1
|
Thaler J, Tripisciano C, Kraemmer D, Hau C, Samadi N, Ruf W, Pabinger I, Knoebl P, Nieuwland R, Ay C. Saliva of persons with hemophilia A triggers coagulation via extrinsic tenase complexes. Blood 2024; 144:2666-2677. [PMID: 39437554 DOI: 10.1182/blood.2024025093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 10/25/2024] Open
Abstract
ABSTRACT Human saliva contains extracellular vesicles (EVs). These EVs expose extrinsic tenase complexes of tissue factor (TF) and activated factor VII (FVIIa), and trigger blood coagulation. Here, we show that EVs exposing extrinsic tenase complexes are also present in saliva of persons with severe hemophilia A, that is, persons with FVIII deficiency. Addition of these salivary EVs to autologous FVIII-deficient blood results in FXa generation, thereby compensating for the lack of FXa generation via intrinsic tenase (FVIIIa/FIXa) complexes. Consistently, in our retrospective analysis of persons with severe hemophilia A who do not receive prophylactic FVIII substitution, oropharyngeal mucosal bleedings are infrequent and self-limited. Conversely, in saliva of persons with severe FVII deficiency, in whom oropharyngeal bleedings are prevalent, functional extrinsic tenase complexes are absent, because EVs lack FVII. Saliva of persons with severe FVII deficiency is unable to restore blood coagulation, which is because of the absence of FVII in both their saliva and blood. Picomolar levels of recombinant FVIIa can restore the coagulant potential of saliva of persons with FVII deficiency. Taken together, our findings may explain the paucity of oropharyngeal bleedings in persons with hemophilia A as well as the occurrence of such bleedings in persons with severe FVII deficiency.
Collapse
Affiliation(s)
- Johannes Thaler
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Carla Tripisciano
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Daniel Kraemmer
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Chi Hau
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Nazanin Samadi
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, University Medical Center, Johannes Gutenberg University Mainz, Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA
| | - Ingrid Pabinger
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Paul Knoebl
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Vesicle Center, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Cihan Ay
- Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
2
|
Tran MN, Jun HJ, Lee S. Identifying the molecular mechanism of blood stasis syndrome through the symptom phenotype-genotype association approach. Medicine (Baltimore) 2024; 103:e40717. [PMID: 39654208 PMCID: PMC11631032 DOI: 10.1097/md.0000000000040717] [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: 06/12/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
In traditional medicine (TM), blood stasis syndrome (BSS) is characterized by insufficient blood flow, resulting in a group of symptoms such as fixed pain, a dark complexion, bleeding, and an astringent pulse. While BSS pathology has been previously explored, its molecular mechanisms remain elusive owing to challenges in linking TM symptoms to genes. Our study aimed to elucidate the mechanisms underlying BSS using a phenotype-genotype association approach. We extracted BSS symptoms from various studies, linked them to medical terms using a Unified Medical Language System, and connected these terms to genes in the DisGeNET database. The molecular network patterns of BSS symptoms were revealed through analyzing protein-protein interactions and symptom-gene associations. Our findings revealed 1325 associations between 16 BSS symptoms comprising 32 concept-unified identifier terms and 937 genes. Network analysis highlighted the centrality of JAK2, ITGB3, and F2, associated with multiple BSS symptoms (≥5 concept-unified identifier terms) and numerous protein interactions (≥20 interactions). Enrichment analysis indicated the involvement of BSS genes in the immune system (P-value = 4.49e-14) and hemostasis (P-value = 1.28e-07) pathways. BSS symptoms were linked to genes regulating blood coagulation, immune responses, blood flow, and inflammatory reactions. This approach may be extended to establish genotype networks for understanding TM pattern identifications, which are composed of diverse groups of symptoms, for personalized diagnosis and treatment.
Collapse
Affiliation(s)
- Minh Nhat Tran
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
- Faculty of Traditional Medicine, Hue University of Medicine and Pharmacy, Hue University, Thua Thien Hue, Vietnam
| | - Hyeong Joon Jun
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Sanghun Lee
- Korean Medicine Data Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
- Korean Convergence Medical Science, University of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
3
|
Gronbeck C, Hadfield MJ, Grant-Kels JM. Dermatologic toxicities of antibody-drug conjugates. J Am Acad Dermatol 2024; 91:1177-1188. [PMID: 39182677 DOI: 10.1016/j.jaad.2024.08.036] [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: 03/17/2024] [Revised: 08/01/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024]
Abstract
Antibody-drug conjugates (ADCs) are a new and emerging category of oncologic treatments that combine the target specificity of a monoclonal antibody with a cytotoxic payload. These drugs are associated with unique cutaneous toxicities that vary across agents. Currently, there are eleven ADCs with regulatory approval for solid and liquid tumors and over 80 ADCs currently in clinical development, it is critical for dermatologists to recognize and appropriately mitigate the cutaneous toxicities associated with these therapies. This clinical review will summarize the novel mechanisms and indications of approved ADCs, discuss dermatologic toxicities demonstrated in clinical trials and postmarketing studies, and impart recognition and management guidance when encountering these reactions to help maintain patients safely and comfortably on their medications.
Collapse
Affiliation(s)
- Christian Gronbeck
- Department of Dermatology, University of Connecticut Health Center, Farmington, Connecticut
| | - Matthew J Hadfield
- Dermatology Medical Oncology, Brown University/Legoretta Cancer Center, Providence, Rhode Island
| | - Jane M Grant-Kels
- Department of Dermatology, University of Connecticut Health Center, Farmington, Connecticut; Department of Dermatology, University of Florida College of Medicine, Gainesville, Florida.
| |
Collapse
|
4
|
Roy A, Kumar Y, Verma N. Coagulopathy in acute liver failure. Best Pract Res Clin Gastroenterol 2024; 73:101956. [PMID: 39709211 DOI: 10.1016/j.bpg.2024.101956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 10/22/2024] [Indexed: 12/23/2024]
Abstract
Acute liver failure (ALF) is a rare but rapidly progressing syndrome, marked by severe liver dysfunction and altered mental status. While definitions of ALF vary across different guidelines, with timelines ranging from 4 to 26 weeks between jaundice onset and encephalopathy, the key defining features remain encephalopathy and coagulopathy. Elevated coagulation markers, particularly prothrombin time and international normalized ratio, have traditionally been associated with bleeding risks. However, emerging evidence suggests a rebalanced state of coagulation in ALF, similar to cirrhosis, where bleeding risks-both spontaneous and procedural-are surprisingly low. Viscoelastic hemostatic assays and thrombin generation assays further confirm this rebalanced hemostatic state. Current guidelines for correcting coagulopathy in ALF remain limited, typically reserved for active bleeding or prior to high-risk invasive procedures.
Collapse
Affiliation(s)
- Akash Roy
- Institute of Gastrosciences and Liver Transplantation, Apollo Multi-speciality Hospitals, Kolkatta, India
| | - Yogendra Kumar
- Department of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nipun Verma
- Department of Hepatology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
5
|
Liu H, Xiong W, Zhong W, Hu Y. NOVEL ACTIVE PROTEINS FOR SEPSIS PROGNOSIS REVEALED THROUGH ScRNA-seq AND QUANTITATIVE PROTEOMICS. Shock 2024; 62:738-745. [PMID: 38888471 DOI: 10.1097/shk.0000000000002408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
ABSTRACT Objective: To uncover critical active proteins influencing sepsis outcomes through multiomics analysis. Methods: This study collected peripheral blood from sepsis patients (NS = 26, SV = 27) and controls (Con = 16). Cellular heterogeneity was assessed using scRNA-seq. Cellular populations were identified through clustering and annotation. Gene set variation analysis was employed to detect pathway alterations in sepsis, while the Viper algorithm estimated protein activity at the single-cell level. Signaling networks were investigated via cell-cell communication analysis. Differentially expressed proteins were identified by DIA proteomics and confirmed through integrated analysis. Prognostic value was evaluated via meta and survival analyses. Results: scRNA-seq of 22,673 features within 34,228 cells identified five cellular clusters and 253 active proteins via Viper, validated by DIA (FC > 2, P < 0.05). Four proteins (SPI1, MEF2A, CBX3, UBTF) with prognostic significance were discovered and mapped onto the cellular landscape. Gene set variation analysis enrichment analysis revealed that the NS group exhibited significant alterations in pathways related to cellular apoptosis and inflammatory responses, while the SV group displayed increased activity in DNA repair and cellular survival pathways. Conclusion: The study's findings advance the understanding of sepsis pathophysiology by linking differentially active proteins to patient prognosis, paving the way for targeted therapeutic strategies.
Collapse
Affiliation(s)
- Hui Liu
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Wei Xiong
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | | | - Yingchun Hu
- Department of Emergency Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
6
|
Yang S, Zhao M, Feng Y, Zhang X, Li Q, Jiang W, Wang D. Exploring the molecular mechanism of Toddalia asiatica (L.) lam on the treatment of thrombosis based on zebrafish models, network pharmacology and experimental verification. Fitoterapia 2024; 179:106224. [PMID: 39321855 DOI: 10.1016/j.fitote.2024.106224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 09/11/2024] [Accepted: 09/21/2024] [Indexed: 09/27/2024]
Abstract
Toddalia asiatica (L.) Lam. (TA) is a traditional folk medicine of ethnic minorities in the southwest of China. It is widely used in the treatment of dispersing blood stasis and activating blood. However, the effective substance and pharmacological mechanism have not been fully elucidated. The zebrafish larvae were treated with Phenylhydrazine (PHZ) to establish a thrombus model, and the staining intensity of zebrafish red blood cells was analyzed. The antithrombotic activity of TA was verified for the first time, and it was found that the inhibition rate of TA on thrombosis was up to 60.85 %. The chemical ingredients of TA were collected by combining UPLC-HRMS analysis and the literature research. Network pharmacology revealed that six key targets were obtained, which including TNF, AKT1, EGFR, PTGS2, PPARG, and IFNG. It showed that the PI3K-Akt pathway was a core signaling pathway. Coagulation factor III(TF), playing an important role in the process of hemostasis and thrombosis, which ranks high in the PPI network. Moreover, the results of molecular docking showed that the active components had a strong binding force with TF, which indicated that TF might be the key target of TA in treating thrombosis. In vitro experiments showed that TA could inhibit TNF-α-induced high expression of TF in EA.hy926 cells. In addition, TA could inhibit TNF-α-activated expression of Akt, IκBα and P65 protein phosphorylation in PI3K-Akt pathway. The results showed that TA had antithrombotic activity and exerted an antithrombotic effect by inhibiting the expression of TF through the PI3K-Akt-NF-κB signaling pathway.
Collapse
Affiliation(s)
- Songqin Yang
- School of Pharmaceutical Sciences, Guizhou University, Guizhou, Guiyang 550025, PR China
| | - Mao Zhao
- School of Pharmaceutical Sciences, Guizhou University, Guizhou, Guiyang 550025, PR China
| | - Yuhan Feng
- School of Pharmaceutical Sciences, Guizhou University, Guizhou, Guiyang 550025, PR China
| | - Xia Zhang
- School of Pharmaceutical Sciences, Guizhou University, Guizhou, Guiyang 550025, PR China
| | - Qiuhong Li
- School of Pharmaceutical Sciences, Guizhou University, Guizhou, Guiyang 550025, PR China
| | - Wenwen Jiang
- School of Pharmaceutical Sciences, Guizhou University, Guizhou, Guiyang 550025, PR China..
| | - Daoping Wang
- Key Laboratory of Natural Products Chemistry of Guizhou Academy of Sciences, Guiyang 550014, China..
| |
Collapse
|
7
|
Protopapas AA, Takardaki A, Protopapa N, Papagiouvanni I, Protopapas AN, Skoura L, Savopoulos C, Goulis I. Microvesicle Tissue Factor Procoagulant Activity Is Elevated and Correlated With Disease Severity in Patients With Cirrhosis. Liver Int 2024. [PMID: 39601330 DOI: 10.1111/liv.16192] [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: 09/02/2024] [Revised: 11/12/2024] [Accepted: 11/17/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND AND AIMS Tissue factor-expressing microvesicles (MV-TF) have been found to correlate with thrombotic complications in various diseases. Simultaneously, there is expanding research regarding the effect of the coagulation cascade on liver fibrosis progression. The aim of our manuscript was to evaluate MV-TF activity in patients with cirrhosis and its correlation with disease severity. METHODS We prospectively enrolled 82 patients [11 with cirrhosis and hepatocellular cancer (Group 1), 50 with cirrhosis (Group 2) and 21 controls (Group 3)]. Extensive workup for disease staging and exclusion criteria was undertaken. Exclusion criteria included thrombophilia, history of thrombosis, recent hospitalisation, ongoing infection, alcohol dependence, cancer, haematological diseases and use of anticoagulant, antiplatelet or contraceptive drugs. Plasma tissue factor antigen concentration and MV-TF activity were assessed. RESULTS MV-TF showed median values of 4.03 [1.57], 3.17 [1.59] and 2.26 [1.23] pg/mL in Groups 1, 2 and 3, respectively. There was a statistically significant difference between Groups 1 and 3 (p < 0.001) and Groups 2 and 3 (p = 0.003), while Group 1 had higher values than Group 2 without statistical significance (p = 0.088). In Group 2, the patients' Child-Pugh (CP) stage was A in 56%, B in 26% and C in 18% of cases. MV-TF activity significantly correlated with decompensated cirrhosis (p = 0.005) and higher CP stage (p = 0.011). Finally, MV-TF activity significantly correlated with 12-month mortality (p = 0.021). CONCLUSIONS MV-TF activity is elevated in patients with cirrhosis, showing a significant correlation with disease severity. MV-TF may play a role in the procoagulant imbalance of liver cirrhosis and their contribution in disease progression should be studied further.
Collapse
Affiliation(s)
- Adonis A Protopapas
- First Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
- Fourth Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Anna Takardaki
- Department of Microbiology, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Nefeli Protopapa
- First Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Ioanna Papagiouvanni
- Fourth Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| | - Andreas N Protopapas
- First Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Lemonia Skoura
- Department of Microbiology, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Christos Savopoulos
- First Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, AHEPA University Hospital, Thessaloniki, Greece
| | - Ioannis Goulis
- Fourth Department of Internal Medicine, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece
| |
Collapse
|
8
|
Wang C, Zhao X, Wang K, Liang H, Chen S, Liu Y, Yao H, Jiang J. Prospective Application of Mesenchymal Stem Cell-Derived Exosomes in the Treatment of Disseminated Intravascular Coagulation. Int J Nanomedicine 2024; 19:11957-11971. [PMID: 39569063 PMCID: PMC11577934 DOI: 10.2147/ijn.s467158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
Disseminated intravascular coagulation (DIC) is an acquired disorder characterized by systemic activation of blood coagulation, which can arise from various causes. Owing to its abrupt onset, rapid progression, and high mortality rate, DIC presents a major clinical challenge. Anticoagulant drugs, such as heparin or low-molecular-weight heparin, are the current gold standard of treatment; however, these interventions pose considerable bleeding risks. Thus, safer and more effective therapeutic strategies are urgently required. Owing to their strong anti-inflammatory and tissue repair capabilities, mesenchymal stem cell-derived exosomes (MSC-Exos) have gained considerable attention as novel therapeutic options for numerous disorders, including DIC. Their stability in diverse pathological states highlights their potential as promising candidates for DIC therapy. This review presents the latest insights on the pathogenesis of DIC and anti-inflammatory and anticoagulant properties of MSC-Exos. We aimed to elucidate the potential mechanisms by which MSC-Exos influence DIC pathogenesis. We speculate that MSC-Exos offer a multifaceted approach to DIC treatment by attenuating neutrophil extracellular trap formation, modulating M1/M2 macrophage polarization, altering Nrf2/NF-κB signalling pathway to downregulate pro-inflammatory factors, and correcting imbalances in the coagulation-fibrinolysis system through anticoagulant routes. This suggests that MSC-Exos are a potential paradigm in DIC therapy, offering novel targets and treatment modalities for DIC management.
Collapse
Affiliation(s)
- Chengran Wang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Xiaoqing Zhao
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Keyan Wang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Huixin Liang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Shuhan Chen
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Yajie Liu
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Hua Yao
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| | - Jinlan Jiang
- Department of Scientific Research Center, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province, People’s Republic of China
| |
Collapse
|
9
|
Zhang Z, Yang M, Zhou T, Chen Y, Zhou X, Long K. Emerging trends and hotspots in intestinal microbiota research in sepsis: bibliometric analysis. Front Med (Lausanne) 2024; 11:1510463. [PMID: 39606629 PMCID: PMC11598531 DOI: 10.3389/fmed.2024.1510463] [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: 10/13/2024] [Accepted: 11/01/2024] [Indexed: 11/29/2024] Open
Abstract
Background The association between the gut microbiota and sepsis has garnered attention in the field of intestinal research in sepsis. This study utilizes bibliometric methods to visualize and analyze the literature on gut microbiota research in sepsis from 2011 to 2024, providing a scientific foundation for research directions and key issues in this domain. Methods Original articles and reviews of gut microbiota research in sepsis, which published in English between 2011 and 2024, were obtained from the Web of Science Core Collection on June 21, 2024. Python, VOSviewer, and CiteSpace software were used for the visual analysis of the retrieved data. Results A total of 1,031 articles were analyzed, originating from 72 countries or regions, 1,614 research institutions, and 6,541 authors. The articles were published in 434 different journals, covering 89 different research fields. The number of publications and citations in this research area showed a significant growth trend from 2011 to 2024, with China, the United States, and the United Kingdom being the main research forces. Asada Leelahavanichkul from Thailand was identified as the most prolific author, making him the most authoritative expert in this field. "Nutrients" had the highest number of publications, while "Frontiers in Cellular and Infection Microbiology," "Frontiers in Immunology" and "the International Journal of Molecular Sciences" have shown increasing attention to this field in the past 2 years. Author keywords appearing more than 100 times included "gut microbiota (GM)," "sepsis" and "microbiota." Finally, this study identified "lipopolysaccharides (LPS)," "short-chain fatty acids (SCFAs)," "probiotics," "fecal microbiota transplantation (FMT)" and "gut-liver axis" as the research hotspots and potential frontier directions in this field. Conclusion This bibliometric study summarizes current important perspectives and offers comprehensive guidance between sepsis and intestinal microbiota, which may help researchers choose the most appropriate research directions.
Collapse
Affiliation(s)
- Zhengyi Zhang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Meijie Yang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tong Zhou
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yingjie Chen
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiujuan Zhou
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Kunlan Long
- Department of Critical Care Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
10
|
Sharma D, Thomas S, Moody TB, Taylor M, Ituarte B, Georgeson CJ, Barrett CD, Wei EX. Laboratory and clinical haemostatic aberrations in primary dermatologic disease: A review. Thromb J 2024; 22:101. [PMID: 39533305 PMCID: PMC11558853 DOI: 10.1186/s12959-024-00665-w] [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: 07/09/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Inflammatory dermatologic diseases have long been viewed as a "skin limited" disease process. Current literature on inflammatory dermatologic diseases investigates their relationship and influence on thromboembolic states and thromboembolic complications and the understanding of their pathophysiology and molecular mechanisms.Studies specifically discuss known inflammatory skin diseases including alopecia areata, vitiligo, psoriasis, hidradenitis suppurativa, atopic dermatitis, chronic spontaneous urticaria, and autoimmune bullous diseases, and their effects on systemic inflammation, associated cardiovascular comorbidities, and thromboembolic or hypercoagulable states. The limited current literature shows potential for links between inflammatory skin diseases and hypercoagulable states. Biomarkers such as F1 + 2, D-dimer, eosinophilic cationic protein, and PAI-1 are currently being studied to outline the mechanisms connecting inflammatory skin disease to the coagulation system. Further study and larger amounts of data are needed to draw definitive conclusions, especially when interpreting biomarkers alone such as PAI-1.The mechanisms, rates of systemic inflammation, and clinical outcomes of traditionally "skin limited" inflammatory diseases remain chronically understudied in dermatology. Many organ systems have well established connections between inflammatory disease and hypercoagulable states, but there are significant gaps in the literature regarding skin diseases. There is a significant need for comprehensive investigation of molecular mechanisms behind inflammatory dermatologic disease and hypercoagulability, how hypercoagulability effects clinical outcomes, and proper intervention to optimize patient outcomes.
Collapse
Affiliation(s)
- Divya Sharma
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| | - Sierra Thomas
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Trace B Moody
- Division of Acute Care Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mitchell Taylor
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- Creighton University School of Medicine, Omaha, NE, USA
| | - Bianca Ituarte
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
- University of Missouri-Kansas City School of Medicine, Kansas City, MO, USA
| | - Corey J Georgeson
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| | - Christopher D Barrett
- Division of Acute Care Surgery, Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Erin X Wei
- Department of Dermatology, University of Nebraska Medical Center, 985645, Omaha, NE, 68198-5645, USA
| |
Collapse
|
11
|
Gastoł J, Paszek E, Bryk-Wiązania A, Matejko B, Undas A. Good metabolic control is associated with decreased circulating factor VIIa- antithrombin complexes in type 2 diabetes: a cross-sectional study. Cardiovasc Diabetol 2024; 23:398. [PMID: 39501309 PMCID: PMC11536800 DOI: 10.1186/s12933-024-02480-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/21/2024] [Indexed: 11/09/2024] Open
Abstract
BACKGROUND Diabetes is associated with a prothrombotic state that contributes to cardiovascular (CV) events in type 2 diabetes (T2DM). Activated factor VII (FVIIa)- antithrombin (AT) complexes are indicative of tissue factor (TF) exposure and have been associated with thromboembolic risk in coronary artery disease. To our knowledge there have been no reports on FVIIa-AT complexes in T2DM, therefore we assessed factors that determine FVIIa-AT complexes in this disease and the impact of higher complexes on a prothrombotic state. METHODS In 108 T2DM patients (mean age 63.8 years, 52.8% men, median HbA1c of 6.9 [interquartile range 6.1-8.2] %) and 83 age- and sex-matched non-diabetic subjects, we measured FVIIa-AT complexes. Metabolic control of T2DM involved fasting glucose, glycated hemoglobin (HbA1c), albumin/creatinine ratio (ACR), and lipid levels. To characterize a prothrombotic state, we determined thrombin generation parameters, fibrinolysis markers, and plasma fibrin clot properties. RESULTS FVII-AT complexes in T2DM patients were similar to controls (73.6 [59.4-91.7] vs. 79.6 [59.2-97.1]pM, respectively, p = 0.30). The T2DM patients with FVIIa-AT in the top vs. the bottom quartile had a larger prevalence of active smoking and insulin use, along with higher fasting glucose (+ 36.4%), HbA1c (+ 27.4%), ACR (+ 72.8%), total cholesterol (+ 34.5%), and LDL-cholesterol (+ 80%). FVIIa-AT complexes showed no associations with in vitro thrombin generation potential, plasma fibrin clot properties, or fibrinolysis variables. On multivariable analysis HbA1c, ACR, and total cholesterol remained independently associated with FVIIa-AT complexes in T2DM. CONCLUSIONS This is the first study to show that in T2DM higher FVIIa-AT complexes are associated with markers of dyslipidemia and glycemia control, indicating that TF-induced coagulation activation could be suppressed by achieving treatment targets.
Collapse
Affiliation(s)
- Joanna Gastoł
- Metabolic Diseases and Diabetology Clinical Department, University Hospital, Kraków, Poland
- Department of Metabolic Diseases, Jagiellonian University Medical College, Kraków, Poland
| | - Elżbieta Paszek
- Clinical Department of Interventional Cardiology, John Paul II Hospital, Kraków, Poland
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St, 31-202, Kraków, Poland
| | - Agata Bryk-Wiązania
- Department of Endocrinology, Jagiellonian University Medical College, Kraków, Poland
- Department of Endocrinology, Oncological Endocrinology and Nuclear Medicine, University Hospital, Kraków, Poland
| | - Bartłomiej Matejko
- Metabolic Diseases and Diabetology Clinical Department, University Hospital, Kraków, Poland
- Department of Metabolic Diseases, Jagiellonian University Medical College, Kraków, Poland
| | - Anetta Undas
- Department of Thromboembolic Disorders, Institute of Cardiology, Jagiellonian University Medical College, 80 Pradnicka St, 31-202, Kraków, Poland.
- Cracow Center for Medical Research and Technology, John Paul II Hospital, Kraków, Poland.
| |
Collapse
|
12
|
Wang X, Liu C, Liu C, Shi Z, Liu X, Huang F. A chitosan macroporous hydrogel integrating enrichment, adsorption and delivery of blood clotting components for rapid hemostasis. Int J Biol Macromol 2024; 281:136482. [PMID: 39406331 DOI: 10.1016/j.ijbiomac.2024.136482] [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: 05/11/2024] [Revised: 09/29/2024] [Accepted: 10/08/2024] [Indexed: 10/18/2024]
Abstract
Traditional hemostatic hydrogels face considerable limitations in achieving rapid control of severe bleedings, a crucial factor in reducing casualties in both military and civilian settings. This study presents a chitosan-based hemostatic hydrogel with interconnected secondary macropores designed to enhance interactions with blood clotting components by reducing diffusion resistance and increasing contact area. The macropores were created using a straightforward process involving NaOH-mediated SiO2 template dissolution and NH3 generation. The resulting macroporous structure increased the hydrogel's overall porosity without compromising its viscoelasticity. Functional studies demonstrated that the macroporous hydrogel effectively concentrated and adsorbed blood clotting components, while also facilitating the delivery of artificially embedded clotting factor to further expedite clot formation. These combined actions resulted in improve hemostatic efficacy, reducing whole blood clotting time by over 94 % in vitro. Furthermore, in vivo studies using rat tail amputation and liver injury models showed a reduction in blood loss by over 65 % and a decrease in bleeding time by over 70 %. Additionally, the porous chitosan hydrogel exhibited minimal biotoxicity and promoted biodegradability in vivo. In conclusion, this work introduces a macroporous chitosan-based hemostatic hydrogel with great potential for rapid hemorrhage control.
Collapse
Affiliation(s)
- Xiaoqiang Wang
- State Key Laboratory of Heavy Oil Processing & College of Chemistry and Chemical Engineering, China University of Petroleum (East China), 66 West Changjiang Road, Qingdao, Shandong 266580, China.
| | - Chang Liu
- State Key Laboratory of Heavy Oil Processing & College of Chemistry and Chemical Engineering, China University of Petroleum (East China), 66 West Changjiang Road, Qingdao, Shandong 266580, China
| | - Chengkun Liu
- State Key Laboratory of Heavy Oil Processing & College of Chemistry and Chemical Engineering, China University of Petroleum (East China), 66 West Changjiang Road, Qingdao, Shandong 266580, China
| | - Zhuang Shi
- State Key Laboratory of Heavy Oil Processing & College of Chemistry and Chemical Engineering, China University of Petroleum (East China), 66 West Changjiang Road, Qingdao, Shandong 266580, China
| | - Xiaodan Liu
- State Key Laboratory of Heavy Oil Processing & College of Chemistry and Chemical Engineering, China University of Petroleum (East China), 66 West Changjiang Road, Qingdao, Shandong 266580, China
| | - Fang Huang
- State Key Laboratory of Heavy Oil Processing & College of Chemistry and Chemical Engineering, China University of Petroleum (East China), 66 West Changjiang Road, Qingdao, Shandong 266580, China.
| |
Collapse
|
13
|
Wu X, Zhao X, Li F, Wang Y, Ou Y, Zhang H, Li X, Wu X, Wang L, Li M, Zhang Y, Liu J, Xing M, Liu H, Tan Y, Wang Y, Xie Y, Zhang H, Luo Y, Li H, Wang J, Sun L, Li Y, Zhang H. MLKL-mediated endothelial necroptosis drives vascular damage and mortality in systemic inflammatory response syndrome. Cell Mol Immunol 2024; 21:1309-1321. [PMID: 39349742 PMCID: PMC11527879 DOI: 10.1038/s41423-024-01217-y] [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: 02/20/2024] [Accepted: 09/11/2024] [Indexed: 11/02/2024] Open
Abstract
The hypersecretion of cytokines triggers life-threatening systemic inflammatory response syndrome (SIRS), leading to multiple organ dysfunction syndrome (MODS) and mortality. Although both coagulopathy and necroptosis have been identified as important factors in the pathogenesis of SIRS, the specific cell types that undergo necroptosis and the interrelationships between coagulopathy and necroptosis remain unclear. In this study, we utilized visualization analysis via intravital microscopy to demonstrate that both anticoagulant heparin and nonanticoagulant heparin (NAH) pretreatment protect mice against TNF-α-induced mortality in SIRS. Moreover, the deletion of Mlkl or Ripk3 resulted in decreased coagulation and reduced mortality in TNF-α-induced SIRS. These findings suggest that necroptosis plays a key role upstream of coagulation in SIRS-related mortality. Furthermore, using a genetic lineage tracing mouse model (Tie2-Cre;Rosa26-tdT), we tracked endothelial cells (ECs) and verified that EC necroptosis is responsible for the vascular damage observed in TNF-α-treated mice. Importantly, Mlkl deletion in vascular ECs in mice had a similar protective effect against lethal SIRS by blocking EC necroptosis to protect the integrity of the endothelium. Collectively, our findings demonstrated that RIPK3-MLKL-dependent necroptosis disrupted vascular integrity, resulting in coagulopathy and multiorgan failure, eventually leading to mortality in SIRS patients. These results highlight the importance of targeting vascular EC necroptosis for the development of effective treatments for SIRS patients.
Collapse
Affiliation(s)
- Xiaoxia Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xiaoming Zhao
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Fang Li
- Department of Anesthesiology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yang Wang
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yangjing Ou
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Haiwei Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xiaoming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Xuanhui Wu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Lingxia Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Ming Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yue Zhang
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jianling Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Mingyan Xing
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Han Liu
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yongchang Tan
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Yangyang Wang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yangyang Xie
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Hanwen Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Yan Luo
- Department of Anesthesiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Hong Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Jing Wang
- Department of Microbiology and Immunology, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Liming Sun
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, PR China
| | - Yu Li
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China
| | - Haibing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, PR China.
| |
Collapse
|
14
|
Tang Y, Jiao Y, An X, Tu Q, Jiang Q. Neutrophil extracellular traps and cardiovascular disease: Associations and potential therapeutic approaches. Biomed Pharmacother 2024; 180:117476. [PMID: 39357329 DOI: 10.1016/j.biopha.2024.117476] [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: 07/14/2024] [Revised: 09/20/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Cardiovascular disease (CVD) is a significant global health concern, ranking among the top five causes of disability-adjusted life-years (DALY) in 190 countries and territories. Neutrophils, key players in the innate immune system, combat infections by releasing neutrophil extracellular traps (NETs) composed of DNA, histones, elastase, myeloperoxidase, and antimicrobial peptides. This paper explores the relationship between NETs and cardiovascular diseases, focusing on conditions such as heart failure, pulmonary hypertension, atrial fibrillation, and ischemia-reperfusion injury. Particularly, it delves into the impact of NETs on atrial fibrillation and pulmonary hypertension, as well as the role of myeloperoxidase (MPO) and neutrophil elastase (NE) in these diseases. Furthermore, the potential of targeting NETs for the treatment of cardiovascular diseases is discussed.
Collapse
Affiliation(s)
- Yiyue Tang
- Department of Cardiovascular Medicine, The First People's Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China; Zunyi Medical University, Zunyi, Guizhou, China
| | - Yang Jiao
- Department of Cardiovascular Medicine, The First People's Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China
| | - Xiaohua An
- Scientific Research Center, The First People's Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China; Zunyi Medical University, Zunyi, Guizhou, China
| | - Qingxian Tu
- Department of Cardiovascular Medicine, The First People's Hospital of Zunyi(The Third Affiliated Hospital of Zunyi Medical University), Zunyi, Guizhou, China.
| | - Qianfeng Jiang
- GuizhouAerospaceHospital,(Affiliated AerospaceHospital of Zunyi Medical University), Zunyi, Guizhou, China.
| |
Collapse
|
15
|
Zhao G, Zhao Y, Liang W, Lu H, Liu H, Deng Y, Zhu T, Guo Y, Chang L, Garcia-Barrio MT, Chen YE, Zhang J. Endothelial KLF11 is a novel protector against diabetic atherosclerosis. Cardiovasc Diabetol 2024; 23:381. [PMID: 39462409 PMCID: PMC11514907 DOI: 10.1186/s12933-024-02473-y] [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: 06/04/2024] [Accepted: 10/14/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Atherosclerotic cardiovascular diseases remain the leading cause of mortality in diabetic patients, with endothelial cell (EC) dysfunction serving as the initiating step of atherosclerosis, which is exacerbated in diabetes. Krüppel-like factor 11 (KLF11), known for its missense mutations leading to the development of diabetes in humans, has also been identified as a novel protector of vascular homeostasis. However, its role in diabetic atherosclerosis remains unexplored. METHODS Diabetic atherosclerosis was induced in both EC-specific KLF11 transgenic and knockout mice in the Ldlr-/- background by feeding a diabetogenic diet with cholesterol (DDC). Single-cell RNA sequencing (scRNA-seq) was utilized to profile EC dysfunction in diabetic atherosclerosis. Additionally, gain- and loss-of-function experiments were conducted to investigate the role of KLF11 in hyperglycemia-induced endothelial cell dysfunction. RESULTS We found that endothelial KLF11 deficiency significantly accelerates atherogenesis under diabetic conditions, whereas KLF11 overexpression remarkably inhibits it. scRNA-seq profiling demonstrates that loss of KLF11 increases endothelial-to-mesenchymal transition (EndMT) during atherogenesis under diabetic conditions. Utilizing gain- and loss-of-function approaches, our in vitro study reveals that KLF11 significantly inhibits EC inflammatory activation and TXNIP-induced EC oxidative stress, as well as Notch1/Snail-mediated EndMT under high glucose exposure. CONCLUSION Our study demonstrates that endothelial KLF11 is an endogenous protective factor against diabetic atherosclerosis. These findings indicate that manipulating KLF11 could be a promising approach for developing novel therapies for diabetes-related cardiovascular complications.
Collapse
Affiliation(s)
- Guizhen Zhao
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, 77204, USA
| | - Yang Zhao
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Wenying Liang
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Haocheng Lu
- School of Medicine, Southern University of Science and Technology, Shenzhen, 518055, People's Republic of China
| | - Hongyu Liu
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Yongjie Deng
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Tianqing Zhu
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Yanhong Guo
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Lin Chang
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Minerva T Garcia-Barrio
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA
| | - Y Eugene Chen
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| | - Jifeng Zhang
- Frankel Cardiovascular Center, Department of Internal Medicine, University of Michigan Medical Center, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
16
|
Bi J, Wang Y, Wang K, Sun Y, Ye F, Wang X, Pan J. FGF1 attenuates sepsis-induced coagulation dysfunction and hepatic injury via IL6/STAT3 pathway inhibition. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167281. [PMID: 38870868 DOI: 10.1016/j.bbadis.2024.167281] [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: 03/10/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND & AIMS Sepsis, a globally prevalent and highly lethal condition, remains a critical medical challenge. This investigation aims to assess the relevance of FGF1 as a potential therapeutic target for sepsis. METHODS Sepsis was induced in C57BL/6 mice through LPS administration to establish an in vivo animal model. Various in vitro assays were conducted using human umbilical vein endothelial cells to elucidate the role of FGF1 in the disruption of the coagulation system and liver injury associated with sepsis, as well as to explore its underlying molecular mechanisms. RESULTS In in vivo experiments, FGF1 ameliorated coagulation system disruption in septic mice by reducing the levels of pro-inflammatory and coagulation-related factors in the bloodstream. FGF1 also enhanced liver function in septic mice, mitigating liver inflammation and cell apoptosis, fostering liver vascular regeneration, increasing liver blood perfusion, and improving mouse survival. In vitro experiments demonstrated that FGF1 could inhibit LPS-induced inflammatory responses and apoptosis in endothelial cells, fortify endothelial cell barrier function, decrease endothelial cell permeability, promote endothelial cell proliferation, and restore endothelial cell tube-forming ability. Both in vivo and in vitro experiments substantiated that FGF1 improved sepsis by inhibiting the IL-6/STAT3 signaling pathway. CONCLUSION In summary, our study indicates that FGF1 mitigates excessive inflammatory responses in sepsis by suppressing the IL-6/STAT3 signaling pathway, thereby improving systemic blood circulation and ameliorating liver damage in septic organisms. Consequently, this research identifies FGF1 as a potential clinical target for the treatment of human sepsis.
Collapse
Affiliation(s)
- Jianing Bi
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Critical Care Medicine, Wenzhou, China; Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, China; School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China.
| | - Yanjing Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Kaicheng Wang
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Critical Care Medicine, Wenzhou, China; Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, China
| | - Yuanyuan Sun
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Critical Care Medicine, Wenzhou, China; Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, China
| | - Fanrong Ye
- Departments of Nuclear Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaojie Wang
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China.
| | - Jingye Pan
- Department of Intensive Care Unit, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China; Zhejiang Key Laboratory of Critical Care Medicine, Wenzhou, China; Wenzhou Key Laboratory of Critical Care and Artificial Intelligence, Wenzhou, China.
| |
Collapse
|
17
|
Noone D, Preston RJS, Rehill AM. The Role of Myeloid Cells in Thromboinflammatory Disease. Semin Thromb Hemost 2024; 50:998-1011. [PMID: 38547918 DOI: 10.1055/s-0044-1782660] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Inflammation contributes to the development of thrombosis, but the mechanistic basis for this association remains poorly understood. Innate immune responses and coagulation pathways are activated in parallel following infection or injury, and represent an important host defense mechanism to limit pathogen spread in the bloodstream. However, dysregulated proinflammatory activity is implicated in the progression of venous thromboembolism and arterial thrombosis. In this review, we focus on the role of myeloid cells in propagating thromboinflammation in acute inflammatory conditions, such as sepsis and coronavirus disease 2019 (COVID-19), and chronic inflammatory conditions, such as obesity, atherosclerosis, and inflammatory bowel disease. Myeloid cells are considered key drivers of thromboinflammation via upregulated tissue factor activity, formation of neutrophil extracellular traps (NETs), contact pathway activation, and aberrant coagulation factor-mediated protease-activated receptor (PAR) signaling. We discuss how strategies to target the intersection between myeloid cell-mediated inflammation and activation of blood coagulation represent an exciting new approach to combat immunothrombosis. Specifically, repurposed anti-inflammatory drugs, immunometabolic regulators, and NETosis inhibitors present opportunities that have the potential to dampen immunothrombotic activity without interfering with hemostasis. Such therapies could have far-reaching benefits for patient care across many thromboinflammatory conditions.
Collapse
Affiliation(s)
- David Noone
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Roger J S Preston
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| | - Aisling M Rehill
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
- National Children's Research Centre, Our Lady's Children's Hospital Crumlin, Dublin, Ireland
| |
Collapse
|
18
|
Zhuang H, Han S, Harris NS, Reeves WH. MEK1/2- and ERK1/2-Mediated Lung Endothelial Injury and Altered Hemostasis Promote Diffuse Alveolar Hemorrhage in Murine Lupus. Arthritis Rheumatol 2024; 76:1538-1551. [PMID: 38923837 PMCID: PMC11421958 DOI: 10.1002/art.42936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/25/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVE About 3% of patients with lupus develop severe diffuse alveolar hemorrhage (DAH) with pulmonary vasculitis. C57BL/6 (B6) mice with pristane-induced lupus also develop DAH, but BALB/c mice are resistant. DAH is independent of Toll-like receptor signaling and other inflammatory pathways. This study examined the role of the MEK1/2 pathway (MEK1/2-ERK1/2, JNK, p38). METHODS B6 and BALB/c mice were treated with pristane with or without inhibitors of MEK1/2 (trametinib/GSK1120212 [GSK]), ERK1/2 (SCH772984 [SCH]), JNK, or p38. Effects on lung hemorrhage and hemostasis were determined. RESULTS GSK and SCH abolished DAH, whereas JNK and p38 inhibitors were ineffective. Apoptotic cells were present in lung samples from pristane-treated mice but not in mice receiving pristane and GSK, and endothelial dysfunction was normalized. Expression of the ERK1/2-regulated transcription factor early growth response 1 increased in pristane-treated B6, but not BALB/c, mice and was normalized by GSK. Pristane also increased expression of the anticoagulant genes Tfpi and Thbd in B6 mice. The ratio of Tfpi to tissue factor (F3) to Tfpi increased in B6 (but not BALB/c) mice and was normalized by GSK. Circulating thrombomodulin protein levels increased in B6 mice and returned to normal after GSK treatment. Consistent with augmented endothelial anticoagulant activity, pristane treatment increased tail bleeding in B6 mice. CONCLUSION Pristane treatment promotes lung endothelial injury and DAH in B6 mice by activating the MEK1/2-ERK1/2 pathway and impairing hemostasis. The hereditary factors determining susceptibility to lung injury and bleeding in pristane-induced lupus are relevant to the pathophysiology of life-threatening DAH in systemic lupus erythematosus and may help to optimize therapy.
Collapse
Affiliation(s)
- Haoyang Zhuang
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida College of Medicine, Gainesville, FL 32610
| | - Shuhong Han
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida College of Medicine, Gainesville, FL 32610
| | - Neil S. Harris
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610
| | - Westley H. Reeves
- Division of Rheumatology, Allergy, & Clinical Immunology, University of Florida College of Medicine, Gainesville, FL 32610
- Department of Pathology, Immunology, and Laboratory Medicine, University of Florida College of Medicine, Gainesville, FL 32610
| |
Collapse
|
19
|
Bonifay A, Mackman N, Hisada Y, Sachetto ATA, Hau C, Gray E, Hogwood J, Aharon A, Badimon L, Barile L, Baudar J, Beckmann L, Benedikter B, Bolis S, Bouriche T, Brambilla M, Burrello J, Camera M, Campello E, Ettelaie C, Faille D, Featherby S, Franco C, Guldenpfennig M, Hansen JB, Judicone C, Kim Y, Kristensen SR, Laakmann K, Langer F, Latysheva N, Lucien F, de Menezes EM, Mullier F, Norris P, Nybo J, Orbe J, Osterud B, Paramo JA, Radu CM, Roncal C, Samadi N, Snir O, Suades R, Wahlund C, Chareyre C, Abdili E, Martinod K, Thaler J, Dignat-George F, Nieuwland R, Lacroix R. Comparison of assays measuring extracellular vesicle tissue factor in plasma samples: communication from the ISTH SSC Subcommittee on Vascular Biology. J Thromb Haemost 2024; 22:2910-2921. [PMID: 38925490 DOI: 10.1016/j.jtha.2024.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 04/23/2024] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
BACKGROUND Scientific and clinical interest in extracellular vesicles (EVs) is growing. EVs that expose tissue factor (TF) bind factor VII/VIIa and can trigger coagulation. Highly procoagulant TF-exposing EVs are detectable in the circulation in various diseases, such as sepsis, COVID-19, or cancer. Many in-house and commercially available assays have been developed to measure EV-TF activity and antigen, but only a few studies have compared some of these assays. OBJECTIVES The International Society on Thrombosis and Haemostasis Scientific and Standardization Committee Subcommittee on Vascular Biology initiated a multicenter study to compare the sensitivity, specificity, and reproducibility of these assays. METHODS Platelet-depleted plasma samples were prepared from blood of healthy donors. The plasma samples were spiked either with EVs from human milk or EVs from TF-positive and TF-negative cell lines. Plasma was also prepared from whole human blood with or without lipopolysaccharide stimulation. Twenty-one laboratories measured EV-TF activity and antigen in the prepared samples using their own assays representing 18 functional and 9 antigenic assays. RESULTS There was a large variability in the absolute values for the different EV-TF activity and antigen assays. Activity assays had higher specificity and sensitivity compared with antigen assays. In addition, there was a large intra-assay and interassay variability. Functional assays that used a blocking anti-TF antibody or immunocapture were the most specific and sensitive. Activity assays that used immunocapture had a lower coefficient of variation compared with assays that isolated EVs by high-speed centrifugation. CONCLUSION Based on this multicenter study, we recommend measuring EV-TF using a functional assay in the presence of an anti-TF antibody.
Collapse
Affiliation(s)
- Amandine Bonifay
- Aix-Marseille University, C2VN, INSERM 1263, INRAE1260, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Yohei Hisada
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Ana Teresa Azevedo Sachetto
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Chi Hau
- Laboratory of Experimental Clinical Chemistry, and Amsterdam Vesicle Center, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Elaine Gray
- National Institute for Biological Standards and Control, Potter's Bar, Hertfordshire, United Kingdom
| | - John Hogwood
- National Institute for Biological Standards and Control, Potter's Bar, Hertfordshire, United Kingdom
| | - Anat Aharon
- Hematology Research Laboratory, Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel; The Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lina Badimon
- Cardiovascular ICCC Program, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Lucio Barile
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland; Faculty of Biomedical Sciences, Università della Svizzera Italiana, Lugano, Switzerland
| | - Justine Baudar
- Université Catholique de Louvain, CHU UCL NAMUR, Namur Thrombosis and Hemostasis Center (NTHC), Yvoir, Belgium
| | - Lennart Beckmann
- Department of Hematology and Oncology, University Cancer Center Hamburg (UCCH), University Medical Center Eppendorf, Hamburg, Germany
| | - Birke Benedikter
- Institute for Lung Research, Universities of Giessen and Marburg Lung Centre, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany; University Eye Clinic Maastricht, MHeNs School for Mental Health and Neuroscience, Maastricht University Medical Center + (MUMC+), Maastricht, the Netherlands
| | - Sara Bolis
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Tarik Bouriche
- Research and Technology Department, BioCytex, Marseille, France
| | | | - Jacopo Burrello
- Cardiovascular Theranostics, Istituto Cardiocentro Ticino, Laboratories for Translational Research, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Marina Camera
- Centro Cardiologico Monzino IRCCS, Milan, Italy; Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| | - Elena Campello
- Department of Medicine, University of Padova, Padua, Italy
| | - Camille Ettelaie
- Biomedical Science, University of Hull/HYMS, Cottingham Road, Hull, United Kingdom
| | - Dorothée Faille
- Université Paris Cité and Université Sorbonne Paris Nord, INSERM, LVTS, Paris, France; Laboratoire d'Hématologie, AP-HP, Hôpital Bichat, Paris, France
| | - Sophie Featherby
- Biomedical Science, University of Hull/HYMS, Cottingham Road, Hull, United Kingdom
| | - Corentin Franco
- Research and Technology Department, BioCytex, Marseille, France
| | - Maite Guldenpfennig
- Université Catholique de Louvain, CHU UCL NAMUR, Namur Thrombosis and Hemostasis Center (NTHC), Yvoir, Belgium
| | - John-Bjarne Hansen
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway; Thrombosis Research Center (TREC), Division of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | | | - Yohan Kim
- epartment of Urology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Soren Risom Kristensen
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - Katrin Laakmann
- Institute for Lung Research, Universities of Giessen and Marburg Lung Centre, Philipps-University Marburg, German Center for Lung Research (DZL), Marburg, Germany
| | - Florian Langer
- Department of Hematology and Oncology, University Cancer Center Hamburg (UCCH), University Medical Center Eppendorf, Hamburg, Germany
| | - Nadezhda Latysheva
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Fabrice Lucien
- epartment of Urology, Department of Immunology, Mayo Clinic, Rochester, Minnesota, USA
| | - Erika Marques de Menezes
- Vitalant Research Institute, San Francisco, California, USA; Department of Laboratory Medicine, University of California, San Francisco, California, USA
| | - François Mullier
- Université Catholique de Louvain, CHU UCL NAMUR, Namur Thrombosis and Hemostasis Center (NTHC), Yvoir, Belgium
| | - Philip Norris
- Vitalant Research Institute, San Francisco, California, USA; Department of Laboratory Medicine, University of California, San Francisco, California, USA; Department of Medicine, UCSF, San Francisco, California, USA
| | - Jette Nybo
- Department of Clinical Biochemistry, Aalborg University Hospital, Aalborg, Denmark
| | - Josune Orbe
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; RICORS-Cerebrovascular Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Bjarne Osterud
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Jose A Paramo
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Claudia M Radu
- Department of Medicine, University of Padova, Padua, Italy
| | - Carmen Roncal
- Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; RICORS-Cerebrovascular Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Nazanin Samadi
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Omri Snir
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Rosa Suades
- Cardiovascular ICCC Program, Research Institute Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red Cardiovascular (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain
| | - Casper Wahlund
- Thrombosis Research Group (TREC), Institute of Clinical Medicine, UiT - The Arctic University of Norway, Tromsø, Norway
| | - Corinne Chareyre
- Aix-Marseille University, C2VN, INSERM 1263, INRAE1260, Marseille, France
| | - Evelyne Abdili
- Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| | - Kimberly Martinod
- Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, KU Leuven, Leuven, Belgium
| | - Johannes Thaler
- Clinical Division of Hematology and Hemostaseology, Department of Medicine I, Medical University of Vienna, Vienna, Austria
| | - Françoise Dignat-George
- Aix-Marseille University, C2VN, INSERM 1263, INRAE1260, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France.
| | - Rienk Nieuwland
- Laboratory of Experimental Clinical Chemistry, and Amsterdam Vesicle Center, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Romaric Lacroix
- Aix-Marseille University, C2VN, INSERM 1263, INRAE1260, Marseille, France; Department of Hematology and Vascular Biology, CHU La Conception, APHM, Marseille, France
| |
Collapse
|
20
|
Garlapati V, Luo Q, Posma J, Aluia M, Nguyen TS, Grunz K, Molitor M, Finger S, Harms G, Bopp T, Ruf W, Wenzel P. Macrophage-Expressed Coagulation Factor VII Promotes Adverse Cardiac Remodeling. Circ Res 2024; 135:841-855. [PMID: 39234697 DOI: 10.1161/circresaha.123.324114] [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: 12/06/2023] [Revised: 08/23/2024] [Accepted: 08/25/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Excess fibrotic remodeling causes cardiac dysfunction in ischemic heart disease, driven by MAP (mitogen-activated protein) kinase-dependent TGF-ß1 (transforming growth factor-ß1) activation by coagulation signaling of myeloid cells. How coagulation-inflammatory circuits can be specifically targeted to achieve beneficial macrophage reprogramming after myocardial infarction (MI) is not completely understood. METHODS Mice with permanent ligation of the left anterior descending artery were used to model nonreperfused MI and analyzed by single-cell RNA sequencing, protein expression changes, confocal microscopy, and longitudinal monitoring of recovery. We probed the role of the tissue factor (TF)-FVIIa (activated factor VII)-integrin ß1-PAR2 (protease-activated receptor 2) signaling complex by utilizing genetic mouse models and pharmacological intervention. RESULTS Cleavage-insensitive PAR2R38E and myeloid cell integrin ß1-deficient mice had improved cardiac function after MI compared with controls. Proximity ligation assays of monocytic cells demonstrated that colocalization of FVIIa with integrin ß1 was diminished in monocyte/macrophage FVII-deficient mice after MI. Compared with controls, F7fl/fl CX3CR1 (CX3C motif chemokine receptor 1)Cre mice showed reduced TGF-ß1 and MAP kinase activation, as well as cardiac dysfunction after MI, despite unaltered overall recruitment of myeloid cells. Single-cell mRNA sequencing of CD45 (cluster of differentiation 45)+ cells 3 and 7 days after MI uncovered a trajectory from recruited monocytes to inflammatory TF+/TREM (triggered receptor expressed on myeloid cells) 1+ macrophages requiring F7. As early as 7 days after MI, macrophage F7 deletion led to an expansion of reparative Olfml 3 (olfactomedin-like protein 3)+ macrophages and, conversely, to a reduction of TF+/TREM1+ macrophages, which were also reduced in PAR2R38E mice. Short-term treatment from days 1 to 5 after nonreperfused MI with a monoclonal antibody inhibiting the macrophage TF-FVIIa-PAR2 signaling complex without anticoagulant activity improved cardiac dysfunction, decreased excess fibrosis, attenuated vascular endothelial dysfunction, and increased survival 28 days after MI. CONCLUSIONS Extravascular TF-FVIIa-PAR2 complex signaling drives inflammatory macrophage polarization in ischemic heart disease. Targeting this signaling complex for specific therapeutic macrophage reprogramming following MI attenuates cardiac fibrosis and improves cardiovascular function.
Collapse
Affiliation(s)
- Venkata Garlapati
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
| | - Qi Luo
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Biochemistry, Cardiovascular Research Maastricht University, the Netherlands (Q.L.)
| | - Jens Posma
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
| | - Melania Aluia
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
| | - Than Son Nguyen
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
| | - Kristin Grunz
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
| | - Michael Molitor
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
| | - Gregory Harms
- Institute of Immunology and Research Center for Immunotherapy (G.H., T.B.), University Medical Center Mainz, Germany
- Cell Biology Unit (G.H.), University Medical Center Mainz, Germany
- Department of Biology, Wilkes University, Wilkes-Barre, PA (G.H.)
| | - Tobias Bopp
- Institute of Immunology and Research Center for Immunotherapy (G.H., T.B.), University Medical Center Mainz, Germany
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Immunology and Microbiology, Scripps Research, La Jolla, CA (W.R.)
| | - Philip Wenzel
- Center for Thrombosis and Hemostasis (V.G., Q.L., J.P., M.A., T.S.N., K.G., M.M., S.F., W.R., P.W.), University Medical Center Mainz, Germany
- Department of Cardiology (V.G., Q.L., M.A., M.M., S.F., P.W.), University Medical Center Mainz, Germany
- German Center for Cardiovascular Research-Partner site Rhine-Main (V.G., Q.L., M.A., M.M., W.R., P.W.), University Medical Center Mainz, Germany
| |
Collapse
|
21
|
Hota S, Kumar M. ErpY-like Protein Interaction with Host Thrombin and Fibrinogen Intervenes the Plasma Coagulation through Extrinsic and Intrinsic Pathways. ACS Infect Dis 2024; 10:3256-3272. [PMID: 39231002 DOI: 10.1021/acsinfecdis.4c00266] [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] [Indexed: 09/06/2024]
Abstract
The survival and proliferation of pathogenic Leptospira within a host are complex phenomena that require careful consideration. The ErpY-like lipoprotein, found on the outer membrane surface of Leptospira, plays a crucial role in enhancing the bacterium's pathogenicity. The rErpY-like protein, in its recombinant form, contributes significantly to spirochete virulence by interacting with various host factors, including host complement regulators. This interaction facilitates the bacterium's evasion of the host complement system, thereby augmenting its overall pathogenicity. The rErpY-like protein exhibits a robust binding affinity to soluble fibrinogen, a vital component of the host coagulation system. In this study, we demonstrate that the rErpY-like protein intervenes in the clotting process of the platelet-poor citrated plasma of bovines and humans in a concentration-dependent manner. It significantly reduces clot density, alters the viscoelastic properties of the clot, and diminishes the average clotting rate in plasma. Furthermore, the ErpY-like protein inhibits thrombin-catalyzed fibrin formation in a dose-dependent manner and exhibits saturable binding to thrombin, suggesting its significant role in leptospiral infection. These findings provide compelling evidence for the anticoagulant effect of the ErpY-like lipoprotein and its significant role in leptospiral infection.
Collapse
Affiliation(s)
- Saswat Hota
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| | - Manish Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam 781039, India
| |
Collapse
|
22
|
Marrufo AM, Flores-Mireles AL. Macrophage fate: to kill or not to kill? Infect Immun 2024; 92:e0047623. [PMID: 38829045 PMCID: PMC11385966 DOI: 10.1128/iai.00476-23] [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] [Indexed: 06/05/2024] Open
Abstract
Macrophages are dynamic innate immune cells that either reside in tissue, serving as sentinels, or recruited as monocytes from bone marrow into inflamed and infected tissue. In response to cues in the tissue microenvironment (TME), macrophages polarize on a continuum toward M1 or M2 with diverse roles in progression and resolution of disease. M1-like macrophages exhibit proinflammatory functions with antimicrobial and anti-tumorigenic activities, while M2-like macrophages have anti-inflammatory functions that generally resolve inflammatory responses and orchestrate a tissue healing process. Given these opposite phenotypes, proper spatiotemporal coordination of macrophage polarization in response to cues within the TME is critical to effectively resolve infectious disease and regulate wound healing. However, if this spatiotemporal coordination becomes disrupted due to persistent infection or dysregulated coagulation, macrophages' inappropriate response to these cues will result in the development of diseases with clinically unfavorable outcomes. Since plasticity and heterogeneity are hallmarks of macrophages, they are attractive targets for therapies to reprogram toward specific phenotypes that could resolve disease and favor clinical prognosis. In this review, we discuss how basic science studies have elucidated macrophage polarization mechanisms in TMEs during infections and inflammation, particularly coagulation. Therefore, understanding the dynamics of macrophage polarization within TMEs in diseases is important in further development of targeted therapies.
Collapse
Affiliation(s)
- Armando M. Marrufo
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, USA
| | | |
Collapse
|
23
|
Ballard-Kordeliski A, Lee RH, O’Shaughnessy EC, Kim PY, Jones SR, Pawlinski R, Flick MJ, Paul DS, Mackman N, Adalsteinsson DA, Bergmeier W. 4D intravital imaging studies identify platelets as the predominant cellular procoagulant surface in a mouse hemostasis model. Blood 2024; 144:1116-1126. [PMID: 38820498 PMCID: PMC11406176 DOI: 10.1182/blood.2023022608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 05/12/2024] [Accepted: 05/13/2024] [Indexed: 06/02/2024] Open
Abstract
ABSTRACT Interplay between platelets, coagulation factors, endothelial cells (ECs), and fibrinolytic factors is necessary for effective hemostatic plug formation. This study describes a 4-dimensional (4D) imaging platform to visualize and quantify hemostatic plug components in mice with high spatiotemporal resolution. Fibrin accumulation after laser-induced vascular injury was observed at the platelet plug-EC interface, controlled by the antagonistic balance between fibrin generation and breakdown. We observed less fibrin accumulation in mice expressing low levels of tissue factor or F12-/-mice compared with controls, whereas increased fibrin accumulation, including on the vasculature adjacent to the platelet plug, was observed in plasminogen-deficient mice or wild-type mice treated with tranexamic acid. Phosphatidylserine (PS), a membrane lipid critical for the assembly of coagulation factors, was first detected at the platelet plug-EC interface, followed by exposure across the endothelium. Impaired PS exposure resulted in a significant reduction in fibrin accumulation in cyclophilin D-/-mice. Adoptive transfer studies demonstrated a key role for PS exposure on platelets, and to a lesser degree on ECs, in fibrin accumulation during hemostatic plug formation. Together, these studies suggest that (1) platelets are the functionally dominant procoagulant cellular surface, and (2) plasmin is critical for limiting fibrin accumulation at the site of a forming hemostatic plug.
Collapse
Affiliation(s)
- Abigail Ballard-Kordeliski
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Robert H. Lee
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Ellen C. O’Shaughnessy
- Department of Cell Biology and Physiology, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Paul Y. Kim
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Thrombosis and Atherosclerosis Research Institute, Hamilton, ON, Canada
| | - Summer R. Jones
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rafal Pawlinski
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Matthew J. Flick
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David S. Paul
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nigel Mackman
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - David A. Adalsteinsson
- Department of Mathematics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Wolfgang Bergmeier
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
24
|
Martinez Bravo G, Annarapu G, Carmona E, Nawarskas J, Clark R, Novelli E, Mota Alvidrez RI. Platelets in Thrombosis and Atherosclerosis: A Double-Edged Sword. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1608-1621. [PMID: 38885926 PMCID: PMC11373056 DOI: 10.1016/j.ajpath.2024.05.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/16/2024] [Accepted: 05/16/2024] [Indexed: 06/20/2024]
Abstract
This review focuses on the dual role of platelets in atherosclerosis and thrombosis, exploring their involvement in inflammation, angiogenesis, and plaque formation, as well as their hemostatic and prothrombotic functions. Beyond their thrombotic functions, platelets engage in complex interactions with diverse cell types, influencing disease resolution and progression. The contribution of platelet degranulation helps in the formation of atheromatous plaque, whereas the reciprocal interaction with monocytes adds complexity. Alterations in platelet membrane receptors and signaling cascades contribute to advanced atherosclerosis, culminating in atherothrombotic events. Understanding these multifaceted roles of platelets will lead to the development of targeted antiplatelet strategies for effective cardiovascular disease prevention and treatment. Understanding platelet functions in atherosclerosis and atherothrombosis at different stages of disease will be critical for designing targeted treatments and medications to prevent or cure the disease Through this understanding, platelets can be targeted at specific times in the atherosclerosis process, possibly preventing the development of atherothrombosis.
Collapse
Affiliation(s)
| | - Gowtham Annarapu
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Emely Carmona
- School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - James Nawarskas
- Pharmaceutical Sciences-Pharmacy Practice, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico
| | - Ross Clark
- Cell Biology and Physiology, University of New Mexico, Albuquerque, New Mexico; Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico
| | - Enrico Novelli
- Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, Pennsylvania; School of Medicine, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Roberto I Mota Alvidrez
- Biomedical Engineering Department, University of New Mexico, Albuquerque, New Mexico; Pharmaceutical Sciences-Pharmacy Practice, College of Pharmacy, University of New Mexico, Albuquerque, New Mexico; Clinical and Translational Science Center, University of New Mexico, Albuquerque, New Mexico.
| |
Collapse
|
25
|
Siti-Zubaidah MZ, Harafinova HS, Liba AN, Nordin ML, Hambali KA, Siti HN. Exploring bradykinin: A common mediator in the pathophysiology of sepsis and atherosclerotic cardiovascular disease. Vascul Pharmacol 2024; 156:107414. [PMID: 39089528 DOI: 10.1016/j.vph.2024.107414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 07/16/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Sepsis and atherosclerotic cardiovascular disease (ASCVD) are major health challenges involving complex processes like inflammation, renin-angiotensin system (RAS) dysregulation, and thrombosis. Despite distinct clinical symptoms, both conditions share mechanisms mediated by bradykinin. This review explores bradykinin's role in inflammation, RAS modulation, and thrombosis in sepsis and ASCVD. In sepsis, variable kininogen-bradykinin levels may correlate with disease severity and progression, though the effect of bradykinin receptor modulation on inflammation remains uncertain. RAS activation is present in both diseases, with sepsis showing variable or low levels of Ang II, ACE, and ACE2, while ASCVD consistently exhibits elevated levels. Bradykinin may act as a mediator for ACE2 and AT2 receptor effects in RAS regulation. It may influence clotting and fibrinolysis in sepsis-associated coagulopathy, but evidence for an antithrombotic effect in ASCVD is insufficient. Understanding bradykinin's role in these shared pathologies could guide therapeutic and monitoring strategies and inform future research.
Collapse
Affiliation(s)
- Mohd Zahari Siti-Zubaidah
- Department of Anaesthesia and Intensive Care, National Heart Institute, Jalan Tun Razak, 50400 Kuala Lumpur, Malaysia.
| | - Harman-Shah Harafinova
- Department of Internal Medicine, Faculty of Medicine, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia.
| | - Abdullahi Nuradeen Liba
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa, Kota Bharu, 16100, Kelantan, Malaysia
| | - Muhammad Luqman Nordin
- Department of Veterinary Clinical Studies, Faculty of Veterinary Medicine, Universiti Malaysia Kelantan, Pengkalan Chepa, Kota Bharu, 16100, Kelantan, Malaysia; Department of Pharmaceutical Technology, Faculty of Pharmacy, Universiti Malaya, 50603 Kuala Lumpur, Malaysia.
| | - Kamarul Ariffin Hambali
- Faculty of Earth Science, Universiti Malaysia Kelantan, Jeli, 17600, Kelantan, Malaysia; Animal and Wildlife Research Group, Faculty of Earth Science, Jeli Campus, Universiti Malaysia Kelantan, 17600, Kelantan, Malaysia.
| | - Hawa Nordin Siti
- Department of Pharmacology, Faculty of Medicine, Universiti Sultan Zainal Abidin, Jalan Sultan Mahmud, 20400 Kuala Terengganu, Terengganu, Malaysia.
| |
Collapse
|
26
|
Cirillo P, Morello M, Titolo G, Marra L, Morello A, De Rosa G, Cozzolino D, Sugraliyev A, Cimmino G. E-Cigarettes induce expression of procoagulant tissue factor in cultivated human endothelial cells. J Thromb Thrombolysis 2024:10.1007/s11239-024-03018-6. [PMID: 39207592 DOI: 10.1007/s11239-024-03018-6] [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] [Accepted: 07/02/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND E-cigarettes (ECIG) are proposed as an alternative for regular tobacco users with less dangerous effects for health. Several studies demonstrated that ECIG exert deleterious cardiovascular effects and promote platelet dependent thrombosis. However, ECIG role on Tissue Factor-dependent thrombosis is still unknown. Dysfunctional endothelial cells (ECs) are known to express Tissue Factor (TF) on their surface. Aim of the present study was to investigate whether ECIG might promote TF expression in ECs, shifting them to a pro thrombotic phenotype. METHODS Human Umbilical Vein Endothelial Cells (HUVEC) were incubated with increasing doses of ECIG (commercially available and mix of propylene glycol/vegetable glycerine/nicotine 18 mg/mL) up to 1.8 mg/mL. TF gene expression and protein levels were assessed at different time points by Real Time PCR and Western Blot, respectively. TF surface expression and activity were also measured by FACS analysis and coagulation assay. Finally, NF-kB translocation was investigated as possible mechanism of action. Potential protective effects by Rosuvastatin were also investigated. RESULTS ECIG significantly increased TF expression at both gene and protein levels in a time and dose dependent manner. Surface expression and procoagulant activity were increased as well. These phenomena appeared modulated by the NF-κB pathway. Rosuvastatin reduced ECIG effects on TF-mRNA. CONCLUSIONS Although in vitro, we indicate that ECIG promote a pro thrombotic phenotype in ECs via expression of functional TF. Data of the present study permit to shed a brighter light on the still partially unresolved issue about the role of ECIG in development of cardiovascular diseases suggesting that they might represent a potential risk factor for thrombotic cardiovascular events.
Collapse
Affiliation(s)
- Plinio Cirillo
- Department of Advanced Biomedical Sciences, Division of Cardiology, University of Naples "Federico II", Via Pansini, 5, Naples, 80131, Italy.
| | - Mariarosaria Morello
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gisella Titolo
- Department of Translational Medical Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Laura Marra
- SC Cell Biology and Biotherapy, Istituto Nazionale Tumori IRCCS, Fondazione G. Pascale, Naples, Italy
| | - Andrea Morello
- Biochemical Unit, Azienda Sanitaria Regionale Molise, Antonio Cardarelli Hospital, Campobasso, Italy
| | - Gennaro De Rosa
- Department of Advanced Biomedical Sciences, Division of Cardiology, University of Naples "Federico II", Via Pansini, 5, Naples, 80131, Italy
| | - Domenico Cozzolino
- Department of Precision Medicine, University of Campania, Caserta, Italy
| | - Akhmetzhan Sugraliyev
- Department of Internal Disease, Kazakh National Medical University, Almaty, Kazakhstan
| | - Giovanni Cimmino
- Department of Advanced Biomedical Sciences, Division of Cardiology, University of Naples "Federico II", Via Pansini, 5, Naples, 80131, Italy
| |
Collapse
|
27
|
Li J, Chen H, Xu C, Hu M, Li J, Chang W. Systemic toxicity of CAR-T therapy and potential monitoring indicators for toxicity prevention. Front Immunol 2024; 15:1422591. [PMID: 39253080 PMCID: PMC11381299 DOI: 10.3389/fimmu.2024.1422591] [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: 04/24/2024] [Accepted: 08/08/2024] [Indexed: 09/11/2024] Open
Abstract
Malignant tumors of the hematologic system have a high degree of malignancy and high mortality rates. Chimeric antigen receptor T cell (CAR-T) therapy has become an important option for patients with relapsed/refractory tumors, showing astonishing therapeutic effects and thus, it has brought new hope to the treatment of malignant tumors of the hematologic system. Despite the significant therapeutic effects of CAR-T, its toxic reactions, such as Cytokine Release Syndrome (CRS) and Immune Effector Cell-Associated Neurotoxicity Syndrome (ICANS), cannot be ignored since they can cause damage to multiple systems, including the cardiovascular system. We summarize biomarkers related to prediction, diagnosis, therapeutic efficacy, and prognosis, further exploring potential monitoring indicators for toxicity prevention. This review aims to summarize the effects of CAR-T therapy on the cardiovascular, hematologic, and nervous systems, as well as potential biomarkers, and to explore potential monitoring indicators for preventing toxicity, thereby providing references for clinical regulation and assessment of therapeutic effects.
Collapse
Affiliation(s)
- Jingxian Li
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Huiguang Chen
- Institute of Infection, Immunology and Tumor Microenvironment, School of Medicine, Wuhan University of Science and Technology, Wuhan, China
| | - Chaoping Xu
- Department of Hematology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Mengci Hu
- Department of Hematology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Jiangping Li
- Department of Blood Transfusion, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Wei Chang
- Department of Hematology, Puren Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
28
|
Song Y, Lu J, Qin P, Chen H, Chen L. Interferon-I modulation and natural products: Unraveling mechanisms and therapeutic potential in severe COVID-19. Cytokine Growth Factor Rev 2024:S1359-6101(24)00066-2. [PMID: 39261232 DOI: 10.1016/j.cytogfr.2024.08.005] [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: 08/06/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to pose a significant global public health threat, particularly to older adults, pregnant women, and individuals with underlying chronic conditions. Dysregulated immune responses to SARS-CoV-2 infection are believed to contribute to the progression of COVID-19 in severe cases. Previous studies indicates that a deficiency in type I interferon (IFN-I) immunity accounts for approximately 15 %-20 % of patients with severe pneumonia caused by COVID-19, highlighting the potential therapeutic importance of modulating IFN-I signals. Natural products and their derivatives, due to their structural diversity and novel scaffolds, play a crucial role in drug discovery. Some of these natural products targeting IFN-I have demonstrated applications in infectious diseases and inflammatory conditions. However, the immunomodulatory potential of IFN-I in critical COVID-19 pneumonia and the natural compounds regulating the related signal pathway remain not fully understood. In this review, we offer a comprehensive assessment of the association between IFN-I and severe COVID-19, exploring its mechanisms and integrating information on natural compounds effective for IFN-I regulation. Focusing on the primary targets of IFN-I, we also summarize the regulatory mechanisms of natural products, their impact on IFNs, and their therapeutic roles in viral infections. Collectively, by synthesizing these findings, our goal is to provide a valuable reference for future research and to inspire innovative treatment strategies for COVID-19.
Collapse
Affiliation(s)
- Yuheng Song
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; School of Pharmacy, Henan University, Kaifeng 475001, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Research Center for Traditional Chinese Medicine, Shanghai Institute of Infectious Diseases and Biosecurity, Fudan University, Shanghai 200032, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
29
|
Chen M, Wu Q, Shao N, Lai X, Lin H, Chen M, Wu Y, Chen J, Lin Q, Huang J, Chen X, Yan W, Chen S, Li H, Wu D, Yang M, Deng C. The significance of CD16+ monocytes in the occurrence and development of chronic thromboembolic pulmonary hypertension: insights from single-cell RNA sequencing. Front Immunol 2024; 15:1446710. [PMID: 39192976 PMCID: PMC11347785 DOI: 10.3389/fimmu.2024.1446710] [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: 06/10/2024] [Accepted: 07/17/2024] [Indexed: 08/29/2024] Open
Abstract
Background Chronic thromboembolic pulmonary hypertension (CTEPH) is a serious pulmonary vascular disease characterized by residual thrombi in the pulmonary arteries and distal pulmonary microvascular remodeling. The pathogenesis of CTEPH remains unclear, but many factors such as inflammation, immunity, coagulation and angiogenesis may be involved. Monocytes are important immune cells that can differentiate into macrophages and dendritic cells and play an important role in thrombus formation. However, the distribution, gene expression profile and differentiation trajectory of monocyte subsets in CTEPH patients have not been systematically studied. This study aims to reveal the characteristics and functions of monocytes in CTEPH patients using single-cell sequencing technology, and to provide new insights for the diagnosis and treatment of CTEPH. Methods Single-cell RNA sequencing (scRNA-seq) were performed to analyze the transcriptomic features of peripheral blood mononuclear cells (PBMCs) from healthy controls, CTEPH patients and the tissues from CTEPH patients after the pulmonary endarterectomy (PEA). We established a CTEPH rat model with chronic pulmonary embolism caused by repeated injection of autologous thrombi through a central venous catheter, and used flow cytometry to detect the proportion changes of monocyte subsets in CTEPH patients and CTEPH rat model. We also observed the infiltration degree of macrophage subsets in thrombus tissue and their differentiation relationship with peripheral blood monocyte subsets by immunofluorescence staining. Results The results showed that the monocyte subsets in peripheral blood of CTEPH patients changed significantly, especially the proportion of CD16+ monocyte subset increased. This monocyte subset had unique functional features at the transcriptomic level, involving processes such as cell adhesion, T cell activation, coagulation response and platelet activation, which may play an important role in pulmonary artery thrombus formation and pulmonary artery intimal remodeling. In addition, we also found that the macrophage subsets in pulmonary endarterectomy tissue of CTEPH patients showed pro-inflammatory and lipid metabolism reprogramming features, which may be related to the persistence and insolubility of pulmonary artery thrombi and the development of pulmonary hypertension. Finally, we also observed that CD16+ monocyte subset in peripheral blood of CTEPH patients may be recruited to pulmonary artery intimal tissue and differentiate into macrophage subset with high expression of IL-1β, participating in disease progression. Conclusion CD16+ monocytes subset had significant gene expression changes in CTEPH patients, related to platelet activation, coagulation response and inflammatory response. And we also found that these cells could migrate to the thrombus and differentiate into macrophages with high expression of IL-1β involved in CTEPH disease progression. We believe that CD16+ monocytes are important participants in CTEPH and potential therapeutic targets.
Collapse
Affiliation(s)
- Maohe Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Qiuxia Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Nan Shao
- Division of Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xingyue Lai
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Huo Lin
- Department of Pulmonary and Critical Care Medicine, Shishi County Hospital, Shishi, China
| | - Min Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Yijing Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Jiafan Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Qinghuang Lin
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Jiahui Huang
- Department of Respiratory and Critical Care Medicine, Fuqing City Hospital Affiliated to Fujian Medical University, Fuzhou, China
| | - Xiaoyun Chen
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Wei Yan
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
| | - Shi Chen
- Department of Respiratory and Critical Care, Wuhan No. 6 Hospital, Affiliated Hospital of Jianghan University, Wuhan, China
| | - Hongli Li
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Dawen Wu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Minxia Yang
- Division of Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Chaosheng Deng
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Institute of Respiratory Disease, Fujian Medical University, Fuzhou, China
- Department of Respiratory and Critical Care Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
30
|
Moruzzi S, Castagna A, Spizzo M, Udali S, Pattini P, Pizzolo F, Friso S, Martinelli N. Activated Factor VII-Antithrombin Complex, a Biomarker of Tissue Factor-Related Pathways in Different Clinical Settings: A Narrative Review from Cardiovascular Diseases to Cancer. Diagnostics (Basel) 2024; 14:1711. [PMID: 39202199 PMCID: PMC11354109 DOI: 10.3390/diagnostics14161711] [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: 07/08/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/03/2024] Open
Abstract
Tissue factor (TF) is a transmembrane glycoprotein that represents the fundamental physiological initiator of the coagulation cascade through its interaction with factor VII. TF belongs to the cytokine receptor protein superfamily and contributes to the transduction of cellular signaling. Therefore, TF-related pathways are involved in multiple pathophysiological processes, not only in coagulation/thrombosis but in a wider mechanisms' panorama, ranging from infective to neoplastic diseases. Consistently, the measurement of TF activity could have a diagnostic and/or prognostic meaning in different clinical conditions. However, the transmembrane localization, the expression on different cellular types and circulating extracellular vesicles, and the different conformations (encrypted and decrypted) and variants (such as the soluble alternatively spliced TF) hamper TF assessment in clinical practice. The activated factor VII-antithrombin (FVIIa-AT) complex is proposed as an indirect biomarker of the TF-FVIIa interaction and, consequently, of the functionally active TF expression. In this narrative review, we evaluate the clinical studies investigating the role of plasma concentration of FVIIa-AT in health and disease. Although without conclusive data, high FVIIa-AT concentrations predict the worst clinical outcomes in different pathologic conditions, such as cardiovascular disease and cancer, thereby suggesting that overactivation of TF-related pathways may play an unfavorable role in various clinical settings.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Nicola Martinelli
- Department of Medicine, University of Verona, 37134 Verona, Italy; (S.M.); (A.C.); (M.S.); (S.U.); (P.P.); (F.P.); (S.F.)
| |
Collapse
|
31
|
Saunderson SC, Halpin JC, Tan GMY, Shrivastava P, McLellan AD. Conversion of anti-tissue factor antibody sequences to chimeric antigen receptor and bi-specific T-cell engager format. Cancer Immunol Immunother 2024; 73:195. [PMID: 39105809 PMCID: PMC11303627 DOI: 10.1007/s00262-024-03778-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND The efficacy of antibody-targeted therapy of solid cancers is limited by the lack of consistent tumour-associated antigen expression. However, tumour-associated antigens shared with non-malignant cells may still be targeted using conditionally activated-antibodies, or by chimeric antigen receptor (CAR) T cells or CAR NK cells activated either by the tumour microenvironment or following 'unlocking' via multiple antigen-recognition. In this study, we have focused on tissue factor (TF; CD142), a type I membrane protein present on a range of solid tumours as a basis for future development of conditionally-activated BiTE or CAR T cells. TF is frequently upregulated on multiple solid tumours providing a selective advantage for growth, immune evasion and metastasis, as well as contributing to the pathology of thrombosis via the extrinsic coagulation pathway. METHODS Two well-characterised anti-TF monoclonal antibodies (mAb) were cloned into expression or transposon vectors to produce single chain (scFv) BiTE for assessment as CAR and CD28-CD3-based CAR or CD3-based BiTE. The affinities of both scFv formats for TF were determined by surface plasmon resonance. Jurkat cell line-based assays were used to confirm the activity of the BiTE or CAR constructs. RESULTS The anti-TF mAb hATR-5 and TF8-5G9 mAb were shown to maintain their nanomolar affinities following conversion into a single chain (scFv) format and could be utilised as CD28-CD3-based CAR or CD3-based BiTE format. CONCLUSION Because of the broad expression of TF on a range of solid cancers, anti-TF antibody formats provide a useful addition for the development of conditionally activated biologics for antibody and cellular-based therapy.
Collapse
Affiliation(s)
- S C Saunderson
- Department of Microbiology and Immunology, University of Otago, Dunedin, 9016, New Zealand
| | - J C Halpin
- Department of Microbiology and Immunology, University of Otago, Dunedin, 9016, New Zealand
- The Children's Hospital Westmead, The Children's Hospital Westmead CRN Hawksbury Road and Hainsworth Street, Westmead, NSW, 2145, Australia
| | - G M Y Tan
- Department of Microbiology and Immunology, University of Otago, Dunedin, 9016, New Zealand
- Molecular and Clinical Cancer Medicine, The University of Liverpool, Crown St., Liverpool, UK
| | - P Shrivastava
- Department of Microbiology and Immunology, University of Otago, Dunedin, 9016, New Zealand
| | - A D McLellan
- Department of Microbiology and Immunology, University of Otago, Dunedin, 9016, New Zealand.
| |
Collapse
|
32
|
Alradwan I, AL Fayez N, Alomary MN, Alshehri AA, Aodah AH, Almughem FA, Alsulami KA, Aldossary AM, Alawad AO, Tawfik YMK, Tawfik EA. Emerging Trends and Innovations in the Treatment and Diagnosis of Atherosclerosis and Cardiovascular Disease: A Comprehensive Review towards Healthier Aging. Pharmaceutics 2024; 16:1037. [PMID: 39204382 PMCID: PMC11360443 DOI: 10.3390/pharmaceutics16081037] [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: 05/26/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 09/04/2024] Open
Abstract
Cardiovascular diseases (CVDs) are classed as diseases of aging, which are associated with an increased prevalence of atherosclerotic lesion formation caused by such diseases and is considered as one of the leading causes of death globally, representing a severe health crisis affecting the heart and blood vessels. Atherosclerosis is described as a chronic condition that can lead to myocardial infarction, ischemic cardiomyopathy, stroke, and peripheral arterial disease and to date, most pharmacological therapies mainly aim to control risk factors in patients with cardiovascular disease. Advances in transformative therapies and imaging diagnostics agents could shape the clinical applications of such approaches, including nanomedicine, biomaterials, immunotherapy, cell therapy, and gene therapy, which are emerging and likely to significantly impact CVD management in the coming decade. This review summarizes the current anti-atherosclerotic therapies' major milestones, strengths, and limitations. It provides an overview of the recent discoveries and emerging technologies in nanomedicine, cell therapy, and gene and immune therapeutics that can revolutionize CVD clinical practice by steering it toward precision medicine. CVD-related clinical trials and promising pre-clinical strategies that would significantly impact patients with CVD are discussed. Here, we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD medicine.
Collapse
Affiliation(s)
- Ibrahim Alradwan
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Nojoud AL Fayez
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Mohammad N. Alomary
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Abdullah A. Alshehri
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Alhassan H. Aodah
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Fahad A. Almughem
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Khulud A. Alsulami
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| | - Ahmad M. Aldossary
- Wellness and Preventative Medicine Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Abdullah O. Alawad
- Healthy Aging Research Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia;
| | - Yahya M. K. Tawfik
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Essam A. Tawfik
- Advanced Diagnostics and Therapeutics Institute, Health Sector, King Abdulaziz City for Science and Technology (KACST), Riyadh 11442, Saudi Arabia; (I.A.); (N.A.F.); (M.N.A.); (A.A.A.); (A.H.A.); (F.A.A.); (K.A.A.)
| |
Collapse
|
33
|
Park JC, Shin D. Current Landscape of Antibody-Drug Conjugate Development in Head and Neck Cancer. JCO Precis Oncol 2024; 8:e2400179. [PMID: 39151109 DOI: 10.1200/po.24.00179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 06/14/2024] [Accepted: 07/22/2024] [Indexed: 08/18/2024] Open
Abstract
Antibody-drug conjugates (ADCs) are fusions of therapeutic drugs and antibodies conjugated by a linker, designed to deliver a therapeutic payload to cells expressing the target antigen. By delivering the highly cytotoxic agent directly to cancer cells, ADCs are designed to enhance safety and broaden the therapeutic window. Recently, ADCs have demonstrated promising efficacy in various solid tumors and are rapidly expanding their indications. The prognosis of patients with advanced head and neck squamous cell carcinoma (HNSCC) remains poor, with no new therapeutics since the advent of anti-PD-1 antibodies in 2016, highlighting a critical need for innovative therapies. Recent preliminary results suggest that ADCs could be promising treatment options for HNSCC as they explore a variety of target antigens, payloads, and linkers. However, for successful adaptation of ADCs in the treatment of HNSCC, addressing key challenges such as payload toxicities, antigen heterogeneity, and adaptive resistance will be essential. Current research focused on new ADC structures, including multispecific antibodies and noncytotoxic payloads, and diverse combination approaches, show promise for future advancements.
Collapse
Affiliation(s)
- Jong Chul Park
- Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Donghoon Shin
- MetroWest Medical Center, Tufts University School of Medicine, Framingham, MA
| |
Collapse
|
34
|
Jia Y, Zhang K, Shi M, Guo D, Yang P, Bu X, Chen J, Wang A, Xu T, He J, Zhu Z, Zhang Y. Associations of Rheumatoid Factor, Rheumatoid Arthritis, and Interleukin-6 Inhibitor with the Prognosis of Ischemic Stroke: a Prospective Multicenter Cohort Study and Mendelian Randomization Analysis. Transl Stroke Res 2024; 15:750-760. [PMID: 37256492 DOI: 10.1007/s12975-023-01161-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 04/23/2023] [Accepted: 05/21/2023] [Indexed: 06/01/2023]
Abstract
Rheumatoid factor (RF), an established diagnostic biomarker for rheumatoid arthritis (RA), is associated with cardiovascular diseases, but its impact on clinical outcomes of ischemic stroke remains unclear. We aimed to investigate the observational associations between serum RF and prognosis of ischemic stroke, and further examined the genetic associations of RA and its therapeutic strategy, interleukin-6 (IL-6) inhibitor, with prognosis of ischemic stroke. We measured serum RF levels in 3474 Chinese ischemic stroke patients from the China Antihypertensive Trial in Acute Ischemic Stroke. The primary outcome was the composite outcome of death or major disability (modified Rankin Scale score ≥3) at 3 months after stroke onset. Mendelian randomization (MR) analyses were performed to examine the associations of genetically predicted RA and IL-6 inhibition with prognosis of ischemic stroke. During 3 months of follow-up, 866 patients (25.43%) experienced death or major disability. After multivariate adjustment, RF-positive was significantly associated with a high risk of primary outcome (OR, 1.47; 95% CI, 1.08-2.00; P =0.016) compared with RF-negative. The two-sample MR analyses suggested that genetically predicted RA was associated with an increased risk of primary outcome (OR, 1.09; 95% CI, 1.01-1.18; P=0.021), while genetically predicted IL-6 inhibition was associated with a decreased risk of primary outcome (OR, 0.88; 95% CI, 0.77-0.99; P=0.041). We found that positive RF was associated with increased risks of adverse outcomes after atherosclerotic ischemic stroke, and genetically predicted RA and IL-6 inhibition increased and decreased the risks of adverse outcomes after ischemic stroke, respectively.
Collapse
Affiliation(s)
- Yiming Jia
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China
| | - Kaixin Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China
| | - Mengyao Shi
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Daoxia Guo
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China
- School of Nursing, Suzhou Medical College of Soochow University, Suzhou, China
| | - Pinni Yang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China
| | - Xiaoqing Bu
- Department of Epidemiology, School of Public health, Chongqing Medical University, Chongqing, China
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Aili Wang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China
| | - Tan Xu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Zhengbao Zhu
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China.
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA.
| | - Yonghong Zhang
- Department of Epidemiology, School of Public Health and Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Suzhou Medical College of Soochow University, 199 Renai Road, Industrial Park District, Suzhou, 215123, Jiangsu Province, China.
| |
Collapse
|
35
|
Hisada Y, Archibald SJ, Bansal K, Chen Y, Dai C, Dwarampudi S, Balas N, Hageman L, Key NS, Bhatia S, Bhatia R, Mackman N, Gangaraju R. Biomarkers of bleeding and venous thromboembolism in patients with acute leukemia. J Thromb Haemost 2024; 22:1984-1996. [PMID: 38574862 PMCID: PMC11214882 DOI: 10.1016/j.jtha.2024.03.020] [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: 10/18/2023] [Revised: 03/01/2024] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Coagulopathy and associated bleeding and deep vein thrombosis (DVT) are major causes of morbidity and mortality in patients with acute leukemia. The underlying mechanisms of these complications have not been fully elucidated. OBJECTIVES To evaluate the associations between biomarker levels and bleeding and DVT in acute leukemia patients. METHODS We examined plasma levels of activators, inhibitors, and biomarkers of the coagulation and fibrinolytic pathways in patients aged ≥18 years with newly diagnosed acute leukemia compared with those of normal controls. Multivariable regression models were used to examine the association of biomarkers with bleeding and DVT in acute leukemia patients. The study included 358 patients with acute leukemia (29 with acute promyelocytic leukemia [APL], 253 with non-APL acute myeloid leukemia, and 76 with acute lymphoblastic leukemia) and 30 normal controls. RESULTS Patients with acute leukemia had higher levels of extracellular vesicle tissue factor (EVTF) activity, phosphatidylserine-positive extracellular vesicles, plasminogen activator inhibitor-1, plasmin-antiplasmin complexes, and cell-free DNA and lower levels of citrullinated histone H3-DNA complexes compared with normal controls. APL patients had the highest levels of EVTF activity and the lowest levels of tissue plasminogen activator among acute leukemia patients. There were 41 bleeding and 23 DVT events in acute leukemia patients. High EVTF activity was associated with increased risk of bleeding (subdistribution hazard ratio, 2.30; 95% CI, 0.99-5.31), whereas high levels of plasminogen activator inhibitor-1 were associated with increased risk of DVT (subdistribution hazard ratio, 3.00; 95% CI, 0.95-9.47) in these patients. CONCLUSION Our study shows alterations in several biomarkers in acute leukemia and identifies biomarkers associated with risk of bleeding and DVT.
Collapse
Affiliation(s)
- Yohei Hisada
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| | - Sierra J Archibald
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Karan Bansal
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chen Dai
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Sindhu Dwarampudi
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nora Balas
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Lindsey Hageman
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nigel S Key
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Ravi Bhatia
- Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Nigel Mackman
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Radhika Gangaraju
- Institute for Cancer Outcomes and Survivorship, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA; Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, USA.
| |
Collapse
|
36
|
Kayalar O, Bayrak BB, Yildirim M, Yanardag R, Oztay F. Retinoic acid reduces kidney injury by regulating oxidative stress, NRF-2, and apoptosis in hyperoxic mice. Cell Biochem Funct 2024; 42:e4094. [PMID: 39001564 DOI: 10.1002/cbf.4094] [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: 04/16/2024] [Revised: 06/03/2024] [Accepted: 07/01/2024] [Indexed: 07/19/2024]
Abstract
Nuclear factor-erythroid-2-related factor-2 (NRF-2) is a cellular resistance protein to oxidants. We investigated the effect of exogenous all-trans retinoic acid (ATRA) on the antioxidant system and NRF-2 in mice kidneys under hyperoxia-induced oxidative stress. Mice were divided into four groups. Daily, two groups were given either peanut-oil/dimethyl sulfoxide (PoDMSO) mixture or 50 mg/kg ATRA. Oxidative stress was induced by hyperoxia in the remaining groups. They were treated with PoDMSO or ATRA as described above, following hyperoxia (100% oxygen) for 72 h. NRF-2 and active-caspase-3 levels, lipid peroxidation (LPO), activities of antioxidant enzymes, xanthine oxidase (XO), paraoxonase1 (PON1), lactate dehydrogenase (LDH), tissue factor (TF), and prolidase were assayed in kidneys. Hyperoxia causes kidney damage induced by oxidative stress and apoptosis. Increased LPO, LDH, TF, and XO activities and decreased PON1 and prolidase activities contributed to kidney damage in hyperoxic mice. After hyperoxia, increases in the activities of antioxidant enzymes and NRF-2 level could not prevent this damage. ATRA attenuated damage via its oxidative stress-lowering effect. The decreased LDH and TF activities increased PON1 and prolidase activities, and normalized antioxidant statuses are indicators of the positive effects of ATRA. We recommend that ATRA can be used as a renoprotective agent against oxidative stress induced-kidney damage.
Collapse
Affiliation(s)
- Ozgecan Kayalar
- Department of Biology, Molecular Biology Division, Science Faculty, Istanbul University, Istanbul, Turkey
- School of Medicine, Koç University Research Center for Translational Medicine (KUTTAM), Koç University, Istanbul, Turkey
| | - Bertan Boran Bayrak
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Merve Yildirim
- Department of Biology, Molecular Biology Division, Science Faculty, Istanbul University, Istanbul, Turkey
| | - Refiye Yanardag
- Department of Chemistry, Faculty of Engineering, Istanbul University-Cerrahpaşa, Istanbul, Turkey
| | - Fusun Oztay
- Department of Biology, Molecular Biology Division, Science Faculty, Istanbul University, Istanbul, Turkey
| |
Collapse
|
37
|
Jiang M, Li Q, Xu B. Spotlight on ideal target antigens and resistance in antibody-drug conjugates: Strategies for competitive advancement. Drug Resist Updat 2024; 75:101086. [PMID: 38677200 DOI: 10.1016/j.drup.2024.101086] [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: 10/24/2023] [Revised: 04/09/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024]
Abstract
Antibody-drug conjugates (ADCs) represent a novel and promising approach in targeted therapy, uniting the specificity of antibodies that recognize specific antigens with payloads, all connected by the stable linker. These conjugates combine the best targeted and cytotoxic therapies, offering the killing effect of precisely targeting specific antigens and the potent cell-killing power of small molecule drugs. The targeted approach minimizes the off-target toxicities associated with the payloads and broadens the therapeutic window, enhancing the efficacy and safety profile of cancer treatments. Within precision oncology, ADCs have garnered significant attention as a cutting-edge research area and have been approved to treat a range of malignant tumors. Correspondingly, the issue of resistance to ADCs has gradually come to the fore. Any dysfunction in the steps leading to the ADCs' action within tumor cells can lead to the development of resistance. A deeper understanding of resistance mechanisms may be crucial for developing novel ADCs and exploring combination therapy strategies, which could further enhance the clinical efficacy of ADCs in cancer treatment. This review outlines the brief historical development and mechanism of ADCs and discusses the impact of their key components on the activity of ADCs. Furthermore, it provides a detailed account of the application of ADCs with various target antigens in cancer therapy, the categorization of potential resistance mechanisms, and the current state of combination therapies. Looking forward, breakthroughs in overcoming technical barriers, selecting differentiated target antigens, and enhancing resistance management and combination therapy strategies will broaden the therapeutic indications for ADCs. These progresses are anticipated to advance cancer treatment and yield benefits for patients.
Collapse
Affiliation(s)
- Mingxia Jiang
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiao Li
- Department of Medical Oncology, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Binghe Xu
- Department of Medical Oncology, State Key Laboratory of Mocelular Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
38
|
Chen Y, Jiao W, Wang Y, Liang Z, Wang L, Li D, Liang Y, Niu H. Microtubule interacting and trafficking domain containing 1 deficiency leads to poor survival via tissue factor-mediated coagulation in bladder cancer. J Thromb Haemost 2024; 22:1956-1972. [PMID: 38554936 DOI: 10.1016/j.jtha.2024.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/08/2024] [Accepted: 03/21/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Patients with cancer are at an increased risk of developing a hypercoagulative phenotype and venous thromboembolism. However, no clinical trial has yet confirmed that anticoagulant therapy improves cancer prognosis, and the mechanism underlying hypercoagulation in patients with bladder cancer is not well understood. OBJECTIVES We hypothesized that the prognostic genes affect tumor progression via tumor-mediated coagulation. METHODS We detected the most significant prognostic genes of bladder cancer with The Cancer Genome Atlas dataset and validated them in 2 Gene Expression Omnibus datasets and 1 ArrayExpress dataset. Immunohistochemical tests were performed on a cohort of 80 individuals to further examine the prognostic genes. For the most reliable prognostic gene, its influence on coagulation was evaluated with gene knockdown followed by next-generation sequencing and cellular and animal experiments. RESULTS Depletion of microtubule interacting and trafficking domain containing 1 (MITD1), a major prognostic gene of bladder cancer, significantly increased the tissue factor (TF) expression. MITD1 deficiency led to cytokinesis arrest, which, in turn, promoted the TF expression via unfolded protein response and c-Jun. The knockdown of IRE1, an essential kinase of unfolded protein response or the inactivation of c-Jun using c-Jun N-terminal kinase inhibitors weakened MITD1 deficiency- or dithiothreitol-induced TF upregulation. Cells lacking MITD1 promoted coagulation and metastasis in the experimental metastasis assay. CONCLUSION Our findings suggest the novel role of tumor prognostic genes upon the development of hypercoagulative phenotype and venous thromboembolism, thereby underlining the importance of anticoagulant therapy and shedding light on the therapeutic value of targeting MITD1 in bladder cancer.
Collapse
Affiliation(s)
- Yuanbin Chen
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wei Jiao
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yonghua Wang
- Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhijuan Liang
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Liping Wang
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Dan Li
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ye Liang
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Haitao Niu
- Qingdao Clinical Medical Research Center for Urinary System Disease, The Affiliated Hospital of Qingdao University, Qingdao, China; Department of Urology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
39
|
Peng C, Li J, Chen Y, Zhang HR, Li TX, Jiang YH, Yang XY, Zhao Y. PCSK9 aggravated carotid artery stenosis in ApoE -/- mice by promoting the expression of tissue factors in endothelial cells via the TLR4/NF-κB pathway. Biochem Pharmacol 2024; 225:116314. [PMID: 38797271 DOI: 10.1016/j.bcp.2024.116314] [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: 02/02/2024] [Revised: 05/09/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
Atherosclerosis, a chronic inflammatory disease, is the most relevant cause of carotid artery stenosis. Vascular endothelial cells (ECs) play a significant role in the development of atherosclerosis. In this chronic inflammatory environment, we aimed to investigate whether PCSK9 could mitigate atherosclerosis progression by reducing tissue factor expression in ECs via in vivo and in vitro assays. In vivo, we investigated the effect of PCSK9 inhibition on preventing atherosclerotic lesion formation in ApoE-/- mice fed a western diet. The results showed that inhibiting PCSK9 could significantly downregulate the protein expression of tissue factor (TF) in ECs to reduce the area of atherosclerotic plaques. In vitro, we incubated human umbilical vein endothelial cells (HUVECs) with lipopolysaccharide (LPS). We found that LPS-induced TF elevation was suppressed by a PCSK9 inhibitor at both the mRNA and protein levels and that the TLR4/NF-κB pathway was also suppressed by a PCSK9 inhibitor. With respect to plasma samples from patients with carotid artery stenosis, we also demonstrated that the expression of TF was positively correlated with that of PCSK9. Thus, in addition to regulating lipid metabolism, the regulation of endothelial cell TF expression through the TLR4/NF-κB pathway may be a potential mechanism of PCSK9 in promoting atherosclerotic carotid stenosis.
Collapse
Affiliation(s)
- Chao Peng
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Jian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yan Chen
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Heng-Rui Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Tian-Xing Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yu-Hang Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Xin-Yu Yang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| | - Yan Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, PR China.
| |
Collapse
|
40
|
Wan J, Dhrolia S, Kasthuri RR, Prokopenko Y, Ilich A, Saha P, Roest M, Wolberg AS, Key NS, Pawlinski R, Bendapudi PK, Mackman N, Grover SP. Plasma kallikrein supports FXII-independent thrombin generation in mouse whole blood. Blood Adv 2024; 8:3045-3057. [PMID: 38593231 PMCID: PMC11215197 DOI: 10.1182/bloodadvances.2024012613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024] Open
Abstract
ABSTRACT Plasma kallikrein (PKa) is an important activator of factor XII (FXII) of the contact pathway of coagulation. Several studies have shown that PKa also possesses procoagulant activity independent of FXII, likely through its ability to directly activate FIX. We evaluated the procoagulant activity of PKa using a mouse whole blood (WB) thrombin-generation (TG) assay. TG was measured in WB from PKa-deficient mice using contact pathway or extrinsic pathway triggers. PKa-deficient WB had significantly reduced contact pathway-initiated TG compared with that of wild-type controls and was comparable with that observed in FXII-deficient WB. PKa-deficient WB supported equivalent extrinsic pathway-initiated TG compared with wild-type controls. Consistent with the presence of FXII-independent functions of PKa, targeted blockade of PKa with either small molecule or antibody-based inhibitors significantly reduced contact pathway-initiated TG in FXII-deficient WB. Inhibition of activated FXII (FXIIa) using an antibody-based inhibitor significantly reduced TG in PKa-deficient WB, consistent with a PKa-independent function of FXIIa. Experiments using mice expressing low levels of tissue factor demonstrated that persistent TG present in PKa- and FXIIa-inhibited WB was driven primarily by endogenous tissue factor. Our work demonstrates that PKa contributes significantly to contact pathway-initiated TG in the complex milieu of mouse WB, and a component of this contribution occurs in an FXII-independent manner.
Collapse
Affiliation(s)
- Jun Wan
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Cyrus Tang Hematology Center, Collaborative Innovation Center of Hematology, Jiangsu Institute of Hematology, Soochow University, Suzhou, China
| | - Sophia Dhrolia
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rohan R. Kasthuri
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Yuriy Prokopenko
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Anton Ilich
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Prakash Saha
- School of Cardiovascular and Metabolic Medicine & Sciences, British Heart Foundation Centre of Research Excellence, King’s College London, London, United Kingdom
| | - Mark Roest
- Synapse Research Institute, Maastricht, The Netherlands
| | - Alisa S. Wolberg
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Department of Pathology and Laboratory Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Nigel S. Key
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Rafal Pawlinski
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Pavan K. Bendapudi
- Division of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Boston, MA
- Division of Hematology and Blood Transfusion Service, Massachusetts General Hospital, Boston, MA
- Harvard Medical School, Boston, MA
| | - Nigel Mackman
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Steven P. Grover
- UNC Blood Research Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC
- Division of Hematology, Department of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC
| |
Collapse
|
41
|
Attah A, Huffman D, Asawa P, Edlukudige Keshava V, Shah D. Asymptomatic COVID-19-Associated Acquired Hemophilia A and Disseminated Intravascular Coagulation From a Bypassing Agent. J Med Cases 2024; 15:106-109. [PMID: 38855293 PMCID: PMC11161180 DOI: 10.14740/jmc4199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/09/2024] [Indexed: 06/11/2024] Open
Abstract
Acquired hemophilia A (AHA) is a clotting disorder characterized by the presence of neutralizing antibodies that inhibit factor VIII, resulting in increased bleeding risk. Known etiologies include malignancy, autoimmune conditions, graft-vs-host disease, and more recently coronavirus disease 2019 (COVID-19) infection. In this case report, we describe an 86-year-old female who was found to have AHA incidentally during preoperative workup for meningioma resection. She was subsequently found to have COVID-19 infection which was the likely cause of her development of AHA. She was treated with factor eight inhibitor bypassing agent (FEIBA) and recombinant factor VII (rVII) for a small hematoma on her right arm along with prednisone and cyclophosphamide. She then developed disseminated intravascular coagulation (DIC) initially secondary to FEIBA and subsequently rFVII. DIC resolved after these factor concentrates were withheld. The aim of this case report was to emphasize the importance of monitoring partial thromboplastin time (PTT) in patients with COVID-19 and proceeding with AHA workup if indicated. It is also imperative to know and understand the potentially life-threatening, albeit rare, adverse effects of DIC associated with the administration of factor concentrates, especially in the elderly population and withholding these factor concentrates once DIC is suspected.
Collapse
Affiliation(s)
- Abraham Attah
- Allegheny Health Network, Internal Medicine Department, Pittsburgh, PA 15212, USA
| | - Deanna Huffman
- Louisiana State University Health Sciences Center New Orleans, New Orleans, LA 70112, USA
| | - Palash Asawa
- Allegheny Health Network, Internal Medicine Department, Pittsburgh, PA 15212, USA
| | - Vinay Edlukudige Keshava
- Division of Hematology/Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA
| | - Deep Shah
- Division of Hematology/Oncology, Allegheny Health Network Cancer Institute, Pittsburgh, PA 15212, USA
| |
Collapse
|
42
|
Du S, Pei X, Huang Y, Wang Y, Li Z, Niu X, Zhang W, Sun W. Hemin/G-quadruplex and AuNPs-MoS 2 based novel dual signal amplification strategy for ultrasensitively sandwich-type electrochemical thrombin aptasensor. Bioelectrochemistry 2024; 157:108635. [PMID: 38185025 DOI: 10.1016/j.bioelechem.2023.108635] [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: 10/06/2023] [Revised: 12/19/2023] [Accepted: 12/23/2023] [Indexed: 01/09/2024]
Abstract
In this work, a novel sandwich-type electrochemical aptasensor based on the dual signal amplification strategy of hemin/G-quadruplex and AuNPs-MoS2 was designed and constructed, which realized the highly sensitive and specific detection of thrombin (TB). In this aptasensor, the 15-mer TB-binding aptamer (TBA-1) modified with thiol group was immobilized on the surface of AuNPs modified glassy carbon electrode (AuNPs/GCE) as capturing elements. Another thiol-modified 29-mer TB-binding aptamer (TBA-2) sequence containing G-quadruplex structure for hemin immobilization was designed. The formed hemin/G-quadruplex/TBA-2 sequence was further combined to the AuNPs decorated flower-like molybdenum disulfide (AuNPs-MoS2) composite surface via Au-S bonds, acting the role of reporter probe. In presence of the target TB, the sandwich-type electrochemical aptamer detection system could be formed properly. With the assistance of the dual signal amplification of AuNPs-MoS2 and hemin/G-quadruplex toward H2O2 reduction, the sandwich-type electrochemical aptasensor was successfully constructed for sensitive detection of TB. The results demonstrate that the fabricated aptasensor displays a wide linear range of 1.0 × 10-6 ∼ 10.0 nM with a low detection limit of 0.34 fM. This proposed aptasensor shows potential application in the detection of TB content in real biological samples with high sensitivity, selectivity, and reliability.
Collapse
Affiliation(s)
- Shina Du
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Xiaoying Pei
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Yan Huang
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Yuebo Wang
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Zhongfang Li
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China
| | - Xueliang Niu
- School of Chemistry and Chemical Engineering, Shandong University of Technology, Zibo 255049, PR China.
| | - Weili Zhang
- College of Pharmacy, Key Laboratory of Biomedical Engineering and Technology in Universities of Shandong, Qilu Medical University, Zibo 255300, PR China.
| | - Wei Sun
- Key Laboratory of Laser Technology and Optoelectronic Functional Materials of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, PR China
| |
Collapse
|
43
|
Zhang W, Bhandari S, Ding Y, Luo J, Feng B, Jiang Y, Chen T, Wei J, Pan X, Weng H, Ding Z, Chen J, Chen X, Gong Y, Li H, Jin S, Hao Y. Polyunsaturated fatty acid-derived lipid mediator Resolvin D1 alleviates sepsis-induced disseminated intravascular coagulation via Caspase-1/Gasdermin D pyroptotic pathway. Clin Nutr 2024; 43:1372-1383. [PMID: 38678822 DOI: 10.1016/j.clnu.2024.04.020] [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: 03/07/2024] [Revised: 04/15/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND & AIMS Sepsis-induced disseminated intravascular coagulation (DIC) is characterised by abnormal blood clotting resulting from severe infection, contributing to organ dysfunction in sepsis. Resolvin D1 (RvD1) is an endogenous lipid mediator, synthesised from the omega-3 polyunsaturated fatty acid (PUFA) docosahexaenoic acid (DHA) through enzymatic processes involving 15-LOX and 5-LOX. RvD1 is recognised for its protective properties against various inflammatory conditions. This study aims to investigate its potential to modulate coagulation dysfunction in sepsis and to evaluate coagulation disorders in septic patients. METHODS Sepsis models were established by intraperitoneal injection LPS (20 mg/kg) or cecal ligation and puncture (CLP) followed by injection of RvD1 (10 μg/kg) or saline. The impact of RvD1 on coagulation dysfunction was assessed by clotting time and coagulation indicators such as TAT, D-dimer, PAI-1, and fibrinogen. The activity of the coagulation system in vivo was observed by evaluating dynamic microcirculation, platelets and thrombin in mice using intravital microscopy. The effect of RvD1 on pyroptosis was investigated by measuring NOD-like receptor protein 3 (NLRP3), Caspase-1, Caspase-11, and Gasdermin D (GSDMD) levels via western blot. Caspase-1 knockout mice, GSDMD knockout mice and bone marrow-derived macrophages (BMDMs) were used to elucidate the underlying mechanisms. Lastly, the concentration of RvD1 in plasma from septic patients was quantified to explore its relationship with coagulation and pyroptosis. RESULTS RvD1 significantly attenuated coagulation dysfunction in septic mice induced by LPS and CLP, and inhibited Caspase-1/GSDMD-dependent pyroptosis in septic mice and bone marrow-derived macrophages. In septic patients, the plasma concentrations of RvD1 was negatively correlated with both coagulation-related indicators and markers of GSDMD activation. CONCLUSION The results suggest that RvD1 can improve coagulation dysfunction in sepsis by regulating the Caspase-1/GSDMD pyroptotic pathway. Additionally, the concentration of RvD1 in septic patient plasma is related to prognosis and DIC development. RvD1 could be a potential biomarker and a promising therapeutic alternative in sepsis-induced DIC.
Collapse
Affiliation(s)
- Wenyan Zhang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China
| | - Suwas Bhandari
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China
| | - Yajun Ding
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China
| | - Jun Luo
- Department of Pharmacy, Taizhou Women and Children's Hospital of Wenzhou Medical University, Taizhou 317599, China
| | - Bo Feng
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China
| | - Yating Jiang
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China
| | - Ting Chen
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China
| | - Jinling Wei
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China
| | - Xiaodong Pan
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China; Department of Emergency Medicine, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China
| | - Haixu Weng
- Department of Critical Care Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Ruian 325207, China
| | - Zhangna Ding
- Department of Critical Care Medicine, The Third Affiliated Hospital of Wenzhou Medical University, Ruian 325207, China
| | - Jie Chen
- Department of Emergency, Third Affiliated Hospital, Wenzhou Medical University, Ruian 325207, China
| | - Xi Chen
- Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China
| | - Yuqiang Gong
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China.
| | - Hui Li
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China.
| | - Shengwei Jin
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China.
| | - Yu Hao
- Department of Anesthesia and Critical Care, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Pediatric Anesthesiology, Ministry of Education, Wenzhou Medical University, Wenzhou 325027, China; Key Laboratory of Anesthesiology of Zhejiang Province, Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
44
|
Yan C, Zhang Y, Jin L, Liu X, Zhu X, Li Q, Wang Y, Hu L, He X, Bao H, Zhu X, Wang Q, Liu WT. Medical ozone alleviates acute lung injury by enhancing phagocytosis targeting NETs via AMPK/SR-A1 axis. J Biomed Res 2024; 38:1-16. [PMID: 38807426 PMCID: PMC11629159 DOI: 10.7555/jbr.38.20240038] [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: 02/16/2024] [Revised: 05/06/2024] [Accepted: 05/10/2024] [Indexed: 05/30/2024] Open
Abstract
Acute lung injury (ALI) linked to sepsis has a high mortality rate, with limited treatment options available. In recent studies, medical ozone has shown promising results in alleviating inflammation and infection. Here, we aimed to evaluate the therapeutic potential of medical ozone in sepsis-induced ALI using a mouse model, measuring behavioral assessments, lung function, and blood flow. Western blot was used to quantify the levels of protein. In vitro, experiments on BMDM cells examine the impact of AMPK inhibitors and agonists on phagocytic activity. Results indicate that medical ozone can enhance the survival rate, ameliorate lung injury, and improve lung function and limb microcirculation in mice with ALI. Notably, it inhibits NETs formation, a crucial player in ALI development. Medical ozone also counteracts elevated TF, MMP-9, and IL-1β levels. In ALI mice, the effects of ozone are nullified and BMDMs exhibit impaired engulfment of NETs following Sr-a1 knockout. Under normal physiological conditions, the use of an AMPK antagonist produces similar effects to Sr-a1 knockout, significantly inhibiting the phagocytosis of NETs by BMDMs. On the contrary, AMPK agonists enhance this phagocytic process. In conclusion, medical ozone can alleviate sepsis-induced lung injury via the AMPK/SR-A1 pathway, thereby enhancing phagocytosis of NETs by macrophages.
Collapse
Affiliation(s)
- Chenxiao Yan
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, the Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yong Zhang
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Lai Jin
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xiaojie Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
- Department of Anesthesiology, the Affiliated Drum Tower Hospital, Medical School of Nanjing University, Nanjing, Jiangsu 210009, China
| | - Xuexian Zhu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Qifeng Li
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yu Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Liang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Xueming He
- Center for Clinical Research and Translational Medicine, Department of Anesthesiology, the Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, Jiangsu 222042, China
- Department of Anesthesiology, the Affiliated Lianyungang Oriental Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu 222042, China
| | - Hongguang Bao
- Department of Anesthesiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu 210006, China
| | - Xia Zhu
- Center for Clinical Research and Translational Medicine, Department of Anesthesiology, the Affiliated Lianyungang Oriental Hospital of Xuzhou Medical University, Lianyungang, Jiangsu 222042, China
- Department of Anesthesiology, the Affiliated Lianyungang Oriental Hospital of Kangda College of Nanjing Medical University, Lianyungang, Jiangsu 222042, China
| | - Qian Wang
- The Key Laboratory of Rare Metabolic Disease, Department of Biochemistry and Molecular Biology, the Key Laboratory of Human Functional Genomics of Jiangsu Province, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Wen-Tao Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
45
|
Hagan CE, Snyder AG, Headley M, Oberst A. Apoptotic cells promote circulating tumor cell survival and metastasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.21.595217. [PMID: 38826267 PMCID: PMC11142129 DOI: 10.1101/2024.05.21.595217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
During tumor progression and especially following cytotoxic therapy, cell death of both tumor and stromal cells is widespread. Despite clinical observations that high levels of apoptotic cells correlate with poorer patient outcomes, the physiological effects of dying cells on tumor progression remain incompletely understood. Here, we report that circulating apoptotic cells robustly enhance tumor cell metastasis to the lungs. Using intravenous metastasis models, we observed that the presence of apoptotic cells, but not cells dying by other mechanisms, supports circulating tumor cell (CTC) survival following arrest in the lung vasculature. Apoptotic cells promote CTC survival by recruiting platelets to the forming metastatic niche. Apoptotic cells externalize the phospholipid phosphatidylserine to the outer leaflet of the plasma membrane, which we found increased the activity of the coagulation initiator Tissue Factor, thereby triggering the formation of platelet clots that protect proximal CTCs. Inhibiting the ability of apoptotic cells to induce coagulation by knocking out Tissue Factor, blocking phosphatidylserine, or administering the anticoagulant heparin abrogated the pro-metastatic effect of apoptotic cells. This work demonstrates a previously unappreciated role for apoptotic cells in facilitating metastasis by establishing CTC-supportive emboli, and suggests points of intervention that may reduce the pro-metastatic effect of apoptotic cells. GRAPHICAL ABSTRACT
Collapse
|
46
|
Yuan X, Ou C, Li X, Zhuang Z, Chen Y. The skin circadian clock gene F3 as a potential marker for psoriasis severity and its bidirectional relationship with IL-17 signaling in keratinocytes. Int Immunopharmacol 2024; 132:111993. [PMID: 38565044 DOI: 10.1016/j.intimp.2024.111993] [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: 01/23/2024] [Revised: 03/21/2024] [Accepted: 03/30/2024] [Indexed: 04/04/2024]
Abstract
OBJECTIVE Psoriasis is an immune-mediated skin disease where the IL-17 signaling pathway plays a crucial role in its development. Chronic circadian rhythm disorder in psoriasis pathogenesis is gaining more attention. The relationship between IL and 17 signaling pathway and skin clock genes remains poorly understood. METHODS GSE121212 with psoriatic lesion and healthy controls was used as the exploration cohort for searching analysis. Datasets GSE54456, GSE13355, GSE14905, GSE117239, GSE51440, and GSE137218 were applied to validation analysis. Single-cell RNA sequencing (scRNA-seq) dataset GSE173706 was used to explore the F3 expression and related pathway activities in single-cell levels. Through intersecting with high-expression DEGs, F3 was selected as the signature skin circadian gene in psoriasis for further investigation. Functional analyses, including correlation analyses, prediction of transcription factors, protein-protein interaction, and single gene GSEA to explore the potential roles of F3. ssGSEA algorithm was performed to uncover the immune-related characteristics of psoriasis. We further explored F3 expression in the specific cell population in scRNA-seq dataset, besides this, AUCell analysis was performed to explore the pathway activities and the results were further compared between the specific cell cluster. Immunohistochemistry experiment, RT-qPCR was used to validate the location and expression of F3, small interfering RNA (siRNA) transfection experiment in HaCaT, and transcriptome sequencing analysis were applied to explore the potential function of F3. RESULTS F3 was significantly down-regulated in psoriasis and interacted with IL-17 signaling pathway. Low expression of F3 could upregulate the receptor of JAK-STAT signaling, thereby promoting keratinocyte inflammation. CONCLUSION Our research revealed a bidirectional link between the skin circadian gene F3 and the IL-17 signaling pathway in psoriasis, suggesting that F3 may interact with the IL-17 pathway by activating JAK-STAT within keratinocytes and inducing abnormal intracellular inflammation.
Collapse
Affiliation(s)
- Xiuqing Yuan
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Caixin Ou
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Xinhui Li
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Zhe Zhuang
- Dermatology Hospital of Southern Medical University, Guangzhou, China
| | - Yongfeng Chen
- Dermatology Hospital of Southern Medical University, Guangzhou, China.
| |
Collapse
|
47
|
Yuan MH, Zhong WX, Wang YL, Liu YS, Song JW, Guo YR, Zeng B, Guo YP, Guo L. Therapeutic effects and molecular mechanisms of natural products in thrombosis. Phytother Res 2024; 38:2128-2153. [PMID: 38400575 DOI: 10.1002/ptr.8151] [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: 08/03/2023] [Revised: 01/03/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024]
Abstract
Thrombotic disorders, such as myocardial infarction and stroke, are the leading cause of death in the global population and have become a health problem worldwide. Drug therapy is one of the main antithrombotic strategies, but antithrombotic drugs are not completely safe, especially the risk of bleeding at therapeutic doses. Recently, natural products have received widespread interest due to their significant efficacy and high safety, and an increasing number of studies have demonstrated their antithrombotic activity. In this review, articles from databases, such as Web of Science, PubMed, and China National Knowledge Infrastructure, were filtered and the relevant information was extracted according to predefined criteria. As a result, more than 100 natural products with significant antithrombotic activity were identified, including flavonoids, phenylpropanoids, quinones, terpenoids, steroids, and alkaloids. These compounds exert antithrombotic effects by inhibiting platelet activation, suppressing the coagulation cascade, and promoting fibrinolysis. In addition, several natural products also inhibit thrombosis by regulating miRNA expression, anti-inflammatory, and other pathways. This review systematically summarizes the natural products with antithrombotic activity, including their therapeutic effects, mechanisms, and clinical applications, aiming to provide a reference for the development of new antithrombotic drugs.
Collapse
Affiliation(s)
- Ming-Hao Yuan
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wen-Xiao Zhong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Lu Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Shi Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jia-Wen Song
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yu-Rou Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Bin Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yi-Ping Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Guo
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
48
|
Hisada Y. Dysregulated hemostasis in acute promyelocytic leukemia. Int J Hematol 2024; 119:526-531. [PMID: 38341391 DOI: 10.1007/s12185-024-03708-0] [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: 11/30/2023] [Revised: 12/26/2023] [Accepted: 01/04/2024] [Indexed: 02/12/2024]
Abstract
Acute promyelocytic leukemia (APL) is associated with a high incidence of early death, which occurs within 30 days of diagnosis. The major cause of early death in APL is severe bleeding, particularly intracranial bleeding. Although APL is known to be associated with activation of coagulation, hyperfibrinolysis, and thrombocytopenia, the precise mechanisms that cause bleeding have not yet been elucidated. I propose that a combination of four pathways may contribute to bleeding in APL: (1) tissue factor, (2) the urokinase plasminogen activator/urokinase plasminogen activator receptor, (3) the annexin A2/S100A100/tissue plasminogen activator, and (4) the podoplanin/C-type lectin-like receptor 2. A better understanding of these pathways will identify new biomarkers to determine which APL patients are at high risk of bleeding and allow the development of new treatments for APL-associated bleeding.
Collapse
Affiliation(s)
- Yohei Hisada
- UNC Blood Research Center, Division of Hematology, Department of Medicine, University of North Carolina at Chapel Hill, 116 Manning Drive, 8004 Mary Ellen Jones Bldg, Campus Box #7035, Chapel Hill, NC, 27599, USA.
| |
Collapse
|
49
|
Collins BEG, Kingsley M, Gordon BA, Zadow EK, Wundersitz DWT. Coagulation activity and thrombotic risk following high-volume endurance exercise in recreationally active cyclists. J Appl Physiol (1985) 2024; 136:1284-1290. [PMID: 38572538 DOI: 10.1152/japplphysiol.00824.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024] Open
Abstract
Despite the prognostic effect of physical activity, acute bouts of high-volume endurance exercise can induce cardiac stress and postexercise hypercoagulation associated with increased thrombotic risk. The aim of this study was to explore the effect of high-volume endurance exercise on coagulation and thrombotic activity in recreational cyclists. Thirty-four recreational cyclists completed 4.8 ± 0.3 h of cycling at 45 ± 5% of maximal power output on a bicycle ergometer. Intravenous blood samples were collected preexercise, immediately postexercise, 24 and 48 h postexercise, and analyzed for brain natriuretic peptide (BNP), cardiac troponin (cTn), C-reactive protein (CRP), D-dimer, thrombin-antithrombin (TAT) complex, tissue factor (TF), tissue factor pathway inhibitor (TFPI), and TF-to-TFPI ratio (TF:TFPI). An increase in cTn was observed postexercise (P < 0.001). CRP concentrations were increased at 24 and 48 h postexercise compared with preexercise concentrations (P ≤ 0.001). TF was elevated at 24 h postexercise (P < 0.031) and TFPI was higher immediately postexercise (P < 0.044) compared with all other time points. TF:TFPI was increased at 24 and 48 h postexercise compared with preexercise (P < 0.025). TAT complex was reduced at 48 h postexercise compared with preexercise (P = 0.015), D-dimer was higher immediately postexercise compared with all other time points (P ≤ 0.013). No significant differences were observed in BNP (P > 0.05). High-volume endurance cycling induced markers of cardiac stress among recreational cyclists. However, plasma coagulation and fibrinolytic activity suggest no increase in thrombotic risk after high-volume endurance exercise.NEW & NOTEWORTHY In this study, a high-volume endurance exercise protocol induced markers of cardiac stress and altered plasma coagulation and fibrinolytic activity for up to 48 h in recreationally active cyclists. However, analysis of coagulation biomarkers indicates no increase in thrombotic risk when appropriate hydration and rest protocols are implemented.
Collapse
Affiliation(s)
- Blake E G Collins
- Holsworth Research Initiative, La Trobe Rural Health School, La Trobe University, Bendigo, Victoria, Australia
| | - Michael Kingsley
- Holsworth Research Initiative, La Trobe Rural Health School, La Trobe University, Bendigo, Victoria, Australia
- Department of Exercise Sciences, Faculty of Science, University of Auckland, Auckland, New Zealand
| | - Brett A Gordon
- Holsworth Research Initiative, La Trobe Rural Health School, La Trobe University, Bendigo, Victoria, Australia
| | - Emma K Zadow
- School of Health Science, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Daniel W T Wundersitz
- Holsworth Research Initiative, La Trobe Rural Health School, La Trobe University, Bendigo, Victoria, Australia
| |
Collapse
|
50
|
Tatsumi K. The pathogenesis of cancer-associated thrombosis. Int J Hematol 2024; 119:495-504. [PMID: 38421488 DOI: 10.1007/s12185-024-03735-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 02/13/2024] [Accepted: 02/18/2024] [Indexed: 03/02/2024]
Abstract
Patients with cancer have a higher risk of venous thromboembolism (VTE), including deep vein thrombosis (DVT) and pulmonary embolism (PE), compared to the general population. Cancer-associated thrombosis (CAT) is a thrombotic event that occurs as a complication of cancer or cancer therapy. Major factors determining VTE risk in cancer patients include not only treatment history and patient characteristics, but also cancer type and site. Cancer types can be broadly divided into three groups based on VTE risk: high risk (pancreatic, ovarian, brain, stomach, gynecologic, and hematologic), intermediate risk (colon and lung), and low risk (breast and prostate). This implies that the mechanism of VTE differs between cancer types and that specific VTE pathways may exist for different cancer types. This review summarizes the specific pathways that contribute to VTE in cancer patients, with a particular focus on leukocytosis, neutrophil extracellular traps (NETs), tissue factor (TF), thrombocytosis, podoplanin (PDPN), plasminogen activator inhibitor-1 (PAI-1), the intrinsic coagulation pathway, and von Willebrand factor (VWF).
Collapse
Affiliation(s)
- Kohei Tatsumi
- Advanced Medical Science of Thrombosis and Hemostasis, Nara Medical University, 840 Shijo-Cho, Kashihara, Nara, 634-8521, Japan.
| |
Collapse
|