1
|
Wojtacha P, Bogdańska-Chomczyk E, Majewski MK, Obremski K, Majewski MS, Kozłowska A. Renal Inflammation, Oxidative Stress, and Metabolic Abnormalities During the Initial Stages of Hypertension in Spontaneously Hypertensive Rats. Cells 2024; 13:1771. [PMID: 39513878 PMCID: PMC11545559 DOI: 10.3390/cells13211771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/03/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Hypertension is a major cause of mortality worldwide. The kidneys play a crucial role in regulating blood pressure and fluid volume. The relationship between the kidneys and hypertension is complex, involving factors such as the renin-angiotensin system, oxidative stress, and inflammation. This study aims to assess the levels of inflammatory markers, oxidative stress, and metabolic factors in the kidneys, focusing on their potential role in early renal damage and their association with the development of hypertension. Methods: This study was designed to compare the levels of selected inflammatory markers, e.g., interleukins, tumor necrosis factor-α (TNF-α), transforming growth factor, and serine/threonine-protein (mTOR); oxidative stress markers such as malondialdehyde, sulfhydryl group, and glucose (GLC); and metabolic markers among other enzymes, such as alanine transaminase (ALT), aspartate transaminase (AST), hexokinase II (HK-II), and hypoxia-inducible factor-1α (HIF-1α), as well as creatinine in the kidneys of spontaneously hypertensive rats (SHR/NCrl, n = 12) and Wistar Kyoto rats (WKY/NCrl, n = 12). Both juvenile (5 weeks old) and maturing (10 weeks old) specimens were examined using spectrophotometric methods, e.g., ELISA. Results: Juvenile SHRs exhibited reduced renal levels of all studied cytokines and chemokines, with lower oxidative stress and deficits in the mTOR and HK-II levels compared to the age-matched WKYs. Maturing SHRs showed increased renal levels of interleukin-1β (IL-1β), IL-6, IL-18, and TNF-α, alongside elevated carbonyl stress and increased HIF-1α as opposed to their control peers. The levels of all other studied markers were normalized in these animals, except for ALT (increased), ALP, and GLC (both reduced). Conclusions: This study underscores the significant impact of inflammatory, oxidative stress, and metabolic marker changes on renal function. Juvenile SHRs display lower marker levels, indicating an immature immune response and potential subclinical kidney damage that may contribute to hypertension development. In contrast, mature SHRs exhibit chronic inflammation, oxidative dysregulation, and metabolic disturbances, suggesting cellular damage. These changes create a feedback loop that worsens kidney function and accelerates hypertension progression, highlighting the kidneys' crucial role in both initiating and exacerbating this condition.
Collapse
Affiliation(s)
- Paweł Wojtacha
- Department of Psychology and Sociology of Health and Public Health, University of Warmia and Mazury, Warszawska Av, 10-082 Olsztyn, Poland
| | - Ewelina Bogdańska-Chomczyk
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| | - Mariusz Krzysztof Majewski
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| | - Kazimierz Obremski
- Department of Veterinary Prevention and Feed Hygiene, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13/29, 10-718 Olsztyn, Poland;
| | - Michał Stanisław Majewski
- Department of Pharmacology and Toxicology, Faculty of Medicine, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland;
| | - Anna Kozłowska
- Department of Human Physiology and Pathophysiology, Collegium Medicum, University of Warmia and Mazury, Warszawska Av, 30, 10-082 Olsztyn, Poland; (E.B.-C.); (M.K.M.)
| |
Collapse
|
2
|
Mosalmanzadeh N, Pence BD. Oxidized Low-Density Lipoprotein and Its Role in Immunometabolism. Int J Mol Sci 2024; 25:11386. [PMID: 39518939 PMCID: PMC11545486 DOI: 10.3390/ijms252111386] [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: 08/19/2024] [Revised: 10/04/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
Modified cholesterols such as oxidized low-density lipoprotein (OxLDL) contribute to atherosclerosis and other disorders through the promotion of foam cell formation and inflammation. In recent years, it has become evident that immune cell responses to inflammatory molecules such as OxLDLs depend on cellular metabolic functions. This review examines the known effects of OxLDL on immunometabolism and immune cell responses in atherosclerosis and several other diseases. We additionally provide context on the relationship between OxLDL and aging/senescence and identify gaps in the literature and our current understanding in these areas.
Collapse
Affiliation(s)
| | - Brandt D. Pence
- College of Health Sciences and Center for Nutraceutical and Dietary Supplement Research, University of Memphis, Memphis, TN 38111, USA
| |
Collapse
|
3
|
Chen Q, Li C, Wei W, Li J, Liu F, Fu Y, Tang L, Han F. Endoplasmic reticulum stress response pathway-mediated cell death in ovarian cancer. Front Oncol 2024; 14:1446552. [PMID: 39319052 PMCID: PMC11420017 DOI: 10.3389/fonc.2024.1446552] [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: 06/10/2024] [Accepted: 08/06/2024] [Indexed: 09/26/2024] Open
Abstract
The endoplasmic reticulum (ER) is one of the largest organelles, and Endoplasmic Reticulum Stress Response Pathway is a series of responses triggered by the homeostatic imbalance of the ER and the state in which unfolded or misfolded proteins accumulate in the ER, which can trigger cell death. Cell death plays a crucial role in the development of diseases such as gynecological oncology. Herein, we review the current research on the response and ovarian cancer, discussing the key sensors (IRE1, PERK, ATF6), and the conditions under which it occurs (Ca2+ homeostasis disruption, hypoxia, others). Using the response as a starting point, provide a comprehensive overview of the relationship with the four types of cell death (apoptosis, autophagy, immunogenic cell death, paraptosis) in an attempt to provide new targeted therapeutic strategies for the organelle-Endoplasmic Reticulum Stress Response Pathway-cell death in ovarian cancer therapy.
Collapse
Affiliation(s)
- Qiaochu Chen
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chan Li
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Wei Wei
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jia Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Fangyuan Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yuqian Fu
- Department of Obstetrics and Gynecology, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Liping Tang
- The Affiliated Tumor Hospital of Harbin Medical University, Harbin, China
| | - Fengjuan Han
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Wang Q, Shi P, Cao L, Li H, Chen X, Wang P, Zhang J. Unveiling the detrimental vicious cycle linking skeletal muscle and COVID-19: A systematic review and meta-analysis. J Evid Based Med 2024; 17:503-525. [PMID: 38975690 DOI: 10.1111/jebm.12629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 06/18/2024] [Indexed: 07/09/2024]
Abstract
OBJECTIVE Skeletal muscle catabolism supports multiple organs and systems during severe trauma and infection, but its role in COVID-19 remains unclear. This study investigates the interactions between skeletal muscle and COVID-19. METHODS The PubMed, EMbase, and The Cochrane Library databases were systematically searched from January 2020 to August 2023 for cohort studies focusing on the impact of skeletal muscle on COVID-19 prevalence and outcomes, and longitudinal studies examining skeletal muscle changes caused by COVID-19. Skeletal muscle quantity (SMQN) and quality (SMQL) were assessed separately. The random-effect model was predominantly utilized for statistical analysis. RESULTS Seventy studies with moderate to high quality were included. Low SMQN/SMQL was associated with an increased risk of COVID-19 infection (OR = 1.62, p < 0.001). Both the low SMQN and SMQL predicted COVID-19-related mortality (OR = 1.53, p = 0.016; OR = 2.18, p = 0.001, respectively). Mortality risk decreased with increasing SMQN (OR = 0.979, p = 0.009) and SMQL (OR = 0.972, p = 0.034). Low SMQN and SMQL were also linked to the need for intensive care unit/mechanical ventilation, increased COVID-19 severity, and longer hospital stays. Significant skeletal muscle wasting, characterized by reduced volume and strength, was observed during COVID-19 infection and the pandemic. CONCLUSIONS This study reveals a detrimental vicious circle between skeletal muscle and COVID-19. Effective management of skeletal muscle could be beneficial for treating COVID-19 infections and addressing the broader pandemic. These findings have important implications for the management of future virus pandemics. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42023395476.
Collapse
Affiliation(s)
- Qin Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peipei Shi
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Lu Cao
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haoran Li
- Department of Thoracic Surgery, Thoracic Oncology Institute, Peking University People's Hospital, Beijing, China
| | - Xiankai Chen
- Department of Thoracic Surgical Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peiyu Wang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Thoracic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Department of Thoracic Surgery, Thoracic Oncology Institute, Peking University People's Hospital, Beijing, China
| | - Jianjiang Zhang
- Department of Pediatrics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
5
|
Cutugno G, Kyriakidou E, Nadjar A. Rethinking the role of microglia in obesity. Neuropharmacology 2024; 253:109951. [PMID: 38615749 DOI: 10.1016/j.neuropharm.2024.109951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/16/2024]
Abstract
Microglia are the macrophages of the central nervous system (CNS), implying their role in maintaining brain homeostasis. To achieve this, these cells are sensitive to a plethora of endogenous and exogenous signals, such as neuronal activity, cellular debris, hormones, and pathological patterns, among many others. More recent research suggests that microglia are highly responsive to nutrients and dietary variations. In this context, numerous studies have demonstrated their significant role in the development of obesity under calorie surfeit. Because many reviews already exist on this topic, we have chosen to present the state of our reflections on various concepts put forth in the literature, bringing a new perspective whenever possible. Our literature review focuses on studies conducted in the arcuate nucleus of the hypothalamus, a key structure in the control of food intake. Specifically, we present the recent data available on the modifications of microglial energy metabolism following the consumption of an obesogenic diet and their consequences on hypothalamic neuron activity. We also highlight the studies unraveling the mechanisms underlying obesity-related sexual dimorphism. The review concludes with a list of questions that remain to be addressed in the field to achieve a comprehensive understanding of the role of microglia in the regulation of body energy metabolism. This article is part of the Special Issue on "Microglia".
Collapse
Affiliation(s)
- G Cutugno
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - E Kyriakidou
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France
| | - A Nadjar
- University of Bordeaux, INSERM, Neurocentre Magendie, Bordeaux, France; Institut Universitaire de France (IUF), France.
| |
Collapse
|
6
|
Yu XW, She PW, Chen FC, Chen YY, Zhou S, Wang XM, Lin XR, Liu QL, Huang ZJ, Qiu Y. Metabolic subtypes and immune landscapes in esophageal squamous cell carcinoma: prognostic implications and potential for personalized therapies. BMC Cancer 2024; 24:230. [PMID: 38373930 PMCID: PMC10875771 DOI: 10.1186/s12885-024-11890-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/16/2024] [Indexed: 02/21/2024] Open
Abstract
BACKGROUND This study aimed to identify metabolic subtypes in ESCA, explore their relationship with immune landscapes, and establish a metabolic index for accurate prognosis assessment. METHODS Clinical, SNP, and RNA-seq data were collected from 80 ESCA patients from the TCGA database and RNA-seq data from the GSE19417 dataset. Metabolic genes associated with overall survival (OS) and progression-free survival (PFS) were selected, and k-means clustering was performed. Immune-related pathways, immune infiltration, and response to immunotherapy were predicted using bioinformatic algorithms. Weighted gene co-expression network analysis (WGCNA) was conducted to identify metabolic genes associated with co-expression modules. Lastly, cell culture and functional analysis were performed using patient tissue samples and ESCA cell lines to verify the identified genes and their roles. RESULTS Molecular subtypes were identified based on the expression profiles of metabolic genes, and univariate survival analysis revealed 163 metabolic genes associated with ESCA prognosis. Consensus clustering analysis classified ESCA samples into three distinct subtypes, with MC1 showing the poorest prognosis and MC3 having the best prognosis. The subtypes also exhibited significant differences in immune cell infiltration, with MC3 showing the highest scores. Additionally, the MC3 subtype demonstrated the poorest response to immunotherapy, while the MC1 subtype was the most sensitive. WGCNA analysis identified gene modules associated with the metabolic index, with SLC5A1, NT5DC4, and MTHFD2 emerging as prognostic markers. Gene and protein expression analysis validated the upregulation of MTHFD2 in ESCA. MTHFD2 promotes the progression of ESCA and may be a potential therapeutic target for ESCA. CONCLUSION The established metabolic index and identified metabolic genes offer potential for prognostic assessment and personalized therapeutic interventions for ESCA, underscoring the importance of targeting metabolism-immune interactions in ESCA. MTHFD2 promotes the progression of ESCA and may be a potential therapeutic target for ESCA.
Collapse
Affiliation(s)
- Xiao-Wan Yu
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China.
| | - Pei-Wei She
- Shengli Clinical Medical College, Fujian Medical University, Fuzhou, Fujian, 350001, P. R. China
- Center for Experimental Research in Clinical Medicine, Fujian Provincial Hospital, 350001, Fuzhou, Fujian, P. R. China
| | - Fang-Chuan Chen
- Stomatology Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Ya-Yu Chen
- Stomatology Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Shuang Zhou
- Central Laboratory, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Xi-Min Wang
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Xiao-Rong Lin
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Qiao-Ling Liu
- Clinical Laboratory Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Zhi-Jun Huang
- Esophageal Surgery Department, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China
| | - Yu Qiu
- Reproductive Center, The Second Affiliated Hospital of Fujian Medical University, 362000, Quanzhou, Fujian, P. R. China.
| |
Collapse
|
7
|
Mu W, Patankar V, Kitchen S, Zhen A. Examining Chronic Inflammation, Immune Metabolism, and T Cell Dysfunction in HIV Infection. Viruses 2024; 16:219. [PMID: 38399994 PMCID: PMC10893210 DOI: 10.3390/v16020219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 01/26/2024] [Accepted: 01/29/2024] [Indexed: 02/25/2024] Open
Abstract
Chronic Human Immunodeficiency Virus (HIV) infection remains a significant challenge to global public health. Despite advances in antiretroviral therapy (ART), which has transformed HIV infection from a fatal disease into a manageable chronic condition, a definitive cure remains elusive. One of the key features of HIV infection is chronic immune activation and inflammation, which are strongly associated with, and predictive of, HIV disease progression, even in patients successfully treated with suppressive ART. Chronic inflammation is characterized by persistent inflammation, immune cell metabolic dysregulation, and cellular exhaustion and dysfunction. This review aims to summarize current knowledge of the interplay between chronic inflammation, immune metabolism, and T cell dysfunction in HIV infection, and also discusses the use of humanized mice models to study HIV immune pathogenesis and develop novel therapeutic strategies.
Collapse
Affiliation(s)
- Wenli Mu
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Vaibhavi Patankar
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Scott Kitchen
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Anjie Zhen
- Division of Hematology/Oncology, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
- UCLA AIDS Institute and the Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
8
|
Hu S, Duan H, Lu Y, Huang S. The genes regulating sensitivity of tumor cells to T cell-mediated killing: could they be potential personalized immunotherapeutic targets in head and neck squamous cell carcinoma? Discov Oncol 2023; 14:199. [PMID: 37926766 PMCID: PMC10625926 DOI: 10.1007/s12672-023-00806-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
OBJECTIVE To identify the pivotal genes, specifically the STTK genes, that govern the sensitivity of tumor cells to T cell-mediated killing in Head and Neck Squamous Cell Carcinoma (HNSC). METHODS The differentially expressed genes (DEGs) in HNSC and STTK genes were overlapped to obtain the DE-STTK genes. Univariate and LASSO regression analyses were conducted to identify the pivotal DE-STTK genes that serve as hubs in HNSC (i.e., hub DE-STTK genes). The risk model was established to divide HNSC tumor samples into high- and low-risk groups based on the hub DE-STTK genes. Further investigations were carried out by examing the expression level, prognostic values, diagnostic values, enriched signaling pathways, correlation with tumor mutation burden (TMB), and association with tumor immune infiltration cells (TIICs). RESULTS A total of 71 genes were found to be overlapped between DEGs in HNSC and STTK genes. Lasso regression analysis identified 9 hub genes which were MYF6, AATF, AURKA, CXCL9, DPM2, MYO1B, NCBP2, TNFRSF12A, and TRAF1. The network analysis of hub DE-STTK genes-pathway reveals that these 9 hub genes exhibit enrichment in multiple signaling pathways, including toll-like receptor signaling, TNF signaling, NF-kappa B signaling, cytokine-cytokine receptor interaction, spliceosome, mRNA surveillance pathway, nucleocytoplasmic transport, GPI-anchor biosynthesis, as well as N-Glycan biosynthesis. The Pearson correlation analysis showed that the majority of correlations between 9 hub DE-STTK genes and immune cells were positive. CONCLUSION The 9 identified hub DE-STTK genes (MYF6, AATF, AURKA, CXCL9, DPM2, MYO1B, NCBP2, TNFRSF12A, and TRAF1) are presumptively implicated in the modulation of tumor immunity in HNSC. These genes, along with their enriched pathways, hold promise as potential personalized immunotherapeutic targets for the treatment of HNSC, thereby offering novel avenues for therapeutic intervention in this malignancy.
Collapse
Affiliation(s)
- Shaonan Hu
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Heng Duan
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China
| | - Yongtao Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China.
| | - Shaohong Huang
- Stomatological Hospital, School of Stomatology, Southern Medical University, 366 Jiangnan South Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
9
|
Wang H, Li Z, Tao Y, Ou S, Ye J, Ran S, Luo K, Guan Z, Xiang J, Yan G, Wang Y, Ma T, Yu S, Song Y, Huang R. Characterization of endoplasmic reticulum stress unveils ZNF703 as a promising target for colorectal cancer immunotherapy. J Transl Med 2023; 21:713. [PMID: 37821882 PMCID: PMC10566095 DOI: 10.1186/s12967-023-04547-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Accepted: 09/20/2023] [Indexed: 10/13/2023] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is one of the most common malignant tumors globally, with high morbidity and mortality. Endoplasmic reticulum is a major organelle responsible for protein synthesis, processing, and transport. Endoplasmic reticulum stress (ERS) refers to the abnormal accumulation of unfolded and misfolded proteins in the endoplasmic reticulum, which are involved in tumorigenesis and cancer immunity. Nevertheless, the clinical significance of ERS remains largely unexplored in CRC. METHODS In present study, we performed an unsupervised clustering to identify two types of ERS-related subtypes [ERS clusters, and ERS-related genes (ERSGs) clusters] in multiple large-scale CRC cohorts. Through the utilization of machine learning techniques, we have successfully developed an uncomplicated yet robust gene scoring system (ERSGs signature). Furthermore, a series of analyses, including GO, KEGG, Tumor Immune Dysfunction and Exclusion (TIDE), the Consensus Molecular Subtypes (CMS), were used to explore the underlying biological differences and clinical significance between these groups. And immunohistochemical and bioinformatics analyses were performed to explore ZNF703, a gene of ERSGs scoring system. RESULTS We observed significant differences in prognosis and tumor immune status between the ERS clusters as well as ERSGs clusters. And the ERSGs scoring system was an independent risk factor for overall survival; and exhibited distinct tumor immune status in multicenter CRC cohorts. Besides, analyses of TNM stages, CMS groups demonstrated that patients in advanced stage and CMS4 had higher ERSGs scores. In addition, the ERSGs scores inversely correlated with positive ICB response predictors (such as, CD8A, CD274 (PD-L1), and TIS), and directly correlated with negative ICB response predictors (such as, TIDE, T cell Exclusion, COX-IS). Notably, immunohistochemical staining and bioinformatics analyses revealed that ZNF70 correlated with CD3 + and CD8 + T cells infiltration. CONCLUSION Based on large-scale and multicenter transcriptomic data, our study comprehensively revealed the essential role of ERS in CRC; and constructed a novel ERSGs scoring system to predict the prognosis of patients and the efficacy of ICB treatment. Furthermore, we identified ZNF703 as a potentially promising target for ICB therapy in CRC.
Collapse
Affiliation(s)
- Hufei Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Zhi Li
- Department of Obstetrics and Gynecology, the Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yangbao Tao
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Suwen Ou
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Jinhua Ye
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Songlin Ran
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Kangjia Luo
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Zilong Guan
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Jun Xiang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Guoqing Yan
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Yang Wang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Tianyi Ma
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China
| | - Shan Yu
- Department of Pathology, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China.
| | - Yanni Song
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Rui Huang
- Department of Colorectal Cancer Surgery, the Second Affiliated Hospital of Harbin Medical University, Harbin, 150080, China.
| |
Collapse
|
10
|
Li Y, Wei Y, Huang Y, Qin G, Zhao C, Ren J, Qu X. Lactate-Responsive Gene Editing to Synergistically Enhance Macrophage-Mediated Cancer Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2301519. [PMID: 37156740 DOI: 10.1002/smll.202301519] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/19/2023] [Indexed: 05/10/2023]
Abstract
Combination therapies involving metabolic regulation and immune checkpoint blockade are considered an encouraging new strategy for cancer therapy. However, the effective utilization of combination therapies for activating tumor-associated macrophages (TAMs) remains challenging. Herein, a lactate-catalyzed chemodynamic approach to activate the therapeutic genome editing of signal-regulatory protein α (SIRPα) to reprogram TAMs and improve cancer immunotherapy is proposed. This system is constructed by encapsulating lactate oxidase (LOx) and clustered regularly interspaced short palindromic repeat-mediated SIRPα genome-editing plasmids in a metal-organic framework (MOF). The genome-editing system is released and activated by acidic pyruvate, which is produced by the LOx-catalyzed oxidation of lactate. The synergy between lactate exhaustion and SIRPα signal blockade can enhance the phagocytic ability of TAMs and promote the repolarization of TAMs to the antitumorigenic M1 phenotype. Lactate exhaustion-induced CD47-SIRPα blockade efficiently improves macrophage antitumor immune responses and effectively reverses the immunosuppressive tumor microenvironment to inhibit tumor growth, as demonstrated by in vitro and in vivo studies. This study provides a facile strategy for engineering TAMs in situ by combining CRISPR-mediated SIRPα knockout with lactate exhaustion for effective immunotherapy.
Collapse
Affiliation(s)
- Yuwei Li
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Yue Wei
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Ying Huang
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Geng Qin
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Chuanqi Zhao
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Jinsong Ren
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| | - Xiaogang Qu
- Laboratory of Chemical Biology and State Key Laboratory of Rare Earth Resource Utilization, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, Anhui, 230026, P. R. China
| |
Collapse
|
11
|
Daoud A, Lema DA, Won T, Čiháková D. Integrative single-cell analysis of cardiac and pulmonary sarcoidosis using publicly available cardiac and bronchoalveolar lavage fluid sequencing datasets. Front Cardiovasc Med 2023; 10:1227818. [PMID: 37576111 PMCID: PMC10419306 DOI: 10.3389/fcvm.2023.1227818] [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: 05/23/2023] [Accepted: 07/03/2023] [Indexed: 08/15/2023] Open
Abstract
Introduction Cardiac presentation of autoimmune sarcoidosis, known as cardiac sarcoidosis (CS), is a poorly understood disease with high mortality and low diagnosis rate. While CS is an immunological syndrome, little is known about how cardiac parenchymal and stromal cells mediate its pathogenesis. Moreover, while most current sarcoidosis research is based on research in pulmonary sarcoidosis (PS), it remains unclear how much both presentations of sarcoidosis overlap. To tackle these concerns, we leveraged publicly available sarcoidosis transcriptomic datasets. Methods Two publicly available bronchoalveolar lavage single-cell RNA sequencing datasets were integrated to analyze PS relative to control. Additionally, two publicly available cardiac single-nucleus RNA sequencing datasets were integrated to analyze CS relative to control. Following integration, we ran cell-cell communication, transcription factor, and differential expression analyses on parenchymal, stromal, and immune subsets identified in our analysis. Results Our analysis revealed that there was an expansion of stromal and immune cells in PS and CS. We also observed upregulation of Th17.1 and attenuated activation transcriptional profiles in the immune cells of CS and PS relative to control. Additionally, we found upregulation of pro-inflammatory and pro-fibrotic transcriptional profiles in the cardiac stromal cells of CS relative to control. We also found that cardiomyocytes exhibited upregulated cardiac stress and proliferation transcriptional profiles in CS relative to control. Conclusions Our integrative transcriptomic analysis shows that despite tissue-specific differences, there are shared transcriptional trends between CS and PS. It also shows that stromal and parenchymal populations exhibit transcriptional trends that could explain their pathogenic role in CS.
Collapse
Affiliation(s)
- Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Diego A. Lema
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Pinho SA, Anjo SI, Cunha-Oliveira T. Metabolic Priming as a Tool in Redox and Mitochondrial Theragnostics. Antioxidants (Basel) 2023; 12:1072. [PMID: 37237939 PMCID: PMC10215850 DOI: 10.3390/antiox12051072] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/05/2023] [Accepted: 05/06/2023] [Indexed: 05/28/2023] Open
Abstract
Theragnostics is a promising approach that integrates diagnostics and therapeutics into a single personalized strategy. To conduct effective theragnostic studies, it is essential to create an in vitro environment that accurately reflects the in vivo conditions. In this review, we discuss the importance of redox homeostasis and mitochondrial function in the context of personalized theragnostic approaches. Cells have several ways to respond to metabolic stress, including changes in protein localization, density, and degradation, which can promote cell survival. However, disruption of redox homeostasis can lead to oxidative stress and cellular damage, which are implicated in various diseases. Models of oxidative stress and mitochondrial dysfunction should be developed in metabolically conditioned cells to explore the underlying mechanisms of diseases and develop new therapies. By choosing an appropriate cellular model, adjusting cell culture conditions and validating the cellular model, it is possible to identify the most promising therapeutic options and tailor treatments to individual patients. Overall, we highlight the importance of precise and individualized approaches in theragnostics and the need to develop accurate in vitro models that reflect the in vivo conditions.
Collapse
Affiliation(s)
- Sónia A. Pinho
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197 Cantanhede, Portugal; (S.A.P.); (S.I.A.)
- PDBEB—PhD Programme in Experimental Biology and Biomedicine, Institute of Interdisciplinary Research (IIIUC), University of Coimbra, 3004-504 Coimbra, Portugal
- IIIUC, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sandra I. Anjo
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197 Cantanhede, Portugal; (S.A.P.); (S.I.A.)
- IIIUC, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Teresa Cunha-Oliveira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3060-197 Cantanhede, Portugal; (S.A.P.); (S.I.A.)
- IIIUC, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
13
|
Hypoxia and Intestinal Inflammation: Common Molecular Mechanisms and Signaling Pathways. Int J Mol Sci 2023; 24:ijms24032425. [PMID: 36768744 PMCID: PMC9917195 DOI: 10.3390/ijms24032425] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
The gastrointestinal tract (GI) has a unique oxygenation profile. It should be noted that the state of hypoxia can be characteristic of both normal and pathological conditions. Hypoxia-inducible factors (HIF) play a key role in mediating the response to hypoxia, and they are tightly regulated by a group of enzymes called HIF prolyl hydroxylases (PHD). In this review, we discuss the involvement of inflammation hypoxia and signaling pathways in the pathogenesis of inflammatory bowel disease (IBD) and elaborate in detail on the role of HIF in multiple immune reactions during intestinal inflammation. We emphasize the critical influence of tissue microenvironment and highlight the existence of overlapping functions and immune responses mediated by the same molecular mechanisms. Finally, we also provide an update on the development of corresponding therapeutic approaches that would be useful for treatment or prophylaxis of inflammatory bowel disease.
Collapse
|
14
|
Review to Understand the Crosstalk between Immunotherapy and Tumor Metabolism. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020862. [PMID: 36677919 PMCID: PMC9863813 DOI: 10.3390/molecules28020862] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/11/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023]
Abstract
Immune checkpoint inhibitors have ushered in a new era of cancer treatment by increasing the likelihood of long-term survival for patients with metastatic disease and by introducing fresh therapeutic indications in cases where the disease is still in its early stages. Immune checkpoint inhibitors that target the proteins cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) or programmed death-1/programmed death ligand-1 have significantly improved overall survival in patients with certain cancers and are expected to help patients achieve complete long-lasting remissions and cures. Some patients who receive immune checkpoint inhibitors, however, either experience therapeutic failure or eventually develop immunotherapy resistance. Such individuals are common, which necessitates a deeper understanding of how cancer progresses, particularly with regard to nutritional regulation in the tumor microenvironment (TME), which comprises metabolic cross-talk between metabolites and tumor cells as well as intracellular metabolism in immune and cancer cells. Combination of immunotherapy with targeted metabolic regulation might be a focus of future cancer research despite a lack of existing clinical evidence. Here, we reviewed the significance of the tumor microenvironment and discussed the most significant immunological checkpoints that have recently been identified. In addition, metabolic regulation of tumor immunity and immunological checkpoints in the TME, including glycolysis, amino acid metabolism, lipid metabolism, and other metabolic pathways were also incorporated to discuss the possible metabolism-based treatment methods being researched in preclinical and clinical settings. This review will contribute to the identification of a relationship or crosstalk between tumor metabolism and immunotherapy, which will shed significant light on cancer treatment and cancer research.
Collapse
|
15
|
Shi Y, Gu L, Zhang X, Chen M. Traditional Chinese medicine mediated tumor suppression via regulating psychological factors. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
16
|
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: 62] [Impact Index Per Article: 31.0] [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
|
17
|
Chen G, Wu K, Li H, Xia D, He T. Role of hypoxia in the tumor microenvironment and targeted therapy. Front Oncol 2022; 12:961637. [PMID: 36212414 PMCID: PMC9545774 DOI: 10.3389/fonc.2022.961637] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 09/01/2022] [Indexed: 11/21/2022] Open
Abstract
Tumor microenvironment (TME), which is characterized by hypoxia, widely exists in solid tumors. As a current research hotspot in the TME, hypoxia is expected to become a key element to break through the bottleneck of tumor treatment. More and more research results show that a variety of biological behaviors of tumor cells are affected by many factors in TME which are closely related to hypoxia. In order to inhibiting the immune response in TME, hypoxia plays an important role in tumor cell metabolism and anti-apoptosis. Therefore, exploring the molecular mechanism of hypoxia mediated malignant tumor behavior and therapeutic targets is expected to provide new ideas for anti-tumor therapy. In this review, we discussed the effects of hypoxia on tumor behavior and its interaction with TME from the perspectives of immune cells, cell metabolism, oxidative stress and hypoxia inducible factor (HIF), and listed the therapeutic targets or signal pathways found so far. Finally, we summarize the current therapies targeting hypoxia, such as glycolysis inhibitors, anti-angiogenesis drugs, HIF inhibitors, hypoxia-activated prodrugs, and hyperbaric medicine.
Collapse
Affiliation(s)
- Gaoqi Chen
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Kaiwen Wu
- Department of Gastroenterology, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, Chengdu, China
| | - Hao Li
- Deparment of Neurology, Affiliated Hospital of Jiangsu University, Jiang Su University, Zhenjiang, China
| | - Demeng Xia
- Luodian Clinical Drug Research Center, Shanghai Baoshan Luodian Hospital, Shanghai University, Shanghai, China
- *Correspondence: Demeng Xia, ; Tianlin He,
| | - Tianlin He
- Department of Hepatobiliary Pancreatic Surgery, Changhai Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
- *Correspondence: Demeng Xia, ; Tianlin He,
| |
Collapse
|
18
|
Yadav S, Dwivedi A, Tripathi A. Biology of macrophage fate decision: Implication in inflammatory disorders. Cell Biol Int 2022; 46:1539-1556. [PMID: 35842768 DOI: 10.1002/cbin.11854] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/04/2022] [Accepted: 06/18/2022] [Indexed: 11/11/2022]
Abstract
The activation of immune cells in response to stimuli present in their microenvironment is regulated by their metabolic profile. Unlike the signal transduction events, which overlap to a huge degree in diverse cellular processes, the metabolome of a cell reflects a more precise picture of cell physiology and function. Different factors governing the cellular metabolome include receptor signaling, macro and micronutrients, normoxic and hypoxic conditions, energy needs, and biomass demand. Macrophages have enormous plasticity and can perform diverse functions depending upon their phenotypic state. This review presents recent updates on the cellular metabolome and molecular patterns associated with M1 and M2 macrophages, also termed "classically activated macrophages" and "alternatively activated macrophages," respectively. M1 macrophages are proinflammatory in nature and predominantly Th1-specific immune responses induce their polarization. On the contrary, M2 macrophages are anti-inflammatory in nature and primarily participate in Th2-specific responses. Interestingly, the same macrophage cell can adapt to the M1 or M2 phenotype depending upon the clues from its microenvironment. We elaborate on the various tissue niche-specific factors, which govern macrophage metabolism and heterogeneity. Furthermore, the current review provides an in-depth account of deregulated macrophage metabolism associated with pathological disorders such as cancer, obesity, and atherosclerosis. We further highlight significant differences in various metabolic pathways governing the cellular bioenergetics and their impact on macrophage effector functions and associated disorders.
Collapse
Affiliation(s)
- Sarika Yadav
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Ashish Dwivedi
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| | - Anurag Tripathi
- Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research, Lucknow, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
19
|
Jiang X, Tian W, Kim D, McQuiston AS, Vinh R, Rockson SG, Semenza GL, Nicolls MR. Hypoxia and Hypoxia-Inducible Factors in Lymphedema. Front Pharmacol 2022; 13:851057. [PMID: 35450048 PMCID: PMC9017680 DOI: 10.3389/fphar.2022.851057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/14/2022] [Indexed: 12/19/2022] Open
Abstract
Lymphedema is a chronic inflammatory disorder characterized by edema, fat deposition, and fibrotic tissue remodeling. Despite significant advances in lymphatic biology research, our knowledge of lymphedema pathology is incomplete. Currently, there is no approved pharmacological therapy for this debilitating disease. Hypoxia is a recognized feature of inflammation, obesity, and fibrosis. Understanding hypoxia-regulated pathways in lymphedema may provide new insights into the pathobiology of this chronic disorder and help develop new medicinal treatments.
Collapse
Affiliation(s)
- Xinguo Jiang
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | - Wen Tian
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | - Dongeon Kim
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | - Alexander S McQuiston
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | - Ryan Vinh
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| | | | - Gregg L Semenza
- Departments of Pediatrics, Medicine, Oncology, Radiation Oncology, and Biological Chemistry, and McKusick-Nathans Institute of Genetic Medicine, Vascular Program, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Mark R Nicolls
- VA Palo Alto Health Care System, Palo Alto, CA, United States.,Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
20
|
Li J, Li X, Li M, Qiu H, Saad C, Zhao B, Li F, Wu X, Kuang D, Tang F, Chen Y, Shu H, Zhang J, Wang Q, Huang H, Qi S, Ye C, Bryant A, Yuan X, Kurts C, Hu G, Cheng W, Mei Q. Differential early diagnosis of benign versus malignant lung cancer using systematic pathway flux analysis of peripheral blood leukocytes. Sci Rep 2022; 12:5070. [PMID: 35332177 PMCID: PMC8948197 DOI: 10.1038/s41598-022-08890-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 03/07/2022] [Indexed: 12/24/2022] Open
Abstract
Early diagnosis of lung cancer is critically important to reduce disease severity and improve overall survival. Newer, minimally invasive biopsy procedures often fail to provide adequate specimens for accurate tumor subtyping or staging which is necessary to inform appropriate use of molecular targeted therapies and immune checkpoint inhibitors. Thus newer approaches to diagnosis and staging in early lung cancer are needed. This exploratory pilot study obtained peripheral blood samples from 139 individuals with clinically evident pulmonary nodules (benign and malignant), as well as ten healthy persons. They were divided into three cohorts: original cohort (n = 99), control cohort (n = 10), and validation cohort (n = 40). Average RNAseq sequencing of leukocytes in these samples were conducted. Subsequently, data was integrated into artificial intelligence (AI)-based computational approach with system-wide gene expression technology to develop a rapid, effective, non-invasive immune index for early diagnosis of lung cancer. An immune-related index system, IM-Index, was defined and validated for the diagnostic application. IM-Index was applied to assess the malignancies of pulmonary nodules of 109 participants (original + control cohorts) with high accuracy (AUC: 0.822 [95% CI: 0.75-0.91, p < 0.001]), and to differentiate between phases of cancer immunoediting concept (odds ratio: 1.17 [95% CI: 1.1-1.25, p < 0.001]). The predictive ability of IM-Index was validated in a validation cohort with a AUC: 0.883 (95% CI: 0.73-1.00, p < 0.001). The difference between molecular mechanisms of adenocarcinoma and squamous carcinoma histology was also determined via the IM-Index (OR: 1.2 [95% CI 1.14-1.35, p = 0.019]). In addition, a structural metabolic behavior pattern and signaling property in host immunity were found (bonferroni correction, p = 1.32e - 16). Taken together our findings indicate that this AI-based approach may be used for "Super Early" cancer diagnosis and amend the current immunotherpay for lung cancer.
Collapse
Affiliation(s)
- Jian Li
- Institute of Molecular Medicine and Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Xiaoyu Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Ming Li
- Department of Oncology, Wuhan Pulmonary Hospital, Wuhan, Hubei, People's Republic of China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Christian Saad
- Department of Computer Science, University of Augsburg, Augsburg, Germany
| | - Bo Zhao
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Fan Li
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Xiaowei Wu
- Department of Thoracic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Dong Kuang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Fengjuan Tang
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Yaobing Chen
- Institute of Pathology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
- Department of Pathology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Hongge Shu
- Radiology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Jing Zhang
- Radiology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Qiuxia Wang
- Radiology Department, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - He Huang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Shankang Qi
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, People's Republic of China
| | - Changkun Ye
- Medical Research Center of Yu Huang Hospital, Yu Huang, Zhejiang, People's Republic of China
| | - Amy Bryant
- Department of Biochemical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, USA
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University Clinic of Rheinische Friedrich-Wilhelms-University, Bonn, Germany
| | - Guangyuan Hu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| | - Weiting Cheng
- Department of Oncology, Wuhan No. 1 Hospital, Wuhan, Hubei, People's Republic of China.
| | - Qi Mei
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China.
| |
Collapse
|
21
|
Munansangu BSM, Kenyon C, Walzl G, Loxton AG, Kotze LA, du Plessis N. Immunometabolism of Myeloid-Derived Suppressor Cells: Implications for Mycobacterium tuberculosis Infection and Insights from Tumor Biology. Int J Mol Sci 2022; 23:ijms23073512. [PMID: 35408873 PMCID: PMC8998693 DOI: 10.3390/ijms23073512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/04/2023] Open
Abstract
The field of immunometabolism seeks to decipher the complex interplay between the immune system and the associated metabolic pathways. The role of small molecules that can target specific metabolic pathways and subsequently alter the immune landscape provides a desirable platform for new therapeutic interventions. Immunotherapeutic targeting of suppressive cell populations, such as myeloid-derived suppressor cells (MDSC), by small molecules has shown promise in pathologies such as cancer and support testing of similar host-directed therapeutic approaches in MDSC-inducing conditions such as tuberculosis (TB). MDSC exhibit a remarkable ability to suppress T-cell responses in those with TB disease. In tumors, MDSC exhibit considerable plasticity and can undergo metabolic reprogramming from glycolysis to fatty acid oxidation (FAO) and oxidative phosphorylation (OXPHOS) to facilitate their immunosuppressive functions. In this review we look at the role of MDSC during M. tb infection and how their metabolic reprogramming aids in the exacerbation of active disease and highlight the possible MDSC-targeted metabolic pathways utilized during M. tb infection, suggesting ways to manipulate these cells in search of novel insights for anti-TB therapies.
Collapse
|
22
|
Shao F, Panahipour L, Sordi MB, Tang F, Liu R, Gruber R. Heartwood of Dalbergia cochinchinensis: 4,7,2'-Trihydroxy-4'-methoxyisoflavanol and 6,4'-Dihydroxy-7-methoxyflavane Reduce Cytokine and Chemokine Expression In Vitro. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041321. [PMID: 35209110 PMCID: PMC8879141 DOI: 10.3390/molecules27041321] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/14/2021] [Accepted: 02/11/2022] [Indexed: 02/02/2023]
Abstract
Dalbergia cochinchinensis has been widely used in traditional medicine because of its flavonoids; however, the impact of the flavonoids to modulate the inflammatory response to oral cells remains to be described. For this aim, we isolated 4,7,2'-trihydroxy-4'-methoxyisoflavanol (472T4MIF) and 6,4'-dihydroxy-7-methoxyflavane (64D7MF) from the heartwood of D. cochinchinensis and confirmed the chemical structure by nuclear magnetic resonance. We show here that both flavonoids are inhibitors of an inflammatory response of murine RAW 264.7 inflammatory macrophages stimulated by LPS. This is indicated by interleukin (IL)1, IL6, and chemokine CCL2 production besides the phosphorylation of p65. Consistently, in primary murine macrophages, both flavonoids decreased the inflammatory response by lowering LPS-induced IL1 and IL6 expression. To introduce oral cells, we have used human gingival fibroblasts and provoked the inflammatory response by exposing them to IL1β and TNFα. Under these conditions, 472T4MIF, but not 64D7MF, reduced the expression of chemokines CXCL1 and CXCL2. Taken together, we identified two flavonoids that can reduce the expression of cytokines and chemokines in macrophages and fibroblastic cells.
Collapse
Affiliation(s)
- Feng Shao
- Department of Oral Biology, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (M.B.S.)
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (F.T.); (R.L.)
- Key Laboratory of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Chinese Medicine, Nanchang 330004, China
- Correspondence: (F.S.); (R.G.)
| | - Layla Panahipour
- Department of Oral Biology, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (M.B.S.)
| | - Mariane Beatriz Sordi
- Department of Oral Biology, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (M.B.S.)
- Department of Dentistry, Federal University of Santa Catarina, Florianopolis 88040-900, Brazil
| | - Fangrui Tang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (F.T.); (R.L.)
| | - Ronghua Liu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, Jiangxi University of Chinese Medicine, Nanchang 330004, China; (F.T.); (R.L.)
| | - Reinhard Gruber
- Department of Oral Biology, Medical University of Vienna, 1090 Vienna, Austria; (L.P.); (M.B.S.)
- Department of Periodontology, School of Dental Medicine, University of Bern, 3012 Bern, Switzerland
- Austrian Cluster for Tissue Regeneration, 1200 Vienna, Austria
- Correspondence: (F.S.); (R.G.)
| |
Collapse
|
23
|
Boyer T, Blaye C, Larmonier N, Domblides C. Influence of the Metabolism on Myeloid Cell Functions in Cancers: Clinical Perspectives. Cells 2022; 11:cells11030554. [PMID: 35159363 PMCID: PMC8834417 DOI: 10.3390/cells11030554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/02/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
Tumor metabolism plays a crucial role in sustaining tumorigenesis. There have been increasing reports regarding the role of tumor metabolism in the control of immune cell functions, generating a potent immunosuppressive contexture that can lead to immune escape. The metabolic reprogramming of tumor cells and the immune escape are two major hallmarks of cancer, with several instances of crosstalk between them. In this paper, we review the effects of tumor metabolism on immune cells, focusing on myeloid cells due to their important role in tumorigenesis and immunosuppression from the early stages of the disease. We also discuss ways to target this specific crosstalk in cancer patients.
Collapse
Affiliation(s)
- Thomas Boyer
- CNRS UMR5164, ImmunoConcEpT, Site de Carreire, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France; (T.B.); (C.B.); (N.L.)
- Department of Life and Medical Sciences, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Céline Blaye
- CNRS UMR5164, ImmunoConcEpT, Site de Carreire, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France; (T.B.); (C.B.); (N.L.)
- Department of Life and Medical Sciences, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France
- Department of Medical Oncology, Bergonié Institute, 229 cours de l’Argonne, 33076 Bordeaux, France
| | - Nicolas Larmonier
- CNRS UMR5164, ImmunoConcEpT, Site de Carreire, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France; (T.B.); (C.B.); (N.L.)
- Department of Life and Medical Sciences, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France
| | - Charlotte Domblides
- CNRS UMR5164, ImmunoConcEpT, Site de Carreire, University of Bordeaux, 146 Rue Léo Saignat, 33076 Bordeaux, France; (T.B.); (C.B.); (N.L.)
- Department of Medical Oncology, Bergonié Institute, 229 cours de l’Argonne, 33076 Bordeaux, France
- Department of Medical Oncology, Hôpital Saint-André, 1 rue Jean Burguet, University Hospital Bordeaux, 33076 Bordeaux, France
- Correspondence:
| |
Collapse
|
24
|
Metabolomics Signatures of SARS-CoV-2 Infection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1376:45-59. [PMID: 34735713 DOI: 10.1007/5584_2021_674] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
For a very long time, viral infections have been considered as one of the most important causes of death and disability around the world. Through the viral infection, viruses as small pathogens enter the host cells and use hosts' biosynthesis machinery to replicate and collect infectious lineages. Moreover, they can modify hosts' metabolic pathways in order to their own purposes. Nowadays (in 2019-2020), the most famous type of viral infection which was caused by a novel type of coronavirus is called COVID-19 disease. It has claimed the lives of many people around the world and is a very serious threat to health. Since investigations of the effects of viruses on host metabolism using metabolomics tools may have given focuses on novel appropriate treatments, in the current review the authors highlighted the virus-host metabolic interactions and metabolomics perspective in COVID-19.
Collapse
|
25
|
Udumula MP, Sakr S, Dar S, Alvero AB, Ali-Fehmi R, Abdulfatah E, Li J, Jiang J, Tang A, Buekers T, Morris R, Munkarah A, Giri S, Rattan R. Ovarian cancer modulates the immunosuppressive function of CD11b +Gr1 + myeloid cells via glutamine metabolism. Mol Metab 2021; 53:101272. [PMID: 34144215 PMCID: PMC8267600 DOI: 10.1016/j.molmet.2021.101272] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 06/03/2021] [Accepted: 06/11/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVE Immature CD11b + Gr1+ myeloid cells that acquire immunosuppressive capability, also known as myeloid-derived suppressor cells (MDSCs), are a heterogeneous population of cells that regulate immune responses. Our study's objective was to elucidate the role of ovarian cancer microenvironment in regulating the immunosuppressive function of CD11b+Gr1+ myeloid cells. METHODS All studies were performed using the intraperitoneal ID8 syngeneic epithelial ovarian cancer mouse model. Myeloid cell depletion and immunotherapy were carried out using anti-Gr1 mAb, gemcitabine treatments, and/or anti-PD1 mAb. The treatment effect was assessed by a survival curve, in situ luciferase-guided imaging, and histopathologic evaluation. Adoptive transfer assays were carried out between congenic CD45.2 and CD45.1 mice. Immune surface and intracellular markers were assessed by flow cytometry. ELISA, western blot, and RT-PCR techniques were employed to assess the protein and RNA expression of various markers. Bone marrow-derived myeloid cells were used for ex-vivo studies. RESULTS The depletion of Gr1+ immunosuppressive myeloid cells alone and in combination with anti-PD1 immunotherapy inhibited ovarian cancer growth. In addition to the adoptive transfer studies, these findings validate the role of immunosuppressive CD11b+Gr1+ myeloid cells in promoting ovarian cancer. Mechanistic investigations showed that ID8 tumor cells and their microenvironments produced recruitment and regulatory factors for immunosuppressive CD11b+Gr1+ myeloid cells. CD11b+Gr1+ myeloid cells primed by ID8 tumors showed increased immunosuppressive marker expression and acquired an energetic metabolic phenotype promoted primarily by increased oxidative phosphorylation fueled by glutamine. Inhibiting the glutamine metabolic pathway reduced the increased oxidative phosphorylation and decreased immunosuppressive markers' expression and function. Dihydrolipoamide succinyl transferase (DLST), a subunit of α-KGDC in the TCA cycle, was found to be the most significantly elevated gene in tumor-primed myeloid cells. The inhibition of DLST reduced oxidative phosphorylation, immunosuppressive marker expression and function in myeloid cells. CONCLUSION Our study shows that the ovarian cancer microenvironment can regulate the metabolism and function of immunosuppressive CD11b + Gr1+ myeloid cells and modulate its immune microenvironment. Targeting glutamine metabolism via DLST in immunosuppressive myeloid cells decreased their activity, leading to a reduction in the immunosuppressive tumor microenvironment. Thus, targeting glutamine metabolism has the potential to enhance the success of immunotherapy in ovarian cancer.
Collapse
Affiliation(s)
- Mary P Udumula
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Sharif Sakr
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Sajad Dar
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Ayesha B Alvero
- Mott Center for Human Growth and Development, Department of Obstetrics and Gynecology, Wayne State University, Detroit, MI, USA
| | - Rouba Ali-Fehmi
- Department of Pathology, Wayne State University and Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Eman Abdulfatah
- Department of Pathology, Wayne State University and Barbara Ann Karmanos Cancer Institute, Detroit, MI, USA
| | - Jing Li
- Metabolomics Core, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Jun Jiang
- Metabolomics Core, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Amy Tang
- Department of Public Health Services, Henry Ford Health System, Detroit, MI, USA
| | - Thomas Buekers
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA; Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Robert Morris
- Department of Gynecology Oncology, Barbara Ann Karmanos Cancer Institute and Wayne State University, Detroit, MI, USA
| | - Adnan Munkarah
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA
| | - Shailendra Giri
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Ramandeep Rattan
- Division of Gynecology Oncology, Department of Women's Health Services, Henry Ford Cancer Institute and Henry Ford Health System, Detroit, MI, USA; Department of Oncology, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
26
|
Li L, Gao H, Wang D, Jiang H, Wang H, Yu J, Jiang X, Huang C. Metabolism-Relevant Molecular Classification Identifies Tumor Immune Microenvironment Characterization and Immunotherapeutic Effect in Cervical Cancer. Front Mol Biosci 2021; 8:624951. [PMID: 34277697 PMCID: PMC8280349 DOI: 10.3389/fmolb.2021.624951] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 06/14/2021] [Indexed: 12/19/2022] Open
Abstract
Cervical cancer (CESC) is a gynecologic malignant tumor associated with high incidence and mortality rates because of its distinctive management complexity. Herein, we characterized the molecular features of CESC based on the metabolic gene expression profile by establishing a novel classification system and a scoring system termed as METAscore. Integrative analysis was performed on human CESC samples from TCGA dataset. Unsupervised clustering of RNA sequencing data on 2,752 formerly described metabolic genes identified three METAclusters. These METAclusters for overall survival time, immune characteristics, metabolic features, transcriptome features, and immunotherapeutic effectiveness existed distinct differences. Then we analyzed 207 DEGs among the three METAclusters and as well identified three geneclusters. Correspondingly, these three geneclusters also differently expressed among the aforementioned features, supporting the reliability of the metabolism-relevant molecular classification. Finally METAscore was constructed which emerged as an independent prognostic biomarker, related to CESC transcriptome features, metabolic features, immune characteristics, and linked to the sensitivity of immunotherapy for individual patient. These findings depicted a new classification and a scoring system in CESC based on the metabolic pattern, thereby furthering the understanding of CESC genetic signatures and aiding in the prediction of the effectiveness to anticancer immunotherapies.
Collapse
Affiliation(s)
- Luyi Li
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China.,The 2 Afflicated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hui Gao
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China.,Key Laboratory of Fertility Preservation and Maintenance of Ministry of Education, Ningxia Medical University, Yinchuan, China
| | - Danhan Wang
- The 2 Afflicated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hao Jiang
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China
| | - Hongzhu Wang
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China
| | - Jiajian Yu
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China
| | - Xin Jiang
- Prenatal Diagnosis Center of NanFang Hospital, The Southern Medical University, Guangzhou, China
| | - Changjiang Huang
- Institude of Environmental Safety and Human Health, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
27
|
Dang CP, Issara-Amphorn J, Charoensappakit A, Udompornpitak K, Bhunyakarnjanarat T, Saisorn W, Sae-Khow K, Leelahavanichkul A. BAM15, a Mitochondrial Uncoupling Agent, Attenuates Inflammation in the LPS Injection Mouse Model: An Adjunctive Anti-Inflammation on Macrophages and Hepatocytes. J Innate Immun 2021; 13:359-375. [PMID: 34062536 PMCID: PMC8613553 DOI: 10.1159/000516348] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 03/30/2021] [Indexed: 11/19/2022] Open
Abstract
Controlof immune responses through the immunometabolism interference is interesting for sepsis treatment. Then, expression of immunometabolism-associated genes and BAM15, a mitochondrial uncoupling agent, was explored in a proinflammatory model using lipopolysaccharide (LPS) injection. Accordingly, the decreased expression of mitochondrial uncoupling proteins was demonstrated by transcriptomic analysis on metabolism-associated genes in macrophages (RAW246.7) and by polymerase chain reaction in LPS-stimulated RAW246.7 and hepatocytes (Hepa 1-6). Pretreatment with BAM15 at 24 h prior to LPS in macrophages attenuated supernatant inflammatory cytokines (IL-6, TNF-α, and IL-10), downregulated genes of proinflammatory M1 polarization (iNOS and IL-1β), upregulated anti-inflammatory M2 polarization (Arg1 and FIZZ), and decreased cell energy status (extracellular flux analysis and ATP production). Likewise, BAM15 decreased expression of proinflammatory genes (IL-6, TNF-α, IL-10, and iNOS) and reduced cell energy in hepatocytes. In LPS-administered mice, BAM15 attenuated serum cytokines, organ injury (liver enzymes and serum creatinine), and tissue cytokines (livers and kidneys), in part, through the enhanced phosphorylated αAMPK, a sensor of ATP depletion with anti-inflammatory property, in the liver, and reduced inflammatory monocytes/macrophages (Ly6C +ve, CD11b +ve) in the liver as detected by Western blot and flow cytometry, respectively. In conclusion, a proof of concept for inflammation attenuation of BAM15 through metabolic interference-induced anti-inflammation on macrophages and hepatocytes was demonstrated as a new strategy of anti-inflammation in sepsis.
Collapse
Affiliation(s)
- Cong Phi Dang
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand,
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand,
| | | | - Awirut Charoensappakit
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kanyarat Udompornpitak
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | | | - Wilasinee Saisorn
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Kritsanawan Sae-Khow
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Translational Research in Inflammation and Immunology Research Unit (TRIRU), Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
28
|
Nie Z, Chen M, Wen X, Gao Y, Huang D, Cao H, Peng Y, Guo N, Ni J, Zhang S. Endoplasmic Reticulum Stress and Tumor Microenvironment in Bladder Cancer: The Missing Link. Front Cell Dev Biol 2021; 9:683940. [PMID: 34136492 PMCID: PMC8201605 DOI: 10.3389/fcell.2021.683940] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 04/28/2021] [Indexed: 02/06/2023] Open
Abstract
Bladder cancer is a common malignant tumor of the urinary system. Despite recent advances in treatments such as local or systemic immunotherapy, chemotherapy, and radiotherapy, the high metastasis and recurrence rates, especially in muscle-invasive bladder cancer (MIBC), have led to the evaluation of more targeted and personalized approaches. A fundamental understanding of the tumorigenesis of bladder cancer along with the development of therapeutics to target processes and pathways implicated in bladder cancer has provided new avenues for the management of this disease. Accumulating evidence supports that the tumor microenvironment (TME) can be shaped by and reciprocally act on tumor cells, which reprograms and regulates tumor development, metastasis, and therapeutic responses. A hostile TME, caused by intrinsic tumor attributes (e.g., hypoxia, oxidative stress, and nutrient deprivation) or external stressors (e.g., chemotherapy and radiation), disrupts the normal synthesis and folding process of proteins in the endoplasmic reticulum (ER), culminating in a harmful situation called ER stress (ERS). ERS is a series of adaptive changes mediated by unfolded protein response (UPR), which is interwoven into a network that can ultimately mediate cell proliferation, apoptosis, and autophagy, thereby endowing tumor cells with more aggressive behaviors. Moreover, recent studies revealed that ERS could also impede the efficacy of anti-cancer treatment including immunotherapy by manipulating the TME. In this review, we discuss the relationship among bladder cancer, ERS, and TME; summarize the current research progress and challenges in overcoming therapeutic resistance; and explore the concept of targeting ERS to improve bladder cancer treatment outcomes.
Collapse
Affiliation(s)
- Zhenyu Nie
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Mei Chen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Xiaohong Wen
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yuanhui Gao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Denggao Huang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Hui Cao
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Yanling Peng
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Na Guo
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| | - Jie Ni
- Cancer Care Center, St. George Hospital, Sydney, NSW, Australia.,St George and Sutherland Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, NSW, Australia
| | - Shufang Zhang
- Central Laboratory, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
| |
Collapse
|
29
|
Candida Administration in Bilateral Nephrectomy Mice Elevates Serum (1→3)-β-D-glucan That Enhances Systemic Inflammation Through Energy Augmentation in Macrophages. Int J Mol Sci 2021; 22:ijms22095031. [PMID: 34068595 PMCID: PMC8126065 DOI: 10.3390/ijms22095031] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/18/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022] Open
Abstract
Systemic inflammation, from gut translocation of organismal molecules, might worsen uremic complications in acute kidney injury (AKI). The monitoring of gut permeability integrity and/or organismal molecules in AKI might be clinically beneficial. Due to the less prominence of Candida albicans in human intestine compared with mouse gut, C. albicans were orally administered in bilateral nephrectomy (BiN) mice. Gut dysbiosis, using microbiome analysis, and gut permeability defect (gut leakage), which was determined by fluorescein isothiocyanate-dextran and intestinal tight-junction immunofluorescent staining, in mice with BiN-Candida was more severe than BiN without Candida. Additionally, profound gut leakage in BiN-Candida also resulted in gut translocation of lipopolysaccharide (LPS) and (1→3)-β-D-glucan (BG), the organismal components from gut contents, that induced more severe systemic inflammation than BiN without Candida. The co-presentation of LPS and BG in mouse serum enhanced inflammatory responses. As such, LPS with Whole Glucan Particle (WGP, a representative BG) induced more severe macrophage responses than LPS alone as determined by supernatant cytokines and gene expression of downstream signals (NFκB, Malt-1 and Syk). Meanwhile, WGP alone did not induced the responses. In parallel, WGP (with or without LPS), but not LPS alone, accelerated macrophage ATP production (extracellular flux analysis) through the upregulation of genes in mitochondria and glycolysis pathway (using RNA sequencing analysis), without the induction of cell activities. These data indicated a WGP pre-conditioning effect on cell energy augmentation. In conclusion, Candida in BiN mice accelerated gut translocation of BG that augmented cell energy status and enhanced pro-inflammatory macrophage responses. Hence, gut fungi and BG were associated with the enhanced systemic inflammation in acute uremia.
Collapse
|
30
|
de Armas LR, George V, Filali-Mouhim A, Steel C, Parmigiani A, Cunningham CK, Weinberg A, Trautmann L, Sekaly RP, Cameron MJ, Pahwa S. Transcriptional and Immunologic Correlates of Response to Pandemic Influenza Vaccine in Aviremic, HIV-Infected Children. Front Immunol 2021; 12:639358. [PMID: 33868267 PMCID: PMC8044856 DOI: 10.3389/fimmu.2021.639358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/09/2021] [Indexed: 11/17/2022] Open
Abstract
People living with HIV (PWH) often exhibit poor responses to influenza vaccination despite effective combination anti-retroviral (ART) mediated viral suppression. There exists a paucity of data in identifying immune correlates of influenza vaccine response in context of HIV infection that would be useful in improving its efficacy in PWH, especially in younger individuals. Transcriptomic data were obtained by microarray from whole blood isolated from aviremic pediatric and adolescent HIV-infected individuals (4-25 yrs) given two doses of Novartis/H1N1 09 vaccine during the pandemic H1N1 influenza outbreak. Supervised clustering and gene set enrichment identified contrasts between individuals exhibiting high and low antibody responses to vaccination. High responders exhibited hemagglutination inhibition antibody titers >1:40 post-first dose and 4-fold increase over baseline. Baseline molecular profiles indicated increased gene expression in metabolic stress pathways in low responders compared to high responders. Inflammation-related and interferon-inducible gene expression pathways were higher in low responders 3 wks post-vaccination. The broad age range and developmental stage of participants in this study prompted additional analysis by age group (e.g. <13yrs and ≥13yrs). This analysis revealed differential enrichment of gene pathways before and after vaccination in the two age groups. Notably, CXCR5, a homing marker expressed on T follicular helper (Tfh) cells, was enriched in high responders (>13yrs) following vaccination which was accompanied by peripheral Tfh expansion. Our results comprise a valuable resource of immune correlates of vaccine response to pandemic influenza in HIV infected children that may be used to identify favorable targets for improved vaccine design in different age groups.
Collapse
Affiliation(s)
- Lesley R de Armas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Varghese George
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | | | - Courtney Steel
- Collaborative Genomics Center, Vaccine and Gene Therapy Institute, Port St. Lucie, FL, United States
| | - Anita Parmigiani
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| | - Coleen K Cunningham
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Adriana Weinberg
- Departments of Medicine, Pathology, and Pediatric Infectious Diseases, University of Colorado School of Medicine, Aurora, CO, United States
| | - Lydie Trautmann
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, OR, United States
| | - Rafick-Pierre Sekaly
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, United States
| | - Mark J Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Savita Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, United States
| |
Collapse
|
31
|
Cruz-Pineda WD, Parra-Rojas I, Rodríguez-Ruíz HA, Illades-Aguiar B, Matia-García I, Garibay-Cerdenares OL. The regulatory role of insulin in energy metabolism and leukocyte functions. J Leukoc Biol 2021; 111:197-208. [PMID: 33724523 PMCID: PMC9291603 DOI: 10.1002/jlb.2ru1220-847r] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/08/2021] [Accepted: 02/10/2021] [Indexed: 12/15/2022] Open
Abstract
Insulin is the hormone responsible for maintaining glucose homeostasis in the body, in addition to participating in lipid metabolism, protein synthesis, and the inhibition of gluconeogenesis. These functions are well characterized in the classic organ target cells that are responsible for general energy regulation: the liver, skeletal muscle, and adipose tissue. However, these actions are not restricted to these tissues because insulin has been shown to affect most cells in the body. This review describes the role of insulin in leukocyte signaling pathways, metabolism and functions, and how insulin resistance could affect this signaling and deteriorate leukocyte metabolism and function, in addition to showing evidence that suggests leukocytes may substantially contribute to the development of systemic insulin resistance.
Collapse
Affiliation(s)
- Walter David Cruz-Pineda
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Isela Parra-Rojas
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Hugo Alberto Rodríguez-Ruíz
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico.,Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Berenice Illades-Aguiar
- Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Inés Matia-García
- Laboratorio de Investigación en Obesidad y Diabetes, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| | - Olga Lilia Garibay-Cerdenares
- CONACyT-Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico.,Laboratorio de Biomedicina Molecular, Facultad de Ciencias Químico-Biológicas, Universidad Autónoma de Guerrero, Chilpancingo, Guerrero, Mexico
| |
Collapse
|
32
|
Zhang W, Liu Z, Xu X. Navigating immune cell immunometabolism after liver transplantation. Crit Rev Oncol Hematol 2021; 160:103227. [PMID: 33675906 DOI: 10.1016/j.critrevonc.2021.103227] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 12/18/2020] [Accepted: 01/16/2021] [Indexed: 11/15/2022] Open
Abstract
Liver transplantation (LT) is the most effective treatment for end-stage liver diseases. The immunometabolism microenvironment undergoes massive changes at the interface of immune functionalities and metabolic regulations after LT. These changes considerably modify post-transplant complications, and immune cells play an influential role in the hepatic immunometabolism microenvironment after LT. Therefore, adequate studies on the complex pathobiology of immune cells are critical to prevent post-transplant complications, and the interplay between cellular metabolism and immune function is evident. Furthermore, immune cells perform their specified functions, such as activation or differentiation, accompanied by alterations in metabolic pathways, such as metabolic reprogramming. This transformation remarkably affects post-transplant complications like rejection. By targeting different metabolic pathways, regulations of metabolism are employed to shape immune responses. These differences of metabolic pathways allow for selective regulation of immune responses to further develop effective therapies that prevent graft loss after LT. This review examines immune cells in the hepatic immunometabolism microenvironment after LT, summarizes possible mechanisms and potential prevention on rejection to acquire immune tolerance, and offers some insight into references for scientific research along with clinical treatment.
Collapse
Affiliation(s)
- Wenhui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University Cancer Center, Hangzhou 310058, China
| | - Zhikun Liu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Xiao Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China; Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China; Zhejiang University Cancer Center, Hangzhou 310058, China.
| |
Collapse
|
33
|
Pillai VB, Gupta MP. Is nuclear sirtuin SIRT6 a master regulator of immune function? Am J Physiol Endocrinol Metab 2021; 320:E399-E414. [PMID: 33308014 PMCID: PMC7988780 DOI: 10.1152/ajpendo.00483.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/03/2020] [Accepted: 12/08/2020] [Indexed: 12/29/2022]
Abstract
The ability to ward off pathogens with minimal damage to the host determines the immune system's robustness. Multiple factors, including pathogen processing, identification, secretion of mediator and effector molecules, and immune cell proliferation and differentiation into various subsets, constitute the success of mounting an effective immune response. Cellular metabolism controls all of these intricate processes. Cells utilize diverse fuel sources and switch back and forth between different metabolic pathways depending on their energy needs. The three most critical metabolic pathways on which immune cells depend to meet their energy needs are oxidative metabolism, glycolysis, and glutaminolysis. Dynamic switching between these metabolic pathways is needed for optimal function of the immune cells. Moreover, switching between these metabolic pathways needs to be tightly regulated to achieve the best results. Immune cells depend on the Warburg effect for their growth, proliferation, secretory, and effector functions. Here, we hypothesize that the sirtuin, SIRT6, could be a negative regulator of the Warburg effect. We also postulate that SIRT6 could act as a master regulator of immune cell metabolism and function by regulating critical signaling pathways.
Collapse
Affiliation(s)
- Vinodkumar B Pillai
- Department of Surgery (Division of Cardiothoracic Surgery), Pritzker School of Medicine, Basic Science Division, University of Chicago, Chicago, Illinois
| | - Mahesh P Gupta
- Department of Surgery (Division of Cardiothoracic Surgery), Pritzker School of Medicine, Basic Science Division, University of Chicago, Chicago, Illinois
| |
Collapse
|
34
|
Peng X, He Y, Huang J, Tao Y, Liu S. Metabolism of Dendritic Cells in Tumor Microenvironment: For Immunotherapy. Front Immunol 2021; 12:613492. [PMID: 33732237 PMCID: PMC7959811 DOI: 10.3389/fimmu.2021.613492] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/25/2021] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) are a type of an antigen-presenting cell which undertake a job on capturing antigens coming from pathogens or tumors and presenting to T cells for immune response. The metabolism of DCs controls its development, polarization, and maturation processes and provides energy support for its functions. However, the immune activity of DCs in tumor microenvironment (TME) is inhibited generally. Abnormal metabolism of tumor cells causes metabolic changes in TME, such as hyperglycolysis, lactate and lipid accumulation, acidification, tryptophan deprivation, which limit the function of DCs and lead to the occurrence of tumor immune escape. Combined metabolic regulation with immunotherapy can strengthen the ability of antigen-presentation and T cell activation of DCs, improve the existing anti-tumor therapy, and overcome the defects of DC-related therapies in the current stage, which has great potential in oncology therapy. Therefore, we reviewed the glucose, lipid, and amino acid metabolism of DCs, as well as the metabolic changes after being affected by TME. Together with the potential metabolic targets of DCs, possible anti-tumor therapeutic pathways were summarized.
Collapse
Affiliation(s)
- Xin Peng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Youe He
- Department of Translational Medicine, Cancer Biological Treatment Center, Xiangya Hospital, Central South University, Changsha, China.,Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Huang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Changsha, China.,Key Laboratory of Carcinogenesis of Ministry of Health, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, China.,Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, China
| | - Shuang Liu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China.,Institute of Medical Sciences, Xiangya Hospital, Central South University, Changsha, China.,Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
35
|
Chen X, Cubillos-Ruiz JR. Endoplasmic reticulum stress signals in the tumour and its microenvironment. Nat Rev Cancer 2021; 21:71-88. [PMID: 33214692 PMCID: PMC7927882 DOI: 10.1038/s41568-020-00312-2] [Citation(s) in RCA: 630] [Impact Index Per Article: 210.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/13/2020] [Indexed: 02/06/2023]
Abstract
Protein handling, modification and folding in the endoplasmic reticulum (ER) are tightly regulated processes that determine cell function, fate and survival. In several tumour types, diverse oncogenic, transcriptional and metabolic abnormalities cooperate to generate hostile microenvironments that disrupt ER homeostasis in malignant and stromal cells, as well as infiltrating leukocytes. These changes provoke a state of persistent ER stress that has been demonstrated to govern multiple pro-tumoural attributes in the cancer cell while dynamically reprogramming the function of innate and adaptive immune cells. Aberrant activation of ER stress sensors and their downstream signalling pathways have therefore emerged as key regulators of tumour growth and metastasis as well as response to chemotherapy, targeted therapies and immunotherapy. In this Review, we discuss the physiological inducers of ER stress in the tumour milieu, the interplay between oncogenic signalling and ER stress response pathways in the cancer cell and the profound immunomodulatory effects of sustained ER stress responses in tumours.
Collapse
Affiliation(s)
- Xi Chen
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, USA.
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, USA.
| | - Juan R Cubillos-Ruiz
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
36
|
Singh Y, Trautwein C, Dhariwal A, Salker MS, Alauddin M, Zizmare L, Pelzl L, Feger M, Admard J, Casadei N, Föller M, Pachauri V, Park DS, Mak TW, Frick JS, Wallwiener D, Brucker SY, Lang F, Riess O. DJ-1 (Park7) affects the gut microbiome, metabolites and the development of innate lymphoid cells (ILCs). Sci Rep 2020; 10:16131. [PMID: 32999308 PMCID: PMC7528091 DOI: 10.1038/s41598-020-72903-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 09/08/2020] [Indexed: 12/21/2022] Open
Abstract
The proper communication between gut and brain is pivotal for the maintenance of health and, dysregulation of the gut-brain axis can lead to several clinical disorders. In Parkinson’s disease (PD) 85% of all patients experienced constipation many years before showing any signs of motor phenotypes. For differential diagnosis and preventive treatment, there is an urgent need for the identification of biomarkers indicating early disease stages long before the disease phenotype manifests. DJ-1 is a chaperone protein involved in the protection against PD and genetic mutations in this protein have been shown to cause familial PD. However, how the deficiency of DJ-1 influences the risk of PD remains incompletely understood. In the present study, we provide evidence that DJ-1 is implicated in shaping the gut microbiome including; their metabolite production, inflammation and innate immune cells (ILCs) development. We revealed that deficiency of DJ-1 leads to a significant increase in two specific genera/species, namely Alistipes and Rikenella. In DJ-1 knock-out (DJ-1-/-) mice the production of fecal calprotectin and MCP-1 inflammatory proteins were elevated. Fecal and serum metabolic profile showed that malonate which influences the immune system was significantly more abundant in DJ-1−/− mice. DJ-1 appeared also to be involved in ILCs development. Further, inflammatory genes related to PD were augmented in the midbrain of DJ-1−/− mice. Our data suggest that metabolites and inflammation produced in the gut could be used as biomarkers for PD detection. Perhaps, these metabolites and inflammatory mediators could be involved in triggering inflammation resulting in PD pathology.
Collapse
Affiliation(s)
- Yogesh Singh
- Institute of Medical Genetics and Applied Genomics, Tübingen University, Calwerstraße 7, 72076, Tübingen, Germany. .,Research Institute of Women's Health, Tübingen University, Calwerstraße 7/6, 72076, Tübingen, Germany.
| | - Christoph Trautwein
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center (WSIC), Tübingen University, Röntgenweg 13, 72076, Tübingen, Germany
| | - Achal Dhariwal
- Department of Oral Biology, University of Oslo, Oslo, Norway
| | - Madhuri S Salker
- Research Institute of Women's Health, Tübingen University, Calwerstraße 7/6, 72076, Tübingen, Germany
| | - Md Alauddin
- Research Institute of Women's Health, Tübingen University, Calwerstraße 7/6, 72076, Tübingen, Germany
| | - Laimdota Zizmare
- Department of Preclinical Imaging and Radiopharmacy, Werner Siemens Imaging Center (WSIC), Tübingen University, Röntgenweg 13, 72076, Tübingen, Germany
| | - Lisann Pelzl
- Department of Vegetative Physiology, Tübingen University, Wilhelmstraße 56, 72076, Tübingen, Germany.,Clinical Transfusion Medicine Centre, Tübingen University, Otfried-Müller-Straße 4/1, 72076, Tübingen, Germany
| | - Martina Feger
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany
| | - Jakob Admard
- Institute of Medical Genetics and Applied Genomics, Tübingen University, Calwerstraße 7, 72076, Tübingen, Germany
| | - Nicolas Casadei
- Institute of Medical Genetics and Applied Genomics, Tübingen University, Calwerstraße 7, 72076, Tübingen, Germany
| | - Michael Föller
- Department of Physiology, University of Hohenheim, Garbenstraße 30, 70599, Stuttgart, Germany
| | - Vivek Pachauri
- Institute of Materials in Electrical Engineering 1, RWTH Aachen University, Aachen, Germany
| | - David S Park
- Health Research Innovation Centre, Hotchkiss Brain Institute, 3330 Hospital Drive NW, Calgary, Alberta, T2N 4N1, Canada
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, UHN, 620 University Ave, Toronto, M5G 2C1, Canada
| | - Julia-Stefanie Frick
- Institute for Medical Microbiology and Hygiene, Tübingen University, Elfriede-Aulhorn-Straße 6, 72076, Tübingen, Germany
| | - Diethelm Wallwiener
- Research Institute of Women's Health, Tübingen University, Calwerstraße 7/6, 72076, Tübingen, Germany
| | - Sara Y Brucker
- Research Institute of Women's Health, Tübingen University, Calwerstraße 7/6, 72076, Tübingen, Germany
| | - Florian Lang
- Department of Vegetative Physiology, Tübingen University, Wilhelmstraße 56, 72076, Tübingen, Germany
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, Tübingen University, Calwerstraße 7, 72076, Tübingen, Germany
| |
Collapse
|
37
|
Molina PE. Thinking through FUNCTION during COVID-19. FUNCTION 2020; 1:zqaa019. [PMID: 35321210 PMCID: PMC7543493 DOI: 10.1093/function/zqaa019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 09/14/2020] [Accepted: 09/24/2020] [Indexed: 01/06/2023] Open
Affiliation(s)
- Patricia E Molina
- Department of Physiology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
38
|
Dang CP, Leelahavanichkul A. Over-expression of miR-223 induces M2 macrophage through glycolysis alteration and attenuates LPS-induced sepsis mouse model, the cell-based therapy in sepsis. PLoS One 2020; 15:e0236038. [PMID: 32658933 PMCID: PMC7357756 DOI: 10.1371/journal.pone.0236038] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/27/2020] [Indexed: 12/31/2022] Open
Abstract
The attenuation of hyper-inflammation in sepsis with the administration of anti-inflammatory macrophages is an interesting adjuvant therapy for sepsis. Because the induction of anti-inflammatory macrophages by microRNA (miR), a regulator of mRNA, has been mentioned, the exploration on miR-induced anti-inflammatory macrophages was performed. The over-expression of miR-223 and miR-146a in RAW264.7 induced M2 macrophage-polarization (anti-inflammatory macrophages) as evaluated by the enhanced expression of Arginase-1 and Fizz. However, miR-223 over-expressed cells demonstrated the more potent anti-inflammatory property against LPS stimulation as lesser iNOS expression, lower supernatant IL-6 and higher supernatant IL-10 compared with miR-146a over-expressed cells. Interestingly, LPS stimulation in miR-223 over-expressed cells, compared with LPS-stimulated control cells, demonstrated lower activity of glycolysis pathway and higher mitochondrial respiration, as evaluated by the extracellular flux analysis, and also down-regulated HIF-1α, an important enzyme of glycolysis pathway. In addition, the administration of miR-223 over-expressed macrophages with IL-4 pre-conditioning, but not IL-4 stimulated control cells, attenuated sepsis severity in LPS injected mice as evaluated by serum creatinine, liver enzymes, lung histology and serum cytokines. In conclusion, miR-223 interfered with the glycolysis pathway through the down-regulation of HIF-1α, resulting in the anti-inflammatory status. The over-expression of miR-223 in macrophages prevented the conversion into M1 macrophage polarization after LPS stimulation. The administration of miR-223 over-expressed macrophages, with IL-4 preconditioning, attenuated sepsis severity in LPS model. Hence, a proof of concept in the induction of anti-inflammatory macrophages through the cell-energy interference for sepsis treatment was proposed as a basis of cell-based therapy in sepsis.
Collapse
Affiliation(s)
- Cong Phi Dang
- Medical Microbiology, Interdisciplinary and International Program, Graduate School, Chulalongkorn University, Bangkok, Thailand
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Asada Leelahavanichkul
- Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Translational Research in Inflammation and Immunology Research Unit (TRIRU), Department of Microbiology, Chulalongkorn University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
39
|
Habel J, Sundrum A. Mismatch of Glucose Allocation between Different Life Functions in the Transition Period of Dairy Cows. Animals (Basel) 2020; 10:E1028. [PMID: 32545739 PMCID: PMC7341265 DOI: 10.3390/ani10061028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 01/04/2023] Open
Abstract
Immune cell functions such as phagocytosis and synthesis of immunometabolites, as well as immune cell survival, proliferation and differentiation, largely depend on an adequate availability of glucose by immune cells. During inflammation, the glucose demands of the immune system may increase to amounts similar to those required for high milk yields. Similar metabolic pathways are involved in the adaptation to both lactation and inflammation, including changes in the somatotropic axis and glucocorticoid response, as well as adipokine and cytokine release. They affect (i) cell growth, proliferation and activation, which determines the metabolic activity and thus the glucose demand of the respective cells; (ii) the overall availability of glucose through intake, mobilization and gluconeogenesis; and (iii) glucose uptake and utilization by different tissues. Metabolic adaptation to inflammation and milk synthesis is interconnected. An increased demand of one life function has an impact on the supply and utilization of glucose by competing life functions, including glucose receptor expression, blood flow and oxidation characteristics. In cows with high genetic merits for milk production, changes in the somatotropic axis affecting carbohydrate and lipid metabolism as well as immune functions are profound. The ability to cut down milk synthesis during periods when whole-body demand exceeds the supply is limited. Excessive mobilization and allocation of glucose to the mammary gland are likely to contribute considerably to peripartal immune dysfunction.
Collapse
Affiliation(s)
- Jonas Habel
- Department of Animal Nutrition and Animal Health, Faculty of Organic Agricultural Sciences, University of Kassel, Nordbahnhofstr. 1a, 37213 Witzenhausen, Germany;
| | | |
Collapse
|
40
|
Abstract
Recent years have witnessed an emergence of interest in understanding metabolic changes associated with immune responses, termed immunometabolism. As oxygen is central to all aerobic metabolism, hypoxia is now recognized to contribute fundamentally to inflammatory and immune responses. Studies from a number of groups have implicated a prominent role for oxygen metabolism and hypoxia in innate immunity of healthy tissue (physiologic hypoxia) and during active inflammation (inflammatory hypoxia). This inflammatory hypoxia emanates from a combination of recruited inflammatory cells (e.g., neutrophils, eosinophils, and monocytes), high rates of oxidative metabolism, and the activation of multiple oxygen-consuming enzymes during inflammation. These localized shifts toward hypoxia have identified a prominent role for the transcription factor hypoxia-inducible factor (HIF) in the regulation of innate immunity. Such studies have provided new and enlightening insight into our basic understanding of immune mechanisms, and extensions of these findings have identified potential therapeutic targets. In this review, we summarize recent literature around the topic of innate immunity and mucosal hypoxia with a focus on transcriptional responses mediated by HIF.
Collapse
Affiliation(s)
- Sean P Colgan
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado 80045, USA;
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Glenn T Furuta
- Mucosal Inflammation Program, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado 80045, USA
| | - Cormac T Taylor
- UCD Conway Institute, Systems Biology Ireland and School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| |
Collapse
|
41
|
Shao G, Zhou C, Ma K, Zhao W, Xiong Q, Yang L, Huang Z, Yang Z. MiRNA-494 enhances M1 macrophage polarization via Nrdp1 in ICH mice model. JOURNAL OF INFLAMMATION-LONDON 2020; 17:17. [PMID: 32351331 PMCID: PMC7183644 DOI: 10.1186/s12950-020-00247-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 04/08/2020] [Indexed: 12/19/2022]
Abstract
Background Ubiquitination-mediated M1/M2 macrophage polarization plays important roles in the pathogenesis of immune disease. However, the regulatory mechanism of ubiquitination during M1/M2 macrophage polarization following intracerebral hemorrhage (ICH) has not been well studied. Methods In the experiment, macrophages were administered with erythrocyte lysates, and then miR-494-, Nrdp1-, and M1/M2-related markers were analyzed. Brain inflammatory response, brain edema, and neurological functions of ICH mice were also assessed. Results We found that miR-494 levels increased while Nrdp1 levels decreased in macrophages after ICH. We also demonstrated that miR-494 inhibited Nrdp1 expression by directly binding its 3′-untranslated region. MiR-494 attenuated C/EBP-β activation and downstream proinflammatory factor production. Upregulation of Nrdp1 in macrophages significantly promoted M2 macrophage polarization via ubiquitinating and activating C/EBP-β. Moreover, the results indicated that miR-494 could enhance M1 macrophage polarization, promote brain edema, and impair neurological functions in ICH mice. Conclusions Taken together, our results demonstrated that Nrdp1 contributed to M1/M2 macrophage polarization and neuroinflammation via ubiquitination and activation of C/EBP-β in ICH. miR-494 may provide a promising therapeutic clue for ICH.
Collapse
Affiliation(s)
- Gaohai Shao
- 1Department of orthopedics, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Changlong Zhou
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Kunlong Ma
- 1Department of orthopedics, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Wang Zhao
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Qijiang Xiong
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Ling Yang
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Zhongyan Huang
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| | - Zhao Yang
- 2Department of Neurology and Chongqing key laboratory of cerebravascular disease, Yongchuan Hospital, Chongqing Medical University, Chongqing, 402160 China
| |
Collapse
|
42
|
Basu A, Kodumudi K. Multimodal approaches to improve immunotherapy in breast cancer. Immunotherapy 2020; 12:161-165. [PMID: 32157934 DOI: 10.2217/imt-2019-0198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Affiliation(s)
- Amrita Basu
- Immunology Program & Department of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Krithika Kodumudi
- Immunology Program & Department of Clinical Science, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA.,Department of Oncological Sciences, USF Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
43
|
Kim SW, Goossens A, Libert C, Van Immerseel F, Staal J, Beyaert R. Phytohormones: Multifunctional nutraceuticals against metabolic syndrome and comorbid diseases. Biochem Pharmacol 2020; 175:113866. [PMID: 32088261 DOI: 10.1016/j.bcp.2020.113866] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 02/18/2020] [Indexed: 12/12/2022]
Abstract
Metabolic syndrome is characterized by the co-occurrence of diverse symptoms initiating the development of type 2 diabetes, cardiovascular diseases, and a variety of comorbid diseases. The complex constellation of numerous comorbidities makes it difficult to develop common therapeutic approaches that ameliorate these pathological features simultaneously. The plant hormones abscisic acid, salicylic acid, auxin, and cytokinins, have shown promising anti-inflammatory and pro-metabolic effects that could mitigate several disorders relevant to metabolic syndrome. Intriguingly, besides plants, human cells and gut microbes also endogenously produce these molecules, indicating a role in the complex interplay between inflammatory responses associated with metabolic syndrome, the gut microbiome, and nutrition. Here, we introduce how bioactive phytohormones can be generated endogenously and through the gut microbiome. These molecules subsequently influence immune responses and metabolism. We also elaborate on how phytohormones can beneficially modulate metabolic syndrome comorbidities, and propose them as nutraceuticals.
Collapse
Affiliation(s)
- Seo Woo Kim
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium; VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Alain Goossens
- VIB-UGent Center for Plant Systems Biology, VIB, Ghent, Belgium; Department of Plant Biotechnology and Bioinformatics, Ghent University, Ghent, Belgium
| | - Claude Libert
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Jens Staal
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium.
| |
Collapse
|
44
|
van de Wetering C, Aboushousha R, Manuel AM, Chia SB, Erickson C, MacPherson MB, van der Velden JL, Anathy V, Dixon AE, Irvin CG, Poynter ME, van der Vliet A, Wouters EFM, Reynaert NL, Janssen-Heininger YMW. Pyruvate Kinase M2 Promotes Expression of Proinflammatory Mediators in House Dust Mite-Induced Allergic Airways Disease. THE JOURNAL OF IMMUNOLOGY 2020; 204:763-774. [PMID: 31924651 DOI: 10.4049/jimmunol.1901086] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022]
Abstract
Asthma is a chronic disorder characterized by inflammation, mucus metaplasia, airway remodeling, and hyperresponsiveness. We recently showed that IL-1-induced glycolytic reprogramming contributes to allergic airway disease using a murine house dust mite model. Moreover, levels of pyruvate kinase M2 (PKM2) were increased in this model as well as in nasal epithelial cells from asthmatics as compared with healthy controls. Although the tetramer form of PKM2 converts phosphoenolpyruvate to pyruvate, the dimeric form of PKM2 has alternative, nonglycolysis functions as a transcriptional coactivator to enhance the transcription of several proinflammatory cytokines. In the current study, we examined the impact of PKM2 on the pathogenesis of house dust mite-induced allergic airways disease in C57BL/6NJ mice. We report, in this study, that activation of PKM2, using the small molecule activator, TEPP46, augmented PKM activity in lung tissues and attenuated airway eosinophils, mucus metaplasia, and subepithelial collagen. TEPP46 attenuated IL-1β-mediated airway inflammation and expression of proinflammatory mediators. Exposure to TEPP46 strongly decreased the IL-1β-mediated increases in thymic stromal lymphopoietin (TSLP) and GM-CSF in primary tracheal epithelial cells isolated from C57BL/6NJ mice. We also demonstrate that IL-1β-mediated increases in nuclear phospho-STAT3 were decreased by TEPP46. Finally, STAT3 inhibition attenuated the IL-1β-induced release of TSLP and GM-CSF, suggesting that the ability of PKM2 to phosphorylate STAT3 contributes to its proinflammatory function. Collectively, these results demonstrate that the glycolysis-inactive form of PKM2 plays a crucial role in the pathogenesis of allergic airways disease by increasing IL-1β-induced proinflammatory signaling, in part, through phosphorylation of STAT3.
Collapse
Affiliation(s)
- Cheryl van de Wetering
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405.,Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, the Netherlands; and
| | - Reem Aboushousha
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Allison M Manuel
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Shi B Chia
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Cuixia Erickson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Maximilian B MacPherson
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Jos L van der Velden
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Anne E Dixon
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Charles G Irvin
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Matthew E Poynter
- Department of Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405
| | - Emiel F M Wouters
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, the Netherlands; and
| | - Niki L Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, the Netherlands; and
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT 05405;
| |
Collapse
|
45
|
Hu C, Pang B, Lin G, Zhen Y, Yi H. Energy metabolism manipulates the fate and function of tumour myeloid-derived suppressor cells. Br J Cancer 2020; 122:23-29. [PMID: 31819182 PMCID: PMC6964679 DOI: 10.1038/s41416-019-0644-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 10/23/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
In recent years, a large number of studies have been carried out in the field of immune metabolism, highlighting the role of metabolic energy reprogramming in altering the function of immune cells. Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells generated during a large array of pathological conditions, such as cancer, inflammation, and infection, and show remarkable ability to suppress T-cell responses. These cells can also change their metabolic pathways in response to various pathogen-derived or inflammatory signals. In this review, we focus on the roles of glucose, fatty acid (FA), and amino acid (AA) metabolism in the differentiation and function of MDSCs in the tumour microenvironment, highlighting their potential as targets to inhibit tumour growth and enhance tumour immune surveillance by the host. We further highlight the remaining gaps in knowledge concerning the mechanisms determining the plasticity of MDSCs in different environments and their specific responses in the tumour environment. Therefore, this review should motivate further research in the field of metabolomics to identify the metabolic pathways driving the enhancement of MDSCs in order to effectively target their ability to promote tumour development and progression.
Collapse
Affiliation(s)
- Cong Hu
- Central Laboratory, The First Hospital of Jilin University, 130031, Changchun, Jilin, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, 130021, Changchun, Jilin, China
- Center for Reproductive Medicine, Center for Prenatal Diagnosis, The First Hospital of Jilin University, 130021, Changchun, Jilin, China
| | - Bo Pang
- Central Laboratory, The First Hospital of Jilin University, 130031, Changchun, Jilin, China
- Department of Cardiology, The First Hospital of Jilin University, 130031, Changchun, Jilin, China
| | - Guangzhu Lin
- Department of Cardiology, The First Hospital of Jilin University, 130031, Changchun, Jilin, China
| | - Yu Zhen
- Department of Dermatology, The First Hospital of Jilin University, 130021, Changchun, Jilin, China
| | - Huanfa Yi
- Central Laboratory, The First Hospital of Jilin University, 130031, Changchun, Jilin, China.
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, 130021, Changchun, Jilin, China.
| |
Collapse
|
46
|
Gao F, Tang Y, Liu WL, Zou MZ, Huang C, Liu CJ, Zhang XZ. Intra/Extracellular Lactic Acid Exhaustion for Synergistic Metabolic Therapy and Immunotherapy of Tumors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1904639. [PMID: 31692128 DOI: 10.1002/adma.201904639] [Citation(s) in RCA: 207] [Impact Index Per Article: 41.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/09/2019] [Indexed: 05/18/2023]
Abstract
Regulating the tumor microenvironment (TME) has been a promising strategy to improve antitumor therapy. Here, a red blood cell membrane (mRBC)-camouflaged hollow MnO2 (HMnO2 ) catalytic nanosystem embedded with lactate oxidase (LOX) and a glycolysis inhibitor (denoted as PMLR) is constructed for intra/extracellular lactic acid exhaustion as well as synergistic metabolic therapy and immunotherapy of tumor. Benefiting from the long-circulation property of the mRBC, the nanosystem can gradually accumulate in a tumor site through the enhanced permeability and retention (EPR) effect. The extracellular nanosystem consumes lactic acid in the TME by catalyzing its oxidation reaction via LOX. Meanwhile, the intracellular nanosystem releases the glycolysis inhibitor to cut off the source of lactic acid, as well as achieve antitumor metabolic therapy through the blockade of the adenosine triphosphate (ATP) supply. Both the extracellular and intracellular processes can be sensitized by O2 , which can be produced during the decomposition of endogenous H2 O2 catalyzed by the PMLR nanosystem. The results show that the PMLR nanosystem can ceaselessly remove lactic acid, and then lead to an immunocompetent TME. Moreover, this TME regulation strategy can effectively improve the antitumor effect of anti-PDL1 therapy without the employment of any immune agonists to avoid the autoimmunity.
Collapse
Affiliation(s)
- Fan Gao
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Ying Tang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Wen-Long Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Mei-Zhen Zou
- The Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
| | - Cui Huang
- State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory of Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, 430079, P. R. China
| | - Chuan-Jun Liu
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
- The Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
47
|
Domínguez-Amorocho O, Takiishi T, da Cunha FF, Camara NOS. Immunometabolism: A target for the comprehension of immune response toward transplantation. World J Transplant 2019; 9:27-34. [PMID: 31363459 PMCID: PMC6656658 DOI: 10.5500/wjt.v9.i2.27] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/11/2019] [Accepted: 01/28/2019] [Indexed: 02/05/2023] Open
Abstract
Organ transplantation is a life-saving procedure, however predicting graft survival is still challenging. Understanding immune-cell pathobiology is critical to the development of effective therapies to prevent rejection. Over the recent years it has become progressively evident that the complex nature of immune cell behavioral dynamics is strongly dependent on cellular metabolism, which in turn, relies on competition for nutrients, oxygen and metabolites with other immune cells and microbiota. Furthermore, the influence of the inflammatory state can lead to substantial changes in conditions within the tissue micro-environment. Considering the context of immunity, alterations in metabolic pathways (glycolysis, the tricarboxylic acid cycle, the pentose phosphate pathway, the fatty acid oxidation and synthesis, and the amino acid metabolic pathways) will influence the production of different sets of cytokines and affect transplantation outcome. It is now known that naïve, resting and effector cells acquire different metabolic profiles and studies have shown that specifically targeting some of these metabolic routes can prevent differentiation of effector T cells in favor of Tregs. Ultimately, to develop effective therapies that will prevent graft loss and understanding how cell metabolism impacts the fate and function of immune cells is now a critical point of discussion. The distinct metabolic features and requirements observed in effector and suppressive cell subsets offer promising opportunities for selective regulation of the immune responses in transplantation and will be discussed in this review.
Collapse
Affiliation(s)
- Omar Domínguez-Amorocho
- Department of Immunology, Biomedical Sciences Institute, University of São Paulo, São Paulo 05508-900, Brazil
| | - Tatiana Takiishi
- Department of Immunology, Biomedical Sciences Institute, University of São Paulo, São Paulo 05508-900, Brazil
| | - Flavia Franco da Cunha
- Department of Immunology, Biomedical Sciences Institute, University of São Paulo, São Paulo 05508-900, Brazil
| | - Niels Olsen Saraiva Camara
- Department of Immunology, Biomedical Sciences Institute, University of São Paulo, São Paulo 05508-900, Brazil
| |
Collapse
|
48
|
Domínguez-Amorocho O, Takiishi T, Cunha FFD, Câmara NOS. Immunometabolism: A target for the comprehension of immune response toward transplantation. World J Transplant 2019. [DOI: 10.5500/wjt.v9.i2.0000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
49
|
Mora J, Mertens C, Meier JK, Fuhrmann DC, Brüne B, Jung M. Strategies to Interfere with Tumor Metabolism through the Interplay of Innate and Adaptive Immunity. Cells 2019; 8:cells8050445. [PMID: 31083487 PMCID: PMC6563030 DOI: 10.3390/cells8050445] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/04/2019] [Accepted: 05/07/2019] [Indexed: 01/19/2023] Open
Abstract
The inflammatory tumor microenvironment is an important regulator of carcinogenesis. Tumor-infiltrating immune cells promote each step of tumor development, exerting crucial functions from initiation, early neovascularization, to metastasis. During tumor outgrowth, tumor-associated immune cells, including myeloid cells and lymphocytes, acquire a tumor-supportive, anti-inflammatory phenotype due to their interaction with tumor cells. Microenvironmental cues such as inflammation and hypoxia are mainly responsible for creating a tumor-supportive niche. Moreover, it is becoming apparent that the availability of iron within the tumor not only affects tumor growth and survival, but also the polarization of infiltrating immune cells. The interaction of tumor cells and infiltrating immune cells is multifaceted and complex, finally leading to different activation phenotypes of infiltrating immune cells regarding their functional heterogeneity and plasticity. In recent years, it was discovered that these phenotypes are mainly implicated in defining tumor outcome. Here, we discuss the role of the metabolic activation of both tumor cells and infiltrating immune cells in order to adapt their metabolism during tumor growth. Additionally, we address the role of iron availability and the hypoxic conditioning of the tumor with regard to tumor growth and we describe the relevance of therapeutic strategies to target such metabolic characteristics.
Collapse
Affiliation(s)
- Javier Mora
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Christina Mertens
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Julia K Meier
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Dominik C Fuhrmann
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
- German Cancer Consortium (DKTK), Partner Site Frankfurt, Germany.
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular Biology and Applied Ecology, 60596 Frankfurt, Germany.
| | - Michaela Jung
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Theodor-Stern-Kai 7, 60590 Frankfurt, Germany.
| |
Collapse
|
50
|
Abstract
Since the pioneering discovery of heat shock proteins in Drosophila by Ferruccio Ritossa in 1960s, a long and exciting journey has been undertaken by molecular biologists and researchers worldwide. Not only lower organisms like worms, yeast, amoeba, and flies but also eukaryotes share common cellular response signals to stressful conditions that can arise from the outside but also from the inside. Moreover, extraordinary interplay between nucleus and subcellular organelles, and between different organelles, like mitochondria and the endoplasmic reticulum called mitochondria-associated endoplasmic reticulum membranes (MAMs), are involved in aging and human diseases like obesity, diabetes, inflammation, neurodegeneration, autoimmune diseases, atherosclerosis, and cancer. Actually, we know that to hit abnormal proteostasis and lipid exchanges in the endoplasmic reticulum is crucial to best guide effective therapies or discover new drugs. Indeed, restoration or impairment of endoplasmic reticulum shape and function lead to cellular homeostasis by autophagy or to final death generally by apoptosis or pyroptosis. This Special Issue collects current valuable articles or reviews on cellular stress research and each contribution opens a new window for further studies and hypothesis. I hope that readers interested in this fascinating topic may be stimulated to know more and more.
Collapse
|