Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Viruses ; 14(6)2022 06 19.
Article in English | MEDLINE | ID: mdl-35746808

ABSTRACT

Sulfated polysaccharides and other polyanions have been promising candidates in antiviral research for decades. These substances gained attention as antivirals when they demonstrated a high inhibitory effect in vitro against human immunodeficiency virus (HIV) and other enveloped viruses. However, that initial interest was followed by wide skepticism when in vivo assays refuted the initial results. In this paper we review the use of sulfated polysaccharides, and other polyanions, in antiviral therapy, focusing on extracellular polymeric substances (EPSs). We maintain that, in spite of those early difficulties, the use of polyanions and, specifically, the use of EPSs, in antiviral therapy should be reconsidered. We base our claim in several points. First, early studies showed that the main disadvantage of sulfated polysaccharides and polyanions is their low bioavailability, but this difficulty can be overcome by the use of adequate administration strategies, such as nebulization of aerosols to gain access to respiratory airways. Second, several sulfated polysaccharides and EPSs have demonstrated to be non-toxic in animals. Finally, these macromolecules are non-specific and therefore they might be used against different variants or even different viruses.


Subject(s)
Antiviral Agents , HIV Infections , Animals , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , Extracellular Polymeric Substance Matrix , HIV Infections/drug therapy , Polysaccharides/pharmacology
2.
Am J Pathol ; 187(7): 1537-1550, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28495592

ABSTRACT

Peritoneal dialysis (PD) is a life-saving form of renal replacement therapy for those with end-stage kidney disease. Mesothelial cells (MCs) line the peritoneal cavity and help define peritoneal response to treatment-associated injury, a major reason for treatment failure. miRNAs are important regulators, but their roles in peritoneal fibrosis are largely unknown. In this study, miR-21 was one of the most abundant miRNAs in primary MCs, and was up-regulated by the profibrotic cytokine transforming growth factor-ß1 and in PD effluent-derived MCs exhibiting mesenchymal phenotypic change. Increased miR-21 was found in peritoneal membrane biopsy specimens from PD patients compared to healthy controls (PD biocompatible, 5.86×, P = 0.0001; PD conventional, 7.09×, P < 0.0001, n = 11 per group). In PD effluent from a cohort of 230 patients, miR-21 was higher in those receiving the therapy long-term compared to new starters (n = 230, miR-21 3.26×, P = 0.001) and associated with icodextrin use (R = 0.52; 95% CI, 0.20-0.84), peritonitis count (R = 0.16; 95% CI, 0.03-0.29), and dialysate cytokines. miR-21 down-regulated programmed cell death 4 and programmed cell death 4 protein was decreased in peritoneal membrane biopsy specimens from PD patients compared to healthy controls. New miR-21 targets were identified that may be important during PD fibrogenesis. These data identify miR-21 as an important effector of fibrosis in the peritoneal membrane, and a promising biomarker in the dialysis effluent for membrane change in patients receiving PD.


Subject(s)
Gene Expression Regulation , Kidney Failure, Chronic/therapy , MicroRNAs/genetics , Peritoneal Fibrosis/genetics , Peritonitis/genetics , Biomarkers/analysis , Cells, Cultured , Cohort Studies , Down-Regulation , Epithelial Cells/metabolism , Epithelium/metabolism , Glucans/therapeutic use , Glucose/therapeutic use , Humans , Icodextrin , MicroRNAs/metabolism , Oligonucleotide Array Sequence Analysis , Peritoneal Dialysis , Peritoneal Fibrosis/metabolism , Peritoneum/metabolism , Peritonitis/metabolism , Treatment Failure , Up-Regulation
3.
Sci Rep ; 7: 44941, 2017 03 22.
Article in English | MEDLINE | ID: mdl-28327551

ABSTRACT

Peritoneal dialysis (PD) is an effective renal replacement therapy, but a significant proportion of patients suffer PD-related complications, which limit the treatment duration. Mesothelial-to-mesenchymal transition (MMT) contributes to the PD-related peritoneal dysfunction. We analyzed the genetic reprograming of MMT to identify new biomarkers that may be tested in PD-patients. Microarray analysis revealed a partial overlapping between MMT induced in vitro and ex vivo in effluent-derived mesothelial cells, and that MMT is mainly a repression process being higher the number of genes that are down-regulated than those that are induced. Cellular morphology and number of altered genes showed that MMT ex vivo could be subdivided into two stages: early/epithelioid and advanced/non-epithelioid. RT-PCR array analysis demonstrated that a number of genes differentially expressed in effluent-derived non-epithelioid cells also showed significant differential expression when comparing standard versus low-GDP PD fluids. Thrombospondin-1 (TSP1), collagen-13 (COL13), vascular endothelial growth factor A (VEGFA), and gremlin-1 (GREM1) were measured in PD effluents, and except GREM1, showed significant differences between early and advanced stages of MMT, and their expression was associated with a high peritoneal transport status. The results establish a proof of concept about the feasibility of measuring MMT-associated secreted protein levels as potential biomarkers in PD.


Subject(s)
Cellular Reprogramming/genetics , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial-Mesenchymal Transition/genetics , Genomics , Peritoneal Dialysis , Biomarkers , Dialysis Solutions/chemistry , Gene Expression Profiling , Genomics/methods , Glycolysis , Humans , Peritoneal Dialysis/adverse effects , Transcriptome
4.
Biomed Res Int ; 2015: 989560, 2015.
Article in English | MEDLINE | ID: mdl-26688823

ABSTRACT

Preservation of peritoneal membrane (PM) is essential for long-term survival in peritoneal dialysis (PD). Continuous presence of PD fluids (PDF) in the peritoneal cavity generates chronic inflammation and promotes changes of the PM, such as fibrosis, angiogenesis, and lymphangiogenesis. Mesothelial-to-mesenchymal transition (MMT) and endothelial-to-mesenchymal transition (Endo-MT) seem to play a central role in this pathogenesis. We speculated that Rapamycin, a potent immunosuppressor, could be beneficial by regulating blood and lymphatic vessels proliferation. We demonstrate that mice undergoing a combined PD and Rapamycin treatment (PDF + Rapa group) presented a reduced PM thickness and lower number of submesothelial blood and lymphatic vessels, as well as decreased MMT and Endo-MT, comparing with their counterparts exposed to PD alone (PDF group). Peritoneal water transport in the PDF + Rapa group remained at control level, whereas PD effluent levels of VEGF, TGF-ß, and TNF-α were lower than in the PDF group. Moreover, the treatment of mesothelial cells with Rapamycin in vitro significantly decreased VEGF synthesis and selectively inhibited the VEGF-C and VEGF-D release when compared with control cells. Thus, Rapamycin has a protective effect on PM in PD through an antifibrotic and antiproliferative effect on blood and lymphatic vessels. Moreover, it inhibits Endo-MT and, at least partially, MMT.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Lymphangiogenesis/drug effects , Membranes, Artificial , Neovascularization, Physiologic/drug effects , Peritoneal Dialysis/adverse effects , Sirolimus/pharmacology , Animals , Cytokines/blood , Female , Mice
5.
J Pathol ; 231(4): 517-31, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24114721

ABSTRACT

Peritoneal dissemination is a frequent metastatic route for cancers of the ovary and gastrointestinal tract. Tumour cells metastasize by attaching to and invading through the mesothelial cell (MC) monolayer that lines the peritoneal cavity. Metastases are influenced by carcinoma-associated fibroblasts (CAFs), a cell population that derives from different sources. Hence, we investigated whether MCs, through mesothelial-mesenchymal transition (MMT), were a source of CAFs during peritoneal carcinomatosis and whether MMT affected the adhesion and invasion of tumour cells. Biopsies from patients with peritoneal dissemination revealed the presence of myofibroblasts expressing mesothelial markers in the proximity of carcinoma implants. Prominent new vessel formation was observed in the peritoneal areas harbouring tumour cells when compared with tumour-free regions. The use of a mouse model of peritoneal dissemination confirmed the myofibroblast conversion of MCs and the increase in angiogenesis at places of tumour implants. Treatment of omentum MCs with conditioned media from carcinoma cell cultures resulted in phenotype changes reminiscent of MMT. Adhesion experiments demonstrated that MMT enhanced the binding of cancer cells to MCs in a ß1-integrin-dependent manner. Scanning electron microscopy imaging showed that the enhanced adhesion was mostly due to increased cell-cell interaction and not to a mere matrix exposure. Invasion assays suggested a reciprocal stimulation of the invasive capacity of tumour cells and MCs. Our results demonstrate that CAFs can derive from mesothelial cells during peritoneal metastasis. We suggest that MMT renders the peritoneum more receptive for tumour cell attachment/invasion and contributes to secondary tumour growth by promoting its vascularization.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Fibroblasts/pathology , Peritoneal Neoplasms/secondary , Animals , Biopsy , Cell Adhesion , Cell Line, Tumor , Colorectal Neoplasms/pathology , Culture Media, Conditioned/pharmacology , Epithelial Cells/pathology , Epithelial Cells/physiology , Epithelial Cells/ultrastructure , Epithelial-Mesenchymal Transition/drug effects , Female , Fibroblasts/physiology , Heterografts , Humans , Mice , Mice, Nude , Microscopy, Electron, Scanning , Neoplasm Invasiveness , Neoplasm Transplantation , Neovascularization, Pathologic/pathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/blood supply , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/ultrastructure
6.
J Am Soc Nephrol ; 22(9): 1682-95, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21742730

ABSTRACT

During peritoneal dialysis (PD), mesothelial cells undergo mesothelial-to-mesenchymal transition (MMT), a process associated with peritoneal-membrane dysfunction. Because TGF-ß1 can induce MMT, we evaluated the efficacy of TGF-ß1-blocking peptides in modulating MMT and ameliorating peritoneal damage in a mouse model of PD. Exposure of the peritoneum to PD fluid induced fibrosis, angiogenesis, functional impairment, and the accumulation of fibroblasts. In addition to expressing fibroblast-specific protein-1 (FSP-1), some fibroblasts co-expressed cytokeratin, indicating their mesothelial origin. These intermediate-phenotype (Cyto(+)/FSP-1(+)) fibroblasts had features of myofibroblasts with fibrogenic capacity. PD fluid treatment triggered the appearance of CD31(+)/FSP-1(+) and CD45(+)/FSP-1(+) cells, suggesting that fibroblasts also originate from endothelial cells and from cells recruited from bone marrow. Administration of blocking peptides significantly ameliorated fibrosis and angiogenesis, improved peritoneal function, and reduced the number of FSP-1(+) cells, especially in the Cyto(+)/FSP-1(+) subpopulation. Conversely, overexpression of TGF-ß1 in the peritoneum by adenovirus-mediated gene transfer led to a marked accumulation of fibroblasts, most of which derived from the mesothelium. Taken together, these results demonstrate that TGF-ß1 drives the peritoneal deterioration induced by dialysis fluid and highlights a role of TGF-ß1-mediated MMT in the pathophysiology of peritoneal-membrane dysfunction.


Subject(s)
Cell Transdifferentiation/drug effects , Peritoneal Dialysis/adverse effects , Peritoneal Fibrosis/etiology , Peritoneum/pathology , Transforming Growth Factor beta1/metabolism , Animals , Biomarkers/metabolism , Cells, Cultured , Dialysis Solutions/adverse effects , Female , Injections, Intraperitoneal , Keratins/metabolism , Mice , Mice, Inbred C57BL , Myofibroblasts/metabolism , Myofibroblasts/pathology , Peptide Fragments/pharmacology , Peptide Fragments/therapeutic use , Peptides/pharmacology , Peptides/therapeutic use , Peritoneal Fibrosis/metabolism , Peritoneal Fibrosis/pathology , Peritoneal Fibrosis/prevention & control , Phenotype , Receptors, Transforming Growth Factor beta/therapeutic use , S100 Calcium-Binding Protein A4 , S100 Proteins/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL