1
|
Niu Y, Li A, Xu W, Zhang R, Mei R, Zhang L, Zhou F, Pan Q, Yan Y. Platelet activation stimulates macrophages to enhance ulcerative colitis through PF4/CXCR3 signaling. Int J Mol Med 2025; 55:78. [PMID: 40084691 PMCID: PMC11936483 DOI: 10.3892/ijmm.2025.5519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 02/19/2025] [Indexed: 03/16/2025] Open
Abstract
Platelets are involved in hemostasis and immune regulation, but little is currently known regarding their role in inflammatory bowel disease. In the present study, the mechanism by which platelet activation affects macrophage C‑X‑C motif chemokine receptor 3 (CXCR3) by releasing platelet factor 4 (PF4), thus aggravating ulcerative colitis (UC) disease progression, was investigated. A dextran sulfate sodium‑induced mouse model showed co‑localization of the platelet marker PF4 with the macrophage M1 marker inducible nitric oxide synthase. Furthermore, co‑culturing platelets with monocytes (THP‑1) in vitro led to the transformation of monocytes into macrophages, as well as the activation of macrophages exhibiting proinflammatory properties. Meanwhile, reverse transcription‑quantitative PCR (RT‑qPCR) showed that inflammatory factors, such as IL‑1β, IL‑6 and TNF‑α were significantly increased in macrophages after platelet co‑culture. It was therefore hypothesized that the PF4/CXCR3 pathway may serve an important role in cell‑to‑cell communication. Furthermore, intervention with PF4 in THP‑1 cells induced the M1 macrophage phenotype and inflammatory cytokine expression, which was consistent with co‑culturing, whereas inhibition of CXCR3 (AMG487) reversed the effects of PF4. In addition, following treatment with PF4, THP‑1 cells were found to be under oxidative stress and apoptosis was enhanced, as determined by detecting reactive oxygen species, mitochondrial membrane potential and Annexin‑V, as well as the classical apoptotic proteins Bcl‑2/Bax/caspase‑3 through western blotting. In addition, changes in MAPK and NF‑κB, two classic inflammatory signaling pathways, were detected. Furthermore, mice were treated with an anti‑platelet medication or CXCR3 inhibitor to observe in vivo inflammatory changes; through phenotypic assessment, immunofluorescence staining, RT‑qPCR and TUNEL assay, it was demonstrated that the PF4/CXCR3 pathway may aggravate inflammation in mice with UC. In conclusion, platelets and macrophages may interact in UC through the PF4/CXCR3 pathway to exacerbate inflammation, providing novel options for the treatment of UC.
Collapse
Affiliation(s)
- Yuxiao Niu
- Graduate School, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Anhong Li
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Zhoupu Hospital, Shanghai 201318, P.R. China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, P.R. China
| | - Weihua Xu
- Department of Pharmacy, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Rong Zhang
- Graduate School, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Ruya Mei
- Graduate School, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Zhoupu Hospital, Shanghai 201318, P.R. China
| | - Langhua Zhang
- School of Medical Technology, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Fenmin Zhou
- Department of Traditional Chinese Medicine, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| | - Qin Pan
- Shanghai Institute of Pediatric Research, Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai 200092, P.R. China
| | - Yuzhong Yan
- Graduate School, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Zhoupu Hospital, Shanghai 201318, P.R. China
- Graduate School, Shanghai University of Traditional Chinese Medicine, Shanghai 200030, P.R. China
- Department of Science Research, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai 201318, P.R. China
| |
Collapse
|
2
|
Alirezaylavasani A, Egner IM, Dahl B, Chopra A, de Matos Kasahara T, Goll GL, Jahnsen J, Grødeland G, Vaage JT, Lund-Johansen F, Holter JC, Halvorsen B, Jørgensen KK, Munthe LA, Kared H. Deficient SARS-CoV-2 hybrid immunity during inflammatory bowel disease. Clin Immunol 2025; 271:110404. [PMID: 39645159 DOI: 10.1016/j.clim.2024.110404] [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: 08/14/2024] [Revised: 10/28/2024] [Accepted: 11/19/2024] [Indexed: 12/09/2024]
Abstract
Patients with Inflammatory Bowel Disease (IBD) undergoing immunosuppressive therapies face heightened susceptibility to severe COVID-19. An in-depth understanding of systemic inflammation and cellular immune responses after SARS-CoV-2 vaccination and breakthrough infections (BTI) is required for optimizing vaccine strategies in this population. While the prevalence of high serological responders post- third COVID-19 vaccine dose was lower, and the antibody waning was higher in IBD patients than in healthy donors (HD), IBD patients showed an increase in anti-RBD Wild Type IgG levels and cross-reactive Spike -specific memory B cells following BTI. However, there was no significant enhancement in cellular immune responses against anti-SARS-CoV-2 post-BTI, with responses instead characterized by activation of SARS-CoV-2 specific and also bystander CD8 T cells. These results suggest a complex interaction between chronic inflammation in IBD and the generation of new immune responses, highlighting the need for tailored vaccine regimens and anti-inflammatory therapies to boost cellular immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Amin Alirezaylavasani
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Ingrid Marie Egner
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway
| | - Børresdatter Dahl
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway
| | - Adity Chopra
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway; ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | | | - Guro Løvik Goll
- Division of Rheumatology and Research, Diakonhjemmet Hospital, Oslo, Norway; Institute of Health and Society, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jørgen Jahnsen
- Department of Gastroenterology, Akershus University Hospital, Lørenskog, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Gunnveig Grødeland
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - John Torgils Vaage
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Fridtjof Lund-Johansen
- Department of Immunology, Oslo University Hospital, Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway; ImmunoLingo Convergence Center, University of Oslo, Oslo, Norway
| | - Jan Cato Holter
- Institute of Clinical Medicine, University of Oslo, Oslo, Norway.; Department of Microbiology, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | | | - Ludvig A Munthe
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| | - Hassen Kared
- Department of Immunology, Oslo University Hospital, Oslo, Norway; KG Jebsen Centre for B cell Malignancies, Institute of Clinical Medicine, University of Oslo, Norway; Precision Immunotherapy Alliance, University of Oslo, Oslo, Norway.
| |
Collapse
|
3
|
Nunes M, Vlok M, Proal A, Kell DB, Pretorius E. Data-independent LC-MS/MS analysis of ME/CFS plasma reveals a dysregulated coagulation system, endothelial dysfunction, downregulation of complement machinery. Cardiovasc Diabetol 2024; 23:254. [PMID: 39014464 PMCID: PMC11253362 DOI: 10.1186/s12933-024-02315-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] [Received: 03/22/2024] [Accepted: 06/16/2024] [Indexed: 07/18/2024] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a debilitating chronic condition that is characterized by unresolved fatigue, post-exertion symptom exacerbation (PESE), cognitive dysfunction, orthostatic intolerance, and other symptoms. ME/CFS lacks established clinical biomarkers and requires further elucidation of disease mechanisms. A growing number of studies demonstrate signs of hematological and cardiovascular pathology in ME/CFS cohorts, including hyperactivated platelets, endothelial dysfunction, vascular dysregulation, and anomalous clotting processes. To build on these findings, and to identify potential biomarkers that can be related to pathophysiology, we measured differences in protein expression in platelet-poor plasma (PPP) samples from 15 ME/CFS study participants and 10 controls not previously infected with SARS-CoV-2, using DIA LC-MS/MS. We identified 24 proteins that are significantly increased in the ME/CFS group compared to the controls, and 21 proteins that are significantly downregulated. Proteins related to clotting processes - thrombospondin-1 (important in platelet activation), platelet factor 4, and protein S - were differentially expressed in the ME/CFS group, suggestive of a dysregulated coagulation system and abnormal endothelial function. Complement machinery was also significantly downregulated, including C9 which forms part of the membrane attack complex. Additionally, we identified a significant upregulation of lactotransferrin, protein S100-A9, and an immunoglobulin variant. The findings from this experiment further implicate the coagulation and immune system in ME/CFS, and bring to attention the pathology of or imposed on the endothelium. This study highlights potential systems and proteins that require further research with regards to their contribution to the pathogenesis of ME/CFS, symptom manifestation, and biomarker potential, and also gives insight into the hematological and cardiovascular risk for ME/CFS individuals affected by diabetes mellitus.
Collapse
Affiliation(s)
- Massimo Nunes
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa
| | - Mare Vlok
- Central Analytical Facility: Mass Spectrometry, Stellenbosch University, Tygerberg Campus, Room 6054, Clinical Building, Francie Van Zijl Drive Tygerberg, Cape Town, 7505, South Africa
| | - Amy Proal
- PolyBio Research Foundation, 7900 SE 28th ST, Suite 412, Mercer Island, DC, 98040, USA
| | - Douglas B Kell
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
- The Novo Nordisk Foundation Centre for Biosustainability, Technical University of Denmark, Building 220, Chemitorvet 200, 2800, Kongens Lyngby, Denmark.
| | - Etheresia Pretorius
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Private Bag X1 Matieland, Stellenbosch, 7602, South Africa.
- Department of Biochemistry and Systems Biology, Institute of Systems, Molecular and Integrative Biology, Faculty of Health and Life Sciences, University of Liverpool, Crown St, Liverpool, L69 7ZB, UK.
| |
Collapse
|
4
|
Xu C, Song Z, Hu LT, Tong YH, Hu JY, Shen H. Abnormal platelet parameters in inflammatory bowel disease: a systematic review and meta-analysis. BMC Gastroenterol 2024; 24:214. [PMID: 38961334 PMCID: PMC11221001 DOI: 10.1186/s12876-024-03305-9] [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: 02/15/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Platelet dysfunction plays a critical role in the pathogenesis of inflammatory bowel disease (IBD). Despite clinical observations indicating abnormalities in platelet parameters among IBD patients, inconsistencies persist, and these parameters lack standardization for diagnosis or clinical assessment. METHODS A comprehensive search was conducted in the PubMed, Embase, Web of Science, and Cochrane Library databases for relevant articles published up to December 16th, 2023. A random-effects model was employed to pool the weighted mean difference (WMD) and 95% confidence interval (95% CI) of platelet count (PLT), mean platelet volume (MPV), platelet distribution width (PDW), and plateletcrit (PCT) between IBD patients and healthy controls, and subgroup analyses were performed. RESULTS The meta-analysis included 79 articles with 8,350 IBD patients and 13,181 healthy individuals. The results revealed significantly increased PLT and PCT levels (WMD: 69.910, 95% CI: 62.177, 77.643 109/L; WMD: 0.046%, 95% CI: 0.031%, 0.061%), and decreased MPV levels (WMD: -0.912, 95% CI: -1.086, -0.739 fL) in IBD patients compared to healthy individuals. No significant difference was found in PDW between the IBD and control groups (WMD: -0.207%, 95% CI: -0.655%, 0.241%). Subgroup analysis by disease type and disease activity showed no change in the differences for PLT, PCT, and MPV in the ulcerative colitis and Crohn's disease groups, as well as the active and inactive groups. Notably, the active group exhibited significantly lower PDW levels than the control group (WMD: -1.138%, 95% CI: -1.535%, -0.741%). CONCLUSIONS Compared with healthy individuals, IBD patients display significantly higher PLT and PCT and significantly lower MPV. Monitoring the clinical manifestations of platelet abnormalities serves as a valuable means to obtain diagnostic and prognostic information. Conversely, proactive measures should be taken to prevent the consequences of platelet abnormalities in individuals with IBD. SYSTEMATIC REVIEW REGISTRATION PROSPERO CRD42023493848.
Collapse
Affiliation(s)
- Cheng Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhen Song
- Nanjing University of Chinese Medicine, Nanjing, China
- Yancheng Binhai Hospital of Traditional Chinese Medicine, Yancheng, China
| | - Li-Ting Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Yi-Heng Tong
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Yi Hu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong Shen
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
5
|
Ngo AT, Skidmore A, Oberg J, Yarovoi I, Sarkar A, Levine N, Bochenek V, Zhao G, Rauova L, Kowalska MA, Eckart K, Mangalmurti NS, Rux A, Cines DB, Poncz M, Gollomp K. Platelet factor 4 limits neutrophil extracellular trap- and cell-free DNA-induced thrombogenicity and endothelial injury. JCI Insight 2023; 8:e171054. [PMID: 37991024 PMCID: PMC10721321 DOI: 10.1172/jci.insight.171054] [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: 03/31/2023] [Accepted: 10/02/2023] [Indexed: 11/23/2023] Open
Abstract
Plasma cell-free DNA (cfDNA), a marker of disease severity in sepsis, is a recognized driver of thromboinflammation and a potential therapeutic target. In sepsis, plasma cfDNA is mostly derived from neutrophil extracellular trap (NET) degradation. Proposed NET-directed therapeutic strategies include preventing NET formation or accelerating NET degradation. However, NET digestion liberates pathogens and releases cfDNA that promote thrombosis and endothelial cell injury. We propose an alternative strategy of cfDNA and NET stabilization with chemokine platelet factor 4 (PF4, CXCL4). We previously showed that human PF4 (hPF4) enhances NET-mediated microbial entrapment. We now show that hPF4 interferes with thrombogenicity of cfDNA and NETs by preventing their cleavage to short-fragment and single-stranded cfDNA that more effectively activates the contact pathway of coagulation. In vitro, hPF4 also inhibits cfDNA-induced endothelial tissue factor surface expression and von Willebrand factor release. In vivo, hPF4 expression reduced plasma thrombin-antithrombin (TAT) levels in animals infused with exogenous cfDNA. Following lipopolysaccharide challenge, Cxcl4-/- mice had significant elevation in plasma TAT, cfDNA, and cystatin C levels, effects prevented by hPF4 infusion. These results show that hPF4 interacts with cfDNA and NETs to limit thrombosis and endothelial injury, an observation of potential clinical benefit in the treatment of sepsis.
Collapse
Affiliation(s)
- Anh T.P. Ngo
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Abigail Skidmore
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jenna Oberg
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Irene Yarovoi
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Amrita Sarkar
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nate Levine
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Veronica Bochenek
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Guohua Zhao
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Lubica Rauova
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - M. Anna Kowalska
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Institute of Medical Biology, Polish Academy of Science, Lodz, Poland
| | | | | | - Ann Rux
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Douglas B. Cines
- Department of Medicine, and
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Mortimer Poncz
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kandace Gollomp
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
6
|
Boccatonda A, Balletta M, Vicari S, Hoxha A, Simioni P, Campello E. The Journey Through the Pathogenesis and Treatment of Venous Thromboembolism in Inflammatory Bowel Diseases: A Narrative Review. Semin Thromb Hemost 2023; 49:744-755. [PMID: 36455617 DOI: 10.1055/s-0042-1758869] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory disorders of the gastrointestinal tract including Crohn's disease and ulcerative colitis, which may result in several extraintestinal complications (∼20-30% of cases), such as increased risk of venous thromboembolism (VTE). The main pathophysiological mechanism of VTE is an inflammation-induced hypercoagulable state, and recent data have shown that endothelial dysregulation due to gut and systemic inflammation may also lead to a prothrombotic state. Several prothrombotic alterations have been described, such as the activation of the coagulation system, platelet abnormalities, and dysregulation of fibrinolysis. Furthermore, the dysregulation of the gut microbiome seems to play a vital role in increasing systemic inflammation and thus inducing a procoagulant state. Our review aims to examine the main correlations between IBD and VTE, the underlying pathophysiology, and current therapeutic options.
Collapse
Affiliation(s)
- Andrea Boccatonda
- Department of Internal Medicine, Bentivoglio Hospital, AUSL Bologna, Bologna, Italy
| | - Marco Balletta
- Department of Internal Medicine, Bologna University, Bologna, Italy
| | - Susanna Vicari
- Department of Internal Medicine, Bentivoglio Hospital, AUSL Bologna, Bologna, Italy
| | - Ariela Hoxha
- Hemorrhagic and Thrombotic Diseases Unit, Department of Medicine (DIMED), Padova University Hospital, Padova, Italy
| | - Paolo Simioni
- Hemorrhagic and Thrombotic Diseases Unit, Department of Medicine (DIMED), Padova University Hospital, Padova, Italy
| | - Elena Campello
- Hemorrhagic and Thrombotic Diseases Unit, Department of Medicine (DIMED), Padova University Hospital, Padova, Italy
| |
Collapse
|
7
|
Ngo ATP, Sarkar A, Yarovoi I, Levine ND, Bochenek V, Zhao G, Rauova L, Kowalska MA, Eckart K, Mangalmurti NS, Rux A, Cines DB, Poncz M, Gollomp K. Neutrophil extracellular trap stabilization by platelet factor 4 reduces thrombogenicity and endothelial cell injury. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.09.522931. [PMID: 36711969 PMCID: PMC9881987 DOI: 10.1101/2023.01.09.522931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Neutrophil extracellular traps (NETs) are abundant in sepsis, and proposed NET-directed therapies in sepsis prevent their formation or accelerate degradation. Yet NETs are important for microbial entrapment, as NET digestion liberates pathogens and NET degradation products (NDPs) that deleteriously promote thrombosis and endothelial cell injury. We proposed an alternative strategy of NET-stabilization with the chemokine, platelet factor 4 (PF4, CXCL4), which we have shown enhances NET-mediated microbial entrapment. We now show that NET compaction by PF4 reduces their thrombogenicity. In vitro, we quantified plasma thrombin and fibrin generation by intact or degraded NETs and cell-free (cf) DNA fragments, and found that digested NETs and short DNA fragments were more thrombogenic than intact NETs and high molecular weight genomic DNA, respectively. PF4 reduced the thrombogenicity of digested NETs and DNA by interfering, in part, with contact pathway activation. In endothelial cell culture studies, short DNA fragments promoted von Willebrand factor release and tissue factor expression via a toll-like receptor 9-dependent mechanism. PF4 blocked these effects. Cxcl4-/- mice infused with cfDNA exhibited higher plasma thrombin anti-thrombin (TAT) levels compared to wild-type controls. Following challenge with bacterial lipopolysaccharide, Cxcl4-/- mice had similar elevations in plasma TAT and cfDNA, effects prevented by PF4 infusion. Thus, NET-stabilization by PF4 prevents the release of short fragments of cfDNA, limiting the activation of the contact coagulation pathway and reducing endothelial injury. These results support our hypothesis that NET-stabilization reduces pathologic sequelae in sepsis, an observation of potential clinical benefit.
Collapse
Affiliation(s)
- Anh T. P. Ngo
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Amrita Sarkar
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Irene Yarovoi
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Nate D. Levine
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Veronica Bochenek
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Guohua Zhao
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Lubica Rauova
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - M. Anna Kowalska
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kaitlyn Eckart
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Nilam S. Mangalmurti
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ann Rux
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Douglas B. Cines
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Mortimer Poncz
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kandace Gollomp
- Division of Hematology, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
8
|
Okeke CO, Amilo GI, Manafa PO, Ibeh NC. Inflammation-mediated changes in haemostatic variables of pulmonary tuberculosis patients during treatment. Tuberculosis (Edinb) 2023; 138:102285. [PMID: 36436460 DOI: 10.1016/j.tube.2022.102285] [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: 09/01/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022]
Abstract
Tuberculosis (TB) disease is usually marked by inflammation which is closely linked to haemostasis both in health and disease. Close monitoring of haemostatic response to inflammatory changes during treatment is important to improve TB management. Here we studied associations between haemostatic markers and inflammatory cytokines in 60 TB-infected individuals, aged 18-65 years who received anti-TB therapy. They were recruited before commencement of therapy and followed up till completion of therapy after 6-months. The TNF-α, IL-6, IL-2 (pro-inflammatory cytokines) and P-selectin, GP IIb/IIIa, thrombopoietin (haemostatic variables) were significantly increased at 2 month into therapy compared to pre-treatment values and decreased at 6 month into therapy. Also at 6 month into therapy in comparison to 2-month into therapy, there were significant increase in IL-10 and TGF-β (anti-inflammatory cytokines) as well as a significant decline in PF-4. There were significant positive correlations between GP IIb/IIIa and TNF-α, IL-6 and PSEL, IL-6 and TPO, PF4 and TGF-β. Conclusively, the changes in the TNF-α, IL-6, IL-2 aligned with changes in the levels of P-selectin, GP IIb/IIIa, and TPO in the course of TB therapy. This may suggest that the levels of inflammatory cytokines are linked to the levels of these haemostatic variables in TB individuals.
Collapse
Affiliation(s)
- C O Okeke
- Department of Medical Laboratory Science, Faculty of Health Sciences and Technology, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus P.M.B. 5001, Anambra State, Nigeria.
| | - G I Amilo
- Department of Haematology, Faculty of Medicine, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus P.M.B. 5001, Anambra State, Nigeria
| | - P O Manafa
- Department of Medical Laboratory Science, Faculty of Health Sciences and Technology, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus P.M.B. 5001, Anambra State, Nigeria
| | - N C Ibeh
- Department of Medical Laboratory Science, Faculty of Health Sciences and Technology, College of Health Sciences, Nnamdi Azikiwe University, Nnewi Campus P.M.B. 5001, Anambra State, Nigeria
| |
Collapse
|
9
|
Furfaro F, Gabbiadini R, D'Amico F, Zilli A, Dal Buono A, Allocca M, Fiorino G, Danese S. Gastrointestinal System: COVID-19 and Potential Mechanisms Associated with Coagulopathy. Curr Drug Targets 2022; 23:1611-1619. [PMID: 36154571 DOI: 10.2174/1389450123666220922095913] [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: 02/16/2022] [Revised: 08/21/2022] [Accepted: 08/25/2022] [Indexed: 01/25/2023]
Abstract
SARS-CoV-2 is a novel coronavirus that expanded worldwide, generating a pandemic of acute respiratory syndrome called "coronavirus disease 2019" (COVID-19), which resulted in a global health crisis. The spectrum of COVID-19 manifestations ranges from none or mild symptoms to severe respiratory failure associated with systemic manifestations, mostly gastrointestinal symptoms. Hypercoagulability is an important feature of COVID-19 disease, which can potentially influence patients' prognosis. Therefore, gastroenterologists should focus on subjects with concomitant hypercoagulable gastrointestinal disorders as they may display a higher risk of thrombotic complications during SARS-CoV-2 infection. The aim of this review is to summarize the available evidence regarding the interplay of the prothrombotic pathogenetic mechanisms of both COVID-19 and hypercoagulable digestive diseases and the possible clinical implications. We summarized the potential interplay of prothrombotic mechanisms of both COVID-19 and hypercoagulable digestive diseases in the graphical abstract.
Collapse
Affiliation(s)
- Federica Furfaro
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | | | - Ferdinando D'Amico
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Alessandra Zilli
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Arianna Dal Buono
- IBD Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Mariangela Allocca
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Gionata Fiorino
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| | - Silvio Danese
- Gastroenterology and Endoscopy, IRCCS Ospedale San Raffaele, Milan, Italy.,University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
10
|
Seo IH, Lee YJ. Usefulness of Complete Blood Count (CBC) to Assess Cardiovascular and Metabolic Diseases in Clinical Settings: A Comprehensive Literature Review. Biomedicines 2022; 10:2697. [PMID: 36359216 PMCID: PMC9687310 DOI: 10.3390/biomedicines10112697] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/07/2022] [Accepted: 10/20/2022] [Indexed: 08/03/2023] Open
Abstract
Complete blood count (CBC) is one of the most common blood tests requested by clinicians and evaluates the total numbers and characteristics of cell components in the blood. Recently, many investigations have suggested that the risk of cancer, cardiovascular disease (CVD), arteriosclerosis, type 2 diabetes (T2DM), and metabolic syndrome can be predicted using CBC components. This review introduces that white blood cell (WBC), neutrophil-to-lymphocyte ratio (NLR), hemoglobin (Hb), mean corpuscular volume (MCV), red cell distribution width (RDW), platelet count, mean platelet volume (MPV), and platelet-to-lymphocyte ratio (PLR) are useful markers to predict CVD and metabolic diseases. Furthermore, we would like to support various uses of CBC by organizing pathophysiology that can explain the relationship between CBC components and diseases.
Collapse
|
11
|
Kirwan DE, Chong DLW, Friedland JS. Platelet Activation and the Immune Response to Tuberculosis. Front Immunol 2021; 12:631696. [PMID: 34093524 PMCID: PMC8170316 DOI: 10.3389/fimmu.2021.631696] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/04/2021] [Indexed: 12/24/2022] Open
Abstract
In 2019 10 million people developed symptomatic tuberculosis (TB) disease and 1.2 million died. In active TB the inflammatory response causes tissue destruction, which leads to both acute morbidity and mortality. Tissue destruction in TB is driven by host innate immunity and mediated via enzymes, chiefly matrix metalloproteinases (MMPs) which are secreted by leukocytes and stromal cells and degrade the extracellular matrix. Here we review the growing evidence implicating platelets in TB immunopathology. TB patients typically have high platelet counts, which correlate with disease severity, and a hypercoagulable profile. Platelets are present in human TB granulomas and platelet-associated gene transcripts are increased in TB patients versus healthy controls. Platelets most likely drive TB immunopathology through their effect on other immune cells, particularly monocytes, to lead to upregulation of activation markers, increased MMP secretion, and enhanced phagocytosis. Finally, we consider current evidence supporting use of targeted anti-platelet agents in the treatment of TB due to growing interest in developing host-directed therapies to limit tissue damage and improve treatment outcomes. In summary, platelets are implicated in TB disease and contribute to MMP-mediated tissue damage via their cellular interactions with other leukocytes, and are potential targets for novel host-directed therapies.
Collapse
Affiliation(s)
- Daniela E Kirwan
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| | - Deborah L W Chong
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| | - Jon S Friedland
- Institute for Infection & Immunity, St. George's, University of London, London, United Kingdom
| |
Collapse
|
12
|
Zamora C, Cantó E, Vidal S. The Dual Role of Platelets in the Cardiovascular Risk of Chronic Inflammation. Front Immunol 2021; 12:625181. [PMID: 33868242 PMCID: PMC8046936 DOI: 10.3389/fimmu.2021.625181] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 02/15/2021] [Indexed: 11/25/2022] Open
Abstract
Patients with chronic inflammatory diseases often exhibit cardiovascular risk. This risk is associated with the systemic inflammation that persists in these patients, causing a sustained endothelial activation. Different mechanisms have been considered responsible for this systemic inflammation, among which activated platelets have been regarded as a major player. However, in recent years, the role of platelets has become controversial. Not only can this subcellular component release pro- and anti-inflammatory mediators, but it can also bind to different subsets of circulating lymphocytes, monocytes and neutrophils modulating their function in either direction. How platelets exert this dual role is not yet fully understood.
Collapse
Affiliation(s)
- Carlos Zamora
- Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Elisabet Cantó
- Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Sílvia Vidal
- Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| |
Collapse
|
13
|
Comprehensive multi-omics analysis of G6PC3 deficiency-related congenital neutropenia with inflammatory bowel disease. iScience 2021; 24:102214. [PMID: 33748703 PMCID: PMC7960940 DOI: 10.1016/j.isci.2021.102214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/29/2020] [Accepted: 02/17/2021] [Indexed: 11/26/2022] Open
Abstract
Autosomal recessive mutations in G6PC3 cause isolated and syndromic congenital neutropenia which includes congenital heart disease and atypical inflammatory bowel disease (IBD). In a highly consanguineous pedigree with novel mutations in G6PC3 and MPL, we performed comprehensive multi-omics analyses. Structural analysis of variant G6PC3 and MPL proteins suggests a damaging effect. A distinct molecular cytokine profile (cytokinome) in the affected proband with IBD was detected. Liquid chromatography-mass spectrometry-based proteomics analysis of the G6PC3-deficient plasma samples identified 460 distinct proteins including 75 upregulated and 73 downregulated proteins. Specifically, the transcription factor GATA4 and LST1 were downregulated while platelet factor 4 (PF4) was upregulated. GATA4 and PF4 have been linked to congenital heart disease and IBD respectively, while LST1 may have perturbed a variety of essential cell functions as it is required for normal cell-cell communication. Together, these studies provide potentially novel insights into the pathogenesis of syndromic congenital G6PC3 deficiency. Multi-omics approaches identify unique signatures Whole-exome sequencing reveals distinct cytokine profiles Expression of GATA4, PF4, and LST1 is dysregulated
Collapse
|
14
|
Anti-CXCL4 Antibody Reactivity Is Present in Systemic Sclerosis (SSc) and Correlates with the SSc Type I Interferon Signature. Int J Mol Sci 2020; 21:ijms21145102. [PMID: 32707718 PMCID: PMC7404208 DOI: 10.3390/ijms21145102] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 07/07/2020] [Accepted: 07/14/2020] [Indexed: 12/26/2022] Open
Abstract
Systemic sclerosis (SSc) is characterized by skin/internal organ fibrosis, vasculopathy and autoimmunity. Chemokine (C-X-C motif) ligand 4 (CXCL4) is an SSc biomarker, predicting unfavorable prognosis and lung fibrosis. CXCL4 binds DNA/RNA and favors interferon (IFN)-α production by plasmacytoid dendritic cells (pDCs), contributing to the type I IFN (IFN-I) signature in SSc patients. However, whether CXCL4 is an autoantigen in SSc is unknown. Here, we show that at least half of SSc patients show consistent antibody reactivity to CXCL4. T-cell proliferation to CXCL4, tested in a limited number of patients, correlates with anti-CXCL4 antibody reactivity. Antibodies to CXCL4 mostly correlate with circulating IFN-α levels and are significantly higher in patients with lung fibrosis in two independent SSc cohorts. Antibodies to CXCL4 implement the CXCL4-DNA complex's effect on IFN-α production by pDCs; CXCL4-DNA/RNA complexes stimulate purified human B-cells to become antibody-secreting plasma cells in vitro. These data indicate that CXCL4 is indeed an autoantigen in SSc and suggest that CXCL4, and CXCL4-specific autoantibodies, can fuel a harmful loop: CXCL4-DNA/RNA complexes induce IFN-α in pDCs and direct B-cell stimulation, including the secretion of anti-CXCL4 antibodies. Anti-CXCL4 antibodies may further increase pDC stimulation and IFN-α release in vivo, creating a vicious cycle which sustains the SSc IFN-I signature and general inflammation.
Collapse
|
15
|
Ma W, Gil HJ, Escobedo N, Benito-Martín A, Ximénez-Embún P, Muñoz J, Peinado H, Rockson SG, Oliver G. Platelet factor 4 is a biomarker for lymphatic-promoted disorders. JCI Insight 2020; 5:135109. [PMID: 32525843 DOI: 10.1172/jci.insight.135109] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 06/03/2020] [Indexed: 01/08/2023] Open
Abstract
Genetic or acquired defects of the lymphatic vasculature often result in disfiguring, disabling, and, occasionally, life-threatening clinical consequences. Advanced forms of lymphedema are readily diagnosed clinically, but more subtle presentations often require invasive imaging or other technologies for a conclusive diagnosis. On the other hand, lipedema, a chronic lymphatic microvascular disease with pathological accumulation of subcutaneous adipose tissue, is often misdiagnosed as obesity or lymphedema; currently there are no biomarkers or imaging criteria available for a conclusive diagnosis. Recent evidence suggests that otherwise-asymptomatic defective lymphatic vasculature likely contributes to an array of other pathologies, including obesity, inflammatory bowel disease, and neurological disorders. Accordingly, identification of biomarkers of lymphatic malfunction will provide a valuable resource for the diagnosis and clinical differentiation of lymphedema, lipedema, obesity, and other potential lymphatic pathologies. In this paper, we profiled and compared blood plasma exosomes isolated from mouse models and from human subjects with and without symptomatic lymphatic pathologies. We identified platelet factor 4 (PF4/CXCL4) as a biomarker that could be used to diagnose lymphatic vasculature dysfunction. Furthermore, we determined that PF4 levels in circulating blood plasma exosomes were also elevated in patients with lipedema, supporting current claims arguing that at least some of the underlying attributes of this disease are also the consequence of lymphatic defects.
Collapse
Affiliation(s)
- Wanshu Ma
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Hyea Jin Gil
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Noelia Escobedo
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois, USA
| | - Alberto Benito-Martín
- Children's Cancer & Blood Foundation Laboratories, Departments of Pediatrics and Cell and Developmental Biology, Weill Cornell Medicine, New York, USA
| | - Pilar Ximénez-Embún
- Proteomics Unit - ProteoRed-ISCIII, Spanish National Cancer Research Centre, Madrid, Spain
| | - Javier Muñoz
- Proteomics Unit - ProteoRed-ISCIII, Spanish National Cancer Research Centre, Madrid, Spain
| | - Héctor Peinado
- Microenvironment & Metastasis Group, Molecular Oncology Program, Spanish National Cancer Research Center, Madrid, Spain
| | - Stanley G Rockson
- Division of Cardiovascular Medicine, Center for Lymphatic and Venous Disorders, Stanford University School of Medicine, Stanford, California, USA
| | - Guillermo Oliver
- Center for Vascular and Developmental Biology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
16
|
Breidert M, Eftekhari P, Louis F, Rotoiu C, Rath T, Neurath MF, Atreya R. Functional Molecular Network Analysis Enables Prediction of Response to Vedolizumab Therapy in Anti-TNF Refractory IBD Patients. CROHNS & COLITIS 360 2020; 2:otaa037. [PMID: 32776006 PMCID: PMC7397736 DOI: 10.1093/crocol/otaa037] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Indexed: 02/07/2023]
Abstract
Background We applied for the first time 2 label-free technologies, physiological intermolecular modulation spectroscopy (PIMS) and nematic protein organization technic (NPOT) in anti-tumor necrosis factor (TNF) refractory inflammatory bowel disease (IBD) patients to identify clinical responders to vedolizumab therapy and elucidate their underlying functional molecular network. Methods PIMS analysis was performed in peripheral blood taken prior to the first vedolizumab application in 20 IBD patients (Crohn disease n = 13; ulcerative colitis n = 7) refractory to at least 1 previous anti-TNF agent therapy. Peripheral blood taken from clinical responders and nonresponders at week 14 of vedolizumab therapy were additionally subjected to NPOT analysis. Response to therapy was assessed by respective clinical disease activity scores (partial Mayo Score and Harvey-Bradshaw Index). Results Clinical response to vedolizumab treatment was observed in 7 of 13 Crohn disease and 4 of 7 ulcerative colitis patients at week 14. Response to therapy was accurately predicted by PIMS blood analysis in 100% of ulcerative colitis and 77% of Crohn disease patients. Overall prediction of clinical response with PIMS blood analysis was achieved with a 89% positive predictive value and a 82% negative predictive value. NPOT analysis revealed the heightened expression of the proteins ITGB7, ITGAV, ITG3, PF4, and ASGH in the peripheral blood of vedolizumab responders compared to nonresponders. Conclusions PIMS analysis of the blood of anti-TNF refractory IBD patients was able to stratify responders to vedolizumab therapy with high accuracy and specificity. NPOT technology could decipher underling molecular networks in the blood of responders, enabling subsequent personalized therapeutic approaches in IBD.
Collapse
Affiliation(s)
- Matthias Breidert
- Department of Gastroenterology and Hepatology, City Hospital Waid and Triemli, Zürich, Switzerland
| | | | - François Louis
- Inoviem Scientific, BIOPARC 3, Illkirch-Graffenstaden, France
| | - Claudia Rotoiu
- Department of Gastroenterology and Hepatology, City Hospital Waid and Triemli, Zürich, Switzerland
| | - Timo Rath
- Department of Medicine I, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine I, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine I, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.,Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-Universität, Erlangen-Nürnberg, Erlangen, Germany.,The Transregio 241 IBDome Consortium
| |
Collapse
|
17
|
Subclinical endometritis in dairy cattle is associated with distinct mRNA expression patterns in blood and endometrium. PLoS One 2019; 14:e0220244. [PMID: 31374089 PMCID: PMC6677313 DOI: 10.1371/journal.pone.0220244] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 07/11/2019] [Indexed: 02/06/2023] Open
Abstract
Cattle with subclinical endometritis (SCE) are sub-fertile and diagnosing subclinical uterine disease remains a challenge. The hypothesis for this study was that endometrial inflammation is reflected in mRNA expression patterns of peripheral blood leucocytes. Transcriptome profiles were evaluated in healthy cows and in cows with SCE using circulating white blood cells (WBC) and endometrial biopsy samples collected from the same animals at 45–55 days postpartum. Bioinformatic analyses of microarray-based transcriptional data identified gene profiles associated with distinct biological functions in circulating WBC and endometrium. In circulating WBC, SCE promotes a pro-inflammatory environment, whereas functions related to tissue remodeling are also affected in the endometrium. Nineteen differentially expressed genes associated with SCE were common to both circulating WBC and the endometrium. Among these genes, transcript abundance of immune factors C3, C2, LTF, PF4 and TRAPPC13 were up-regulated in SCE cows at 45–55 days postpartum. Moreover, mRNA expression of C3, CXCL8, LTF, TLR2 and TRAPPC13 was temporally regulated during the postpartum period in circulating WBC of healthy cows compared with SCE cows. This observation might indicate an advantageous modulation of the immune system in healthy animals. The transcript abundance of these genes represents a potential source of indicators for postpartum uterine health.
Collapse
|
18
|
Boshagh MA, Foroutan P, Moloudi MR, Fakhari S, Malakouti P, Nikkhoo B, Jalili A. ELR positive CXCL chemokines are highly expressed in an animal model of ulcerative colitis. J Inflamm Res 2019; 12:167-174. [PMID: 31417300 PMCID: PMC6599894 DOI: 10.2147/jir.s203714] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 05/16/2019] [Indexed: 12/21/2022] Open
Abstract
Background: The presence of neutrophil-rich inflammation in colon tissues of patients with ulcerative colitis (UC) is one of the most important histological characteristics of this disease. However, the expression of CXCL chemokines governing the infiltration of neutrophils in UC has not been well elucidated. Materials and methods: In this experimental study, the UC model was induced in Wistar rats by administration of 2 mL 4% acetic acid into the large colon through the rectum. Animals were anesthetized after 48 hrs; their colon tissue samples were isolated for macroscopic and histopathological examinations. The expression of CXCL family was assessed by reverse transcription polymerase chain reaction (qRT-PCR) technique. Results: Heavy infiltration of neutrophils, coagulation necrosis, and ulcers were observed in H&E staining, which pathologically proved the UC model. qRT-PCR results showed that ELR+ CXC chemokines such as CXCL6 and CXCL3 had the highest expression in the UC group, which was 49 and 28 times higher than that of the control group, respectively. In addition, other chemokines of this group including CXCL1, CXCL2, and CXCL7 had a significant increase compared to the control group (P≤0.05). However, ELR− CXC chemokines such as CXCL4, CXCL13, and CXCL16 showed a smaller upregulation, while CXCL14 chemokine showed a significant decrease compared to the control group (P≤0.05). However, the expression of CXCL9-12 and CXCL17 did not change. Conclusion: The results showed that the ELR+ CXC chemokines, especially CXCL6 and CXCL3, many involved in the pathogenesis of UC; therefore, CXCL6 and CXCL3 chemokines can be used as therapeutic targets for UC, although more studies using human samples are required.
Collapse
Affiliation(s)
- Mohammad Amin Boshagh
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Immunology & Hematology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Poorya Foroutan
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Immunology & Hematology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Mohammad Raman Moloudi
- Liver and Digestive Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Shohreh Fakhari
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Parisa Malakouti
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Bahram Nikkhoo
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Ali Jalili
- Cancer and Immunology Research Center, Kurdistan University of Medical Sciences, Sanandaj, Iran.,Department of Immunology & Hematology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
19
|
Lande R, Lee EY, Palazzo R, Marinari B, Pietraforte I, Santos GS, Mattenberger Y, Spadaro F, Stefanantoni K, Iannace N, Dufour AM, Falchi M, Bianco M, Botti E, Bianchi L, Alvarez M, Riccieri V, Truchetet ME, C.L. Wong G, Chizzolini C, Frasca L. CXCL4 assembles DNA into liquid crystalline complexes to amplify TLR9-mediated interferon-α production in systemic sclerosis. Nat Commun 2019; 10:1731. [PMID: 31043596 PMCID: PMC6494823 DOI: 10.1038/s41467-019-09683-z] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 03/23/2019] [Indexed: 01/17/2023] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune disease characterized by fibrosis and vasculopathy. CXCL4 represents an early serum biomarker of severe SSc and likely contributes to inflammation via chemokine signaling pathways, but the exact role of CXCL4 in SSc pathogenesis is unclear. Here, we elucidate an unanticipated mechanism for CXCL4-mediated immune amplification in SSc, in which CXCL4 organizes "self" and microbial DNA into liquid crystalline immune complexes that amplify TLR9-mediated plasmacytoid dendritic cell (pDC)-hyperactivation and interferon-α production. Surprisingly, this activity does not require CXCR3, the CXCL4 receptor. Importantly, we find that CXCL4-DNA complexes are present in vivo and correlate with type I interferon (IFN-I) in SSc blood, and that CXCL4-positive skin pDCs coexpress IFN-I-related genes. Thus, we establish a direct link between CXCL4 overexpression and the IFN-I-gene signature in SSc and outline a paradigm in which chemokines can drastically modulate innate immune receptors without being direct agonists.
Collapse
Affiliation(s)
- Roberto Lande
- National Center for Drug Research and Evaluation, Pharmacological research and experimental therapy UNIT, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Ernest Y. Lee
- Department of Bioengineering, Department of Chemistry & Biochemistry, and California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Raffaella Palazzo
- National Center for Drug Research and Evaluation, Pharmacological research and experimental therapy UNIT, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Barbara Marinari
- Dermatology Unit, Department of Systems Medicine, University of Tor Vergata, Rome, 00133 Italy
| | - Immacolata Pietraforte
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy
| | - Giancarlo Santiago Santos
- Department of Bioengineering, Department of Chemistry & Biochemistry, and California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Yves Mattenberger
- Department of Microbiol and Molecular Medicine, University of Geneva, CH-1211 Geneva, Switzerland
| | - Francesca Spadaro
- Istituto Superiore di Sanità, Confocal Microscopy Unit, Core Facilities, Rome, 00161 Italy
| | - Katia Stefanantoni
- Division of Rheumatology, Internal Medicine and Medical Specialties, University La Sapienza, 00161 Rome, Italy
| | - Nicoletta Iannace
- Division of Rheumatology, Internal Medicine and Medical Specialties, University La Sapienza, 00161 Rome, Italy
| | - Aleksandra Maria Dufour
- Immunology & Allergy and Immunology & Pathology, University Hospital and School of Medicine, CH-1211 Geneva, Switzerland
| | - Mario Falchi
- Istituto Superiore di Sanità, National AIDS Center, Rome, 00161 Italy
| | - Manuela Bianco
- National Center for Drug Research and Evaluation, Pharmacological research and experimental therapy UNIT, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
| | - Elisabetta Botti
- Dermatology Unit, Department of Systems Medicine, University of Tor Vergata, Rome, 00133 Italy
| | - Luca Bianchi
- Dermatology Unit, Department of Systems Medicine, University of Tor Vergata, Rome, 00133 Italy
| | - Montserrat Alvarez
- Immunology & Allergy and Immunology & Pathology, University Hospital and School of Medicine, CH-1211 Geneva, Switzerland
| | - Valeria Riccieri
- Division of Rheumatology, Internal Medicine and Medical Specialties, University La Sapienza, 00161 Rome, Italy
| | - Marie-Elise Truchetet
- Division of Rheumatology and immunoConcept, University Hospital, Bordeaux, 33076 France
| | - Gerard C.L. Wong
- Department of Bioengineering, Department of Chemistry & Biochemistry, and California NanoSystems Institute, University of California, Los Angeles, CA 90095 USA
| | - Carlo Chizzolini
- Immunology & Allergy and Immunology & Pathology, University Hospital and School of Medicine, CH-1211 Geneva, Switzerland
| | - Loredana Frasca
- National Center for Drug Research and Evaluation, Pharmacological research and experimental therapy UNIT, Istituto Superiore di Sanità (ISS), 00161 Rome, Italy
- Immunology & Allergy and Immunology & Pathology, University Hospital and School of Medicine, CH-1211 Geneva, Switzerland
| |
Collapse
|
20
|
Marlicz W, Skonieczna-Żydecka K, Dabos KJ, Łoniewski I, Koulaouzidis A. Emerging concepts in non-invasive monitoring of Crohn's disease. Therap Adv Gastroenterol 2018; 11:1756284818769076. [PMID: 29707039 PMCID: PMC5912292 DOI: 10.1177/1756284818769076] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Inflammatory bowel disease (IBD) is an umbrella term for Crohn's disease (CD) and ulcerative colitis (UC). In light of evolving epidemiology of CD, its clinical management is still complex and remains a challenge for contemporary physicians. With the advent of new diagnostic and treatment paradigms, there is a growing need for new biomarkers to guide decision-making, differential diagnosis, disease activity monitoring, as well as prognosis. However, both clinical and endoscopic scoring systems, widely utilized for disease monitoring and prognosis, have drawbacks and limitations. In recent years, biochemical peptides have become available for IBD monitoring and more frequently used as surrogate markers of gut inflammation. Emerging concepts that revolve around molecular, stem cell, epigenetic, microbial or metabolomic pathways associated with vascular and epithelial gut barrier could lead to development of new CD biomarkers. Measurement of cell-derived microvesicles (MVs) in the blood of IBD patients is another emerging concept helpful in future disease management. In this review, we discuss novel concepts of non-invasive biomarkers, which may become useful in monitoring of CD activity and prognosis. We discuss metabolomics as a new powerful tool for clinicians to guide differential IBD diagnosis. In the coming years, new developments of prognostic tools are expected, aiming for breakthroughs in the management of patients with CD.
Collapse
Affiliation(s)
- Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University, Unii Lubelskiej 1, 71-252 Szczecin, Poland
| | | | | | - Igor Łoniewski
- Department of Biochemistry and Human Nutrition, Pomeranian Medical University, Szczecin, Poland
- Sanprobi Sp. z o.o. Sp. K., Szczecin, Poland
| | | |
Collapse
|
21
|
Chen GL, Xiong DQ, Jiang ZY, Zhang CM, Hu QY. Diagnostic value of platelet-to-lymphocyte ratio and neutrophil-to-lymphocyte ratio in Crohn's disease. Shijie Huaren Xiaohua Zazhi 2018; 26:93-98. [DOI: 10.11569/wcjd.v26.i2.93] [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] [Indexed: 02/06/2023] Open
Abstract
AIM To investigate the diagnostic efficacy of platelet-to-lymphocyte ratio (PLR) and neutrophil-to-lymphocyte ratio (NLR) in patients with Crohn's disease (CD).
METHODS EDTA anticoagulant venous blood was collected from CD patients (n = 60) and healthy controls (n = 60), and routine blood test was performed to obtain the neutrophilic granulocyte count, lymphocyte count, and platelet count. Then, the PLR and NLR were calculated. The sensitivity (Sen), specificity (Spe), and cut-off values were calculated by receiver operator characteristic (ROC) curve analysis.
RESULTS CD patients had significantly higher PLR (181 ± 45.12 vs 86 ± 32.78) and NLR (3.07 ± 1.02 vs 2.52 ± 1.14) than healthy controls (P < 0.05). The cut-off values for PLR and NLR were 133.12 (Sen: 76.21%; Spe: 84.53%) and 2.85 (Sen: 69.16%; Spe: 76.24%), respectively, and the areas under the ROC curves for PLR and NLR were 0.764 and 0.828, respectively.
CONCLUSION PLR and NLR might be effective and low-cost biomarkers for distinguishing CD patients from healthy controls.
Collapse
Affiliation(s)
- Gao-Li Chen
- Department of Laboratory Medicine, Teaching Hospital of Chengdu University of TCM, Chengdu 610072, Sichuan Province, China
| | - Da-Qian Xiong
- Department of Laboratory Medicine, Teaching Hospital of Chengdu University of TCM, Chengdu 610072, Sichuan Province, China
| | - Ze-You Jiang
- Department of Laboratory Medicine, Teaching Hospital of Chengdu University of TCM, Chengdu 610072, Sichuan Province, China
| | - Chao-Ming Zhang
- Department of Laboratory Medicine, Teaching Hospital of Chengdu University of TCM, Chengdu 610072, Sichuan Province, China
| | - Qiong-Ying Hu
- Department of Laboratory Medicine, Teaching Hospital of Chengdu University of TCM, Chengdu 610072, Sichuan Province, China
| |
Collapse
|