiple intracellular signals, which converge to cause the activation of mitogen-activated protein kinases (MAPKs). COX-2 expression can be regulated by several factors, such as estrogen, hypoxia, proinflammatory cytokines, environmental pollutants, metabolites and metabolic enzymes, and platelets. High concentrations of COX-2 lead to high cell proliferation, a low level of apoptosis, high invasion, angiogenesis, EMS-related pain and infertility. COX-2-derived PGE2 performs a crucial function in EMS development by binding to EP2 and EP4 receptors. These basic findings have contributed to COX-2-targeted treatment in EMS, including COX-2 inhibitors, hormone drugs and glycyrrhizin. In this review, we summarize the most recent basic research in detail and provide a short summary of COX-2-targeted treatment.
Collapse
Affiliation(s)
- Zhen-Zhen Lai
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Hui-Li Yang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Si-Yao Ha
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Kai-Kai Chang
- Department of Gynecology, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, People's Republic of China
| | - Jie Mei
- Reproductive Medicine Center, Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, People's Republic of China
| | - We-Jie Zhou
- Clinical and Translational Research Center, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai 201204, People's Republic of China
| | - Xue-Min Qiu
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Xiao-Qiu Wang
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Rui Zhu
- Center for Human Reproduction and Genetics, Suzhou Municipal Hospital, Suzhou 215008, People's Republic of China
| | - Da-Jin Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China
| | - Ming-Qing Li
- NHC Key Lab of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200080, People's Republic of China.,Shanghai Key Laboratory of Female Reproductive Endocrine Related Diseases, Hospital of Obstetrics and Gynecology, Fudan University, Shanghai 200011, People's Republic of China
| |
Collapse
|
42
|
Li J, Van Valkenburgh J, Hong X, Conti PS, Zhang X, Chen K. Small molecules as theranostic agents in cancer immunology. Theranostics 2019; 9:7849-7871. [PMID: 31695804 PMCID: PMC6831453 DOI: 10.7150/thno.37218] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 09/10/2019] [Indexed: 12/24/2022] Open
Abstract
With further research into the molecular mechanisms and roles linking immune suppression and restraint of (pre)malignancies, immunotherapies have revolutionized clinical strategies in the treatment of cancer. However, nearly 70% of patients who received immune checkpoint therapeutics showed no response. Complementary and/or synergistic effects may occur when extracellular checkpoint antibody blockades combine with small molecules targeting intracellular signal pathways up/downstream of immune checkpoints or regulating the innate and adaptive immune response. After radiolabeling with radionuclides, small molecules can also be used for estimating treatment efficacy of immune checkpoint blockades. This review not only highlights some significant intracellular pathways and immune-related targets such as the kynurenine pathway, purinergic signaling, the kinase signaling axis, chemokines, etc., but also summarizes some attractive and potentially immunosuppression-related small molecule agents, which may be synergistic with extracellular immune checkpoint blockade. In addition, opportunities for small molecule-based theranostics in cancer immunology will be discussed.
Collapse
Affiliation(s)
- Jindian Li
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Juno Van Valkenburgh
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| | - Xingfang Hong
- Laboratory of Pathogen Biology, School of Basic Medical Sciences, Dali University, Dali 671000, China
| | - Peter S. Conti
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| | - Xianzhong Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Kai Chen
- Molecular Imaging Center, Department of Radiology, Keck School of Medicine, University of Southern California, 2250 Alcazar Street, CSC103, Los Angeles, CA 90033, USA
| |
Collapse
|
43
|
Lorenzo-Sanz L, Muñoz P. Tumor-Infiltrating Immunosuppressive Cells in Cancer-Cell Plasticity, Tumor Progression and Therapy Response. CANCER MICROENVIRONMENT 2019; 12:119-132. [PMID: 31583529 DOI: 10.1007/s12307-019-00232-2] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Accepted: 09/01/2019] [Indexed: 12/16/2022]
Abstract
In most tumors, cancer cells show the ability to dynamically transit from a non-cancer stem-like cell to a cancer stem-like cell (CSC) state and vice versa. This cell plasticity has been associated with the epithelial-to-mesenchymal transition program (EMT) and can be regulated by tumor cell-intrinsic mechanisms and complex interactions with various tumor microenvironment (TME) components. These interactions favor the generation of a specific "CSC niche" that helps maintain the main properties, phenotypic plasticity and metastatic potential of this subset of tumor cells. For this reason, TME has been recognized as an important promoter of tumor progression and therapy resistance. Tumors have evolved a network of immunosuppressive mechanisms that limits the cytotoxic T cell response to cancer cells. Some key players in this network are tumor-associated macrophages, myeloid-derived suppressor cells and regulatory T cells, which not only favor a pro-tumoral and immunosuppressive environment that supports tumor growth and immune evasion, but also negatively influences immunotherapy. Here, we review the relevance of cytokines and growth factors provided by immunosuppressive immune cells in regulating cancer-cell plasticity. We also discuss how cancer cells remodel their own niche to promote proliferation, stemness and EMT, and escape immune surveillance. A better understanding of CSC-TME crosstalk signaling will enable the development of effective targeted or immune therapies that block tumor growth and metastasis.
Collapse
Affiliation(s)
- Laura Lorenzo-Sanz
- Aging and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Vía de L'Hospitalet 199-203, 08908, Barcelona, Spain
| | - Purificación Muñoz
- Aging and Cancer Group, Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Av. Gran Vía de L'Hospitalet 199-203, 08908, Barcelona, Spain.
| |
Collapse
|
44
|
Xu J, Su Y, Hu L, Cain A, Gu Y, Liu B, Wu R, Wang S, Wang H. Effect of Bone Morphogenetic Protein 6 on Immunomodulatory Functions of Salivary Gland-Derived Mesenchymal Stem Cells in Sjögren's Syndrome. Stem Cells Dev 2019; 27:1540-1548. [PMID: 30132383 DOI: 10.1089/scd.2017.0161] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Sjögren's syndrome (SS) is characterized by autoimmune activation and loss of function in the salivary glands. Recent studies reported that bone morphogenetic protein 6 (BMP6), which is a member of transforming growth factor beta (TGF-β) superfamily, was highly expressed in SS patients. To investigate the role of BMP6 in SS, we treated the salivary gland-derived mesenchymal stem cells (SGMSCs) with BMP6 and found that BMP6 could impair immunomodulatory properties of normal SGMSCs by downregulating the Prostaglandin E2 synthase through DNA-binding protein inhibitor-1. Neutralizing the BMP6 could significantly restore the SGMSC's immunoregulatory function in vitro and delay the SS disease activity in vivo. In conclusion, BMP6 could not only affect the secreting function of epithelial cells in the salivary gland but also influence the immunomodulatory properties of SGMSCs, which may trigger or enhance the autoimmune reflection in SS.
Collapse
Affiliation(s)
- Junji Xu
- 1 Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China .,2 Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda, Maryland
| | - Yingying Su
- 3 Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University , Beijing, China
| | - Lei Hu
- 1 Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China
| | - Alexander Cain
- 2 Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health , Bethesda, Maryland
| | - Yi Gu
- 4 Department of Pediatrics, Beijing Chaoyang Hospital, Capital Medical University , Beijing, China
| | - Bowen Liu
- 1 Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China
| | - Ruiqing Wu
- 3 Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University , Beijing, China
| | - Songlin Wang
- 1 Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, Capital Medical University School of Stomatology , Beijing, China
| | - Hao Wang
- 3 Department of Stomatology, Beijing Tiantan Hospital, Capital Medical University , Beijing, China
| |
Collapse
|
45
|
Maseda D, Ricciotti E, Crofford LJ. Prostaglandin regulation of T cell biology. Pharmacol Res 2019; 149:104456. [PMID: 31553935 DOI: 10.1016/j.phrs.2019.104456] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/06/2019] [Accepted: 09/13/2019] [Indexed: 12/26/2022]
Abstract
Prostaglandins (PG) are pleiotropic bioactive lipids involved in the control of many physiological processes, including key roles in regulating inflammation. This links PG to the modulation of the quality and magnitude of immune responses. T cells, as a core part of the immune system, respond readily to inflammatory cues from their environment, and express a diverse array of PG receptors that contribute to their function and phenotype. Here we put in context our knowledge about how PG affect T cell biology, and review advances that bring light into how specific T cell functions that have been newly discovered are modulated through PG. We will also comment on drugs that target PG metabolism and sensing, their effect on T cell function during disease, and we will finally discuss how we can design new approaches that modulate PG in order to maximize desired therapeutic T cell effects.
Collapse
Affiliation(s)
- Damian Maseda
- Department of Microbiology, University of Pennsylvania School of Medicine, 8-138 Smillow Center for Translational Research, Philadelphia, PA, USA.
| | - Emanuela Ricciotti
- Department of Systems Pharmacology and Translational Therapeutics, Institute for Translational Medicine and Therapeutics, University of Pennsylvania, Philadelphia, PA, USA
| | - Leslie J Crofford
- Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
46
|
Khan S, Andrews KL, Chin-Dusting JPF. Cyclo-Oxygenase (COX) Inhibitors and Cardiovascular Risk: Are Non-Steroidal Anti-Inflammatory Drugs Really Anti-Inflammatory? Int J Mol Sci 2019; 20:ijms20174262. [PMID: 31480335 PMCID: PMC6747368 DOI: 10.3390/ijms20174262] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 08/08/2019] [Indexed: 12/15/2022] Open
Abstract
Cyclo-oxygenase (COX) inhibitors are among the most commonly used drugs in the western world for their anti-inflammatory and analgesic effects. However, they are also well-known to increase the risk of coronary events. This area is of renewed significance given alarming new evidence suggesting this effect can occur even with acute usage. This contrasts with the well-established usage of aspirin as a mainstay for cardiovascular prophylaxis, as well as overwhelming evidence that COX inhibition induces vasodilation and is protective for vascular function. Here, we present an updated review of the preclinical and clinical literature regarding the cardiotoxicity of COX inhibitors. While studies to date have focussed on the role of COX in influencing renal and vascular function, we suggest an interaction between prostanoids and T cells may be a novel factor, mediating elevated cardiovascular disease risk with NSAID use.
Collapse
Affiliation(s)
- Shanzana Khan
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia.
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia.
| | - Karen L Andrews
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| | - Jaye P F Chin-Dusting
- Department of Pharmacology, Monash University, Clayton, Victoria 3800, Australia
- Baker IDI Heart and Diabetes Institute, Melbourne, Victoria 3004, Australia
| |
Collapse
|
47
|
Shimizu K, Iyoda T, Okada M, Yamasaki S, Fujii SI. Immune suppression and reversal of the suppressive tumor microenvironment. Int Immunol 2019; 30:445-454. [PMID: 29939325 DOI: 10.1093/intimm/dxy042] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
Most tumors employ multiple strategies to attenuate T-cell-mediated immune responses. In particular, immune suppression surrounding the tumor is achieved by interfering with antigen-presenting cells and effector T cells. Controlling both the tumor and the tumor microenvironment (TME) is critical for cancer treatment. Checkpoint blockade therapy can overcome tumor-induced immune suppression, but more than half of the patients fail to respond to this treatment; therefore, more effective cancer immunotherapies are needed. Generation of an anti-tumor immune response is a multi-step process of immune activation against the tumor that requires effector T cells to recognize and exert toxic effects against tumor cells, for which two strategies are employed-inhibition of various types of immune suppressor cells, such as myeloid cells and regulatory T cells, and establishment of anti-tumor immune surveillance including, activation of natural killer cells and cytotoxic T cells. It was recently shown that anti-cancer drugs not only directly kill tumor cells, but also influence the immune response to cancer by promoting immunogenic cell death, enhancing antigen presentation or depleting immunosuppressive cells. Herein, we review the mechanisms by which tumors exert immune suppression as well as their regulation. We then discuss how the complex reciprocal interactions between immunosuppressive and immunostimulatory cells influence immune cell dynamics in the TME. Finally, we highlight the new therapies that can reverse immune suppression in the TME and promote anti-tumor immunity.
Collapse
Affiliation(s)
- Kanako Shimizu
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Tomonori Iyoda
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Masahiro Okada
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Satoru Yamasaki
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| | - Shin-Ichiro Fujii
- Laboratory for Immunotherapy, RIKEN Center for Integrative Medical Sciences, Tsurumi-ku, Yokohama, Kanagawa, Japan
| |
Collapse
|
48
|
Polanczyk MJ, Walker E, Haley D, Guerrouahen BS, Akporiaye ET. Blockade of TGF-β signaling to enhance the antitumor response is accompanied by dysregulation of the functional activity of CD4 +CD25 +Foxp3 + and CD4 +CD25 -Foxp3 + T cells. J Transl Med 2019; 17:219. [PMID: 31288845 PMCID: PMC6617864 DOI: 10.1186/s12967-019-1967-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/04/2019] [Indexed: 12/26/2022] Open
Abstract
Background The pleiotropic cytokine, transforming growth factor (TGF)-β, and CD4+CD25+Foxp3+ regulatory T cells (Tregs) play a critical role in actively suppressing antitumor immune responses. Evidence shows that TGF-β produced by tumor cells promotes tolerance via expansion of Tregs. Our group previously demonstrated that blockade of TGF-β signaling with a small molecule TGF-β receptor I antagonist (SM16) inhibited primary and metastatic tumor growth in a T cell dependent fashion. In the current study, we evaluated the effect of SM16 on Treg generation and function. Methods Using BALB/c, FoxP3eGFP and Rag−/− mice, we performed FACS analysis to determine if SM16 blocked de novo TGF-β-induced Treg generation in vitro and in vivo. CD4+ T cells from lymph node and spleen were isolated from control mice or mice maintained on SM16 diet, and flow cytometry analysis was used to detect the frequency of CD4+CD25−FoxP3+ and CD4+CD25+FoxP3+ T cells. In vitro suppression assays were used to determine the ability to suppress naive T cell proliferation in vitro of both CD4+CD25+FoxP3+ and CD4+CD25−FoxP3+ T cell sub-populations. We then examined whether SM16 diet exerted an inhibitory effect on primary tumor growth and correlated with changes in FoxP3+expression. ELISA analysis was used to measure IFN-γ levels after 72 h co-culture of CD4+CD25+ T cells from tumor-bearing mice on control or SM16 diet with CD4+CD25− T cells from naive donors. Results SM16 abrogates TGF-β-induced Treg generation in vitro but does not prevent global homeostatic expansion of CD4+ T cell sub-populations in vivo. Instead, SM16 treatment causes expansion of a population of CD4+CD25−Foxp3+ Treg-like cells without significantly altering the overall frequency of Treg in lymphoreplete naive and tumor-bearing mice. Importantly, both the CD4+CD25−Foxp3+ T cells and the CD4+CD25+Foxp3+ Tregs in mice receiving SM16 diet exhibited diminished ability to suppress naive T cell proliferation in vitro compared to Treg from mice on control diet. Conclusions These findings suggest that blockade of TGF-β signaling is a potentially useful strategy for blunting Treg function to enhance the anti-tumor response. Our data further suggest that the overall dampening of Treg function may involve the expansion of a quiescent Treg precursor population, which is CD4+CD25−Foxp3+.
Collapse
Affiliation(s)
| | - Edwin Walker
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA.,Veana Therapeutics, Inc., Portland, OR, USA
| | - Daniel Haley
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA
| | | | - Emmanuel T Akporiaye
- Earle A. Chiles Research Institute, Providence Cancer Center, Portland, OR, USA. .,Veana Therapeutics, Inc., Portland, OR, USA.
| |
Collapse
|
49
|
Taechangam N, Iyer SS, Walker NJ, Arzi B, Borjesson DL. Mechanisms utilized by feline adipose-derived mesenchymal stem cells to inhibit T lymphocyte proliferation. Stem Cell Res Ther 2019; 10:188. [PMID: 31238978 PMCID: PMC6593543 DOI: 10.1186/s13287-019-1300-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/20/2019] [Accepted: 06/07/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Feline adipose-derived mesenchymal stem cells (ASCs) have been successfully used in clinical trials for the treatment of immune-mediated diseases with T cell dysregulation. However, the immunomodulatory pathways utilized by feline ASCs to suppress T cell activation have not been fully determined. We investigated the mechanisms used by feline ASCs to inhibit T cell proliferation, including the soluble factors and the cell-cell contact ligands responsible for ASC-T cell interaction. METHODS The immunomodulatory activity of feline ASCs was evaluated via cell cycle analysis and in vitro mixed leukocyte reaction using specific immunomodulatory inhibitors. Cell-cell interactions were assessed with static adhesion assays, also with inhibitors. RESULTS Feline ASCs decrease T cell proliferation by causing cell cycle arrest in G0-G1. Blocking prostaglandin (PGE2), but not IDO, partially restored lymphocyte proliferation. Although PDL-1 and CD137L are both expressed on activated feline ASCs, only the interaction of intercellular adhesion molecule 1 (ICAM-1, CD54) with its ligand, lymphocyte function-associated antigen 1 (LFA-1, CD11a/CD18), was responsible for ASC-T cell adhesion. Blocking this interaction reduced cell-cell adhesion and mediator (IFN-γ) secretion and signaling. CONCLUSIONS Feline ASCs utilize PGE2 and ICAM-1/LFA-1 ligand interaction to inhibit T cell proliferation with a resultant cell cycle arrest in G0-G1. These data further elucidate the mechanisms by which feline ASCs interact with T cells, help define appropriate T cell-mediated disease targets in cats that may be amenable to ASC therapy, and may also inform potential translational models for human diseases.
Collapse
Affiliation(s)
- Nopmanee Taechangam
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Smita S Iyer
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA
| | - Naomi J Walker
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Boaz Arzi
- Department of Surgical and Radiological Sciences, University of California, Davis, CA, 95616, USA.,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA
| | - Dori L Borjesson
- Department of Pathology, Microbiology and Immunology, Vet Med 3A, University of California, 1285 Veterinary Medicine Mall, Davis, CA, 95616, USA. .,Veterinary Institute for Regenerative Cures, School of Veterinary Medicine, University of California, Davis, CA, 95616, USA.
| |
Collapse
|
50
|
Cui G, Li Z, Ren J, Yuan A. IL-33 in the tumor microenvironment is associated with the accumulation of FoxP3-positive regulatory T cells in human esophageal carcinomas. Virchows Arch 2019; 475:579-586. [DOI: 10.1007/s00428-019-02579-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 04/07/2019] [Accepted: 04/18/2019] [Indexed: 12/13/2022]
|