1
|
Chen HJ, Sévin DC, Griffith GR, Vappiani J, Booty LM, van Roomen CPAA, Kuiper J, Dunnen JD, de Jonge WJ, Prinjha RK, Mander PK, Grandi P, Wyspianska BS, de Winther MPJ. Integrated metabolic-transcriptomic network identifies immunometabolic modulations in human macrophages. Cell Rep 2024; 43:114741. [PMID: 39276347 DOI: 10.1016/j.celrep.2024.114741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 06/08/2024] [Accepted: 08/26/2024] [Indexed: 09/17/2024] Open
Abstract
Macrophages exhibit diverse phenotypes and respond flexibly to environmental cues through metabolic remodeling. In this study, we present a comprehensive multi-omics dataset integrating intra- and extracellular metabolomes with transcriptomic data to investigate the metabolic impact on human macrophage function. Our analysis establishes a metabolite-gene correlation network that characterizes macrophage activation. We find that the concurrent inhibition of tryptophan catabolism by IDO1 and IL4I1 inhibitors suppresses the macrophage pro-inflammatory response, whereas single inhibition leads to pro-inflammatory activation. We find that a subset of anti-inflammatory macrophages activated by Fc receptor signaling promotes glycolysis, challenging the conventional concept of reduced glycolysis preference in anti-inflammatory macrophages. We demonstrate that cholesterol accumulation suppresses macrophage IFN-γ responses. Our integrated network enables the discovery of immunometabolic features, provides insights into macrophage functional metabolic reprogramming, and offers valuable resources for researchers exploring macrophage immunometabolic characteristics and potential therapeutic targets for immune-related disorders.
Collapse
Affiliation(s)
- Hung-Jen Chen
- Department of Medical Biochemistry, Experimental Vascular Biology, Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | | | - Guillermo R Griffith
- Department of Medical Biochemistry, Experimental Vascular Biology, Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | | | - Lee M Booty
- Immunology Network, Immunology Research Unit, GSK, SG1 2NY Stevenage, UK
| | - Cindy P A A van Roomen
- Department of Medical Biochemistry, Experimental Vascular Biology, Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, 2333 CL Leiden, the Netherlands
| | - Jeroen den Dunnen
- Center for Experimental and Molecular Medicine, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands
| | - Wouter J de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 BK Amsterdam, the Netherlands
| | - Rab K Prinjha
- Immunology Research Unit, GSK Medicines Research Centre, SG1 2NY Stevenage, UK
| | - Palwinder K Mander
- Immunology Research Unit, GSK Medicines Research Centre, SG1 2NY Stevenage, UK
| | | | - Beata S Wyspianska
- Immunology Research Unit, GSK Medicines Research Centre, SG1 2NY Stevenage, UK
| | - Menno P J de Winther
- Department of Medical Biochemistry, Experimental Vascular Biology, Atherosclerosis and Ischemic Syndromes, Amsterdam Cardiovascular Sciences, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center, Location AMC, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
2
|
Wu C, Zhang G, Wang L, Hu J, Ju Z, Tao H, Li Q, Li J, Zhang W, Sheng J, Hou X, Hu Y. Spatial proteomic profiling elucidates immune determinants of neoadjuvant chemo-immunotherapy in esophageal squamous cell carcinoma. Oncogene 2024; 43:2751-2767. [PMID: 39122893 DOI: 10.1038/s41388-024-03123-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 08/01/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Esophageal squamous cell carcinoma (ESCC) presents significant clinical and therapeutic challenges due to its aggressive nature and generally poor prognosis. We initiated a Phase II clinical trial (ChiCTR1900027160) to assess the efficacy of a pioneering neoadjuvant chemo-immunotherapy regimen comprising programmed death-1 (PD-1) blockade (Toripalimab), nanoparticle albumin-bound paclitaxel (nab-paclitaxel), and the oral fluoropyrimidine derivative S-1, in patients with locally advanced ESCC. This study uniquely integrates clinical outcomes with advanced spatial proteomic profiling using Imaging Mass Cytometry (IMC) to elucidate the dynamics within the tumor microenvironment (TME), focusing on the mechanistic interplay of resistance and response. Sixty patients participated, receiving the combination therapy prior to surgical resection. Our findings demonstrated a major pathological response (MPR) in 62% of patients and a pathological complete response (pCR) in 29%. The IMC analysis provided a detailed regional assessment, revealing that the spatial arrangement of immune cells, particularly CD8+ T cells and B cells within tertiary lymphoid structures (TLS), and S100A9+ inflammatory macrophages in fibrotic regions are predictive of therapeutic outcomes. Employing machine learning approaches, such as support vector machine (SVM) and random forest (RF) analysis, we identified critical spatial features linked to drug resistance and developed predictive models for drug response, achieving an area under the curve (AUC) of 97%. These insights underscore the vital role of integrating spatial proteomics into clinical trials to dissect TME dynamics thoroughly, paving the way for personalized and precise cancer treatment strategies in ESCC. This holistic approach not only enhances our understanding of the mechanistic basis behind drug resistance but also sets a robust foundation for optimizing therapeutic interventions in ESCC.
Collapse
Affiliation(s)
- Chao Wu
- Department of Medical Oncology, Senior Department of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, Beijing, China
| | - Guoqing Zhang
- Department of Medical Oncology, Senior Department of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, Beijing, China
| | - Lin Wang
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Jinlong Hu
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhongjian Ju
- Department of Radiation Oncology, Chinese PLA General Hospital, The First Medical Center, Beijing, China
| | - Haitao Tao
- Department of Medical Oncology, Senior Department of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, Beijing, China
| | - Qing Li
- The Shapingba Affiliated Hospital, Chongqing University, Chongqing, China
| | - Jian Li
- Chengdu Medical College, Chengdu, China
| | - Wei Zhang
- Department of Medical Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Jianpeng Sheng
- College of Artificial Intelligence, Nanjing University of Aeronautics and Astronautics, Nanjing, China.
- Chinese Institutes for Medical Research, Beijing, China.
| | - Xiaobin Hou
- Department of Thoracic Surgery, Chinese PLA General Hospital, The First Medical Center, Beijing, China.
| | - Yi Hu
- Department of Medical Oncology, Senior Department of Oncology, Chinese PLA General Hospital, The Fifth Medical Center, Beijing, China.
| |
Collapse
|
3
|
Chang K, Jiao Y, Zhang B, Hou L, He X, Wang D, Li D, Li R, Wang Z, Fan P, Zhang J. MGP + and IDO1 + tumor-associated macrophages facilitate immunoresistance in breast cancer revealed by single-cell RNA sequencing. Int Immunopharmacol 2024; 131:111818. [PMID: 38460300 DOI: 10.1016/j.intimp.2024.111818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 03/11/2024]
Abstract
Immunotherapy is widely applied for the treatment of breast cancer, but to which some patients respond poorly or develop resistance. Therefore, the mechanism needs to be further studied. Transcriptomic data of 31 breast cancer patients treated with anti-programmed death receptor 1 (PD-1) was downloaded from the VIB-KULeuven Center for Cancer Biology to analyze the changes in myeloid cells in tumor tissues before and after immunotherapy. And 24 cell populations that may be immune-related were further identified. Representative cell populations were also screened and validated through cellular and animal experiments to evaluate the relevant molecular expression and pathways of tumor-associated macrophages (TAMs) in the tumor microenvironment. The results demonstrated that MGP+ TAMs and IDO1+ TAMs influenced the efficacy of immunotherapy in breast cancer patients. After anti-PD-1 treatment, Increased numbers of MGP+ TAMs and IDO1+ TAMs in breast cancer patients upregulated pro-tumorigenic factors associated with resistance to immunosuppressive therapy. This study provides new biomarkers for immunotherapy to predict therapeutic responses and overcome potential resistance to immunotherapy. It is an important complement to the immunosuppression caused by TAMs after immunotherapy for breast cancer.
Collapse
Affiliation(s)
- Kexin Chang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Yangchi Jiao
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Zhang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Lan Hou
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiangmei He
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Donghui Wang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Danxi Li
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Ruolei Li
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhe Wang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Pengyu Fan
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Juliang Zhang
- Department of Thyroid, Breast and Vascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
4
|
Mestiri S, El-Ella DMA, Fernandes Q, Bedhiafi T, Almoghrabi S, Akbar S, Inchakalody V, Assami L, Anwar S, Uddin S, Gul ARZ, Al-Muftah M, Merhi M, Raza A, Dermime S. The dynamic role of immune checkpoint molecules in diagnosis, prognosis, and treatment of head and neck cancers. Biomed Pharmacother 2024; 171:116095. [PMID: 38183744 DOI: 10.1016/j.biopha.2023.116095] [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: 10/26/2023] [Revised: 12/21/2023] [Accepted: 12/26/2023] [Indexed: 01/08/2024] Open
Abstract
Head and neck cancer (HNC) is the sixth most common cancer type, accounting for approximately 277,597 deaths worldwide. Recently, the Food and Drug Administration (FDA) has approved immune checkpoint blockade (ICB) agents targeting programmed death-1 (PD-1) and programmed death-ligand 1 (PD-L1) as a treatment regimen for head and neck squamous cell carcinomas (HNSCC). Studies have reported the role of immune checkpoint inhibitors as targeted therapeutic regimens that unleash the immune response against HNSCC tumors. However, the overall response rates to immunotherapy vary between 14-32% in recurrent or metastatic HNSCC, with clinical response and treatment success being unpredictable. Keeping this perspective in mind, it is imperative to understand the role of T cells, natural killer cells, and antigen-presenting cells in modulating the immune response to immunotherapy. In lieu of this, these immune molecules could serve as prognostic and predictive biomarkers to facilitate longitudinal monitoring and understanding of treatment dynamics. These immune biomarkers could pave the path for personalized monitoring and management of HNSCC. In this review, we aim to provide updated immunological insight on the mechanism of action, expression, and the clinical application of immune cells' stimulatory and inhibitory molecules as prognostic and predictive biomarkers in HNC. The review is focused mainly on CD27 and CD137 (members of the TNF-receptor superfamily), natural killer group 2 member D (NKG2D), tumor necrosis factor receptor superfamily member 4 (TNFRSF4 or OX40), S100 proteins, PD-1, PD-L1, PD-L2, T cell immunoglobulin and mucin domain 3 (TIM-3), cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), lymphocyte-activation gene 3 (LAG-3), indoleamine-pyrrole 2,3-dioxygenase (IDO), B and T lymphocyte attenuator (BTLA). It also highlights the importance of T, natural killer, and antigen-presenting cells as robust biomarker tools for understanding immune checkpoint inhibitor-based treatment dynamics. Though a comprehensive review, all aspects of the immune molecules could not be covered as they were beyond the scope of the review; Further review articles can cover other aspects to bridge the knowledge gap.
Collapse
Affiliation(s)
- Sarra Mestiri
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Dina Moustafa Abo El-Ella
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Queenie Fernandes
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; College of Medicine, Qatar University, Doha, Qatar
| | - Takwa Bedhiafi
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Salam Almoghrabi
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shayista Akbar
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Varghese Inchakalody
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Laila Assami
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shaheena Anwar
- Department of Biosciences, Salim Habib University, Karachi, Pakistan
| | - Shahab Uddin
- Translational Research Institute and Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha, Qatar; Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Abdul Rehman Zar Gul
- National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Mariam Al-Muftah
- Translational Cancer and Immunity Centre, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar; College of Health and Life Sciences, Hamad Bin Khalifa University, Doha, Qatar
| | - Maysaloun Merhi
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Afsheen Raza
- Department of Biomedical Sciences, College of Health Science, Abu Dhabi University, Abu Dhabi, United Arab Emirates
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research/ Translational Research Institute, Hamad Medical Corporation, Doha, Qatar; National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
5
|
Qin MC, Li JJ, Zheng YT, Li YJ, Zhang YX, Ou RX, He WY, Zhao JM, Liu ST, Liu MH, Lin HY, Gao L. Naringin ameliorates liver fibrosis in zebrafish by modulating IDO1-mediated lipid metabolism and inflammatory infiltration. Food Funct 2023; 14:10347-10361. [PMID: 37930368 DOI: 10.1039/d3fo03858k] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Liver fibrosis (LF) is an important reparative process in response to acute or chronic hepatic injury, which has the potential to advance towards cirrhosis and hepatocellular carcinoma. Dietary naringin consumption contributes to protection against LF in animal studies, while the exact protective mechanism of naringin remains unclear. This study aimed to investigate the molecular mechanisms behind the potential protective effect of naringin against TAA-induced LF in zebrafish. In this study, we utilized zebrafish to create the LF model and investigate the therapeutic mechanism of naringin. Firstly, we evaluated the changes in hepatic fibrosis and lipid accumulation in the liver following naringin treatment with oil red O, Nile red, and Sirius red and immunohistochemistry. In addition, we employed an ROS probe to directly measure oxidative stress and monitor inflammatory cell migration in a zebrafish transgenic line. Morpholino was used in the knockdown of IDO1 in order to verify its vital role in LF. Our findings demonstrated that naringin exhibited anti-inflammatory and anti-fibrotic action in conjunction with a reversal in lipid accumulation, oxidative stress and suppression of macrophage infiltration and activation of hepatic stellate cells. Furthermore, the results showed that the antifibrotic effect of naringin was removed upon IDO1 knockdown, proving that naringin exerts a protective effect by regulating IDO1. Naringin demonstrates remarkable protective effects against LF, effectively counteracting inflammation and hepatic steatosis in zebrafish liver. These findings suggest that naringin may function as an effective IDO1 inhibitor, holding the potential for clinical translation as a therapeutic agent for the treatment of LF.
Collapse
Affiliation(s)
- Meng-Chen Qin
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jun-Jie Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yan-Tao Zheng
- Emergency Department, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yun-Jia Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Yu-Xue Zhang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Rou-Xuan Ou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Wei-Yi He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Jia-Min Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | - Su-Tong Liu
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China.
| | - Ming-Hao Liu
- The First Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, China.
| | - Hai-Yan Lin
- Shenzhen Hospital, University of Chinese Academy of Sciences, Shenzhen, China.
| | - Lei Gao
- Emergency Department, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| |
Collapse
|
6
|
Nogueira CO, Rocha T, Messor DF, Souza INO, Clarke JR. Fundamental neurochemistry review: Glutamatergic dysfunction as a central mechanism underlying flavivirus-induced neurological damage. J Neurochem 2023; 166:915-927. [PMID: 37603368 DOI: 10.1111/jnc.15935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/06/2023] [Accepted: 07/26/2023] [Indexed: 08/22/2023]
Abstract
The Flaviviridae family comprises positive-sense single-strand RNA viruses mainly transmitted by arthropods. Many of these pathogens are especially deleterious to the nervous system, and a myriad of neurological symptoms have been associated with infections by Zika virus (ZIKV), West Nile virus (WNV), and Japanese encephalitis virus (JEV) in humans. Studies suggest that viral replication in neural cells and the massive release of pro-inflammatory mediators lead to morphological alterations of synaptic spine structure and changes in the balance of excitatory/inhibitory neurotransmitters and receptors. Glutamate is the predominant excitatory neurotransmitter in the brain, and studies propose that either enhanced release or impaired uptake of this amino acid contributes to brain damage in several conditions. Here, we review existing evidence suggesting that glutamatergic dysfunction-induced by flaviviruses is a central mechanism for neurological damage and clinical outcomes of infection. We also discuss current data suggesting that pharmacological approaches that counteract glutamatergic dysfunction show benefits in animal models of such viral diseases.
Collapse
Affiliation(s)
- Clara O Nogueira
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Tamires Rocha
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel F Messor
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Isis N O Souza
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Julia R Clarke
- Programa de Pós-graduação em Ciências Farmacêuticas, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
7
|
Lin L, Wei J, Zhu C, Hao G, Xue J, Zhu Y, Wu R. Sema3A alleviates viral myocarditis by modulating SIRT1 to regulate cardiomyocyte mitophagy. ENVIRONMENTAL TOXICOLOGY 2023; 38:1305-1317. [PMID: 36880403 DOI: 10.1002/tox.23765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/25/2022] [Accepted: 02/20/2023] [Indexed: 05/18/2023]
Abstract
Viral myocarditis (VMC) is a common myocardial inflammatory disease characterized by inflammatory cell infiltration and cardiomyocyte necrosis. Sema3A was reported to reduce cardiac inflammation and improve cardiac function after myocardial infarction, but its role in VMC remains to be explored. Here, a VMC mouse model was established by infection with CVB3, and Sema3A was overexpressed in vivo by intraventricular injection of an adenovirus-mediated Sema3A expression vector (Ad-Sema3A). We found that Sema3A overexpression attenuated CVB3-induced cardiac dysfunction and tissue inflammation. And Sema3A also reduced macrophage accumulation and NLRP3 inflammasome activation in the myocardium of VMC mice. In vitro, LPS was used to stimulate primary splenic macrophages to mimic the macrophage activation state in vivo. Activated macrophages were co-cultured with primary mouse cardiomyocytes to evaluate macrophage infiltration-induced cardiomyocyte damage. Ectopic expression of Sema3A in cardiomyocytes effectively protected cardiomyocytes from activated macrophage-induced inflammation, apoptosis, and ROS accumulation. Mechanistically, cardiomyocyte-expressed Sema3A mitigated macrophage infiltration-caused cardiomyocyte dysfunction by promoting cardiomyocyte mitophagy and hindering NLRP3 inflammasome activation. Furthermore, NAM (a SIRT1 inhibitor) reversed the protective effect of Sema3A against activated macrophage-induced cardiomyocyte dysfunction by suppressing cardiomyocyte mitophagy. In conclusion, Sema3A promoted cardiomyocyte mitophagy and suppressed inflammasome activation by regulating SIRT1, thereby attenuating macrophage infiltration-induced cardiomyocyte injury in VMC.
Collapse
Affiliation(s)
- Lin Lin
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jin Wei
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Canzhan Zhu
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Guanghua Hao
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jiahong Xue
- Cardiovascular Hospital of the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yanhe Zhu
- Department of Medicine, School of Public Health, Institute of Endemic Diseases, Xi'an Jiaotong University, Xi'an, China
| | - Ruiyun Wu
- Department of Medicine, School of Public Health, Institute of Endemic Diseases, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
8
|
Mannarino MR, Bianconi V, Scalisi G, Franceschini L, Manni G, Cucci A, Bagaglia F, Mencarelli G, Giglioni F, Ricciuti D, Figorilli F, Pieroni B, Cosentini E, Padiglioni E, Colangelo C, Fuchs D, Puccetti P, Follenzi A, Pirro M, Gargaro M, Fallarino F. A tryptophan metabolite prevents depletion of circulating endothelial progenitor cells in systemic low-grade inflammation. Front Immunol 2023; 14:964660. [PMID: 37081894 PMCID: PMC10110845 DOI: 10.3389/fimmu.2023.964660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 03/20/2023] [Indexed: 04/07/2023] Open
Abstract
BackgroundChronic systemic inflammation reduces the bioavailability of circulating endothelial progenitor cells (EPCs). Indoleamine 2,3-dioxygenase 1 (IDO1), a key enzyme of immune tolerance catalyzing the initial step of tryptophan degradation along the so-called l-kynurenine (l-kyn) pathway, that is induced by inflammatory stimuli and exerts anti-inflammatory effects. A specific relationship between IDO1 activity and circulating EPC numbers has not yet been investigated.MethodsIn this study, circulating EPCs were examined in mice treated with low doses of lipopolysaccharide (LPS) to mimic low-grade inflammation. Moreover, the association between IDO1 activity and circulating EPCs was studied in a cohort of 277 patients with variable systemic low-grade inflammation.ResultsRepeated low doses of LPS caused a decrease in circulating EPCs and l-kyn supplementation, mimicking IDO1 activation, significantly increased EPC numbers under homeostatic conditions preventing EPC decline in low-grade endotoxemia. Accordingly, in patients with variable systemic low-grade inflammation, there was a significant interaction between IDO1 activity and high-sensitivity C-reactive protein (hs-CRP) in predicting circulating EPCs, with high hs-CRP associated with significantly lower EPCs at low IDO1 activity but not at high IDO1 activity.InterpretationOverall, these findings demonstrate that systemic low-grade inflammation reduces circulating EPCs. However, high IDO1 activity and l-kyn supplementation limit circulating EPC loss in low-grade inflammation.
Collapse
Affiliation(s)
| | - Vanessa Bianconi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Vanessa Bianconi, ; Marco Gargaro, ; Francesca Fallarino,
| | - Giulia Scalisi
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Luca Franceschini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giorgia Manni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessia Cucci
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Francesco Bagaglia
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Giulia Mencarelli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Giglioni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Doriana Ricciuti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Filippo Figorilli
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Benedetta Pieroni
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elena Cosentini
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | | | - Cecilia Colangelo
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Dietmar Fuchs
- Division of Biological Chemistry, Biocenter, Innsbruck Medical University, Innsbruck, Austria
| | - Paolo Puccetti
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Antonia Follenzi
- Department of Health Sciences, School of Medicine, University of Piemonte Orientale, Novara, Italy
| | - Matteo Pirro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Marco Gargaro
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Vanessa Bianconi, ; Marco Gargaro, ; Francesca Fallarino,
| | - Francesca Fallarino
- Department of Medicine and Surgery, University of Perugia, Perugia, Italy
- *Correspondence: Vanessa Bianconi, ; Marco Gargaro, ; Francesca Fallarino,
| |
Collapse
|
9
|
Nori P, Haghshenas R, Aftabi Y, Akbari H. Comparison of moderate-intensity continuous training and high-intensity interval training effects on the Ido1-KYN-Ahr axis in the heart tissue of rats with occlusion of the left anterior descending artery. Sci Rep 2023; 13:3721. [PMID: 36879035 PMCID: PMC9988842 DOI: 10.1038/s41598-023-30847-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Myocardial infarction (MI) affects many molecular pathways in heart cells, including the Ido1-KYN-Ahr axis. This pathway has recently been introduced as a valuable therapeutic target in infarction. We examined the effects of moderate-intensity continuous training (MICT) and high-intensity interval training (HIIT) on the axis in the heart tissue of male Wistar rats with occluded left anterior descending (OLAD). Thirty rats (age 10-12 weeks, mean weight 275 ± 25 g) were divided into five groups with 6 animals: Control (Ct) group, MICT group, rats with OLAD as MI group, rats with OLAD treated with MICT (MIMCT group) and rats with OLAD treated with HIIT (MIHIIT group). Rats performed the training protocols for 8 weeks, 5 days a week. HIIT included 7 sets of 4 min running with an intensity of 85-90% VO2max and 3 min of recovery activation between sets. MICT included continuous running at the same distance as HIIT with an intensity of 50-60% VO2max for 50 min. The expressions of Ahr, Cyp1a1, and Ido1 were assayed by real-time PCR. Malondialdehyde (MDA) and Kynurenine levels, and AHR, CYP1A1, and IDO1 proteins were detected using ELISA. Data were analyzed using the ANOVA and MANOVA tests. Compared to the CT group, MI caused an increase in all studied factors, but only statistically significant (P < 0.05) for MDA and IDO1. With a greater effect of HIIT, both protocols significantly lowered the proteins expressions in the MIHIIT and MIMCT groups, compared with the MI group (P < 0.001). In healthy rats, only AHR protein significantly decreased in the MICT group compared to the Ct group (P < 0.05). HIIT and MICT protocols significantly reduced the gene and protein expression of Cyp1a1 (P < 0.05) and Ido1 (P < 0.01), and HIIT had a greater effect. In conclusion, both protocols were effective at reducing the levels of Ido1-Kyn-Ahr axis components and oxidative stress in the infarcted heart tissue and HIIT had a higher significant effect.
Collapse
Affiliation(s)
- Pouria Nori
- Department of Sport Sciences, Faculty of Humanities, Semnan University, Semnan, Iran
| | - Rouhollah Haghshenas
- Associate Professor of Exercise Physiology, Department of Sport Sciences, Faculty of Humanities, Semnan University, Semnan, Iran.
| | - Younes Aftabi
- Tuberculosis and Lung Diseases Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hakimeh Akbari
- Assistant Professor of Exercise Physiology, Department of Sport Sciences, Faculty of Humanities, Semnan University, Semnan, Iran
| |
Collapse
|
10
|
Mujalli A, Alghamdi KS, Nasser KK, Al-Rayes N, Banaganapalli B, Shaik NA, Elango R. Bioinformatics insights into the genes and pathways on severe COVID-19 pathology in patients with comorbidities. Front Physiol 2022; 13:1045469. [PMID: 36589459 PMCID: PMC9795193 DOI: 10.3389/fphys.2022.1045469] [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: 09/15/2022] [Accepted: 11/15/2022] [Indexed: 12/23/2022] Open
Abstract
Background: Coronavirus disease (COVID-19) infection is known for its severe clinical pathogenesis among individuals with pre-existing comorbidities. However, the molecular basis of this observation remains elusive. Thus, this study aimed to map key genes and pathway alterations in patients with COVID-19 and comorbidities using robust systems biology approaches. Methods: The publicly available genome-wide transcriptomic datasets from 120 COVID-19 patients, 281 patients suffering from different comorbidities (like cardiovascular diseases, atherosclerosis, diabetes, and obesity), and 252 patients with different infectious diseases of the lung (respiratory syncytial virus, influenza, and MERS) were studied using a range of systems biology approaches like differential gene expression, gene ontology (GO), pathway enrichment, functional similarity, mouse phenotypic analysis and drug target identification. Results: By cross-mapping the differentially expressed genes (DEGs) across different datasets, we mapped 274 shared genes to severe symptoms of COVID-19 patients or with comorbidities alone. GO terms and functional pathway analysis highlighted genes in dysregulated pathways of immune response, interleukin signaling, FCGR activation, regulation of cytokines, chemokines secretion, and leukocyte migration. Using network topology parameters, phenotype associations, and functional similarity analysis with ACE2 and TMPRSS2-two key receptors for this virus-we identified 17 genes with high connectivity (CXCL10, IDO1, LEPR, MME, PTAFR, PTGS2, MAOB, PDE4B, PLA2G2A, COL5A1, ICAM1, SERPINE1, ABCB1, IL1R1, ITGAL, NCAM1 and PRKD1) potentially contributing to the clinical severity of COVID-19 infection in patients with comorbidities. These genes are predicted to be tractable and/or with many existing approved inhibitors, modulators, and enzymes as drugs. Conclusion: By systemic implementation of computational methods, this study identified potential candidate genes and pathways likely to confer disease severity in COVID-19 patients with pre-existing comorbidities. Our findings pave the way to develop targeted repurposed therapies in COVID-19 patients.
Collapse
Affiliation(s)
- Abdulrahman Mujalli
- Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia,Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia,Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia,*Correspondence: Abdulrahman Mujalli, ; Ramu Elango,
| | - Kawthar Saad Alghamdi
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia,Department of Biology, Faculty of Science, University of Hafr Al Batin, Hafar Al-Batin, Saudi Arabia
| | - Khalidah Khalid Nasser
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nuha Al-Rayes
- Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Babajan Banaganapalli
- Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia,Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Noor Ahmad Shaik
- Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia,Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ramu Elango
- Department of Genetic Medicine, King Abdulaziz University, Jeddah, Saudi Arabia,Princess Al-Jawhara Center of Excellence in Research of Hereditary Disorders (PACER-HD), King Abdulaziz University, Jeddah, Saudi Arabia,*Correspondence: Abdulrahman Mujalli, ; Ramu Elango,
| |
Collapse
|
11
|
Ala M, Eftekhar SP. The Footprint of Kynurenine Pathway in Cardiovascular Diseases. Int J Tryptophan Res 2022; 15:11786469221096643. [PMID: 35784899 PMCID: PMC9248048 DOI: 10.1177/11786469221096643] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/28/2022] [Indexed: 12/30/2022] Open
Abstract
Kynurenine pathway is the main route of tryptophan metabolism and produces several metabolites with various biologic properties. It has been uncovered that several cardiovascular diseases are associated with the overactivation of kynurenine pathway and kynurenine and its metabolites have diagnostic and prognostic value in cardiovascular diseases. Furthermore, it was found that several kynurenine metabolites can differently affect cardiovascular health. For instance, preclinical studies have shown that kynurenine, xanthurenic acid and cis-WOOH decrease blood pressure; kynurenine and 3-hydroxyanthranilic acid prevent atherosclerosis; kynurenic acid supplementation and kynurenine 3-monooxygenase (KMO) inhibition improve the outcome of stroke. Indoleamine 2,3-dioxygenase (IDO) overactivity and increased kynurenine levels improve cardiac and vascular transplantation outcomes, whereas exacerbating the outcome of myocardial ischemia, post-ischemic myocardial remodeling, and abdominal aorta aneurysm. IDO inhibition and KMO inhibition are also protective against viral myocarditis. In addition, dysregulation of kynurenine pathway is observed in several conditions such as senescence, depression, diabetes, chronic kidney disease (CKD), cirrhosis, and cancer closely connected to cardiovascular dysfunction. It is worth defining the exact effect of each metabolite of kynurenine pathway on cardiovascular health. This narrative review is the first review that separately discusses the involvement of kynurenine pathway in different cardiovascular diseases and dissects the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Moein Ala
- School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Seyed Parsa Eftekhar
- Student Research Committee, Health Research Center, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
12
|
Huang Y, Guo L, Chen J, Wu M, Zhang C, Liu Z, Li J, Li K, Xiong Z, Wu Q, Li Z, Luo K, Yuan W, Wu X. Serum Lactate Dehydrogenase Level as a Prognostic Factor for COVID-19: A Retrospective Study Based on a Large Sample Size. Front Med (Lausanne) 2022; 8:671667. [PMID: 35059407 PMCID: PMC8763698 DOI: 10.3389/fmed.2021.671667] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 11/29/2021] [Indexed: 01/08/2023] Open
Abstract
Background: In this study, we investigated the relationship between serum lactate dehydrogenase (LDH) level and disease progression and prognosis of patients with COVID-19. Methods: We retrospectively reviewed the information of 1,751 patients with COVID-19 from Leishenshan Hospital in Wuhan, China. Univariate and multivariate Cox regression analyses as well as Logistics regression analyses, and Kaplan-Meier curves were used to determine the association between LDH levels and the prognosis of COVID-19 patients. Results: LDH was an independent risk factor for in-hospital death no matter it was taken as classified variable and continuous variable (all P = 0.001) but not for severe or critical illness status. The Kaplan-Meier curves for LDH level showed that an elevated level of LDH was associated with in-hospital death. Conclusions: In patients with COVID-19, the increased LDH level is associated with a higher risk of negative clinical prognosis and higher mortality. This will provide a reference for clinicians and researchers to understand, diagnose, and treat patients with COVID-19. Further prospective studies with larger sample sizes are needed to verify these findings.
Collapse
Affiliation(s)
- Yihui Huang
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Liang Guo
- Department of Plastic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jiwei Chen
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Meng Wu
- Department of Ultrasound, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chao Zhang
- Department of Cardiovascular Surgery, Tongji Medical College, Union Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zeming Liu
- Department of Plastic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinpeng Li
- Department of Thyroid and Breast Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kun Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhongwei Xiong
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Qian Wu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhengwei Li
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kuan Luo
- Department of Neurosurgery, Wuhan Puren Hospital, Wuhan, China
| | - Weiwei Yuan
- Department of Plastic and Cosmetic Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Xiaohui Wu
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
13
|
Liu Y, Li S, Gao Z, Li S, Tan Q, Li Y, Wang D, Wang Q. Indoleamine 2,3-Dioxygenase 1 (IDO1) Promotes Cardiac Hypertrophy via a PI3K-AKT-mTOR-Dependent Mechanism. Cardiovasc Toxicol 2021; 21:655-668. [PMID: 34021461 PMCID: PMC8211584 DOI: 10.1007/s12012-021-09657-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/04/2021] [Indexed: 01/25/2023]
Abstract
Indoleamine 2,3-dioxygenase 1 (IDO1) is an enzyme for tryptophan metabolism, involved in immune cell differentiation/maturation and cancer biology. IDO1 is also expressed in cardiomyocytes, but its roles in the cardiovascular system are not fully understood. Here, we reported the functions of IDO1 during cardiac hypertrophy. Quantitative real-time PCR and Western blot experiments demonstrated the upregulation of IDO1 mRNA and protein levels in human and hypertrophic mouse hearts, as well as in angiotensin II (Ang II)-induced hypertrophic rat cardiomyocytes. IDO1 activity and metabolite product kynurenine were upregulated in rodent hypertrophic hearts and cardiomyocytes. Inhibition of IDO1 activity with PF-06840003 reduced Ang II-induced cardiac hypertrophy and rescued cardiac function in mice. siRNA-mediated knockdown of Ido1 repressed Ang II-induced growth in cardiomyocyte size and overexpression of hypertrophy-associated genes atrial natriuretic peptide (Anp or Nppa), brain natriuretic peptide (Bnp or Nppb), β-myosin heavy chain (β-Mhc or Myh7). By contrast, adenovirus-mediated rat Ido1 overexpression in cardiomyocytes promoted hypertrophic growth induced by Ang II. Mechanism analysis showed that IDO1 overexpression was associated with PI3K-AKT-mTOR signaling to activate the ribosomal protein S6 kinase 1 (S6K1), which promoted protein synthesis in Ang II-induced hypertrophy of rat cardiomyocytes. Finally, we provided evidence that inhibition of PI3K with pictilisib, AKT with perifosine, or mTOR with rapamycin, blocked the effects of IDO1 on protein synthesis and cardiomyocyte hypertrophy in Ang II-treated cells. Collectively, our findings identify that IDO1 promotes cardiomyocyte hypertrophy partially via PI3K-AKT-mTOR-S6K1 signaling.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Cardiomegaly/drug therapy
- Cardiomegaly/enzymology
- Cardiomegaly/pathology
- Case-Control Studies
- Cells, Cultured
- Disease Models, Animal
- Female
- Humans
- Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors
- Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Kynurenine/metabolism
- Male
- Mice
- Middle Aged
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/enzymology
- Myocytes, Cardiac/pathology
- Phosphatidylinositol 3-Kinase/metabolism
- Protein Kinase Inhibitors/pharmacology
- Proto-Oncogene Proteins c-akt/antagonists & inhibitors
- Proto-Oncogene Proteins c-akt/metabolism
- Rats, Sprague-Dawley
- Signal Transduction
- TOR Serine-Threonine Kinases/antagonists & inhibitors
- TOR Serine-Threonine Kinases/metabolism
- Rats
Collapse
Affiliation(s)
- Yang Liu
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Shuang Li
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Zhanqun Gao
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Shuangjia Li
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Qingyun Tan
- Department of Anesthesiology, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Yanmei Li
- Emergency Department, First Affiliated Hospital of Jiamusi University, Jiamusi, China
| | - Dongwei Wang
- Department of Anesthesiology, First Affiliated Hospital of Jiamusi University, Jiamusi, China.
| | - Qingdong Wang
- Department of Anesthesiology, First Affiliated Hospital of Jiamusi University, Jiamusi, China.
| |
Collapse
|
14
|
Mo C, Xie S, Liu B, Zhong W, Zeng T, Huang S, Lai Y, Deng G, Zhou C, Yan W, Chen Y, Huang S, Gao L, Lv Z. Indoleamine 2,3-dioxygenase 1 limits hepatic inflammatory cells recruitment and promotes bile duct ligation-induced liver fibrosis. Cell Death Dis 2021; 12:16. [PMID: 33414436 PMCID: PMC7791029 DOI: 10.1038/s41419-020-03277-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Revised: 11/18/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023]
Abstract
Liver fibrosis is a course of chronic liver dysfunction, can develop into cirrhosis and hepatocellular carcinoma. Inflammatory insult owing to pathogenic factors plays a crucial role in the pathogenesis of liver fibrosis. Indoleamine 2,3-dioxygenase 1 (IDO1) can affect the infiltration of immune cells in many pathology processes of diseases, but its role in liver fibrosis has not been elucidated completely. Here, the markedly elevated protein IDO1 in livers was identified, and dendritic cells (DCs) immune-phenotypes were significantly altered after BDL challenge. A distinct hepatic population of CD11c+DCs was decreased and presented an immature immune-phenotype, reflected by lower expression levels of co-stimulatory molecules (CD40, MHCII). Frequencies of CD11c+CD80+, CD11c+CD86+, CD11c+MHCII+, and CD11c+CD40+ cells in splenic leukocytes were reduced significantly. Notably, IDO1 overexpression inhibited hepatic, splenic CD11c+DCs maturation, mature DCs-mediated T-cell proliferation and worsened liver fibrosis, whereas above pathological phenomena were reversed in IDO1-/- mice. Our data demonstrate that IDO1 affects the process of immune cells recruitment via inhibiting DCs maturation and subsequent T cells proliferation, resulting in the promotion of hepatic fibrosis. Thus, amelioration of immune responses in hepatic and splenic microenvironment by targeting IDO1 might be essential for the therapeutic effects on liver fibrosis.
Collapse
Affiliation(s)
- Chan Mo
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Shuwen Xie
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Bin Liu
- Department of Emergency, Guangzhou Red Cross Hospital, Medical College, Jinan University, 510220, Guangzhou, China
| | - Weichao Zhong
- Shenzhen Traditional Chinese Medicine Hospital, No.1, Fuhua Road, Futian District, 518033, Shenzhen, Guangdong, People's Republic of China
| | - Ting Zeng
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Sha Huang
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Yuqi Lai
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Guanghui Deng
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Weixin Yan
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Shaohui Huang
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China.
- The Key Laboratory of Molecular Biology, State Administration of Traditional Chinese Medicine, School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China.
- Guangdong Provincial Key Laboratory of Shock and Microcirculation, Southern Medical University, 510515, Guangzhou, People's Republic of China.
| | - Zhiping Lv
- School of Traditional Chinese Medicine, Southern Medical University, 510515, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
15
|
Jiang N, Zhang L, Zhao G, Lin J, Wang Q, Xu Q, Li C, Hu L, Peng X, Yu F, Xu M. Indoleamine 2,3-Dioxygenase Regulates Macrophage Recruitment, Polarization and Phagocytosis in Aspergillus Fumigatus Keratitis. Invest Ophthalmol Vis Sci 2021; 61:28. [PMID: 32692841 PMCID: PMC7425693 DOI: 10.1167/iovs.61.8.28] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Purpose To explore the influence of indoleamine 2,3-dioxygenase (IDO) on macrophage recruitment, polarization and phagocytosis in Aspergillus fumigatus keratitis. Methods A murine model of A. fumigatus keratitis and peritoneal macrophages incubated with the hyphae of A. fumigatus were used. Macrophage recruitment in corneas was evaluated using immunofluorescence staining. The polarization of macrophages, which was stimulated by A. fumigatus and pretreatment with or without 1-methyltryptophan (1-MT), interferon gamma (IFNG), extracellular regulated protein kinases (ERK) antagonist, and p38 antagonist, was determined using reverse-transcription polymerase chain reaction and flow cytometry. P38 and ERK levels were determined using Western blotting. Macrophage phagocytosis was examined using colony-forming units. Results Compared with the A.F. group, recruitment of macrophages increased, tumor necrosis factor–α (TNF-α) and inducible nitric oxide synthase (iNOS) expression decreased, whereas arginase-1 (Arg-1) and interleukin-10 (IL-10) expression increased in the mouse corneas of the 1-MT+A.F. group. The ratio of CD206+/CD86+ macrophages in the corneas and spleens of 1-MT+A.F. group increased. Furthermore, in peritoneal macrophages stimulated by A. fumigatus, 1-MT promoted Arg-1 and IL-10 expression while upregulating the ratio of CD206+/CD86+ macrophages. Conversely, IDO agonist IFNG promoted TNF-α and iNOS expression, inhibited Arg-1 and IL-10 expression and downregulated the ratio of CD206+/CD86+ macrophages. The role of IFNG was reversed by the antagonist of P38 or ERK. P38 and ERK levels were downregulated in corneas of 1-MT+A.F. group. Besides, IFNG inhibited macrophage phagocytosis. Conclusions IDO inhibited macrophage recruitment and phagocytosis in A. fumigatus keratitis. Mechanistically, IDO is involved in M1 macrophage polarization in A. fumigatus keratitis through a MAPK/ERK-dependent pathway.
Collapse
|
16
|
Hua TR, Zhang SY. Cardiomyopathies in China: A 2018-2019 state-of-the-art review. Chronic Dis Transl Med 2020; 6:224-238. [PMID: 33336168 PMCID: PMC7729112 DOI: 10.1016/j.cdtm.2020.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Indexed: 11/02/2022] Open
Abstract
Cardiomyopathies are diseases of the cardiac muscle and are often characterized by ventricular dilation, hypertrophy, and cardiac arrhythmia. Patients with cardiomyopathies often experience sudden death and cardiac failure and require cardiac transplantation during the course of disease progression. Early diagnosis, differential diagnosis, and genetic consultation depend on imaging techniques, genetic testing, and new emerging diagnostic tools such as serum biomarkers. The molecular genetics of cardiomyopathies has been widely studied recently. The discovery of mechanisms underlying heterogeneity and overlapping of the phenotypes of cardiomyopathies has revealed the existence of disease modifiers, and this has led to the emergence of novel disease-modifying therapy. This 2018-2019 state-of-the-art review outlines the pathogenesis, diagnosis, and treatment of cardiomyopathies in China.
Collapse
Affiliation(s)
- Tian-Rui Hua
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Shu-Yang Zhang
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| |
Collapse
|
17
|
Zeng T, Deng G, Zhong W, Gao Z, Ma S, Mo C, Li Y, Huang S, Zhou C, Lai Y, Xie S, Xie Z, Chen Y, He S, Lv Z, Gao L. Indoleamine 2, 3-dioxygenase 1enhanceshepatocytes ferroptosis in acute immune hepatitis associated with excess nitrative stress. Free Radic Biol Med 2020; 152:668-679. [PMID: 31945497 DOI: 10.1016/j.freeradbiomed.2020.01.009] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 01/09/2020] [Indexed: 12/16/2022]
Abstract
Ferroptosis is a recently recognized form of regulated cell death that is characterized by lipid peroxidation. However, the molecular mechanisms of ferroptosis in acute immune hepatitis (AIH) are largely unknown. In this study, we investigated the classical ferroptotic events in the livers of mice with concanavalin A (ConA) to induce AIH. The dramatically upregulated gene indoleamine 2, 3-dioxygenase 1 (IDO1) was identified with AIH, and its role in generation of ferroptosis and reactive nitrogen species (RNS) was assessed both in vitro and in vivo by genetic deletion or pharmacologic inhibition of IDO1. We observed that ferroptosis contributed to the ConA-induced hepatic damage, which was confirmed by the therapeutical effects of ferroptosis inhibitor (ferrostatin-1). Noteworthy, upregulation of hepatic IDO1 and nitrative stress in ConA-induced hepatic damage were also remarkably inhibited by the ferroptosis abolishment. Additionally, IDO1 deficiency contributed to ferroptosis resistance by activating solute carrier family 7 member 11 (SLC7A11; also known as xCT) expression, accompanied with the reductions of murine liver lesions and RNS. Meanwhile, IDO inhibitor 1-methyl tryptophan alleviated murine liver damage with the reduction of inducible nitric oxide synthase and 3-nitrotyrosine expression. Consistent with the results in vivo, hepatocytes-specific knockdown of IDO1 led to ferroptosis resistance upon exposure to ferroptosis-inducing compound (Erastin) in vitro, whereas IDO1 overexpression aggravated the classical ferroptotic events, and the RNS stress. Overall, these results revealed a novel molecular mechanism of ferroptosis with the key feature of nitrative stress in ConA-induced liver injury, and also identified IDO1-dependent ferroptosis as a potential target for the treatment of AIH.
Collapse
Affiliation(s)
- Ting Zeng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Guanghui Deng
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Weichao Zhong
- Shenzhen Traditional Chinese Medicine Hospital, No.1, Fuhua Road, Futian District, Shenzhen, Guangdong, China
| | - Zhuowei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuoyi Ma
- Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Chan Mo
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yunjia Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Sha Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Chuying Zhou
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuqi Lai
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Shuwen Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Zeping Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuyao Chen
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China
| | - Songqi He
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Zhiping Lv
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| | - Lei Gao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|