Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
2.
Int J Mol Sci ; 20(24)2019 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-31817940

RESUMEN

In systemic sclerosis (SSc), the possible involvement of lymphatic microcirculation and lymphangiogenesis has traditionally been overshadowed by the greater emphasis placed on dysfunctional blood vascular system and angiogenesis. In the present in vitro study, we explore for the first time whether the SSc microenvironment may interfere with lymphangiogenesis, a complex, multi-step process in which lymphatic microvascular endothelial cells (LMVECs) sprout, migrate, and proliferate to generate new lymphatic capillaries. Normal human adult dermal LMVECs from three donors were treated with serum from SSc patients (n = 8), serum from healthy individuals (n = 8), or recombinant human vascular endothelial growth factor (VEGF)-C as a positive control for lymphangiogenesis. Cell proliferation, Boyden chamber Matrigel chemoinvasion, wound healing capacity, and lymphatic capillary morphogenesis on Geltrex were assayed. VEGF-C serum levels were measured by enzyme-linked immunosorbent assay. Gene and protein expression levels of the lymphangiogenic orchestrators VEGF receptor-3 (VEGFR-3)/Flt-4 and neuropilin-2 (NRP-2) were determined by real-time PCR and Western blotting, respectively. Conditioning with SSc serum significantly inhibited LMVEC proliferation, Matrigel invasion, and wound healing capacity with respect to healthy serum. The ability of LMVECs to form lymphatic tubes on Geltrex was also severely compromised in the presence of SSc serum. VEGF-C levels were comparable in SSc and healthy sera. Treatment with SSc serum resulted in a significant downregulation of both VEGFR-3/Flt-4 and NRP-2 mRNA and protein levels. In SSc, the pathologic environment severely hampers every lymphangiogenesis step, likely through the reduction of pro-lymphangiogenic VEGFR-3/NRP-2 co-receptor signaling. The impairment of the lymphangiogenic process opens a new scenario underlying SSc vascular pathophysiology, which is worth investigating further.


Asunto(s)
Medios de Cultivo Condicionados/farmacología , Células Endoteliales/patología , Linfangiogénesis , Neovascularización Patológica/patología , Esclerodermia Sistémica/sangre , Esclerodermia Sistémica/patología , Microambiente Tumoral , Adulto , Apoptosis , Estudios de Casos y Controles , Movimiento Celular , Proliferación Celular , Células Cultivadas , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Humanos , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Neuropilina-2/genética , Neuropilina-2/metabolismo , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/metabolismo , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/metabolismo , Adulto Joven
3.
Acta Histochem ; 121(8): 151442, 2019 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-31540712

RESUMEN

Telocytes (TCs), also known as CD34+ stromal/interstitial cells, have recently been identified within the connective tissue of a variety of organs including the normal human testis. Testicular TCs appear to constitute a widespread reticular network distributed either in the peritubular or in the intertubular stromal spaces where they have been suggested to play different roles, such as participation to testis morphogenesis, postnatal preservation of the normal tissue/organ three-dimensional structure, and regulation of spermatogenesis and androgen hormone secretion and release. Although increasing evidence indicates that TCs may be involved in the pathophysiology of various diseases, no study has yet reported possible changes in these cells within the stromal compartment of seminoma, one of the most frequent malignant testicular cancers in humans. Therefore, here we carried out the first investigation of the presence and tissue distribution of TCs/CD34+ stromal cells in human testicular seminoma in comparison with normal human testis using either CD34 immunohistochemistry or CD34/CD31 and CD34/α-smooth muscle actin (α-SMA) double immunofluorescence analyses. In seminoma tissue sections, we observed an overall loss of TCs (CD34+/CD31- stromal cells) accompanying a severe degeneration of the normal architecture of seminiferous tubules and stromal tissue associated with dense cellularity increase and presence of interstitial fibrosis. Noteworthy, in the seminoma tissue the disappearance of TCs was paralleled by an expansion of α-SMA+ myoid cells. Moreover, the CD34+/CD31+ blood vessel network was greatly expanded, while steroidogenic Leydig cells were undetectable in seminoma specimens. Since TCs are emerging as important regulators of tissue and organ homeostasis, collectively the present findings indicate that the possible pathophysiologic implications of the loss of TCs in human testicular seminoma should not be further overlooked.


Asunto(s)
Actinas/metabolismo , Antígenos CD34/metabolismo , Células Mieloides , Proteínas de Neoplasias/metabolismo , Seminoma , Telocitos , Neoplasias Testiculares , Adulto , Humanos , Masculino , Células Mieloides/metabolismo , Células Mieloides/patología , Seminoma/metabolismo , Seminoma/patología , Telocitos/metabolismo , Telocitos/patología , Neoplasias Testiculares/metabolismo , Neoplasias Testiculares/patología
4.
J Clin Med ; 8(7)2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31261789

RESUMEN

Hemophilic arthropathy (HA) typically begins with proliferative synovitis that shares some similarities with inflammatory arthritides, in which the proinflammatory cytokine tumor necrosis factor (TNF)-α has a crucial pathogenetic role. Inappropriate release of TNF-α was shown to contribute to arthropathy development following intra-articular bleeding in hemophilic mice. Here, we were interested in determining whether systemic levels of TNF-α and synovial tissue expression of the TNF-α/TNF receptor (TNF-R) system could be increased and related to joint damage in hemophilia A patients with severe HA. Serum levels of TNF-α measured by quantitative enzyme-linked immunosorbent assay (ELISA) were significantly increased in HA patients (n = 67) compared to healthy controls (n = 20). In HA patients, elevated TNF-α levels were significantly associated with the number of hemarthroses, the grade of synovial hypertrophy, and both the clinical World Federation of Hemophilia score and ultrasound score. The expression of TNF-α, TNF-R1, and TNF-R2 was strongly increased in HA synovium (n = 10) compared to the non-inflamed osteoarthritis control synovium (n = 8), as assessed by both immunohistochemistry and Western blotting. Increased protein levels of TNF-α, TNF-R1, and TNF-R2 were retained in vitro by HA fibroblast-like synoviocytes (n = 6) with respect to osteoarthritis control fibroblast-like synoviocytes (n = 6). Stimulation with TNF-α resulted in a significant increase in HA fibroblast-like synoviocyte proliferation quantified by the water-soluble tetrazolium (WST)-1 assay, while it had no relevant effect on osteoarthritis fibroblast-like synoviocytes. Quantification of active/cleaved caspase-3 by ELISA demonstrated that TNF-α did not induce apoptosis either in HA or in osteoarthritis fibroblast-like synoviocytes. The TNF-α/TNF-R system may represent a crucial mediator of proliferative synovitis and, therefore, a new attractive target for the prevention and treatment of joint damage in HA patients. Our findings provide the groundwork for further clinical investigation of anti-TNF-α therapeutic feasibility in hemophiliacs.

5.
Sci Rep ; 9(1): 5858, 2019 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-30971762

RESUMEN

Telocytes have recently emerged as unique interstitial cells defined by their extremely long, thin and moniliform prolongations termed telopodes. Despite growing evidence that these cells consistently reside in the stromal compartment of various organs from human beings, studies dealing with telocytes in structures of the oral cavity are scarce. Hence, the present morphologic study was undertaken to explore for the first time the presence and specific localization of telocytes within tissues of the normal human tongue, a complex muscular organ whose main functions include taste, speech, and food manipulation in the oral cavity. Telocytes were initially identified by CD34 immunostaining and confirmed by CD34/PDGFRα double immunofluorescence and transmission electron microscopy. CD34+/PDGFRα+ telocytes were organized in interstitial meshworks either in the tongue lamina propria or in the underlying striated muscle. Lingual telocytes were immunonegative for CD31, c-kit and α-SMA. Telopodes were finely distributed throughout the stromal space and concentrated beneath the lingual epithelium and around CD31+ vessels, skeletal muscle bundles/fibers, and intramuscular nerves and ganglia. They also enveloped salivary gland units outside the α-SMA+ myoepithelial cells and delimited lymphoid aggregates. These findings establish telocytes as a previously overlooked interstitial cell population worth investigating further in the setting of human tongue pathophysiology.


Asunto(s)
Telocitos/metabolismo , Lengua/metabolismo , Adulto , Anciano , Antígenos CD34/metabolismo , Femenino , Humanos , Inmunofenotipificación , Masculino , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Persona de Mediana Edad , Membrana Mucosa/citología , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Telocitos/patología , Telocitos/ultraestructura , Telopodos/metabolismo , Telopodos/patología , Lengua/patología
6.
Ann Rheum Dis ; 77(11): 1665-1674, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30021803

RESUMEN

OBJECTIVE: In systemic sclerosis (SSc), early microvascular injury is followed by impaired angiogenesis and peripheral capillary loss. Here, we investigated the possible contribution of the neurovascular guidance molecule Slit2 and its Roundabout (Robo) receptors to SSc-related endothelial cell dysfunction. METHODS: Circulating Slit2 levels were measured in patients with SSc and healthy controls. Slit2, Robo1 and Robo4 expression was investigated in SSc and healthy skin biopsies and explanted dermal microvascular endothelial cells (MVECs). Slit2/Robo4 function in MVEC angiogenesis was studied by cell viability, wound healing and capillary-like tube formation assays. RESULTS: Circulating Slit2 was significantly increased in either SSc or patients with a very early diagnosis of SSc (VEDOSS) compared with controls. Interestingly, serum Slit2 levels were raised in patients with VEDOSS with nailfold videocapillaroscopy (NVC) abnormalities, while they were similar in VEDOSS with normal NVC and controls. In SSc, Slit2 and Robo4 expression was upregulated in clinically affected skin and explanted MVECs in respect to controls. The angiogenic performance of healthy MVECs was significantly reduced after challenge with recombinant human Slit2 or SSc sera. These inhibitory effects were significantly attenuated when SSc sera were preincubated with an anti-Slit2 blocking antibody. In vitro angiogenesis was severely compromised in SSc-MVECs and could be significantly ameliorated by Slit2 neutralisation or ROBO4 gene silencing. Slit2/Robo4 axis interfered with angiogenesis through the inhibition of Src kinase phosphorylation. CONCLUSIONS: In SSc, increased circulating levels of Slit2 and activation of the Slit2/Robo4 antiangiogenic axis may contribute to peripheral microangiopathy since the very early phase of the disease.


Asunto(s)
Péptidos y Proteínas de Señalización Intercelular/fisiología , Neovascularización Patológica/sangre , Proteínas del Tejido Nervioso/fisiología , Receptores de Superficie Celular/fisiología , Esclerodermia Sistémica/patología , Adulto , Anciano , Supervivencia Celular/fisiología , Células Cultivadas , Células Endoteliales/fisiología , Endotelio Vascular/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intercelular/sangre , Masculino , Persona de Mediana Edad , Neovascularización Patológica/patología , Proteínas del Tejido Nervioso/sangre , Receptores de Superficie Celular/sangre , Esclerodermia Sistémica/sangre , Esclerodermia Sistémica/fisiopatología , Piel/irrigación sanguínea , Cicatrización de Heridas/fisiología
7.
Adv Exp Med Biol ; 998: 237-254, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28936744

RESUMEN

Among cardiac interstitial cells, the recently described telocytes (TCs) display the unique ability to build a supportive three-dimensional network formed by their very long and thin prolongations named telopodes. Cardiac TCs are increasingly regarded as pivotal regulators in intercellular signaling with multiple cell types, such as cardiomyocytes, stem/progenitor cells, microvessels, nerve endings, fibroblasts and immune cells, thus converting the cardiac stromal compartment into an integrated system that may drive either heart development or maintenance of cardiac homeostasis in post-natal life. Besides direct intercellular communications between TCs and neighboring cells, different types of TC-released extracellular vesicles (EVs), namely exosomes, ectosomes and multivesicular cargos, may act as shuttles for paracrine molecular signal exchange between cardiac TCs and cardiomyocytes or putative cardiomyocyte progenitors. In this review, we summarize the recent research findings on cardiac TCs and their EVs. We first provide an overview of the general features of TCs, including their peculiar morphological traits and immunophenotypes, intercellular signaling mechanisms and possible functional roles. Thereafter, we describe the distribution of TCs in normal and diseased hearts, as well as their role as intercellular communicators via the release of exosomes and other types of EVs. Finally, the involvement of cardiac TCs in cardiovascular diseases and the potential utility of TC transplantation and TC-derived exosomes in cardiac regeneration and repair are discussed.


Asunto(s)
Enfermedades Cardiovasculares/fisiopatología , Exosomas/metabolismo , Miocardio/metabolismo , Transducción de Señal , Telocitos/metabolismo , Animales , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/cirugía , Comunicación Celular , Exosomas/trasplante , Exosomas/ultraestructura , Humanos , Miocardio/ultraestructura , Fenotipo , Recuperación de la Función , Regeneración , Telocitos/trasplante , Telocitos/ultraestructura
8.
J Cell Mol Med ; 21(12): 3602-3611, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28714595

RESUMEN

Telocytes (TC) are typically defined as cells with telopodes by their ultrastructural features. Their presence was reported in the interstitium of various organs in vertebrates, including humans. However, no study has yet described the presence of TC in the human eye and in particular, within the stromal compartment of the cornea. To address this issue, samples of normal and pathologic (keratoconic) human corneas were tested by immunohistochemistry for CD34, platelet-derived growth factor receptor α (PDGFRα) and c-kit/CD117 or examined by transmission electron microscopy. We found that TC coexpressing CD34 and PDGFRα were distributed throughout the whole normal corneal stroma with different TC subtypes being distinguishable on the basis of the expression of the stemness marker c-kit (i.e. c-kit-positive and c-kit-negative TC subpopulations). Transmission electron microscopy examination confirmed the existence of spindle-shaped and bipolar TC typically displaying two long and thin moniliform telopodes establishing intercellular contacts formed by gap junctions. Keratoconic corneas were characterized by ultrastructural damages and patchy loss of TC with an almost complete depletion of the c-kit-positive TC subpopulation. We propose that TC may contribute to the maintenance of corneal stromal homoeostasis and that, in particular, the c-kit-positive TC subtype might have stemness capacity participating in corneal regeneration and repair processes. Further studies are needed to clarify the differential roles of corneal TC subtypes as well as the possible therapeutic applications of TC in degenerative corneal disorders such as keratoconus.


Asunto(s)
Linaje de la Célula/fisiología , Córnea/ultraestructura , Uniones Comunicantes/ultraestructura , Queratocono/patología , Proteínas Proto-Oncogénicas c-kit/genética , Telocitos/ultraestructura , Antígenos CD34/genética , Antígenos CD34/metabolismo , Biomarcadores/metabolismo , Forma de la Célula , Córnea/metabolismo , Uniones Comunicantes/metabolismo , Expresión Génica , Humanos , Inmunohistoquímica , Queratocono/genética , Queratocono/metabolismo , Microscopía Electrónica de Transmisión , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Telocitos/clasificación , Telocitos/metabolismo
9.
Adv Exp Med Biol ; 913: 51-76, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27796880

RESUMEN

Telocytes are a peculiar stromal (interstitial) cell type implicated in tissue homeostasis and development, as well as in the pathophysiology of several disorders. Severe damage and reduction of telocytes have been reported during fibrotic remodeling of multiple organs in various diseases, including scleroderma, Crohn's disease, ulcerative colitis, and liver fibrosis, as well as in chronic inflammatory lesions like those of primary Sjögren's syndrome and psoriasis. Owing to their close relationship with stem cells, telocytes are also supposed to contribute to tissue repair/regeneration. Indeed, telocytes are universally considered as "connecting cells" mostly oriented to intercellular signaling. On the basis of recent promising experimental findings, in the near future, telocyte transplantation might represent a novel therapeutic opportunity to control the evolution of chronic inflammatory and fibrotic diseases. Notably, there is evidence to support that telocytes could help in preventing abnormal activation of immune cells and fibroblasts, as well as in attenuating the altered matrix organization during the fibrotic process. By targeting telocytes alone or in tandem with stem cells, we might be able to promote regeneration and prevent the evolution to irreversible tissue injury. Besides exogenous transplantation, exploring pharmacological or non-pharmacological methods to enhance the growth and/or survival of telocytes could be an additional therapeutic strategy for many disorders.


Asunto(s)
Colitis Ulcerosa/terapia , Enfermedad de Crohn/terapia , Cirrosis Hepática/terapia , Psoriasis/terapia , Esclerodermia Sistémica/terapia , Síndrome de Sjögren/terapia , Telocitos/trasplante , Comunicación Celular , Enfermedad Crónica , Colitis Ulcerosa/metabolismo , Colitis Ulcerosa/patología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Humanos , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Linfocitos/citología , Linfocitos/metabolismo , Terapia Molecular Dirigida/métodos , Psoriasis/metabolismo , Psoriasis/patología , Regeneración/fisiología , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Transducción de Señal , Síndrome de Sjögren/metabolismo , Síndrome de Sjögren/patología , Células Madre/citología , Células Madre/metabolismo , Telocitos/citología , Telocitos/metabolismo
10.
J Cell Mol Med ; 20(4): 613-22, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26828975

RESUMEN

Although lymphatic neovascularization may be a key feature of chronic inflammation, it is almost unexplored in primary Sjögren's syndrome (pSS). A recent study revealed a pro-lymphangiogenic function of interleukin (IL)-17, a leading player in pSS pathogenesis. The aims of the study were to investigate lymphangiogenic mediators and lymphatic vasculature in pSS, as well as their possible association with IL-17. Circulating lymphatic endothelial precursor cells (LEPCs) and Th17 cells were enumerated in pSS patients and healthy donors. VEGF-C and IL-17 levels were assessed in paired serum samples. Lymphatic vasculature, VEGF-C/VEGF receptor (VEGFR)-3 and IL-17 were evaluated in pSS minor salivary glands (MSGs) and compared with normal and non-specific chronic sialadenitis (NSCS) MSGs. Circulating LEPCs were expanded in pSS and correlated with circulating Th17 cells, IL-17 and VEGF-C. In pSS MSGs, a newly formed lymphatic capillary network was found within periductal inflammatory infiltrates and the number of interlobular lymphatic vessels was significantly increased compared with normal and NSCS MSGs. Strong VEGF-C expression was detected in pSS ductal epithelial cells and periductal inflammatory cells. Numerous VEGFR-3(+) infiltrating mononuclear cells were exclusively observed in pSS MSGs. VEGFR-3 expression was strongly increased in lymphatic capillaries of pSS MSGs. IL-17(+) inflammatory cells were preferentially observed around lymphatic vessels in pSS MSGs. This study supports the notion that lymphvasculogenesis and lymphangiogenesis are active in pSS, thereby unmasking a novel aspect of disease pathogenesis. In addition, our results suggest another possible pathogenic role of IL-17 in pSS, further supporting its therapeutic targeting in this disease.


Asunto(s)
Células Endoteliales/patología , Interleucina-17/inmunología , Neovascularización Patológica/patología , Glándulas Salivales Menores/patología , Sialadenitis/diagnóstico , Síndrome de Sjögren/diagnóstico , Estudios de Casos y Controles , Diagnóstico Diferencial , Células Endoteliales/inmunología , Femenino , Regulación de la Expresión Génica , Humanos , Interleucina-17/genética , Linfangiogénesis/genética , Linfangiogénesis/inmunología , Vasos Linfáticos/inmunología , Vasos Linfáticos/patología , Masculino , Persona de Mediana Edad , Neovascularización Patológica/genética , Neovascularización Patológica/inmunología , Glándulas Salivales Menores/inmunología , Sialadenitis/genética , Sialadenitis/inmunología , Sialadenitis/patología , Transducción de Señal , Síndrome de Sjögren/genética , Síndrome de Sjögren/inmunología , Síndrome de Sjögren/patología , Células Th17/inmunología , Células Th17/patología , Factor C de Crecimiento Endotelial Vascular/genética , Factor C de Crecimiento Endotelial Vascular/inmunología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 3 de Factores de Crecimiento Endotelial Vascular/inmunología
11.
Haematologica ; 101(2): 219-25, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26494839

RESUMEN

Recent evidence suggests that patients with severe hemophilia B may have a less severe disease compared to severe hemophilia A. To investigate clinical, radiological, laboratory and histological differences in the arthropathy of severe hemophilia A and hemophilia B, 70 patients with hemophilia A and 35 with hemophilia B with at least one joint bleeding were consecutively enrolled. Joint bleedings (<10, 10-50, >50), regimen of treatment (prophylaxis/on demand), World Federation of Hemophilia, Pettersson and ultrasound scores, serum soluble RANK ligand and osteoprotegerin were assessed in all patients. RANK, RANK ligand and osteoprotegerin expression was evaluated in synovial tissue from 18 hemophilia A and 4 hemophilia B patients. The percentage of patients with either 10-50 or more than 50 hemarthrosis was greater in hemophilia A than in hemophilia B (P<0.001 and P=0.03, respectively), while that with less than 10 hemarthrosis was higher in hemophilia B (P<0.0001). World Federation of Hemophilia (36.6 vs. 20.2; P<0.0001) and ultrasound (10.9 vs. 4.3; P<0.0001) score mean values were significantly higher in hemophilia A patients. Serum osteoprotegerin and soluble RANK ligand were decreased in hemophilia A versus hemophilia B (P<0.0001 and P=0.006, respectively). Osteoprotegerin expression was markedly reduced in synovial tissue from hemophilia A patients. In conclusion, the reduced number of hemarthrosis, the lower World Federation of Hemophilia and ultrasound scores, and higher osteoprotegerin expression in serum and synovial tissue in hemophilia B suggest that hemophilia B is a less severe disease than hemophilia A. Osteoprotegerin reduction seems to play a pivotal role in the progression of arthropathy in hemophilia A.


Asunto(s)
Hemartrosis/patología , Hemofilia A/patología , Hemofilia B/patología , Osteoprotegerina/genética , Adolescente , Adulto , Anciano , Niño , Preescolar , Progresión de la Enfermedad , Femenino , Expresión Génica , Hemartrosis/complicaciones , Hemartrosis/diagnóstico por imagen , Hemartrosis/genética , Hemofilia A/complicaciones , Hemofilia A/diagnóstico por imagen , Hemofilia A/genética , Hemofilia B/complicaciones , Hemofilia B/diagnóstico por imagen , Hemofilia B/genética , Humanos , Cápsula Articular/química , Cápsula Articular/patología , Masculino , Persona de Mediana Edad , Osteoprotegerina/sangre , Ligando RANK/sangre , Ligando RANK/genética , Receptor Activador del Factor Nuclear kappa-B/sangre , Receptor Activador del Factor Nuclear kappa-B/genética , Índice de Severidad de la Enfermedad , Ultrasonografía
12.
Ann Rheum Dis ; 75(8): 1541-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26359450

RESUMEN

OBJECTIVES: In systemic sclerosis (SSc), vascular involvement is characterised by vascular endothelial growth factor (VEGF)-A/VEGF receptor (VEGFR) system disturbances. Neuropilin-1 (NRP1), a receptor for both class-3 semaphorins (Sema3s) and VEGF-A, is required for optimal VEGF-A/VEGFR-2 signalling. Here, we investigated the possible involvement of Sema3A/NRP1 axis in SSc. METHODS: Circulating Sema3A and soluble NRP1 (sNRP1) were measured in patients with SSc and controls. NRP1 and Sema3A expression in skin biopsies was evaluated by immunofluorescence and western blotting. NRP1 expression was assessed in SSc and healthy dermal microvascular endothelial cells (SSc-MVECs and H-MVECs), and in SSc and control endothelial progenitor cell (EPC)-derived endothelial cells (ECs). The possible impact of transcription factor Friend leukaemia integration 1 (Fli1) deficiency on endothelial NRP1 expression was investigated by gene silencing. The binding of Fli1 to NRP1 gene promoter was evaluated using chromatin immunoprecipitation. Capillary morphogenesis was performed on Matrigel. RESULTS: Decreased sNRP1 levels in SSc were associated with active and late nailfold videocapillaroscopy patterns and digital ulcers. No difference in Sema3A was found between patients and controls. NRP1 was significantly decreased in SSc-MVECs both ex vivo and in vitro. NRP1 and Fli1 significantly decreased in H-MVECs challenged with SSc sera, while they were not different in SSc and control EPC-derived ECs. Fli1 occupied the NRP1 gene promoter and Fli1 gene silencing reduced NRP1 expression in H-MVECs. NRP1 gene silencing in H-MVECs resulted in a significantly impaired angiogenic capacity comparable to that of cells treated with SSc sera. CONCLUSION: In SSc, NRP1 deficiency may be an additional factor in the perturbed VEGF-A/VEGFR-2 system contributing to peripheral microvasculopathy and defective angiogenesis.


Asunto(s)
Neovascularización Patológica/metabolismo , Neuropilina-1/metabolismo , Enfermedades Vasculares Periféricas/metabolismo , Esclerodermia Sistémica/metabolismo , Adulto , Anciano , Anciano de 80 o más Años , Biopsia , Células Cultivadas , Regulación hacia Abajo/fisiología , Células Endoteliales/metabolismo , Femenino , Humanos , Masculino , Angioscopía Microscópica/métodos , Persona de Mediana Edad , Neuropilina-1/deficiencia , Neuropilina-1/genética , Enfermedades Vasculares Periféricas/etiología , Proteína Proto-Oncogénica c-fli-1/deficiencia , Esclerodermia Sistémica/complicaciones , Esclerodermia Sistémica/patología , Semaforina-3A/sangre , Piel/irrigación sanguínea , Piel/patología
13.
PLoS One ; 10(6): e0130166, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26076019

RESUMEN

In systemic sclerosis (SSc), dermal capillaries are progressively lost with consequent chronic tissue hypoxia insufficiently compensated by angiogenesis. Clinical studies reported that intravenous cyclophosphamide (CYC) may improve SSc-related peripheral microvascular damage. Recently, we showed that CYC treatment may normalize SSc sera-induced abnormalities in endothelial cell-matrix interactions. Our objective was to evaluate in vitro the effects of sera from treatment-naïve or CYC-treated SSc patients on dermal blood microvascular endothelial cell (dMVEC) angiogenesis, migration, proliferation and apoptosis. dMVECs were challenged with sera from 21 SSc patients, treatment-naïve (n = 8) or under CYC treatment (n = 13), and 8 healthy controls. Capillary morphogenesis on Geltrex matrix was significantly reduced upon challenge with sera from naïve SSc patients compared with healthy controls. When dMVECs were challenged with sera from CYC-treated SSc patients, their angiogenic capacity was comparable to that of cells treated with healthy sera. Wound healing capacity and chemotaxis in Boyden chamber were both significantly decreased in the presence either of naïve or CYC-treated SSc sera compared with healthy sera. WST-1 assay revealed that cell proliferation was significantly decreased in dMVECs challenged with sera from naïve SSc patients compared with healthy sera. Conversely, dMVEC proliferation was not impaired in the presence of sera from CYC-treated SSc patients. Accordingly, the percentage of TUNEL-positive apoptotic dMVECs was significantly higher in the presence of sera from naïve SSc patients than healthy controls, while CYC-treated SSc sera did not induce dMVEC apoptosis. Levels of the angiostatic mediators endostatin, pentraxin 3, angiostatin and matrix metalloproteinase-12 were all significantly elevated in sera from naïve SSc patients compared with sera from both healthy controls and CYC-treated SSc patients. In SSc, CYC treatment might boost angiogenesis and consequently improve peripheral microangiopathy through the normalization of the endothelial cell-matrix interactions, reduction of endothelial cell apoptosis and rebalance of dysregulated angiostatic factors.


Asunto(s)
Ciclofosfamida/farmacología , Dermis/patología , Endotelio Vascular/patología , Neovascularización Patológica/patología , Esclerodermia Sistémica/patología , Antineoplásicos Alquilantes/farmacología , Apoptosis/efectos de los fármacos , Western Blotting , Estudios de Casos y Controles , Adhesión Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Dermis/efectos de los fármacos , Dermis/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Neovascularización Patológica/tratamiento farmacológico , Neovascularización Patológica/metabolismo , Pronóstico , Esclerodermia Sistémica/sangre , Esclerodermia Sistémica/tratamiento farmacológico , Cicatrización de Heridas
14.
Mediators Inflamm ; 2015: 751793, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25918479

RESUMEN

In recent years several studies investigated the role of T lymphocyte subpopulations in the pathogenesis of rheumatoid arthritis (RA). Pathogenic Th17 cells mediate pannus growth, osteoclastogenesis, and synovial neoangiogenesis; hence they are key players in the development of the disease. On the other hand, regulatory T (Treg) cells are a T cell subset whose peculiar function is to suppress autoreactive lymphocytes. The imbalance between Th17 and Treg cells has been identified as a crucial event in the pathogenesis of RA. In addition, the effects of currently employed RA therapeutic strategies on these lymphocyte subpopulations have been extensively investigated. This review article aims to discuss current knowledge on Treg and Th17 cells in RA and possible implications of their therapeutic targeting in this disorder.


Asunto(s)
Artritis Reumatoide/sangre , Artritis Reumatoide/terapia , Linfocitos T Reguladores/citología , Células Th17/citología , Abatacept/uso terapéutico , Corticoesteroides/uso terapéutico , Animales , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antirreumáticos/uso terapéutico , Epigénesis Genética , Humanos , Inflamación/inmunología , Linfocitos/citología , Ratones , Membrana Sinovial/metabolismo , Linfocitos T Reguladores/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
15.
J Cell Mol Med ; 19(7): 1689-96, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25753463

RESUMEN

It has been recently reported that telocytes, a stromal (interstitial) cell subset involved in the control of local tissue homeostasis, are hampered in the target organs of inflammatory/autoimmune disorders. Since no data concerning telocytes in minor salivary glands (MSGs) are currently available, aim of the study was to evaluate telocyte distribution in MSGs with normal architecture, non-specific chronic sialadenitis (NSCS) and primary Sjögren's syndrome (pSS)-focal lymphocytic sialadenitis. Twelve patients with pSS and 16 sicca non-pSS subjects were enrolled in the study. MSGs were evaluated by haematoxylin and eosin staining and immunofluorescence for CD3/CD20 and CD21 to assess focus score, Tarpley biopsy score, T/B cell segregation and germinal center (GC)-like structures. Telocytes were identified by immunoperoxidase-based immunohistochemistry for CD34 and CD34/platelet-derived growth factor receptor α double immunofluorescence. Telocytes were numerous in the stromal compartment of normal MSGs, where their long cytoplasmic processes surrounded vessels and encircled both the excretory ducts and the secretory units. In NSCS, despite the presence of a certain degree of inflammation, telocytes were normally represented. Conversely, telocytes were markedly reduced in MSGs from pSS patients compared to normal and NSCS MSGs. Such a decrease was associated with both worsening of glandular inflammation and progression of ectopic lymphoid neogenesis, periductal telocytes being reduced in the presence of smaller inflammatory foci and completely absent in the presence of GC-like structures. Our findings suggest that a loss of MSG telocytes might have important pathophysiological implications in pSS. The specific pro-inflammatory cytokine milieu of pSS MSGs might be one of the causes of telocyte loss.


Asunto(s)
Inflamación/patología , Tejido Linfoide/patología , Glándulas Salivales Menores/patología , Síndrome de Sjögren/patología , Telocitos/patología , Femenino , Centro Germinal/patología , Humanos , Sialadenitis/patología
16.
Clin Exp Rheumatol ; 33(2): 195-200, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25664492

RESUMEN

OBJECTIVES: The role of the lymphatic system in the connection between spondyloarthritis (SpA) and Crohn's disease (CD) remains yet to be elucidated. The aim of the present study was to investigate the circulating levels of lymphatic endothelial progenitor cells (LEPCs) and vascular endothelial growth factor-C (VEGF-C) and their possible correlation with clinical parameters in SpA, SpA associated with CD (SC), and CD. METHODS: Peripheral blood samples from SpA (n=36), SC (n=20) and CD (n=28) patients and 20 age- and sex-matched healthy controls were collected and used for quantification of circulating LEPCs and VEGF-C. LEPCs were identified by fluorescence-activated cell sorting using FITC-CD34, APC-CD133 and PE-VEGFR-3 antibodies. Serum levels of VEGF-C were measured by enzyme-linked immunosorbent assay. The possible correlations between disease duration (< or >10 years; < or >20 years) and clinical activity (BASDAI for SpA or CDAI for CD) and LEPC counts and VEGF-C levels were analysed. RESULTS: Circulating LEPC levels were significantly increased in SpA (p=0.0006) and SC (p=0.0058) patients compared with controls. In CD patients, LEPC counts negatively correlated with disease duration, with lower levels in longstanding disease (>20 years, p=0.018), but were not different from controls. No significant difference in VEGF-C levels was found in SpA, SC and CD compared with controls. Both LEPC and VEGF-C levels were independent of BASDAI and CDAI. CONCLUSIONS: On the basis of our observations, an active mobilisation of lymphatic endothelial cell precursors was observed only for spondylitis involvement.


Asunto(s)
Enfermedad de Crohn/diagnóstico , Células Progenitoras Endoteliales/patología , Endotelio Linfático/patología , Espondiloartritis/diagnóstico , Factor C de Crecimiento Endotelial Vascular/sangre , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Estudios de Casos y Controles , Recuento de Células , Separación Celular/métodos , Enfermedad de Crohn/sangre , Enfermedad de Crohn/patología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Estado de Salud , Humanos , Masculino , Persona de Mediana Edad , Valor Predictivo de las Pruebas , Índice de Severidad de la Enfermedad , Espondiloartritis/sangre , Espondiloartritis/patología , Encuestas y Cuestionarios , Factores de Tiempo
17.
Ann Rheum Dis ; 73(9): 1700-9, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23852693

RESUMEN

OBJECTIVE: Urokinase-type plasminogen activator receptor (uPAR) is a key component of the fibrinolytic system involved in extracellular matrix remodelling and angiogenesis. The cleavage/inactivation of uPAR is a crucial step in fibroblast-to-myofibroblast transition and has been implicated in systemic sclerosis (SSc) microvasculopathy. In the present study, we investigated whether uPAR gene inactivation in mice could result in tissue fibrosis and peripheral microvasculopathy resembling human SSc. METHODS: The expression of the native full-length form of uPAR in human skin biopsies was determined by immunohistochemistry. Skin and lung sections from uPAR-deficient (uPAR(-/-)) and wild-type (uPAR(+/+)) mice at 12 and 24 weeks of age were stained with haematoxylin-eosin, Masson's trichrome and Picrosirius red. Dermal thickness and hydroxyproline content in skin and lungs were quantified. Dermal myofibroblast and microvessel counts were determined by immunohistochemistry for α-smooth muscle actin and CD31, respectively. Endothelial cell apoptosis was assessed by TUNEL/CD31 immunofluorescence assay. RESULTS: Full-length uPAR expression was significantly downregulated in SSc dermis, especially in fibroblasts and endothelial cells. Dermal thickness, collagen content and myofibroblast counts were significantly greater in uPAR(-/-) than in uPAR(+/+) mice. In uPAR(-/-) mice, dermal fibrosis was paralleled by endothelial cell apoptosis and severe loss of microvessels. Lungs from uPAR(-/-) mice displayed non-specific interstitial pneumonia-like pathological features, both with inflammation and collagen deposition. Pulmonary pathology worsened significantly from 12 to 24 weeks, as shown by a significant increase in alveolar septal width and collagen content. CONCLUSIONS: uPAR(-/-) mice are a new animal model closely mimicking the histopathological features of SSc. This model warrants future studies.


Asunto(s)
Modelos Animales de Enfermedad , Fibrosis Pulmonar/genética , Receptores del Activador de Plasminógeno Tipo Uroquinasa/genética , Esclerodermia Sistémica/genética , Piel/patología , Adulto , Anciano , Animales , Apoptosis , Biopsia , Colágeno/metabolismo , Citocinas/metabolismo , Progresión de la Enfermedad , Endotelio Vascular/patología , Femenino , Fibrosis , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microvasos/patología , Persona de Mediana Edad , Miofibroblastos/patología , Enfermedades Vasculares Periféricas/genética , Enfermedades Vasculares Periféricas/metabolismo , Enfermedades Vasculares Periféricas/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/deficiencia , Receptores del Activador de Plasminógeno Tipo Uroquinasa/metabolismo , Esclerodermia Sistémica/metabolismo , Esclerodermia Sistémica/patología , Piel/irrigación sanguínea , Piel/metabolismo
18.
J Cell Mol Med ; 17(12): 1525-36, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24251911

RESUMEN

Crohn's disease (CD) is a relapsing chronic inflammatory disorder that may involve all the gastrointestinal tract with a prevalence of terminal ileum. Intestinal lesions have a characteristic discontinuous and segmental distribution and may affect all layers of the gut wall. Telocytes (TC), a peculiar type of stromal cells, have been recently identified in a variety of tissues and organs, including gastrointestinal tract of humans and mammals. Several roles have been proposed for TC, including mechanical support, spatial relationships with different cell types, intercellular signalling and modulation of intestinal motility. The aim of our study was to investigate the presence and distribution of TC in disease-affected and -unaffected ileal specimens from CD patients compared with controls. TC were identified by CD34/PDGFRα immunohistochemistry. In affected CD specimens TC disappeared, particularly where fibrosis and architectural derangement of the intestinal wall were observed. In the thickened muscularis mucosae and submucosa, few TC entrapped in the fibrotic extracellular matrix were found. A discontinuous network of TC was present around smooth muscle bundles, ganglia and enteric strands in the altered muscularis propria. At the myenteric plexus, the loss of TC network was paralleled by the loss of interstitial cells of Cajal network. In the unaffected CD specimens, TC were preserved in their distribution. Our results suggest that in CD the loss of TC might have important pathophysiological implications contributing to the architectural derangement of the intestinal wall and gut dysmotility. Further functional studies are necessary to better clarify the role of TC loss in CD pathophysiology.


Asunto(s)
Enfermedad de Crohn/patología , Íleon/patología , Adulto , Antígenos CD34/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Persona de Mediana Edad , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Células del Estroma/patología
19.
Arthritis Res Ther ; 15(5): R165, 2013 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-24286167

RESUMEN

INTRODUCTION: Microvascular damage and defective angiogenesis and vasculogenesis have a major role in the pathogenesis of systemic sclerosis (SSc). Epidermal growth factor-like domain 7 (EGFL7) is a proangiogenic molecule which is predominantly expressed and secreted by endothelial cells and their progenitors and controls vascular development and integrity. In this study, we investigated the possible involvement of EGFL7 in SSc. METHODS: Serum EGFL7 levels from 60 patients with SSc and 35 age- and sex-matched healthy controls were examined by colorimetric sandwich enzyme-linked immunosorbent assay. The expression of EGFL7 in forearm skin biopsies (n = 16 SSc, n = 10 controls), cultured dermal microvascular endothelial cells (MVECs) (n = 3 SSc, n = 3 controls) and late-outgrowth peripheral blood endothelial progenitor cell (EPC)-derived endothelial cells (n = 15 SSc, n = 8 controls) was investigated by immunofluorescence and Western blotting. RESULTS: Serum EGFL7 levels were detectable in 68.6% of healthy controls and 45% of SSc cases (P < 0.05). Circulating levels of EGFL7 were significantly decreased in SSc patients compared with healthy controls (P = 0.01). Serum levels of EGFL7 were significantly lower in both limited cutaneous SSc and diffuse cutaneous SSc patients than in controls (P = 0.02 and P = 0.04, respectively). In SSc, decreased serum EGFL7 levels were significantly correlated with the severity of nailfold capillary abnormalities. Patients with the most severe capillary changes and digital ulcers had serum EGFL7 levels significantly lower than healthy controls, while the EGFL7 levels did not differ significantly between controls and SSc patients with less capillary damage and lack of digital ulcers. Endothelial EGFL7 expression was strongly downregulated or even almost completely undetectable in SSc-affected dermis compared with controls (P < 0.001). In cultured SSc dermal MVECs and late-outgrowth peripheral blood EPC-derived endothelial cells, EGFL7 was significantly downregulated compared with cells obtained from healthy subjects (P < 0.01 and P < 0.001, respectively). CONCLUSIONS: Our findings suggest that the loss of EGFL7 expression in endothelial cells and their progenitors might play a role in the development and progression of peripheral microvascular damage and the defective vascular repair process characteristic of SSc.


Asunto(s)
Factores de Crecimiento Endotelial/genética , Neovascularización Patológica/genética , Esclerodermia Sistémica/genética , Esclerodermia Sistémica/fisiopatología , Adulto , Anciano , Biomarcadores/sangre , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/fisiopatología , Western Blotting , Proteínas de Unión al Calcio , Estudios de Casos y Controles , Regulación hacia Abajo , Familia de Proteínas EGF , Factores de Crecimiento Endotelial/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Regulación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Valores de Referencia , Esclerodermia Sistémica/metabolismo , Sensibilidad y Especificidad , Índice de Severidad de la Enfermedad , Estadísticas no Paramétricas
20.
J Cell Mol Med ; 17(9): 1099-108, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24151977

RESUMEN

Telocytes (TC), a cell population located in the connective tissue of many organs of humans and laboratory mammals, are characterized by a small cell body and extremely long and thin processes. Different TC subpopulations share unique ultrastructural features, but express different markers. In the gastrointestinal (GI) tract, cells with features of TC were seen to be CD34-positive/c-kit-negative and several roles have been proposed for them. Other interstitial cell types with regulatory roles described in the gut are the c-kit-positive/CD34-negative/platelet-derived growth factor receptor α (PDGFRα)-negative interstitial cells of Cajal (ICC) and the PDGFRα-positive/c-kit-negative fibroblast-like cells (FLC). As TC display the same features and locations of the PDGFRα-positive cells, we investigated whether TC and PDGFRα-positive cells could be the same cell type. PDGFRα/CD34, PDGFRα/c-kit and CD34/c-kit double immunolabelling was performed in full-thickness specimens from human oesophagus, stomach and small and large intestines. All TC in the mucosa, submucosa and muscle coat were PDGFRα/CD34-positive. TC formed a three-dimensional network in the submucosa and in the interstitium between muscle layers, and an almost continuous layer at the submucosal borders of muscularis mucosae and circular muscle layer. Moreover, TC encircled muscle bundles, nerve structures, blood vessels, funds of gastric glands and intestinal crypts. Some TC were located within the muscle bundles, displaying the same location of ICC and running intermingled with them. ICC were c-kit-positive and CD34/PDGFRα-negative. In conclusion, in the human GI tract the TC are PDGFRα-positive and, therefore, might correspond to the FLC. We also hypothesize that in human gut, there are different TC subpopulations probably playing region-specific roles.


Asunto(s)
Células del Tejido Conectivo/metabolismo , Tracto Gastrointestinal/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Adulto , Anciano , Antígenos CD34/metabolismo , Células del Tejido Conectivo/citología , Humanos , Inmunohistoquímica , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-kit/metabolismo , Ubiquitina Tiolesterasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA