1
|
Caetano J, Rodrigues LM, Alves JD. What have we learned on pre, very early, and early systemic sclerosis microcirculatory pathophysiology? A scoping review. Autoimmun Rev 2024; 23:103540. [PMID: 38604463 DOI: 10.1016/j.autrev.2024.103540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/03/2024] [Accepted: 04/03/2024] [Indexed: 04/13/2024]
Abstract
OBJECTIVE Microvascular dysfunction is an early event in the pathogenesis of systemic sclerosis (SSc). The objective of this scoping review is to update the current information and the level of knowledge about the mechanisms of microvascular dysfunction in pre-SSc, very early diagnosis of SSc (VEDOSS) and early SSc. METHODS A PubMed® database search allowed us to include original data from full-length articles in English in which the main topic was microvascular dysfunction in pre-SSC, VEDOSS or early SSc. Data was extracted using a customized form. RESULTS In the present review 437 articles were identified, and 42 studies included, reporting data from a total of 1069 patients with pre-SSc, VEDOSS or early-SSc. Distinct mechanisms of microvascular injury were identified comprising, angiogenesis and vasculogenesis, cell surface proteins and adhesion, molecules expression, cytokines profile, inflammatory and oxidation pathways, and skin perfusion determinants. Most of the studies were conducted in early SSc, with a reduced number in pre-disease stages, in which the prompt recognition of specific mechanisms and biomarkers may allow targeted treatment to prevent disease progression. CONCLUSIONS Although different molecular expression patterns and signaling pathways related to microvascular dysfunction in pre-SSc, VEDOSS, and early SSc were identified, additional prospective longitudinal studies and combined work with functional evaluation of peripheral skin perfusion are needed.
Collapse
Affiliation(s)
- Joana Caetano
- CBIOS-Research Center for Biosciences and Health Technologies, Universidade Lusófona Lisboa, 1749-024 Lisbon, Portugal; Systemic Autoimmune Diseases Unit, Department of Medicine IV, Fernando Fonseca Hospital, 2720-276 Amadora, Portugal; Nova Medical School, 1169-056 Lisboa, Portugal.
| | - Luís Monteiro Rodrigues
- CBIOS-Research Center for Biosciences and Health Technologies, Universidade Lusófona Lisboa, 1749-024 Lisbon, Portugal
| | - José Delgado Alves
- Systemic Autoimmune Diseases Unit, Department of Medicine IV, Fernando Fonseca Hospital, 2720-276 Amadora, Portugal; Nova Medical School, 1169-056 Lisboa, Portugal.
| |
Collapse
|
2
|
Jones OY, McCurdy D. Cell Based Treatment of Autoimmune Diseases in Children. Front Pediatr 2022; 10:855260. [PMID: 35615628 PMCID: PMC9124972 DOI: 10.3389/fped.2022.855260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/14/2022] [Indexed: 11/28/2022] Open
Abstract
Mesenchymal stem cells have recently been recoined as medicinal signaling cells (MSC) for their ability to promote tissue homeostasis through immune modulation, angiogenesis and tropism. During the last 20 years, there has been a plethora of publications using MSC in adults and to lesser extent neonates on a variety of illnesses. In parts of the world, autologous and allogeneic MSCs have been purified and used to treat a range of autoimmune conditions, including graft versus host disease, Crohn's disease, multiple sclerosis, refractory systemic lupus erythematosus and systemic sclerosis. Generally, these reports are not part of stringent clinical trials but are of note for good outcomes with minimal side effects. This review is to summarize the current state of the art in MSC therapy, with a brief discussion of cell preparation and safety, insights into mechanisms of action, and a review of published reports of MSC treatment of autoimmune diseases, toward the potential application of MSC in treatment of children with severe autoimmune diseases using multicenter clinical trials and treatment algorithms.
Collapse
Affiliation(s)
- Olcay Y. Jones
- Division of Pediatric Rheumatology, Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, MD, United States
| | - Deborah McCurdy
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
3
|
Höppner J, Bruni C, Distler O, Robson SC, Burmester GR, Siegert E, Distler JHW. Purinergic signaling in systemic sclerosis. Rheumatology (Oxford) 2021; 61:2770-2782. [PMID: 34849624 DOI: 10.1093/rheumatology/keab859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 11/13/2022] Open
Abstract
Systemic sclerosis (SSc) is a chronic autoimmune rheumatic disease that involves numerous organs and presents major management challenges. The histopathologic hallmarks of SSc include vasculopathy, fibrosis and autoimmune phenomena involving both innate and adaptive immune systems. Purinergic signalling is a pathway that may be implicated in the pathophysiology of several of these disease manifestations. Extracellular purines are potent signalling mediators, which have been shown to be dysregulated in SSc. As examples, purines can exacerbate vasculopathy and provoke platelet dysfunction; as well as contributing to immune dysregulation. Elements of purinergic signalling further promote organ and tissue fibrosis in several disease models. Here, we provide an overview of extracellular purine metabolism in purinergic signalling and link disorders of these to the molecular pathology of SSc. We also discuss targeting the purinergic signalling and explore the translational applications for new therapeutic options in SSc.
Collapse
Affiliation(s)
- Jakob Höppner
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, Division of Rheumatology, Careggi University Hospital, University of Florence, Florence, Italy.,Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Oliver Distler
- Department of Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Simon C Robson
- Departments of Anesthesia and Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Gerd R Burmester
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Elise Siegert
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Jörg H W Distler
- Department of Internal Medicine 3-Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and University Hospital Erlangen, Erlangen, Germany
| |
Collapse
|
4
|
Pulito-Cueto V, Remuzgo-Martínez S, Genre F, Atienza-Mateo B, Mora-Cuesta VM, Iturbe-Fernández D, Lera-Gómez L, Pérez-Fernández R, Prieto-Peña D, Portilla V, Blanco R, Corrales A, Gualillo O, Cifrián JM, López-Mejías R, González-Gay MA. Endothelial Progenitor Cells: Relevant Players in the Vasculopathy and Lung Fibrosis Associated with the Presence of Interstitial Lung Disease in Systemic Sclerosis Patients. Biomedicines 2021; 9:biomedicines9070847. [PMID: 34356910 PMCID: PMC8301775 DOI: 10.3390/biomedicines9070847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/16/2021] [Accepted: 07/19/2021] [Indexed: 12/16/2022] Open
Abstract
Endothelial progenitor cells (EPC), which are key effectors in the physiologic vascular network, have been described as relevant players in autoimmune diseases. We previously showed that EPC frequency may help to identify the presence of interstitial lung disease (ILD) in rheumatoid arthritis patients. Given that ILD constitutes the main cause of mortality in systemic sclerosis (SSc) patients, we aimed to determine the EPC contribution to the pathogenic processes of vasculopathy and lung fibrosis in SSc-ILD+. EPC quantification was performed by flow cytometry on blood from 83 individuals: 21 SSc-ILD+ patients and subjects from comparative groups (20 SSc-ILD− and 21 idiopathic pulmonary fibrosis (IPF) patients and 21 healthy controls (HC)). EPC were considered as CD34+, CD45low, CD309+, and CD133+. A significant increase in EPC frequency was found in SSc-ILD+ patients when compared to HC (p < 0.001). SSc-ILD+ patients exhibited a higher EPC frequency than SSc-ILD− patients (p = 0.012), whereas it was markedly reduced compared to IPF patients (p < 0.001). EPC frequency was higher in males (p = 0.04) and negatively correlated to SSc duration (p = 0.04) in SSc-ILD+ patients. Our results indicate a role of EPC in the processes of vasculopathy and lung fibrosis in SSc-ILD+. EPC frequency may be considered as a biomarker of ILD in SSc patients.
Collapse
Affiliation(s)
- Verónica Pulito-Cueto
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Sara Remuzgo-Martínez
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Fernanda Genre
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Belén Atienza-Mateo
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- López Albo’ Post-Residency Programme, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Víctor M. Mora-Cuesta
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - David Iturbe-Fernández
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Leticia Lera-Gómez
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Raquel Pérez-Fernández
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Diana Prieto-Peña
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Virginia Portilla
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Ricardo Blanco
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Alfonso Corrales
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
| | - Oreste Gualillo
- Servizo Galego de Saude and Instituto de Investigación Sanitaria, Hospital Clinico Universitario de Santiago, 15706 Santiago de Compostela, Spain;
| | - José M. Cifrián
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
- School of Medicine, Universidad de Cantabria, 39011 Santander, Spain
| | - Raquel López-Mejías
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Correspondence: (R.L.-M.); (M.A.G.-G.); Tel.: +34-942-315-515 (R.L.-M. & M.A.G.-G.); Fax: +34-942-31-55-17 (R.L.-M. & M.A.G.-G.)
| | - Miguel A. González-Gay
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, 39011 Santander, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (B.A.-M.); (V.M.M.-C.); (D.I.-F.); (L.L.-G.); (R.P.-F.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, 39008 Santander, Spain
- School of Medicine, Universidad de Cantabria, 39011 Santander, Spain
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2050, South Africa
- Correspondence: (R.L.-M.); (M.A.G.-G.); Tel.: +34-942-315-515 (R.L.-M. & M.A.G.-G.); Fax: +34-942-31-55-17 (R.L.-M. & M.A.G.-G.)
| |
Collapse
|
5
|
Pulito-Cueto V, Remuzgo-Martínez S, Genre F, Mora-Cuesta VM, Iturbe-Fernández D, Fernández-Rozas S, Atienza-Mateo B, Lera-Gómez L, Alonso-Lecue P, Rodríguez-Carrio J, Prieto-Peña D, Portilla V, Blanco R, Corrales A, Gualillo O, Cifrián JM, López-Mejías R, González-Gay MA. Endothelial Progenitor Cells as a Potential Biomarker in Interstitial Lung Disease Associated with Rheumatoid Arthritis. J Clin Med 2020; 9:jcm9124098. [PMID: 33353104 PMCID: PMC7766338 DOI: 10.3390/jcm9124098] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/09/2020] [Accepted: 12/14/2020] [Indexed: 12/16/2022] Open
Abstract
Interstitial lung disease (ILD) increases morbidity and mortality in patients with rheumatoid arthritis (RA). Although the pathogenesis of ILD associated with RA (RA-ILD+) remains poorly defined, vascular tissue is crucial in lung physiology. In this context, endothelial progenitor cells (EPC) are involved in endothelial tissue repair. However, little is known about their implication in RA-ILD+. Accordingly, we aimed to investigate the potential role of EPC related to endothelial damage in RA-ILD+. EPC quantification in peripheral blood from 80 individuals (20 RA-ILD+ patients, 25 RA-ILD- patients, 21 idiopathic pulmonary fibrosis (IPF) patients, and 14 healthy controls) was performed by flow cytometry. EPC were considered as CD34+, CD45low, CD309+ and CD133+. A significant increase in EPC frequency in RA-ILD+ patients, as well as in RA-ILD- and IPF patients, was found when compared with controls (p < 0.001, p = 0.02 and p < 0.001, respectively). RA-ILD+ patients exhibited a higher EPC frequency than the RA-ILD- ones (p = 0.003), but lower than IPF patients (p < 0.001). Our results suggest that EPC increase may represent a reparative compensatory mechanism in patients with RA-ILD+. The degree of EPC frequency may help to identify the presence of ILD in RA patients and to discriminate RA-ILD+ from IPF.
Collapse
Affiliation(s)
- Verónica Pulito-Cueto
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Sara Remuzgo-Martínez
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Fernanda Genre
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Víctor M. Mora-Cuesta
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - David Iturbe-Fernández
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - Sonia Fernández-Rozas
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - Belén Atienza-Mateo
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- López Albo’ Post-Residency Programme, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - Leticia Lera-Gómez
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
| | - Pilar Alonso-Lecue
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - Javier Rodríguez-Carrio
- Department of Functional Biology, Immunology Area, Faculty of Medicine, Universidad de Oviedo, Oviedo, 33006 Asturias, Spain;
| | - Diana Prieto-Peña
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - Virginia Portilla
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - Ricardo Blanco
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - Alfonso Corrales
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
| | - Oreste Gualillo
- SERGAS (Servizo Galego de Saude) and IDIS (Instituto de Investigación Sanitaria de Santiago), NEIRID Lab (Neuroendocrine Interactions in Rheumatology and Inflammatory Diseases), Research Laboratory 9, Santiago University Clinical Hospital, Santiago de Compostela, 15706 A Coruña, Spain;
| | - José M. Cifrián
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Pneumology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
- School of Medicine, Universidad de Cantabria, Santander, 39005 Cantabria, Spain
| | - Raquel López-Mejías
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Correspondence: (R.L.-M.); (M.A.G.-G.); Tel.: +34-942-315-515 (R.L.-M. & M.A.G.-G.); Fax: +34-942-31-55-17 (R.L.-M. & M.A.G.-G.)
| | - Miguel A. González-Gay
- Research Group on Genetic Epidemiology and Atherosclerosis in Systemic Diseases and in Metabolic Bone Diseases of the Musculoskeletal System, IDIVAL, Santander, 39011 Cantabria, Spain; (V.P.-C.); (S.R.-M.); (F.G.); (V.M.M.-C.); (D.I.-F.); (S.F.-R.); (B.A.-M.); (L.L.-G.); (P.A.-L.); (D.P.-P.); (V.P.); (R.B.); (A.C.); (J.M.C.)
- Department of Rheumatology, Hospital Universitario Marqués de Valdecilla, Santander, 39008 Cantabria, Spain
- School of Medicine, Universidad de Cantabria, Santander, 39005 Cantabria, Spain
- Cardiovascular Pathophysiology and Genomics Research Unit, School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg 2050, South Africa
- Correspondence: (R.L.-M.); (M.A.G.-G.); Tel.: +34-942-315-515 (R.L.-M. & M.A.G.-G.); Fax: +34-942-31-55-17 (R.L.-M. & M.A.G.-G.)
| |
Collapse
|
6
|
Del Papa N, Pignataro F. The Role of Endothelial Progenitors in the Repair of Vascular Damage in Systemic Sclerosis. Front Immunol 2018; 9:1383. [PMID: 29967618 PMCID: PMC6015881 DOI: 10.3389/fimmu.2018.01383] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Accepted: 06/04/2018] [Indexed: 01/17/2023] Open
Abstract
Systemic sclerosis (SSc) is a connective tissue disease characterized by a complex pathological process where the main scenario is represented by progressive loss of microvascular bed, with the consequent progressive fibrotic changes in involved organ and tissues. Although most aspects of vascular injury in scleroderma are poorly understood, recent data suggest that the scleroderma impairment of neovascularization could be related to both angiogenesis and vasculogenesis failure. Particularly, compensatory angiogenesis does not occur normally in spite of an important increase in many angiogenic factors either in SSc skin or serum. Besides insufficient angiogenesis, the contribution of defective vasculogenesis to SSc vasculopathy has been extensively studied. Over the last decades, our understanding of the processes responsible for the formation of new vessels after tissue ischemia has increased. In the past, adult neovascularization was thought to depend mainly on angiogenesis (a process by which new vessels are formed by the proliferation and migration of mature endothelial cells). More recently, increased evidence suggests that stem cells mobilize from the bone marrow into the peripheral blood (PB), differentiate in circulating endothelial progenitors (EPCs), and home to site of ischemia to contribute to de novo vessel formation. Significant advances have been made in understanding the biology of EPCs, and molecular mechanisms regulating EPC function. Autologous EPCs now are becoming a novel treatment option for therapeutic vascularization and vascular repair, mainly in ischemic diseases. However, different diseases, such as cardiovascular diseases, diabetes, and peripheral artery ischemia are related to EPC dysfunction. Several studies have shown that EPCs can be detected in the PB of patients with SSc and are impaired in their function. Based on an online literature search (PubMed, EMBASE, and Web of Science, last updated December 2017) using keywords related to “endothelial progenitor cells” and “Systemic Sclerosis,” “scleroderma vasculopathy,” “angiogenesis,” “vasculogenesis,” this review gives an overview on the large body of data of current research in this issue, including controversies over the identity and functions of EPCs, their meaning as biomarker of SSc microangiopathy and their clinical potency.
Collapse
|
7
|
The Assessment of Serum Endocan Levels in Children With Juvenile Idiopathic Arthritis. Arch Rheumatol 2017; 33:168-173. [PMID: 30207559 DOI: 10.5606/archrheumatol.2018.6528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 08/31/2017] [Indexed: 02/03/2023] Open
Abstract
Objectives This study aims to evaluate the levels of serum endocan in children with juvenile idiopathic arthritis (JIA). Patients and methods Sixty-seven children with JIA (30 males, 37 females; mean age 10.4±4.9 years; range 2 to 18 years) and a sex- and age- matched healthy control group of 39 children (16 males, 23 females; mean age 9.3±4.1 years; range 1 to 17 years) were recruited. Patients with JIA were divided into two groups as the clinically active JIA group (n=27) and inactive JIA group (n=40). Results The median serum endocan level in patients with JIA was significantly higher than in the control group (633.75 ng/L vs. 379.76 ng/L, p<0.01). Comparison between patients with active JIA and inactive JIA was not significant in terms of endocan levels (618.70 ng/L vs. 687.36 ng/L, p=0.34). There was a weak negative correlation between Childhood Health Assessment Questionnaire scores of patients with JIA and serum endocan levels. Conclusion The high level of serum endocan highlighted the endothelial damage in patients with JIA.
Collapse
|
8
|
No Significant Reduction of Circulating Endothelial-Derived and Platelet-Derived Microparticles in Patients with Psoriasis Successfully Treated with Anti-IL12/23. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3242143. [PMID: 27144162 PMCID: PMC4842038 DOI: 10.1155/2016/3242143] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/17/2016] [Accepted: 03/20/2016] [Indexed: 12/20/2022]
Abstract
Psoriasis is associated with atherosclerosis, in which circulating microparticles play an important role. In severe psoriasis, there was an increase of endothelial- and platelet- microparticles which could be decreased by anti-TNFα. However, whether anti-IL-12/23 treatment would decrease the level of microparticles remains unknown. Our study showed that, despite the clinical improvement of psoriasis after IL-12/13 blockage, the increased levels of circulating CD41a and CD31 microparticles were unchanged after anti-IL-12/23. This result suggested that anti-IL12/23 treatment may not alter the development of cardiovascular disease in patients with psoriasis.
Collapse
|
9
|
Red cell distribution width is associated with endothelial progenitor cell depletion and vascular-related mediators in rheumatoid arthritis. Atherosclerosis 2015; 240:131-6. [DOI: 10.1016/j.atherosclerosis.2015.03.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 03/03/2015] [Accepted: 03/03/2015] [Indexed: 12/31/2022]
|
10
|
Martini G, Biscaro F, Boscaro E, Calabrese F, Lunardi F, Facco M, Agostini C, Zulian F, Fadini GP. Reduced levels of circulating progenitor cells in juvenile idiopathic arthritis are counteracted by anti TNF-α therapy. BMC Musculoskelet Disord 2015; 16:103. [PMID: 25925313 PMCID: PMC4418050 DOI: 10.1186/s12891-015-0555-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 04/16/2015] [Indexed: 02/07/2023] Open
Abstract
Background Endothelial progenitor cells (EPC) promote angiogenesis and vascular repair. Though reduced EPC levels have been shown in rheumatoid arthritis, no study has so far evaluated EPCs in children with juvenile idiopathic arthritis (JIA). We aimed to study circulating EPCs in children with JIA, their relation to disease activity, and effects of anti TNF-α treatment. Methods Circulating EPCs were quantified by flow cytometry based on CD34, CD133 and KDR expression in peripheral blood of 22 patients with oligoarticular JIA and 29 age-matched controls. EPCs were re-assessed in children with methotrexate-resistant oligo-extended JIA before and up to 12 month after initiation of anti-TNF-alpha therapy. Plasma concentrations of inflammatory and EPC-regulating factors were measured using a multiplex array. Confocal immunofluorescence was used to demonstrate EPCs in synovial tissues. Results Children with active JIA showed a significant reduction of relative and absolute counts of circulating progenitor cells and EPCs compared to age-matched healthy controls. CD34+ cell levels were modestly and inversely correlated to disease activity. A strong inverse correlation was found between serum TNF-α and EPC levels. In 8 patients treated with anti TNF-α agents, the number of EPCs rose to values similar to healthy controls. CD34+KDR+ EPCs were found in the synovial tissue of JIA children, but not in control. Conclusions Children with JIA have reduced levels of the vasculoprotective and proangiogenic EPCs. While EPCs may contribute to synovial tissue remodelling, EPC pauperization may indicate an excess cardiovascular risk if projected later in life.
Collapse
Affiliation(s)
- Giorgia Martini
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Francesca Biscaro
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Elisa Boscaro
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Fiorella Calabrese
- Department of Cardiovascular and Thoracic Sciences, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Francesca Lunardi
- Department of Cardiovascular and Thoracic Sciences, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Monica Facco
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Carlo Agostini
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| | - Francesco Zulian
- Paediatric Rheumatology Unit, Department of Paediatrics, 35128, Padova, Italy.
| | - Gian Paolo Fadini
- Department of Medicine, University of Padova, Via Giustiniani, 2, 35128, Padova, Italy.
| |
Collapse
|
11
|
Rusak M, Radzikowska U, Glowinska-Olszewska B, Dobrenko E, Piotrowska-Jastrzebska J, Dabrowska M, Bodzenta-Lukaszyk A, Bossowski A, Moniuszko M. Endothelial progenitor cell levels in juvenile idiopathic arthritis patients: effects of anti-inflammatory therapies. Pediatr Rheumatol Online J 2015; 13:6. [PMID: 25705139 PMCID: PMC4336757 DOI: 10.1186/s12969-015-0001-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 01/27/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Juvenile idiopathic arthritis (JIA), similarly to other arthritides, can be associated with damage of endothelial layer of which structure and function is dependent on reparative properties of endothelial progenitor cells (EPC). To date, it remained unknown whether EPC numbers are altered in young JIA patients and whether on-going anti-inflammatory therapies could exert positive effects on these progenitor cells. METHODS We performed a quantitative analysis of EPC numbers in 25 patients diagnosed with JIA according to International League of Associations for Rheumatism (ILAR) criteria [age 11.50 (7.50-15.00) years] in a broad context of inflammatory and cardiovascular parameters as well as different types of anti-inflammatory treatments. 11 healthy children [age 13.00 (11.00-14.00) years] were recruited as a control group. RESULTS We demonstrated that EPC numbers were similar in JIA patients and control subjects (0.02% vs. 0.05%, respectively, p = 0.37). EPC levels in JIA patients were negatively correlated with index of insulin resistance (rho = -0.458, p = 0.021), endogenous insulin (rho = -0.472, p = 0.017), triglyceride (rho = -0.438, p = 0.029) and TNF-alpha levels (rho = -0.446, p = 0.026). Notably, glucocorticoid (GC) therapy, was associated with detection of decreased EPC levels in JIA patients (p = 0.023). In contrast, methothrexate (MTX) and etanercept therapy in JIA patients did not affect EPC levels (p = 0.92 and p = 0.08, respectively). CONCLUSIONS We found that EPC numbers are maintained at normal levels in JIA patients and are not enhanced by disease-specific anti-inflammatory treatments.
Collapse
Affiliation(s)
- Malgorzata Rusak
- Department of Hematological Diagnostics, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Urszula Radzikowska
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland
| | - Barbara Glowinska-Olszewska
- Department of Pediatrics, Endocrinology, Diabetology with Cardiology Division, Medical University of Bialystok, 15-274 Białystok, Poland
| | - Elzbieta Dobrenko
- Department of Pediatrics and Developmental Disorders, Medical University of Bialystok, 15-274 Białystok, Poland
| | | | - Milena Dabrowska
- Department of Hematological Diagnostics, Medical University of Bialystok, 15-274 Bialystok, Poland
| | - Anna Bodzenta-Lukaszyk
- Department of Allergology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Artur Bossowski
- Department of Pediatrics, Endocrinology, Diabetology with Cardiology Division, Medical University of Bialystok, 15-274 Białystok, Poland
| | - Marcin Moniuszko
- Department of Regenerative Medicine and Immune Regulation, Medical University of Bialystok, 15-269 Bialystok, Poland ,Department of Allergology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| |
Collapse
|
12
|
Andrigueti FV, Arismendi MI, Ebbing PCC, Kayser C. Decreased numbers of endothelial progenitor cells in patients in the early stages of systemic sclerosis. Microvasc Res 2015; 98:82-7. [PMID: 25596148 DOI: 10.1016/j.mvr.2015.01.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 12/12/2014] [Accepted: 01/05/2015] [Indexed: 01/25/2023]
Abstract
INTRODUCTION Microangiopathy and endothelial dysfunction are present in the early stages of systemic sclerosis (SSc). Defective vasculogenesis mediated by bone marrow-derived endothelial progenitor cells (EPCs) might be involved in the vascular abnormalities found in SSc. OBJECTIVES To evaluate the circulating EPC levels and EPC subtypes via flow cytometry and early outgrowth colony-forming units (CFUs) in patients with SSc compared to healthy subjects. METHODS Thirty-nine female SSc patients (30 in the early stages of SSc) and 44 age-matched healthy women were included. Peripheral blood EPCs were quantified using flow cytometry and by counting the early outgrowth CFUs. RESULTS The EPCs quantified with flow cytometry and the CFU numbers were significantly lower in SSc patients than in control subjects (155.1 ± 95.1 vs. 241.3 ± 184.2 EPC/10(6) lymphomononuclear cells, p=0.011; 15.4 ± 8.6 vs. 23.5 ± 10.9 CFU, p<0.001; respectively), as well as in the group of patients in the early stages of SSc compared to the controls. Patients with digital ulcers had significantly higher CFU counts than those without ulcers (p=0.013). Among patients with the scleroderma pattern on nailfold capillaroscopy, patients with the late pattern had significantly lower EPC levels than those with the early and active patterns (p=0.046). There were no significant correlations of EPCs or CFU levels with RP duration. CONCLUSIONS The present study revealed decreased EPCs in SSc patients, including those with early disease onset. These findings suggest that defective vasculogenesis occurs in the early phases of the disease. Therefore, EPCs might be an important therapeutic target for the prevention of vascular complications in SSc patients.
Collapse
Affiliation(s)
- Fernando V Andrigueti
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Maria I Arismendi
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Pâmela C C Ebbing
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil
| | - Cristiane Kayser
- Rheumatology Division, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, Brazil.
| |
Collapse
|
13
|
Noort AR, van Zoest KPM, Weijers EM, Koolwijk P, Maracle CX, Novack DV, Siemerink MJ, Schlingemann RO, Tak PP, Tas SW. NF-κB-inducing kinase is a key regulator of inflammation-induced and tumour-associated angiogenesis. J Pathol 2014; 234:375-85. [PMID: 25043127 PMCID: PMC4194146 DOI: 10.1002/path.4403] [Citation(s) in RCA: 71] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/23/2014] [Accepted: 07/02/2014] [Indexed: 01/08/2023]
Abstract
Angiogenesis is essential during development and in pathological conditions such as chronic inflammation and cancer progression. Inhibition of angiogenesis by targeting vascular endothelial growth factor (VEGF) blocks disease progression, but most patients eventually develop resistance which may result from compensatory signalling pathways. In endothelial cells (ECs), expression of the pro-angiogenic chemokine CXCL12 is regulated by non-canonical nuclear factor (NF)-κB signalling. Here, we report that NF-κB-inducing kinase (NIK) and subsequent non-canonical NF-κB signalling regulate both inflammation-induced and tumour-associated angiogenesis. NIK is highly expressed in endothelial cells (ECs) in tumour tissues and inflamed rheumatoid arthritis synovial tissue. Furthermore, non-canonical NF-κB signalling in human microvascular ECs significantly enhanced vascular tube formation, which was completely blocked by siRNA targeting NIK. Interestingly, Nik(-/-) mice exhibited normal angiogenesis during development and unaltered TNFα- or VEGF-induced angiogenic responses, whereas angiogenesis induced by non-canonical NF-κB stimuli was significantly reduced. In addition, angiogenesis in experimental arthritis and a murine tumour model was severely impaired in these mice. These studies provide evidence for a role of non-canonical NF-κB signalling in pathological angiogenesis, and identify NIK as a potential therapeutic target in chronic inflammatory diseases and tumour neoangiogenesis.
Collapse
Affiliation(s)
- Ae R Noort
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Katinka PM van Zoest
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Ester M Weijers
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU Medical CenterAmsterdam, The Netherlands
| | - Pieter Koolwijk
- Department of Physiology, Institute for Cardiovascular Research (ICaR-VU), VU Medical CenterAmsterdam, The Netherlands
| | - Chrissta X Maracle
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Deborah V Novack
- Division of Bone and Mineral Diseases, Departments of Medicine and Pathology, Washington University School of MedicineSt Louis, Missouri, USA
| | - Martin J Siemerink
- Ocular Angiogenesis Group, Department of Ophthalmology and Department of Cell Biology and Histology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Reinier O Schlingemann
- Ocular Angiogenesis Group, Department of Ophthalmology and Department of Cell Biology and Histology, Academic Medical Center/University of AmsterdamThe Netherlands
| | - Paul P Tak
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Medicine, University of CambridgeCambridge, UK
| | - Sander W Tas
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of AmsterdamThe Netherlands
- Department of Experimental Immunology, Academic Medical Center/University of AmsterdamThe Netherlands
| |
Collapse
|
14
|
Verma I, Syngle A, Krishan P. Endothelial progenitor cell biology in ankylosing spondylitis. Int J Rheum Dis 2014; 18:336-40. [PMID: 25351377 DOI: 10.1111/1756-185x.12487] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
AIM Endothelial progenitor cells (EPCs) are unique populations which have reparative potential in overcoming endothelial damage and reducing cardiovascular risk. Patients with ankylosing spondylitis (AS) have increased risk of cardiovascular morbidity and mortality. The aim of this study was to investigate the endothelial progenitor cell population in AS patients and its potential relationships with disease variables. METHODS Endothelial progenitor cells were measured in peripheral blood samples from 20 AS and 20 healthy controls by flow cytometry on the basis of CD34 and CD133 expression. Disease activity was evaluated by using Bath Ankylosing Spondylitis Disease Activity Index (BASDAI). Functional ability was monitored by using Bath Ankylosing Spondylitis Functional Index (BASFI). RESULTS EPCs were depleted in AS patients as compared to healthy controls (CD34(+) /CD133(+) : 0.027 ± 0.010% vs. 0.044 ± 0.011%, P < 0.001). EPC depletions were significantly associated with disease duration (r = -0.52, P = 0.01), BASDAI (r = -0.45, P = 0.04) and C-reactive protein (r = -0.5, P = 0.01). CONCLUSION This is the first study to demonstrate endothelial progenitor cell depletion in AS patients. EPC depletions inversely correlate with disease duration, disease activity and inflammation, suggesting the pivotal role of inflammation in depletion of EPCs. EPC would possibly also serve as a therapeutic target for preventing cardiovascular disease in AS.
Collapse
Affiliation(s)
- Inderjeet Verma
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, India
| | | | | |
Collapse
|
15
|
Inhibitor of DNA binding 1 as a secreted angiogenic transcription factor in rheumatoid arthritis. Arthritis Res Ther 2014; 16:R68. [PMID: 24620998 PMCID: PMC4060463 DOI: 10.1186/ar4507] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2013] [Accepted: 03/04/2014] [Indexed: 12/17/2022] Open
Abstract
Introduction Rheumatoid arthritis (RA) is characterized by enhanced blood vessel development in joint synovium. This involves the recruitment of endothelial progenitor cells (EPCs), allowing for de novo vessel formation and pro-inflammatory cell infiltration. Inhibitor of DNA Binding 1 (Id1) is a transcription factor characteristic of EPCs that influences cell maturation. Method Enzyme-linked immunosorbant assay (ELISA) and polymerase chain reaction (PCR) were used to examine Id1 levels in synovial fluid (SF) and endothelial cells (ECs), respectively. Immunohistology was used to determine the expression of Id1 in synovial tissue (ST). Human dermal microvascular EC (HMVEC) migration and tube forming assays were used to determine if recombinant human Id1 (rhuId1) and/or RA SF immunodepleted Id1 showed angiogenic activity. We also utilized the RA ST severe combined immunodeficient (SCID) mouse chimera to examine if Id1 recruits EPCs to RA synovium. Results ST samples immunostained for Id1 showed heightened expression in RA compared to osteoarthritis (OA) and normal (NL) ST. By immunofluorescence staining, we found significantly more Id1 in RA compared to OA and NL vasculature, showing that Id1 expressing cells, and therefore EPCs, are most active in vascular remodeling in the RA synovium. We also detected significantly more Id1 in RA compared to OA and other arthritis SFs by ELISA, which correlates highly with Chemokine (C-X-C motif) ligand 16 (CXCL16) levels. In vitro chemotaxis assays showed that Id1 is highly chemotactic for HMVECs and can be attenuated by inhibition of Nuclear Factor κB and phosphoinositide 3-kinase. Using in vitro Matrigel assays, we found that HMVECs form tubes in response to rhuId1 and that Id1 immunodepleted from RA SF profoundly decreases tube formation in Matrigel in vitro. PCR showed that Id1 mRNA could be up-regulated in EPCs compared to HMVECs in response to CXCL16. Finally, using the K/BxN serum induced arthritis model, we found that EC CXCR6 correlated with Id1 expression by immunohistochemistry. Conclusions We conclude that Id1 correlates highly with CXCL16 expression, EPC recruitment, and blood vessel formation in the RA joint, and that Id1 is potently angiogenic and can be up-regulated in EPCs by CXCL16.
Collapse
|
16
|
Takeshita J, Mohler ER, Krishnamoorthy P, Moore J, Rogers WT, Zhang L, Gelfand JM, Mehta NN. Endothelial cell-, platelet-, and monocyte/macrophage-derived microparticles are elevated in psoriasis beyond cardiometabolic risk factors. J Am Heart Assoc 2014; 3:e000507. [PMID: 24584739 PMCID: PMC3959700 DOI: 10.1161/jaha.113.000507] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background Psoriasis, especially when severe, is a risk factor for cardiometabolic disease beyond traditional risk factors. The mechanism of atherogenesis in psoriasis remains unknown. Cell membrane vesicles (ie, microparticles), released upon cell activation or apoptosis, have recently been associated with cardiometabolic disease and may play a pathogenic role. Microparticle levels, particularly from endothelial cells and platelets, are elevated in patients with cardiovascular disorders, metabolic syndrome, other inflammatory diseases, autoimmune conditions, and have been shown to be predictive of cardiovascular outcomes. Methods and Results Concentrations of microparticles with positive expression for any of 7 cell surface markers (Annexin V, CD3, CD31, CD41a, CD64, CD105, and CD144) were measured in blood samples from psoriasis patients (n=53) and control subjects without psoriasis (n=41). Platelet‐free plasma was separated from whole blood by one‐step centrifugation for microparticle analysis. Microparticles were fluorescently labeled and characterized by flow cytometry. Higher concentrations of CD105 (5.5/μL versus 2.5/μL, P<0.001), CD31 (31/μL versus 18/μL, P=0.002), CD41a (50/μL versus 22/μL, P<0.001), and CD64 (5.0/μL versus 4.1/μL, P=0.02) singly positive microparticles corresponding to endothelial cell‐, platelet‐, and monocyte/macrophage‐derived microparticles, respectively, were found in psoriasis patients compared with controls. These differences persisted after adjustment for traditional cardiometabolic risk factors including body mass index. Conclusions Increased microparticle concentrations, independent of cardiometabolic risk factors, in patients with psoriasis suggest that the presence of increased endothelial cell, platelet, and monocyte/macrophage activation with cell turnover may contribute to the heightened atherogenesis associated with psoriasis.
Collapse
Affiliation(s)
- Junko Takeshita
- Section of Vascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Isozaki T, Arbab AS, Haas CS, Amin MA, Arendt MD, Koch AE, Ruth JH. Evidence that CXCL16 is a potent mediator of angiogenesis and is involved in endothelial progenitor cell chemotaxis : studies in mice with K/BxN serum-induced arthritis. ACTA ACUST UNITED AC 2013; 65:1736-46. [PMID: 23633118 DOI: 10.1002/art.37981] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Accepted: 04/11/2013] [Indexed: 01/14/2023]
Abstract
OBJECTIVE To examine the possibility that CXCL16 recruits endothelial cells (ECs) to developing neovasculature in rheumatoid arthritis (RA) synovium. METHODS We utilized the RA synovial tissue SCID mouse chimera system to examine human microvascular EC (HMVEC) and human endothelial progenitor cell (EPC) recruitment into engrafted human synovium that was injected intragraft with CXCL16-immunodepleted RA synovial fluid (SF). CXCR6-deficient and wild-type (WT) C57BL/6 mice were primed to develop K/BxN serum-induced arthritis and evaluated for angiogenesis. HMVECs and EPCs from human cord blood were also examined for CXCR6 expression, by immunofluorescence and assessment of CXCL16 signaling activity. RESULTS CXCR6 was prominently expressed on human EPCs and HMVECs, and its expression on HMVECs could be up-regulated by interleukin-1β. SCID mice injected with CXCL16-depleted RA SF exhibited a significant reduction in EPC recruitment. In experiments using the K/BxN serum-induced inflammatory arthritis model, CXCR6(-/-) mice showed profound reductions in hemoglobin levels, which correlated with reductions in monocyte and T cell recruitment to arthritic joint tissue compared to that observed in WT mice. Additionally, HMVECs and EPCs responded to CXCL16 stimulation, but exhibited unique signal transduction pathways and homing properties. CONCLUSION These results indicate that CXCL16 and its receptor CXCR6 may be a central ligand/receptor pair that is closely associated with EPC recruitment and blood vessel formation in the RA joint.
Collapse
Affiliation(s)
- Takeo Isozaki
- University of Michigan Medical School, Ann Arbor, MI, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Balbir-Gurman A, Braun-Moscovici Y. Scleroderma - new aspects in pathogenesis and treatment. Best Pract Res Clin Rheumatol 2013; 26:13-24. [PMID: 22424190 DOI: 10.1016/j.berh.2012.01.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2012] [Accepted: 01/10/2012] [Indexed: 12/20/2022]
Abstract
Systemic sclerosis (SSc) is a multisystem disease with a variable clinical course and a poor prognosis corresponding to extent of microangiopathy and skin and internal organ fibrosis. Microvascular damage provokes immune cells to produce autoantibodies, pro-inflammatory and pro-fibrotic cytokines and chemokines. The hallmark of SSc is excessive collagen production by activated fibroblasts and myofibroblasts, and its accumulation in skin and internal organs. Better understanding of SSc pathogenesis resulted in the development of drugs, such as prostanoids, endothelin-1 and phosphodiesterase inhibitors, for treatment of pulmonary arterial hypertension and digital ulcers. The use of biological therapies and anti-fibrotic agents is under investigation. Stem cell transplantation seems to be promising in restarting the immune system to diminish fibrosis and restore microvasculature. Future research will be directed at genetic factors, diagnostic and prognostic markers for fibrosis and microangiopathy, and development of drugs directed to pathogenic key cells and mediators.
Collapse
|
19
|
|
20
|
Batycka-Baran A, Paprocka M, Krawczenko A, Kantor A, Duś D, Szepietowski JC. Reduced number of circulating endothelial progenitor cells (CD133+/KDR+) in patients with plaque psoriasis. Dermatology 2012; 225:88-92. [PMID: 22986416 DOI: 10.1159/000341534] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 06/20/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Psoriasis is associated with an increased cardiovascular risk. Circulating endothelial progenitor cells (CEPCs) play a significant role in the maintenance of vascular homeostasis. OBJECTIVE The aim of this study was to evaluate the number of CEPCs in patients with psoriasis compared to controls and assess possible correlations between the number of these cells and the plasma levels of vascular endothelial growth factor (VEGF), soluble vascular endothelial growth factor receptor-1 (sVEGFR-1) and clinical features of psoriasis. METHODS The number of CEPCs, identified as CD133+/KDR+ cells, was determined with flow cytometry in peripheral blood of psoriatic patients (n = 63) and controls (n = 31). The plasma levels of VEGF and sVEGFR-1 were measured with enzyme-linked immunosorbent assay. RESULTS The number of CEPCs was significantly reduced in psoriatic patients compared with controls (p = 0.000026) and inversely correlated with disease severity (R = -0.283; p = 0.0248). CONCLUSION A reduced number of CEPCs may contribute to endothelial dysfunction in patients with psoriasis.
Collapse
Affiliation(s)
- Aleksandra Batycka-Baran
- Department of Dermatology, Venereology and Allergology, Wroclaw Medical University, Wroclaw, Poland.
| | | | | | | | | | | |
Collapse
|
21
|
|
22
|
Endothelial progenitor cell-dependent angiogenesis requires localization of the full-length form of uPAR in caveolae. Blood 2011; 118:3743-55. [PMID: 21803847 DOI: 10.1182/blood-2011-02-338681] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Endothelial urokinase-type plasminogen activator receptor (uPAR) is thought to provide a regulatory mechanism in angiogenesis. Here we studied the proangiogenic role of uPAR in endothelial colony-forming cells (ECFCs), a cell population identified in human umbilical blood that embodies all of the properties of an endothelial progenitor cell matched with a high proliferative rate. By using caveolae-disrupting agents and by caveolin-1 silencing, we have shown that the angiogenic properties of ECFCs depend on caveolae integrity and on the presence of full-length uPAR in such specialized membrane invaginations. Inhibition of uPAR expression by antisense oligonucleotides promoted caveolae disruption, suggesting that uPAR is an inducer of caveolae organization. Vascular endothelial growth factor (VEGF) promoted accumulation of uPAR in ECFC caveolae in its undegraded form. We also demonstrated that VEGF-dependent ERK phosphorylation required integrity of caveolae as well as caveolar uPAR expression. VEGF activity depends on inhibition of ECFC MMP12 production, which results in impairment of MMP12-dependent uPAR truncation. Further, MMP12 overexpression in ECFC inhibited vascularization in vitro and in vivo. Our data suggest that intratumor homing of ECFCs suitably engineered to overexpress MMP12 could have the chance to control uPAR-dependent activities required for tumor angiogenesis and malignant cells spreading.
Collapse
|
23
|
Gasparyan AY, Ayvazyan L, Blackmore H, Kitas GD. Writing a narrative biomedical review: considerations for authors, peer reviewers, and editors. Rheumatol Int 2011; 31:1409-17. [PMID: 21800117 DOI: 10.1007/s00296-011-1999-3] [Citation(s) in RCA: 468] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2011] [Accepted: 07/10/2011] [Indexed: 12/20/2022]
Abstract
Review articles comprehensively covering a specific topic are crucial for successful research and academic projects. Most editors consider review articles for special and regular issues of journals. Writing a review requires deep knowledge and understanding of a field. The aim of this review is to analyze the main steps in writing a narrative biomedical review and to consider points that may increase the chances of success. We performed a comprehensive search through MEDLINE, EMBASE, Scopus, and Web of Science using the following keywords: review of the literature, narrative review, title, abstract, authorship, ethics, peer review, research methods, medical writing, scientific writing, and writing standards. Opinions expressed in the review are also based on personal experience as authors, peer reviewers, and editors.
Collapse
Affiliation(s)
- Armen Yuri Gasparyan
- Department of Rheumatology, Clinical Research Unit, Dudley Group of Hospitals NHS Foundation Trust (Teaching Trust of University of Birmingham, UK), Russell's Hall Hospital, North Block, Dudley, West Midlands, DY1 2HQ, United Kingdom.
| | | | | | | |
Collapse
|
24
|
Distler JHW, Akhmetshina A, Dees C, Jüngel A, Stürzl M, Gay S, Pisetsky DS, Schett G, Distler O. Induction of apoptosis in circulating angiogenic cells by microparticles. ACTA ACUST UNITED AC 2011; 63:2067-77. [DOI: 10.1002/art.30361] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
25
|
Bartoloni E, Shoenfeld Y, Gerli R. Inflammatory and autoimmune mechanisms in the induction of atherosclerotic damage in systemic rheumatic diseases: Two faces of the same coin. Arthritis Care Res (Hoboken) 2011; 63:178-83. [DOI: 10.1002/acr.20322] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Baker JF, Zhang L, Imadojemu S, Sharpe A, Patil S, Moore JS, Mohler ER, Von Feldt J. Circulating endothelial progenitor cells are reduced in SLE in the absence of coronary artery calcification. Rheumatol Int 2011; 32:997-1002. [PMID: 21246370 DOI: 10.1007/s00296-010-1730-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 12/30/2010] [Indexed: 12/15/2022]
Abstract
Circulating endothelial progenitor cells (EPCs) are reduced in patients with systemic lupus erythematosus (SLE). A reduced number of EPCs are associated with the presence of atherosclerosis in other populations. We sought to determine whether the reduction in EPC numbers in SLE is dependent on the presence of advanced coronary artery calcification (CAC). Patients with SLE had previous coronary calcium scores which placed them in either the >75th percentile or <25th percentile for their age. Seventeen patients with SLE and 13 healthy controls (HC) were included in the study. White blood cells were stained for EPC and progenitor cell markers including CD34, CD133, and VEGFR and analyzed by flow cytometry. SLE patients had repeated coronary imaging as well as carotid ultrasound. There was no difference in age between groups. SLE patients with advanced CAC were more likely to be hypertensive, to be smokers, and to have longer disease duration than SLE patients without CAC. SLE patients without evidence of CAC had a significantly lower number of EPCs (CD34+/CD133+/VEGFR+) compared to HC (median (IQR)) 0 (0, 6.7) vs. 10.2 (5.8, 12.3) (P = 0.02). Total numbers of PCs (CD133+/CD34+) were not significantly decreased in patients with SLE ((mean ± SEM) 1,007 ± 154 vs. 824 ± 170 (P = 0.20)). No significant difference was seen in EPC number between SLE patients without CAC and those with advanced CAC. Increased carotid intima-media thickness did not correlate with CAC or EPC number in SLE patients. Reduced numbers of EPCs in SLE patients may be observed compared to HC even in the absence of CAC. Differences in measured risk factor profiles and depletion of total circulating PCs do not fully explain this finding.
Collapse
Affiliation(s)
- Joshua F Baker
- Division of Rheumatology, Department of Medicine, Hospital of the University of Pennsylvania, 8 Penn Tower Building, 34th and Civic Center Blvd., Philadelphia, PA 19104, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Jones OY, Gok F, Rushing EJ, Horkayne-Szakaly I, Ahmed AA. Engraftment of donor mesenchymal stem cells in chimeric BXSB includes vascular endothelial cells and hepatocytes. STEM CELLS AND CLONING-ADVANCES AND APPLICATIONS 2011; 4:73-8. [PMID: 24198532 PMCID: PMC3781759 DOI: 10.2147/sccaa.s23014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Somatic tissue engraftment was studied in BXSB mice treated with mesenchymal stem cell transplantation. Hosts were conditioned with nonlethal radiation prior to introducing donor cells from major histocompatibility complex-matched green fluorescent protein transgenic mice. Transplant protocols differed for route of injection, ie, intravenous (i.v.) versus intraperitoneal (i.p.), and source of mesenchymal stem cells, ie, unfractionated bone marrow cells, ex vivo expanded mesenchymal stem cells, or bone chips. Tissue chimerism was determined after short (10–12 weeks) or long (62 weeks) posttransplant follow-up by immunohistochemistry for green fluorescent protein. Engraftment of endothelial cells was seen in several organs including liver sinusoidal cells in i.v. treated mice with ex vivo expanded mesenchymal stem cells or with unfractionated bone marrow cells. Periportal engraftment of liver hepatocytes, but not engraftment of endothelial cells, was found in mice injected i.p. with bone chips. Engraftment of adipocytes was a common denominator in both i.v. and i.p. routes and occurred during early phases post-transplant. Disease control was more robust in mice that received both i.v. bone marrow and i.p. bone chips compared to mice that received i.v. bone marrow alone. Thus, the data support potential use of mesenchymal stem cell transplant for treatment of severe lupus. Future studies are needed to optimize transplant conditions and tailor protocols that may in part be guided by fat and endothelial biomarkers. Furthermore, the role of liver chimerism in disease control and the nature of cellular communication among donor hematopoietic and mesenchymal stem cells in a chimeric host merit further investigation.
Collapse
Affiliation(s)
- Olcay Y Jones
- Department of Pediatrics, Walter Reed National Military Medical Center, Bethesda, MD, USA
| | | | | | | | | |
Collapse
|
28
|
Bartoloni E, Alunno A, Luccioli F, Moscatelli S, Biscontini D, Santoboni G, Gerli R. Atherosclerotic vascular damage and rheumatoid arthritis: a complex but intriguing link. Expert Rev Cardiovasc Ther 2010; 8:1309-16. [PMID: 20828353 DOI: 10.1586/erc.10.123] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Rheumatoid arthritis is a chronic inflammatory disease characterized by a reduced life expectancy mainly due to cardiovascular disease. In long-standing disease, it has been widely demonstrated that both traditional cardiovascular risk and disease-related factors, including chronic inflammation and immune-mediated mechanisms, play a key role in accelerating atherosclerotic damage of the arterial wall. The short- and long-term effects of immunosuppressive treatment on cardiovascular disease outcome is, however, uncertain and a multidisciplinary approach appears to represent the best management of cardiovascular risk in these patients.
Collapse
Affiliation(s)
- Elena Bartoloni
- Rheumatology Unit, Department of Clinical & Experimental Medicine, University of Perugia, Perugia 06100, Italy
| | | | | | | | | | | | | |
Collapse
|
29
|
Petrelli A, Maestroni A, Fadini GP, Belloni D, Venturini M, Albiero M, Kleffel S, Mfarrej BG, Maschio AD, Maffi P, Avogaro A, Ferrero E, Zerbini G, Secchi A, Fiorina P. Improved function of circulating angiogenic cells is evident in type 1 diabetic islet-transplanted patients. Am J Transplant 2010; 10:2690-700. [PMID: 21114646 DOI: 10.1111/j.1600-6143.2010.03309.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Circulating angiogenic cells (CACs) are vascular-committed bone marrow-derived cells that are dysfunctional in type 1 diabetes (T1D). Here we studied whether restoration of normoglycemia following islet transplantation is associated with better CAC function. We carried out a cross-sectional study of 18 T1D patients, 14 insulin-independent islet-transplanted patients (ITA) and 14 healthy controls (C) evaluating in vivo and in vitro CACs viability and function. We found that the percentage of CACs in vivo did not differ among the three groups while the number of CAC colonies obtained from T1D, but not from ITA, was reduced compared to C (C = 7.3 ± 1.9, T1D = 0.9 ± 0.4 and ITA = 4.7 ± 1.9; p < 0.05 T1D vs. all). In vitro CAC migration/differentiation were similar, while in vivo an improved angiogenic ability of ITA compared to T1D was shown (capillary density: C = 93.5 ± 22.1, T1D = 19.2 ± 2.8 and ITA = 44.0 ± 10.5, p < 0.05 T1D vs. all). Increased apoptosis and lesser IL-8 secretion were evident in CACs obtained from T1D compared to C and ITA. in vitro addition of anti-hIL-8 reduced the number of colonies obtained from C. Finally, T1D, but not ITA, had a lower endothelial-dependent dilatation (EDD) compared with C. These data suggest that CAC function is altered in T1D and may be improved after islet transplantation.
Collapse
Affiliation(s)
- A Petrelli
- Transplantation Research Center, Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bartoloni E, Alunno A, Bistoni O, Gerli R. How early is the atherosclerotic risk in rheumatoid arthritis? Autoimmun Rev 2010; 9:701-7. [DOI: 10.1016/j.autrev.2010.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Accepted: 06/02/2010] [Indexed: 12/25/2022]
|
31
|
Manetti M, Guiducci S, Ibba-Manneschi L, Matucci-Cerinic M. Mechanisms in the loss of capillaries in systemic sclerosis: angiogenesis versus vasculogenesis. J Cell Mol Med 2010; 14:1241-54. [PMID: 20132409 PMCID: PMC3828842 DOI: 10.1111/j.1582-4934.2010.01027.x] [Citation(s) in RCA: 100] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Systemic sclerosis (SSc, scleroderma) is a chronic, multisystem connective tissue disorder affecting the skin and various internal organs. Although the disease is characterized by a triad of widespread microangiopathy, fibrosis and autoimmunity, increasing evidence indicates that vascular damage is a primary event in the pathogenesis of SSc. The progressive vascular injury includes persistent endothelial cell activation/damage and apoptosis, intimal thickening, delamination, vessel narrowing and obliteration. These profound vascular changes lead to vascular tone dysfunction and reduced capillary blood flow, with consequent tissue ischemia and severe clinical manifestations, such as digital ulceration or amputation, pulmonary arterial hypertension and scleroderma renal crisis. The resulting tissue hypoxia induces complex cellular and molecular mechanisms in the attempt to recover endothelial cell function and tissue perfusion. Nevertheless, in SSc patients there is no evidence of significant angiogenesis and the disease evolves towards chronic tissue ischemia, with progressive and irreversible structural changes in multiple vascular beds culminating in the loss of capillaries. A severe imbalance between pro-angiogenic and angiostatic factors may also lead to impaired angiogenic response during SSc. Besides insufficient angiogenesis, defective vasculogenesis with altered numbers and functional defects of bone marrow-derived endothelial progenitor cells may contribute to the vascular pathogenesis of SSc. The purpose of this article is to review the contribution of recent studies to the understanding of the complex mechanisms of impaired vascular repair in SSc. Indeed, understanding the pathophysiology of SSc-associated vascular disease may be the key in dissecting the disease pathogenesis and developing novel therapies. Either angiogenic or vasculogenic mechanisms may potentially become in the future the target of therapeutic strategies to promote capillary regeneration in SSc.
Collapse
Affiliation(s)
- Mirko Manetti
- Department of Anatomy, Histology and Forensic Medicine, University of Florence, Florence, Italy.
| | | | | | | |
Collapse
|