1
|
Ishiyama A, Suda K, Rao X, Sun M, Lane GJ, Yamataka A, Koga H. Angiopoietin-1 attenuates lipopolysaccharide-induced endotoxemia in a Hirschsprung's disease murine model by improving intestinal vascular integrity: implications for treating postoperative Hirschsprung-associated enterocolitis. Pediatr Surg Int 2024; 40:277. [PMID: 39466437 DOI: 10.1007/s00383-024-05867-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/18/2024] [Indexed: 10/30/2024]
Abstract
PURPOSE Angiopoietin-1 (Ang1) mitigates inflammation as a proangiogenic growth factor. Action of Ang1 on lipopolysaccharide (LPS)-induced endotoxemic inflammation was investigated in endothelin receptor-B null Hirschsprung's disease mice (KO). METHODS LPS or saline was injected intraperitoneally in KO (KO-LPS; n = 9, KO-sal; n = 5) and wild-type (WT) (WT-LPS; n = 6, WT-sal; n = 6) pups obtained within 24 h of birth. Normoganglionic terminal ileum harvested 6 h after LPS was used for RNA extraction and histology. IL-1β, SELE, VEGFA, Ang1, Angiopoietin-2 (Ang2), and TIE2 expression analyzed by quantitative polymerase chain reaction (qPCR), vascular permeability assessed by the Miles assay, severity of inflammation, and immunofluorescence for phospho-TIE2 and VE-cadherin were used to assess endothelial cell contact integrity and compared with KO pups pretreated with intraperitoneal Ang1 [Ang1(KO-LPS); n = 5] or saline [sal(KO-LPS); n = 6] 2 h before LPS. RESULTS KO-LPS pups showed significantly increased inflammation (p < 0.05) and expression of IL-1β, SELE, VEGFA, and Ang2 (p = 0.019, 0.003, 0.008 and < 0.0001, respectively); expression of Ang1 and TIE2 remained unchanged when compared with KO-saline. In Ang1(KO-LPS) ileum, changes seen in sal(KO-LPS) were eliminated and phospho-TIE2 and VE-cadherin fluorescence increased. CONCLUSION Ang1 successfully attenuated LPS-induced normoganglionic intestinal inflammation, downregulated pro-inflammatory genes, and improved vascular barrier integrity in KO pups.
Collapse
Affiliation(s)
- Asuka Ishiyama
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Kazuto Suda
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan.
| | - Xuxuan Rao
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Minhua Sun
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Geoffrey J Lane
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Atsuyuki Yamataka
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Hiroyuki Koga
- Department of Pediatric General and Urogenital Surgery, Juntendo University School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| |
Collapse
|
2
|
Silva Z, Soares CO, Barbosa M, Palma AS, Marcelo F, Videira PA. The role of sialoglycans in modulating dendritic cell function and tumour immunity. Semin Immunol 2024; 74-75:101900. [PMID: 39461124 DOI: 10.1016/j.smim.2024.101900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/15/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
Dendritic cells (DCs) are crucial for initiating immune responses against tumours by presenting antigens to T cells. Glycosylation, particularly sialylation, plays a significant role in regulating cell functions, by modulating protein folding and signalling. This review aimed to provide a comprehensive overview of how sialic acids influence key aspects of DC biology, including maturation, migration, antigen presentation, and T cell interactions. Sialic acids influence DC endocytosis, affecting their ability to uptake and present antigens, while guiding their migration to lymph nodes and inflamed tissues. Removing sialic acids enhances DC-mediated antigen presentation to T cells, potentially boosting immune responses. Additionally, sialylated glycans on DCs modulate immune checkpoints, which can impact tumour immunity. Hypersialylation of tumour mucins further promotes immune evasion by interacting with DCs. Understanding the interplay between sialylation and DC functions offers promising avenues for enhancing cancer immunotherapy.
Collapse
Affiliation(s)
- Zélia Silva
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Cátia O Soares
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Mariana Barbosa
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Angelina S Palma
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Filipa Marcelo
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal
| | - Paula A Videira
- Institute for Health and Bioeconomy (i4HB), NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal; CDG & Allies - Professionals and Patient Associations International Network (CDG & Allies - PPAIN), Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Caparica 2829-516, Portugal.
| |
Collapse
|
3
|
Avdonin PP, Blinova MS, Serkova AA, Komleva LA, Avdonin PV. Immunity and Coagulation in COVID-19. Int J Mol Sci 2024; 25:11267. [PMID: 39457048 PMCID: PMC11508857 DOI: 10.3390/ijms252011267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/23/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Discovered in late 2019, the SARS-CoV-2 coronavirus has caused the largest pandemic of the 21st century, claiming more than seven million lives. In most cases, the COVID-19 disease caused by the SARS-CoV-2 virus is relatively mild and affects only the upper respiratory tract; it most often manifests itself with fever, chills, cough, and sore throat, but also has less-common mild symptoms. In most cases, patients do not require hospitalization, and fully recover. However, in some cases, infection with the SARS-CoV-2 virus leads to the development of a severe form of COVID-19, which is characterized by the development of life-threatening complications affecting not only the lungs, but also other organs and systems. In particular, various forms of thrombotic complications are common among patients with a severe form of COVID-19. The mechanisms for the development of thrombotic complications in COVID-19 remain unclear. Accumulated data indicate that the pathogenesis of severe COVID-19 is based on disruptions in the functioning of various innate immune systems. The key role in the primary response to a viral infection is assigned to two systems. These are the pattern recognition receptors, primarily members of the toll-like receptor (TLR) family, and the complement system. Both systems are the first to engage in the fight against the virus and launch a whole range of mechanisms aimed at its rapid elimination. Normally, their joint activity leads to the destruction of the pathogen and recovery. However, disruptions in the functioning of these innate immune systems in COVID-19 can cause the development of an excessive inflammatory response that is dangerous for the body. In turn, excessive inflammation entails activation of and damage to the vascular endothelium, as well as the development of the hypercoagulable state observed in patients seriously ill with COVID-19. Activation of the endothelium and hypercoagulation lead to the development of thrombosis and, as a result, damage to organs and tissues. Immune-mediated thrombotic complications are termed "immunothrombosis". In this review, we discuss in detail the features of immunothrombosis associated with SARS-CoV-2 infection and its potential underlying mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Pavel V. Avdonin
- Koltzov Institute of Developmental Biology RAS, ul. Vavilova, 26, 119334 Moscow, Russia; (P.P.A.)
| |
Collapse
|
4
|
Bhavani SV, Spicer A, Sinha P, Malik A, Lopez-Espina C, Schmalz L, Watson GL, Bhargava A, Khan S, Urdiales D, Updike L, Dagan A, Davila H, Demarco C, Evans N, Gosai F, Iyer K, Kurtzman N, Palagiri AV, Sims M, Smith S, Syed A, Sarma D, Reddy B, Verhoef PA, Churpek MM. Distinct immune profiles and clinical outcomes in sepsis subphenotypes based on temperature trajectories. Intensive Care Med 2024:10.1007/s00134-024-07669-0. [PMID: 39382693 DOI: 10.1007/s00134-024-07669-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 09/21/2024] [Indexed: 10/10/2024]
Abstract
PURPOSE Sepsis is a heterogeneous syndrome. Identification of sepsis subphenotypes with distinct immune profiles could lead to targeted therapies. This study investigates the immune profiles of patients with sepsis following distinct body temperature patterns (i.e., temperature trajectory subphenotypes). METHODS Hospitalized patients from four hospitals between 2018 and 2022 with suspicion of infection were included. A previously validated temperature trajectory algorithm was used to classify study patients into temperature trajectory subphenotypes. Microbiological profiles, clinical outcomes, and levels of 31 biomarkers were compared between these subphenotypes. RESULTS The 3576 study patients were classified into four temperature trajectory subphenotypes: hyperthermic slow resolvers (N = 563, 16%), hyperthermic fast resolvers (N = 805, 23%), normothermic (N = 1693, 47%), hypothermic (N = 515, 14%). The mortality rate was significantly different between subphenotypes, with the highest rate in hypothermics (14.2%), followed by hyperthermic slow resolvers 6%, normothermic 5.5%, and lowest in hyperthermic fast resolvers 3.6% (p < 0.001). After multiple testing correction for the 31 biomarkers tested, 20 biomarkers remained significantly different between temperature trajectories: angiopoietin-1 (Ang-1), C-reactive protein (CRP), feline McDonough sarcoma-like tyrosine kinase 3 ligand (Flt-3l), granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage colony stimulating factor (GM-CSF), interleukin (IL)-15, IL-1 receptor antagonist (RA), IL-2, IL-6, IL-7, interferon gamma-induced protein 10 (IP-10), monocyte chemoattractant protein-1 (MCP-1), human macrophage inflammatory protein 3 alpha (MIP-3a), neutrophil gelatinase-associated lipocalin (NGAL), pentraxin-3, thrombomodulin, tissue factor, soluble triggering receptor expressed on myeloid cells-1 (sTREM-1), and vascular cellular adhesion molecule-1 (vCAM-1).The hyperthermic fast and slow resolvers had the highest levels of most pro- and anti-inflammatory cytokines. Hypothermics had suppressed levels of most cytokines but the highest levels of several coagulation markers (Ang-1, thrombomodulin, tissue factor). CONCLUSION Sepsis subphenotypes identified using the universally available measurement of body temperature had distinct immune profiles. Hypothermic patients, who had the highest mortality rate, also had the lowest levels of most pro- and anti-inflammatory cytokines.
Collapse
Affiliation(s)
- Sivasubramanium V Bhavani
- School of Medicine, Emory University, Atlanta, GA, USA.
- Emory Critical Care Center, Atlanta, GA, USA.
| | - Alexandra Spicer
- Department of Medicine, University of Wisconsin, Madison, WI, USA
| | - Pratik Sinha
- School of Medicine, Washington University, St. Louis, MO, USA
| | - Albahi Malik
- School of Medicine, Emory University, Atlanta, GA, USA
| | | | | | | | | | | | | | | | - Alon Dagan
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | | | - Neil Evans
- Davis School of Medicine, University of California, Sacramento, CA, USA
| | - Falgun Gosai
- OSF Saint Francis Medical Center, Peoria, IL, USA
| | | | - Niko Kurtzman
- School of Medicine, Emory University, Atlanta, GA, USA
| | | | | | | | | | - Deesha Sarma
- Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | | | - Philip A Verhoef
- University of Hawaii John A. Burns School of Medicine, Honolulu, HI, USA
| | | |
Collapse
|
5
|
Moser S, Araschmid L, Panagiotou A, Bonati LH, Breidthardt T, Fahrni G, Kaiser C, Jeger R, Trendelenburg M, Osthoff M. Association of Endothelial Cell Activation with Acute Kidney Injury during Coronary Angiography and the Influence of Recombinant Human C1 Inhibitor-A Secondary Analysis of a Randomized, Placebo-Controlled, Double-Blind Trial. Biomedicines 2024; 12:1956. [PMID: 39335470 PMCID: PMC11428207 DOI: 10.3390/biomedicines12091956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Acute kidney injury (AKI) as a result of iodinated contrast media (CM) has been linked to CM-induced renal ischemia and toxic effects on endothelial cells (EC). The recombinant human C1 inhibitor (rhC1INH) has been shown to influence EC activation. METHODS Secondary analysis of 74/77 (96%) participants of a double-blind, randomized, and placebo-controlled study that assessed the effect of rhC1INH on AKI. E-selectin, intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule (VCAM-1), and CC-chemokin-ligand-5 (CCL5) were determined in frozen blood samples over 48 h and analyzed according to the treatment group and renal outcomes. RESULTS The mean age was 76.7 years, and 37 patients each received rhC1INH and placebo, respectively. In the entire study population, minor differences in median EC activation markers/CCL5 concentrations during the first 48 h compared to baseline were observed (e.g., E-selectin 27.5 ng/mL at baseline vs. 29.7 ng/mL on day 1, CCL5: 17.7 ng/mL at baseline vs. 32.2 ng/mL on day 2). Absolute changes in ICAM-1/E-selectin concentrations correlated with a higher peak change in urinary NGAL concentrations. However, AKI was not associated with significant changes in EC markers/CCL5. Last, no significant differences in serum concentrations of EC activation markers/CCL5 were evident between the placebo and the rhC1INH group. CONCLUSIONS CM administration during coronary angiography only mildly activated ECs within the first 48 h, which does not explain subsequent AKI. The administration of rhC1INH was not associated with a reduction of EC activation or CCL5.
Collapse
Affiliation(s)
- Stephan Moser
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
| | - Laura Araschmid
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Anneza Panagiotou
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Leo H. Bonati
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
- Research Department, Reha Rheinfelden, 4310 Rheinfelden, Switzerland
| | - Tobias Breidthardt
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
| | - Gregor Fahrni
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
- Department of Cardiology, Stadtspital Triemli, 8063 Zürich, Switzerland
| | - Christoph Kaiser
- Department of Cardiology, University Hospital Basel, 4031 Basel, Switzerland
| | - Raban Jeger
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
- Department of Cardiology, Stadtspital Triemli, 8063 Zürich, Switzerland
| | - Marten Trendelenburg
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
| | - Michael Osthoff
- Division of Internal Medicine, University Hospital Basel, 4031 Basel, Switzerland
- Department of Clinical Research, University of Basel, 4001 Basel, Switzerland
- Division of General Internal Medicine, Cantonal Hospital Winterthur, 8400 Winterthur, Switzerland
| |
Collapse
|
6
|
Barai A, Piplani N, Saha SK, Dutta S, Gomathi V, Ghogale MM, Kumar S, Kulkarni M, Sen S. Bulky glycocalyx drives cancer invasiveness by modulating substrate-specific adhesion. PNAS NEXUS 2024; 3:pgae335. [PMID: 39211517 PMCID: PMC11358709 DOI: 10.1093/pnasnexus/pgae335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 07/30/2024] [Indexed: 09/04/2024]
Abstract
The majority of the eukaryotic cell surface is decorated with a layer of membrane-attached polysaccharides and glycoproteins collectively referred to as the glycocalyx. While the formation of a bulky glycocalyx has been associated with the cancer progression, the mechanisms by which the glycocalyx regulates cancer invasiveness are incompletely understood. We address this question by first documenting subtype-specific expression of the major glycocalyx glycoprotein Mucin-1 (MUC1) in breast cancer patient samples and breast cancer cell lines. Strikingly, glycocalyx disruption led to inhibition of 2D motility, loss of 3D invasion, and reduction of clonal scattering in breast cancer cells at the population level. Tracking of 2D cell motility and 3D invasiveness of MUC1-based sorted subpopulations revealed the fastest motility and invasiveness in intermediate MUC1-expressing cells, with glycocalyx disruption abolishing these effects. While differential sensitivity in 2D motility is attributed to a nonmonotonic dependence of focal adhesion size on MUC1 levels, higher MUC1 levels enhance 3D invasiveness via increased traction generation. In contrast to inducing cell rounding on collagen-coated substrates, high MUC1 level promotes cell adhesion and confers resistance to shear flow on substrates coated with the endothelial surface protein E-selectin. Collectively, our findings illustrate how MUC1 drives cancer invasiveness by differentially regulating cell-substrate adhesion in a substrate-dependent manner.
Collapse
Affiliation(s)
- Amlan Barai
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Niyati Piplani
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Sumon Kumar Saha
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Sarbajeet Dutta
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - V Gomathi
- Center for Translational Cancer Research, IISER Pune and PCCM Pune, Pune 411008, India
| | - Mayank M Ghogale
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Sushil Kumar
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| | - Madhura Kulkarni
- Center for Translational Cancer Research, IISER Pune and PCCM Pune, Pune 411008, India
| | - Shamik Sen
- Department of Biosciences and Bioengineering, IIT Bombay, Mumbai 400076, India
| |
Collapse
|
7
|
Zhang J, Huang S, Zhu Z, Gatt A, Liu J. E-selectin in vascular pathophysiology. Front Immunol 2024; 15:1401399. [PMID: 39100681 PMCID: PMC11294169 DOI: 10.3389/fimmu.2024.1401399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/05/2024] [Indexed: 08/06/2024] Open
Abstract
Selectins are a group of Ca2+-dependent, transmembrane type I glycoproteins which attract cell adhesion and migration. E-selectin is exclusively expressed in endothelial cells, and its expression is strongly enhanced upon activation by pro-inflammatory cytokines. The interaction of E-selectin with its ligands on circulating leukocytes captures and slows them down, further facilitating integrin activation, firm adhesion to endothelial cells and transmigration to tissues. Oxidative stress induces endothelial cell injury, leading to aberrant expression of E-selectin. In addition, the elevated level of E-selectin is positively related to high risk of inflammation. Dysregulation of E-selectin has been found in several pathological conditions including acute kidney injury (AKI), pulmonary diseases, hepatic pathology, Venous thromboembolism (VTE). Deletion of the E-selectin gene in mice somewhat ameliorates these complications. In this review, we describe the mechanisms regulating E-selectin expression, the interaction of E-selectin with its ligands, the E-selectin physiological and pathophysiological roles, and the therapeutical potential of targeting E-selectin.
Collapse
Affiliation(s)
- Jinjin Zhang
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Shengshi Huang
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Jinan, China
| | - Zhiying Zhu
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Alex Gatt
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Msida, Malta
- Haematology Laboratory, Department of Pathology, Mater Dei Hospital, Msida, Malta
| | - Ju Liu
- Department of Laboratory Medicine, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Key Medical and Health Laboratory of Translational Medicine in Microvascular Aging, Jinan, China
| |
Collapse
|
8
|
Haage TR, Charakopoulos E, Bhuria V, Baldauf CK, Korthals M, Handschuh J, Müller P, Li J, Harit K, Nishanth G, Frey S, Böttcher M, Fischer KD, Dudeck J, Dudeck A, Lipka DB, Schraven B, Green AR, Müller AJ, Mougiakakos D, Fischer T. Neutrophil-specific expression of JAK2-V617F or CALRmut induces distinct inflammatory profiles in myeloproliferative neoplasia. J Hematol Oncol 2024; 17:43. [PMID: 38853260 PMCID: PMC11163796 DOI: 10.1186/s13045-024-01562-5] [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: 03/22/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024] Open
Abstract
BACKGROUND Neutrophils play a crucial role in inflammation and in the increased thrombotic risk in myeloproliferative neoplasms (MPNs). We have investigated how neutrophil-specific expression of JAK2-V617F or CALRdel re-programs the functions of neutrophils. METHODS Ly6G-Cre JAK2-V617F and Ly6G-Cre CALRdel mice were generated. MPN parameters as blood counts, splenomegaly and bone marrow histology were compared to wild-type mice. Megakaryocyte differentiation was investigated using lineage-negative bone marrow cells upon in vitro incubation with TPO/IL-1β. Cytokine concentrations in serum of mice were determined by Mouse Cytokine Array. IL-1α expression in various hematopoietic cell populations was determined by intracellular FACS analysis. RNA-seq to analyse gene expression of inflammatory cytokines was performed in isolated neutrophils from JAK2-V617F and CALR-mutated mice and patients. Bioenergetics of neutrophils were recorded on a Seahorse extracellular flux analyzer. Cell motility of neutrophils was monitored in vitro (time lapse microscopy), and in vivo (two-photon microscopy) upon creating an inflammatory environment. Cell adhesion to integrins, E-selectin and P-selection was investigated in-vitro. Statistical analysis was carried out using GraphPad Prism. Data are shown as mean ± SEM. Unpaired, two-tailed t-tests were applied. RESULTS Strikingly, neutrophil-specific expression of JAK2-V617F, but not CALRdel, was sufficient to induce pro-inflammatory cytokines including IL-1 in serum of mice. RNA-seq analysis in neutrophils from JAK2-V617F mice and patients revealed a distinct inflammatory chemokine signature which was not expressed in CALR-mutant neutrophils. In addition, IL-1 response genes were significantly enriched in neutrophils of JAK2-V617F patients as compared to CALR-mutant patients. Thus, JAK2-V617F positive neutrophils, but not CALR-mutant neutrophils, are pathogenic drivers of inflammation in MPN. In line with this, expression of JAK2-V617F or CALRdel elicited a significant difference in the metabolic phenotype of neutrophils, suggesting a stronger inflammatory activity of JAK2-V617F cells. Furthermore, JAK2-V617F, but not CALRdel, induced a VLA4 integrin-mediated adhesive phenotype in neutrophils. This resulted in reduced neutrophil migration in vitro and in an inflamed vessel. This mechanism may contribute to the increased thrombotic risk of JAK2-V617F patients compared to CALR-mutant individuals. CONCLUSIONS Taken together, our findings highlight genotype-specific differences in MPN-neutrophils that have implications for the differential pathophysiology of JAK2-V617F versus CALR-mutant disease.
Collapse
Affiliation(s)
- Tobias Ronny Haage
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Emmanouil Charakopoulos
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Vikas Bhuria
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
| | - Conny K Baldauf
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Mark Korthals
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Juliane Handschuh
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Müller
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Juan Li
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, GB, England
| | - Kunjan Harit
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Gopala Nishanth
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Stephanie Frey
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Martin Böttcher
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Klaus-Dieter Fischer
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Biochemistry and Cell Biology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Jan Dudeck
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Anne Dudeck
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniel B Lipka
- Section of Translational Cancer Epigenomics, Division of Translational Medical Oncology, German Cancer Research Center (DKFZ), National Center for Tumor Diseases (NCT) Heidelberg, Heidelberg, Germany
- Faculty of Medicine, Otto-von-Guericke University, Magdeburg, Germany
| | - Burkhart Schraven
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
| | - Anthony R Green
- Cambridge Stem Cell Institute, Department of Haematology, University of Cambridge, Cambridge, GB, England
| | - Andreas J Müller
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dimitrios Mougiakakos
- Department of Hematology, Oncology, and Cell Therapy, Medical Faculty, Otto-von-Guericke University, Leipziger Str. 44, 39120, Magdeburg, Germany
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany
| | - Thomas Fischer
- Healthcampus Immunology, Inflammation and Infectiology (GC-I, Otto-von-Guericke-University, Magdeburg, Germany.
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
- Center for Health and Medical Prevention - CHaMP, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
9
|
Shenoy US, Adiga D, Alhedyan F, Kabekkodu SP, Radhakrishnan R. HOXA9 transcription factor is a double-edged sword: from development to cancer progression. Cancer Metastasis Rev 2024; 43:709-728. [PMID: 38062297 PMCID: PMC11156722 DOI: 10.1007/s10555-023-10159-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/30/2023] [Indexed: 04/02/2024]
Abstract
The HOXA9 transcription factor serves as a molecular orchestrator in cancer stemness, epithelial-mesenchymal transition (EMT), metastasis, and generation of the tumor microenvironment in hematological and solid malignancies. However, the multiple modes of regulation, multifaceted functions, and context-dependent interactions responsible for the dual role of HOXA9 as an oncogene or tumor suppressor in cancer remain obscure. Hence, unravelling its molecular complexities, binding partners, and interacting signaling molecules enables us to comprehend HOXA9-mediated transcriptional programs and molecular crosstalk. However, it is imperative to understand its central role in fundamental biological processes such as embryogenesis, foetus implantation, hematopoiesis, endothelial cell proliferation, and tissue homeostasis before designing targeted therapies. Indeed, it presents an enormous challenge for clinicians to selectively target its oncogenic functions or restore tumor-suppressive role without altering normal cellular functions. In addition to its implications in cancer, the present review also focuses on the clinical applications of HOXA9 in recurrence and drug resistance, which may provide a broader understanding beyond oncology, open new avenues for clinicians for accurate diagnoses, and develop personalized treatment strategies. Furthermore, we have also discussed the existing therapeutic options and accompanying challenges in HOXA9-targeted therapies in different cancer types.
Collapse
Affiliation(s)
- U Sangeetha Shenoy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Divya Adiga
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Faisal Alhedyan
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj, 11942, Saudi Arabia
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom
| | - Shama Prasada Kabekkodu
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal Academy of Higher Education, Manipal, 576104, India.
- Unit of Oral and Maxillofacial Pathology, School of Clinical Dentistry, The University of Sheffield, Sheffield, United Kingdom.
| |
Collapse
|
10
|
Bartold M, Ivanovski S. Biological processes and factors involved in soft and hard tissue healing. Periodontol 2000 2024. [PMID: 38243683 DOI: 10.1111/prd.12546] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 11/12/2023] [Accepted: 11/23/2023] [Indexed: 01/21/2024]
Abstract
Wound healing is a complex and iterative process involving myriad cellular and biologic processes that are highly regulated to allow satisfactory repair and regeneration of damaged tissues. This review is intended to be an introductory chapter in a volume focusing on the use of platelet concentrates for tissue regeneration. In order to fully appreciate the clinical utility of these preparations, a sound understanding of the processes and factors involved in soft and hard tissue healing. This encompasses an appreciation of the cellular and biological mediators of both soft and hard tissues in general as well as specific consideration of the periodontal tissues. In light of good advances in this basic knowledge, there have been improvements in clinical strategies and therapeutic management of wound repair and regeneration. The use of platelet concentrates for tissue regeneration offers one such strategy and is based on the principles of cellular and biologic principles of wound repair discussed in this review.
Collapse
Affiliation(s)
- Mark Bartold
- University of Queensland, Brisbane, Queensland, Australia
| | - Saso Ivanovski
- University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
Mostmans Y, Maurer M, Richert B, Smith V, Melsens K, De Maertelaer V, Saidi I, Corazza F, Michel O. Chronic spontaneous urticaria: Evidence of systemic microcirculatory changes. Clin Transl Allergy 2024; 14:e12335. [PMID: 38282194 PMCID: PMC10821744 DOI: 10.1002/clt2.12335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/24/2023] [Accepted: 01/18/2024] [Indexed: 01/30/2024] Open
Abstract
BACKGROUND Chronic spontaneous urticaria (CSU) is a chronic inflammatory skin disease where activation of endothelial cells (ECs) at sites of skin lesions leads to increased blood flow, leakage of fluid into the skin, cellular infiltration, and vascular remodeling. To understand the disease duration and the sometimes vague systemic symptoms accompanying flares, the objective of this study was to examine if CSU comes with systemic vascular changes at the microcirculatory level. METHODS We investigated CSU patients (n = 49) and healthy controls (HCs, n = 44) for microcirculatory differences by nailfold videocapillaroscopy (NVC) and for blood levels of the soluble EC biomarkers serum vascular endothelial growth factor (VEGF), soluble E-selectin, and stem cell factor (SCF). Patients were also assessed for clinical characteristics, disease activity, and markers of autoimmune CSU (aiCSU). RESULTS CSU patients had significantly lower capillary density, more capillary malformations, and more irregular capillary dilations than HCs on NVC. Serum levels of VEGF, soluble E selectin and SCF were similar in CSU patients and HCs. CSU patients with higher VEGF levels had significantly more abnormal capillaries. Patients with markers of aiCSU, that is, low IgE levels or increased anti-TPO levels, had significantly more capillaries and less capillary dilations than those without. CONCLUSION Our results suggest that CSU comes with systemic microcirculatory changes, which may be driven, in part, by VEGF.
Collapse
Affiliation(s)
- Yora Mostmans
- Department of Immunology‐AllergologyCHU BrugmannUniversité Libre de Bruxelles (ULB)LakenBelgium
- Department of DermatologyCHU BrugmannULBLakenBelgium
| | - Marcus Maurer
- Institute of AllergologyCharité–Universitätsmedizin BerlinCorporate Member of Freie Universität Berlin and Humboldt‐Universität zu BerlinBerlinGermany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMPAllergology and ImmunologyBerlinGermany
| | | | - Vanessa Smith
- Department of Internal MedicineGhent UniversityGhentBelgium
- Department of RheumatologyGhent University HospitalGhentBelgium
- Unit for Molecular Immunology and InflammationVIB Inflammation Research Center (IRC)GhentBelgium
| | - Karin Melsens
- Department of Internal MedicineGhent UniversityGhentBelgium
- Department of RheumatologyGhent University HospitalGhentBelgium
- Unit for Molecular Immunology and InflammationVIB Inflammation Research Center (IRC)GhentBelgium
- Department of RheumatologyVrije Universiteit Brussel (VUB)Universitair Ziekenhuis Brussel (UZ Brussel)BrusselsBelgium
| | | | - Ines Saidi
- Department of DermatologyCHU BrugmannULBLakenBelgium
| | - Francis Corazza
- Department of ImmunologyLaboratoire Hospitalier Universitaire de Bruxelles – Universitair Laboratorium Brussels (LHUB‐ULB)BrusselsBelgium
| | - Olivier Michel
- Department of Immunology‐AllergologyCHU BrugmannUniversité Libre de Bruxelles (ULB)LakenBelgium
| |
Collapse
|
12
|
Rajan SS, Chandran R, Abrahamse H. Overcoming challenges in cancer treatment: Nano-enabled photodynamic therapy as a viable solution. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1942. [PMID: 38456341 DOI: 10.1002/wnan.1942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/09/2024] [Accepted: 01/17/2024] [Indexed: 03/09/2024]
Abstract
Cancer presents a formidable challenge, necessitating innovative therapies that maximize effectiveness while minimizing harm to healthy tissues. Nanotechnology has emerged as a transformative force in cancer treatment, particularly through nano-enabled photodynamic therapy (NE-PDT), which leverages precise and targeted interventions. NE-PDT capitalizes on photosensitizers activated by light to generate reactive oxygen species (ROS) that initiate apoptotic pathways in cancer cells. Nanoparticle enhancements optimize this process, improving drug delivery, selectivity, and ROS production within tumors. This review dissects NE-PDT's mechanistic framework, showcasing its potential to harness apoptosis as a potent tool in cancer therapy. Furthermore, the review explores the synergy between NE-PDT and complementary treatments like chemotherapy, immunotherapy, and targeted therapies, highlighting the potential to amplify apoptotic responses, enhance immune recognition of cancer cells, and inhibit resistance mechanisms. Preclinical and clinical advancements in NE-PDT demonstrate its efficacy across various cancer types. Challenges in translating NE-PDT into clinical practice are also addressed, emphasizing the need for optimizing nanoparticle design, refining dosimetry, and ensuring long-term safety. Ultimately, NE-PDT represents a promising approach in cancer therapy, utilizing the intricate mechanisms of apoptosis to address therapeutic hurdles. The review underscores the importance of understanding the interplay between nanoparticles, ROS generation, and apoptotic pathways, contributing to a deeper comprehension of cancer biology and novel therapeutic strategies. As interdisciplinary collaborations continue to thrive, NE-PDT offers hope for effective and targeted cancer interventions, where apoptosis manipulation becomes central to conquering cancer. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Sheeja S Rajan
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Rahul Chandran
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| | - Heidi Abrahamse
- Laser Research Centre, Faculty of Health Sciences, University of Johannesburg, Johannesburg, South Africa
| |
Collapse
|
13
|
Stelling-Férez J, Cappellacci I, Pandolfi A, Gabaldón JA, Pipino C, Nicolás FJ. Oleanolic acid rescues critical features of umbilical vein endothelial cells permanently affected by hyperglycemia. Front Endocrinol (Lausanne) 2023; 14:1308606. [PMID: 38192424 PMCID: PMC10773851 DOI: 10.3389/fendo.2023.1308606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/23/2023] [Indexed: 01/10/2024] Open
Abstract
Skin wound healing is a physiological process that involves several cell types. Among them, endothelial cells are required for inflammation resolution and neo-angiogenesis, both necessary for tissue restoration after injury. Primary human umbilical vein endothelial cells (C-HUVECs) are derived from the umbilical cord. When women develop gestational diabetes, chronic exposure to hyperglycemia induces epigenetic modifications in these cells (GD-HUVECs), leading to a permanent pro-inflammatory phenotype and impaired angiogenesis in contrast to control cells. Oleanolic acid (OA) is a bioactive triterpenoid known for its epithelial cell migration promotion stimulation and higher tensile strength of wounds. However, the potentially anti-inflammatory and pro-angiogenic properties of OA are still under investigation. We tested OA on C- and GD-HUVECs under inflammatory conditions induced by low levels of the inflammatory cytokine TNF-α. Reduced expression of adhesion molecules VCAM1, ICAM1, and SELE was obtained in OA-pre-treated C- and GD-HUVECs. Additionally, protein VCAM1 levels were also decreased by OA. Coherently, monocyte adhesion assays showed that a lower number of monocytes adhered to GD-HUVEC endothelium under OA pre-treatment when compared to untreated ones. It is noteworthy that OA improved angiogenesis parameters in both phenotypes, being especially remarkable in the case of GD-HUVECs, since OA strongly rescued their poor tube formation behavior. Moreover, endothelial cell migration was improved in C- and GD-HUVECs in scratch assays, an effect that was further confirmed by focal adhesion (FA) remodeling, revealed by paxillin staining on immunocytochemistry assays. Altogether, these results suggest that OA could be an emergent wound healing agent due to its capacity to rescue endothelial malfunction caused by hyperglycemia.
Collapse
Affiliation(s)
- Javier Stelling-Férez
- Department of Nutrition and Food Technology, Health Sciences PhD Program, Universidad Católica de Murcia (UCAM), Murcia, Spain
- Regeneration, Molecular Oncology, and TGF-β, IMIB-Pascual Parrilla, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| | - Ilaria Cappellacci
- Department of Medical, Oral and Biotechnological Sciences, StemTeCh Group, Center for Advanced Studies and Technology-CAST (ex CeSI-MeT), University G. D’Annunzio Chieti-Pescara, Chieti, Italy
| | - Assunta Pandolfi
- Department of Medical, Oral and Biotechnological Sciences, StemTeCh Group, Center for Advanced Studies and Technology-CAST (ex CeSI-MeT), University G. D’Annunzio Chieti-Pescara, Chieti, Italy
| | - José Antonio Gabaldón
- Department of Nutrition and Food Technology, Health Sciences PhD Program, Universidad Católica de Murcia (UCAM), Murcia, Spain
| | - Caterina Pipino
- Department of Medical, Oral and Biotechnological Sciences, StemTeCh Group, Center for Advanced Studies and Technology-CAST (ex CeSI-MeT), University G. D’Annunzio Chieti-Pescara, Chieti, Italy
| | - Francisco José Nicolás
- Regeneration, Molecular Oncology, and TGF-β, IMIB-Pascual Parrilla, Hospital Clínico Universitario Virgen de la Arrixaca, Murcia, Spain
| |
Collapse
|
14
|
Kmezic I, Gustafsson R, Fink K, Svenningsson A, Samuelsson K, Ingre C, Olsson T, Hansson M, Kockum I, Adzemovic MZ, Press R. Validation of elevated levels of interleukin-8 in the cerebrospinal fluid, and discovery of new biomarkers in patients with GBS and CIDP using a proximity extension assay. Front Immunol 2023; 14:1241199. [PMID: 38077366 PMCID: PMC10702497 DOI: 10.3389/fimmu.2023.1241199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 11/01/2023] [Indexed: 12/18/2023] Open
Abstract
Background Biomarkers for diagnosis of inflammatory neuropathies, assessment of prognosis, and treatment response are lacking. Methods CSF and EDTA plasma from patients with Guillain-Barré syndrome (GBS), chronic inflammatory demyelinating polyneuropathy (CIDP), healthy controls (HC) and disease controls were analyzed with Olink multiplex proximity extension assay (PEA) from two independent cohorts. Levels of interleukin-8 (IL8) were further analyzed with ELISA in patients with GBS, CIDP, paraproteinemia-related demyelinating polyneuropathy (PDN), multifocal motor neuropathy (MMN), HC and disease controls. ROC analysis was performed. Outcome was measured with the GBS-disability score (GBS-ds) or Inflammatory Neuropathy Cause and Treatment (INCAT) score. Results In CSF, multiplex PEA analysis revealed up-regulation of IL8 in GBS compared to CIDP and HC respectively, and CIDP compared to HC. In addition, levels of IL2RA were upregulated in GBS compared to both HC and CIDP, SELE in GBS compared to HC, and ITGAM, IL6, and NRP1 in GBS compared to CIDP. In plasma, levels of MMP3, THBD and ITGAM were upregulated in CIDP compared to HC. Validation of multiplex IL8 results using ELISA, revealed increased levels of IL8 in CSF in patients with GBS and CIDP versus HC and non-inflammatory polyneuropathies (NIP) respectively, as well as in PDN versus NIP and HC. Levels of IL8 in CSF correlated with impairment in the acute phase of GBS as well as outcome at 6-months follow up. Conclusion IL8 in CSF is validated as a diagnostic biomarker in GBS and CIDP, and a prognostic biomarker in GBS. Multiplex PEA hereby identifies several potential biomarkers in GBS and CIDP.
Collapse
Affiliation(s)
- Ivan Kmezic
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Rasmus Gustafsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Katharina Fink
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Anders Svenningsson
- Department of Clinical Sciences, Karolinska Institutet Danderyd Hospital, Stockholm, Sweden
| | - Kristin Samuelsson
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Caroline Ingre
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tomas Olsson
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Magnus Hansson
- Department of Clinical Chemistry, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine H5, Karolinska Institutet, Stockholm, Sweden
| | - Ingrid Kockum
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Milena Z. Adzemovic
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Centre for Neurology, Academic Specialist Centre, Stockholm Health Services, Stockholm, Sweden
| | - Rayomand Press
- Department of Neurology, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
15
|
Liang C, Zhang B, Li R, Guo S, Fan X. Network pharmacology -based study on the mechanism of traditional Chinese medicine in the treatment of glioblastoma multiforme. BMC Complement Med Ther 2023; 23:342. [PMID: 37759283 PMCID: PMC10523639 DOI: 10.1186/s12906-023-04174-7] [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: 01/25/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND Glioblastoma multiforme (GBM) is one of the most common primary malignant brain tumors. Yi Qi Qu Yu Jie Du Fang (YYQQJDF) is a traditional Chinese medicine (TCM) prescription for GBM. The present study aimed to use a network pharmacology method to analyze the underlying mechanism of YQQYJDF in treating GBM. METHODS GBM sample data, active ingredients and potential targets of YQQYJDF were obtained from databases. R language was used to screen differentially expressed genes (DEGs) between GBM tissues and normal tissues, and to perform enrichment analysis and weighted gene coexpression network analysis (WGCNA). The Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database was used to perform a protein‒protein interaction (PPI) analysis. A Venn diagram was used to obtain the core target genes of YQQYJDF for GBM treatment. Molecular docking was used to verify the binding between the active ingredient molecules and the proteins corresponding to the core target genes. Cell proliferation assays and invasion assays were used to verify the effect of active ingredients on the proliferation and invasion of glioma cells. RESULTS A total of 73 potential targets of YQQYJDF in the treatment of GBM were obtained. Enrichment analyses showed that the biological processes and molecular functions involved in these target genes were related to the activation of the G protein-coupled receptor (GPCR) signaling pathway and the regulation of hypoxia. The neuroactive ligand‒receptor pathway, the cellular senescence pathway, the calcium signaling pathway, the cell cycle pathway and the p53 signaling pathway might play important roles. Combining the results of WGCNA and PPI analysis, five core target genes and their corresponding four core active ingredients were screened. Molecular docking indicated that the core active ingredient molecules and the proteins corresponding to the core target genes had strong binding affinities. Cell proliferation and invasion assays showed that the core active ingredients of YQQYJDF significantly inhibited the proliferation and invasion of glioma cells (P < 0.01). CONCLUSIONS The present study predicted the possible active ingredients and targets of YQQYJDF in treating GBM, and analyzed its possible mechanism. These results may provide a basis and ideas for further research.
Collapse
Affiliation(s)
- Chen Liang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China.
- Division of Medical Physics, Department of Diagnostic and Interventional Radiology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, 79108, Freiburg, Germany.
| | - Binbin Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Ruichun Li
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Shiwen Guo
- Department of Neurosurgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Xiaoxuan Fan
- Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China.
| |
Collapse
|
16
|
Dutsch A, Uhlig C, Bock M, Graesser C, Schuchardt S, Uhlig S, Schunkert H, Joner M, Holdenrieder S, Lechner K. Multi-Omic Candidate Screening for Markers of Severe Clinical Courses of COVID-19. J Clin Med 2023; 12:6225. [PMID: 37834869 PMCID: PMC10573369 DOI: 10.3390/jcm12196225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/17/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
BACKGROUND Severe coronavirus disease 2019 (COVID-19) disease courses are characterized by immuno-inflammatory, thrombotic, and parenchymal alterations. Prediction of individual COVID-19 disease courses to guide targeted prevention remains challenging. We hypothesized that a distinct serologic signature precedes surges of IL-6/D-dimers in severely affected COVID-19 patients. METHODS We performed longitudinal plasma profiling, including proteome, metabolome, and routine biochemistry, on seven seropositive, well-phenotyped patients with severe COVID-19 referred to the Intensive Care Unit at the German Heart Center. Patient characteristics were: 65 ± 8 years, 29% female, median CRP 285 ± 127 mg/dL, IL-6 367 ± 231 ng/L, D-dimers 7 ± 10 mg/L, and NT-proBNP 2616 ± 3465 ng/L. RESULTS Based on time-series analyses of patient sera, a prediction model employing feature selection and dimensionality reduction through least absolute shrinkage and selection operator (LASSO) revealed a number of candidate proteins preceding hyperinflammatory immune response (denoted ΔIL-6) and COVID-19 coagulopathy (denoted ΔD-dimers) by 24-48 h. These candidates are involved in biological pathways such as oxidative stress/inflammation (e.g., IL-1alpha, IL-13, MMP9, C-C motif chemokine 23), coagulation/thrombosis/immunoadhesion (e.g., P- and E-selectin), tissue repair (e.g., hepatocyte growth factor), and growth factor response/regulatory pathways (e.g., tyrosine-protein kinase receptor UFO and low-density lipoprotein receptor (LDLR)). The latter are host- or co-receptors that promote SARS-CoV-2 entry into cells in the absence of ACE2. CONCLUSIONS Our novel prediction model identified biological and regulatory candidate networks preceding hyperinflammation and coagulopathy, with the most promising group being the proteins that explain changes in D-dimers. These biomarkers need validation. If causal, our work may help predict disease courses and guide personalized treatment for COVID-19.
Collapse
Affiliation(s)
- Alexander Dutsch
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Carsten Uhlig
- Institute for Laboratory Medicine, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
| | - Matthias Bock
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Christian Graesser
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Sven Schuchardt
- Fraunhofer Institute for Toxicology and Experimental Medicine ITEM, 30625 Hannover, Germany
| | - Steffen Uhlig
- QuoData Gesellschaft für Qualitätsmanagement und Statistik, Fabeckstr. 43, 14195 Berlin, Germany
| | - Heribert Schunkert
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Michael Joner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| | - Stefan Holdenrieder
- Institute for Laboratory Medicine, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
| | - Katharina Lechner
- Department of Cardiology, German Heart Centre Munich, Technical University of Munich, Lazarettstraße 36, 80636 Munich, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Munich, Munich Heart Alliance, 80336 Munich, Germany
| |
Collapse
|
17
|
Viúdez-Pareja C, Kreft E, García-Caballero M. Immunomodulatory properties of the lymphatic endothelium in the tumor microenvironment. Front Immunol 2023; 14:1235812. [PMID: 37744339 PMCID: PMC10512957 DOI: 10.3389/fimmu.2023.1235812] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 08/08/2023] [Indexed: 09/26/2023] Open
Abstract
The tumor microenvironment (TME) is an intricate complex and dynamic structure composed of various cell types, including tumor, stromal and immune cells. Within this complex network, lymphatic endothelial cells (LECs) play a crucial role in regulating immune responses and influencing tumor progression and metastatic dissemination to lymph node and distant organs. Interestingly, LECs possess unique immunomodulatory properties that can either promote or inhibit anti-tumor immune responses. In fact, tumor-associated lymphangiogenesis can facilitate tumor cell dissemination and metastasis supporting immunoevasion, but also, different molecular mechanisms involved in LEC-mediated anti-tumor immunity have been already described. In this context, the crosstalk between cancer cells, LECs and immune cells and how this communication can shape the immune landscape in the TME is gaining increased interest in recent years. In this review, we present a comprehensive and updated report about the immunomodulatory properties of the lymphatic endothelium within the TME, with special focus on primary tumors and tumor-draining lymph nodes. Furthermore, we outline emerging research investigating the potential therapeutic strategies targeting the lymphatic endothelium to enhance anti-tumor immune responses. Understanding the intricate mechanisms involved in LEC-mediated immune modulation in the TME opens up new possibilities for the development of innovative approaches to fight cancer.
Collapse
Affiliation(s)
- Cristina Viúdez-Pareja
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| | - Ewa Kreft
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| | - Melissa García-Caballero
- Department of Molecular Biology and Biochemistry, Faculty of Sciences, Andalucía Tech, University of Málaga, Málaga, Spain
- IBIMA (Biomedical Research Institute of Málaga)-Plataforma BIONAND, Málaga, Spain
| |
Collapse
|
18
|
Pingili S, Makkena VK, Jaramillo AP, Awosusi BL, Ayyub J, Dabhi KN, Gohil NV, Tanveer N, Hussein S, Hamid P. The Role of Non-genetic Therapies to Reduce the Incidence of Sickle Cell Crisis: A Systematic Review. Cureus 2023; 15:e42785. [PMID: 37664256 PMCID: PMC10469705 DOI: 10.7759/cureus.42785] [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: 07/10/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Sickle cell anemia is a hemoglobinopathy that causes complications such as Vaso-Occlusive Crisis (VOC), stroke, priapism, Acute Chest Syndromes (ACS), and bone infarcts due to blood vessel occlusion, resulting in hypoxia, ischemia, and inflammation. Preventing these incidents improves the quality of life and lowers mortality rates in Sickle Cell Disease (SCD) patients. This systematic review aims to describe the drugs, their mechanisms of action, dosages, changes in hemoglobin parameters, decrease in VOCs, delay the time for the next VOC, decrease in the length of hospital stay, and side effects associated with these drugs. This review adheres to the Preferred Reporting Items for Systematic Review and Meta-Analysis (PRISMA) 2020 guidelines. For this review, we searched the PubMed, Google Scholar, and Cochrane databases and screened them for full free texts published in English and studied in humans in the last five years beginning in 2018. Randomized clinical trials (RCT), observational studies, meta-analyses, systemic reviews, and traditional reviews were all included in the search. According to the type of study, quality assessment tools are used, and eight papers are chosen. Full-text articles from these papers are studied, analyzed, and tabulated. We discussed seven interventions that are used to treat sickle cell disease. Voxelotor, crizanlizumab, L-glutamate, long-term blood transfusions, Zinc (Zn), Niprisan®, and Ciklavit* were found to reduce the number and severity of VOC. We discovered that VOCs containing L -glutamate reduced the length of hospitalization. Magnesium (Mg) did not affect the number and severity of VOCs. This review includes a few articles for the study. Future papers on this subject should include a large sample size and many papers. More clinical trials are required to evaluate the dosages and outcomes of using these drugs in combination to prevent VOCs.
Collapse
Affiliation(s)
- Shravya Pingili
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Vijaya Krishna Makkena
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Arturo P Jaramillo
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Babatope L Awosusi
- Pathology and Laboratory Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Javaria Ayyub
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Karan Nareshbhai Dabhi
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Namra V Gohil
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Nida Tanveer
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Sally Hussein
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Pousette Hamid
- Neurology, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
19
|
Ogrodzinski L, Platt S, Goulding J, Alexander C, Farr TD, Woolard J, Hill SJ, Kilpatrick LE. Probing expression of E-selectin using CRISPR-Cas9-mediated tagging with HiBiT in human endothelial cells. iScience 2023; 26:107232. [PMID: 37496673 PMCID: PMC10366498 DOI: 10.1016/j.isci.2023.107232] [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: 12/09/2022] [Revised: 05/30/2023] [Accepted: 06/23/2023] [Indexed: 07/28/2023] Open
Abstract
E-selectin is expressed on endothelial cells in response to inflammatory cytokines and mediates leukocyte rolling and extravasation. However, studies have been hampered by lack of experimental approaches to monitor expression in real time in living cells. Here, NanoLuc Binary Technology (NanoBiT) in conjunction with CRISPR-Cas9 genome editing was used to tag endogenous E-selectin in human umbilical vein endothelial cells (HUVECs) with the 11 amino acid nanoluciferase fragment HiBiT. Addition of the membrane-impermeable complementary fragment LgBiT allowed detection of cell surface expression. This allowed the effect of inflammatory mediators on E-selectin expression to be monitored in real time in living endothelial cells. NanoBiT combined with CRISPR-Cas9 gene editing allows sensitive monitoring of real-time changes in cell surface expression of E-selectin and offers a powerful tool for future drug discovery efforts aimed at this important inflammatory protein.
Collapse
Affiliation(s)
- Lydia Ogrodzinski
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, NG7 2UH Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, Nottingham, UK
| | - Simon Platt
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, NG7 2UH Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, Nottingham, UK
| | - Joelle Goulding
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, NG7 2UH Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, Nottingham, UK
| | - Cameron Alexander
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, Boots Building, University of Nottingham, NG7 2RD Nottingham, UK
| | - Tracy D. Farr
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, NG7 2UH Nottingham, UK
| | - Jeanette Woolard
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, NG7 2UH Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, Nottingham, UK
| | - Stephen J. Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, NG7 2UH Nottingham, UK
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, Nottingham, UK
| | - Laura E. Kilpatrick
- Centre of Membrane Proteins and Receptors, University of Birmingham and Nottingham, The Midlands, Nottingham, UK
- Division of Bimolecular Science and Medicinal Chemistry, School of Pharmacy, Biodiscovery Institute, University of Nottingham, NG7 2RD Nottingham, UK
| |
Collapse
|
20
|
Khoshbin M, Ahmadi SAY, Cheraghi M, Nouryazdan N, Birjandi M, Shahsavari G. Association of E-Selectin gene polymorphisms and serum E-Selectin level with risk of coronary artery disease in lur population of Iran. Arch Physiol Biochem 2023; 129:387-392. [PMID: 33022186 DOI: 10.1080/13813455.2020.1828481] [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] [Indexed: 10/23/2022]
Abstract
BACKGROUND Adhesion molecules like E-selectin have important role in pathogenesis of atherosclerosis. C1901T and G98T polymorphisms of E-selectin gene and E-selectin serum level may affect the risk of coronary artery disease (CAD). METHODS A total of 145 normal individuals and 154 patients diagnosed with CAD from the Lur population of Iran undergoing coronary angiography were enrolled. Genetic polymorphisms of E-selectin were determined using PCR-RFLP. Serum level of soluble E-selectin was measured using Elisa. RESULTS T allele in C1901T polymorphism was significantly associated with an increased risk of atherosclerosis (P = 0.018). No significant association was observed for G98T polymorphism. The mean serum level of soluble E-selectin in the patient group was significantly higher than the control group (P < 0.001). CONCLUSIONS Allele type in C1901T polymorphism plays a role in increasing the risk of developing CAD. Furthermore, since serum E-selectin level is associated with systemic inflammation, it contributes to the increased risk of the disease.
Collapse
Affiliation(s)
- Mobin Khoshbin
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department of Clinical Biochemistry, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Seyyed Amir Yasin Ahmadi
- Pediatric Growth and Development Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Mostafa Cheraghi
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department of Cardiology, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Negar Nouryazdan
- Department of Clinical Biochemistry, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mehdi Birjandi
- Department of Epidemiology and Biostatistics, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Gholamreza Shahsavari
- Cardiovascular Research Center, Shahid Rahimi Hospital, Lorestan University of Medical Sciences, Khorramabad, Iran
- Department of Clinical Biochemistry, Lorestan University of Medical Sciences, Khorramabad, Iran
| |
Collapse
|
21
|
Sialyl Lewis X/A and Cytokeratin Crosstalk in Triple Negative Breast Cancer. Cancers (Basel) 2023; 15:cancers15030731. [PMID: 36765690 PMCID: PMC9913872 DOI: 10.3390/cancers15030731] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/31/2022] [Accepted: 01/23/2023] [Indexed: 01/27/2023] Open
Abstract
Triple-negative breast cancer (TNBC) encompasses multiple entities and is generally highly aggressive and metastatic. We aimed to determine the clinical and biological relevance of Sialyl-Lewis X and A (sLeX/A)-a fucosylated glycan involved in metastasis-in TNBC. Here, we studied tissues from 50 TNBC patients, transcripts from a TNBC dataset from The Cancer Genome Atlas (TCGA) database, and a primary breast cancer cell line. All 50 TNBC tissue samples analysed expressed sLeX/A. Patients with high expression of sLeX/A had 3 years less disease-free survival than patients with lower expression. In tissue, sLeX/A negatively correlated with cytokeratins 5/6 (CK5/6, which was corroborated by the inverse correlation between fucosyltransferases and CK5/6 genes. Our observations were confirmed in vitro when inhibition of sLeX/A remarkably increased expression of CK5/6, followed by a decreased proliferation and invasion capacity. Among the reported glycoproteins bearing sLeX/A and based on the STRING tool, α6 integrin showed the highest interaction score with CK5/6. This is the first report on the sLeX/A expression in TNBC, highlighting its association with lower disease-free survival and its inverse crosstalk with CK5/6 with α6 integrin as a mediator. All in all, sLeX/A is critical for TNBC malignancy and a potential prognosis biomarker and therapeutic target.
Collapse
|
22
|
Scholz GA, Fux M, Christ L, Iype J, Banz Y, Villiger PM. Divergent regulatory T cell responses to high-dose methylprednisolone and tocilizumab in giant cell arteritis. J Autoimmun 2022; 133:102909. [PMID: 36115211 DOI: 10.1016/j.jaut.2022.102909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/28/2022] [Accepted: 08/31/2022] [Indexed: 12/13/2022]
Affiliation(s)
- Godehard A Scholz
- University Clinic of Rheumatology and Immunology, Inselspital, University Hospital Bern, Switzerland.
| | - Michaela Fux
- Institute of Social and Preventive Medicine, University of Bern, Switzerland.
| | - Lisa Christ
- University Clinic of Rheumatology and Immunology, Inselspital, University Hospital Bern, Switzerland.
| | - Joseena Iype
- University Institute of Clinical Chemistry, Inselspital, University Hospital Bern, Switzerland.
| | - Yara Banz
- Institute of Pathology, University of Bern, Switzerland.
| | | |
Collapse
|
23
|
Engelbrecht E, Kooistra T, Knipe RS. The Vasculature in Pulmonary Fibrosis. CURRENT TISSUE MICROENVIRONMENT REPORTS 2022; 3:83-97. [PMID: 36712832 PMCID: PMC9881604 DOI: 10.1007/s43152-022-00040-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 02/02/2023]
Abstract
Purpose of Review The current paradigm of idiopathic pulmonary fibrosis (IPF) pathogenesis involves recurrent injury to a sensitive alveolar epithelium followed by impaired repair responses marked by fibroblast activation and deposition of extracellular matrix. Multiple cell types are involved in this response with potential roles suggested by advances in single-cell RNA sequencing and lung developmental biology. Notably, recent work has better characterized the cell types present in the pulmonary endothelium and identified vascular changes in patients with IPF. Recent Findings Lung tissue from patients with IPF has been examined at single-cell resolution, revealing reductions in lung capillary cells and expansion of a population of vascular cells expressing markers associated with bronchial endothelium. In addition, pre-clinical models have demonstrated a fundamental role for aging and vascular permeability in the development of pulmonary fibrosis. Summary Mounting evidence suggests that the endothelium undergoes changes in the context of fibrosis, and these changes may contribute to the development and/or progression of pulmonary fibrosis. Additional studies will be needed to further define the functional role of these vascular changes.
Collapse
Affiliation(s)
| | - Tristan Kooistra
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Rachel S. Knipe
- Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
24
|
Zhong M, Huang J, Wu Z, Chan KG, Wang L, Li J, Lee LH, Law JWF. Potential Roles of Selectins in Periodontal Diseases and Associated Systemic Diseases: Could They Be Targets for Immunotherapy? Int J Mol Sci 2022; 23:14280. [PMID: 36430760 PMCID: PMC9698067 DOI: 10.3390/ijms232214280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/19/2022] Open
Abstract
Periodontal diseases are predisposing factors to the development of many systemic disorders, which is often initiated via leukocyte infiltration and vascular inflammation. These diseases could significantly affect human health and quality of life. Hence, it is vital to explore effective therapies to prevent disease progression. Periodontitis, which is characterized by gingival bleeding, disruption of the gingival capillary's integrity, and irreversible destruction of the periodontal supporting bone, appears to be caused by overexpression of selectins in periodontal tissues. Selectins (P-, L-, and E-selectins) are vital members of adhesion molecules regulating inflammatory and immune responses. They are mainly located in platelets, leukocytes, and endothelial cells. Furthermore, selectins are involved in the immunopathogenesis of vascular inflammatory diseases, such as cardiovascular disease, diabetes, cancers, and so on, by mediating leukocyte recruitment, platelet activation, and alteration of endothelial barrier permeability. Therefore, selectins could be new immunotherapeutic targets for periodontal disorders and their associated systemic diseases since they play a crucial role in immune regulation and endothelium dysfunction. However, the research on selectins and their association with periodontal and systemic diseases remains limited. This review aims to discuss the critical roles of selectins in periodontitis and associated systemic disorders and highlights the potential of selectins as therapeutic targets.
Collapse
Affiliation(s)
- Mei Zhong
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Jiangyong Huang
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Zhe Wu
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Kok-Gan Chan
- Division of Genetics and Molecular Biology, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur 50603, Malaysia
- International Genome Centre, Jiangsu University, Zhenjiang 212013, China
| | - Lijing Wang
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Vascular Biology Research Institute, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Jiang Li
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
- Department of Prosthodontics, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
- Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| | - Jodi Woan-Fei Law
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Selangor Darul Ehsan, Malaysia
| |
Collapse
|
25
|
Shivatare SS, Shivatare VS, Wong CH. Glycoconjugates: Synthesis, Functional Studies, and Therapeutic Developments. Chem Rev 2022; 122:15603-15671. [PMID: 36174107 PMCID: PMC9674437 DOI: 10.1021/acs.chemrev.1c01032] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Glycoconjugates are major constituents of mammalian cells that are formed via covalent conjugation of carbohydrates to other biomolecules like proteins and lipids and often expressed on the cell surfaces. Among the three major classes of glycoconjugates, proteoglycans and glycoproteins contain glycans linked to the protein backbone via amino acid residues such as Asn for N-linked glycans and Ser/Thr for O-linked glycans. In glycolipids, glycans are linked to a lipid component such as glycerol, polyisoprenyl pyrophosphate, fatty acid ester, or sphingolipid. Recently, glycoconjugates have become better structurally defined and biosynthetically understood, especially those associated with human diseases, and are accessible to new drug, diagnostic, and therapeutic developments. This review describes the status and new advances in the biological study and therapeutic applications of natural and synthetic glycoconjugates, including proteoglycans, glycoproteins, and glycolipids. The scope, limitations, and novel methodologies in the synthesis and clinical development of glycoconjugates including vaccines, glyco-remodeled antibodies, glycan-based adjuvants, glycan-specific receptor-mediated drug delivery platforms, etc., and their future prospectus are discussed.
Collapse
Affiliation(s)
- Sachin S Shivatare
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Vidya S Shivatare
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Chi-Huey Wong
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| |
Collapse
|
26
|
Cetin-Atalay R, Meliton AY, Sun KA, Glass ME, Woods PS, Peng YJ, Fang Y, Hamanaka RB, Prabhakar NR, Mutlu GM. Intermittent hypoxia inhibits epinephrine-induced transcriptional changes in human aortic endothelial cells. Sci Rep 2022; 12:17167. [PMID: 36229484 PMCID: PMC9561121 DOI: 10.1038/s41598-022-21614-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Accepted: 09/29/2022] [Indexed: 02/02/2023] Open
Abstract
Obstructive sleep apnea (OSA) is an independent risk factor for cardiovascular disease. While intermittent hypoxia (IH) and catecholamine release play an important role in this increased risk, the mechanisms are incompletely understood. We have recently reported that IH causes endothelial cell (EC) activation, an early phenomenon in the development of cardiovascular disease, via IH-induced catecholamine release. Here, we investigated the effects of IH and epinephrine on gene expression in human aortic ECs using RNA-sequencing. We found a significant overlap between IH and epinephrine-induced differentially expressed genes (DEGs) including enrichment in leukocyte migration, cytokine-cytokine receptor interaction, cell adhesion and angiogenesis. Epinephrine caused higher number of DEGs compared to IH. Interestingly, IH when combined with epinephrine had an inhibitory effect on epinephrine-induced gene expression. Combination of IH and epinephrine induced MT1G (Metallothionein 1G), which has been shown to be highly expressed in ECs from parts of aorta (i.e., aortic arch) where atherosclerosis is more likely to occur. In conclusion, epinephrine has a greater effect than IH on EC gene expression in terms of number of genes and their expression level. IH inhibited the epinephrine-induced transcriptional response. Further investigation of the interaction between IH and epinephrine is needed to better understand how OSA causes cardiovascular disease.
Collapse
Affiliation(s)
- Rengul Cetin-Atalay
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637 USA
| | - Angelo Y. Meliton
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637 USA
| | - Kaitlyn A. Sun
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637 USA
| | - Mariel E. Glass
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637 USA
| | - Parker S. Woods
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637 USA
| | - Ying-Jie Peng
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Emergency Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Institute for Integrative Physiology, University of Chicago, Chicago, IL USA
| | - Yun Fang
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637 USA ,grid.170205.10000 0004 1936 7822Institute for Integrative Physiology, University of Chicago, Chicago, IL USA
| | - Robert B. Hamanaka
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637 USA ,grid.170205.10000 0004 1936 7822Institute for Integrative Physiology, University of Chicago, Chicago, IL USA
| | - Nanduri R. Prabhakar
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Emergency Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Institute for Integrative Physiology, University of Chicago, Chicago, IL USA
| | - Gökhan M. Mutlu
- grid.170205.10000 0004 1936 7822Department of Medicine, University of Chicago, Chicago, IL USA ,grid.170205.10000 0004 1936 7822Section of Pulmonary and Critical Care Medicine, University of Chicago, 5841 S. Maryland Avenue, MC6026, Chicago, IL 60637 USA ,grid.170205.10000 0004 1936 7822Institute for Integrative Physiology, University of Chicago, Chicago, IL USA
| |
Collapse
|
27
|
Kalinin RE, Korotkova NV, Suchkov IA, Mzhavanadze ND, Ryabkov AN. Selectins and their involvement in the pathogenesis of cardiovascular diseases. KAZAN MEDICAL JOURNAL 2022; 103:617-627. [DOI: 10.17816/kmj2022-617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
The review presents current data on the structure and functional role of cell adhesion molecules belonging to the selectin family (selectins P, L and E), and their involvement in the pathogenesis of cardiovascular diseases. On the one hand, intercellular adhesion molecules of the vascular wall endothelium, platelets and leukocytes are an important link in the processes of vasculogenesis, development and regeneration of the vascular system. On the other hand, these molecules participate in the earliest stages of endothelial dysfunction with the subsequent development of pathology. For this reason, figuring out the mechanisms of activity of this group of molecules is very important for understanding the molecular basis of the cardiovascular diseases pathogenesis. The adhesion of molecules, both between cells and between cells and a component of the extracellular matrix, is the most important stage of physiological and biochemical processes. According to present knowledge, five classes of intercellular adhesion molecules are known: integrins, cadherins, immunoglobulins (including nectins), selectins and addressins. All of them are bonded to a cytoplasmic membrane and provide the interaction of cells with each other. Some of them are transmembrane and associated with the cytoskeleton of the cell. On the cell surface, intercellular adhesion molecules can be located in clusters, forming multipoint binding sites and thereby determining the degree of avidity. One of the most significant functions of selectins is participation in the initial stage of the leukocyte adhesion cascade, which results in their binding to the endothelium, rolling and further extravasation into tissues. The first stage of this process is mediated by specific non-covalent interactions between selectins and their glycan ligands, with the glycans functioning as an interface between leukocytes or cancer cells and the endothelium. Targeting these interactions remains one of the main strategies aimed at developing new methods of treating immune, inflammatory and oncological diseases.
Collapse
|
28
|
Transcriptomics of angiotensin II-induced long noncoding and coding RNAs in endothelial cells. J Hypertens 2022; 40:1303-1313. [PMID: 35762471 DOI: 10.1097/hjh.0000000000003140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE Angiotensin II (Ang II)-induced endothelial dysfunction plays an important role in the pathogenesis of cardiovascular diseases such as systemic hypertension, cardiac hypertrophy and atherosclerosis. Recently, long noncoding RNAs (lncRNAs) have been shown to play an essential role in the pathobiology of cardiovascular diseases; however, the effect of Ang II on lncRNAs and coding RNAs expression in endothelial cells has not been evaluated. Accordingly, we sought to evaluate the expression profiles of lncRNAs and coding RNAs in endothelial cells following treatment with Ang II. METHODS Human umbilical vein endothelial cells (HUVECs) were cultured and treated with Ang II (10-6 mol/l) for 24 h. The cells were then profiled for the expression of lncRNAs and mRNAs using the Arraystar Human lncRNA Expression Microarray V3.0. RESULTS In HUVECs following Ang II treatment, from a total of 30 584 lncRNA targets screened, 25 targets were significantly upregulated, while 69 were downregulated. In the same HUVECs samples, from 26 106 mRNA targets screened, 28 targets were significantly upregulated and 67 were downregulated. Of the differentially expressed lncRNAs, RP11-354P11.2 and RP11-360F5.1 were the most upregulated (11-fold) and downregulated (three-fold) lncRNAs, respectively. Assigning the differentially regulated genes into functional groups using bioinformatics reveals numerous genes involved in the nucleotide excision repair and ECM-receptor interaction. CONCLUSION This is the first study to profile the Ang II-induced differentially expressed lncRNAs and mRNAs in human endothelial cells. Our results reveal novel targets and substantially extend the list of potential candidate genes involved in Ang II-induced endothelial dysfunction and cardiovascular diseases.
Collapse
|
29
|
Cho I, Lee KN, Joo E, Kim YM, Kim TE, Park KH. Plasma E-selectin and kallistatin as predictive markers of histologic chorioamnionitis in women with preterm premature rupture of membranes. Am J Reprod Immunol 2022; 88:e13584. [PMID: 35772987 DOI: 10.1111/aji.13584] [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: 02/18/2022] [Revised: 05/23/2022] [Accepted: 06/13/2022] [Indexed: 12/01/2022] Open
Abstract
PROBLEM We aimed to assess the predictive potential of 12 plasma biomarkers to predict acute histologic chorioamnionitis (HCA) in women with preterm premature rupture of membranes (PPROM) and to develop multi-biomarker panels based on these biomarkers in combination with widely used conventional laboratory markers. METHOD OF STUDY This was a retrospective cohort study involving 81 singleton pregnant women (24-34 weeks of gestation) who delivered within 96 h of blood sampling. White blood cell (WBC) count, differential counts, and C-reactive protein (CRP) levels were measured at admission. The levels of DKK-3, Fas, haptoglobin, IGFBP-2, kallistatin, MIP-1α, MMP-2, MMP-8, pentraxin 3, progranulin, E-selectin, and P-selectin were evaluated by ELISA using stored plasma samples. The primary outcome measure was acute HCA. RESULTS Multivariate analyses showed that low plasma E-selectin and kallistatin levels were independently associated with HCA occurrence after adjusting for gestational age. Using a stepwise regression analysis, a multi-biomarker panel comprising plasma E-selectin, serum CRP, and WBC was developed, which provided a good prediction of acute HCA in women with PPROM (area under the curve [AUC], 0.899), with a significantly higher AUC than that of any single variable included in the panel (P<0.05). The plasma levels of DKK-3, Fas, haptoglobin, IGFBP-2, MIP-1α, MMP-2, MMP-8, pentraxin 3, and P-selectin were not significantly associated with HCA occurrence. CONCLUSIONS This study identified E-selectin and kallistatin as potential plasma biomarkers associated with acute HCA in women with PPROM. Their combined analysis with serum CRP and WBC counts significantly improved acute HCA diagnosis. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Iseop Cho
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyong-No Lee
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Eunwook Joo
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Yu Mi Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Tae Eun Kim
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Kyo Hoon Park
- Department of Obstetrics and Gynecology, Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| |
Collapse
|
30
|
Radovani B, Gudelj I. N-Glycosylation and Inflammation; the Not-So-Sweet Relation. Front Immunol 2022; 13:893365. [PMID: 35833138 PMCID: PMC9272703 DOI: 10.3389/fimmu.2022.893365] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/30/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic inflammation is the main feature of many long-term inflammatory diseases such as autoimmune diseases, metabolic disorders, and cancer. There is a growing number of studies in which alterations of N-glycosylation have been observed in many pathophysiological conditions, yet studies of the underlying mechanisms that precede N-glycome changes are still sparse. Proinflammatory cytokines have been shown to alter the substrate synthesis pathways as well as the expression of glycosyltransferases required for the biosynthesis of N-glycans. The resulting N-glycosylation changes can further contribute to disease pathogenesis through modulation of various aspects of immune cell processes, including those relevant to pathogen recognition and fine-tuning the inflammatory response. This review summarizes our current knowledge of inflammation-induced N-glycosylation changes, with a particular focus on specific subsets of immune cells of innate and adaptive immunity and how these changes affect their effector functions, cell interactions, and signal transduction.
Collapse
Affiliation(s)
- Barbara Radovani
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
| | - Ivan Gudelj
- Department of Biotechnology, University of Rijeka, Rijeka, Croatia
- Genos Glycoscience Research Laboratory, Zagreb, Croatia
| |
Collapse
|
31
|
Liu H, Liu D, Ji M, Xiao P, Qin Y, Zhao J, Wang N, Gou J, Yin T, He H, Chen G, Zhang Y, Tang X. Inflammation-targeted sialic acid-dexamethasone conjugates for reducing the side effects of glucocorticoids. Int J Pharm 2022; 622:121900. [PMID: 35690305 DOI: 10.1016/j.ijpharm.2022.121900] [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: 02/24/2022] [Revised: 06/02/2022] [Accepted: 06/04/2022] [Indexed: 11/30/2022]
Abstract
As a potent glucocorticoid drug (GCs), Dexamethasone (Dex) is widely used clinically for the treatment of inflammatory diseases. However, such side effects as Cushing's syndrome and osteoporosis caused severe distress to patients. Herein, a sialic acid (SA)-modified dexamethasone conjugate (Dex-SA) was synthesized successfully to reduce side effects by targeting inflammatory diseases. The solubility of Dex-SA in water reached 58 times that of Dex, which meets the need for intravenous administration. The excellent stability of Dex-SA in plasma also laid a foundation for targeting disease sites. According to cellular uptake and biodistribution experiments, Dex-SA was more readily to be taken up by inflammatory cells and accumulated in diseased kidneys compared to Dex, which is attributed to the interaction of SA with E-selectin receptors overexpressed on the surface of inflammatory vascular endothelial cells. Besides, the pharmacodynamics studies of acute kidney injury showed that Dex-SA and Dex could produce comparable therapeutic effects. More importantly, Dex-SA was found to significantly reduce Dex-related side effects, as measured by blood glucose, red blood cells and immune cells, etc. At last, molecular docking results were obtained to confirm that Dex-SA could enter the cells by binding specifically with the E-selectin receptor, for combination with glucocorticoid receptors in the cytoplasm to exert pharmacological effects. Our study is expected to contribute a new strategy to the safe and effective targeting treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Hongbing Liu
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| | - Dongdong Liu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, Liaoning 110016, China
| | - Muse Ji
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| | - Peifu Xiao
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| | - Yi Qin
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, Liaoning 110016, China
| | - Jiansong Zhao
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| | - Na Wang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| | - Jingxin Gou
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| | - Haibing He
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| | - Guoliang Chen
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, Liaoning 110016, China.
| | - Yu Zhang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China.
| | - Xing Tang
- Department of Pharmaceutics, Shenyang Pharmaceutical University, Wenhua Road No.103, Shenyang, China
| |
Collapse
|
32
|
Suryawanshi H, Yang H, Lubetzky M, Morozov P, Lagman M, Thareja G, Alonso A, Li C, Snopkowski C, Belkadi A, Mueller FB, Lee JR, Dadhania DM, Salvatore SP, Seshan SV, Sharma VK, Suhre K, Suthanthiran M, Tuschl T, Muthukumar T. Detection of infiltrating fibroblasts by single-cell transcriptomics in human kidney allografts. PLoS One 2022; 17:e0267704. [PMID: 35657798 PMCID: PMC9165878 DOI: 10.1371/journal.pone.0267704] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
We tested the hypothesis that single-cell RNA-sequencing (scRNA-seq) analysis of human kidney allograft biopsies will reveal distinct cell types and states and yield insights to decipher the complex heterogeneity of alloimmune injury. We selected 3 biopsies of kidney cortex from 3 individuals for scRNA-seq and processed them fresh using an identical protocol on the 10x Chromium platform; (i) HK: native kidney biopsy from a living donor, (ii) AK1: allograft kidney with transplant glomerulopathy, tubulointerstitial fibrosis, and worsening graft function, and (iii) AK2: allograft kidney after successful treatment of active antibody-mediated rejection. We did not study T-cell-mediated rejections. We generated 7217 high-quality single cell transcriptomes. Taking advantage of the recipient-donor sex mismatches revealed by X and Y chromosome autosomal gene expression, we determined that in AK1 with fibrosis, 42 months after transplantation, more than half of the kidney allograft fibroblasts were recipient-derived and therefore likely migratory and graft infiltrative, whereas in AK2 without fibrosis, 84 months after transplantation, most fibroblasts were donor-organ-derived. Furthermore, AK1 was enriched for tubular progenitor cells overexpressing profibrotic extracellular matrix genes. AK2, eight months after successful treatment of rejection, contained plasmablast cells with high expression of immunoglobulins, endothelial cell elaboration of T cell chemoattractant cytokines, and persistent presence of cytotoxic T cells. In addition to these key findings, our analysis revealed unique cell types and states in the kidney. Altogether, single-cell transcriptomics yielded novel mechanistic insights, which could pave the way for individualizing the care of transplant recipients.
Collapse
Affiliation(s)
- Hemant Suryawanshi
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, United States of America
| | - Hua Yang
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Michelle Lubetzky
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
- Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY, United States of America
| | - Pavel Morozov
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, United States of America
| | - Mila Lagman
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Gaurav Thareja
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Doha, Qatar
| | - Alicia Alonso
- Epigenomics Core Facility, Weill Cornell Medical College, New York, NY, United States of America
| | - Carol Li
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Catherine Snopkowski
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Aziz Belkadi
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Doha, Qatar
| | - Franco B. Mueller
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - John R. Lee
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
- Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY, United States of America
| | - Darshana M. Dadhania
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
- Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY, United States of America
| | - Steven P. Salvatore
- Division of Renal Pathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Surya V. Seshan
- Division of Renal Pathology, Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Vijay K. Sharma
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
| | - Karsten Suhre
- Department of Physiology and Biophysics, Weill Cornell Medical College in Qatar, Doha, Qatar
| | - Manikkam Suthanthiran
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
- Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY, United States of America
| | - Thomas Tuschl
- Laboratory of RNA Molecular Biology, The Rockefeller University, New York, NY, United States of America
| | - Thangamani Muthukumar
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medical College, New York, NY, United States of America
- Department of Transplantation Medicine, New York Presbyterian Hospital-Weill Cornell Medical College, New York, NY, United States of America
| |
Collapse
|
33
|
Nashine S, Cohen P, Wan J, Kenney C. Effect of Humanin G (HNG) on inflammation in age-related macular degeneration (AMD). Aging (Albany NY) 2022; 14:4247-4269. [PMID: 35576057 PMCID: PMC9186758 DOI: 10.18632/aging.204074] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/25/2022] [Indexed: 12/03/2022]
Abstract
Inflammation plays a crucial role in the etiology and pathogenesis of AMD (Age-related Macular Degeneration). Humanin G (HNG) is a Mitochondrial Derived Peptide (MDP) that is cytoprotective in AMD and can protect against mitochondrial and cellular stress induced by damaged AMD mitochondria. The goal of this study was to test our hypothesis that inflammation-associated marker protein levels are increased in AMD and treatment with HNG leads to reduction in their protein levels. Humanin protein levels were measured in the plasma of AMD patients and normal subjects using ELISA assay. Humanin G was added to AMD and normal (control) cybrids which had identical nuclei from mitochondria-deficient ARPE-19 cells but differed in mitochondrial DNA (mtDNA) content derived from clinically characterized AMD patients and normal (control) subjects. Cell lysates were extracted from untreated and HNG-treated AMD and normal cybrids, and the Luminex XMAP multiplex assay was used to measure the levels of inflammatory proteins. AMD plasma showed reduced Humanin protein levels, but higher protein levels of inflammation markers compared to control plasma samples. In AMD RPE cybrid cells, Humanin G reduced the CD62E/ E-Selectin, CD62P/ P-Selectin, ICAM-1, TNF-α, MIP-1α, IFN–γ, IL-1β, IL-13, and IL-17A protein levels, thereby suggesting that Humanin G may rescue from mtDNA-mediated inflammation in AMD cybrids. In conclusion, we present novel findings that: A) show reduced Humanin protein levels in AMD plasma vs. normal plasma; B) suggest the role of inflammatory markers in AMD pathogenesis, and C) highlight the positive effects of Humanin G in reducing inflammation in AMD.
Collapse
Affiliation(s)
- Sonali Nashine
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA
| | - Pinchas Cohen
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Junxiang Wan
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90007, USA
| | - Cristina Kenney
- Department of Ophthalmology, Gavin Herbert Eye Institute, University of California Irvine, Irvine, CA 92697, USA.,Department of Pathology and Laboratory Medicine, University of California, Irvine, CA 92697, USA
| |
Collapse
|
34
|
Landreth S, Teague AM, Jensen ME, Gulati S, Tryggestad JB. Impact of maternal diabetes exposure on soluble adhesion molecules in the offspring. Nutr Metab Cardiovasc Dis 2022; 32:1253-1258. [PMID: 35256229 PMCID: PMC9018574 DOI: 10.1016/j.numecd.2022.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 11/29/2021] [Accepted: 01/31/2022] [Indexed: 10/19/2022]
Abstract
BACKGROUND AND AIMS Soluble adhesion molecules are associated with cardiovascular disease and increased in individuals with diabetes. This study assesses the impact of diabetes exposure in utero on the abundance of circulating adhesion molecules in cord serum and soluble adhesion molecules released from human umbilical vein endothelial cells (HUVEC) exposed to high glucose concentrations. METHODS AND RESULTS Women with and without diabetes were recruited. DM was diagnosed based on the American Diabetes Association criteria. Primary cultures of HUVEC were cultured in 5 mM and 25 mM glucose with 25 mM mannitol osmotic control. The soluble adhesion molecules, intracellular adhesion molecule (ICAM), vascular cell adhesion molecule (VCAM) and E-selectin were measured by ELISA in the cord blood serum and conditioned HUVEC media. The mothers with DM were older with higher BMI (p = 0.027 and 0.008, respectively). In a fully adjusted model, VCAM was significantly increased in the cord serum of infants born to mothers with diabetes (p = 0.046), but ICAM and E-selectin were not different. ICAM was also significantly correlated with maternal HbA1c (r2 = 0.16, p = 0.004) and cord serum non-esterified fatty acids (r2 = 0.08, p = 0.013). From the HUVEC media, the abundance of adhesion molecules was not different based on DM or high glucose exposure; however, VCAM abundance in the HUVEC supernatant was significantly correlated with ICAM (r2 = 0.27, p = 0.010) and cord serum c-peptide (R2 = 0.19, p = 0.043). CONCLUSIONS Alterations in soluble adhesion molecule abundance in infants exposed to the diabetic milieu of pregnancy may reflect early alterations in vascular function predicting future cardiovascular disease.
Collapse
Affiliation(s)
- Samantha Landreth
- Department of Pediatrics, Section of Diabetes/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - April M Teague
- Department of Pediatrics, Section of Diabetes/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Mary E Jensen
- Department of Pediatrics, Section of Diabetes/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shelly Gulati
- Department of Pediatrics, Section of Diabetes/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jeanie B Tryggestad
- Department of Pediatrics, Section of Diabetes/Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
35
|
Szlasa W, Wilk K, Knecht-Gurwin K, Gurwin A, Froń A, Sauer N, Krajewski W, Saczko J, Szydełko T, Kulbacka J, Małkiewicz B. Prognostic and Therapeutic Role of CD15 and CD15s in Cancer. Cancers (Basel) 2022; 14:cancers14092203. [PMID: 35565333 PMCID: PMC9101515 DOI: 10.3390/cancers14092203] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 02/06/2023] Open
Abstract
Simple Summary CD15 (Lewis X) is a typical myeloid antigen presented in myeloid and monocytic lineages of cells. This molecule interacts with E-, L- and P-selectins, which allows for adhesion with endothelial cells. CD15 is found on various cancer cells, including renal cancer, prostate and bladder cancers, acute leukaemias, hepatocellular carcinoma, breast cancer and melanoma cells. Its high expression can serve as a prognostic marker for patients and is a potentially valuable target for immunotherapy against cancer. Blockage of the antigen’s function results in reduced metastatic potential and it may be an immunotherapeutic target. CD15s is a sialyl derivative of CD15; however, unlike the high expression of CD15, which is a prognostic factor in Hodgkin lymphoma, CD15s relates to poor prognosis for patients. CD15 is considered a marker of cancer stem cells. This review presents a comprehensive description of the prognostic role of CD15 and CD15s and their use in anticancer therapy. Abstract CD15 (Lewis X/Lex) is a fucosyl (3-fucosly-N-acetyl-lactosamine) moiety found on membrane proteins of various cancer cells. These cancers include renal cancer, prostate and bladder cancers, acute leukaemias, hepatocellular carcinoma, breast cancer and melanoma. The biological role of CD15 is interaction with E-, L- and P-selectins (adhesion molecules), allowing for adhesion with endothelial cells. In this way, cancer cells start to interact with the endothelia of blood vessels and consequently move out from the blood flow to the surrounding tissues. Blockage of the antigen’s function results in reduced metastatic potential. Moreover, the molecule may be a therapeutic target against cancer in monoclonal antibody-based therapies. CD15 may serve as a prognostic marker for patients and there are high hopes for its use in the immunotherapeutic treatment of tumours. CD15s is a sialyl derivative of CD15 that possesses its own unique characteristics. Its soluble form may act as a competitive inhibitor of the interaction of cancer cells with epithelial cells and thus disallow migration through the vessels. However, the prognostic relevance of CD15 and CD15s expression is very complex. This review presents a comprehensive description of the role of CD15 and CD15s in cancer development and metastasis and overviews its significance for clinical applications.
Collapse
Affiliation(s)
- Wojciech Szlasa
- Department of Minimally Invasive and Robotic Urology, University Center of Excellence in Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (A.G.); (A.F.); (W.K.); (T.S.)
- Correspondence: (W.S.); (B.M.)
| | - Karol Wilk
- Department of Minimally Invasive and Robotic Urology, University Center of Excellence in Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (A.G.); (A.F.); (W.K.); (T.S.)
| | - Klaudia Knecht-Gurwin
- Department of Dermatology, Venerology and Allergology, Faculty of Medicine, Wroclaw Medical University, 50-368 Wroclaw, Poland;
| | - Adam Gurwin
- Department of Minimally Invasive and Robotic Urology, University Center of Excellence in Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (A.G.); (A.F.); (W.K.); (T.S.)
| | - Anita Froń
- Department of Minimally Invasive and Robotic Urology, University Center of Excellence in Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (A.G.); (A.F.); (W.K.); (T.S.)
| | - Natalia Sauer
- Department of Drugs Form Technology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland;
| | - Wojciech Krajewski
- Department of Minimally Invasive and Robotic Urology, University Center of Excellence in Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (A.G.); (A.F.); (W.K.); (T.S.)
| | - Jolanta Saczko
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.S.); (J.K.)
| | - Tomasz Szydełko
- Department of Minimally Invasive and Robotic Urology, University Center of Excellence in Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (A.G.); (A.F.); (W.K.); (T.S.)
| | - Julita Kulbacka
- Department of Molecular and Cellular Biology, Faculty of Pharmacy, Wroclaw Medical University, 50-556 Wroclaw, Poland; (J.S.); (J.K.)
| | - Bartosz Małkiewicz
- Department of Minimally Invasive and Robotic Urology, University Center of Excellence in Urology, Wroclaw Medical University, 50-556 Wroclaw, Poland; (K.W.); (A.G.); (A.F.); (W.K.); (T.S.)
- Correspondence: (W.S.); (B.M.)
| |
Collapse
|
36
|
Santana-Gonçalves M, Zanin-Silva D, Henrique-Neto Á, Moraes DA, Kawashima- Vasconcelos MY, Lima-Júnior JR, Dias JBE, Bragagnollo V, de Azevedo JTC, Covas DT, Malmegrim KCR, Ramalho L, Oliveira MC. Autologous hematopoietic stem cell transplantation modifies specific aspects of systemic sclerosis-related microvasculopathy. Ther Adv Musculoskelet Dis 2022; 14:1759720X221084845. [PMID: 35368373 PMCID: PMC8966069 DOI: 10.1177/1759720x221084845] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 02/14/2022] [Indexed: 11/15/2022] Open
Abstract
Objective: Autologous hematopoietic stem cell transplantation (AHSCT) is a therapeutic option for patients with severe and progressive systemic sclerosis (SSc). Here, we aimed to investigate how AHSCT affects the vasculopathy of SSc patients. Methods: Twenty-seven SSc patients were retrospectively assessed, before and after AHSCT, for vessel morphology (nailfold capillaroscopy), skin expression of endothelial markers and serum levels of markers of inflammation, angiogenesis and endothelial activation. Skin biopsies were analyzed by immunohistochemistry (IHC) for expression of CD31, VE-cadherin, E-selectin, angiopoietin-1 (Ang1), angiopoietin-2 (Ang2), Tie-2, vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor receptor 2 (VEGFR2), and endothelin-1 before and 12 months post-AHSCT. Serum samples from SSc patients were assessed before and up to 36 months after AHSCT for IL-6, von Willebrand factor (vWF), CXC Motif Chemokine Ligand 8 (CXCL8), Endothelin-1, epidermal growth factor (EGF), VEGFA, Pentraxin-3, Intercellular Adhesion Molecule 1 (ICAM-1), E-selectin, P-selectin, Thrombomodulin and IL-18 levels, and compared to healthy control samples. Results: On nailfold capillaroscopy, the number of capillaries increased at 1 year, while giant capillaries decreased at 6 months and 1 year after AHSCT. In the skin biopsies, expression of E-selectin notably decreased and Ang1 increased after AHSCT. At baseline, all vascular markers evaluated in the serum were significantly higher in SSc patients when compared to healthy controls, except for ICAM-1. When compared at different time points after AHSCT, Thrombomodulin, Pentraxin-3, vWF, and IL-18 levels remained generally stable at high levels until 36 months after AHSCT. Conclusion: Our results suggest that AHSCT contributes to improvements of the vessel morphology and dermal microvasculopathy, but does not normalize elevated levels of serum vascular markers in SSc patients. Additional vascular therapeutic approaches might contribute to more effectively treat the endothelial injury.
Collapse
Affiliation(s)
- Maynara Santana-Gonçalves
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Graduate Program in Oncology, Stem Cell and Cell-Therapy, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Djúlio Zanin-Silva
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Graduate Program in Basic and Applied Immunology Program, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Álvaro Henrique-Neto
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Graduate Program in Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Daniela A. Moraes
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Marianna Y. Kawashima- Vasconcelos
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Graduate Program in Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - João R. Lima-Júnior
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Images, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Juliana B. E. Dias
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Vinícius Bragagnollo
- Department of Internal Medicine, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Júlia T. C. de Azevedo
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Images, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Dimas T. Covas
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Medical Images, Hematology and Oncology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Kelen C. R. Malmegrim
- Center for Cell-based Therapy, Regional Hemotherapy Center of the Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- Department of Clinical, Toxicological and Bromatological Analysis, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Leandra Ramalho
- Department of Pathology and Legal Medicine, Ribeirão Preto Medical School, University of São Paulo, Brazil
| | | |
Collapse
|
37
|
Early unhealthy eating habits underlie morpho-functional changes in the liver and adipose tissue in male rats. Histochem Cell Biol 2022; 157:657-669. [PMID: 35344087 DOI: 10.1007/s00418-022-02092-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2022] [Indexed: 12/26/2022]
Abstract
Early-life consumption of high-fat and sugar-rich foods is recognized as a major contributor for the onset of metabolic dysfunction and its related disorders, including diabetes and nonalcoholic fatty liver disease. The lifelong impact of early unhealthy eating habits that start at younger ages remains unclear. Therefore, to better understand the effects of diet, it is essential to evaluate the structural and functional changes induced in metabolic organs and potential mechanisms underlying those changes. To investigate the long-term effects of eating habits, young male rats were exposed to high-sugar and high-energy diets. After 14 weeks, body composition was assessed, and histopathological changes were analyzed in the liver and adipose tissue. Serum biochemical parameters were also determined. Expression of inflammatory markers in the liver was evaluated by immunohistochemistry. Our results revealed that serum levels of glucose, creatinine, aspartate transaminase (AST), alanine transaminase (ALT), and lipid profile were increased in rats red high-sugar and high-energy diets. Histopathological alterations were observed, including abnormal hepatocyte organization and lipid droplet accumulation in the liver, and abnormal structure of adipocytes. In both unhealthy diet groups, hepatic expression of Toll-like receptor 4 (TLR4), cyclooxygenase 2 (COX-2), and E-selectin were increased, as well as a biomarker of oxidative stress. Together, our data demonstrated that unhealthy diets induced functional and structural changes in the metabolic organs, suggesting that proinflammatory and oxidative stress mechanisms trigger the hepatic alterations and metabolic dysfunction.
Collapse
|
38
|
A single-cell atlas of non-haematopoietic cells in human lymph nodes and lymphoma reveals a landscape of stromal remodelling. Nat Cell Biol 2022; 24:565-578. [PMID: 35332263 PMCID: PMC9033586 DOI: 10.1038/s41556-022-00866-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 02/10/2022] [Indexed: 12/14/2022]
Abstract
The activities of non-haematopoietic cells (NHCs), including mesenchymal stromal cells and endothelial cells, in lymphomas are reported to underlie lymphomagenesis. However, our understanding of lymphoma NHCs has been hampered by unexplained NHC heterogeneity, even in normal human lymph nodes (LNs). Here we constructed a single-cell transcriptome atlas of more than 100,000 NHCs collected from 27 human samples, including LNs and various nodal lymphomas, and it revealed 30 distinct subclusters, including some that were previously unrecognized. Notably, this atlas was useful for comparative analyses with lymphoma NHCs, which revealed an unanticipated landscape of subcluster-specific changes in gene expression and interaction with malignant cells in follicular lymphoma NHCs. This facilitates our understanding of stromal remodelling in lymphoma and highlights potential clinical biomarkers. Our study largely updates NHC taxonomy in human LNs and analysis of disease status, and provides a rich resource and deeper insights into LN and lymphoma biology to advance lymphoma management and therapy. Abe et al. profile, characterize and compare non-haematopoietic cells in normal human lymph nodes versus nodal lymphomas from patients, providing insights into stromal modelling in health and disease.
Collapse
|
39
|
Bandzerewicz A, Gadomska-Gajadhur A. Into the Tissues: Extracellular Matrix and Its Artificial Substitutes: Cell Signalling Mechanisms. Cells 2022; 11:914. [PMID: 35269536 PMCID: PMC8909573 DOI: 10.3390/cells11050914] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
The existence of orderly structures, such as tissues and organs is made possible by cell adhesion, i.e., the process by which cells attach to neighbouring cells and a supporting substance in the form of the extracellular matrix. The extracellular matrix is a three-dimensional structure composed of collagens, elastin, and various proteoglycans and glycoproteins. It is a storehouse for multiple signalling factors. Cells are informed of their correct connection to the matrix via receptors. Tissue disruption often prevents the natural reconstitution of the matrix. The use of appropriate implants is then required. This review is a compilation of crucial information on the structural and functional features of the extracellular matrix and the complex mechanisms of cell-cell connectivity. The possibilities of regenerating damaged tissues using an artificial matrix substitute are described, detailing the host response to the implant. An important issue is the surface properties of such an implant and the possibilities of their modification.
Collapse
|
40
|
Deng X, Yang F, Zhang L, Wang J, Liu B, Liang W, Tang J, Xie Y, He L. ECO: An Integrated Gene Expression Omnibus for Mouse Endothelial Cells In Vivo. Front Genet 2022; 13:844544. [PMID: 35309132 PMCID: PMC8931405 DOI: 10.3389/fgene.2022.844544] [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: 12/28/2021] [Accepted: 02/15/2022] [Indexed: 11/30/2022] Open
Abstract
Endothelial cell (EC) plays critical roles in vascular physiological and pathological processes. With the development of high-throughput technologies, transcriptomics analysis of EC has increased dramatically and a large amount of informative data have been generated. The dynamic patterns of gene expression in ECs under various conditions were revealed. Unfortunately, due to the lack of bioinformatics infrastructures, reuse of these large-scale datasets is challenging for many scientists. Here, by systematic re-analyzing, integrating, and standardizing of 203 RNA sequencing samples from freshly isolated mouse ECs under 71 conditions, we constructed an integrated mouse EC gene expression omnibus (ECO). The ECO database enables one-click retrieval of endothelial expression profiles from different organs under different conditions including disease models, genetic modifications, and clinically relevant treatments in vivo. The EC expression profiles are visualized with user-friendly bar-plots. It also provides a convenient search tool for co-expressed genes. ECO facilitates endothelial research with an integrated tool and resource for transcriptome analysis. The ECO database is freely available at https://heomics.shinyapps.io/ecodb/.
Collapse
Affiliation(s)
- Xiangyi Deng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Fan Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Lei Zhang
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, China
- Precision Medicine Center, the Second People’s Hospital of Huaihua, Huaihua, China
| | - Jianhao Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
| | - Boxuan Liu
- Precision Medicine Center, the Second People’s Hospital of Huaihua, Huaihua, China
| | - Wei Liang
- Precision Medicine Center, the Second People’s Hospital of Huaihua, Huaihua, China
| | - Jiefu Tang
- Trauma Center, First Affiliated Hospital of Hunan University of Medicine, Huaihua, China
| | - Yuan Xie
- Key Laboratory of Ministry of Education for Medicinal Plant Resource and Natural Pharmaceutical Chemistry, National Engineering Laboratory for Resource Developing of Endangered Chinese Crude Drugs in Northwest of China, College of Life Sciences, Shaanxi Normal University, Xi’an, China
| | - Liqun He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- *Correspondence: Liqun He,
| |
Collapse
|
41
|
Homologous targeting nanoparticles for enhanced PDT against osteosarcoma HOS cells and the related molecular mechanisms. J Nanobiotechnology 2022; 20:83. [PMID: 35177075 PMCID: PMC8851855 DOI: 10.1186/s12951-021-01201-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 12/09/2021] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND No prominent advancements in osteosarcoma (OS) treatment have been made in the past 20 years. Although photodynamic therapy (PDT) is an emerging technique for cancer therapy, the lack of targeted photosensitizers for OS treatment severely limits its applications. RESULTS In this study, we constructed a potential theranostic nanoplatform by using (poly (lactic-co-glycolic) acid (PLGA) nanoparticles (NPs) encapsulating IR780 into the shell (PLGA-IR780 NPs), which were further camouflaged with human OS cell membranes from the HOS cell line (MH-PLGA-IR780 NPs). These constructed NPs showed the capacity for homologous targeting with excellent photoacoustic (PA)/fluorescence (FL) imaging ability. Benefitting from their homologous targeting capacity, MH-PLGA-IR780 NPs obviously promoted cell endocytosis in vitro and tumor accumulation in vivo, which could further improve PDT performance under near-infrared (NIR) irradiation. In addition, to their homologous targeting and PA/FL dual-mode imaging ability, MH-PLGA-IR780 NPs had advantages in penetrating deeper into tumor tissues and in real-time dynamic distribution monitoring in vivo, which laid a foundation for further clinical applications in OS. Moreover, we demonstrated that PDT guided by the constructed NPs could significantly induce HOS cells apoptosis and ferroptosis via excessive accumulation of reactive oxygen species (ROS), and further determined that the potential anticancer molecular mechanism of apoptosis was triggered by the release of cytochrome c-activated mitochondrial apoptosis (endogenous apoptosis), and that ferroptosis caused the activation of nuclear receptor coactivator 4 (NCOA4)-mediated ferritinophagy and the inactivation of glutathione peroxidase 4 (GPX4), synergistically leading to excessive accumulation of Lipid-ROS and Lipid peroxides (LPOs). Concurrently, MH-PLGA-IR780 NPs-guided PDT also showed an obvious inhibitory effect on tumor growth in vivo. CONCLUSION These results suggest that this homologous targeting-based theranostic nanoplatform provides an effective method to improve PDT performance in OS and contributes a new and promising approach for OS therapy.
Collapse
|
42
|
Jiang Y, Lin W, Zhu L. Targeted Drug Delivery for the Treatment of Blood Cancers. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27041310. [PMID: 35209102 PMCID: PMC8880555 DOI: 10.3390/molecules27041310] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/02/2022] [Accepted: 02/08/2022] [Indexed: 01/11/2023]
Abstract
Blood cancers are a type of liquid tumor which means cancer is present in the body fluid. Multiple myeloma, leukemia, and lymphoma are the three common types of blood cancers. Chemotherapy is the major therapy of blood cancers by systemic administration of anticancer agents into the blood. However, a high incidence of relapse often happens, due to the low efficiency of the anticancer agents that accumulate in the tumor site, and therefore lead to a low survival rate of patients. This indicates an urgent need for a targeted drug delivery system to improve the safety and efficacy of therapeutics for blood cancers. In this review, we describe the current targeting strategies for blood cancers and recently investigated and approved drug delivery system formulations for blood cancers. In addition, we also discuss current challenges in the application of drug delivery systems for treating blood cancers.
Collapse
Affiliation(s)
- Yao Jiang
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK;
- Institute of Clinical Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Weifeng Lin
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Sciences, Rehovot 761001, Israel;
| | - Linyi Zhu
- Arthritis Research UK Centre for Osteoarthritis Pathogenesis, Kennedy Institute of Rheumatology, University of Oxford, Oxford OX3 7FY, UK
- Correspondence:
| |
Collapse
|
43
|
Rajasinghe LD, Bates MA, Benninghoff AD, Wierenga KA, Harkema JR, Pestka JJ. Silica Induction of Diverse Inflammatory Proteome in Lungs of Lupus-Prone Mice Quelled by Dietary Docosahexaenoic Acid Supplementation. Front Immunol 2022; 12:781446. [PMID: 35126352 PMCID: PMC8813772 DOI: 10.3389/fimmu.2021.781446] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 12/06/2021] [Indexed: 11/18/2022] Open
Abstract
Repeated short-term intranasal instillation of lupus-prone mice with crystalline silica (cSiO2) induces inflammatory gene expression and ectopic lymphoid neogenesis in the lung, leading to early onset of systemic autoimmunity and rapid progression to glomerulonephritis. These responses are suppressed by dietary supplementation with the ω-3 polyunsaturated fatty acid docosahexaenoic acid (DHA). Here, we tested the hypothesis that dietary DHA supplementation suppresses cSiO2-induced inflammatory proteins in bronchoalveolar alveolar lavage fluid (BALF) and plasma of lupus-prone mice. Archived tissue fluid samples were used from a prior investigation in which 6 wk-old lupus-prone female NZBWF1 mice were fed isocaloric diets containing 0 or 10 g/kg DHA for 2 wks and then intranasally instilled with 1 mg cSiO2 or vehicle once weekly for 4 wks. Cohorts were terminated at 1, 5, 9 or 13 wk post-instillation (PI). BALF and plasma from each cohort were analyzed by high density multiplex array profiling of 200 inflammatory proteins. cSiO2 time-dependently induced increases in the BALF protein signatures that were highly reflective of unresolved lung inflammation, although responses in the plasma were much less robust. Induced proteins in BALF included chemokines (e.g., MIP-2, MCP-5), enzymes (e.g., MMP-10, granzyme B), adhesion molecules (e.g., sE-selectin, sVCAM-1), co-stimulatory molecules (e.g., sCD40L, sCD48), TNF superfamily proteins (e.g., sTNFRI, sBAFF-R), growth factors (e.g., IGF-1, IGFBP-3), and signal transduction proteins (e.g., MFG-E8, FcgRIIB), many of which were blocked or delayed by DHA supplementation. The BALF inflammatory proteome correlated positively with prior measurements of gene expression, pulmonary ectopic lymphoid tissue neogenesis, and induction of autoantibodies in the lungs of the control and treatment groups. Ingenuity Pathway Analysis (IPA) revealed that IL-1β, TNF-α, and IL-6 were among the top upstream regulators of the cSiO2-induced protein response. Furthermore, DHA's effects were associated with downregulation of cSiO2-induced pathways involving i) inhibition of ARE-mediated mRNA decay, ii) bacterial and viral pattern recognition receptor activation, or iii) TREM1, STAT3, NF-κB, and VEGF signaling and with upregulation of PPAR, LXR/RXR and PPARα/RXRα signaling. Altogether, these preclinical findings further support the contention that dietary DHA supplementation could be applicable as an intervention against inflammation-driven autoimmune triggering by cSiO2 or potentially other environmental agents.
Collapse
Affiliation(s)
- Lichchavi D. Rajasinghe
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Melissa A. Bates
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
| | - Abby D. Benninghoff
- Department of Animal, Dairy and Veterinary Sciences, School of Veterinary Medicine, Utah State University, Logan, UT, United States
| | - Kathryn A. Wierenga
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI, United States
| | - Jack R. Harkema
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, MI, United States
| | - James J. Pestka
- Department of Food Science and Human Nutrition, Michigan State University, East Lansing, MI, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, United States
- Department of Microbiology and Molecular Genetics, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
44
|
Goel I, Noiri M, Yamauchi Y, Kato K, Chung UI, Teramura Y. Enhancement of intercellular interaction between iPSC-derived neural progenitor cells and activated endothelial cells using cell surface modification with functional oligopeptides. Biomater Sci 2022; 10:925-938. [PMID: 35014994 DOI: 10.1039/d1bm01503f] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Cell-based therapy has been used to treat stroke related disorders, which have no treatment options available 4.5 hours after onset. Although the administration of tissue plasminogen activator and mechanical thrombectomy are potent treatments, their clinical implementation is limited within the available time. Here, we aimed to use induced pluripotent stem cell-derived neural progenitor cells (NPCs) for stroke treatment with higher delivery efficiency in stroke areas, which will improve the therapeutic effect. E-selectin binding oligopeptide (Esbp) was conjugated with poly(ethylene glycol)-conjugated-lipid (Esbp-PEG-lipid) with different molecular weights of PEG (5 and 40 kDa) for cell surface modification. Then, we optimized the cell surface modification of NPCs by studying cell-binding ability onto the model surfaces of stroke areas, such as recombinant E-selectin-immobilized surfaces and TNF-α activated endothelium. As a result, the cell surface modification of NPCs with Esbp-PEG-lipid was found to induce specific intercellular interactions with the activated endothelium through the binding of Esbp with E-selectin. Additionally, the shorter PEG spacer was suitable for intercellular interactions. Thus, our technique shows potential for use in cell therapy with enhanced cell accumulation in infarct areas.
Collapse
Affiliation(s)
- Isha Goel
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Makoto Noiri
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yuka Yamauchi
- Department of Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan.,Department of Orthodontics, Division of Oral Health and Development, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8551, Japan
| | - Koichi Kato
- Department of Biomaterials, Graduate School of Biomedical and Health Sciences, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, 734-8553, Japan
| | - Ung-Il Chung
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Yuji Teramura
- Cellular and Molecular Biotechnology Research Institute (CMB), National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba Central fifth, 1-1-1 Higashi, Tsukuba, Ibaraki 305-8565, Japan. .,Department of Immunology, Genetics and Pathology (IGP), Uppsala University, Dag Hammarskjölds väg 20, SE-751 85, Uppsala, Sweden
| |
Collapse
|
45
|
Saiki P, Yoshihara M, Kawano Y, Miyazaki H, Miyazaki K. Anti-Inflammatory Effects of Heliangin from Jerusalem Artichoke (Helianthus tuberosus) Leaves Might Prevent Atherosclerosis. Biomolecules 2022; 12:biom12010091. [PMID: 35053238 PMCID: PMC8774036 DOI: 10.3390/biom12010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 12/27/2021] [Accepted: 01/04/2022] [Indexed: 11/16/2022] Open
Abstract
Atherosclerosis is considered the major cause of cardiovascular and cerebrovascular diseases, which are the leading causes of death worldwide. Excessive nitric oxide production and inflammation result in dysfunctional vascular endothelial cells, which are critically involved in the initiation and progression of atherosclerosis. The present study aimed to identify a bioactive compound from Jerusalem artichoke leaves with anti-inflammatory activity that might prevent atherosclerosis. We isolated bioactive heliangin that inhibited NO production in LPS-induced macrophage-like RAW 264.7 cells. Heliangin suppressed ICAM-1, VCAM-1, E-selectin, and MCP-1 expression, as well as NF-κB and IκBα phosphorylation, in vascular endothelial cells stimulated with TNF-α. These results suggested that heliangin suppresses inflammation by inhibiting excessive NO production in macrophages and the expression of the factors leading to the development of atherosclerosis via the NF-κB signaling pathway in vascular endothelial cells. Therefore, heliangin in Jerusalem artichoke leaves could function in the prevention of atherosclerosis that is associated with heart attacks and strokes.
Collapse
Affiliation(s)
- Papawee Saiki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
- Correspondence: ; Tel.: +81-29-861-4304
| | - Mizuki Yoshihara
- Graduate School of Life and Environment Sciences, University of Tsukuba, Tsukuba 305-8577, Japan; (M.Y.); (H.M.)
| | - Yasuhiro Kawano
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
| | - Hitoshi Miyazaki
- Graduate School of Life and Environment Sciences, University of Tsukuba, Tsukuba 305-8577, Japan; (M.Y.); (H.M.)
| | - Koyomi Miyazaki
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advance Industrial Science and Technology, Tsukuba 305-8566, Japan; (Y.K.); (K.M.)
| |
Collapse
|
46
|
Tvaroška I. Glycosyltransferases as targets for therapeutic intervention in cancer and inflammation: molecular modeling insights. CHEMICAL PAPERS 2022. [DOI: 10.1007/s11696-021-02026-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Udaya R, Sivakanesan R. Synopsis of Biomarkers of Atheromatous Plaque Formation, Rupture and Thrombosis in the Diagnosis of Acute Coronary Syndromes. Curr Cardiol Rev 2022; 18:53-62. [PMID: 35410616 PMCID: PMC9896418 DOI: 10.2174/1573403x18666220411113450] [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: 08/18/2021] [Revised: 12/08/2021] [Accepted: 01/15/2022] [Indexed: 11/22/2022] Open
Abstract
Acute coronary syndrome is the main cause of mortality and morbidity worldwide and early diagnosis is a challenge for clinicians. Though cardiac Troponin, the most commonly used biomarker, is the gold standard for myocardial necrosis, it is blind for ischemia without necrosis. Therefore, ideal biomarkers are essential in the care of patients presenting with symptoms suggestive of cardiac ischemia. The ideal biomarker or group of biomarkers of atheromatous plaque formation, rupture and thrombosis for timely and accurate diagnosis of acute coronary syndrome is a current need. Therefore, we discuss the existing understanding and future of biomarkers of atheromatous plaque formation, rupture and thrombosis of acute coronary syndrome in this review. Keywords were searched from Medline, ISI, IBSS and Google Scholar databases. Further, the authors conducted a manual search of other relevant journals and reference lists of primary articles. The development of high-sensitivity troponin assays facilitates earlier exclusion of acute coronary syndrome, contributing to a reduced length of stay at the emergency department, and earlier treatment resulting in better outcomes. Although researchers have investigated biomarkers of atheromatous plaque formation, rupture and thrombosis to help early diagnosis of cardiac ischemia, most of them necessitate validation from further analysis. Among these biomarkers, pregnancy-associated plasma protein-A, intercellular adhesion molecule-1, and endothelial cell-specific molecule- 1(endocan) have shown promising results in the early diagnosis of acute coronary syndrome but need further evaluation. However, the use of a combination of biomarkers representing varying pathophysiological mechanisms of cardiac ischemia will support risk assessment, diagnosis and prognosis in these patients and this is the way forward.
Collapse
Affiliation(s)
- Ralapanawa Udaya
- Address correspondence to this author at the Department of Medicine, University of Peradeniya, Galaha Rd, 20400, Sri Lanka; Tel: 0718495682; E-mail:
| | | |
Collapse
|
48
|
Ma H, Liu Z, Yang Y, Feng D, Dong Y, Garbutt TA, Hu Z, Wang L, Luan C, Cooper CD, Li Y, Welch JD, Qian L, Liu J. Functional coordination of non-myocytes plays a key role in adult zebrafish heart regeneration. EMBO Rep 2021; 22:e52901. [PMID: 34523214 DOI: 10.15252/embr.202152901] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 07/07/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022] Open
Abstract
Cardiac regeneration occurs primarily through proliferation of existing cardiomyocytes, but also involves complex interactions between distinct cardiac cell types including non-cardiomyocytes (non-CMs). However, the subpopulations, distinguishing molecular features, cellular functions, and intercellular interactions of non-CMs in heart regeneration remain largely unexplored. Using the LIGER algorithm, we assemble an atlas of cell states from 61,977 individual non-CM scRNA-seq profiles isolated at multiple time points during regeneration. This analysis reveals extensive non-CM cell diversity, including multiple macrophage (MC), fibroblast (FB), and endothelial cell (EC) subpopulations with unique spatiotemporal distributions, and suggests an important role for MC in inducing the activated FB and EC subpopulations. Indeed, pharmacological perturbation of MC function compromises the induction of the unique FB and EC subpopulations. Furthermore, we developed computational algorithm Topologizer to map the topological relationships and dynamic transitions between functional states. We uncover dynamic transitions between MC functional states and identify factors involved in mRNA processing and transcriptional regulation associated with the transition. Together, our single-cell transcriptomic analysis of non-CMs during cardiac regeneration provides a blueprint for interrogating the molecular and cellular basis of this process.
Collapse
Affiliation(s)
- Hong Ma
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Ziqing Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yuchen Yang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA.,Department of Genetics, University of North Carolina, Chapel Hill, NC, USA
| | - Dong Feng
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Yanhan Dong
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Tiffany A Garbutt
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Zhiyuan Hu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Li Wang
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Changfei Luan
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Cynthia D Cooper
- School of Molecular Biosciences, Washington State University Vancouver, Vancouver, WA, USA
| | - Yun Li
- Department of Genetics, University of North Carolina, Chapel Hill, NC, USA.,Department of Biostatistics, University of North Carolina, Chapel Hill, NC, USA.,Department of Computer Science, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua D Welch
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Li Qian
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Jiandong Liu
- McAllister Heart Institute, University of North Carolina, Chapel Hill, NC, USA.,Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
49
|
Šmak P, Tvaroška I, Koča J. The catalytic reaction mechanism of tyrosylprotein sulfotransferase-1. Phys Chem Chem Phys 2021; 23:23850-23860. [PMID: 34647946 DOI: 10.1039/d1cp03718h] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Tyrosine sulfation alters the biological activity of many proteins involved in different physiological and pathophysiological conditions, such as non-specific immune reaction, response to inflammation and ischemia, targeting of leukocytes and stem cells, or the formation of cancer metastases. Tyrosine sulfation is catalyzed by the enzymes tyrosylprotein sulfotransferases (TPST). In this study, we used QM/MM Car-Parrinello metadynamics simulations together with QM/MM potential energy calculations to investigate the catalytic mechanism of isoform TPST-1. The structural changes along the reaction coordinate are analyzed and discussed. Furthermore, both the methods supported the SN2 type of catalytic mechanism. The reaction barrier obtained from CPMD metadynamics was 12.8 kcal mol-1, and the potential energy scan led to reaction barriers of 11.6 kcal mol-1 and 13.7 kcal mol-1 with the B3LYP and OPBE functional, respectively. The comparison of the two methods (metadynamics and potential energy scan) may be helpful for future mechanistic studies. The insight into the reaction mechanism of TPST-1 might help with the rational design of transition-state TPST inhibitors.
Collapse
Affiliation(s)
- Pavel Šmak
- National Center for Biomolecular Research (NCBR), Faculty of Science, Masaryk University, Brno, Czech Republic
| | - Igor Tvaroška
- National Center for Biomolecular Research (NCBR), Faculty of Science, Masaryk University, Brno, Czech Republic.,Institute of Chemistry, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | - Jaroslav Koča
- National Center for Biomolecular Research (NCBR), Faculty of Science, Masaryk University, Brno, Czech Republic.,Central European Institute of Technology (CEITEC), Masaryk University, Brno, Czech Republic
| |
Collapse
|
50
|
Jarahian M, Marofi F, Maashi MS, Ghaebi M, Khezri A, Berger MR. Re-Expression of Poly/Oligo-Sialylated Adhesion Molecules on the Surface of Tumor Cells Disrupts Their Interaction with Immune-Effector Cells and Contributes to Pathophysiological Immune Escape. Cancers (Basel) 2021; 13:5203. [PMID: 34680351 PMCID: PMC8534074 DOI: 10.3390/cancers13205203] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/11/2021] [Accepted: 10/12/2021] [Indexed: 12/28/2022] Open
Abstract
Glycans linked to surface proteins are the most complex biological macromolecules that play an active role in various cellular mechanisms. This diversity is the basis of cell-cell interaction and communication, cell growth, cell migration, as well as co-stimulatory or inhibitory signaling. Our review describes the importance of neuraminic acid and its derivatives as recognition elements, which are located at the outermost positions of carbohydrate chains linked to specific glycoproteins or glycolipids. Tumor cells, especially from solid tumors, mask themselves by re-expression of hypersialylated neural cell adhesion molecule (NCAM), neuropilin-2 (NRP-2), or synaptic cell adhesion molecule 1 (SynCAM 1) in order to protect themselves against the cytotoxic attack of the also highly sialylated immune effector cells. More particularly, we focus on α-2,8-linked polysialic acid chains, which characterize carrier glycoproteins such as NCAM, NRP-2, or SynCam-1. This characteristic property correlates with an aggressive clinical phenotype and endows them with multiple roles in biological processes that underlie all steps of cancer progression, including regulation of cell-cell and/or cell-extracellular matrix interactions, as well as increased proliferation, migration, reduced apoptosis rate of tumor cells, angiogenesis, and metastasis. Specifically, re-expression of poly/oligo-sialylated adhesion molecules on the surface of tumor cells disrupts their interaction with immune-effector cells and contributes to pathophysiological immune escape. Further, sialylated glycoproteins induce immunoregulatory cytokines and growth factors through interactions with sialic acid-binding immunoglobulin-like lectins. We describe the processes, which modulate the interaction between sialylated carrier glycoproteins and their ligands, and illustrate that sialic acids could be targets of novel therapeutic strategies for treatment of cancer and immune diseases.
Collapse
Affiliation(s)
- Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| | - Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5165665931, Iran;
| | - Marwah Suliman Maashi
- Stem Cells and Regenerative Medicine Unit at King Fahad Medical Research Centre, Jeddah 11211, Saudi Arabia;
| | - Mahnaz Ghaebi
- Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan 4513956184, Iran;
| | - Abdolrahman Khezri
- Department of Biotechnology, Inland Norway University of Applied Sciences, 2418 Hamar, Norway;
| | - Martin R. Berger
- German Cancer Research Center, Toxicology and Chemotherapy Unit Heidelberg, 69120 Heidelberg, Germany;
| |
Collapse
|