Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 23(9)2022 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-35563220

RESUMO

Peritoneal dialysis (PD) is an efficient renal replacement therapy for patients with end-stage renal disease. Even if it ensures an outcome equivalent to hemodialysis and a better quality of life, in the long-term, PD is associated with the development of peritoneal fibrosis and the consequents patient morbidity and PD technique failure. This unfavorable effect is mostly due to the bio-incompatibility of PD solution (mainly based on high glucose concentration). In the present review, we described the mechanisms and the signaling pathway that governs peritoneal fibrosis, epithelial to mesenchymal transition of mesothelial cells, and angiogenesis. Lastly, we summarize the present and future strategies for developing more biocompatible PD solutions.


Assuntos
Diálise Peritoneal , Fibrose Peritoneal , Soluções para Diálise/metabolismo , Transição Epitelial-Mesenquimal , Humanos , Diálise Peritoneal/efeitos adversos , Fibrose Peritoneal/etiologia , Fibrose Peritoneal/metabolismo , Fibrose Peritoneal/terapia , Peritônio/patologia , Qualidade de Vida
2.
Semin Cancer Biol ; 62: 86-98, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31348993

RESUMO

Heparanase (HPSE) is an endoglycosidase that catalyses the cutting of the side chains of heparan-sulphate proteoglycans (HS), thus determining the remodelling of the extracellular matrix and basement membranes, as well as promoting the release of different HS-related molecules as growth factors, cytokines and enzymes. Ever since the HPSE was identified in the late 1980s, several experimental studies have shown that its overexpression was instrumental in increasing tumor growth, metastatic dissemination, angiogenesis and inflammation. More recently, HPSE involvment has also been demonstrated in mediating tumor-host crosstalk, in inducing gene transcription, in the activation of signaling pathways and in the formation of exosomes and in autophagy. All of these activities (enzymatic and non-enzymatic) together make heparanase a multifunctional molecule that increases the aggressiveness and chemo-resistance of tumor cells. Conversely, heparanase gene-silencing or tumor treatment with compounds that inhibit heparanase activity have been shown to significantly attenuate tumor progression in different animal models of tumorigenesis, further emphasizing the therapeutic potential of anti-heparanase therapy for several types of neoplasms. This review focuses on present knowledge and recent development in the study of heparanase in cancer progression as well as on novel mechanisms by which heparanase regulates tumor metastasis and chemo-resistance. Moreover, recent advances in strategies for its inhibition as a potential therapeutic option will be discussed.


Assuntos
Glucuronidase/genética , Glucuronidase/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Animais , Autofagia , Coagulação Sanguínea , Gerenciamento Clínico , Progressão da Doença , Suscetibilidade a Doenças , Resistencia a Medicamentos Antineoplásicos , Exossomos/metabolismo , Matriz Extracelular/metabolismo , Glucuronidase/antagonistas & inibidores , Humanos , Inflamação/complicações , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Espaço Intracelular/metabolismo , Terapia de Alvo Molecular , Neoplasias/patologia , Neoplasias/terapia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Microambiente Tumoral
3.
Int J Mol Sci ; 22(6)2021 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-33804258

RESUMO

The endothelial glycocalyx, the gel layer covering the endothelium, is composed of glycosaminoglycans, proteoglycans, and adsorbed plasma proteins. This structure modulates vessels' mechanotransduction, vascular permeability, and leukocyte adhesion. Thus, it regulates several physiological and pathological events. In the present review, we described the mechanisms that disturb glycocalyx stability such as reactive oxygen species, matrix metalloproteinases, and heparanase. We then focused our attention on the role of glycocalyx degradation in the induction of profibrotic events and on the possible pharmacological strategies to preserve this delicate structure.


Assuntos
Endotélio/química , Fibrose/genética , Glicocálix/química , Mecanotransdução Celular/genética , Proteínas Sanguíneas/química , Proteínas Sanguíneas/genética , Permeabilidade Capilar/genética , Endotélio/ultraestrutura , Fibrose/patologia , Glucuronidase/efeitos adversos , Glicocálix/genética , Glicocálix/ultraestrutura , Glicosaminoglicanos/química , Glicosaminoglicanos/genética , Humanos , Metaloproteinases da Matriz/efeitos adversos , Proteoglicanas/química , Proteoglicanas/genética , Espécies Reativas de Oxigênio/efeitos adversos
4.
Adv Exp Med Biol ; 1221: 669-684, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32274731

RESUMO

Organ fibrosis is defined as a deregulated wound-healing process characterized by a progressive accumulation of fibrous tissue and by reduced remodeling that can lead to the loss of functionality of the affected organ. This pathological process is quite common in several parenchymal organs such as kidneys, liver, and lungs and represents a real health emergency in developed western countries since a real anti-fibrotic therapy is not yet available in most cases. Heparanase (HPSE), which is the enzyme that cuts off the side chains of heparan sulfate (HS) proteoglycan, appears to be involved in the aetiopathogenesis of fibrosis in all these organs, even if with different mechanisms. Here we discuss how the interplay between HPSE and components of the immune and inflammatory responses can activate recruitment, proliferation, and activation of myofibroblasts which represent the main cell type responsible for the deposition of fibrous matrix. Finally, bearing in mind that once the activity of HPSE is inhibited no other molecule is able to perform the same function, it is desirable that this enzyme could prove to be a suitable pharmacological target in anti-fibrotic therapy.


Assuntos
Fibrose/enzimologia , Fibrose/patologia , Glucuronidase/metabolismo , Fibrose/metabolismo , Proteoglicanas de Heparan Sulfato , Humanos , Miofibroblastos , Especificidade de Órgãos , Cicatrização
5.
J Transl Med ; 17(1): 12, 2019 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-30616602

RESUMO

BACKGROUND: The epithelial to mesenchymal transition (EMT) is a multi-factorial biological mechanism involved in renal and hepatic fibrosis and the IL-1 beta has been assumed as a mediator of this process although data are not exhaustive. Therefore, the aim of our study was to evaluate the role of this cytokine in the EMT of renal proximal tubular epithelial cells (HK-2) and stellate cells (LX-2) and the protective/anti-fibrotic effect of its inhibition by Canakinumab (a specific human monoclonal antibody targeted against IL-1beta). METHODS: Both cell types were treated with IL-1 beta (10 ng/ml) for 6 and 24 h with and without Canakinumab (5 µg/ml). As control we used TGF-beta (10 ng/ml). Expression of EMT markers (vimentin, alpha-SMA, fibronectin) were evaluated through western blotting and immunofluorescence. Genes expression for matrix metalloproteinases (MMP)-2 was measured by Real-Time PCR and enzymatic activity by zymography. Cellular motility was assessed by scratch assay. RESULTS: IL-1 beta induced a significant up-regulation of EMT markers in both cell types and increased the MMP-2 protein expression and enzymatic activity, similarly to TGF-beta. Moreover, IL-1 beta induced a higher rate of motility in HK-2. Canakinumab prevented all these modifications in both cell types. CONCLUSIONS: Our results clearly demonstrate the role of IL-1 beta in the EMT of renal/stellate cells and it underlines, for the first time, the therapeutic potential of its specific inhibition on the prevention/minimization of organ fibrosis.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Células Estreladas do Fígado/patologia , Interleucina-1beta/farmacologia , Túbulos Renais/patologia , Anticorpos Monoclonais Humanizados/farmacologia , Biomarcadores/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Células Estreladas do Fígado/efeitos dos fármacos , Células Estreladas do Fígado/enzimologia , Humanos , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/enzimologia , Metaloproteinase 2 da Matriz/metabolismo , Fator de Crescimento Transformador beta1/farmacologia
6.
FASEB J ; 32(2): 742-756, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-28970256

RESUMO

Heparanase (HPSE) is part of the biologic network triggered by ischemia/reperfusion (I/R) injury, a complication of renal transplantation and acute kidney injury. During this period, the kidney or graft undergoes a process of macrophages recruitment and activation. HPSE may therefore control these biologic effects. We measured the ability of HPSE and its inhibitor, SST0001, to regulate macrophage polarization and the crosstalk between macrophages and HK-2 renal tubular cells during in vitro hypoxia/reoxygenation (H/R). Furthermore, we evaluated in vivo renal inflammation, macrophage polarization, and histologic changes in mice subjected to monolateral I/R and treated with SST0001 for 2 or 7 d. The in vitro experiments showed that HPSE sustained M1 macrophage polarization and modulated apoptosis, the release of damage associated molecular patterns in post-H/R tubular cells, the synthesis of proinflammatory cytokines, and the up-regulation of TLRs on both epithelial cells and macrophages. HPSE also regulated M1 polarization induced by H/R-injured tubular cells and the partial epithelial-mesenchymal transition of these epithelial cells by M1 macrophages. All these effects were prevented by inhibiting HPSE. Furthermore, the inhibition of HPSE in vivo reduced inflammation and M1 polarization in mice undergoing I/R injury, partially restored renal function and normal histology, and reduced apoptosis. These results show for the first time that HPSE regulates macrophage polarization as well as renal damage and repair after I/R. HPSE inhibitors could therefore provide a new pharmacologic approach to minimize acute kidney injury and to prevent the chronic profibrotic damages induced by I/R.-Masola, V., Zaza, G., Bellin, G., Dall'Olmo, L., Granata, S., Vischini, G., Secchi, M. F., Lupo, A., Gambaro, G., Onisto, M. Heparanase regulates the M1 polarization of renal macrophages and their crosstalk with renal epithelial tubular cells after ischemia/reperfusion injury.


Assuntos
Células Epiteliais/enzimologia , Glucuronidase/metabolismo , Nefropatias/enzimologia , Túbulos Renais/enzimologia , Macrófagos/enzimologia , Traumatismo por Reperfusão/enzimologia , Animais , Células Epiteliais/patologia , Nefropatias/patologia , Túbulos Renais/lesões , Túbulos Renais/patologia , Macrófagos/patologia , Camundongos , Traumatismo por Reperfusão/patologia
7.
Nephrol Dial Transplant ; 32(7): 1145-1154, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-28064160

RESUMO

BACKGROUND: Epithelial-to-mesenchymal transition (EMT) of peritoneal mesothelial cells induced by high glucose (HG) levels is a major biological mechanism leading to myofibroblast accumulation in the omentum of patients on peritoneal dialysis (PD). Heparanase (HPSE), an endoglycosidase that cleaves heparan sulfate chains, is involved in the EMT of several cell lines, and may have a major role in this pro-fibrotic process potentially responsible for the failure of dialysis. Its specific inhibition may therefore plausibly minimize this pathological condition. METHODS: An in vitro study employing several biomolecular strategies was conducted to assess the role of HPSE in the HG-induced mesothelial EMT process, and to measure the effects of its specific inhibition by SST0001, a N-acetylated glycol-split heparin with a strong anti-HPSE activity. Rat mesothelial cells were grown for 6 days in HG (200 mM) culture medium with or without SST0001. Then EMT markers (VIM, α-SMA, TGF-ß) and vascular endothelial growth factor (VEGF) (a factor involved in neoangiogenesis) were measured by real-time PCR and immunofluorescence/western blotting. As a functional analysis, trans-epithelial resistance (TER) and permeability to albumin were also measured in our in vitro model using a Millicell-ERS ohmmeter and a spectrophotometer, respectively. RESULTS: Our results showed that 200 mM of glucose induced a significant gene and protein up-regulation of VEGF and all EMT markers after 6 days of culture. Intriguingly, adding SST0001 on day 3 reversed these biological and cellular effects. HPSE inhibition also restored the normal TER and permeability lost during the HG treatment. CONCLUSION: Taken together, our data confirm that HG can induce EMT of mesothelial cells, and that HPSE plays a central part in this process. Our findings also suggest that pharmacological HPSE inhibition could prove a valuable therapeutic tool for minimizing fibrosis and avoiding a rapid decline in the efficacy of dialysis in patients on PD, though clinical studies and/or trials would be needed to confirm the clinical utility of this treatment.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Epitélio/efeitos dos fármacos , Fibrose/fisiopatologia , Glucose/farmacologia , Glucuronidase/antagonistas & inibidores , Heparina/análogos & derivados , Peritônio/efeitos dos fármacos , Animais , Células Cultivadas , Epitélio/metabolismo , Epitélio/patologia , Heparina/farmacologia , Peritônio/metabolismo , Peritônio/patologia , Ratos , Fator de Crescimento Transformador beta/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
J Pathol ; 238(1): 7-9, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26387558

RESUMO

Vitamin D deficiency is inevitable in chronic kidney diseases. Clinical and experimental therapies with vitamin D supplements or analogues have demonstrated nephroprotective effects, which vitamin D exerts partly by controlling the renin-angiotensin-aldosterone system, but also by modulating other signalling pathways. In recent work published in the Journal of Pathology, Garsen and colleagues identified heparanase as a novel target of vitamin D and its antiproteinuric activity. Heparanase is an endoglycosidase with a role in remodelling the extracellular matrix through its ability to degrade heparan sulphate, and is involved in the pathogenesis of several proteinuric and fibrotic renal diseases. The new evidence that vitamin D inhibits heparanase expression sets the stage for a better understanding of the vitamin's kidney-protecting effects and its possible application to proteinuric and non-proteinuric chronic kidney diseases.


Assuntos
Calcitriol/farmacologia , Glucuronidase/metabolismo , Podócitos/enzimologia , Proteinúria/metabolismo , Animais
9.
Biochim Biophys Acta ; 1843(9): 2122-8, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24937189

RESUMO

Epithelial-mesenchymal transition (EMT) of tubular cells is one of the mechanisms which contribute to renal fibrosis and transforming growth factor-ß (TGF-ß) is one of the main triggers. Heparanase (HPSE) is an endo-ß-D-glucuronidase that cleaves heparan-sulfate thus regulating the bioavailability of growth factors (FGF-2, TGF-ß). HPSE controls FGF-2-induced EMT in tubular cells and is necessary for the development of diabetic nephropathy in mice. The aim of this study was to investigate whether HPSE can modulate the expression and the effects of TGF-ß in tubular cells. First we proved that the lack of HPSE or its inhibition prevents the increased synthesis of TGF-ß by tubular cells in response to pro-fibrotic stimuli such as FGF-2, advanced glycosylation end products (AGE) and albumin overload. Second, since TGF-ß may derive from sources different from tubular cells, we investigated whether HPSE modulates tubular cell response to exogenous TGF-ß. HPSE does not prevent EMT induced by TGF-ß although it slows its onset; indeed in HPSE-silenced cells the acquisition of a mesenchymal phenotype does not develop as quickly as in wt cells. Additionally, TGF-ß induces an autocrine loop to sustain its signal, whereas the lack of HPSE partially interferes with this autocrine loop. Overall these data confirm that HPSE is a key player in renal fibrosis since it interacts with the regulation and the effects of TGF-ß. HPSE is needed for pathological TGF-ß overexpression in response to pro-fibrotic factors. Furthermore, HPSE modulates TGF-ß-induced EMT: the lack of HPSE delays tubular cell transdifferentiation, and impairs the TGF-ß autocrine loop.


Assuntos
Glucuronidase/metabolismo , Rim/metabolismo , Rim/patologia , Fator de Crescimento Transformador beta/genética , Actinas/genética , Actinas/metabolismo , Animais , Comunicação Autócrina , Biomarcadores/metabolismo , Linhagem Celular , Fibronectinas/genética , Fibronectinas/metabolismo , Fibrose , Regulação da Expressão Gênica , Glucuronidase/genética , Humanos , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Mesoderma/metabolismo , Mesoderma/patologia , Camundongos , Fator de Crescimento Transformador beta/metabolismo , Vimentina/genética , Vimentina/metabolismo
10.
J Transl Med ; 13: 181, 2015 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-26040666

RESUMO

Tubulo-interstitial fibrosis has been recognized as the hallmark of progression of chronic kidney disease, but, despite intensive research studies, there are currently no biomarkers or effective treatments for this condition. In this context, a promising candidate could be heparanase-1 (HPSE), an endoglycosidase that cleaves heparan sulfate chains and thus takes part in extracellular matrix remodeling. As largely described, it has a central role in the pathogenesis of cancer and inflammation, and it participates in the complex biological machinery involved in the onset of different renal proteinuric diseases (e.g., diabetic nephropathy, glomerulonephritis). Additionally, HPSE may significantly influence the progression of chronic kidney damage trough its major role in the biological pathway of renal fibrogenesis. Here, we briefly summarize data supporting the role of HPSE in renal damage, focusing on recent evidences that demonstrate the capability of this enzyme to modulate the signaling of pro-fibrotic factors such as FGF-2 and TGF-ß and consequently to control the epithelial-mesenchymal transition in renal tubular cells. We also emphasize the need of the research community to undertake studies and clinical trials to assess the potential clinical employment of this enzyme as diagnostic and prognostic tool and/or its role as therapeutic target for new pharmacological interventions.


Assuntos
Glucuronidase/metabolismo , Rim/enzimologia , Rim/patologia , Biomarcadores/metabolismo , Transição Epitelial-Mesenquimal , Fibrose , Humanos , Terapia de Alvo Molecular
11.
J Biol Chem ; 287(2): 1478-88, 2012 Jan 06.
Artigo em Inglês | MEDLINE | ID: mdl-22102278

RESUMO

The epithelial-mesenchymal transition (EMT) of proximal tubular epithelial cells (PTECs) into myofibroblasts contributes to the establishment of fibrosis that leads to end stage renal disease. FGF-2 induces EMT in PTECs. Because the interaction between FGF-2 and its receptor is mediated by heparan sulfate (HS) and syndecans, we speculated that a deranged HS/syndecans regulation impairs FGF-2 activity. Heparanase is crucial for the correct turnover of HS/syndecans. The aim of the present study was to assess the role of heparanase on epithelial-mesenchymal transition induced by FGF-2 in renal tubular cells. In human kidney 2 (HK2) PTEC cultures, although FGF-2 induces EMT in the wild-type clone, it is ineffective in heparanase-silenced cells. The FGF-2 induced EMT is through a stable activation of PI3K/AKT which is only transient in heparanase-silenced cells. In PTECs, FGF-2 induces an autocrine loop which sustains its signal through multiple mechanisms (reduction in syndecan-1, increase in heparanase, and matrix metalloproteinase 9). Thus, heparanase is necessary for FGF-2 to produce EMT in PTECs and to sustain FGF-2 intracellular signaling. Heparanase contributes to a synergistic loop for handling syndecan-1, facilitating FGF-2 induced-EMT. In conclusion, heparanase plays a role in the tubular-interstitial compartment favoring the FGF-2-dependent EMT of tubular cells. Hence, heparanase is an interesting pharmacological target for the prevention of renal fibrosis.


Assuntos
Transição Epitelial-Mesenquimal , Fator 2 de Crescimento de Fibroblastos/metabolismo , Glucuronidase/metabolismo , Falência Renal Crônica/metabolismo , Túbulos Renais/metabolismo , Sindecana-1/metabolismo , Comunicação Autócrina/genética , Linhagem Celular , Ativação Enzimática/genética , Fator 2 de Crescimento de Fibroblastos/genética , Fibrose , Glucuronidase/genética , Humanos , Falência Renal Crônica/patologia , Túbulos Renais/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/genética , Sindecana-1/genética
12.
J Transl Med ; 11: 292, 2013 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-24256696

RESUMO

BACKGROUND: Everolimus (EVE) is a drug widely used in several renal transplant protocols. Although characterized by a relatively low nephrotoxicity, it may induce several adverse effects including severe fibro-interstitial pneumonitis. The exact molecular/biological mechanism associated to these pro-fibrotic effects is unknown, but epithelial to mesenchymal transition (EMT) may have a central role. Additionally, heparanase, an enzyme recently associated with the progression of chronic allograft nephropathy, could contribute to activate this machinery in renal cells. METHODS: Several biomolecular strategies (RT-PCR, immunofluorescence, zymography and migration assay) have been used to assess the capability of EVE (10, 100, 200 and 500 nM) to induce an in vitro heparanase-mediated EMT in wild-type (WT) and Heparanase (HPSE)-silenced immortalized human renal epithelial proximal tubular cells (HK-2). Additionally, microarray technology was used to find additional biological elements involved in EVE-induced EMT. RESULTS: Biomolecular experiments demonstrated a significant up-regulation (more than 1.5 fold increase) of several genes encoding for well known EMT markers [(alpha-smooth muscle actin (α-SMA), Vimentin (VIM), Fibronectin (FN) and matrix metalloproteinase-9 (MMP9)], enhancement of MMP9 protein level and increment of cells motility in WT HK2 cells treated with high concentrations of EVE (higher than 100 nM). Similarly, immunofluorescence analysis showed that 100 nM of EVE increased α-SMA, VIM and FN protein expression in WT HK2 cells. All these effects were absent in both HPSE- and AKT-silenced cell lines. AKT is a protein having a central role in EMT. Additionally, microarray analysis identified other 2 genes significantly up-regulated in 100 nM EVE-treated cells (p < 0.005 and FDR < 5%): transforming growth factor beta-2 (TGFß2) and epidermal growth factor receptor (EGFR). Real-time PCR analysis validated microarray. CONCLUSIONS: Our in vitro study reveals new biological/cellular aspects of the pro-fibrotic activity of EVE and it demonstrates, for the first time, that an heparanase-mediated EMT of renal tubular cells may be activated by high doses of this drug. Additionally, our results suggest that clinicians should administer the adequate dosage of EVE in order to increase efficacy and reduce adverse effects. Finally heparanase could be a new potential therapeutic target useful to prevent/minimize drug-related systemic fibrotic adverse effects.


Assuntos
Transição Epitelial-Mesenquimal/efeitos dos fármacos , Glucuronidase/metabolismo , Imunossupressores/farmacologia , Túbulos Renais Proximais/efeitos dos fármacos , Sirolimo/análogos & derivados , Actinas/genética , Linhagem Celular Transformada , Everolimo , Fibronectinas/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/enzimologia , Metaloproteinase 9 da Matriz/genética , Sirolimo/farmacologia , Vimentina/genética
13.
Ann Anat ; 250: 152115, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37315628

RESUMO

BACKGROUND: accumulating evidence indicates that during tail regeneration in lizards the initial stage of regenerative blastema is a tumor-like proliferative outgrowth that rapidly elongates into a new tail composed of fully differentiated tissues. Both oncogenes and tumor-suppressors are expressed during regeneration, and it has been hypothesized that an efficient control of cell proliferation avoids that the blastema is turned into a tumor outgrowth. METHODS: in order to determine whether functional tumor-suppressors are present in the growing blastema we have utilized protein extracts collected from early regenerating tails of 3-5 mm that have been tested for a potential anti-tumor effect on in-vitro culture by using cancer cell lines from human mammary gland (MDA-MB-231) and prostate cancer (DU145). RESULTS: at specific dilutions, the extract determines a reduction of viability in cancer cells after 2-4 days of culture, as supported by statistical and morphological analyses. While control cells appear viable, treated cells result damaged and produce an intense cytoplasmic granulation and degeneration. CONCLUSIONS: this negative effect on cell viability and proliferation is absent using tissues from the original tail supporting the hypothesis that only regenerating tissues synthesize tumor-suppressor molecules. The study suggests that the regenerating tail of lizard at the stages here selected contains some molecules that determine inhibition of cell viability on the cancer cells analyzed.


Assuntos
Lagartos , Neoplasias , Masculino , Animais , Humanos , Lagartos/fisiologia , Regeneração/fisiologia , Diferenciação Celular
14.
Biomedicines ; 11(7)2023 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-37509428

RESUMO

We investigated how the extracellular matrix (ECM) affects LoVo colorectal cancer cells behavior during a spatiotemporal invasion. Epithelial-to-mesenchymal transition (EMT) markers, matrix-degrading enzymes, and morphological phenotypes expressed by LoVo-S (doxorubicin-sensitive) and higher aggressive LoVo-R (doxorubicin-resistant) were evaluated in cells cultured for 3 and 24 h on Millipore filters covered by Matrigel, mimicking the basement membrane, or type I Collagen reproducing a desmoplastic lamina propria. EMT and invasiveness were investigated with RT-qPCR, Western blot, and scanning electron microscopy. As time went by, most gene expressions decreased, but in type I Collagen samples, a strong reduction and high increase in MMP-2 expression in LoVo-S and -R cells occurred, respectively. These data were confirmed by the development of an epithelial morphological phenotype in LoVo-S and invading phenotypes with invadopodia in LoVo-R cells as well as by protein-level analysis. We suggest that the duration of culturing and type of substrate influence the morphological phenotype and aggressiveness of both these cell types differently. In particular, the type I collagen meshwork, consisting of large fibrils confining inter fibrillar micropores, affects the two cell types differently. It attenuates drug-sensitive LoVo-S cell aggressiveness but improves a proteolytic invasion in drug-resistant LoVo-R cells as time goes by. Experimental studies on CRC cells should examine the peri-tumoral ECM components, as well as the dynamic physical conditions of TME, which affect the behavior and aggressiveness of both drug-sensitive and drug-resistant LoVo cells differently.

15.
Biochim Biophys Acta ; 1813(8): 1475-82, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21600934

RESUMO

Diabetic nephropathy is one of the main causes of end-stage renal disease, in which the development of tubular damage depends on factors such as high glucose levels, albuminuria and advanced glycation end-product. In this study, we analyzed the involvement of heparanase, a heparan sulfate glycosidase, in the homeostasis of proximal tubular epithelial cells in the diabetic milieu. In vitro studies were performed on a wild-type and stably heparanase-silenced adult tubular line (HK2) and HEK293. Gene and protein expression analyses were performed in the presence and absence of diabetic mediators. Albumin and advanced glycation end-product, but not high glucose levels, increased heparanase expression in adult tubular cells via the AKT/PI3K signaling pathway. This over-expression of heparanase is then responsible for heparan sulfate reduction via its endoglycosidase activity and its capacity to regulate the heparan sulfate-proteoglycans core protein. In fact, heparanase regulates the gene expression of syndecan-1, the most abundant heparan sulfate-proteoglycans in tubular cells. We showed that heparanase is a target gene of the diabetic nephropathy mediators albumin and advanced glycation end-product, so it may be relevant to the progression of diabetic nephropathy. It could take part in several processes, e.g. extracellular-matrix remodeling and cell-cell crosstalk, via its heparan sulfate endoglycosidase activity and capacity to regulate the expression of the heparan sulfate-proteoglycan syndecan-1.


Assuntos
Albuminas/metabolismo , Glucuronidase/metabolismo , Produtos Finais de Glicação Avançada/metabolismo , Túbulos Renais Proximais/metabolismo , Sequência de Bases , Linhagem Celular , Nefropatias Diabéticas/complicações , Nefropatias Diabéticas/genética , Nefropatias Diabéticas/metabolismo , Glucose/metabolismo , Glucuronidase/antagonistas & inibidores , Glucuronidase/genética , Células HEK293 , Heparitina Sulfato/metabolismo , Homeostase , Humanos , Falência Renal Crônica/etiologia , Falência Renal Crônica/genética , Falência Renal Crônica/metabolismo , Túbulos Renais Proximais/citologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Albumina Sérica/metabolismo , Soroalbumina Bovina/metabolismo , Sindecana-1/genética , Sindecana-1/metabolismo , Ativação Transcricional , Albumina Sérica Glicada
16.
J Transl Med ; 10: 213, 2012 Oct 24.
Artigo em Inglês | MEDLINE | ID: mdl-23095131

RESUMO

BACKGROUND: Epithelial-mesenchymal transition of tubular cells is a widely recognized mechanism that sustains interstitial fibrosis in diabetic nephropathy (DN). The signaling of FGF-2, a growth factor involved in this mechanism, is regulated by glycosaminoglycans. Heparanase-1, an endoglycosidase that cleaves heparan sulfate, is implicated in the pathogenesis of diabetic nephropathy and is necessary to FGF-2 for the induction of tubular cells transition. Well known Heparanase-1 inhibitors are heparin(s) and sulodexide, a low-molecular weight heparin - dermatan sulphate blend, which is effective in the treatment of DN. METHODS: We have investigated the inhibition by sulodexide and its components of Heparanase-1 by an ELISA assay. We have analyzed its effect on the epithelial-mesenchymal transition of tubular cells by real time gene expression analysis, zymography and migration assay. RESULTS: Results show that sulodexide is an effective heparanase-1 inhibitor, exclusively in virtue to the heparin component, with an IC50 of 5 µg/ml. In FGF-2 treated tubular cells, sulodexide also prevents the over-expression of the mesenchymal markers αSMA, vimentin and fibronectin and the motility increase, i.e. the epithelial-mesenchymal transition of tubular cells. Moreover, sulodexide prevents FGF-2 induced heparanase-1 and MMP9 increase switching off the autocrine loop that FGF-2 activates to support its signal. CONCLUSIONS: The findings highlight the capacity of sulodexide to inhibit heparanase-1 and to control tubular fibrosis triggered by epithelial-mesenchymal transition. In conclusion, these sulodexide activities support the value of this agent in controlling the progression of nephropathy to renal failure.


Assuntos
Nefropatias Diabéticas/tratamento farmacológico , Nefropatias Diabéticas/enzimologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Fator 2 de Crescimento de Fibroblastos/farmacologia , Glucuronidase/antagonistas & inibidores , Glicosaminoglicanos/uso terapêutico , Rim/patologia , Biomarcadores/metabolismo , Linhagem Celular , Movimento Celular/efeitos dos fármacos , Nefropatias Diabéticas/patologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucuronidase/metabolismo , Glicosaminoglicanos/farmacologia , Humanos , Rim/efeitos dos fármacos , Rim/enzimologia , Metaloproteinase 9 da Matriz/genética , Metaloproteinase 9 da Matriz/metabolismo , Mesoderma/efeitos dos fármacos , Mesoderma/metabolismo , Sindecana-1/genética , Sindecana-1/metabolismo
17.
Lung ; 190(4): 419-30, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22430123

RESUMO

BACKGROUND: Moderate normobaric hyperoxia causes alveolar and vascular lung derangement in the newborn rat. Endogenous nitric oxide (NO), which promotes lung growth, is produced from the metabolism of L-arginine to L-citrulline in endothelial cells. We investigated whether administering L-citrulline by raising the serum levels of L-arginine and enhancing NO endogenous synthesis attenuates moderate hyperoxia-induced lung injury. METHODS: Newborn rats were exposed to FiO(2) = 0.6 or room air for 14 days to induce lung derangement and then were administered L-citrulline or a vehicle (sham). Lung histopathology was studied with morphometric features. Lung tissues and bronchoalveolar lavage fluid (BALF) were collected for analysis. Lung vascular endothelial growth factor (VEGF), nitric oxide synthase (eNOS), and matrix metalloproteinase 2 (MMP2) gene and protein expressions were assessed. RESULTS: Serum L-arginine rose in the L-citr + hyperoxia group (p = 0.05), as well as the Von Willebrand factor stained vessels count (p = 0.0008). Lung VEGF immune staining, localized on endothelial cells, was weaker in the sections under hyperoxia than the L-citr + hyperoxia and room air groups. This pattern was comparable with the VEGF gene and protein expression profiles. Mean alveolar size increased in the untreated hyperoxia and sham-treated groups compared with the groups reared in room air or treated with L-citrulline under exposure to hyperoxia (p = 0.0001). Lung VEGF and eNOS increased in the L-citrulline-treated rats, though this treatment did not change MMP2 gene expression but regulated the MMP2 active protein, which rose in BALF (p = 0.003). CONCLUSIONS: We conclude that administering L: -citrulline proved effective in improving alveolar and vascular growth in a model of oxygen-induced pulmonary damage, suggesting better lung growth and matrix regulation than in untreated groups.


Assuntos
Citrulina/uso terapêutico , Endotélio Vascular/patologia , Hiperóxia/complicações , Lesão Pulmonar/etiologia , Lesão Pulmonar/prevenção & controle , Pulmão/irrigação sanguínea , Alvéolos Pulmonares/patologia , Animais , Animais Recém-Nascidos , Arginina/metabolismo , Citrulina/farmacologia , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Pulmão/metabolismo , Pulmão/patologia , Lesão Pulmonar/patologia , Metaloproteinase 2 da Matriz/metabolismo , Óxido Nítrico/metabolismo , Alvéolos Pulmonares/efeitos dos fármacos , Alvéolos Pulmonares/metabolismo , Ratos , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Fator A de Crescimento do Endotélio Vascular/metabolismo
18.
Cell Death Dis ; 13(11): 977, 2022 11 19.
Artigo em Inglês | MEDLINE | ID: mdl-36402749

RESUMO

The activation of TNF receptors can lead to cell death with a mechanism of cell necrosis regulated genetically and distinct from apoptosis which is defined as necroptosis. Necroptosis has been one of the most studied emerging cell death/signaling pathways in recent years, especially in light of the role of this process in human disease. However, not all regulatory components of TNF signaling have been identified in relation to both physiological and pathological conditions. In 2008, Spata2 (Spermatogenesis-associated protein 2) was identified as one of the seven fundamental genes for the cellular signaling network that regulates necroptosis and apoptosis. This gene had been cloned by our group and named Spata2 as its expression was found to be elevated in the testis compared to other tissues, localized at the Sertoli cell level and FSH-dependent. More recently, it has been demonstrated that deletion of Spata2 gene causes increased inhibin α expression and attenuated fertility in male mice. However, more importantly, five recently published reports have highlighted that SPATA2 is crucial for recruiting CYLD to the TNFR1 signaling complex thus promoting its activation leading to TNF-induced cell death. Loss of SPATA2 increases transcriptional activation of NF-kB and limits TNF-induced necroptosis. Here we will discuss these important findings regarding SPATA2 and, in particular, focus attention on the evidence that suggests a role for this protein in the TNF signaling pathway.


Assuntos
Neoplasias , Espermatogênese , Humanos , Masculino , Camundongos , Animais , Espermatogênese/genética , Transdução de Sinais , NF-kappa B/genética , NF-kappa B/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Proteínas
19.
Matrix Biol Plus ; 13: 100097, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35036899

RESUMO

The surface of all animal cells is coated with a layer of carbohydrates linked in various ways to the outer side of the plasma membrane. These carbohydrates are mainly bound to proteins in the form of glycoproteins and proteoglycans and together with the glycolipids constitute the so-called glycocalyx. In particular, the endothelial glycocalyx that covers the luminal layer of the endothelium is composed of glycosaminoglycans (heparan sulphate -HS and hyaluronic acid -HA), proteoglycans (syndecans and glypicans) and adsorbed plasma proteins. Thanks to its ability to absorb water, this structure contributes to making the surface of the vessels slippery but at the same time acts by modulating the mechano-transduction of the vessels, the vascular permeability and the adhesion of leukocytes in thus regulating several physiological and pathological events. Among the various enzymes involved in the degradation of the glycocalyx, heparanase (HPSE) has been shown to be particularly involved. This enzyme is responsible for the cutting of heparan sulfate (HS) chains at the level of the proteoglycans of the endothelial glycocalyx whose dysfunction appears to have a role in organ fibrosis, sepsis and viral infection. In this mini-review, we describe the mechanisms by which HPSE contributes to glycocalyx remodeling and then examine the role of glycocalyx degradation in the development of pathological conditions and pharmacological strategies to preserve glycocalyx during disease pathogenesis.

20.
Front Oncol ; 12: 918419, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35965510

RESUMO

Prostate cancer displays a certain phenotypic plasticity that allows for the transition of cells from the epithelial to the mesenchymal state. This process, known as epithelial-mesenchymal transition (EMT), is one of the factors that give the tumor cells greater invasive and migratory capacity with subsequent formation of metastases. In addition, many cancers, including prostate cancer, are derived from a cell population that shows the properties of stem cells. These cells, called cancer stem cells (CSCs) or tumor-initiating cells, not only initiate the tumor process and growth but are also able to mediate metastasis and drug resistance. However, the impact of EMT and CSCs in prostate cancer progression and patient survival is still far from fully understood. Heparanase (HPSE), the sole mammalian endoglycosidase capable of degrading heparan sulfate (HS), is also involved in prostate cancer progression. We had previously proved that HPSE regulates EMT in non-cancerous pathologies. Two prostate cancer cell lines (DU145 and PC3) were silenced and overexpressed for HPSE. Expression of EMT and stemness markers was evaluated. Results showed that the expression of several EMT markers are modified by HPSE expression in both the prostate cancer cell lines analyzed. In the same way, the stemness markers and features are also modulated by HPSE expression. Taken together, the present findings seem to prove a new mechanism of action of HPSE in sustaining prostate cancer growth and diffusion. As for other tumors, these results highlight the importance of HPSE as a potential pharmacological target in prostate cancer treatment.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA