1
|
Gao L, Ding H, Shi Q, Wang X, Zhang L, Yang L. Pregnancy-induced changes in the toll-like receptor pathway in the ovine duodenum. Vet Immunol Immunopathol 2025; 282:110900. [PMID: 39985901 DOI: 10.1016/j.vetimm.2025.110900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/01/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
Toll-like receptor (TLR) signaling plays a key role in the intestinal innate and adaptive immune responses, and pregnancy has effects on intestinal length and villus height. Nevertheless, it is unclear if early pregnancy modulates the expression of TLR members in the duodenum of ewes. In this study, ovine duodenums were obtained at day 16 of the estrous cycle (N16), and days 13, 16, and 25 of gestation (P13, P16, and P25) from ewes, and mRNA and protein expression of TLR members were analyzed. The results showed that early pregnancy increased the expression of TLR3, TLR5, and TRAF6, but did not influence TLR4 expression. In addition, there were increases in expression of TLR2, MyD88, and IRAK1 at P13 and P16, or only at P16 compared to N16 and P25. In summary, these data suggest that early pregnancy modulates the TLR signal pathway in the maternal duodenum, which may be related to the immunoregulation and adaptation of the maternal duodenum in the ovine.
Collapse
Affiliation(s)
- Linna Gao
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Haiquan Ding
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Qianqian Shi
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Xinxin Wang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Leying Zhang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China.
| | - Ling Yang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China.
| |
Collapse
|
2
|
Yu G, Song M, Wu C, Ma X, Zhang L, Yang L. Modulation of the Toll-like Receptor Pathway in Ovine Endometria During Early Pregnancy. Animals (Basel) 2025; 15:917. [PMID: 40218311 PMCID: PMC11987747 DOI: 10.3390/ani15070917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 02/14/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Correct immunological dialogue between the maternal uterus and conceptus is essential during implantation, and Toll-like receptors (TLRs) participate in maternal immune tolerance during pregnancy. This study aimed to analyze the effect of early pregnancy on Toll-like receptor pathways in the ovine endometrium. Ovine endometria were obtained on day 16 of the estrous cycle (N16) and days 13, 16, and 25 of pregnancy (P13, P16, and P25), and expression profiles of TLR members, including TLRs, tumor necrosis factor receptor-associated factor 6 (TRAF6), interleukin 1 receptor-associated kinase 1 (IRAK1), and myeloid differentiation primary response gene 88 (MyD88), were detected by quantitative real-time PCR, Western blot analysis, and immunohistochemistry analysis. The data of this study showed that the expression of TLR2 and TLR5 was gradually increased during early pregnancy compared to N16, and TLR3 expression was greater at P16 and P25 than at N16 and P13. However, the expression levels of TLR4 and TRAF6 were weaker at P13 and P16, and the expression of MyD88 was inhibited by early pregnancy. Furthermore, early pregnancy regulated IRAK1 expression. These findings corroborated that the TLR pathway was modulated in the ovine endometrium during early pregnancy, which may be involved in maternal immunoregulation.
Collapse
Affiliation(s)
| | | | | | | | | | - Ling Yang
- School of of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China; (G.Y.); (M.S.); (C.W.); (X.M.); (L.Z.)
| |
Collapse
|
3
|
Gray OA, Witonsky DB, Jousma J, Sobreira DR, Van Alstyne A, Huang RT, Fang Y, Di Rienzo A. Transcriptomic analysis of iPSC-derived endothelium reveals adaptations to high altitude hypoxia in energy metabolism and inflammation. PLoS Genet 2025; 21:e1011570. [PMID: 39928692 PMCID: PMC11809796 DOI: 10.1371/journal.pgen.1011570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 01/10/2025] [Indexed: 02/12/2025] Open
Abstract
Tibetan adaptation to high-altitude hypoxia remains a classic example of Darwinian selection in humans. Amongst Tibetan populations, alleles in the EPAS1 gene - whose protein product, HIF-2α, is a central regulator of the hypoxia response - have repeatedly been shown to carry some of the strongest signals of positive selection in humans. However, selective sweep signals alone may only account for some of the phenotypes that differentiate high-altitude adapted populations from closely related lowlanders. Therefore, there is a pressing need to functionally probe adaptive alleles and their impact at both the locus-specific and genome-wide levels and across cell types to uncover the full range of beneficial traits. To this end, we established a library of induced pluripotent stem cells (iPSCs) derived from Tibetan and Han Chinese individuals, a robust model system allowing precise exploration of allelic effects on transcriptional responses, and we differentiated them into vascular endothelium. Using this system, we focus first on a hypoxia-dependent enhancer (ENH5) that contributes to the regulation of EPAS1 to investigate its locus-specific effects in endothelium. Then, to cast a wider net, we harness the same experimental system to compare the transcriptome of Tibetan and Han Chinese cells in hypoxia and find evidence that angiogenesis, energy metabolism and immune pathways differ between these two populations with different histories of long-term residence at high altitude. Coupled with evidence of polygenic adaptations targeting the same pathways, these results suggests that the observed transcriptional differences between the two populations were shaped by natural selection.
Collapse
Affiliation(s)
- Olivia A. Gray
- Department of Human Genetics, University of Chicago Division of the Biological Sciences, Chicago, Illinois, United States of America
| | - David B. Witonsky
- Department of Human Genetics, University of Chicago Division of the Biological Sciences, Chicago, Illinois, United States of America
| | - Jordan Jousma
- Department of Human Genetics, University of Chicago Division of the Biological Sciences, Chicago, Illinois, United States of America
| | - Débora R. Sobreira
- Department of Human Genetics, University of Chicago Division of the Biological Sciences, Chicago, Illinois, United States of America
| | - Alexander Van Alstyne
- Department of Human Genetics, University of Chicago Division of the Biological Sciences, Chicago, Illinois, United States of America
| | - Ru-Ting Huang
- Department of Medicine, Section of Pulmonary and Intensive Care, University of Chicago Hospital: The University of Chicago Medicine, Chicago, Illinois, United States of America
| | - Yun Fang
- Department of Medicine, Section of Pulmonary and Intensive Care, University of Chicago Hospital: The University of Chicago Medicine, Chicago, Illinois, United States of America
| | - Anna Di Rienzo
- Department of Human Genetics, University of Chicago Division of the Biological Sciences, Chicago, Illinois, United States of America
| |
Collapse
|
4
|
Chucair-Elliott AJ, Pham K, Cleuren ACA, Schafer CM, Griffin CT, Ocanas SR, Freeman WM, Elliott MH. Comparative analysis of In vivo endothelial cell translatomes across central nervous system vascular beds. Exp Eye Res 2024; 248:110101. [PMID: 39303842 PMCID: PMC11532013 DOI: 10.1016/j.exer.2024.110101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/06/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Endothelial cells (ECs) display organ- and tissue-specific heterogeneity. In the eye, the retinal and choroidal vascular beds are distinct networks with different molecular and morphological properties that serve location-specific functions, i.e., the former maintaining a tight barrier and the latter, a permeable fenestrated vasculature. Given that retinal health critically relies on the function of these vascular beds and that their dysfunction is implicated in a variety of retinal diseases, a molecular understanding of both physiological and pathophysiological characteristics of these distinct vasculatures is critical. Given their interspersed anatomic distribution among parenchymal cells, the study of EC gene expression, in vivo, has been hampered by the challenge of isolating pure populations of ocular ECs in sufficient quantities for large-scale transcriptomics. To address this challenge, we present a methodological and analytical workflow to facilitate inter-tissue comparisons of the in vivo EC translatome isolated from choroid, retina, and brain using the Cre-inducible NuTRAP flox construct and two widely-used endothelial Cre mouse lines: constitutive Tie2-Cre and tamoxifen-inducible Cdh5-CreERT2. For each Cre line, inter-tissue comparison of TRAP-RNAseq enrichment (TRAP-isolated translatome vs input transcriptome) showed tissue-specific gene enrichments with differential pathway representation. For each mouse model, inter-tissue comparison of the EC translatome (choroid vs brain, choroid vs retina, and brain vs retina) showed over 50% overlap of differentially expressed genes (DEGs) between the three paired comparisons, with differential pathway representation for each tissue. Pathway analysis of DEGs in the Cdh5-NuTRAP vs Tie2-NuTRAP comparison for retina, choroid, and brain predicted inhibition of processes related to myeloid cell function and activation, consistent with more specific targeting of ECs in the Cdh5-NuTRAP than in the Tie2-NuTRAP model which also targets hematopoietic progenitors giving rise to immune cells. Indeed, while TRAP enriches for EC transcripts in both models, myeloid transcripts were also captured in the Tie2-NuTRAP model which was confirmed using cell sorting. We suggest experimental/analytical considerations should be taken when selecting Cre-lines to target ECs.
Collapse
Affiliation(s)
- Ana J Chucair-Elliott
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Kevin Pham
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Audrey C A Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Christopher M Schafer
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Sarah R Ocanas
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Willard M Freeman
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| | - Michael H Elliott
- Department of Ophthalmology, McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
5
|
Matuszewska J, Krawiec A, Radziemski A, Uruski P, Tykarski A, Mikuła-Pietrasik J, Książek K. Alterations of receptors and insulin-like growth factor binding proteins in senescent cells. Eur J Cell Biol 2024; 103:151438. [PMID: 38945074 DOI: 10.1016/j.ejcb.2024.151438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 06/24/2024] [Accepted: 06/25/2024] [Indexed: 07/02/2024] Open
Abstract
The knowledge about cellular senescence expands dynamically, providing more and more conclusive evidence of its triggers, mechanisms, and consequences. Senescence-associated secretory phenotype (SASP), one of the most important functional traits of senescent cells, is responsible for a large extent of their context-dependent activity. Both SASP's components and signaling pathways are well-defined. A literature review shows, however, that a relatively underinvestigated aspect of senescent cell autocrine and paracrine activity is the change in the production of proteins responsible for the reception and transmission of SASP signals, i.e., receptors and binding proteins. For this reason, we present in this article the current state of knowledge regarding senescence-associated changes in cellular receptors and insulin-like growth factor binding proteins. We also discuss the role of these alterations in senescence induction and maintenance, pro-cancerogenic effects of senescent cells, and aging-related structural and functional malfunctions.
Collapse
Affiliation(s)
- Julia Matuszewska
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Adrianna Krawiec
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Artur Radziemski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Paweł Uruski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Andrzej Tykarski
- Poznan University of Medical Sciences, Department of Hypertensiology, Długa 1/2 Str., Poznań 61-848, Poland
| | - Justyna Mikuła-Pietrasik
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland
| | - Krzysztof Książek
- Poznan University of Medical Sciences, Department of Pathophysiology of Ageing and Civilization Diseases, Święcickiego 4 Str., Poznań 60-781, Poland.
| |
Collapse
|
6
|
Jiang D, Yue H, Liang WT, Wu Z. Developmental endothelial locus 1: the present and future of an endogenous factor in vessels. Front Physiol 2024; 15:1347888. [PMID: 39206385 PMCID: PMC11350114 DOI: 10.3389/fphys.2024.1347888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Developmental Endothelial Locus-1 (DEL-1), also known as EGF-like repeat and discoidin I-like domain-3 (EDIL3), is increasingly recognized for its multifaceted roles in immunoregulation and vascular biology. DEL-1 is a protein that is mainly produced by endothelial cells. It interacts with various integrins to regulate the behavior of immune cells, such as preventing unnecessary recruitment and inflammation. DEL-1 also helps in resolving inflammation by promoting efferocytosis, which is the process of clearing apoptotic cells. Its potential as a therapeutic target in immune-mediated blood disorders, cardiovascular diseases, and cancer metastasis has been spotlighted due to its wide-ranging implications in vascular integrity and pathology. However, there are still unanswered questions about DEL-1's precise functions and mechanisms. This review provides a comprehensive examination of DEL-1's activity across different vascular contexts and explores its potential clinical applications. It underscores the need for further research to resolve existing controversies and establish the therapeutic viability of DEL-1 modulation.
Collapse
Affiliation(s)
| | | | - Wei-Tao Liang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhong Wu
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Li X, Ding X, He Y, Yi W, Zhu Y, Han W, Liao B, Han X, Bai D. Ultrasound Tissue Engineering Technology for Regulating Immune Microenvironment. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202400656] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Indexed: 01/06/2025]
Abstract
AbstractThe immune microenvironment is critical for the occurrence, progression, and treatment of diseases. Ultrasound tissue engineering technology utilizes ultrasound and the principles of tissue engineering to repair, regenerate, and functionally reconstruct biological tissues. Ultrasound therapy is a non‐invasive treatment modality that regulates the immune microenvironment and maintains homeostasis through various characteristic effects. Ultrasound‐responsive biomaterials utilize biological properties or drug/gene delivery to regulate the immune microenvironment under ultrasound stimulation for targeted and purposeful treatment. This article comprehensively and systematically reviews advancements in ultrasound tissue engineering technology for regulating the immune microenvironment. First, the changes in the immune microenvironment at different stages of the disease is briefly illustrated. It is then reviewed the regulation of the immune microenvironment by ultrasound and ultrasound‐responsive biomaterials in five types of diseases: tumor, cardiovascular system diseases, nervous system diseases, musculoskeletal diseases, and wound. Finally, the prospects of the ultrasound tissue engineering technology for regulating the immune microenvironment is summarized.
Collapse
Affiliation(s)
- Xinhe Li
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Xiaoqian Ding
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Yi He
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Weiwei Yi
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Ying Zhu
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Wang Han
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Bo Liao
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Xiaoyu Han
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Dingqun Bai
- Department of Rehabilitation Medicine The First Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
- State Key Laboratory of Ultrasound in Medicine and Engineering Chongqing Medical University Chongqing 400016 P. R. China
| |
Collapse
|
8
|
Liu WS, Chen Z, Lu ZM, Dong JH, Wu JH, Gao J, Deng D, Li M. Multifunctional hydrogels based on photothermal therapy: A prospective platform for the postoperative management of melanoma. J Control Release 2024; 371:406-428. [PMID: 38849093 DOI: 10.1016/j.jconrel.2024.06.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/22/2024] [Accepted: 06/01/2024] [Indexed: 06/09/2024]
Abstract
Preventing the recurrence of melanoma after surgery and accelerating wound healing are among the most challenging aspects of melanoma management. Photothermal therapy has been widely used to treat tumors and bacterial infections and promote wound healing. Owing to its efficacy and specificity, it may be used for postoperative management of tumors. However, its use is limited by the uncontrollable distribution of photosensitizers and the likelihood of damage to the surrounding normal tissue. Hydrogels provide a moist environment with strong biocompatibility and adhesion for wound healing owing to their highly hydrophilic three-dimensional network structure. In addition, these materials serve as excellent drug carriers for tumor treatment and wound healing. It is possible to combine the advantages of both of these agents through different loading modalities to provide a powerful platform for the prevention of tumor recurrence and wound healing. This review summarizes the design strategies, research progress and mechanism of action of hydrogels used in photothermal therapy and discusses their role in preventing tumor recurrence and accelerating wound healing. These findings provide valuable insights into the postoperative management of melanoma and may guide the development of promising multifunctional hydrogels for photothermal therapy.
Collapse
Affiliation(s)
- Wen-Shang Liu
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, People's Republic of China
| | - Zhuo Chen
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, People's Republic of China
| | - Zheng-Mao Lu
- Department of Gastrointestinal Surgery, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, People's Republic of China
| | - Jin-Hua Dong
- Women and Children Hospital Affiliated to Jiaxing University, 2468 Middle Ring Eastern Road, Jiaxing City, Zhejiang 314000, People's Republic of China
| | - Jin-Hui Wu
- Ophthalmology Department of the Third Affiliated Hospital of Naval Medical University, Shanghai 201805, People's Republic of China.
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai 200433, People's Republic of China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai 200433, People's Republic of China.
| | - Dan Deng
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, People's Republic of China.
| | - Meng Li
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai 200127, People's Republic of China.
| |
Collapse
|
9
|
Xiong L, Zhevlakova I, West XZ, Gao D, Murtazina R, Horak A, Brown JM, Molokotina I, Podrez EA, Byzova TV. TLR2 regulates hair follicle cycle and regeneration via BMP signaling. eLife 2024; 12:RP89335. [PMID: 38483447 PMCID: PMC10939499 DOI: 10.7554/elife.89335] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2024] Open
Abstract
The etiology of hair loss remains enigmatic, and current remedies remain inadequate. Transcriptome analysis of aging hair follicles uncovered changes in immune pathways, including Toll-like receptors (TLRs). Our findings demonstrate that the maintenance of hair follicle homeostasis and the regeneration capacity after damage depend on TLR2 in hair follicle stem cells (HFSCs). In healthy hair follicles, TLR2 is expressed in a cycle-dependent manner and governs HFSCs activation by countering inhibitory BMP signaling. Hair follicles in aging and obesity exhibit a decrease in both TLR2 and its endogenous ligand carboxyethylpyrrole (CEP), a metabolite of polyunsaturated fatty acids. Administration of CEP stimulates hair regeneration through a TLR2-dependent mechanism. These results establish a novel connection between TLR2-mediated innate immunity and HFSC activation, which is pivotal to hair follicle health and the prevention of hair loss and provide new avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Luyang Xiong
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Irina Zhevlakova
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Xiaoxia Z West
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Rakhilya Murtazina
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Anthony Horak
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - J Mark Brown
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Iuliia Molokotina
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Eugene A Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland ClinicClevelandUnited States
| |
Collapse
|
10
|
Shi X, Yu J, Lu C, Luo Q, Xu C, Li J, Wang W. Screening of the shared pathogenic genes of ulcerative colitis and colorectal cancer by integrated bioinformatics analysis. J Cell Mol Med 2024; 28:e17878. [PMID: 37494129 PMCID: PMC10902564 DOI: 10.1111/jcmm.17878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 07/08/2023] [Accepted: 07/19/2023] [Indexed: 07/28/2023] Open
Abstract
Ulcerative colitis (UC) is one of the high-risk pathogenic factors for colorectal cancer (CRC). However, the shared gene and signalling mechanisms between UC and CRC remain unclear. The goal of this study was to delve more into the probable causal relationship between UC and CRC. CRC and UC datasets were downloaded from the Gene Expression Omnibus database. Using R software and Perl, differentially expressed genes (DEGs) in both UC and CRC tissues were re-annotated and screened. The biological activities and signalling pathways involved in DEGs were investigated using Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment analyses. The STRING database and Cytoscape software were used to construct the gene interaction network. A total of 384 DEGs were selected for further investigation, and functional analysis revealed that inflammatory and immunological responses were crucial in the development of the two diseases. Moreover, the top 15 key genes involved in the UC and CRC were screened using cytoHubba, including IL1B, CXCL10, CCL20, MMP9, ICAM1, CCL4, CXCR1, MMP3, TLR2, PTGS2, IL1RN, IL6, COL1A2, TIMP1 and CXCL1. The identification of these genes in the present study may provide a novel perspective for the prediction, prevention and personalized medicine of UC and CRC patients.
Collapse
Affiliation(s)
- Xu Shi
- Department of OrthopaedicsThe Affiliated People's Hospital of Jiangsu UniversityZhenjiangChina
| | - Jun Yu
- Department of PaediatricsAffiliated Hospital of Nanjing University of Chinese Medicine, Taicang Hospital of Traditional Chinese MedicineTaicangChina
| | - Chen Lu
- Department of General SurgerySiyang HospitalSuqianChina
| | - Qian Luo
- Department of General SurgerySiyang HospitalSuqianChina
| | - Caihong Xu
- Department of Obstetrics and GynaecologyNanjing Tongren Hospital, School of Medicine, Southeast UniversityNanjingChina
| | - Jie Li
- Department of General SurgerySiyang HospitalSuqianChina
| | - Wei Wang
- Department of Clinical LaboratoryLianshui County People's HospitalHuai'anChina
| |
Collapse
|
11
|
Frias MA, Pagano S, Bararpour N, Sidibé J, Kamau F, Fétaud-Lapierre V, Hudson P, Thomas A, Lecour S, Strijdom H, Vuilleumier N. People living with HIV display increased anti-apolipoprotein A1 auto-antibodies, inflammation, and kynurenine metabolites: a case-control study. Front Cardiovasc Med 2024; 11:1343361. [PMID: 38414919 PMCID: PMC10896987 DOI: 10.3389/fcvm.2024.1343361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 01/25/2024] [Indexed: 02/29/2024] Open
Abstract
Objective This study aimed to study the relationship between auto-antibodies against apolipoprotein A1 (anti-apoA1 IgG), human immunodeficiency virus (HIV) infection, anti-retroviral therapy (ART), and the tryptophan pathways in HIV-related cardiovascular disease. Design This case-control study conducted in South Africa consisted of control volunteers (n = 50), people living with HIV (PLWH) on ART (n = 50), and untreated PLWH (n = 44). Cardiovascular risk scores were determined, vascular measures were performed, and an extensive biochemical characterisation (routine, metabolomic, and inflammatory systemic profiles) was performed. Methods Anti-apoA1 IgG levels were assessed by an in-house ELISA. Inflammatory biomarkers were measured with the Meso Scale Discovery® platform, and kynurenine pathway metabolites were assessed using targeted metabolomic profiling conducted by liquid chromatography-multiple reaction monitoring/mass spectrometry (LC-MRM/MS). Results Cardiovascular risk scores and vascular measures exhibited similarities across the three groups, while important differences were observed in systemic inflammatory and tryptophan pathways. Anti-apoA1 IgG seropositivity rates were 15%, 40%, and 70% in control volunteers, PLWH ART-treated, and PLWH ART-naïve, respectively. Circulating anti-apoA1 IgG levels were significantly negatively associated with CD4+ cell counts and positively associated with viremia and pro-inflammatory biomarkers (IFNγ, TNFα, MIPα, ICAM-1, VCAM-1). While circulating anti-apoA1 IgG levels were associated with increased levels of kynurenine in both control volunteers and PLWH, the kynurenine/tryptophan ratio was significantly increased in PLWH ART-treated. Conclusion HIV infection increases the humoral response against apoA1, which is associated with established HIV severity criteria and kynurenine pathway activation.
Collapse
Affiliation(s)
- Miguel A. Frias
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sabrina Pagano
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Nasim Bararpour
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Genetics, Stanford University, Stanford, CA, United States
- Stanford Center for Genomics and Personalized Medicine, Stanford, CA, United States
| | - Jonathan Sidibé
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Festus Kamau
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Vanessa Fétaud-Lapierre
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Peter Hudson
- Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Aurélien Thomas
- Faculty Unit of Toxicology, CURML, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Unit of Forensic Toxicology and Chemistry, CURML, Lausanne and Geneva University Hospitals, Lausanne, Geneva, Switzerland
| | - Sandrine Lecour
- Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Hans Strijdom
- Centre for Cardiometabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nicolas Vuilleumier
- Division of Laboratory Medicine, Diagnostic Department, Geneva University Hospitals, Geneva, Switzerland
- Department of Medical Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
12
|
Li A, Fang J. Anti‐angiogenic therapy enhances cancer immunotherapy: Mechanism and clinical application. INTERDISCIPLINARY MEDICINE 2024; 2. [DOI: 10.1002/inmd.20230025] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/18/2023] [Indexed: 01/04/2025]
Abstract
AbstractImmunotherapy, specifically immune checkpoint inhibitors, is revolutionizing cancer treatment, achieving durable control of previously incurable or advanced tumors. However, only a certain group of patients exhibit effective responses to immunotherapy. Anti‐angiogenic therapy aims to block blood vessel growth in tumors by depriving them of essential nutrients and effectively impeding their growth. Emerging evidence shows that tumor vessels exhibit structural and functional abnormalities, resulting in an immunosuppressive microenvironment and poor response to immunotherapy. Both preclinical and clinical studies have used anti‐angiogenic agents to enhance the effectiveness of immunotherapy against cancer. In this review, we concentrate on the synergistic effect of anti‐angiogenic and immune therapies in cancer management, dissect the direct effects and underlying mechanisms of tumor vessels on recruiting and activating immune cells, and discuss the potential of anti‐angiogenic agents to improve the effectiveness of immunotherapy. Lastly, we outline challenges and opportunities for the anti‐angiogenic strategy to enhance immunotherapy. Considering the increasing approval of the combination of anti‐angiogenic and immune therapies in treating cancers, this comprehensive review would be timely and important.
Collapse
Affiliation(s)
- An‐Qi Li
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University Guangzhou China
| | - Jian‐Hong Fang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism Guangdong Provincial Key Laboratory of New Drug Screening School of Pharmaceutical Sciences Southern Medical University Guangzhou China
- Department of Hepatobiliary Surgery I General Surgery Center Zhujiang Hospital Southern Medical University Guangzhou China
| |
Collapse
|
13
|
Shin J, Hong J, Edwards-Glenn J, Krukovets I, Tkachenko S, Adelus ML, Romanoski CE, Rajagopalan S, Podrez E, Byzova TV, Stenina-Adongravi O, Cherepanova OA. Unraveling the Role of Sex in Endothelial Cell Dysfunction: Evidence From Lineage Tracing Mice and Cultured Cells. Arterioscler Thromb Vasc Biol 2024; 44:238-253. [PMID: 38031841 PMCID: PMC10842863 DOI: 10.1161/atvbaha.123.319833] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Biological sex differences play a vital role in cardiovascular diseases, including atherosclerosis. The endothelium is a critical contributor to cardiovascular pathologies since endothelial cells (ECs) regulate vascular tone, redox balance, and inflammatory reactions. Although EC activation and dysfunction play an essential role in the early and late stages of atherosclerosis development, little is known about sex-dependent differences in EC. METHODS We used human and mouse aortic EC as well as EC-lineage tracing (Cdh5-CreERT2 Rosa-YFP [yellow fluorescence protein]) atherosclerotic Apoe-/- mice to investigate the biological sexual dimorphism of the EC functions in vitro and in vivo. Bioinformatics analyses were performed on male and female mouse aortic EC and human lung and aortic EC. RESULTS In vitro, female human and mouse aortic ECs showed more apoptosis and higher cellular reactive oxygen species levels than male EC. In addition, female mouse aortic EC had lower mitochondrial membrane potential (ΔΨm), lower TFAM (mitochondrial transcription factor A) levels, and decreased angiogenic potential (tube formation, cell viability, and proliferation) compared with male mouse aortic EC. In vivo, female mice had significantly higher lipid accumulation within the aortas, impaired glucose tolerance, and lower endothelial-mediated vasorelaxation than males. Using the EC-lineage tracing approach, we found that female lesions had significantly lower rates of intraplaque neovascularization and endothelial-to-mesenchymal transition within advanced atherosclerotic lesions but higher incidents of missing EC lumen coverage and higher levels of oxidative products and apoptosis. RNA-seq analyses revealed that both mouse and human female EC had higher expression of genes associated with inflammation and apoptosis and lower expression of genes related to angiogenesis and oxidative phosphorylation than male EC. CONCLUSIONS Our study delineates critical sex-specific differences in EC relevant to proinflammatory, pro-oxidant, and angiogenic characteristics, which are entirely consistent with a vulnerable phenotype in females. Our results provide a biological basis for sex-specific proatherosclerotic mechanisms.
Collapse
Affiliation(s)
- Junchul Shin
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Junyoung Hong
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jonnelle Edwards-Glenn
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Irene Krukovets
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Svyatoslav Tkachenko
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH, USA
| | - Maria L. Adelus
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
- Clinical Translational Sciences Graduate Program, The University of Arizona, Tucson, AZ, USA
| | - Casey E. Romanoski
- Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ, USA
| | - Sanjay Rajagopalan
- Department of Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Eugene Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tatiana V. Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olga Stenina-Adongravi
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Olga A. Cherepanova
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
14
|
Xu X, Zhang D, Zhao K, Liu Z, Ren X, Zhang X, Lu Z, Qin C, Wang J, Wang S. Comprehensive analysis of the impact of emerging flame retardants on prostate cancer progression: The potential molecular mechanisms and immune infiltration landscape. Toxicology 2024; 501:153681. [PMID: 38006928 DOI: 10.1016/j.tox.2023.153681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/18/2023] [Accepted: 11/21/2023] [Indexed: 11/27/2023]
Abstract
Emerging flame retardants have been used to replace traditional flame retardants, but their potential impact on cancer, especially prostate cancer, is not well understood. Our study aimed to explore the link between flame retardants and prostate cancer, and identify potential carcinogenic mechanisms among populations exposed to emerging flame retardants. We screened flame retardant interacting genes differentially expressed in prostate cancer patients and identified hub genes by protein-protein interaction (PPI) analysis based on the STRING database. Univariate and multivariate Cox regression analyses were performed to construct risk models and identify flame retardant-related prognostic genes. We calculated the proportion of immune cell infiltration to explore the potential mechanism of the prognostic gene, and verified the target cell population of the prognostic gene in the single-cell transcriptome dataset. Our study revealed a significant link between emerging flame retardants and prostate cancer. We constructed a risk model with good predictive ability for prostate cancer prognosis using TCGA dataset, and identified six flame retardant-related prognostic genes validated in the GSE70769 dataset. We found that the expression of M2 macrophages was up-regulated in patients with high expression of prognostic genes, and the single-cell dataset confirmed the expression of prognostic genes in macrophages. Our study confirms the link between emerging flame retardants and prostate cancer, and highlights the role of immune-related pathways in the high-risk population exposed to these flame retardants.
Collapse
Affiliation(s)
- Xinchi Xu
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China; Department of Urology, The Second People's Hospital of Wuhu, Wuhu, Anhui Province 241000, China
| | - Dong Zhang
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Kai Zhao
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Zhanpeng Liu
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Xiaohan Ren
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Xu Zhang
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Zhongwen Lu
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Chao Qin
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China
| | - Jiawei Wang
- Department of Urology, The Second People's Hospital of Wuhu, Wuhu, Anhui Province 241000, China.
| | - Shangqian Wang
- The State Key Lab of Reproductive, Department of Urology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu Province 210029, China.
| |
Collapse
|
15
|
Tan S, Yu H, Zhang Z, Liu Y, Lou G. Hypoxic tumour-derived exosomal miR-1225-5p regulates M2 macrophage polarisation via toll-like receptor 2 to promote ovarian cancer progress. Autoimmunity 2023; 56:2281226. [PMID: 38010845 DOI: 10.1080/08916934.2023.2281226] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 11/05/2023] [Indexed: 11/29/2023]
Abstract
Tumor-secreted exosomes are critical for the functional regulation of tumor-associated macrophages (TAMs). This study aimed to explore how exosomes secreted by ovarian carcinoma cells regulate the phenotype and function of macrophages. Hypoxic treatment of A2780 cells was postulated to mimic the tumor microenvironment, and exosomes were co-cultured with TAMs. miR-1225-5p was enriched in hypoxic exosomes and contributed to M2 macrophage polarizationby modulating Toll-like receptor 2 expression (TLR2). Furthermore, hypoxia-treated macrophages promote ovarian cancer cell viability, migration, and invasion via the wnt/β-catenin pathway. This study clarified that exosomal miR-1225-5p promotes macrophage M2-like polarization by targeting TLR2 to promote ovarian cancer, which may via the wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Shu Tan
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hao Yu
- Nangang District of Heilongjiang Provincial Hospital, Harbin, China
| | - Zhaocong Zhang
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Yiming Liu
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Ge Lou
- Department of Gynecology Oncology, Harbin Medical University Cancer Hospital, Harbin, China
| |
Collapse
|
16
|
Zhu S, Yu Y, Qu M, Qiu Z, Zhang H, Miao C, Guo K. Neutrophil extracellular traps contribute to immunothrombosis formation via the STING pathway in sepsis-associated lung injury. Cell Death Discov 2023; 9:315. [PMID: 37626060 PMCID: PMC10457383 DOI: 10.1038/s41420-023-01614-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/04/2023] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Neutrophil extracellular traps (NETs) are involved in the activation and dysfunction of multiple overlapping and interacting pathways, including the immune response to injury, inflammation, and coagulation, which contribute to the pathogenesis of sepsis-induced acute lung injury (SI-ALI). However, how NETs mediate the relationship between inflammation and coagulation has not been fully clarified. Here, we found that NETs, through stimulator of interferon genes (STING) activation, induced endothelial cell damage with abundant production of tissue factor (TF), which magnified the dysregulation between inflammatory and coagulant responses and resulted in poor prognosis of SI-ALI model mice. Disruption of NETs and inhibition of STING improved the outcomes of septic mice and reduced the inflammatory response and coagulation. Furthermore, Toll-like receptor 2 (TLR2) on the surface of endothelial cells was involved in the interaction between NETs and the STING pathway. Collectively, these findings demonstrate that NETs activate the coagulant cascade in endothelial cells in a STING-dependent manner in the development of SI-ALI.
Collapse
Affiliation(s)
- Shuainan Zhu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Ying Yu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Mengdi Qu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Zhiyun Qiu
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China
| | - Hao Zhang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| | - Kefang Guo
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Shanghai Key Laboratory of Perioperative Stress and Protection, Shanghai, China.
| |
Collapse
|
17
|
Bi W, Guo W, Fan G, Xie L, Jiang C. Identification and validation of a novel overall survival prediction model for immune-related genes in bone metastases of prostate cancer. Aging (Albany NY) 2023; 15:7161-7186. [PMID: 37494663 PMCID: PMC10415549 DOI: 10.18632/aging.204900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 07/06/2023] [Indexed: 07/28/2023]
Abstract
Immunotherapy has become a revolutionary treatment for cancer and brought new vitality to tumor immunity. Bone metastases are the most prevalent metastatic site for advanced prostate cancer (PCa). Therefore, finding new immunotherapy targets in PCa patients with bone metastasis is urgently needed. We conducted an elaborative bioinformatics study of immune-related genes (IRGs) and tumor-infiltrating immune cells (TIICs) in PCa bone metastases. Databases were integrated to obtain RNA-sequencing data and clinical prognostic information. Univariate and multivariate Cox regression analyses were conducted to construct an overall survival (OS) prediction model. GSE32269 was analyzed to acquire differentially expressed IRGs. The OS prediction model was established by employing six IRGs (MAVS, HSP90AA1, FCGR3A, CTSB, FCER1G, and CD4). The CIBERSORT algorithm was adopted to assess the proportion of TIICs in each group. Furthermore, Transwell, MTT, and wound healing assays were employed to determine the effect of MAVS on PCa cells. High-risk patients had worse OS compared to the low-risk patients in the training and validation cohorts. Meanwhile, clinically practical nomograms were generated using these identified IRGs to predict the 3- and 5-year survival rates of patients. The infiltration percentages of some TIICs were closely linked to the risk score of the OS prediction model. Some tumor-infiltrating immune cells were related to the OS. FCGR3A was closely correlated with some TIICs. In vitro experiments verified that up-regulation of MAVS suppressed the proliferation and metastatic abilities of PCa cells. Our work presented a thorough interpretation of TIICs and IRGs for illustrating and discovering new potential immune checkpoints in bone metastases of PCa.
Collapse
Affiliation(s)
- Wen Bi
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Weiming Guo
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Gang Fan
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Lei Xie
- Department of Urology, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Changqing Jiang
- Department of Sports Medicine, Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| |
Collapse
|
18
|
Tarawneh R. Microvascular Contributions to Alzheimer Disease Pathogenesis: Is Alzheimer Disease Primarily an Endotheliopathy? Biomolecules 2023; 13:830. [PMID: 37238700 PMCID: PMC10216678 DOI: 10.3390/biom13050830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer disease (AD) models are based on the notion that abnormal protein aggregation is the primary event in AD, which begins a decade or longer prior to symptom onset, and culminates in neurodegeneration; however, emerging evidence from animal and clinical studies suggests that reduced blood flow due to capillary loss and endothelial dysfunction are early and primary events in AD pathogenesis, which may precede amyloid and tau aggregation, and contribute to neuronal and synaptic injury via direct and indirect mechanisms. Recent data from clinical studies suggests that endothelial dysfunction is closely associated with cognitive outcomes in AD and that therapeutic strategies which promote endothelial repair in early AD may offer a potential opportunity to prevent or slow disease progression. This review examines evidence from clinical, imaging, neuropathological, and animal studies supporting vascular contributions to the onset and progression of AD pathology. Together, these observations support the notion that the onset of AD may be primarily influenced by vascular, rather than neurodegenerative, mechanisms and emphasize the importance of further investigations into the vascular hypothesis of AD.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Center for Memory and Aging, University of New Mexico, Albuquerque, NM 87106, USA
| |
Collapse
|
19
|
Tumor Vasculature as an Emerging Pharmacological Target to Promote Anti-Tumor Immunity. Int J Mol Sci 2023; 24:ijms24054422. [PMID: 36901858 PMCID: PMC10002465 DOI: 10.3390/ijms24054422] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/20/2023] [Accepted: 02/22/2023] [Indexed: 02/25/2023] Open
Abstract
Tumor vasculature abnormality creates a microenvironment that is not suitable for anti-tumor immune response and thereby induces resistance to immunotherapy. Remodeling of dysfunctional tumor blood vessels by anti-angiogenic approaches, known as vascular normalization, reshapes the tumor microenvironment toward an immune-favorable one and improves the effectiveness of immunotherapy. The tumor vasculature serves as a potential pharmacological target with the capacity of promoting an anti-tumor immune response. In this review, the molecular mechanisms involved in tumor vascular microenvironment-modulated immune reactions are summarized. In addition, the evidence of pre-clinical and clinical studies for the combined targeting of pro-angiogenic signaling and immune checkpoint molecules with therapeutic potential are highlighted. The heterogeneity of endothelial cells in tumors that regulate tissue-specific immune responses is also discussed. The crosstalk between tumor endothelial cells and immune cells in individual tissues is postulated to have a unique molecular signature and may be considered as a potential target for the development of new immunotherapeutic approaches.
Collapse
|
20
|
Song C, Pan S, Li D, Hao B, Lu Z, Lai K, Li N, Geng Q. Comprehensive analysis reveals the potential value of inflammatory response genes in the prognosis, immunity, and drug sensitivity of lung adenocarcinoma. BMC Med Genomics 2022; 15:198. [PMID: 36117156 PMCID: PMC9484176 DOI: 10.1186/s12920-022-01340-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 08/16/2022] [Indexed: 11/18/2022] Open
Abstract
Background Although the relationship between inflammatory response and tumor has been gradually recognized, the potential implications of of inflammatory response genes in lung adenocarcinoma (LUAD) remains poorly investigated. Methods RNA sequencing and clinical data were obtained from multiple independent datasets (GSE29013, GSE30219, GSE31210, GSE37745, GSE42127, GSE50081, GSE68465, GSE72094, TCGA and GTEx). Unsupervised clustering analysis was used to identify different tumor subtypes, and LASSO and Cox regression analysis were applied to construct a novel scoring tool. We employed multiple algorithms (ssGSEA, CIBERSORT, MCP counter, and ESTIMATE) to better characterize the LUAD tumor microenvironment (TME) and immune landscapes. GSVA and Metascape analysis were performed to investigate the biological processes and pathway activity. Furthermore, ‘pRRophetic’ R package was used to evaluate the half inhibitory concentration (IC50) of each sample to infer drug sensitivity. Results We identified three distinct tumor subtypes, which were related to different clinical outcomes, biological pathways, and immune characteristics. A scoring tool called inflammatory response gene score (IRGS) was established and well validated in multiple independent cohorts, which could well divide patients into two subgroups with significantly different prognosis. High IRGS patients, characterized by increased genomic variants and mutation burden, presented a worse prognosis, and might show a more favorable response to immunotherapy and chemotherapy. Additionally, based on the cross-talk between TNM stage, IRGS and patients clinical outcomes, we redefined the LUAD stage, which was called ‘IRGS-Stage’. The novel staging system could distinguish patients with different prognosis, with better predictive ability than the conventional TNM staging. Conclusions Inflammatory response genes present important potential value in the prognosis, immunity and drug sensitivity of LUAD. The proposed IRGS and IRGS-Stage may be promising biomarkers for estimating clinical outcomes in LUAD patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12920-022-01340-7.
Collapse
|
21
|
Man SM, Jenkins BJ. Context-dependent functions of pattern recognition receptors in cancer. Nat Rev Cancer 2022; 22:397-413. [PMID: 35355007 DOI: 10.1038/s41568-022-00462-5] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/01/2022] [Indexed: 02/07/2023]
Abstract
The immune system plays a critical role in shaping all facets of cancer, from the early initiation stage through to metastatic disease and resistance to therapy. Our understanding of the importance of the adaptive arm of the immune system in antitumour immunity has led to the implementation of immunotherapy with immune checkpoint inhibitors in numerous cancers, albeit with differing efficacy. By contrast, the clinical utility of innate immunity in cancer has not been exploited, despite dysregulated innate immunity being a feature of at least one-third of all cancers associated with tumour-promoting chronic inflammation. The past two decades have seen innate immune pattern recognition receptors (PRRs) emerge as critical regulators of the immune response to microbial infection and host tissue damage. More recently, it has become apparent that in many cancer types, PRRs play a central role in modulating a vast array of tumour-inhibiting and tumour-promoting cellular responses both in immune cells within the tumour microenvironment and directly in cancer cells. Herein, we provide a comprehensive overview of the fast-evolving field of PRRs in cancer, and discuss the potential to target PRRs for drug development and biomarker discovery in a wide range of oncology settings.
Collapse
Affiliation(s)
- Si Ming Man
- Division of Immunity, Inflammation and Infection, The John Curtin School of Medical Research, The Australian National University, Canberra, ACT, Australia
| | - Brendan J Jenkins
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia.
- Department of Molecular and Translational Science, School of Clinical Sciences, Monash University, Clayton, VIC, Australia.
| |
Collapse
|
22
|
Cai C, Guo Z, Chang X, Li Z, Wu F, He J, Cao T, Wang K, Shi N, Zhou H, Toan S, Muid D, Tan Y. Empagliflozin attenuates cardiac microvascular ischemia/reperfusion through activating the AMPKα1/ULK1/FUNDC1/mitophagy pathway. Redox Biol 2022; 52:102288. [PMID: 35325804 PMCID: PMC8938627 DOI: 10.1016/j.redox.2022.102288] [Citation(s) in RCA: 133] [Impact Index Per Article: 44.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/07/2022] [Accepted: 03/12/2022] [Indexed: 02/07/2023] Open
Abstract
Mitophagy preserves microvascular structure and function during myocardial ischemia/reperfusion (I/R) injury. Empagliflozin, an anti-diabetes drug, may also protect mitochondria. We explored whether empagliflozin could reduce cardiac microvascular I/R injury by enhancing mitophagy. In mice, I/R injury induced luminal stenosis, microvessel wall damage, erythrocyte accumulation and perfusion defects in the myocardial microcirculation. Additionally, I/R triggered endothelial hyperpermeability and myocardial neutrophil infiltration, which upregulated adhesive factors and endothelin-1 but downregulated vascular endothelial cadherin and endothelial nitric oxide synthase in heart tissue. In vitro, I/R impaired the endothelial barrier function and integrity of cardiac microvascular endothelial cells (CMECs), while empagliflozin preserved CMEC homeostasis and thus maintained cardiac microvascular structure and function. I/R activated mitochondrial fission, oxidative stress and apoptotic signaling in CMECs, whereas empagliflozin normalized mitochondrial fission and fusion, neutralized supraphysiologic reactive oxygen species concentrations and suppressed mitochondrial apoptosis. Empagliflozin exerted these protective effects by activating FUNDC1-dependent mitophagy through the AMPKα1/ULK1 pathway. Both in vitro and in vivo, genetic ablation of AMPKα1 or FUNDC1 abolished the beneficial effects of empagliflozin on the myocardial microvasculature and CMECs. Taken together, the preservation of mitochondrial function through an activation of the AMPKα1/ULK1/FUNDC1/mitophagy pathway is the working mechanism of empagliflozin in attenuating cardiac microvascular I/R injury. Empagliflozin reduces I/R-induced microvascular damage. Empagliflozin suppresses I/R-induced endothelial cell damage. Empagliflozin activates FUNDC1-dependent mitophagy through the AMPKα1/ULK1 pathway. Ablation of FUNDC1 or AMPKα1 abolishes the protective effects of empagliflozin against I/R-induced microvascular damage.
Collapse
|
23
|
Xiong L, McCoy M, Murtazina R, Podrez EA, Byzova TV. Timely Wound Healing is Dependent upon Endothelial but not Hair Follicle Stem Cell Toll-like Receptor 2 Signaling. J Invest Dermatol 2022; 142:3082-3092.e1. [PMID: 35561753 DOI: 10.1016/j.jid.2022.04.018] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/08/2022] [Accepted: 04/22/2022] [Indexed: 11/18/2022]
Abstract
As a part of innate immunity, Toll-like receptor 2 (TLR2) plays an important function in most defensive responses of the organism, including but not limited to infections. Cutaneous injury, one of the most common challenges for mammals, mobilizes a number of cell types, including epithelial, immune, and vascular cells for timely tissue repair. However, in contrast to immune cells, little is known about TLR2 function on non-immune cells during skin regeneration. Here, we used two tissue-specific conditional TLR2 knockout mouse lines to address the impact of TLR2 in endothelial and hair follicle stem cells (HFSCs) on cutaneous wound healing. The loss of TLR2 on endothelial cells diminishes their ability to migrate, sprout, and proliferate in response to specific TLR2 ligands, and also reduces the secretion of key pro-angiogenic factors. Lack of TLR2 on endothelial cells prolongs wound healing due to diminished angiogenesis. TLR2 is expressed in key structures of hair follicle including HFSCs, secondary hair germ, and dermal papilla. Despite the prominent role for HFSCs in skin regeneration, excision of TLR2 from HFSCs has no impact on their proliferation or wound healing potential. Our study demonstrates that timely tissue regeneration after skin injury is dependent upon endothelial TLR2 for robust angiogenesis, while HFSC TLR2 is dispensable.
Collapse
Affiliation(s)
- Luyang Xiong
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Michael McCoy
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Rakhilya Murtazina
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA; Department of Biochemistry and Molecular Genetics, College of Medicine, University of Illinois Chicago, Chicago, Illinois, USA
| | - Eugene A Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.
| |
Collapse
|
24
|
Liu W, Xiong S, Du J, Song Y, Wang T, Zhang Y, Dong C, Huang Z, He Q, Yu Z, Ma X. Deciphering Key Foreign Body Reaction-Related Transcription Factors and Genes Through Transcriptome Analysis. Front Mol Biosci 2022; 9:843391. [PMID: 35350715 PMCID: PMC8958039 DOI: 10.3389/fmolb.2022.843391] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Silicone implants are widely used in the field of plastic surgery for wound repair and cosmetic augmentation. However, molecular mechanisms and signaling pathways underlying the foreign body reaction (FBR) of a host tissue to the silicone require further elucidation. The purpose of this study was to identify key FBR-related transcription factors (TFs) and genes through transcriptome analysis.Methods: We used a rat model with a subcutaneous silicone implant in the scalp and performed high throughput sequencing to determine the transcriptional profiles involved in the FBR. The function was analyzed by Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway-enrichment analysis. A protein-protein interaction (PPI) network of differentially expressed mRNAs (DEmRNAs) was constructed to identify the hub genes and key modules and to determine the regulatory TF-mRNA relationships. In addition, the hub gene and transcript expression levels were determined by Quantitative Reverse Transcription polymerase Chain Reaction (qRT-PCR). Myofibroblasts differentiation and macrophage recruitment were identified by immunofluorescence. The protein expression of MMP9 was detected by immunohistochemistry and Western blot.Results: We identified ten hub genes (Fos, Spp1, Fn1, Ctgf, Tlr2, Itgb2, Itgax, Ccl2, Mmp9, and Serpine1) and 3 TFs (FOS, IRF4, and SPI1) that may be crucial (particularly FOS) for the FBR. Furthermore, we identified multiple differentially expressed genes involved in several important biological processes, including leukocyte migration, cytokine‒ cytokine receptor interaction, phagocytosis, extracellular matrix (ECM) organization, and angiogenesis. We also identified potentially significant signaling pathways, including cytokine‒cytokine receptor interaction, phagosome, ECM‒receptor interaction, complement and coagulation cascades, the IL-17 signaling pathway, and the PI3K‒Akt signaling pathway. In addition, qRT-PCR confirmed the expression patterns of the TFs and hub genes, Western blot and immunohistochemistry validated the expression patterns of MMP9.Conclusion: We generated a comprehensive overview of the gene networks underlying the FBR evoked by silicone implants. Moreover, we identified specific molecular and signaling pathways that may perform key functions in the silicone implant-induced FBR. Our results provide significant insights into the molecular mechanisms underlying silicone-induced FBR and determine novel therapeutic targets to reduce complications related to silicone implantation.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Zhou Yu
- *Correspondence: Zhou Yu, ; Xianjie Ma,
| | | |
Collapse
|
25
|
Vellasco L, Svensjö E, Bulant CA, Blanco PJ, Nogueira F, Domont G, de Almeida NP, Nascimento CR, Silva-dos-Santos D, Carvalho-Pinto CE, Medei EH, Almeida IC, Scharfstein J. Sheltered in Stromal Tissue Cells, Trypanosoma cruzi Orchestrates Inflammatory Neovascularization via Activation of the Mast Cell Chymase Pathway. Pathogens 2022; 11:pathogens11020187. [PMID: 35215131 PMCID: PMC8878313 DOI: 10.3390/pathogens11020187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/04/2022] Open
Abstract
Microangiopathy may worsen the clinical outcome of Chagas disease. Given the obstacles to investigating the dynamics of inflammation and angiogenesis in heart tissues parasitized by Trypanosoma cruzi, here we used intravital microscopy (IVM) to investigate microcirculatory alterations in the hamster cheek pouch (HCP) infected by green fluorescent protein-expressing T. cruzi (GFP-T. cruzi). IVM performed 3 days post-infection (3 dpi) consistently showed increased baseline levels of plasma extravasation. Illustrating the reciprocal benefits that microvascular leakage brings to the host-parasite relationship, these findings suggest that intracellular amastigotes, acting from inside out, stimulate angiogenesis while enhancing the delivery of plasma-borne nutrients and prosurvival factors to the infection foci. Using a computer-based analysis of images (3 dpi), we found that proangiogenic indexes were positively correlated with transcriptional levels of proinflammatory cytokines (pro-IL1β and IFN-γ). Intracellular GFP-parasites were targeted by delaying for 24 h the oral administration of the trypanocidal drug benznidazole. A classification algorithm showed that benznidazole (>24 h) blunted angiogenesis (7 dpi) in the HCP. Unbiased proteomics (3 dpi) combined to pharmacological targeting of chymase with two inhibitors (chymostatin and TY-51469) linked T. cruzi-induced neovascularization (7 dpi) to the proangiogenic activity of chymase, a serine protease stored in secretory granules from mast cells.
Collapse
Affiliation(s)
- Lucas Vellasco
- Department of Immunobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (E.S.); (C.R.N.); (D.S.-d.-S.); (E.H.M.)
| | - Erik Svensjö
- Department of Immunobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (E.S.); (C.R.N.); (D.S.-d.-S.); (E.H.M.)
| | - Carlos Alberto Bulant
- Department of Mathematical and Computational Methods, National Laboratory for Scientific Computing, Petrópolis 25651-075, Brazil; (C.A.B.); (P.J.B.)
| | - Pablo Javier Blanco
- Department of Mathematical and Computational Methods, National Laboratory for Scientific Computing, Petrópolis 25651-075, Brazil; (C.A.B.); (P.J.B.)
| | - Fábio Nogueira
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (F.N.); (G.D.); (N.P.d.A.)
| | - Gilberto Domont
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (F.N.); (G.D.); (N.P.d.A.)
| | - Natália Pinto de Almeida
- Department of Biochemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil; (F.N.); (G.D.); (N.P.d.A.)
| | - Clarissa Rodrigues Nascimento
- Department of Immunobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (E.S.); (C.R.N.); (D.S.-d.-S.); (E.H.M.)
| | - Danielle Silva-dos-Santos
- Department of Immunobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (E.S.); (C.R.N.); (D.S.-d.-S.); (E.H.M.)
| | | | - Emiliano Horácio Medei
- Department of Immunobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (E.S.); (C.R.N.); (D.S.-d.-S.); (E.H.M.)
| | - Igor C. Almeida
- Department of Biological Sciences, Border Biomedical Research Center, University of Texas at El Paso, El Paso, TX 79968, USA;
| | - Julio Scharfstein
- Department of Immunobiology, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (L.V.); (E.S.); (C.R.N.); (D.S.-d.-S.); (E.H.M.)
- Correspondence:
| |
Collapse
|
26
|
Xiong L, McCoy M, Komuro H, West XZ, Yakubenko V, Gao D, Dudiki T, Milo A, Chen J, Podrez EA, Trapp B, Byzova TV. Inflammation-dependent oxidative stress metabolites as a hallmark of amyotrophic lateral sclerosis. Free Radic Biol Med 2022; 178:125-133. [PMID: 34871763 PMCID: PMC8744315 DOI: 10.1016/j.freeradbiomed.2021.11.031] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/24/2021] [Accepted: 11/22/2021] [Indexed: 01/03/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease, with poor prognosis and no cure. Substantial evidence implicates inflammation and associated oxidative stress as a potential mechanism for ALS, especially in patients carrying the SOD1 mutation and, therefore, lacking anti-oxidant defense. The brain is particularly vulnerable to oxidation due to the abundance of polyunsaturated fatty acids, such as docosahexaenoic acid (DHA), which can give rise to several oxidized metabolites. Accumulation of a DHA peroxidation product, CarboxyEthylPyrrole (CEP) is dependent on activated inflammatory cells and myeloperoxidase (MPO), and thus marks areas of inflammation-associated oxidative stress. At the same time, generation of an alternative inactive DHA peroxidation product, ethylpyrrole, does not require cell activation and MPO activity. While absent in normal brain tissues, CEP is accumulated in the central nervous system (CNS) of ALS patients, reaching particularly high levels in individuals carrying a SOD1 mutation. ALS brains are characterized by high levels of MPO and lowered anti-oxidant activity (due to the SOD1 mutation), thereby aiding CEP generation and accumulation. Due to DHA oxidation within the membranes, CEP marks cells with the highest oxidative damage. In all ALS cases CEP is present in nearly all astrocytes and microglia, however, only in individuals carrying a SOD1 mutation CEP marks >90% of neurons, thereby emphasizing an importance of CEP accumulation as a potential hallmark of oxidative damage in neurodegenerative diseases.
Collapse
Affiliation(s)
- Luyang Xiong
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Michael McCoy
- Department of Medicine, Cardiovascular Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Hitoshi Komuro
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Xiaoxia Z West
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Valentin Yakubenko
- Department of Biomedical Sciences, Center of Excellence for Inflammation, Infectious Disease and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN, 37684, USA
| | - Detao Gao
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Tejasvi Dudiki
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Amanda Milo
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jacqueline Chen
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Eugene A Podrez
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Bruce Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Tatiana V Byzova
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA.
| |
Collapse
|
27
|
Ren B, Ramchandran R, Yang X. Editorial: Molecular Mechanisms and Signaling in Endothelial Cell Biology and Vascular Heterogeneity. Front Cell Dev Biol 2021; 9:821100. [PMID: 34977049 PMCID: PMC8718799 DOI: 10.3389/fcell.2021.821100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 12/14/2022] Open
Affiliation(s)
- Bin Ren
- Department of Surgery, O’Neal Comprehensive Cancer Center, and Comprehensive Cardiovascular Center, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
- *Correspondence: Bin Ren,
| | - Ramani Ramchandran
- Department of Pediatrics and Children’s Research Institute, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Xiaofeng Yang
- Department of Cardiovascular Sciences, Centers for Cardiovascular Research and Metabolic Disease Research, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
28
|
Eteshola EO, Landa K, Rempel RE, Naqvi IA, Hwang ES, Nair SK, Sullenger BA. Breast cancer-derived DAMPs enhance cell invasion and metastasis, while nucleic acid scavengers mitigate these effects. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 26:1-10. [PMID: 34513289 PMCID: PMC8408553 DOI: 10.1016/j.omtn.2021.06.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/25/2021] [Indexed: 12/23/2022]
Abstract
Breast cancer (BC) is the most common malignancy in women. Particular subtypes with aggressive behavior are major contributors to poor outcomes. Triple-negative breast cancer (TNBC) is difficult to treat, pro-inflammatory, and highly metastatic. We demonstrate that TNBC cells express TLR9 and are responsive to TLR9 ligands, and treatment of TNBC cells with chemotherapy increases the release of nucleic-acid-containing damage-associated molecular patterns (NA DAMPs) in cell culture. Such culture-derived and breast cancer patient-derived NA DAMPs increase TLR9 activation and TNBC cell invasion in vitro. Notably, treatment with the polyamidoamine dendrimer generation 3.0 (PAMAM-G3) behaved as a nucleic acid scavenger (NAS) and significantly mitigates such effects. In mice that develop spontaneous BC induced by polyoma middle T oncoprotein (MMTV-PyMT), treatment with PAMAM-G3 significantly reduces lung metastasis. Thus, NAS treatment mitigates cancer-induced inflammation and metastasis and represents a novel therapeutic approach for combating breast cancer.
Collapse
Affiliation(s)
- Elias O.U. Eteshola
- Duke University School of Medicine, Department of Pharmacology and Cancer Biology, Durham, NC 27710, USA
- Duke University Medical Center, Department of Surgery, Durham, NC 27710, USA
| | - Karenia Landa
- Duke University Medical Center, Department of Surgery, Durham, NC 27710, USA
| | - Rachel E. Rempel
- Duke University Medical Center, Department of Surgery, Durham, NC 27710, USA
| | - Ibtehaj A. Naqvi
- Duke University Medical Center, Department of Surgery, Durham, NC 27710, USA
| | - E. Shelley Hwang
- Duke University Medical Center, Department of Surgery, Durham, NC 27710, USA
- Duke Cancer Institute, Durham, NC 27710, USA
| | - Smita K. Nair
- Duke University Medical Center, Department of Surgery, Durham, NC 27710, USA
- Duke Cancer Institute, Durham, NC 27710, USA
| | - Bruce A. Sullenger
- Duke University School of Medicine, Department of Pharmacology and Cancer Biology, Durham, NC 27710, USA
- Duke University Medical Center, Department of Surgery, Durham, NC 27710, USA
- Duke Cancer Institute, Durham, NC 27710, USA
| |
Collapse
|
29
|
Reeves KM, Song PN, Angermeier A, Della Manna D, Li Y, Wang J, Yang ES, Sorace AG, Larimer BM. 18F-FMISO PET Imaging Identifies Hypoxia and Immunosuppressive Tumor Microenvironments and Guides Targeted Evofosfamide Therapy in Tumors Refractory to PD-1 and CTLA-4 Inhibition. Clin Cancer Res 2021; 28:327-337. [PMID: 34615724 DOI: 10.1158/1078-0432.ccr-21-2394] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/25/2021] [Accepted: 10/04/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE Hypoxia is a common characteristic of many tumor microenvironments, and it has been shown to promote suppression of anti-tumor immunity. Despite strong biological rationale, longitudinal correlation of hypoxia and response to immunotherapy has not been investigated. EXPERIMENTAL DESIGN In this study, we probed the tumor and its surrounding microenvironment with 18F-FMISO PET imaging to non-invasively quantify tumor hypoxia in vivo prior to and during PD-1 and CTLA-4 checkpoint blockade in preclinical models of breast and colon cancer. RESULTS Longitudinal imaging identified hypoxia as an early predictive biomarker of therapeutic response (prior to anatomic changes in tumor volume) with a decreasing standard uptake value (SUV) ratio in tumors that effectively respond to therapy. PET signal correlated with ex vivo markers of tumor immune response including cytokines (Ifng, Gzmb, and Tnf), damage-associated molecular pattern receptors (Tlr2/4) and immune cell populations (macrophages, dendritic cells, and cytotoxic T cells). Responding tumors were marked by increased inflammation that were spatially distinct from hypoxic regions, providing a mechanistic understanding of the immune signaling pathways activated. To exploit image-guided combination therapy, hypoxia signal from PET imaging was used to guide the addition of a hypoxia targeted treatment to non-responsive tumors, which ultimately provided therapeutic synergy and rescued response as determined by longitudinal changes in tumor volume. CONCLUSIONS The results generated from this work provide an immediately translatable paradigm for measuring and targeting hypoxia to increase response to immune checkpoint therapy and using hypoxia imaging to guide combinatory therapies.
Collapse
Affiliation(s)
| | | | - Allyson Angermeier
- Cellular, Molecular, and Developmental Biology, University of Alabama at Birmingham
| | | | - Yufeng Li
- Division of Preventive Medicine, University of Alabama at Birmingham
| | - Jianbo Wang
- Cellular, Developmental and Integrative Biology, University of Alabama at Birmingham
| | - Eddy S Yang
- Department of Radiation Oncology, University of Alabama at Birmingham
| | - Anna G Sorace
- Radiology and Biomedical Engineering, University of Alabama at Birmingham
| | | |
Collapse
|
30
|
Abstract
TLR2 located in the endothelium promotes angiogenesis and inflammation.
Collapse
Affiliation(s)
- Samantha Mahfoud
- Department of Oncology, University of Lausanne, and Ludwig Institute for Cancer Research Lausanne, 1066 Epalinges, Switzerland
| | - Tatiana V Petrova
- Department of Oncology, University of Lausanne, and Ludwig Institute for Cancer Research Lausanne, 1066 Epalinges, Switzerland
| |
Collapse
|