1
|
Li Y, Chen K, Wang QF. Immunological face of megakaryocytes. Front Med 2024; 18:988-1001. [PMID: 39542989 DOI: 10.1007/s11684-024-1087-1] [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: 12/22/2023] [Accepted: 05/17/2024] [Indexed: 11/17/2024]
Abstract
Megakaryocytes (MKs), which are traditionally known for their role in platelet production, are now emerging as unique immune cells with diverse capabilities. They express immune receptors, participate in pathogen recognition and response, phagocytose pathogens, contribute to antigen presentation, and interact with various immune cell types. When encountering inflammatory challenges, MKs exhibit intricate immune functions that can either promote or inhibit inflammation. These responses are mediated through mechanisms, such as the secretion of either anti-inflammatory or pro-inflammatory cytokines and release of immunomodulatory platelets according to specific conditions. This intricate array of responses necessitates a detailed exploration to determine whether the immune functions of MKs are carried out by the entire MK population or by a specific subpopulation. Breakthroughs in single-cell RNA sequencing have uncovered a unique "immune MK" subpopulation, revealing its distinct characteristics and immunoregulatory functions. This review provides latest insights into MKs' immune attributes and their roles in physiological and pathological contexts and emphasizes the discovery and functions of "immune MKs".
Collapse
Affiliation(s)
- Yueying Li
- China National Center for Bioinformation, Beijing, 100101, China.
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100101, China.
| | - Kunying Chen
- China National Center for Bioinformation, Beijing, 100101, China
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Qian-Fei Wang
- China National Center for Bioinformation, Beijing, 100101, China.
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
2
|
Kim JG, Koo BS, Lee JH, Yoon BY. Anemia as an indicator of a higher retention rate for tocilizumab versus tumor necrosis factor inhibitors in patients with rheumatoid arthritis from a Korean multi-center registry. JOURNAL OF RHEUMATIC DISEASES 2024; 31:212-222. [PMID: 39355549 PMCID: PMC11439637 DOI: 10.4078/jrd.2024.0026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 10/03/2024]
Abstract
Objective To examine whether simple laboratory tests can guide selection between tocilizumab (TCZ) and tumor necrosis factor inhibitors (TNFi) in biologic-naive patients with rheumatoid arthritis (RA), by investigating their influence on drug retention. Methods Data of RA patients prescribed TCZ or TNFi as the initial biologics from March 2013 to December 2021 were obtained from the KOrean College of Rheumatology BIOlogics and Targeted Therapy (KOBIO) registry. Propensity score matching was performed to adjust for baseline confounding factors. Hazards of drug discontinuation for TCZ were calculated compared to those for TNFi. Interaction analyses with a Bonferroni-corrected p-value threshold were conducted to determine whether the hemoglobin level, C-reactive protein level, erythrocyte sedimentation rate, and platelet count affected the hazards of drug discontinuation. Results Overall, 893 patients were analyzed, of whom 315 and 578 were treated with TCZ and TNFi, respectively. The hazards of drug discontinuation in all patients were lower for TCZ than for TNFi (hazard ratio [HR] 0.53, 95% confidence interval [CI] 0.44~0.66). Notably, only the presence of anemia indicated a significant interaction (p for interaction=0.010); the HRs for drug discontinuation were 0.41 (95% CI 0.30~0.55) and 0.70 (95% CI 0.53~0.92) in the anemic and non-anemic groups, respectively. In the anemic subgroup, biologics were discontinued because of a lack of efficacy in 35.0% of TNFi initiators and 7.4% of TCZ initiators.Conclusion The drug discontinuation rate in biologic-naïve patients with RA was significantly lower for TCZ than for TNFi, particularly in those with anemia.
Collapse
Affiliation(s)
- Jung Gon Kim
- Division of Rheumatology, Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Bon San Koo
- Division of Rheumatology, Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Joo-Hyun Lee
- Division of Rheumatology, Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| | - Bo Young Yoon
- Division of Rheumatology, Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Korea
| |
Collapse
|
3
|
Gao YH, Liu YT, Zhang MY, Li SY, Fajgenbaum DC, Zhang L, Li J. Idiopathic multicentric Castleman disease (iMCD)-idiopathic plasmacytic lymphadenopathy: A distinct subtype of iMCD-not otherwise specified with different clinical features and better survival. Br J Haematol 2024; 204:1830-1837. [PMID: 38356434 PMCID: PMC11090736 DOI: 10.1111/bjh.19334] [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: 12/24/2023] [Revised: 01/21/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Idiopathic multicentric Castleman disease (iMCD) is subclassified into iMCD-thrombocytopenia, anasarca, reticulin fibrosis, renal dysfunction, organomegaly (TAFRO) and iMCD-not otherwise specified (NOS) according to the Castleman Disease Collaborative Network (CDCN) consensus criteria. With a deeper understanding of iMCD, a group of patients with iMCD-NOS characterised by polyclonal hypergammaglobulinaemia, plasmacytic/mixed-type lymph node histopathology and thrombocytosis has attracted attention. This group of patients has been previously described as having idiopathic plasmacytic lymphadenopathy (IPL). Whether these patients should be excluded from the current classification system lacks sufficient evidence. This retrospective analysis of 228 patients with iMCD-NOS identified 103 (45.2%) patients with iMCD-IPL. The clinical features and outcomes of patients with iMCD-IPL and iMCD-NOS without IPL were compared. Patients with iMCD-IPL showed a significantly higher inflammatory state but longer overall survival. No significant difference in overall survival was observed between severe and non-severe patients in the iMCD-IPL group according to the CDCN severity classification. Compared with lymphoma-like treatments, multiple myeloma-like and IL-6-blocking treatment approaches in the iMCD-IPL group resulted in significantly higher response rates and longer time to the next treatment. These findings highlight the particularities of iMCD-IPL and suggest that it should be considered a new subtype of iMCD-NOS.
Collapse
Affiliation(s)
- Yu-han Gao
- Department of Haematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan-ting Liu
- Department of Haematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Miao-yan Zhang
- Department of Haematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Si-yuan Li
- Department of Haematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - David C. Fajgenbaum
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Lu Zhang
- Department of Haematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jian Li
- Department of Haematology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Rubenstein AI, Pierson SK, Shyamsundar S, Sarmiento Bustamante M, Gonzalez MV, Milller ID, Brandstadter JD, Mumau MD, Fajgenbaum DC. Immune-mediated thrombocytopenia and IL-6-mediated thrombocytosis observed in idiopathic multicentric Castleman disease. Br J Haematol 2024; 204:921-930. [PMID: 38168727 PMCID: PMC10998476 DOI: 10.1111/bjh.19279] [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: 10/02/2023] [Revised: 11/30/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Idiopathic multicentric Castleman disease (iMCD) is a rare haematological disorder characterized by generalized lymphadenopathy with atypical histopathological features and systemic inflammation caused by a cytokine storm involving interleukin-6 (IL-6). Three clinical subtypes are recognized: thrombocytopenia, anasarca, fever, renal dysfunction, organomegaly (iMCD-TAFRO); idiopathic plasmacytic lymphadenopathy (iMCD-IPL), involving thrombocytosis and hypergammaglobulinaemia; and iMCD-not otherwise specified (iMCD-NOS), which includes patients who do not meet criteria for the other subtypes. Disease pathogenesis is poorly understood, with potential involvement of infectious, clonal and/or autoimmune mechanisms. To better characterize iMCD clinicopathology and gain mechanistic insights into iMCD, we analysed complete blood counts, other clinical laboratory values and blood smear morphology among 63 iMCD patients grouped by clinical subtype. Patients with iMCD-TAFRO had large platelets, clinical severity associated with lower platelet counts and transfusion-resistant thrombocytopenia, similar to what is observed with immune-mediated destruction of platelets in immune thrombocytopenic purpura. Conversely, elevated platelet counts in iMCD-IPL were associated with elevated IL-6 and declined following anti-IL-6 therapy. Our data suggest that autoimmune mechanisms contribute to the thrombocytopenia in at least a portion of iMCD-TAFRO patients whereas IL-6 drives thrombocytosis in iMCD-IPL, and these mechanisms likely contribute to disease pathogenesis.
Collapse
Affiliation(s)
- Ayelet I. Rubenstein
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sheila K. Pierson
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Saishravan Shyamsundar
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mateo Sarmiento Bustamante
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michael V. Gonzalez
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ira D. Milller
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Joshua D. Brandstadter
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Melanie D. Mumau
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David C. Fajgenbaum
- Center for Cytokine Storm Treatment & Laboratory, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Chun Y, Jo JH, Park JW. Effects of physical activity levels on characteristic pain in temporomandibular dysfunctions: a cross-sectional study. Head Face Med 2024; 20:6. [PMID: 38238755 PMCID: PMC10795209 DOI: 10.1186/s13005-024-00407-3] [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: 07/18/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Physical activity is known to influence the symptoms of a variety of pain disorders including fibromyalgia and osteoarthritis although the underlying mechanism is not fully understood. In spite of the high prevalence of temporomandibular disorders (TMD), no previous study has objectively evaluated the relationship between TMD and general physical activity. This study aims to investigate the influence of physical activity on pain and disability from TMD, considering various confounders including sleep, systemic inflammation, psychosocial disturbances, and widespread pain. METHODS This observational cross-sectional study is based on consecutive samples of 100 TMD patients (22 with high pain disability and 78 with low pain disability level). Physical activity levels were assessed with actigraph. Level of pain and disability were evaluated using the Graded Chronic Pain Scale. Hematologic examinations including inflammatory biomarkers were assessed and comorbidities were investigated with validated questionnaires. Differences were analyzed according to disability level. RESULTS Patients with high disability level spent significantly more time doing both moderate (p = 0.033) and vigorous (p = 0.039) level physical activity. Light physical activity, on the other hand, was associated with low disability but the difference did not reach statistical significance. Time spent in light physical activity was significantly associated with high levels of pain and disability (p = 0.026, β = -0.001) and time spent in vigorous physical activity had significant predictive power (cutoff value 2.5 min per week, AUC 0.643, p = 0.041). Scores of the Jaw Function Limitation Score-20 (p = 0.001), present McGill Pain Score (p = 0.010), and number of people potentially diagnosed with fibromyalgia (p = 0.033) were significantly higher in the high disability group. CONCLUSIONS Moderate or vigorous physical activity is associated with worse TMD symptoms while light physical activity may be beneficial. Further research related to the amount and frequency of physical activity is necessary to establish clinical guidelines for TMD. TRIAL REGISTRATION clinical trial registration of the Clinical Research Information Service of Republic of Korea (number KCT0007107).
Collapse
Affiliation(s)
- Youngwoo Chun
- Department of Oral Medicine, Seoul National University Dental Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Jung Hwan Jo
- Department of Oral Medicine, Seoul National University Dental Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea
| | - Ji Woon Park
- Department of Oral Medicine, Seoul National University Dental Hospital, 101, Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
- Department of Oral Medicine and Oral Diagnosis, School of Dentistry, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
- Dental Research Institute, Seoul National University, 101 Daehak-ro, Jongno-gu, Seoul, 03080, South Korea.
| |
Collapse
|
6
|
Khan S. Interleukin 6 Antagonists in Severe COVID-19 Disease: Cardiovascular and Respiratory Outcomes. Protein Pept Lett 2024; 31:178-191. [PMID: 38375841 DOI: 10.2174/0109298665266730240118054023] [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: 07/18/2023] [Revised: 12/17/2023] [Accepted: 12/27/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND Inhibitors of interleukin 6 [IL-6] have been utilized to treat severe COVID-19 disease. Their immunosuppressive or immunomodulating impact may be beneficial in COVID-19. OBJECTIVES To discuss the role of IL-6 inhibitors and assess various trials conducted to evaluate the efficacy of IL-6 inhibitors in COVID-19 disease. SUMMARY Two of the most common causes of mortality in COVID-19-infected critically ill individuals are acute respiratory distress syndrome (ARDS) and multiorgan failure. Increased levels of inflammatory cytokines suggest that a cytokine storm, also known as cytokine release syndrome (CRS), is involved in the etiology of COVID-19. Most tissue damage, sepsis, and pulmonary and cardiovascular problems are caused mainly by the host defense system. Therefore, regulating this inflammatory cascade using immunomodulators is a prudent strategy. Although corticosteroids, as immunomodulators, are routinely used in COVID-19 management, interleukin (IL) inhibitors, especially IL-6 inhibitors, are also tested in many trials. Many studies have demonstrated that IL-6 inhibitors improve disease outcomes and decrease mortality, whereas others have shown that they are ineffective. In this paper, we briefly examined the role of IL-6 in COVID-19 pathogenesis and trials that support or refute the use of IL-6 inhibitors in treating COVID-19 disease. RESULTS Though mixed results are coming from trials regarding the adjuvant use of IL-6 inhibitors and standard anti-viral therapy with dexamethasone, a consensus favors using IL-6 inhibitors in severely ill COVID-19 patients regardless of the outcome.
Collapse
Affiliation(s)
- Shahzad Khan
- Department of Biomedical Sciences, College of Clinical Pharmacy, King Faisal University, Al Hofuf, Saudi Arabia
| |
Collapse
|
7
|
Menghoum N, Beauloye C, Lejeune S, Badii MC, Gruson D, van Dievoet MA, Pasquet A, Vancraeynest D, Gerber B, Bertrand L, Horman S, Pouleur AC. Mean platelet volume: a prognostic marker in heart failure with preserved ejection fraction. Platelets 2023; 34:2188965. [PMID: 37157842 DOI: 10.1080/09537104.2023.2188965] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Heart failure (HF) with preserved ejection fraction (HFpEF) is associated with high burden of comorbidities known to increase the mean platelet volume (MPV). This parameter has been associated with morbidity and mortality in HF. However, the role of platelets and the prognostic relevance of MPV in HFpEF remain largely unexplored. We aimed to evaluate the clinical usefulness of MPV as a prognostic marker in HFpEF. We prospectively enrolled 228 patients with HFpEF (79 ± 9 years; 66% females) and 38 controls of similar age and gender (78 ± 5 years; 63% females). All subjects underwent two-dimensional echocardiography and MPV measurements. Patients were followed-up for a primary end point of all-cause mortality or first HF hospitalization. The prognostic impact of MPV was determined using Cox proportional hazard models. Mean MPV was significantly higher in HFpEF patients compared with controls (MPV: 10.7 ± 1.1fL vs. 10.1 ± 1.1fL, p = .005). HFpEF patients (n = 56) with MPV >75th percentile (11.3 fL) displayed more commonly a history of ischemic cardiomyopathy. Over a median follow-up of 26 months, 136 HFpEF patients reached the composite endpoint. MPV >75th percentile was a significant predictor of the primary endpoint (HR: 1.70 [1.08; 2.67], p = .023) adjusted for NYHA class, chronic obstructive pulmonary disease, loop diuretics, renal function, and hemoglobin. We demonstrated that MPV was significantly higher in HFpEF patients compared with controls of similar age and gender. Elevated MPV was a strong and independent predictor of poor outcome in HFpEF patients and may be relevant for clinical use.
Collapse
Affiliation(s)
- Nassiba Menghoum
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Christophe Beauloye
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Sibille Lejeune
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Maria Chiara Badii
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Damien Gruson
- Clinical Biology Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | | | - Agnès Pasquet
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - David Vancraeynest
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Bernhard Gerber
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Luc Bertrand
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Sandrine Horman
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| | - Anne-Catherine Pouleur
- Cardiovascular Department, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Pôle de Recherche Cardiovasculaire (CARD), Institut de Recherche Expérimentale et Clinique (IREC), Université Catholique de Louvainr (UCLouvain), Brussels, Belgium
| |
Collapse
|
8
|
Aguilar-Cazares D, Chavez-Dominguez R, Marroquin-Muciño M, Perez-Medina M, Benito-Lopez JJ, Camarena A, Rumbo-Nava U, Lopez-Gonzalez JS. The systemic-level repercussions of cancer-associated inflammation mediators produced in the tumor microenvironment. Front Endocrinol (Lausanne) 2022; 13:929572. [PMID: 36072935 PMCID: PMC9441602 DOI: 10.3389/fendo.2022.929572] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 08/01/2022] [Indexed: 12/15/2022] Open
Abstract
The tumor microenvironment is a dynamic, complex, and redundant network of interactions between tumor, immune, and stromal cells. In this intricate environment, cells communicate through membrane-membrane, ligand-receptor, exosome, soluble factors, and transporter interactions that govern cell fate. These interactions activate the diverse and superfluous signaling pathways involved in tumor promotion and progression and induce subtle changes in the functional activity of infiltrating immune cells. The immune response participates as a selective pressure in tumor development. In the early stages of tumor development, the immune response exerts anti-tumor activity, whereas during the advanced stages, the tumor establishes mechanisms to evade the immune response, eliciting a chronic inflammation process that shows a pro-tumor effect. The deregulated inflammatory state, in addition to acting locally, also triggers systemic inflammation that has repercussions in various organs and tissues that are distant from the tumor site, causing the emergence of various symptoms designated as paraneoplastic syndromes, which compromise the response to treatment, quality of life, and survival of cancer patients. Considering the tumor-host relationship as an integral and dynamic biological system, the chronic inflammation generated by the tumor is a communication mechanism among tissues and organs that is primarily orchestrated through different signals, such as cytokines, chemokines, growth factors, and exosomes, to provide the tumor with energetic components that allow it to continue proliferating. In this review, we aim to provide a succinct overview of the involvement of cancer-related inflammation at the local and systemic level throughout tumor development and the emergence of some paraneoplastic syndromes and their main clinical manifestations. In addition, the involvement of these signals throughout tumor development will be discussed based on the physiological/biological activities of innate and adaptive immune cells. These cellular interactions require a metabolic reprogramming program for the full activation of the various cells; thus, these requirements and the by-products released into the microenvironment will be considered. In addition, the systemic impact of cancer-related proinflammatory cytokines on the liver-as a critical organ that produces the leading inflammatory markers described to date-will be summarized. Finally, the contribution of cancer-related inflammation to the development of two paraneoplastic syndromes, myelopoiesis and cachexia, will be discussed.
Collapse
Affiliation(s)
- Dolores Aguilar-Cazares
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Rodolfo Chavez-Dominguez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Mario Marroquin-Muciño
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Mario Perez-Medina
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Laboratorio de Quimioterapia Experimental, Departamento de Bioquimica, Escuela Nacional de Ciencias Biologicas, Instituto Politecnico Nacional, Mexico City, Mexico
| | - Jesus J. Benito-Lopez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
- Posgrado en Ciencias Biologicas, Universidad Nacional Autonoma de Mexico, Mexico City, Mexico
| | - Angel Camarena
- Laboratorio de Human Leukocyte Antigen (HLA), Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Uriel Rumbo-Nava
- Clinica de Neumo-Oncologia, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| | - Jose S. Lopez-Gonzalez
- Laboratorio de Investigacion en Cancer Pulmonar, Departamento de Enfermedades Cronico-Degenerativas, Instituto Nacional de Enfermedades Respiratorias “Ismael Cosio Villegas”, Mexico City, Mexico
| |
Collapse
|
9
|
von Stemann JH, Pedersen OBV, Hjalgrim H, Erikstrup C, Ullum H, Dowsett J, Thørner LW, Larsen MAH, Sørensen E, Hansen MB, Ostrowski SR. IL-6 Autoantibodies Predict Lower Platelet Counts and Altered Plasma Cytokine Profiles in Healthy Blood Donors: Results From the Danish Blood Donor Study. Front Med (Lausanne) 2022; 9:914262. [PMID: 35814772 PMCID: PMC9263719 DOI: 10.3389/fmed.2022.914262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/02/2022] [Indexed: 12/03/2022] Open
Abstract
Cytokine-specific autoantibodies (c-aAb) represent a novel type of immune dysfunction. Though they have been detected in both patient cohorts and healthy individuals, and have immunomodulatory properties, the full extent of their influence remains unknown. Based on the critical role of several cytokines in thrombopoiesis, we investigated if there is an association between c-aAb and platelet variables in healthy individuals, with a specific focus on c-aAb against a known thrombopoietic cytokine, IL-6. Using platelet count and mean platelet volume in 3,569 healthy participants of the Danish Blood Donor Study as dependent variables, we performed a series of multivariate regression analyses using five cytokine autoantibodies, including IL-6 c-aAb, as independent variables. In men, high titers of IL-6 c-aAb were negatively associated with platelet counts (β = −24 *109/l (95% confidence interval −43 to −6), p = 0.008) and positively associated with mean platelet volume (β = 0.4 fL (95% confidence interval 0.0–0.7) p = 0.043). These associations were exacerbated when adjusting for undetectable C-reactive protein levels, which we used as a proxy for c-aAb mediated IL-6 inhibition in vivo. Furthermore, in a smaller subgroup, individuals with high vs. low titer IL-6 c-aAb had different profiles of plasma IL-6, IL-10, TNFα and TPO, further suggesting a functional inhibition of IL-6 by high titers of circulating IL-6 c-aAb. We therefore speculate that in addition to their immunomodulatory potential IL-6 c-aAb may interfere with thrombopoiesis – directly or indirectly – under normal physiological conditions. This study is the first to suggest an influence of c-aAb on platelets in healthy individuals, beyond their apparent effects on immune competence.
Collapse
Affiliation(s)
- Jakob Hjorth von Stemann
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- *Correspondence: Jakob Hjorth von Stemann
| | - Ole Birger Vesterager Pedersen
- Department of Clinical Immunology, Zealand University Hospital, Køge, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Henrik Hjalgrim
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Epidemiology Research, Statens Serum Institut, Copenhagen, Denmark
- Centre for Cancer Research, Danish Cancer Society, Copenhagen, Denmark
- Department of Haematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Christian Erikstrup
- Department of Clinical Immunology, Aarhus University Hospital, Aarhus, Denmark
| | | | - Joseph Dowsett
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Lise Wegner Thørner
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Margit Anita Hørup Larsen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Erik Sørensen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Morten Bagge Hansen
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
10
|
Exploring Endothelial Colony-Forming Cells to Better Understand the Pathophysiology of Disease: An Updated Review. Stem Cells Int 2022; 2022:4460041. [PMID: 35615696 PMCID: PMC9126670 DOI: 10.1155/2022/4460041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 04/20/2022] [Accepted: 05/09/2022] [Indexed: 12/12/2022] Open
Abstract
Endothelial cell (EC) dysfunction has been implicated in a variety of pathological conditions. The collection of ECs from patients is typically conducted postmortem or through invasive procedures, such as surgery and interventional procedures, hampering efforts to clarify the role of ECs in disease onset and progression. In contrast, endothelial colony-forming cells (ECFCs), also termed late endothelial progenitor cells, late outgrowth endothelial cells, blood outgrowth endothelial cells, or endothelial outgrowth cells, are obtained in a minimally invasive manner, namely, by the culture of human peripheral blood mononuclear cells in endothelial growth medium. ECFCs resemble mature ECs phenotypically, genetically, and functionally, making them excellent surrogates for ECs. Numerous studies have been performed that examined ECFC function in conditions such as coronary artery disease, diabetes mellitus, hereditary hemorrhagic telangiectasia, congenital bicuspid aortic valve disease, pulmonary arterial hypertension, venous thromboembolic disease, and von Willebrand disease. Here, we provide an updated review of studies using ECFCs that were performed to better understand the pathophysiology of disease. We also discuss the potential of ECFCs as disease biomarkers and the standardized methods to culture, quantify, and evaluate ECFCs and suggest the future direction of research in this field.
Collapse
|
11
|
La Manna MP, Orlando V, Badami GD, Tamburini B, Azgomi MS, Presti EL, Del Nonno F, Petrone L, Belmonte B, Falasca L, Carlo PD, Dieli F, Goletti D, Caccamo N. Platelets accumulate in lung lesions of tuberculosis patients and inhibit T-cell responses and Mycobacterium tuberculosis replication in macrophages. Eur J Immunol 2022; 52:784-799. [PMID: 35338775 PMCID: PMC9325462 DOI: 10.1002/eji.202149549] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 12/24/2021] [Accepted: 03/23/2022] [Indexed: 12/05/2022]
Abstract
Platelets regulate human inflammatory responses that lead to disease. However, the role of platelets in tuberculosis (TB) pathogenesis is still unclear. Here, we show that patients with active TB have a high number of platelets in peripheral blood and a low number of lymphocytes leading to a high platelets to lymphocytes ratio (PL ratio). Moreover, the serum concentration of different mediators promoting platelet differentiation or associated with platelet activation is increased in active TB. Immunohistochemistry analysis shows that platelets localise around the lung granuloma lesions in close contact with T lymphocytes and macrophages. Transcriptomic analysis of caseous tissue of human pulmonary TB granulomas, followed by Gene Ontology analysis, shows that 53 platelet activation‐associated genes are highly expressed compared to the normal lung tissue. In vitro activated platelets (or their supernatants) inhibit BCG‐induced T‐ lymphocyte proliferation and IFN‐γ production. Likewise, platelets inhibit the growth of intracellular macrophages of Mycobacterium (M.) tuberculosis. Soluble factors released by activated platelets mediate both immunological and M. tuberculosis replication activities. Furthermore, proteomic and neutralisation studies (by mAbs) identify TGF‐β and PF4 as the factors responsible for inhibiting T‐cell response and enhancing the mycobactericidal activity of macrophages, respectively. Altogether these results highlight the importance of platelets in TB pathogenesis.
Collapse
Affiliation(s)
- Marco P La Manna
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Valentina Orlando
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Giusto D Badami
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Bartolo Tamburini
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Elena Lo Presti
- Institute for Biomedical Research and Innovation, National Research Council, Palermo, Italy
| | - Franca Del Nonno
- Pathology Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Linda Petrone
- Translational research Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Science, Human Pathology Section, University of Palermo School of Medicine, Palermo, Italy
| | - Laura Falasca
- Pathology Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Paola Di Carlo
- Department of Sciences for Health Promotion and Mother-Child Care "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| | - Delia Goletti
- Translational research Unit, National Institute for Infectious Diseases L. Spallanzani-IRCCS, Rome, Italy
| | - Nadia Caccamo
- Central Laboratory of Advanced Diagnosis and Biomedical Research (CLADIBIOR).,Department of Biomedicine, Neurosciences and Advanced Diagnostic (Bi.N.D.), University of Palermo, Palermo, 90127, Italy
| |
Collapse
|
12
|
Swan D, Thachil J. Management of haemostatic complications of chimaeric antigen receptor T-cell therapy. Br J Haematol 2022; 197:250-259. [PMID: 35146749 DOI: 10.1111/bjh.18045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 11/28/2022]
Affiliation(s)
- Dawn Swan
- Department of Haematology, St James' Hospital, Dublin, Republic of Ireland
| | - Jecko Thachil
- Department of Haematology, Central Manchester University Hospitals NHS Foundation Trust, Manchester, UK
| |
Collapse
|
13
|
Khatib-Massalha E, Méndez-Ferrer S. Megakaryocyte Diversity in Ontogeny, Functions and Cell-Cell Interactions. Front Oncol 2022; 12:840044. [PMID: 35186768 PMCID: PMC8854253 DOI: 10.3389/fonc.2022.840044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 01/17/2022] [Indexed: 12/12/2022] Open
Abstract
Hematopoietic stem cells (HSCs) rely on local interactions in the bone marrow (BM) microenvironment with stromal cells and other hematopoietic cells that facilitate their survival and proliferation, and also regulate their functions. HSCs and multipotent progenitor cells differentiate into lineage-specific progenitors that generate all blood and immune cells. Megakaryocytes (Mks) are hematopoietic cells responsible for producing blood platelets, which are essential for normal hemostasis and blood coagulation. Although the most prominent function of Mks is platelet production (thrombopoiesis), other increasingly recognized functions include HSC maintenance and host immune response. However, whether and how these diverse programs are executed by different Mk subpopulations remains poorly understood. This Perspective summarizes our current understanding of diversity in ontogeny, functions and cell-cell interactions. Cumulative evidence suggests that BM microenvironment dysfunction, partly caused by mutated Mks, can induce or alter the progression of a variety of hematologic malignancies, including myeloproliferative neoplasms (MPNs) and other disorders associated with tissue scarring (fibrosis). Therefore, as an example of the heterogeneous functions of Mks in malignant hematopoiesis, we will discuss the role of Mks in the onset and progression of BM fibrosis. In this regard, abnormal interactions between of Mks and other immune cells might directly contribute to fibrotic diseases. Overall, further understanding of megakaryopoiesis and how Mks interact with HSCs and immune cells has potential clinical implications for stem cell transplantation and other therapies for hematologic malignancies, as well as for treatments to stimulate platelet production and prevent thrombocytopenia.
Collapse
Affiliation(s)
- Eman Khatib-Massalha
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
| | - Simón Méndez-Ferrer
- Wellcome-Medical Research Council (MRC) Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Hematology, University of Cambridge, Cambridge, United Kingdom
- National Health Service Blood and Transplant, Cambridge Biomedical Campus, Cambridge, United Kingdom
- Instituto de Biomedicina de Sevilla-IBiS, Hospitales Universitarios Virgen del Rocío y Macarena/Spanish National Research Council (CSIC)/Universidad de Sevilla, Seville, Spain
- Departamento de Fisiología Médica y Biofísica, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
14
|
Belyaeva E, Rubenstein A, Pierson SK, Dalldorf D, Frank D, Lim MS, Fajgenbaum DC. Bone Marrow Findings of Idiopathic Multicentric Castleman Disease: A Histopathologic Analysis and Systematic Literature Review. Hematol Oncol 2022; 40:191-201. [PMID: 35104370 DOI: 10.1002/hon.2969] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 11/12/2022]
Abstract
Idiopathic multicentric Castleman disease (iMCD) is a polyclonal lymphoproliferative disorder characterized by constitutional symptoms, generalized lymphadenopathy, cytopenias, and multi-organ dysfunction due to excessive cytokines, notably Interleukin-6. iMCD is often sub-classified into iMCD-TAFRO, which is associated with thrombocytopenia (T), anasarca (A), fever/elevated C-reactive protein (F), renal dysfunction (R), and organomegaly (O), and iMCD-NOS, which is typically associated with thrombocytosis and hypergammaglobulinemia. The diagnosis of iMCD is challenging as consensus clinico-pathological diagnostic criteria were only recently established and include several non-specific lymph node histopathological features. Identification of further clinico-pathological features commonly found in iMCD could contribute to more accurate and timely diagnoses. We set out to characterize bone marrow (BM) histopathological features in iMCD, assess differences between iMCD-TAFRO and iMCD-NOS, and determine if these findings are specific to iMCD. Examination of BM specimens from 24 iMCD patients revealed a high proportion with hypercellularity, megakaryocytic atypia, reticulin fibrosis, and plasmacytosis across patients with both iMCD-NOS and iMCD-TAFRO with significantly more megakaryocytic hyperplasia (p=0.001) in the iMCD-TAFRO cases. These findings were also consistent with bone marrow findings from 185 published cases of iMCD-NOS and iMCD-TAFRO. However, these findings are relatively nonspecific as they can be seen in various other infectious, malignant, and autoimmune diseases. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Elizaveta Belyaeva
- Department of Pathology and Laboratory Medicine, Tulane University, New Orleans, Usa, 70112
| | - Ayelet Rubenstein
- Department of Medicine, Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, Usa, 19104
| | - Sheila K Pierson
- Department of Medicine, Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, Usa, 19104
| | - Delaney Dalldorf
- Castleman Disease Collaborative Network, Philadelphia, Usa, 19104
| | - Dale Frank
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Usa, 19104
| | - Megan S Lim
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Usa, 19104
| | - David C Fajgenbaum
- Department of Medicine, Center for Cytokine Storm Treatment & Laboratory, University of Pennsylvania, Philadelphia, Usa, 19104
| |
Collapse
|
15
|
Discovery of a novel megakaryopoiesis enhancer, ingenol, promoting thrombopoiesis through PI3K-Akt signaling independent of thrombopoietin. Pharmacol Res 2022; 177:106096. [PMID: 35077844 DOI: 10.1016/j.phrs.2022.106096] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Revised: 01/08/2022] [Accepted: 01/20/2022] [Indexed: 01/09/2023]
Abstract
Thrombocytopenia, a most common complication of radiotherapy and chemotherapy, is an important cause of morbidity and mortality in cancer patients. However, there are still no approved agents for the treatment of radiation- and chemotherapy-induced thrombocytopenia (RIT and CIT, respectively). In this study, a drug screening model for predicting compounds with activity in promoting megakaryocyte (MK) differentiation and platelet production was established based on machine learning (ML), and a natural product ingenol was predicted as a potential active compound. Then, in vitro experiments showed that ingenol significantly promoted MK differentiation in K562 and HEL cells. Furthermore, a RIT mice model and c-MPL knock-out (c-MPL-/-) mice constructed by CRISPR/Cas9 technology were used to assess the therapeutic action of ingenol on thrombocytopenia. The results showed that ingenol accelerated megakaryopoiesis and thrombopoiesis both in RIT mice and c-MPL-/- mice. Next, RNA-sequencing (RNA-seq) was carried out to analyze the gene expression profile induced by ingenol during MK differentiation. Finally, through experimental verifications, we demonstrated that the activation of PI3K/Akt signaling pathway was involved in ingenol-induced MK differentiation. Blocking PI3K/Akt signaling pathway abolished the promotion of ingenol on MK differentiation. Nevertheless, inhibition of TPO/c-MPL signaling pathway could not suppress ingenol-induced MK differentiation. In conclusion, our study builds a drug screening model to discover active compounds against thrombocytopenia, reveals the critical roles of ingenol in promoting MK differentiation and platelet production, and provides a promising avenue for the treatment of RIT.
Collapse
|
16
|
A dry immersion model of microgravity modulates platelet phenotype, miRNA signature, and circulating plasma protein biomarker profile. Sci Rep 2021; 11:21906. [PMID: 34753989 PMCID: PMC8578674 DOI: 10.1038/s41598-021-01335-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 10/26/2021] [Indexed: 11/08/2022] Open
Abstract
Ground based research modalities of microgravity have been proposed as innovative methods to investigate the aetiology of chronic age-related conditions such as cardiovascular disease. Dry Immersion (DI), has been effectively used to interrogate the sequelae of physical inactivity (PI) and microgravity on multiple physiological systems. Herein we look at the causa et effectus of 3-day DI on platelet phenotype, and correlate with both miRomic and circulating biomarker expression. The miRomic profile of platelets is reflective of phenotype, which itself is sensitive and malleable to the exposome, undergoing responsive transitions in order to fulfil platelets role in thrombosis and haemostasis. Heterogeneous platelet subpopulations circulate at any given time, with varying degrees of sensitivity to activation. Employing a DI model, we investigate the effect of acute PI on platelet function in 12 healthy males. 3-day DI resulted in a significant increase in platelet count, plateletcrit, platelet adhesion, aggregation, and a modest elevation of platelet reactivity index (PRI). We identified 15 protein biomarkers and 22 miRNA whose expression levels were altered after DI. A 3-day DI model of microgravity/physical inactivity induced a prothrombotic platelet phenotype with an unique platelet miRNA signature, increased platelet count and plateletcrit. This correlated with a unique circulating protein biomarker signature. Taken together, these findings highlight platelets as sensitive adaptive sentinels and functional biomarkers of epigenetic drift within the cardiovascular compartment.
Collapse
|
17
|
Jarlborg M, Gabay C. Systemic effects of IL-6 blockade in rheumatoid arthritis beyond the joints. Cytokine 2021; 149:155742. [PMID: 34688020 DOI: 10.1016/j.cyto.2021.155742] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 09/13/2021] [Accepted: 10/05/2021] [Indexed: 12/15/2022]
Abstract
Interleukin (IL)-6 is produced locally in response to an inflammatory stimulus, and is able to induce systemic manifestations at distance from the site of inflammation. Its unique signaling mechanism, including classical and trans-signaling pathways, leads to a major expansion in the number of cell types responding to IL-6. This pleiotropic cytokine is a key factor in the pathogenesis of rheumatoid arthritis (RA) and is involved in many extra-articular manifestations that accompany the disease. Thus, IL-6 blockade is associated with various biological effects beyond the joints. In this review, the systemic effects of IL-6 in RA comorbidities and the consequences of its blockade will be discussed, including anemia of chronic disease, cardiovascular risks, bone and muscle functions, and neuro-psychological manifestations.
Collapse
Affiliation(s)
- Matthias Jarlborg
- Division of Rheumatology, University Hospital of Geneva, and Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland; VIB-UGent Center for Inflammation Research and Ghent University, Ghent, Belgium
| | - Cem Gabay
- Division of Rheumatology, University Hospital of Geneva, and Department of Pathology and Immunology, University of Geneva School of Medicine, Geneva, Switzerland.
| |
Collapse
|
18
|
Abstract
Thrombocytopoiesis is a complex process beginning at the level of hematopoietic stem cells, which ultimately generate megakaryocytes, large marrow cells with a distinctive morphology, and then, through a process of terminal maturation, megakaryocytes shed thousands of platelets into the circulation. This process is controlled by intrinsic and extrinsic factors. Emerging data indicate that an important intrinsic control on the late stages of thrombopoiesis is exerted by integrins, a family of transmembrane receptors composed of one α and one β subunit. One β subunit expressed by megakaryocytes is the β1 integrin, the role of which in the regulation of platelet formation is beginning to be clarified. Here, we review recent data indicating that activation of β1 integrin by outside-in and inside-out signaling regulates the interaction of megakaryocytes with the endosteal niche, which triggers their maturation, while its inactivation by galactosylation determines the migration of these cells to the perivascular niche, where they complete their terminal maturation and release platelets in the bloodstream. Furthermore, β1 integrin mediates the activation of transforming growth factor β (TGF-β), a protein produced by megakaryocytes that may act in an autocrine fashion to halt their maturation and affect the composition of their surrounding extracellular matrix. These findings suggest that β1 integrin could be a therapeutic target for inherited and acquired disorders of platelet production.
Collapse
Affiliation(s)
- Maria Mazzarini
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Italy
| | - Paola Verachi
- Biomedical and Neuromotor Sciences, Alma Mater University Bologna, Italy
| | - Fabrizio Martelli
- National Center for Preclinical and Clinical Research and Evaluation of Pharmaceutical Drugs, Rome, Italy
| | - Anna Rita Migliaccio
- University Campus Biomedico, Rome, Italy
- Myeloproliferative Neoplasm-Research Consortium, New York, NY, USA
| |
Collapse
|
19
|
Abstract
IL-6 is involved both in immune responses and in inflammation, hematopoiesis, bone metabolism and embryonic development. IL-6 plays roles in chronic inflammation (closely related to chronic inflammatory diseases, autoimmune diseases and cancer) and even in the cytokine storm of corona virus disease 2019 (COVID-19). Acute inflammation during the immune response and wound healing is a well-controlled response, whereas chronic inflammation and the cytokine storm are uncontrolled inflammatory responses. Non-immune and immune cells, cytokines such as IL-1β, IL-6 and tumor necrosis factor alpha (TNFα) and transcription factors nuclear factor-kappa B (NF-κB) and signal transducer and activator of transcription 3 (STAT3) play central roles in inflammation. Synergistic interactions between NF-κB and STAT3 induce the hyper-activation of NF-κB followed by the production of various inflammatory cytokines. Because IL-6 is an NF-κB target, simultaneous activation of NF-κB and STAT3 in non-immune cells triggers a positive feedback loop of NF-κB activation by the IL-6-STAT3 axis. This positive feedback loop is called the IL-6 amplifier (IL-6 Amp) and is a key player in the local initiation model, which states that local initiators, such as senescence, obesity, stressors, infection, injury and smoking, trigger diseases by promoting interactions between non-immune cells and immune cells. This model counters dogma that holds that autoimmunity and oncogenesis are triggered by the breakdown of tissue-specific immune tolerance and oncogenic mutations, respectively. The IL-6 Amp is activated by a variety of local initiators, demonstrating that the IL-6-STAT3 axis is a critical target for treating diseases.
Collapse
Affiliation(s)
- Toshio Hirano
- National Institutes for Quantum and Radiological Science and Technology, Anagawa, Inage-ku, Chiba, Japan
- Division of Molecular Psychoimmunology, Institute for Genetic Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
20
|
Role of thrombopoiesis in leishmaniasis. Cytokine 2020; 147:155310. [PMID: 33127256 DOI: 10.1016/j.cyto.2020.155310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 11/21/2022]
Abstract
The blood vascular system of mammals is unique in nature; inhabited with a pool of tiny small cell fragments called platelets; attributed with the most important patrolling tasks to check integrity of the entire endothelial landscape. Their production is tightly coupled with hematopoietic system where everything starts from self renewable multipotent hematopoietic stem cells (HSCs) which eventually undergo dual step (megakaryopoiesis-thrombopoiesis) thrombocytes production. Several cytokines tune the fate of every progenitor cells during hematopoiesis through temporal activation of specific transcription factors. Though platelets generated through steady state hematopoiesis are involved in the regulation of vascular homeostasis, these cells can sense pathogens through its innate immune sensors and can mount crucial responses against the invading pathogen. For this, the primary aim of many infections including Leishmania is to induce thrombocytopenia within infected host. But the underlying mechanism of this induced thrombocytopenia in Leishmania infection has not been evaluated. Elucidation of these mechanisms will be fruitful to design new chemotherapeutic strategies.
Collapse
|
21
|
Tocilizumab for the treatment of TAFRO syndrome: a systematic literature review. Ann Hematol 2020; 99:2463-2475. [DOI: 10.1007/s00277-020-04275-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/17/2020] [Indexed: 02/07/2023]
|
22
|
Interleukin-6 in Rheumatoid Arthritis. Int J Mol Sci 2020; 21:ijms21155238. [PMID: 32718086 PMCID: PMC7432115 DOI: 10.3390/ijms21155238] [Citation(s) in RCA: 132] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 07/18/2020] [Accepted: 07/20/2020] [Indexed: 01/08/2023] Open
Abstract
The role of interleukin (IL)-6 in health and disease has been under a lot of scrutiny in recent years, particularly during the recent COVID-19 pandemic. The inflammatory pathways in which IL-6 is involved are also partly responsible of the development and progression of rheumatoid arthritis (RA), opening interesting perspectives in terms of therapy. Anti-IL-6 drugs are being used with variable degrees of success in other diseases and are being tested in RA. Results have been encouraging, particularly when anti-IL-6 has been used with other drugs, such as metothrexate (MTX). In this review we discuss the main immunologic aspects that make anti-IL-6 a good candidate in RA, but despite the main therapeutic options available to target IL-6, no gold standard treatment has been established so far.
Collapse
|
23
|
Recent Advances in Electrochemical and Optical Biosensors Designed for Detection of Interleukin 6. SENSORS 2020; 20:s20030646. [PMID: 31979357 PMCID: PMC7038342 DOI: 10.3390/s20030646] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/06/2019] [Accepted: 12/18/2019] [Indexed: 12/11/2022]
Abstract
Interleukin 6 (IL-6), being a major component of homeostasis, immunomodulation, and hematopoiesis, manifests multiple pathological conditions when upregulated in response to viral, microbial, carcinogenic, or autoimmune stimuli. High fidelity immunosensors offer real-time monitoring of IL-6 and facilitate early prognosis of life-threatening diseases. Different approaches to augment robustness and enhance overall performance of biosensors have been demonstrated over the past few years. Electrochemical- and fluorescence-based detection methods with integrated electronics have been subjects of intensive research due to their ability to offer a better signal-to-noise ratio, high specificity, ultra-sensitivity, and wide dynamic range. In this review, the pleiotropic role of IL-6 and its clinical significance is discussed in detail, followed by detection schemes devised so far for their quantitative analysis. A critical review on underlying signal amplification strategies and performance of electrochemical and optical biosensors is presented. In conclusion, we discuss the reliability and feasibility of the proposed detection technologies for commercial applications.
Collapse
|
24
|
Wang P, Feng YB, Wang L, Li Y, Fan C, Song Q, Yu SY. Interleukin-6: Its role and mechanisms in rescuing depression-like behaviors in rat models of depression. Brain Behav Immun 2019; 82:106-121. [PMID: 31394209 DOI: 10.1016/j.bbi.2019.08.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 07/27/2019] [Accepted: 08/04/2019] [Indexed: 12/12/2022] Open
Abstract
Neuronal injury within specific brain regions is considered a critical risk factor in the pathophysiology of depression. However, the underlying mechanisms of this process, and thus the potential for development of novel therapeutic strategies in the treatment of depression, remain largely unknown. Here, we report that Il-6 protects against neuronal anomalies related with depression, in part, by suppressing oxidative stress and consequent autophagic and apoptotic hyperactivity. Specifically, we show that IL-6 is downregulated within the CA1 hippocampus in two animal models of depression and upregulated by antidepressants. Increasing levels of IL-6 in the CA1 region result in pleiotropic protective actions including reductions in oxidative stress and modulation of autophagy, anti-immuno-inflammatory activation and anti-apoptotic effects in CA1 neurons, all of which are associated with the rescue of depression-like behaviors. In contrast, IL-6 downregulation exacerbates neuronal anomalies within the CA1 region and facilitates the genesis of depression phenotypes in rats. Interestingly, in addition to attenuating oxidative damage, the antioxidant, N-acetylcysteine (NAC), is also associated with significantly decreased neuronal deficits and the display of depressive behaviors in rats. These results suggest that IL-6 may exert neuroprotection within CA1 neurons via pleiotropic mechanisms and may serve as a potential therapeutic target for the treatment of depression.
Collapse
Affiliation(s)
- Peng Wang
- Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, Shandong Province 250012, PR China
| | - Ya-Bo Feng
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong University, Jingwuweiqi Road 423#, Jinan, Shandong Province 250012, PR China
| | - Liyan Wang
- Morphological Experimental Center, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, Shandong Province 250012, PR China
| | - Ye Li
- Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, Shandong Province 250012, PR China
| | - Cuiqin Fan
- Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, Shandong Province 250012, PR China
| | - Qiqi Song
- Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, Shandong Province 250012, PR China
| | - Shu Yan Yu
- Department of Physiology, Shandong University, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, Shandong Province 250012, PR China; Shandong Provincial Key Laboratory of Mental Disorders, School of Basic Medical Sciences, 44 Wenhuaxilu Road, Jinan, Shandong Province 250012, PR China.
| |
Collapse
|
25
|
Karkhur S, Hasanreisoglu M, Vigil E, Halim MS, Hassan M, Plaza C, Nguyen NV, Afridi R, Tran AT, Do DV, Sepah YJ, Nguyen QD. Interleukin-6 inhibition in the management of non-infectious uveitis and beyond. J Ophthalmic Inflamm Infect 2019; 9:17. [PMID: 31523783 PMCID: PMC6745304 DOI: 10.1186/s12348-019-0182-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 08/02/2019] [Indexed: 02/06/2023] Open
Abstract
Background Uveitis consists of a spectrum of inflammatory disorders characterized by ocular inflammation. The underlying pathophysiology consists of a complex interplay of various inflammatory pathways. Interleukin 6 is an important mediator of inflammation in uveitis and constitutes focus of research toward development of newer biological therapies in the management of non-infectious uveitis. Main body Pan-blockade of the inflammatory pathways with steroids is generally the first step in the management of acute non-infectious uveitis. However, long-term therapy with steroids is associated with systemic and ocular side effects, thereby necessitating the need for development of steroid sparing agents. IL-6 is a cytokine produced by various immune cells, in response to molecular patterns and affects multiple inflammatory cells. In particular, IL-6 is involved in differentiation of CD-4 cells into Th-17 cells that have been shown to play a significant role in various immune-mediated diseases such as uveitis. This broad-spectrum immunomodulatory activity makes IL-6 an excellent target for immunomodulatory therapy. Tocilizumab was the first IL-6 inhibitor to demonstrate efficacy in humans. It inhibits IL-6 from binding to both membrane-bound and soluble receptor and can be administered via intravenous (IV) and subcutaneous (SC) routes. It has been FDA approved for treatment of rheumatoid arthritis (RA) and juvenile idiopathic arthritis (JIA). Following the approval in systemic diseases, its efficacy was demonstrated in various uveitis studies including a phase 2 clinical trial (STOP-Uveitis). Overall, tocilizumab has shown a good safety profile with the risk of malignancy consistent with that expected in patients with rheumatoid arthritis. However, tocilizumab therapy has been shown to increase the risk for gastrointestinal perforation and dose-dependent neutropenia. Following the success of tocilizumab, several other agents targeting the IL-6 pathway are in the pipeline. These include sirukumab, siltuximab, olokizumab, clazakizumab, and EBI-031 which target IL-6; Sarilumab and ALX-0061 act on the IL-6 receptor. Conclusion Studies have shown that IL-6 inhibitors can be effective in the management of NIU. In addition, the levels of IL-6 are elevated in other ocular vascular diseases such as retinal vein occlusion and diabetic macular edema. The roles of IL-6 inhibition may be broadened in the future to include the management of retinal vascular diseases and non-uveitic macular edema.
Collapse
Affiliation(s)
- Samendra Karkhur
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.,Department of Ophthalmology, All India Institute of Medical Sciences Bhopal, Bhopal, Madhya Pradesh, India
| | - Murat Hasanreisoglu
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.,Department of Ophthalmology, School of Medicine, Gazi University, Ankara, Turkey
| | - Erin Vigil
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.,University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Muhammad Sohail Halim
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Muhammad Hassan
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Carlos Plaza
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.,Department of Ophthalmology, Hospital Universitario de León, León, Spain
| | - Nam V Nguyen
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.,University of Nebraska, Lincoln, USA
| | - Rubbia Afridi
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Anh T Tran
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Diana V Do
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Yasir J Sepah
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA
| | - Quan Dong Nguyen
- Byers Eye Institute, Spencer Center for Vision Research, Stanford University, 2370 Watson Court, Suite 200, Palo Alto, CA, 94303, USA.
| |
Collapse
|
26
|
Houck KL, Yuan H, Tian Y, Solomon M, Cramer D, Liu K, Zhou Z, Wu X, Zhang J, Oehler V, Dong JF. Physical proximity and functional cooperation of glycoprotein 130 and glycoprotein VI in platelet membrane lipid rafts. J Thromb Haemost 2019; 17:1500-1510. [PMID: 31145836 DOI: 10.1111/jth.14525] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/28/2019] [Indexed: 12/01/2022]
Abstract
OBJECTIVE Clinical and laboratory studies have demonstrated that platelets become hyperactive and prothrombotic in conditions of inflammation. We have previously shown that the proinflammatory cytokine interleukin (IL)-6 forms a complex with soluble IL-6 receptor α (sIL-6Rα) to prime platelets for activation by subthreshold concentrations of collagen. Upon being stimulated with collagen, the transcription factor signal transducer and activator of transcription (STAT) 3 in platelets is phosphorylated and dimerized to act as a protein scaffold to facilitate the catalytic action between the kinase Syk and the substrate phospholipase Cγ2 (PLCγ2) in collagen-induced signaling. However, it remains unknown how collagen induces phosphorylation and dimerization of STAT3. METHODS AND RESULTS We conducted complementary in vitro experiments to show that the IL-6 receptor subunit glycoprotein 130 (GP130) was in physical proximity to the collagen receptor glycoprotein VI (GPVI in membrane lipid rafts of platelets. This proximity allows collagen to induce STAT3 activation and dimerization, and the IL-6-sIL-6Rα complex to activate the kinase Syk and the substrate PLCγ2 in the GPVI signal pathway, resulting in an enhanced platelet response to collagen. Disrupting lipid rafts or blocking GP130-Janus tyrosine kinase (JAK)-STAT3 signaling abolished the cross-activation and reduced platelet reactivity to collagen. CONCLUSION These results demonstrate cross-talk between collagen and IL-6 signal pathways. This cross-talk could potentially provide a novel mechanism for inflammation-induced platelet hyperactivity, so the IL-6-GP130-JAK-STAT3 pathway has been identified as a potential target to block this hyperactivity.
Collapse
Affiliation(s)
| | - Hengjie Yuan
- Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China
| | - Ye Tian
- Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China
| | | | - Drake Cramer
- Bloodworks Research Institute, Seattle, Washington
| | - Kitty Liu
- Bloodworks Research Institute, Seattle, Washington
| | - Zhou Zhou
- State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Fuwai Hospital, Beijing, China
| | - Xiaoping Wu
- Bloodworks Research Institute, Seattle, Washington
| | - Jianning Zhang
- Tianjin Neurological Institute, General Hospital, Tianjin Medical University, Tianjin, China
| | - Vivian Oehler
- Clinical Research Division, Hutchison Cancer Center, Seattle, Washington
- Seattle Cancer Alliances, Seattle, Washington
- Division of Hematology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| | - Jing-Fei Dong
- Bloodworks Research Institute, Seattle, Washington
- Division of Hematology, Department of Medicine, School of Medicine, University of Washington, Seattle, Washington
| |
Collapse
|
27
|
Involvement of circulating inflammatory factors in prognosis and risk of cardiovascular disease. J Mol Cell Cardiol 2019; 132:110-119. [DOI: 10.1016/j.yjmcc.2019.05.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 05/09/2019] [Accepted: 05/12/2019] [Indexed: 12/11/2022]
|
28
|
Couldwell G, Machlus KR. Modulation of megakaryopoiesis and platelet production during inflammation. Thromb Res 2019; 179:114-120. [PMID: 31128560 DOI: 10.1016/j.thromres.2019.05.008] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2019] [Revised: 04/19/2019] [Accepted: 05/13/2019] [Indexed: 12/24/2022]
Abstract
Megakaryocytes (MKs) are widely known as the progenitor cells of platelets. These large, polyploid cells are a derivative of the hematopoietic stem cell (HSC), and reside in the bone marrow, lining blood vessel walls where they release their platelet progeny into circulation. Although little is known about how MKs differ under various environmental stressors, both chronic and acute inflammation alter the differentiation and molecular content of MKs. Furthermore, evidence suggests that the release of inflammatory cytokines may induce MK rupture and rapid release of platelets as a mechanism to quickly replenish diminished platelet counts in response to inflammation. Similarities between MKs and their close relatives, white blood cells, have introduced the notion that MKs may play a role in combating infection by engulfing and presenting antigens, and passing this information to circulating platelets. In addition, MKs exposed to varying bone marrow environments produce different platelets which enter circulation primed to respond to and combat inflammation, infection, or injury. This review focuses on how inflammation alters MK production, maturation, and platelet production. In addition, it introduces the idea that inflammation reprograms MKs to create different, more pathogenic platelets and leads them to take on different roles as responders to deleterious conditions. In the future, studies determining how platelets are altered in disease states may lead to novel MK- and platelet-based therapeutic targets to mitigate inflammation-related morbidity and mortality.
Collapse
Affiliation(s)
| | - Kellie R Machlus
- Division of Hematology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
29
|
O'Neill L, McCormick J, Gao W, Veale DJ, McCarthy GM, Murphy CC, Fearon U, Molloy ES. Interleukin-6 does not upregulate pro-inflammatory cytokine expression in an ex vivo model of giant cell arteritis. Rheumatol Adv Pract 2019; 3:rkz011. [PMID: 31431999 PMCID: PMC6649906 DOI: 10.1093/rap/rkz011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 03/17/2019] [Indexed: 11/14/2022] Open
Abstract
Objective The aim of this study was to examine the pro-inflammatory effects of IL-6 in ex vivo temporal artery explant cultures. Methods Patients meeting 1990 ACR classification criteria for GCA were prospectively recruited. Temporal artery biopsies were obtained and temporal artery explants cultured ex vivo with IL-6 (10-40 ng/ml) in the presence or absence of its soluble receptor (sIL-6R; 20 ng/ml) for 24 h. Explant supernatants were harvested after 24 h and assayed for IFN-γ, TNF-α, Serum amyloid A, IL-1β, IL-17, IL-8, angiotensin II and VEGF by ELISA. Myofibroblast outgrowths, cytoskeletal rearrangement and wound repair assays were performed. Results IL-6 augmented production of VEGF, but not of any of the other pro-inflammatory mediators assayed. No differences were observed in the explants cultured in the presence or absence of the sIL-6R or between those with a positive (n = 11) or negative (n = 17) temporal artery biopsy. IL-6 did not enhance myofibroblast proliferation or migration. Western blot analysis confirmed signalling activation, with increased expression of pSTAT3 in response to IL-6+sIL-6R. Conclusion IL-6 stimulation of temporal artery explants from patients with GCA neither increased expression of key pro-inflammatory mediators nor influenced myofibroblast proliferation or migration.
Collapse
Affiliation(s)
- Lorraine O'Neill
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Royal College of Surgeons, Ireland
| | - Jennifer McCormick
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Royal College of Surgeons, Ireland
| | - Wei Gao
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Royal College of Surgeons, Ireland
| | - Douglas J Veale
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Royal College of Surgeons, Ireland
| | - Geraldine M McCarthy
- Mater Misericordiae University Hospital, Dublin Academic Medical Centre, Royal College of Surgeons, Ireland
| | - Conor C Murphy
- Department of Ophthalmology, Royal Victoria Eye and Ear Hospital, Royal College of Surgeons, Ireland
| | - Ursula Fearon
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Royal College of Surgeons, Ireland
| | - Eamonn S Molloy
- Centre for Arthritis and Rheumatic Diseases, St Vincent's University Hospital, Dublin Academic Medical Centre, Royal College of Surgeons, Ireland
| |
Collapse
|
30
|
Fu W, Wang J, Jiang H, Hu X. Myocardial infarction induces bone marrow megakaryocyte proliferation, maturation and platelet production. Biochem Biophys Res Commun 2019; 510:456-461. [PMID: 30732856 DOI: 10.1016/j.bbrc.2019.01.129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 01/27/2019] [Accepted: 01/29/2019] [Indexed: 01/29/2023]
Abstract
Platelet, apart from its classic role of homeostasis, serves also as a crucial immune cell component that contributes to the aggravation of atherosclerosis. It has been reported that myocardial infarction (MI) triggers leukocytosis in the bone marrow and spleen, which accelerates post-MI atherosclerosis. However, it remains unclear whether thrombopoiesis is also enhanced after MI. Here, using flow cytometry and bone marrow whole-mount immunofluorescence staining combined with three-dimensional (3D) reconstruction, we for the first time demonstrated an enhanced thrombopoiesis and megakaryopoiesis in a mouse model of coronary artery ligation as a mimic of MI. We showed that MI leads to increasing number of peripheral platelets, as well as elevating number and larger size of bone marrow MKs. We also observed more proplatelets and fragmented MKs, and a closer spatial distribution of MK populations to the bone marrow vascular niche after MI. This study provides direct evidence that MI induces bone marrow megakaryocyte proliferation, maturation and platelet production. It opens a new scope that targeting platelet production might become a novel therapeutic approach for attenuating post-MI atherosclerosis.
Collapse
Affiliation(s)
- Wenwen Fu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Jichun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Cardiology, Wuhan, PR China
| | - Hong Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China; Cardiovascular Research Institute, Wuhan University, Wuhan, PR China; Hubei Key Laboratory of Cardiology, Wuhan, PR China.
| | - Xiaorong Hu
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, PR China.
| |
Collapse
|
31
|
The Two-Faced Cytokine IL-6 in Host Defense and Diseases. Int J Mol Sci 2018; 19:ijms19113528. [PMID: 30423923 PMCID: PMC6274717 DOI: 10.3390/ijms19113528] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 10/30/2018] [Accepted: 11/06/2018] [Indexed: 02/07/2023] Open
Abstract
Interleukein-6 (IL-6), is produced locally from infectious or injured lesions and is delivered to the whole body via the blood stream, promptly activating the host defense system to perform diverse functions. However, excessive or sustained production of IL-6 is involved in various diseases. In diseases, the IL-6 inhibitory strategy begins with the development of the anti-IL-6 receptor antibody, tocilizumab (TCZ). This antibody has shown remarkable effects on Castleman disease, rheumatoid arthritis and juvenile idiopathic arthritis. In 2017, TCZ was proven to work effectively against giant cell arteritis, Takayasu arteritis and cytokine releasing syndrome, initiating a new era for the treatment of these diseases. In this study, the defensive functions of IL-6 and various pathological conditions are compared. Further, the diseases of which TCZ has been approved for treatment are summarized, the updated results of increasing off-label use of TCZ for various diseases are reviewed and the conditions for which IL-6 inhibition might have a beneficial role are discussed. Given the involvement of IL-6 in many pathologies, the diseases that can be improved by IL-6 inhibition will expand. However, the important role of IL-6 in host defense should always be kept in mind in clinical practice.
Collapse
|
32
|
Abstract
Platelets are anuclear blood cells required for haemostasis and are implicated in other processes including inflammation and metastasis. Platelets are produced by megakaryocytes, specialized cells that are themselves generated by a process of controlled differentiation and maturation of bone-marrow stem and progenitor cells. This process of megakaryopoiesis involves the coordinated interplay of transcription factor-controlled cellular programming with extra-cellular cues produced locally in supporting niches or as circulating factors. This review focuses on these external cues, the cytokines and chemokines, that drive production of megakaryocytes and support the terminal process of platelet release. Emphasis is given to thrombopoietin (Tpo), the major cytokine regulator of steady-state megakaryopoiesis, and its specific cell surface receptor, the Mpl protein, including normal and pathological roles as well as clinical application. The potential for alternative or supplementary regulatory mechanisms for platelet production, particularly in times of acute need, or in states of infection or inflammation are also discussed.
Collapse
Affiliation(s)
- Kira Behrens
- a The Walter and Eliza Hall Institute of Medical Research , Parkville , Australia
- b Department of Medical Biology , University of Melbourne , Melbourne , Australia
| | - Warren S Alexander
- a The Walter and Eliza Hall Institute of Medical Research , Parkville , Australia
- b Department of Medical Biology , University of Melbourne , Melbourne , Australia
| |
Collapse
|
33
|
Kim HJ, Jung SM, Song JJ, Park YB, Lee SW. Mean platelet volume can estimate the current vasculitis activity of microscopic polyangiitis. Rheumatol Int 2018; 38:1095-1101. [PMID: 29556749 DOI: 10.1007/s00296-018-4011-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 03/15/2018] [Indexed: 12/27/2022]
Abstract
Mean platelet volume (MPV) was known to be associated with the inflammatory burdens in various diseases, but not microscopic polyangiitis (MPA) yet. In this study, we investigated the association between MPV and Birmingham vasculitis activity score (BVAS) in 88 patients with MPA. We retrospectively reviewed the medical records of 88 patients with MPA. Clinical and laboratory data at diagnosis and partially at follow-up were collected. Linear regression analyses were used to clarify the association between MPV and BVAS. We also stratified MPA patients into three groups according to tertile of BVAS and define the lower limit of the highest tertile as the cut-off of BVAS for severe MPA. The area under the receiver operator characteristic curve (AUROC) was used to identify the cut-off of MPV for severe MPA (8.85 fL). The mean age was 60.5 years and 25 patients were male (28.4%). In univariable linear regression analysis, MPV was negatively correlated with BVAS (r = - 0.326). In multivariable linear regression analysis, MPA was independently associated with BVAS (β = - 0.260). When we classified MPA patients into two groups based on the cutoff of MPV for severe MPA, patients having MPV ≤ 8.85 fL exhibited the higher frequency of severe MPA than those having MPV > 8.85 fL (RR 5.000). In addition, MPV increased along with improvement of vasculitis activity during the follow-up. In conclusion, MPV is significantly associated with BVAS and it can estimate the current BVAS in patients with MPA.
Collapse
Affiliation(s)
- Ho Jae Kim
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jason Jungsik Song
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Yong-Beom Park
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sang-Won Lee
- Division of Rheumatology, Department of Internal Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea. .,Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
34
|
Hernández Vera R, Padró T, Vilahur G, Badimon L. Antithrombotic therapy in obesity. Thromb Haemost 2017; 110:681-8. [DOI: 10.1160/th12-12-0928] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Accepted: 03/11/2013] [Indexed: 12/17/2022]
Abstract
summaryClinical management of obese subjects to reduce their risk of suffering cardiovascular events is complex. Obese patients typically require preventive strategies, life-style modifications, and multi-drug therapy to address obesity-induced co-morbidities. Data regarding the effects of excess weight on the pharmacokinetics of most drugs is scarce as these individuals are often excluded from clinical trials. However, the physiological alterations observed in obese patients and their lower response to some antiplatelet agents and anticoagulants have suggested that dosage regimes need to be adjusted for these subjects. In this review we will briefly discuss platelet alterations that can contributeto increased thrombotic risk, analyse existing data regarding the effects of obesity on drug pharmacokinetics focusing on antiplatelet agents and anticoagulants, and we will describe the beneficial effects of weight loss on thrombosis.
Collapse
|
35
|
Larsen SB, Grove EL, Würtz M, Neergaard-Petersen S, Hvas AM, Kristensen SD. The influence of low-grade inflammation on platelets in patients with stable coronary artery disease. Thromb Haemost 2017; 114:519-29. [DOI: 10.1160/th14-12-1007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 05/07/2015] [Indexed: 12/19/2022]
Abstract
SummaryInflammation is likely to be involved in all stages of atherosclerosis. Numerous inflammatory biomarkers are currently being studied, and even subtle increases in inflammatory biomarkers have been associated with increased risk of cardiovascular events in patients with coronary artery disease (CAD). Low-grade inflammation may influence both platelet production and platelet activation potentially leading to enhanced platelet aggregation. Thrombopoietin is considered the primary regulator of platelet production, but it likely acts in conjunction with numerous cytokines, of which many have altered levels in CAD. Previous studies have shown that high-sensitive C-reactive protein (CRP) independently predicts increased platelet aggregation in stable CAD patients. Increased levels of CRP, fibrinogen, interleukin-6, stromal cell-derived factor-1, CXC motif ligand 16, macrophage migration inhibitory factor, RANTES, calprotectin, and copeptin have been associated with increased risk of cardiovascular events in CAD patients. Additionally, some of these inflammatory markers have been associated with enhanced platelet activation and aggregation. However, CRP and other inflammatory markers provide only limited additional predictive value to classical risk factors such as smoking, blood pressure, and cholesterol levels. Existing data do not clarify whether inflammation simply accompanies CAD and increased production and aggregation of platelets, or whether a causal relationship exists. In this review, we provide a comprehensive overview of inflammatory markers in stable CAD with particular emphasis on platelet production, activation, and aggregation in CAD patients.
Collapse
|
36
|
Contribution of Interleukin-6 to the Pathogenesis of Systemic Sclerosis. JOURNAL OF SCLERODERMA AND RELATED DISORDERS 2017. [DOI: 10.5301/jsrd.5000258] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Systemic sclerosis (SSc) is a connective tissue disease of unknown etiology, manifesting in patients as tissue fibrosis, endothelial dysfunction, and inflammation. The disease is characterized by autoantibodies, a hallmark of autoimmunity. Various cytokines and growth factors are elevated in the systemic circulation and fibrotic lesions of patients with SSc. In particular, several studies over the past 2 decades have shown that interleukin-6 (IL-6) appears to be involved in the pathogenesis of SSc. Based on the association between aberrant IL-6 production and tissue fibrosis in patients with SSc, the anti-IL-6 receptor antibody, tocilizumab, is being investigated in clinical trials. This article reviews the biological features of IL-6 and the IL-6 receptor; the role of IL-6 in the pathogenesis of SSc; and the potential for IL-6 inhibition to be used in the treatment of patients with SSc.
Collapse
|
37
|
Kostyak JC, Liverani E, Kunapuli SP. PKC-epsilon deficiency alters progenitor cell populations in favor of megakaryopoiesis. PLoS One 2017; 12:e0182867. [PMID: 28783756 PMCID: PMC5544228 DOI: 10.1371/journal.pone.0182867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Accepted: 07/25/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND It has long been postulated that Protein Kinase C (PKC) is an important regulator of megakaryopoiesis. Recent contributions to the literature have outlined the functions of several individual PKC isoforms with regard to megakaryocyte differentiation and platelet production. However, the exact role of PKCε remains elusive. OBJECTIVE To delineate the role of PKCε in megakaryopoiesis. APPROACH AND RESULTS We used a PKCε knockout mouse model to examine the effect of PKCε deficiency on platelet mass, megakaryocyte mass, and bone marrow progenitor cell distribution. We also investigated platelet recovery in PKCε null mice and TPO-mediated signaling in PKCε null megakaryocytes. PKCε null mice have higher platelet counts due to increased platelet production compared to WT littermate controls (p<0.05, n = 8). Furthermore, PKCε null mice have more bone marrow megakaryocyte progenitor cells than WT littermate control mice. Additionally, thrombopoietin-mediated signaling is perturbed in PKCε null mice as Akt and ERK1/2 phosphorylation are enhanced in PKCε null megakaryocytes stimulated with thrombopoietin. Finally, in response to immune-induced thrombocytopenia, PKCε null mice recovered faster and had higher rebound thrombocytosis than WT littermate control mice. CONCLUSIONS Enhanced platelet recovery could be due to an increase in megakaryocyte progenitor cells found in PKCε null mice as well as enhanced thrombopoietin-mediated signaling observed in PKCε deficient megakaryocytes. These data suggest that PKCε is a negative regulator of megakaryopoiesis.
Collapse
Affiliation(s)
- John C. Kostyak
- Sol Sherry Thrombosis Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Elisabetta Liverani
- Center for Inflammation, Translational and Clinical Lung Research, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
| | - Satya P. Kunapuli
- Sol Sherry Thrombosis Research Center, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Pharmacology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
- Department of Physiology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
38
|
Mesquida M, Molins B, Llorenç V, de la Maza MS, Adán A. Targeting interleukin-6 in autoimmune uveitis. Autoimmun Rev 2017; 16:1079-1089. [PMID: 28778705 DOI: 10.1016/j.autrev.2017.08.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 07/21/2017] [Indexed: 01/01/2023]
Abstract
Interleukin-6 (IL-6) is a key cytokine that is strongly up-regulated during infection and inflammation. Featuring pleiotropic activity, IL-6 is responsible for the induction of hepatic acute-phase proteins, trafficking of acute and chronic inflammatory cells, differentiation of adaptive T cell responses, homeostatic regulation, and tissue regeneration. Dysregulated IL-6 production has been associated with the development of a wide variety of systemic immune-mediated, chronic diseases, and even certain types of cancer. From the ocular perspective, significant elevation of IL-6 has been found in ocular fluids derived from diabetic macular edema, retinal vein occlusion, and refractory/chronic uveitis patients. During the last decade, tocilizumab, a neutralizing monoclonal antibody (mAb) that targets the IL-6 receptor (IL-6R), has been approved for the treatment of rheumatoid arthritis in >100 countries worldwide. Furthermore, it has been reported to be effective for the treatment of a number of autoimmune diseases including uveitis and its associated macular edema. Currently numerous candidate molecular strategies targeting the IL-6 signaling pathways are in progress through clinical trials in various disorders. Herein we discuss the basic biology of IL-6 and its pathological role in the development of immune-mediated conditions, particularly focusing on inflammatory eye diseases. It also provides an overview of the on-going clinical trials with the new anti-IL-6 mAbs and their potential use in the clinical practice.
Collapse
Affiliation(s)
- Marina Mesquida
- Uveitis Unit, Institut Clinic d'Oftalmologia, Hospital Clinic de Barcelona, Universitat de Barcelona, Spain; Fundació Clínic per a la Recerca Biomèdica, IDIBAPS, Barcelona, Spain.
| | - Blanca Molins
- Fundació Clínic per a la Recerca Biomèdica, IDIBAPS, Barcelona, Spain
| | - Víctor Llorenç
- Uveitis Unit, Institut Clinic d'Oftalmologia, Hospital Clinic de Barcelona, Universitat de Barcelona, Spain
| | - Maite Sáinz de la Maza
- Uveitis Unit, Institut Clinic d'Oftalmologia, Hospital Clinic de Barcelona, Universitat de Barcelona, Spain
| | - Alfredo Adán
- Uveitis Unit, Institut Clinic d'Oftalmologia, Hospital Clinic de Barcelona, Universitat de Barcelona, Spain
| |
Collapse
|
39
|
Iwaki N, Gion Y, Kondo E, Kawano M, Masunari T, Moro H, Nikkuni K, Takai K, Hagihara M, Hashimoto Y, Yokota K, Okamoto M, Nakao S, Yoshino T, Sato Y. Elevated serum interferon γ-induced protein 10 kDa is associated with TAFRO syndrome. Sci Rep 2017; 7:42316. [PMID: 28205564 PMCID: PMC5304226 DOI: 10.1038/srep42316] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 01/05/2017] [Indexed: 01/09/2023] Open
Abstract
Multicentric Castleman disease (MCD) is a heterogeneous lymphoproliferative disorder. It is characterized by inflammatory symptoms, and interleukin (IL)-6 contributes to the disease pathogenesis. Human herpesvirus 8 (HHV-8) often drives hypercytokinemia in MCD, although the etiology of HHV-8-negative MCD is idiopathic (iMCD). A distinct subtype of iMCD that shares a constellation of clinical features including thrombocytopenia (T), anasarca (A), fever (F), reticulin fibrosis (R), and organomegaly (O) has been reported as TAFRO-iMCD, however the differences in cytokine profiles between TAFRO-iMCD and iMCD have not been established. We retrospectively compared levels of serum interferon γ-induced protein 10 kDa (IP-10), platelet-derived growth factor (PDGF)-AA, interleukin (IL)-10, and other cytokines between 11 cases of TAFRO-iMCD, 6 cases of plasma cell type iMCD, and 21 healthy controls. During flare-ups, patients with TAFRO-iMCD had significantly higher serum IP-10 and tended to have lower PDGF-AA levels than the other 2 groups. In addition, serum IL-10, IL-23, and vascular endothelial growth factor-A were elevated in both TAFRO-iMCD and iMCD. Elevated serum IP-10 is associated with inflammatory diseases including infectious diseases. There was a strong correlation between high serum IP-10 and the presence of TAFRO-iMCD, suggesting that IP-10 might be involved in the pathogenesis of TAFRO-iMCD.
Collapse
Affiliation(s)
- Noriko Iwaki
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.,Hematology/Respiratory Medicine Kanazawa University Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| | - Yuka Gion
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Eisei Kondo
- Department of General Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Mitsuhiro Kawano
- Division of Rheumatology, Kanazawa University Hospital, Kanazawa, Japan
| | - Taro Masunari
- Department of Hematology, Chugoku Central Hospital, Fukuyama, Japan
| | - Hiroshi Moro
- Division of Clinical Infection Control and Prevention, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Koji Nikkuni
- Division of Hematology, Niigata City General Hospital, Niigata, Japan
| | - Kazue Takai
- Division of Hematology, Niigata City General Hospital, Niigata, Japan
| | - Masao Hagihara
- Department of Hematology, Eiju General Hospital, Tokyo, Japan
| | - Yuko Hashimoto
- Department of Diagnostic Pathology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Kenji Yokota
- Division of Pathophysiology, Okayama University Graduate School of Health Sciences, Okayama, Japan
| | - Masataka Okamoto
- Department of Hematology and Medical Oncology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Shinji Nakao
- Hematology/Respiratory Medicine Kanazawa University Faculty of Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa, Japan
| | - Tadashi Yoshino
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Yasuharu Sato
- Department of Pathology, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan.,Division of Pathophysiology, Okayama University Graduate School of Health Sciences, Okayama, Japan
| |
Collapse
|
40
|
Beauchemin H, Shooshtarizadeh P, Vadnais C, Vassen L, Pastore YD, Möröy T. Gfi1b controls integrin signaling-dependent cytoskeleton dynamics and organization in megakaryocytes. Haematologica 2017; 102:484-497. [PMID: 28082345 PMCID: PMC5394960 DOI: 10.3324/haematol.2016.150375] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 01/11/2017] [Indexed: 12/27/2022] Open
Abstract
Mutations in GFI1B are associated with inherited bleeding disorders called GFI1B-related thrombocytopenias. We show here that mice with a megakaryocyte-specific Gfi1b deletion exhibit a macrothrombocytopenic phenotype along a megakaryocytic dysplasia reminiscent of GFI1B-related thrombocytopenia. GFI1B deficiency increases megakaryocyte proliferation and affects their ploidy, but also abrogates their responsiveness towards integrin signaling and their ability to spread and reorganize their cytoskeleton. Gfi1b-null megakaryocytes are also unable to form proplatelets, a process independent of integrin signaling. GFI1B-deficient megakaryocytes exhibit aberrant expression of several components of both the actin and microtubule cytoskeleton, with a dramatic reduction of α-tubulin. Inhibition of FAK or ROCK, both important for actin cytoskeleton organization and integrin signaling, only partially restored their response to integrin ligands, but the inhibition of PAK, a regulator of the actin cytoskeleton, completely rescued the responsiveness of Gfi1b-null megakaryocytes to ligands, but not their ability to form proplatelets. We conclude that Gfi1b controls major functions of megakaryocytes such as integrin-dependent cytoskeleton organization, spreading and migration through the regulation of PAK activity whereas the proplatelet formation defect in GFI1B-deficient megakaryocytes is due, at least partially, to an insufficient α-tubulin content.
Collapse
Affiliation(s)
| | | | - Charles Vadnais
- Institut de Recherches Cliniques de Montréal, IRCM, QC, Canada
| | - Lothar Vassen
- Institut de Recherches Cliniques de Montréal, IRCM, QC, Canada
| | - Yves D Pastore
- Département de Pédiatrie, Service d'Hématologie et Oncologie, CHU Ste-Justine, Montréal, QC, Canada
| | - Tarik Möröy
- Institut de Recherches Cliniques de Montréal, IRCM, QC, Canada .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, QC, Canada.,Division of Experimental Medicine, McGill University, Montréal, QC, Canada
| |
Collapse
|
41
|
Nishimori M, Tsunemine H, Maruoka H, Itoh K, Kodaka T, Matsuoka H, Takahashi T. Marked Thrombocytosis in Chronic Eosinophilic Pneumonia and Analysis of Cytokine Mechanism. J Clin Exp Hematop 2016; 55:97-102. [PMID: 26490522 DOI: 10.3960/jslrt.55.97] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
A 47-year-old woman with marked thrombocytosis of 1,650 × 10(9)/L was diagnosed with chronic eosinophilic pneumonia (CEP) based on imaging of the lung and abundant eosinophils in bronchoalveolar lavage fluid. Known gene abnormalities that cause eosinophilia were not detected in bone marrow cells. Treatment with oral prednisolone at 20 mg/day relieved the CEP and resolved the laboratory abnormalities, including eosinophilia and thrombocytosis. Serum concentrations of interleukin (IL)-5 and IL-6 were elevated to 9.6 and 14.0 pg/mL, respectively. The megakaryocyte-potentiating activity of IL-6 and possibly, that of IL-1β, which is known to be secreted by activated eosinophils, may have caused the marked thrombocytosis in this patient.
Collapse
Affiliation(s)
- Makoto Nishimori
- Departments of Hematology Kobe City Medical Center General Hospital
| | | | | | | | | | | | | |
Collapse
|
42
|
Hammond WP, Wun T, Kaplan A, Kaplan S, Paglieroni T, Kaushansky K, Foster DC. High Concentrations of Thrombopoietin Activate Platelets In Vitro. Clin Appl Thromb Hemost 2016. [DOI: 10.1177/107602969800400306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Thrombopoietin (TPO), the ligand for the proto- oncogene c-mpl, has been cloned and expressed from both human and murine sources. Thrombopoietin increases platelet counts when given in vivo and acts on progenitor cells to in crease their proliferation and maturation into megakaryocytes. The effects of TPO on mature platelets were investigated by evaluating platelet aggregation and platelet activation- dependent antigen expression. Platelet aggregation score, a quantitative representation of aggregation, showed potentiation of response to ADP-induced aggregation but no direct agonist response to TPO alone. Soluble c-mpl blocked the effect of TPO on the platelet aggregation score. Flow cytometry showed that TPO at concentrations >250 U/ml (50 ng/ml) caused a minority population of platelets to express the activation mark ers CD62, CD63 and activated glycoprotein IIb/IIIa. While stem cell factor and interleukins-3 and -6 did not affect platelet activation antigen expression, interleukin-11 increased CD62 expression on platelets in vitro. The effects of TPO on antigenic expression and aggregability were partially inhibited in vitro by preincubation with aspirin. We conclude that high concentra tions of TPO promote platelet activation antigen expression on a proportion of platelets and potentiate platelet aggregability to ADP in vitro by a process that is partially inhibited by aspirin.
Collapse
Affiliation(s)
- William P. Hammond
- Departments of Chemistry/Hematology and Molecular Biology and Division of Applied Mathematics and Engineering, The Hope Heart Institute, Providence Seattle Medical Center, Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle
| | - Theodore Wun
- Sacramento Medical Foundation Center for Blood Research, the University of California, Davis Cancer Center, Sacramento, California
| | - Alexander Kaplan
- Departments of Chemistry/Hematology and Molecular Biology and Division of Applied Mathematics and Engineering, The Hope Heart Institute, Providence Seattle Medical Center
| | - Svetlana Kaplan
- Departments of Chemistry/Hematology and Molecular Biology and Division of Applied Mathematics and Engineering, The Hope Heart Institute, Providence Seattle Medical Center
| | - Teresa Paglieroni
- Sacramento Medical Foundation Center for Blood Research, the University of California, Davis Cancer Center, Sacramento, California
| | - Kenneth Kaushansky
- Division of Hematology, Department of Medicine, University of Washington School of Medicine, Seattle
| | | |
Collapse
|
43
|
Bone marrow niche in immune thrombocytopenia: a focus on megakaryopoiesis. Ann Hematol 2016; 95:1765-76. [PMID: 27236577 DOI: 10.1007/s00277-016-2703-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Accepted: 05/23/2016] [Indexed: 12/18/2022]
Abstract
Immune thrombocytopenia (ITP) is an autoimmune disorder characterized by increased bleeding tendency and thrombocytopenia. In fact, the precise pathogenesis of this disease is still not clear. Megakaryopoiesis involves complete differentiation of megakaryocyte (MK) progenitors to functional platelets. This complex process occurs in specific bone marrow (BM) niches composed of several hematopoietic and non-hematopoietic cell types, soluble factors, and extracellular matrix proteins. These specialized microenvironments sustain MK maturation and localization to sinusoids as well as platelet release into circulation. However, MKs in ITP patients show impaired maturation and signs of degradation. Intrinsic defects in MKs and their extrinsic environment have been implicated in altered megakaryopoiesis in this disease. In particular, aberrant expression of miRNAs directing MK proliferation, differentiation, and platelet production; defective MK apoptosis; and reduced proliferation and differentiation rate of the MSC compartment observed in these patients may account for BM defects in ITP. Furthermore, insufficient production of thrombopoietin is another likely reason for ITP development. Therefore, identifying the signaling pathways and transcription factors influencing the interaction between MKs and BM niche in ITP patients will contribute to increased platelet production in order to prevent incomplete MK maturation and destruction as well as BM fibrosis and apoptosis in ITP. In this review, we will examine the interaction and role of BM niches in orchestrating megakaryopoiesis in ITP patients and discuss how these factors can be exploited to improve the quality of patient treatment and prognosis.
Collapse
|
44
|
|
45
|
Iwaki N, Fajgenbaum DC, Nabel CS, Gion Y, Kondo E, Kawano M, Masunari T, Yoshida I, Moro H, Nikkuni K, Takai K, Matsue K, Kurosawa M, Hagihara M, Saito A, Okamoto M, Yokota K, Hiraiwa S, Nakamura N, Nakao S, Yoshino T, Sato Y. Clinicopathologic analysis of TAFRO syndrome demonstrates a distinct subtype of HHV-8-negative multicentric Castleman disease. Am J Hematol 2016; 91:220-6. [PMID: 26805758 DOI: 10.1002/ajh.24242] [Citation(s) in RCA: 206] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/05/2015] [Accepted: 11/10/2015] [Indexed: 12/22/2022]
Abstract
Multicentric Castleman disease (MCD) describes a heterogeneous group of disorders involving systemic inflammation, characteristic lymph node histopathology, and multi-organ dysfunction because of pathologic hypercytokinemia. Whereas Human Herpes Virus-8 (HHV-8) drives the hypercytokinemia in a cohort of immunocompromised patients, the etiology of HHV-8-negative MCD is idiopathic (iMCD). Recently, a limited series of iMCD cases in Japan sharing a constellation of clinical features, including thrombocytopenia (T), anasarca (A), fever (F), reticulin fibrosis (R), and organomegaly (O) has been described as TAFRO syndrome. Herein, we report clinicopathological findings on 25 patients (14 males and 11 females; 23 Japanese-born and two US-born), the largest TAFRO syndrome case series, including the first report of cases from the USA. The median age of onset was 50 years old (range: 23-72). The frequency of each feature was as follows: thrombocytopenia (21/25), anasarca (24/25), fever (21/25), organomegaly (25/25), and reticulin fibrosis (13/16). These patients frequently demonstrated abdominal pain, elevated serum alkaline phosphatase levels, and acute kidney failure. Surprisingly, none of the cases demonstrated marked hypergammoglobulinemia, which is frequently reported in iMCD. Lymph node biopsies revealed atrophic germinal centers with enlarged nuclei of endothelial cells and proliferation of endothelial venules in interfollicular zone. 23 of 25 cases were treated initially with corticosteroids; 12 patients responded poorly and required further therapy. Three patients died during the observation period (median: 9 months) because of disease progression or infections. TAFRO syndrome is a unique subtype of iMCD that demonstrates characteristic clinicopathological findings. Further study to clarify prognosis, pathophysiology, and appropriate treatment is needed.
Collapse
Affiliation(s)
- Noriko Iwaki
- Department of Pathology; Okayama University Graduate School of Medicine, Dentistry and, Pharmaceutical Sciences; Okayama Japan
- Department of Cellular Transplantation Biology (Hematology/Oncology and Respiratory Medicine), Division of Cancer Medicine; Graduate School of Medical Sciences Kanazawa University; Kanazawa Japan
| | - David C. Fajgenbaum
- Department of Medicine, Division of Hematology & Oncology, Raymond & Ruth Perelman School of Medicine; University of Pennsylvania; Philadelphia Pennsylvania
| | - Christopher S. Nabel
- Department of Medicine, Division of Hematology & Oncology, Raymond & Ruth Perelman School of Medicine; University of Pennsylvania; Philadelphia Pennsylvania
| | - Yuka Gion
- Department of Pathology; Okayama University Graduate School of Medicine, Dentistry and, Pharmaceutical Sciences; Okayama Japan
| | - Eisei Kondo
- Department of General Medicine; Okayama University Graduate School of Medicine, Dentistry and, Pharmaceutical Sciences; Okayama Japan
| | - Mitsuhiro Kawano
- Division of Rheumatology; Kanazawa University Hospital; Kanazawa Japan
| | - Taro Masunari
- Department of Hematology; Chugoku Central Hospital; Fukuyama Japan
| | - Isao Yoshida
- Department of Hematologic Oncology; National Hospital Organization Shikoku Cancer Center; Matsuyama Japan
| | - Hiroshi Moro
- Division of Clinical Infection Control and Prevention; Niigata University Graduate School of Medical and Dental Sciences; Niigata Japan
| | - Koji Nikkuni
- Division of Hematology; Niigata City General Hospital; Niigata Japan
| | - Kazue Takai
- Division of Hematology; Niigata City General Hospital; Niigata Japan
| | - Kosei Matsue
- Division of Hematology and Oncology, Department of Medicine; Kameda Medical Center; Kamogawa Japan
| | - Mitsutoshi Kurosawa
- Department of Hematology; National Hospital Organization Hokkaido Cancer Center; Sapporo Japan
| | - Masao Hagihara
- Department of Hematology; Eiju General Hospital; Tokyo Japan
| | - Akio Saito
- Department of Hematology; National Hospital Organization Nishigunma National Hospital; Shibukawa Japan
| | - Masataka Okamoto
- Department of Hematology and Medical Oncology; Fujita Health University School of Medicine; Toyoake Japan
| | - Kenji Yokota
- Division of Pathophysiology; Okayama University Graduate School of Health Sciences; Okayama Japan
| | - Shinichiro Hiraiwa
- Department of Pathology; Tokai University School of Medicine; Kanagawa Japan
| | - Naoya Nakamura
- Department of Pathology; Tokai University School of Medicine; Kanagawa Japan
| | - Shinji Nakao
- Cellular Transplantation Biology; Division of Medicine, Kanazawa University Institutes of Medical, Pharmaceutical, and Health Sciences; Kanazawa Japan
| | - Tadashi Yoshino
- Department of Pathology; Okayama University Graduate School of Medicine, Dentistry and, Pharmaceutical Sciences; Okayama Japan
| | - Yasuharu Sato
- Department of Pathology; Okayama University Graduate School of Medicine, Dentistry and, Pharmaceutical Sciences; Okayama Japan
- Division of Pathophysiology; Okayama University Graduate School of Health Sciences; Okayama Japan
| |
Collapse
|
46
|
Schumacher A, Denecke B, Braunschweig T, Stahlschmidt J, Ziegler S, Brandenburg LO, Stope MB, Martincuks A, Vogt M, Görtz D, Camporeale A, Poli V, Müller-Newen G, Brümmendorf TH, Ziegler P. Angptl4 is upregulated under inflammatory conditions in the bone marrow of mice, expands myeloid progenitors, and accelerates reconstitution of platelets after myelosuppressive therapy. J Hematol Oncol 2015; 8:64. [PMID: 26054961 PMCID: PMC4460974 DOI: 10.1186/s13045-015-0152-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 05/07/2015] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Upon inflammation, myeloid cell generation in the bone marrow (BM) is broadly enhanced by the action of induced cytokines which are produced locally and at multiple sites throughout the body. METHODS Using microarray studies, we found that Angptl4 is upregulated in the BM during systemic inflammation. RESULTS Recombinant murine Angptl4 (rmAngptl4) stimulated the proliferation of myeloid colony-forming units (CFUs) in vitro. Upon repeated in vivo injections, rmAngptl4 increased BM progenitor cell frequency and this was paralleled by a relative increase in phenotypically defined granulocyte-macrophage progenitors (GMPs). Furthermore, in vivo treatment with rmAngptl4 resulted in elevated platelet counts in steady-state mice while allowing a significant acceleration of reconstitution of platelets after myelosuppressive therapy. The administration of rmAngptl4 increased the number of CD61(+)CD41(low)-expressing megakaryocytes (MK) in the BM of steady-state and in the spleen of transplanted mice. Furthermore, rmAngptl4 improved the in vitro differentiation of immature MKs from hematopoietic stem and progenitor cells. Mechanistically, using a signal transducer and activator of transcription 3 (STAT3) reporter knockin model, we show that rmAngptl4 induces de novo STAT3 expression in immature MK which could be important for the effective expansion of MKs after myelosuppressive therapy. CONCLUSION Whereas the definitive role of Angptl4 in mediating the effects of lipopolysaccharide (LPS) on the BM has to be demonstrated by further studies involving multiple cytokine knockouts, our data suggest that Angptl4 plays a critical role during hematopoietic, especially megakaryopoietic, reconstitution following stem cell transplantation.
Collapse
Affiliation(s)
- Anne Schumacher
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research IZKF Aachen, RWTH Aachen University Hospital, Aachen, Germany.
| | - Till Braunschweig
- Institute of Pathology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Jasmin Stahlschmidt
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Susanne Ziegler
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Lars-Ove Brandenburg
- Department of Anatomy and Cell Biology, RWTH Aachen University, Wendlingweg 2, 52074, Aachen, Germany.
| | - Matthias B Stope
- Department of Urology, University Medicine Greifswald, Greifswald, Germany.
| | - Antons Martincuks
- Department of Biochemistry and Molecular Biology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Michael Vogt
- Institute for Laboratory Animal Science, University Hospital, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Dieter Görtz
- Department of Biochemistry and Molecular Biology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Annalisa Camporeale
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10126, Turin, Italy.
| | - Valeria Poli
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, 10126, Turin, Italy.
| | - Gerhard Müller-Newen
- Department of Biochemistry and Molecular Biology, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Tim H Brümmendorf
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
| | - Patrick Ziegler
- Department of Oncology, Hematology and Stem Cell Transplantation, University Hospital Aachen, RWTH Aachen University, Pauwelsstrasse 30, 52074, Aachen, Germany.
- Institute for Occupational and Social Medicine, Aachen University, Aachen, Germany.
| |
Collapse
|
47
|
Malara A, Abbonante V, Di Buduo CA, Tozzi L, Currao M, Balduini A. The secret life of a megakaryocyte: emerging roles in bone marrow homeostasis control. Cell Mol Life Sci 2015; 72:1517-36. [PMID: 25572292 PMCID: PMC4369169 DOI: 10.1007/s00018-014-1813-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 12/15/2014] [Accepted: 12/19/2014] [Indexed: 12/19/2022]
Abstract
Megakaryocytes are rare cells found in the bone marrow, responsible for the everyday production and release of millions of platelets into the bloodstream. Since the discovery and cloning, in 1994, of their principal humoral factor, thrombopoietin, and its receptor c-Mpl, many efforts have been directed to define the mechanisms underlying an efficient platelet production. However, more recently different studies have pointed out new roles for megakaryocytes as regulators of bone marrow homeostasis and physiology. In this review we discuss the interaction and the reciprocal regulation of megakaryocytes with the different cellular and extracellular components of the bone marrow environment. Finally, we provide evidence that these processes may concur to the reconstitution of the bone marrow environment after injury and their deregulation may lead to the development of a series of inherited or acquired pathologies.
Collapse
Affiliation(s)
- Alessandro Malara
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
- Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Vittorio Abbonante
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
- Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Christian A. Di Buduo
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
- Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Lorenzo Tozzi
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA USA
| | - Manuela Currao
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
- Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Via Forlanini 6, 27100 Pavia, Italy
- Laboratory of Biotechnology, IRCCS San Matteo Foundation, Pavia, Italy
- Department of Biomedical Engineering, Tufts University, Medford, MA USA
| |
Collapse
|
48
|
Badimon L, Hernández Vera R, Vilahur G. Atherothrombotic risk in obesity. Hamostaseologie 2014; 33:259-68. [PMID: 24190286 DOI: 10.5482/hamo-13-07-0034] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/02/2013] [Indexed: 12/28/2022] Open
Abstract
A link between obesity and coronary artery disease development has been repeatedly proposed, possibly in part due to the development of a proinflammatory and prothrombotic state in obese subjects. Adipocytes secrete numerous hormones and cytokines (adipokines) which influence gene expression and cell functions in endothelial cells, arterial smooth muscle cells, and monocytes/macrophages favouring the development of an atherosclerotic vulnerable plaque. Moreover, the release of such biologically active molecules also promotes endothelial function impairment, disturbs the haemostatic and fibrinolytic systems, and produces alterations in platelet function affecting the initiation, progression, and stabilization of thrombus formation upon atherosclerotic plaque rupture. In this review we will discuss the pathophysiological mechanisms by which obesity contributes to increase atherothrombosis paying special attention to its effects over thrombosis.
Collapse
Affiliation(s)
- L Badimon
- Prof. Lina Badimon, Cardiovascular Research Center, C/ Sant Antoni Mª Claret 167, 08025 Barcelona, Spain, Tel. +34/93/556 58 80; Fax +34/93/556 55 59, E-mail:
| | | | | |
Collapse
|
49
|
Langdon JM, Yates SC, Femnou LK, McCranor BJ, Cheadle C, Xue QL, Vaulont S, Civin CI, Walston JD, Roy CN. Hepcidin-dependent and hepcidin-independent regulation of erythropoiesis in a mouse model of anemia of chronic inflammation. Am J Hematol 2014; 89:470-9. [PMID: 24415655 PMCID: PMC4200395 DOI: 10.1002/ajh.23670] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 01/02/2014] [Accepted: 01/08/2014] [Indexed: 12/21/2022]
Abstract
Increased hepcidin antimicrobial peptide correlates with hypoferremia and anemia in various disease states, but its requirement for anemia of inflammation has not been adequately demonstrated. Anemia of inflammation is usually described as normocytic and normochromic, while diseases associated with over expression of hepcidin, alone, are often microcytic and hypochromic. These differences in erythrocyte parameters suggest anemia in many inflammatory states may not be fully explained by hepcidin-mediated iron sequestration. We used turpentine-induced sterile abscesses to model chronic inflammation in mice with targeted disruption of Hepcidin 1 [Hepc1 (-/-)] or its positive regulator, Interleukin-6 [IL-6 (-/-)], to determine whether these genes are required for features characteristic of anemia of inflammation. Although hemoglobin levels did not decline in Hepc1 (-/-) mice with sterile abscesses, erythrocyte numbers were significantly reduced compared to untreated Hepc1 (-/-) mice. In contrast, both hemoglobin concentration and erythrocyte number declined significantly in wild type and IL-6 (-/-) mice with sterile abscesses. Both Hepc1 (-/-) and IL-6 (-/-) mice had increased erythrocyte mean cell volume and mean cell hemoglobin following sterile abscesses, while wild types had no change. Thus, IL-6 (-/-) mice with sterile abscesses exhibit an intermediate phenotype between wild type and Hepc1 (-/-). Our results demonstrate the requirement of Hepc1 for the development of anemia in this rodent model. Simultaneously, our results demonstrate hepcidin-independent effects of inflammation on the suppression of erythropoiesis. Our results suggest chronic anemia associated with inflammation may benefit from interventions protecting erythrocyte number in addition to anti-hepcidin interventions aimed at enhancing iron availability.
Collapse
Affiliation(s)
- Jacqueline M. Langdon
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Saiah C. Yates
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Laurette K. Femnou
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Bryan J. McCranor
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Chris Cheadle
- Lowe Family Genomics Core, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qian-Li Xue
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Sophie Vaulont
- Institut Cochin, Institut National de la Santé et de la Recherche Medicale U1016, Paris, France
| | - Curt I. Civin
- Center for Stem Cell Biology and Regenerative Medicine, Department of Pediatrics, University of Maryland, Baltimore, Maryland
- Center for Stem Cell Biology and Regenerative Medicine, Department of Physiology, University of Maryland, Baltimore, Maryland
| | - Jeremy D. Walston
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Cindy N. Roy
- Division of Geriatric Medicine and Gerontology, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
50
|
Platelet activation and platelet-leukocyte aggregation elicited in experimental colitis are mediated by interleukin-6. Inflamm Bowel Dis 2014; 20:353-62. [PMID: 24390064 PMCID: PMC3947085 DOI: 10.1097/01.mib.0000440614.83703.84] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
There is growing evidence for an interdependence of inflammation, coagulation, and thrombosis in acute and chronic inflammatory diseases. Inflammatory bowel diseases (IBD) are associated with a hypercoagulable state and an increased risk of thromboembolism. Although the IBD-associated prothrombogenic state has been linked to the inflammatory response, the mediators that link these 2 conditions remain unclear. Recent evidence suggests that interleukin-6 (IL-6) may be important in this regard. The objective of this study was to more fully define the contribution of IL-6 to the altered platelet function that occurs during experimental colitis. The number of immature and mature platelets, activated platelets, and platelet-leukocyte aggregates were measured in wild-type and IL-6 mice with dextran sodium sulfate (DSS)-induced colonic inflammation. DSS treatment of WT mice was associated with significant increases in the number of both immature and mature platelets, activated platelets, and platelet-leukocyte aggregates. These platelet responses to DSS were not observed in IL-6 mice. Chronic IL-6 infusion (through an Alzet pump) in WT mice reproduced all of the platelet abnormalities observed in DSS-colitic mice. IL-6-infused mice also exhibited an acceleration of thrombus formation in arterioles, similar to DSS. These findings implicate IL-6 in the platelet activation and enhanced platelet-leukocyte aggregate formation associated with experimental colitis, and support a role for this cytokine as a mediator of the enhanced thrombogenesis in IBD.
Collapse
|