1
|
Fanoodi A, Maharati A, Akhlaghipour I, Rahimi HR, Moghbeli M. MicroRNAs as the critical regulators of tumor angiogenesis in liver cancer. Pathol Res Pract 2023; 251:154913. [PMID: 37931431 DOI: 10.1016/j.prp.2023.154913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/25/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
Liver cancer is one of the most common malignancies in human digestive system. Despite the recent therapeutic methods, there is a high rate of mortality among liver cancer patients. Late diagnosis in the advanced tumor stages can be one of the main reasons for the poor prognosis in these patients. Therefore, investigating the molecular mechanisms of liver cancer can be helpful for the early stage tumor detection and treatment. Vascular expansion in liver tumors can be one of the important reasons for poor prognosis and aggressiveness. Therefore, anti-angiogenic drugs are widely used in liver cancer patients. MicroRNAs (miRNAs) have key roles in the regulation of angiogenesis in liver tumors. Due to the high stability of miRNAs in body fluids, these factors are widely used as the non-invasive diagnostic and prognostic markers in cancer patients. Regarding, the importance of angiogenesis during liver tumor growth and invasion, in the present review, we discussed the role of miRNAs in regulation of angiogenesis in these tumors. It has been reported that miRNAs mainly exert an anti-angiogenic function by regulation of tumor microenvironment, transcription factors, and signaling pathways in liver tumors. This review can be an effective step to suggest the miRNAs for the non-invasive early detection of malignant and invasive liver tumors.
Collapse
Affiliation(s)
- Ali Fanoodi
- Student Research Committee, School of Medicine, Birjand University of Medical Sciences, Birjand, Iran
| | - Amirhosein Maharati
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Iman Akhlaghipour
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamid Reza Rahimi
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Meysam Moghbeli
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Ullah A, Zhao J, Singla RK, Shen B. Pathophysiological impact of CXC and CX3CL1 chemokines in preeclampsia and gestational diabetes mellitus. Front Cell Dev Biol 2023; 11:1272536. [PMID: 37928902 PMCID: PMC10620730 DOI: 10.3389/fcell.2023.1272536] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Diabetes-related pathophysiological alterations and various female reproductive difficulties were common in pregnant women with gestational diabetes mellitus (GDM), who had 21.1 million live births. Preeclampsia (PE), which increases maternal and fetal morbidity and mortality, affects approximately 3%-5% of pregnancies worldwide. Nevertheless, it is unclear what triggers PE and GDM to develop. Therefore, the development of novel moderator therapy approaches is a crucial advancement. Chemokines regulate physiological defenses and maternal-fetal interaction during healthy and disturbed pregnancies. Chemokines regulate immunity, stem cell trafficking, anti-angiogenesis, and cell attraction. CXC chemokines are usually inflammatory and contribute to numerous reproductive disorders. Fractalkine (CX3CL1) may be membrane-bound or soluble. CX3CL1 aids cell survival during homeostasis and inflammation. Evidence reveals that CXC and CX3CL1 chemokines and their receptors have been the focus of therapeutic discoveries for clinical intervention due to their considerable participation in numerous biological processes. This review aims to give an overview of the functions of CXC and CX3CL1 chemokines and their receptors in the pathophysiology of PE and GDM. Finally, we examined stimulus specificity for CXC and CX3CL1 chemokine expression and synthesis in PE and GDM and preclinical and clinical trials of CXC-based PE and GDM therapies.
Collapse
Affiliation(s)
- Amin Ullah
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jing Zhao
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Rajeev K. Singla
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Bairong Shen
- Joint Laboratory of Artificial Intelligence for Critical Care Medicine, Department of Critical Care Medicine, Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Hassanshahi F, Noroozi Karimabad M, Miranzadeh E, Hassanshahi G, Torabizadeh SA, Jebali A. The Serum Level of CXCL9, CXCL10, and CXCL11 and the Expression of CXCR3 of Peripheral Blood Mononuclear Cells in Brucellosis Patients. Curr Microbiol 2023; 80:201. [PMID: 37140634 DOI: 10.1007/s00284-023-03230-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 02/14/2023] [Indexed: 05/05/2023]
Abstract
Brucella spp. can replicate in human endothelial cells, inducing an inflammatory response with increased expression of chemokines. Although Brucella infects humans, its ability to induce the production of chemokines by lung cells is unknown. Therefore, the current investigation was designed to examine the association between brucellosis and CXCL9, 10, and 11 chemokines. The patient group included 71 patients suffering from Brucella infection and the control group consisted of 50 healthy ranchers from the same geographical area. Serum levels of CXCL9, CXCL10, and CXCL11 were analyzed by ELISA. The fold changes of CXCR3 expression against β-actin were determined by real-time-PCR technique. Western blotting analysis was also applied for evaluating the expression of CXCR3 at protein level. The results of this study showed that the serum levels of CXCL9, CXCL10, and CXCL11 are significantly increased in acute brucellosis patients in comparison to control as indicated by ELISA test, mRNA levels of CXCR3 by Real-time PCR as well as protein levels of CXCR3 by Western blot analysis. According to findings, these chemokines have the potential to serve as markers for brucellosis patients. Taken together, cytokine/chemokine network was active in acute brucellosis patients, and it is suggested to evaluate other cytokines in future studies.
Collapse
Affiliation(s)
- Farzaneh Hassanshahi
- Faculty of Veterinary Medicine, Islamic Azad university Shahr-E-Kord -Branch, Shahr-e-kord, Iran
| | - Mojgan Noroozi Karimabad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Science, Rafsanjan, Iran.
| | - Elahe Miranzadeh
- Faculty of Veterinary Medicine, Islamic Azad university Shahr-E-Kord -Branch, Shahr-e-kord, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Science, Rafsanjan, Iran
| | - Seyedeh Atekeh Torabizadeh
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Jebali
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Science, Rafsanjan, Iran
| |
Collapse
|
4
|
Recent Emerging Immunological Treatments for Primary Brain Tumors: Focus on Chemokine-Targeting Immunotherapies. Cells 2023; 12:cells12060841. [PMID: 36980182 PMCID: PMC10046911 DOI: 10.3390/cells12060841] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/01/2023] [Accepted: 03/05/2023] [Indexed: 03/11/2023] Open
Abstract
Primary brain tumors are a leading cause of death worldwide and are characterized by extraordinary heterogeneity and high invasiveness. Current drug and radiotherapy therapies combined with surgical approaches tend to increase the five-year survival of affected patients, however, the overall mortality rate remains high, thus constituting a clinical challenge for which the discovery of new therapeutic strategies is needed. In this field, novel immunotherapy approaches, aimed at overcoming the complex immunosuppressive microenvironment, could represent a new method of treatment for central nervous system (CNS) tumors. Chemokines especially are a well-defined group of proteins that were so named due to their chemotactic properties of binding their receptors. Chemokines regulate the recruitment and/or tissue retention of immune cells as well as the mobilization of tumor cells that have undergone epithelial–mesenchymal transition, promoting tumor growth. On this basis, this review focuses on the function and involvement of chemokines and their receptors in primary brain tumors, specifically examining chemokine-targeting immunotherapies as one of the most promising strategies in neuro-oncology.
Collapse
|
5
|
Karimabad MN, Hassanshahi G, Kounis NG, Mplani V, Roditis P, Gogos C, Lagadinou M, Assimakopoulos SF, Dousdampanis P, Koniari I. The Chemokines CXC, CC and C in the Pathogenesis of COVID-19 Disease and as Surrogates of Vaccine-Induced Innate and Adaptive Protective Responses. Vaccines (Basel) 2022; 10:vaccines10081299. [PMID: 36016187 PMCID: PMC9416781 DOI: 10.3390/vaccines10081299] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/08/2022] [Accepted: 08/08/2022] [Indexed: 02/07/2023] Open
Abstract
COVID-19 is one of the progressive viral pandemics that originated from East Asia. COVID-19 or SARS-CoV-2 has been shown to be associated with a chain of physio-pathological mechanisms that are basically immunological in nature. In addition, chemokines have been proposed as a subgroup of chemotactic cytokines with different activities ranging from leukocyte recruitment to injury sites, irritation, and inflammation to angiostasis and angiogenesis. Therefore, researchers have categorized the chemotactic elements into four classes, including CX3C, CXC, CC, and C, based on the location of the cysteine motifs in their structures. Considering the severe cases of COVID-19, the hyperproduction of particular chemokines occurring in lung tissue as well as pro-inflammatory cytokines significantly worsen the disease prognosis. According to the studies conducted in the field documenting the changing expression of CXC and CC chemokines in COVID-19 cases, the CC and CXC chemokines contribute to this pandemic, and their impact could reflect the development of reasonable strategies for COVID-19 management. The CC and the CXC families of chemokines are important in host immunity to viral infections and along with other biomarkers can serve as the surrogates of vaccine-induced innate and adaptive protective responses, facilitating the improvement of vaccine efficacy. Furthermore, the immunogenicity elicited by the chemokine response to adenovirus vector vaccines may constitute the basis of vaccine-induced immune thrombotic thrombocytopaenia.
Collapse
Affiliation(s)
- Mojgan Noroozi Karimabad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan 7717933777, Iran
| | - Nicholas G. Kounis
- Department of Internal Medicine, Division of Cardiology, University of Patras Medical School, 26500 Patras, Greece
- Correspondence:
| | - Virginia Mplani
- Intensive Care Unit, Patras University Hospital, 26500 Patras, Greece
| | - Pavlos Roditis
- Department of Cardiology, Mamatsio Kozanis General Hospital, 50100 Kozani, Greece
| | - Christos Gogos
- COVID-19 Unit, Papageorgiou General Hospital, 56403 Thessaloniki, Greece
| | - Maria Lagadinou
- Department of Internal Medicine, Division of Infectious Diseases, University of Patras Medical School, 26500 Patras, Greece
| | - Stelios F. Assimakopoulos
- Department of Internal Medicine, Division of Infectious Diseases, University of Patras Medical School, 26500 Patras, Greece
| | - Periklis Dousdampanis
- Department of Nephrology, Saint Andrews State General Hospital, 26221 Patras, Greece
| | - Ioanna Koniari
- Department of Cardiology, University Hospital of South Manchester, NHS Foundation Trust, Manchester M23 9LT, UK
| |
Collapse
|
6
|
Cunningham CM, Li M, Ruffenach G, Doshi M, Aryan L, Hong J, Park J, Hrncir H, Medzikovic L, Umar S, Arnold AP, Eghbali M. Y-Chromosome Gene, Uty, Protects Against Pulmonary Hypertension by Reducing Proinflammatory Chemokines. Am J Respir Crit Care Med 2022; 206:186-196. [PMID: 35504005 PMCID: PMC9887415 DOI: 10.1164/rccm.202110-2309oc] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Rationale: Idiopathic pulmonary arterial hypertension (PAH) is a terminal pulmonary vascular disease characterized by increased pressure, right ventricular failure, and death. PAH exhibits a striking sex bias and is up to four times more prevalent in females. Understanding the molecular basis behind sex differences could help uncover novel therapies. Objectives: We previously discovered that the Y chromosome is protective against hypoxia-induced experimental pulmonary hypertension (PH), which may contribute to sex differences in PAH. Here, we identify the gene responsible for Y-chromosome protection, investigate key downstream autosomal genes, and demonstrate a novel preclinical therapy. Methods: To test the effect of Y-chromosome genes on PH development, we knocked down each Y-chromosome gene expressed in the lung by means of intratracheal instillation of siRNA in gonadectomized male mice exposed to hypoxia and monitored changes in right ventricular and pulmonary artery hemodynamics. We compared the lung transcriptome of Uty knockdown mouse lungs to those of male and female PAH patient lungs to identify common downstream pathogenic chemokines and tested the effects of these chemokines on human pulmonary artery endothelial cells. We further inhibited the activity of these chemokines in two preclinical pulmonary hypertension models to test the therapeutic efficacy. Measurements and Main Results: Knockdown of the Y-chromosome gene Uty resulted in more severe PH measured by increased right ventricular pressure and decreased pulmonary artery acceleration time. RNA sequencing revealed an increase in proinflammatory chemokines Cxcl9 and Cxcl10 as a result of Uty knockdown. We found CXCL9 and CXCL10 significantly upregulated in human PAH lungs, with more robust upregulation in females with PAH. Treatment of human pulmonary artery endothelial cells with CXCL9 and CXCL10 triggered apoptosis. Inhibition of Cxcl9 and Cxcl10 expression in male Uty knockout mice and CXCL9 and CXCL10 activity in female rats significantly reduced PH severity. Conclusions:Uty is protective against PH. Reduction of Uty expression results in increased expression of proinflammatory chemokines Cxcl9 and Cxcl10, which trigger endothelial cell death and PH. Inhibition of CLXC9 and CXLC10 rescues PH development in multiple experimental models.
Collapse
Affiliation(s)
- Christine M. Cunningham
- Division of Molecular Medicine, Department of Anesthesiology,,School of Medicine, Stanford University, Stanford, California;,VA Palo Alto Health Care System, Palo Alto, California; and
| | - Min Li
- Division of Molecular Medicine, Department of Anesthesiology
| | | | - Mitali Doshi
- Division of Molecular Medicine, Department of Anesthesiology,,University of Massachusetts Medical School, Worcester, Massachusetts
| | - Laila Aryan
- Division of Molecular Medicine, Department of Anesthesiology
| | - Jason Hong
- Division of Molecular Medicine, Department of Anesthesiology,,Division of Pulmonary and Critical Care Medicine, Department of Medicine, and
| | - John Park
- Division of Molecular Medicine, Department of Anesthesiology
| | - Haley Hrncir
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, California
| | | | - Soban Umar
- Division of Molecular Medicine, Department of Anesthesiology
| | - Arthur P. Arnold
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, California
| | | |
Collapse
|
7
|
The Involvement of CXC Motif Chemokine Ligand 10 (CXCL10) and Its Related Chemokines in the Pathogenesis of Coronary Artery Disease and in the COVID-19 Vaccination: A Narrative Review. Vaccines (Basel) 2021; 9:vaccines9111224. [PMID: 34835155 PMCID: PMC8623875 DOI: 10.3390/vaccines9111224] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022] Open
Abstract
Coronary artery disease (CAD) and coronary heart disease (CHD) constitute two of the leading causes of death in Europe, USA and the rest of the world. According to the latest reports of the Iranian National Health Ministry, CAD is the main cause of death in Iranian patients with an age over 35 years despite a significant reduction in mortality due to early interventional treatments in the context of an acute coronary syndrome (ACS). Inflammation plays a fundamental role in coronary atherogenesis, atherosclerotic plaque formation, acute coronary thrombosis and CAD establishment. Chemokines are well-recognized mediators of inflammation involved in several bio-functions such as leucocyte migration in response to inflammatory signals and oxidative vascular injury. Different chemokines serve as chemo-attractants for a wide variety of cell types including immune cells. CXC motif chemokine ligand 10 (CXCL10), also known as interferon gamma-induced protein 10 (IP-10/CXLC10), is a chemokine with inflammatory features whereas CXC chemokine receptor 3 (CXCR3) serves as a shared receptor for CXCL9, 10 and 11. These chemokines mediate immune responses through the activation and recruitment of leukocytes, eosinophils, monocytes and natural killer (NK) cells. CXCL10, interleukin (IL-15) and interferon (IFN-g) are increased after a COVID-19 vaccination with a BNT162b2 mRNA (Pfizer/BioNTech) vaccine and are enriched by tumor necrosis factor alpha (TNF-α) and IL-6 after the second vaccination. The aim of the present study is the presentation of the elucidation of the crucial role of CXCL10 in the patho-physiology and pathogenesis of CAD and in identifying markers associated with the vaccination resulting in antibody development.
Collapse
|
8
|
Guan C, Zhao F, Yang Z, Tang Q, Wang L, Li X, Zhang L, Deng Z, Hou H, Wang J, Xu Y, Zhang R, Lin Y, Tan P, Zhang Y, Liu S, Zhang L. A review of key cytokines based on gene polymorphism in the pathogenesis of pre-eclampsia. Am J Reprod Immunol 2021; 87:e13503. [PMID: 34599631 DOI: 10.1111/aji.13503] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/26/2021] [Accepted: 09/28/2021] [Indexed: 12/15/2022] Open
Abstract
Although a number of theories have been suggested, including roles for oxidative stress, an abnormal maternal-fetal interface, and genetic and environmental factors, the etiopathology of pre-eclampsia (PE) remains unclear. Maternal immune tolerance is important for maintaining pregnancy, and researchers have increasingly focused on the critical roles of cytokines in the pathogenesis of PE in recent years. The assessment of candidate genetic polymorphisms in PE could partially elucidate the mechanisms of susceptibility to disease, and contribute to seeking for new diagnosis and treatment methods of PE. PE can lead to severe complications, and even the death of both mother and fetus. Although the complex pathology is not yet clear, some evidence suggested that the occurrence of PE is related to inflammatory factors. We reviewed the current understandings of roles of cytokines in PE, and provided an extensive overview of the role of single nucleotide chain polymorphisms (SNPs) in the genes potentially underlying the pathophysiology of PE.
Collapse
Affiliation(s)
- Chengcheng Guan
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Fei Zhao
- Department of gynecology and obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhencui Yang
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Qian Tang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ling Wang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xueli Li
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lixia Zhang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ziwen Deng
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Huabin Hou
- Clinical Laboratory, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jingli Wang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yinglei Xu
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ru Zhang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Lin
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ping Tan
- Department of gynecology and obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yan Zhang
- Department of gynecology and obstetrics, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shiguo Liu
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lu Zhang
- Medical genetic department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Prenatal Diagnosis Center, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
9
|
Zolfaghari MA, Arefnezhad R, Parhizkar F, Hejazi MS, Motavalli Khiavi F, Mahmoodpoor A, Yousefi M. T lymphocytes and preeclampsia: The potential role of T-cell subsets and related MicroRNAs in the pathogenesis of preeclampsia. Am J Reprod Immunol 2021; 86:e13475. [PMID: 34043850 DOI: 10.1111/aji.13475] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 05/11/2021] [Accepted: 05/22/2021] [Indexed: 12/13/2022] Open
Abstract
Innate and adaptive immune systems have a crucial role in initiating and progressing some pregnancy disorders such as preeclampsia (PE), which is one of the pregnancy-specific disorders that could result in neonatal and maternal morbidity and mortality. The dysregulation of the spiral artery and inadequate trophoblast invasion lead to PE symptoms through producing various inflammatory cytokines and anti-angiogenic factors from the placenta. T lymphocytes play a special role in the epithelium and stroma of the human endometrium. CD4+ T helper (Th) cells, Th1/Th2, and Th17/T regulatory (Treg) balance mainly contribute to the establishment of a pregnancy-favorable environment. This review examined the dysregulation of some cytokines produced from T cells, the dysregulation of the transcription factors of Th cells, the expression of chemokine receptors on T cells, as well as the effects of some factors including vitamin D on the activity of T cells, and finally, the dysregulation of various miRNAs related to T cells, which could cause PE.
Collapse
Affiliation(s)
- Mohammad Ali Zolfaghari
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Arefnezhad
- Department of Anatomy, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Forough Parhizkar
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Saeid Hejazi
- Department of Molecular Medicine, Faculty of Advanced Biomedical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Farhad Motavalli Khiavi
- Medical Biotechnology Research Center, AJA University of Medical Sciences, Tehran, Iran.,Pasteur Institute of Iran, Department of Virology, Tehran, Iran
| | - Ata Mahmoodpoor
- Department of Anesthesiology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Yousefi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
10
|
MiR-325-3p mediate the CXCL17/CXCR8 axis to regulate angiogenesis in hepatocellular carcinoma. Cytokine 2021; 141:155436. [PMID: 33515898 DOI: 10.1016/j.cyto.2021.155436] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 12/27/2020] [Accepted: 01/02/2021] [Indexed: 12/13/2022]
Abstract
INTRODUCTION MicroRNA-325-3p (miR-325-3p) is involved in the progression of a great number of tumors. However, the regulatory mechanism of miR-325-3p on hepatocellular carcinoma (HCC) remains unclear. AIM In this paper, we aim to investigate the underlying mechanism by which miR-325-3p regulate the progression of HCC. METHODS RT-qPCR was performed to detect the levels of miR-325-3p, CXCL17, and CXCR8. Western bolt was conducted to determine the levels of pro-angiogenic factors VEGF, FGF2, Ang-1 and PDGF-B. Immunohistochemistry was carried to detect the distribution and expression of Ki-67 and CD34 in HCC tissues. MTT and colony formation were carried to evaluate cell proliferation, endothelial tube-formation assay was used detect tubule formation, and transwell assay was performed to evaluate cell migration and invasion ability. Dual-luciferase activity assay was used to verify the relationship between miR-325-3p and CXCL17. RESULTS MiR-325-3p was down-regulated in HCC cells and tissues, miR-325-3p overexpression inhibited the proliferation, migration and invasion of HCC cells. Besides, miR-325-3p overexpression inhibited angiogenesis of HCC. CXCL17 is a direct target of miR-325-3p and partially mediates the effect of miR-325-3p on proliferation, migration, invasion and angiogenesis of HCC. CONCLUSION MiR-325-3p regulated angiogenesis of HCC via mediating CXCL17/CXCR8 axis, indicating miR-325-3p may serve as a promising therapy biomarker for HCC.
Collapse
|
11
|
Hao W, Li M, Pang Y, Du W, Huang X. Increased chemokines levels in patients with chronic obstructive pulmonary disease: correlation with quantitative computed tomography metrics. Br J Radiol 2020; 94:20201030. [PMID: 33237823 DOI: 10.1259/bjr.20201030] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
OBJECTIVE We sought to explore the relationships between multiple chemokines with spirometry, inflammatory mediators and CT findings of emphysema, small airways disease and bronchial wall thickness. METHODS All patients with COPD (n = 65) and healthy control subjects (n = 23) underwent high-resolution CT, with image analysis determining the low attenuation area (LAA), ratio of mean lung attenuation on expiratory and inspiratory scans (E/I MLD) and bronchial wall thickness of inner perimeter of a 10-mm diameter airway (Pi10). At enrollment, subjects underwent pulmonary function studies, chemokines and inflammatory mediators measurements. RESULTS Multiple chemokines (CCL2, CCL3, CCL5, CX3CL1, CXCL8, CXCL9, CXCL10, CXCL11 and CXCL12) and inflammatory mediators (MMP-9, MMP-12, IL-18 and neutrophil count) were markedly increased in the serum of COPD patients compared with healthy controls. There were associations between small airway disease (E/I MLD) and CCL11, CXCL8, CXCL10, CXCL11, CXCL12 and CX3CL1. Especially CXCL8 and CX3CL1 are strongly associated with E/I MLD (r = 0.74, p < 0.001; r = 0.76, p < 0.001, respectively). CXCL8, CXCL12 and CX3CL1 were moderately positively correlated with emphysema (%LAA) (r = 0.49, p < 0.05; r = 0.51, p < 0.05; r = 0.54, p < 0.01, respectively). Bronchial wall thickness (Pi10)showed no significant differences between the COPD and healthy controls,,but there was an association between Pi10 and FEV1% in COPD patients (r=-0.420, p = 0.048). Our statistical results showed that there were not any associations between airway wall thickness (Pi10) and chemokines. CONCLUSION Pulmonary chemokines levels are closely associated with the extent of gas trapping, small airways disease and emphysema identified on high-resolution chest CT scan. ADVANCES IN KNOWLEDGE This study combines quantitative CT analysis with multiplex chemokines and inflammatory mediators to identify a new role of pathological changes in COPD.
Collapse
Affiliation(s)
- Wendong Hao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'anJiaotong University, Xi'an, China.,Department of Respiratory Medicine, The Affiliated Hospital of Yan'an University, Yan'an, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'anJiaotong University, Xi'an, China
| | - Yamei Pang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Xi'anJiaotong University, Xi'an, China
| | - Weiping Du
- Clinical Laboratory Diagnosis Department, The Affiliated Hospital of Yan'an University, Yan'an, China
| | - Xiaoqi Huang
- Department of Radiology, The Affiliated Hospital of Yan'an University, Yan'an, China
| |
Collapse
|
12
|
Marinello WP, Mohseni ZS, Cunningham SJ, Crute C, Huang R, Zhang JJ, Feng L. Perfluorobutane sulfonate exposure disrupted human placental cytotrophoblast cell proliferation and invasion involving in dysregulating preeclampsia related genes. FASEB J 2020; 34:14182-14199. [PMID: 32901980 DOI: 10.1096/fj.202000716rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/15/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022]
Abstract
We reported that maternal PFBS, an emerging pollutant, exposure is positively associated with preeclampsia which can result from aberrant trophoblasts invasion and subsequent placental ischemia. In this study, we investigated the effects of PFBS on trophoblasts proliferation/invasion and signaling pathways. We exposed a human trophoblast line, HTR8/SVneo, to PFBS. Cell viability, proliferation, and cell cycle were evaluated by the MTS assay, Ki-67 staining, and flow cytometry, respectively. We assessed cell migration and invasion with live-cell imaging-based migration assay and matrigel invasion assay, respectively. Signaling pathways were examined by Western blot, RNA-seq, and qPCR. PFBS exposure interrupted cell proliferation and invasion in a dose-dependent manner. PFBS (100 μM) did not cause cell death but instead significant cell proliferation without cell cycle disruption. PFBS (10 and 100 μM) decreased cell migration and invasion, while PFBS (0.1 μM) significantly increased cell invasion but not migration. Further, RNA-seq analysis identified dysregulated HIF-1α target genes that are relevant to cell proliferation/invasion and preeclampsia, while Western Blot data showed the activation of HIF-1α, but not Notch, ERK1/2, (PI3K)AKT, and P38 pathways. PBFS exposure altered trophoblast cell proliferation/invasion which might be mediated by preeclampsia-related genes, suggesting a possible association between prenatal PFBS exposure and adverse placentation.
Collapse
Affiliation(s)
- William P Marinello
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Zahra S Mohseni
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA
| | - Sarah J Cunningham
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Christine Crute
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,Integrated Toxicology and Environmental Health Program, Nicholas School of the Environment, Duke University, Durham, NC, USA
| | - Rong Huang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Jun J Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, USA.,MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
13
|
Cruz JDO, Conceição IMCA, Tosatti JAG, Gomes KB, Luizon MR. Global DNA methylation in placental tissues from pregnant with preeclampsia: A systematic review and pathway analysis. Placenta 2020; 101:97-107. [PMID: 32942147 DOI: 10.1016/j.placenta.2020.09.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 08/24/2020] [Accepted: 09/01/2020] [Indexed: 12/28/2022]
Abstract
Pre-eclampsia (PE) is the major cause of fetal and maternal mortality and can be classified according to gestational age of onset into early-onset (EOPE, <34 weeks of gestation) and late- (LOPE, ≥34 weeks of gestation). DNA methylation (DNAm) may help to understand the abnormal placentation in PE. Therefore, we performed a systematic review to assess the role of global DNAm on pathophysiology of PE, focused on fetal and maternal tissues of placenta from pregnant with PE, including EOPE and LOPE. We searched the databases EMBASE, Medline/PubMed, Cochrane Central Register of Controlled Trials, Scopus, Lilacs, Scielo and Google Scholar, and followed the MOOSE guidelines. Moreover, we performed pathway analysis with the overlapping genes from the included studies. Twelve out of 24 included studies in the qualitative analysis considered the classification into EOPE and LOPE. We did not found heterogeneity in the criteria used for diagnosis of PE, and a few studies evaluated whether confounding factors would influence placental DNAm. Fourteen out of 24 included studies showed hypomethylation in placental tissue from pregnant with PE compared to controls. The differences in DNAm are specific to genes or differentially methylated regions, and more evident in EOPE and preterm PE compared to controls, rather than LOPE and term PE. The overlapping genes from included studies revealed pathways relevant to pathophysiology of PE. Our findings highlighted the heterogeneous results of the included studies, mainly focused on North America and China. Replication studies in different populations should use the same placental tissues, techniques to assess DNAm and pipelines for bioinformatic analysis.
Collapse
Affiliation(s)
- Juliana de O Cruz
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Izabela M C A Conceição
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Jéssica A G Tosatti
- Department of Clinical and Toxicological Analyzes, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Karina B Gomes
- Department of Clinical and Toxicological Analyzes, Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil
| | - Marcelo R Luizon
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil; Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, 31270-901, Brazil.
| |
Collapse
|
14
|
van Boeckel SR, Macpherson H, Norman JE, Davidson DJ, Stock SJ. Inflammation-mediated generation and inflammatory potential of human placental cell-free fetal DNA. Placenta 2020; 93:49-55. [PMID: 32250739 PMCID: PMC7146537 DOI: 10.1016/j.placenta.2020.02.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 02/21/2020] [Accepted: 02/22/2020] [Indexed: 11/16/2022]
Abstract
Introduction Circulating DNA can be pro-inflammatory when detected by leukocytes via toll-like receptor 9 (TLR9). Cell-free fetal DNA (cff-DNA) of placental origin, circulates in pregnancy, and increased concentrations are seen in conditions associated with placental and maternal inflammation such as pre-eclampsia. However, whether cff-DNA is directly pro-inflammatory in pregnant women and what regulates cff-DNA levels in pregnancy are unknown. Methods Using a human term placental explant model, we examined whether induction of placental inflammation can promote cff-DNA release, and the capacity of this cff-DNA to stimulate peripheral blood mononuclear cells (PBMCs) from pregnant women. Results We demonstrate lipopolysaccharide (LPS)-mediated inflammation in placental explants and induced apoptosis after 24 h. However, this did not increase levels of cff-DNA generation compared to controls. Furthermore, the methylation status of the cff-DNA, was not altered by LPS-induced inflammation. Cff-DNA did not elicit production of inflammatory cytokines from PBMCs, in contrast to exposure to LPS or the TLR9 agonist CpG-ODN. Finally, we demonstrate that cff-DNA acquired directly from pregnant women did not differ in methylation status from placental extracted DNA, or from placental explant generated cell-free DNA, and that, unlike Escherichia coli DNA, this cff-DNA has a low level of unmethylated CpG sequences. Discussion Our data suggest that placental inflammation does not increase release of cff-DNA and that placental cff-DNA is not pro-inflammatory to circulating PBMCs. It thus seems unlikely that high levels of cff-DNA are either a direct consequence or cause of inflammation observed in obstetric complications. Cell-free fetal DNA was generated using a human placental explant model. Lipopolysaccharide causes inflammation and cell death in placental explants. Inflammation does not increase cell-free fetal DNA release from placental explants. Generated DNA does not elicit inflammation from blood cells from pregnant women.
Collapse
Affiliation(s)
- Sara R van Boeckel
- Tommy's Centre for Maternal and Fetal Health at the MRC Centre for Reproductive Health, University of Edinburgh, QMRI, Edinburgh, United Kingdom.
| | - Heather Macpherson
- Tommy's Centre for Maternal and Fetal Health at the MRC Centre for Reproductive Health, University of Edinburgh, QMRI, Edinburgh, United Kingdom
| | - Jane E Norman
- Tommy's Centre for Maternal and Fetal Health at the MRC Centre for Reproductive Health, University of Edinburgh, QMRI, Edinburgh, United Kingdom
| | - Donald J Davidson
- University of Edinburgh Centre for Inflammation Research, QMRI, Edinburgh, United Kingdom
| | - Sarah J Stock
- Tommy's Centre for Maternal and Fetal Health at the MRC Centre for Reproductive Health, University of Edinburgh, QMRI, Edinburgh, United Kingdom; Usher Institute, University of Edinburgh NINE Edinburgh BioQuarter, 9 Little France Road, Edinburgh, EH16 4UX, United Kingdom
| |
Collapse
|
15
|
Darakhshan S, Fatehi A, Hassanshahi G, Mahmoodi S, Hashemi MS, Karimabad MN. Serum concentration of angiogenic (CXCL1, CXCL12) and angiostasis (CXCL9, CXCL10) CXC chemokines are differentially altered in normal and gestational diabetes mellitus associated pregnancies. J Diabetes Metab Disord 2019; 18:371-378. [PMID: 31890662 PMCID: PMC6915176 DOI: 10.1007/s40200-019-00421-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 06/28/2019] [Indexed: 02/07/2023]
Abstract
BACKGROUND The present study was aimed and designed to determine the serum levels of CXCL1 and CXCL12 as angiogenesis along with CXCL9 and CXCL10 as angiostasis, chemokines in, Gestational diabetes mellitus mothers (GDMM) and normal pregnancy mothers (NPM) and neonates who delivered by them. METHODS We have recruited 63 pregnant GDMM and 63 normal pregnant mothers at the third trimester of pregnancy to this cross-sectional study. Cord blood specimens were obtained from neonates who were delivered from GDMM and NPM. The serum and cord blood levels of chemokines were measured by ELISA in studied groups. Data were analyzed by chi-square and student's t test between two groups. The P-values less than 0.05 were considered significant. RESULTS Our results revealed that the serum levels of CXCL1, CXCL9 and CXCL12 were increased in GDMM, while no alteration was found in the serum levels of CXCL10 when compared to NPM. We have observed that in neonates the serum levels of angiogeneic chemokines followed an inverse fashion when compared to angiostasis chemokines. Interestingly, CXCL1 and CXCL12 were both increased in neonates who were delivered by GDMM, while, CXCL9 and CXCL10 were decreased in neonates delivered by GDMM.
Collapse
Affiliation(s)
- Shokoofeh Darakhshan
- Department of Pediatrics, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Abbas Fatehi
- Department of Pediatrics, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamhossein Hassanshahi
- Department of Hematology, Faculty of Biomed, Biomedical Sciences Kerman University of Medical Sciences, Kerman, Iran
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Soodabeh Mahmoodi
- Department of Pediatrics, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Monireh Seyed Hashemi
- Department of Pediatrics, Faculty of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mojgan Noroozi Karimabad
- Molecular Medicine Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|