Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Int J Mol Sci ; 25(2)2024 Jan 11.
Article in English | MEDLINE | ID: mdl-38255995

ABSTRACT

Vessel co-option (VCO) is a non-angiogenic mechanism of vascularization that has been associated to anti-angiogenic therapy. In VCO, cancer cells hijack the pre-existing blood vessels and use them to obtain oxygen and nutrients and invade adjacent tissue. Multiple primary tumors and metastases undergo VCO in highly vascularized tissues such as the lungs, liver or brain. VCO has been associated with a worse prognosis. The cellular and molecular mechanisms that undergo VCO are poorly understood. Recent studies have demonstrated that co-opted vessels show a quiescent phenotype in contrast to angiogenic tumor blood vessels. On the other hand, it is believed that during VCO, cancer cells are adhered to basement membrane from pre-existing blood vessels by using integrins, show enhanced motility and a mesenchymal phenotype. Other components of the tumor microenvironment (TME) such as extracellular matrix, immune cells or extracellular vesicles play important roles in vessel co-option maintenance. There are no strategies to inhibit VCO, and thus, to eliminate resistance to anti-angiogenic therapy. This review summarizes all the molecular mechanisms involved in vessel co-option analyzing the possible therapeutic strategies to inhibit this process.


Subject(s)
Immunotherapy , Neoplasms , Humans , Neovascularization, Pathologic/drug therapy , Basement Membrane , Brain , Cell Division , Neoplasms/drug therapy
3.
MAGMA ; 36(3): 419-426, 2023 Jul.
Article in English | MEDLINE | ID: mdl-35986790

ABSTRACT

OBJECTIVE: Speech production MRI benefits from lower magnetic fields due to reduced off-resonance effects at air-tissue interfaces and from the use of dedicated receiver coils due to higher SNR and parallel imaging capability. Here we present a custom designed upper airway coil for 1H imaging at 0.55 Tesla and evaluate its performance in comparison with a vendor-provided prototype 16-channel head/neck coil. MATERIALS AND METHODS: Four adult volunteers were scanned with both custom speech and prototype head-neck coils. We evaluated SNR gains of each of the coils over eleven upper airway volumes-of-interest measured relative to the integrated body coil. We evaluated parallel imaging performance of both coils by computing g-factors for SENSE reconstruction of uniform and variable density Cartesian sampling schemes with R = 2, 3, and 4. RESULTS: The dedicated coil shows approximately 3.5-fold SNR efficiency compared to the head-neck coil. For R = 2 and 3, both uniform and variable density samplings have g-factor values below 1.1 in the upper airway region. For R = 4, g-factor values are higher for both trajectories. DISCUSSION: The dedicated coil configuration allows for a significant SNR gain over the head-neck coil in the articulators. This, along with favorable g values, makes the coil useful in speech production MRI.


Subject(s)
Magnetic Resonance Imaging , Speech , Adult , Humans , Signal-To-Noise Ratio , Magnetic Resonance Imaging/methods , Head , Volunteers , Phantoms, Imaging
4.
Cancers (Basel) ; 14(23)2022 Nov 28.
Article in English | MEDLINE | ID: mdl-36497340

ABSTRACT

Liquid biopsy has improved significantly over the last decade and is attracting attention as a tool that can complement tissue biopsy to evaluate the genetic landscape of solid tumors. In the present study, we evaluated the usefulness of liquid biopsy in daily oncology practice in different clinical contexts. We studied ctDNA and tissue biopsy to investigate EGFR, KRAS, NRAS, and BRAF mutations from 199 cancer patients between January 2016 and March 2021. The study included 114 male and 85 female patients with a median age of 68 years. A total of 122 cases were lung carcinoma, 53 were colorectal carcinoma, and 24 were melanoma. Liquid biopsy was positive for a potentially druggable driver mutation in 14 lung and colorectal carcinoma where tissue biopsy was not performed, and in two (3%) lung carcinoma patients whose tissue biopsy was negative. Liquid biopsy identified nine (45%) de novo EGFR-T790M mutations during TKI-treatment follow-up in lung carcinoma. BRAF-V600 mutation resurgence was detected in three (12.5%) melanoma patients during follow-up. Our results confirm the value of liquid biopsy in routine clinical oncologic practice for targeted therapy, diagnosis of resistance to treatment, and cancer follow-up.

5.
Front Pharmacol ; 13: 843732, 2022.
Article in English | MEDLINE | ID: mdl-35770075

ABSTRACT

Renal tubulo-interstitial fibrosis is characterized by the excessive accumulation of extracellular matrix (ECM) in the tubular interstitium during chronic kidney disease. The main source of ECM proteins are emerging and proliferating myofibroblasts. The sources of myofibroblasts in the renal tubular interstitium have been studied during decades, in which the epithelial contribution of the myofibroblast population through the epithelial-to-mesenchymal (EMT) process was assumed to be the major mechanism. However, it is now accepted that the EMT contribution is very limited and other mechanisms such as the proliferation of local resident fibroblasts or the transdifferentiation of endothelial cells seem to be more relevant. Activin receptor-like kinase 1 (ALK1) is a type I receptor which belongs to the transforming growth factor beta (TGF-ß) superfamily, with a key role in tissue fibrosis and production of ECM by myofibroblast. Predominantly expressed in endothelial cells, ALK1 also plays an important role in angiogenesis and vessel maturation, but the relation of these processes with kidney fibrosis is not fully understood. We show that after 3 days of unilateral ureteral obstruction (UUO), ALK1 heterozygous mice (Alk1 +/- ) display lower levels of kidney fibrosis associated to a lower number of myofibroblasts. Moreover, Alk1 +/- mice have a lower degree of vascular rarefaction, showing improved peritubular microvasculature after UUO. All these data suggest an important role of ALK1 in regulating vascular rarefaction and emergence of myofibroblasts.

6.
Cancers (Basel) ; 13(21)2021 Oct 28.
Article in English | MEDLINE | ID: mdl-34771575

ABSTRACT

The improvement of cancer therapy efficacy, the extension of patient survival and the reduction of adverse side effects are major challenges in cancer research. Targeting blood vessels has been considered a promising strategy in cancer therapy. Since the tumor vasculature is disorganized, leaky and triggers immunosuppression and tumor hypoxia, several strategies have been studied to modify tumor vasculature for cancer therapy improvement. Anti-angiogenesis was first described as a mechanism to prevent the formation of new blood vessels and prevent the oxygen supply to tumor cells, showing numerous limitations. Vascular normalization using low doses of anti-angiogenic drugs was purposed to overcome the limitations of anti-angiogenic therapies. Other strategies such as vascular promotion or the induction of high endothelial venules are being studied now to improve cancer therapy. Bone morphogenetic protein 9 (BMP9) exerts a dual effect through the activin receptor-like kinase 1 (ALK1) receptor in blood vessel maturation or activation phase of angiogenesis. Thus, it is an interesting pathway to target in combination with chemotherapies or immunotherapies. This review manuscript explores the effect of the BMP9-ALK1 pathway in tumor angiogenesis and the possible usefulness of targeting this pathway in anti-angiogenesis, vascular normalization or vascular promotion therapies.

7.
Cancers (Basel) ; 13(20)2021 Oct 17.
Article in English | MEDLINE | ID: mdl-34680355

ABSTRACT

Several strategies have been developed to modulate the tumour vasculature for cancer therapy including anti-angiogenesis and vascular normalisation. Vasculature modulation results in changes to the tumour microenvironment including oxygenation and immune cell infiltration, therefore lending itself to combination with cancer therapy. The development of immunotherapies has led to significant improvements in cancer treatment. Particularly promising are immune checkpoint blockade and CAR T cell therapies, which use antibodies against negative regulators of T cell activation and T cells reprogrammed to better target tumour antigens, respectively. However, while immunotherapy is successful in some patients, including those with advanced or metastatic cancers, only a subset of patients respond. Therefore, better predictors of patient response and methods to overcome resistance warrant investigation. Poor, or periphery-limited, T cell infiltration in the tumour is associated with poor responses to immunotherapy. Given that (1) lymphocyte recruitment requires leucocyte-endothelial cell adhesion and (2) the vasculature controls tumour oxygenation and plays a pivotal role in T cell infiltration and activation, vessel targeting strategies including anti-angiogenesis and vascular normalisation in combination with immunotherapy are providing possible new strategies to enhance therapy. Here, we review the progress of vessel modulation in enhancing immunotherapy efficacy.

8.
Genes (Basel) ; 12(6)2021 05 24.
Article in English | MEDLINE | ID: mdl-34073961

ABSTRACT

Many different regulatory mechanisms of renal fibrosis are known to date, and those related to transforming growth factor-ß1 (TGF-ß1)-induced signaling have been studied in greater depth. However, in recent years, other signaling pathways have been identified, which contribute to the regulation of these pathological processes. Several studies by our team and others have revealed the involvement of small Ras GTPases in the regulation of the cellular processes that occur in renal fibrosis, such as the activation and proliferation of myofibroblasts or the accumulation of extracellular matrix (ECM) proteins. Intracellular signaling mediated by TGF-ß1 and Ras GTPases are closely related, and this interaction also occurs during the development of renal fibrosis. In this review, we update the available in vitro and in vivo knowledge on the role of Ras and its main effectors, such as Erk and Akt, in the cellular mechanisms that occur during the regulation of kidney fibrosis (ECM synthesis, accumulation and activation of myofibroblasts, apoptosis and survival of tubular epithelial cells), as well as the therapeutic strategies for targeting the Ras pathway to intervene on the development of renal fibrosis.


Subject(s)
Renal Insufficiency, Chronic/metabolism , ras Proteins/metabolism , Animals , Extracellular Matrix Proteins/metabolism , Fibrosis , Humans , Renal Insufficiency, Chronic/pathology , Signal Transduction , ras Proteins/genetics
9.
Nat Commun ; 11(1): 2810, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32499572

ABSTRACT

The overexpression of the protein tyrosine kinase, Focal adhesion kinase (FAK), in endothelial cells has implicated its requirement in angiogenesis and tumour growth, but how pericyte FAK regulates tumour angiogenesis is unknown. We show that pericyte FAK regulates tumour growth and angiogenesis in multiple mouse models of melanoma, lung carcinoma and pancreatic B-cell insulinoma and provide evidence that loss of pericyte FAK enhances Gas6-stimulated phosphorylation of the receptor tyrosine kinase, Axl with an upregulation of Cyr61, driving enhanced tumour growth. We further show that pericyte derived Cyr61 instructs tumour cells to elevate expression of the proangiogenic/protumourigenic transmembrane receptor Tissue Factor. Finally, in human melanoma we show that when 50% or more tumour blood vessels are pericyte-FAK negative, melanoma patients are stratified into those with increased tumour size, enhanced blood vessel density and metastasis. Overall our data uncover a previously unknown mechanism of tumour growth by pericytes that is controlled by pericyte FAK.


Subject(s)
Cysteine-Rich Protein 61/metabolism , Focal Adhesion Kinase 1/metabolism , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/metabolism , Neovascularization, Pathologic , Pericytes/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Aorta, Thoracic/pathology , Carcinoma, Lewis Lung/metabolism , Cell Adhesion , Cell Proliferation , Female , Focal Adhesion Kinase 1/genetics , Humans , Lymphokines/metabolism , Male , Melanoma/blood supply , Melanoma/metabolism , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/pathology , Placenta Growth Factor/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis/metabolism , Signal Transduction , Tumor Microenvironment , Vascular Endothelial Growth Factor A/metabolism , Axl Receptor Tyrosine Kinase
10.
Cell ; 181(6): 1346-1363.e21, 2020 06 11.
Article in English | MEDLINE | ID: mdl-32473126

ABSTRACT

Enhanced blood vessel (BV) formation is thought to drive tumor growth through elevated nutrient delivery. However, this observation has overlooked potential roles for mural cells in directly affecting tumor growth independent of BV function. Here we provide clinical data correlating high percentages of mural-ß3-integrin-negative tumor BVs with increased tumor sizes but no effect on BV numbers. Mural-ß3-integrin loss also enhances tumor growth in implanted and autochthonous mouse tumor models with no detectable effects on BV numbers or function. At a molecular level, mural-cell ß3-integrin loss enhances signaling via FAK-p-HGFR-p-Akt-p-p65, driving CXCL1, CCL2, and TIMP-1 production. In particular, mural-cell-derived CCL2 stimulates tumor cell MEK1-ERK1/2-ROCK2-dependent signaling and enhances tumor cell survival and tumor growth. Overall, our data indicate that mural cells can control tumor growth via paracrine signals regulated by ß3-integrin, providing a previously unrecognized mechanism of cancer growth control.


Subject(s)
Integrin beta3/metabolism , Neoplasms/metabolism , Tumor Burden/physiology , Animals , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Female , Humans , Male , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
11.
EMBO Mol Med ; 12(2): e11663, 2020 02 07.
Article in English | MEDLINE | ID: mdl-31858727

ABSTRACT

Pathological angiogenesis contributes to tumour progression as well as to chronic inflammatory diseases. In this issue of EMBO Molecular Medicine, Esteban and co-workers identify endothelial cell MT1-MMP as a key regulator of intussusceptive angiogenesis (IA) in inflammatory colitis. Thrombospondin 1 (TSP1) cleavage by MT1-MMP results in the binding of the c-terminal fragment of TSP1 to αvß3 integrin, which induces nitric oxide (NO) production, vasodilation and further initiation of IA. This novel control mechanism of inflammatory IA points towards promising new therapeutic targets for inflammatory bowel disease.


Subject(s)
Matrix Metalloproteinase 14 , Metalloendopeptidases , Endothelial Cells , Humans , Matrix Metalloproteinases, Membrane-Associated , Neovascularization, Pathologic
12.
ACS Pharmacol Transl Sci ; 2(6): 387-401, 2019 Dec 13.
Article in English | MEDLINE | ID: mdl-32259072

ABSTRACT

The integrin αVß3 receptor has been implicated in several important diseases, but no antagonists are approved for human therapy. One possible limitation of current small-molecule antagonists is their ability to induce a major conformational change in the receptor that induces it to adopt a high-affinity ligand-binding state. In response, we used structural inferences from a pure peptide antagonist to design the small-molecule pure antagonists TDI-4161 and TDI-3761. Both compounds inhibit αVß3-mediated cell adhesion to αVß3 ligands, but do not induce the conformational change as judged by antibody binding, electron microscopy, X-ray crystallography, and receptor priming studies. Both compounds demonstrated the favorable property of inhibiting bone resorption in vitro, supporting potential value in treating osteoporosis. Neither, however, had the unfavorable property of the αVß3 antagonist cilengitide of paradoxically enhancing aortic sprout angiogenesis at concentrations below its IC50, which correlates with cilengitide's enhancement of tumor growth in vivo.

13.
Acta Physiol (Oxf) ; 226(2): e13247, 2019 06.
Article in English | MEDLINE | ID: mdl-30589223

ABSTRACT

AIM: Chronic kidney disease is characterized by tubulointerstitial fibrosis involving inflammation, tubular apoptosis, fibroblast proliferation and extracellular matrix accumulation. Cardiotrophin-1, a member of the interleukin-6 family of cytokines, protects several organs from damage by promoting survival and anti-inflammatory effects. However, whether cardiotrophin-1 participates in the response to chronic kidney injury leading to renal fibrosis is unknown. METHODS: We hypothesized and assessed the potential role of cardiotrophin-1 in a mice model of tubulointerstitial fibrosis induced by unilateral ureteral obstruction (UUO). RESULTS: Three days after UUO, obstructed kidneys from cardiotrophin-1-/- mice show higher expression of inflammatory markers IL-1ß, Cd68, ICAM-1, COX-2 and iNOs, higher activation of NF-κB, higher amount of myofibroblasts and higher severity of tubular damage and apoptosis, compared with obstructed kidneys from wild-type littermates. In a later stage, obstructed kidneys from cardiotrophin-1-/- mice show higher fibrosis than obstructed kidneys from wild-type mice. Interestingly, administration of exogenous cardiotrophin-1 prevents the increased fibrosis resulting from the genetic knockout of cardiotrophin-1 upon UUO, and supplementation of wild-type mice with exogenous cardiotrophin-1 further reduces the renal fibrosis induced by UUO. In vitro, renal myofibroblasts from cardiotrophin-1-/- mice have higher collagen I and fibronectin expression and higher NF-κB activation than wild-type cells. CONCLUSIONS: Cardiotrophin-1 participates in the endogenous response that opposes renal damage by counteracting the inflammatory, apoptotic and fibrotic processes. And exogenous cardiotrophin-1 is proposed as a candidate for the treatment and prevention of chronic renal fibrosis.


Subject(s)
Cytokines/metabolism , Fibrosis/metabolism , Kidney/metabolism , Renal Insufficiency, Chronic/metabolism , Ureteral Obstruction/metabolism , Animals , Cytokines/genetics , Disease Models, Animal , Gene Expression Regulation/physiology , Mice, Knockout , Renal Insufficiency, Chronic/genetics , Renal Insufficiency, Chronic/pathology , Ureteral Obstruction/genetics
14.
J Cell Mol Med ; 22(1): 302-314, 2018 01.
Article in English | MEDLINE | ID: mdl-28857467

ABSTRACT

Renal fibrosis and anaemia are two of the most relevant events in chronic kidney disease. Fibrosis is characterized by the accumulation of extracellular matrix proteins in the glomeruli and tubular interstitium. Anaemia is the consequence of a decrease in erythropoietin production in fibrotic kidneys. This work analyses the possibility that the accumulation of abnormal collagens in kidney interstitium could be one of the mechanisms responsible for erythropoietin decreased synthesis. In renal interstitial fibroblast grown on collagen I, erythropoietin mRNA expression and HIF-2α protein decreased, whereas focal adhesion kinase protein (FAK) phosphorylation and proteasome activity increased, compared to cells grown on collagen IV. Proteasome inhibition or FAK inactivation in cells plated on collagen I restored erythropoietin and HIF-2α expression. FAK inhibition also decreased the collagen I-dependent proteasome activation. In a model of tubulointerstitial fibrosis induced by unilateral ureteral obstruction in mice, increased collagen I protein content and an almost complete disappearance of erythropoietin mRNA expression were observed in the ureteral ligated kidney with respect to the contralateral control. Interestingly, erythropoietin synthesis was recovered in obstructed mice treated with proteasome inhibitor. These data suggest that reduced kidney erythropoietin synthesis could be caused by the accumulation of abnormal extracellular matrix proteins.


Subject(s)
Erythropoietin/biosynthesis , Extracellular Matrix/metabolism , Renal Insufficiency, Chronic/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line , Collagen Type I/pharmacology , Down-Regulation/drug effects , Erythropoietin/genetics , Erythropoietin/metabolism , Extracellular Matrix/drug effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibrosis , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Humans , Kidney/pathology , Mice, Inbred C57BL , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Proteolysis/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Renal Insufficiency, Chronic/pathology , Ureteral Obstruction/pathology
15.
Angew Chem Int Ed Engl ; 56(51): 16405-16409, 2017 12 18.
Article in English | MEDLINE | ID: mdl-29072809

ABSTRACT

A highly systematic approach for the development of both orally bioavailable and bioactive cyclic N-methylated hexapeptides as high affinity ligands for the integrin αvß3 is based on two concepts: a) screening of systematically designed libraries with spatial diversity and b) masking of the peptide charge with a lipophilic protecting group. The key steps of the method are 1) initial design of a combinatorial library of N-methylated analogues of the stem peptide cyclo(d-Ala-Ala5 ); 2) selection of cyclic peptides with the highest intestinal permeability; 3) design of sublibraries with the bioactive RGD sequence in all possible positions; 4) selection of the best ligands for RGD-recognizing integrin subtypes; 5) fine-tuning of the affinity and selectivity by additional Ala to Xaa substitutions; 6) protection of the charged functional groups according to the prodrug concept to regain intestinal and oral permeability; 7) proof of biological effects in mice after oral administration.


Subject(s)
Drug Design , Integrin alphaVbeta3/metabolism , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/pharmacology , Administration, Oral , Animals , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Injections, Intraperitoneal , Ligands , Mice , Peptides, Cyclic/chemical synthesis , Protein Conformation , Xenograft Model Antitumor Assays
16.
J Cell Sci ; 130(9): 1583-1595, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28289267

ABSTRACT

The α6ß1-integrin is a major laminin receptor, and formation of a laminin-rich basement membrane is a key feature in tumour blood vessel stabilisation and pericyte recruitment, processes that are important in the growth and maturation of tumour blood vessels. However, the role of pericyte α6ß1-integrin in angiogenesis is largely unknown. We developed mice where the α6-integrin subunit is deleted in pericytes and examined tumour angiogenesis and growth. These mice had: (1) reduced pericyte coverage of tumour blood vessels; (2) reduced tumour blood vessel stability; (3) increased blood vessel diameter; (4) enhanced blood vessel leakiness, and (5) abnormal blood vessel basement membrane architecture. Surprisingly, tumour growth, blood vessel density and metastasis were not altered. Analysis of retinas revealed that deletion of pericyte α6-integrin did not affect physiological angiogenesis. At the molecular level, we provide evidence that pericyte α6-integrin controls PDGFRß expression and AKT-mTOR signalling. Taken together, we show that pericyte α6ß1-integrin regulates tumour blood vessels by both controlling PDGFRß and basement membrane architecture. These data establish a novel dual role for pericyte α6-integrin as modulating the blood vessel phenotype during pathological angiogenesis.


Subject(s)
Blood Vessels/metabolism , Integrin alpha6beta1/metabolism , Neoplasms/blood supply , Pericytes/metabolism , Animals , Basement Membrane/drug effects , Basement Membrane/metabolism , Becaplermin , Blood Vessels/drug effects , Blood Vessels/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Integrases/metabolism , Mice , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology , Pericytes/drug effects , Proto-Oncogene Proteins c-sis/pharmacology , Receptor, Platelet-Derived Growth Factor beta/metabolism
17.
Biochim Biophys Acta ; 1862(9): 1801-14, 2016 09.
Article in English | MEDLINE | ID: mdl-27321931

ABSTRACT

Transforming growth factor beta 1 (TGF-ß1) is one of the most studied cytokines involved in renal tubulo-interstitial fibrosis, which is characterized by myofibroblast abundance and proliferation, and high buildup of extracellular matrix in the tubular interstitium leading to organ failure. Endoglin (Eng) is a 180-kDa homodimeric transmembrane protein that regulates a great number of TGF-ß1 actions in different biological processes, including ECM synthesis. High levels of Eng have been observed in experimental models of renal fibrosis or in biopsies from patients with chronic kidney disease. In humans and mice, two Eng isoforms are generated by alternative splicing, L-Eng and S-Eng that differ in the length and composition of their cytoplasmic domains. We have previously described that L-Eng overexpression promotes renal fibrosis after unilateral ureteral obstruction (UUO). However, the role of S-Eng in renal fibrosis is unknown and its study would let us analyze the possible function of the cytoplasmic domain of Eng in this process. For this purpose, we have generated a mice strain that overexpresses S-Eng (S-ENG(+)) and we have performed an UUO in S-ENG(+) and their wild type (WT) control mice. Our results indicate that obstructed kidney of S-ENG(+) mice shows lower levels of tubulo-interstitial fibrosis, less inflammation and less interstitial cell proliferation than WT littermates. Moreover, S-ENG(+) mice show less activation of Smad1 and Smad2/3 pathways. Thus, S-Eng overexpression reduces UUO-induced renal fibrosis and some associated mechanisms. As L-Eng overexpression provokes renal fibrosis we conclude that Eng-mediated induction of renal fibrosis in this model is dependent on its cytoplasmic domain.


Subject(s)
Endoglin/genetics , Endoglin/metabolism , Kidney/metabolism , Kidney/pathology , Nephritis/prevention & control , Ureteral Obstruction/metabolism , Animals , Cell Proliferation , Collagen Type I/metabolism , Disease Models, Animal , Fibronectins/metabolism , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Mice, Transgenic , Models, Biological , Myofibroblasts/metabolism , Myofibroblasts/pathology , Nephritis/metabolism , Nephritis/pathology , Protein Isoforms/genetics , Protein Isoforms/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation , Ureteral Obstruction/complications , Ureteral Obstruction/pathology
18.
Cell Signal ; 28(9): 1252-1261, 2016 09.
Article in English | MEDLINE | ID: mdl-27208502

ABSTRACT

Upregulated synthesis of extracellular matrix (ECM) proteins by myofibroblasts is a common phenomenon in the development of fibrosis. Although the role of TGF-ß in fibrosis development has been extensively studied, the involvement of other members of this superfamily of cytokines, the bone morphogenetic proteins (BMPs) in organ fibrosis has given contradictory results. BMP9 is the main ligand for activin receptor-like kinase-1 (ALK1) TGF-ß1 type I receptor and its effect on fibrosis development is unknown. Our purpose was to study the effect of BMP9 in ECM protein synthesis in fibroblasts, as well as the involved receptors and signaling pathways. In cultured mice fibroblasts, BMP9 induces an increase in collagen, fibronectin and connective tissue growth factor expression, associated with Smad1/5/8, Smad2/3 and Erk1/2 activation. ALK5 inhibition with SB431542 or ALK1/2/3/6 with dorsomorphin-1, inhibition of Smad3 activation with SIS3, and inhibition of the MAPK/Erk1/2 with U0126, demonstrates the involvement of these pathways in BMP9-induced ECM synthesis in MEFs. Whereas BMP9 induced Smad1/5/8 phosphorylation through ALK1, it also induces Smad2/3 phosphorylation through ALK5 but only in the presence of ALK1. Summarizing, this is the first study that accurately identifies BMP9 as a profibrotic factor in fibroblasts that promotes ECM protein expression through ALK1 and ALK5 receptors.


Subject(s)
Growth Differentiation Factor 2/metabolism , Activin Receptors, Type I/metabolism , Animals , Embryo, Mammalian/cytology , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibrosis , Heterozygote , Humans , Mice , Models, Biological , NIH 3T3 Cells , Phosphorylation , Protein Biosynthesis , Signal Transduction , Smad Proteins/metabolism
19.
J Cell Physiol ; 231(10): 2224-35, 2016 10.
Article in English | MEDLINE | ID: mdl-26873620

ABSTRACT

The involvement of Ras-GTPases in the development of renal fibrosis has been addressed in the last decade. We have previously shown that H- and N-Ras isoforms participate in the regulation of fibrosis. Herein, we assessed the role of K-Ras in cellular processes involved in the development of fibrosis: proliferation, migration, and extracellular matrix (ECM) proteins synthesis. K-Ras knockout (KO) mouse embryonic fibroblasts (K-ras(-/-) ) stimulated with transforming growth factor-ß1 (TGF-ß1) exhibited reduced proliferation and impaired mobility than wild-type fibroblasts. Moreover, an increase on ECM production was observed in K-Ras KO fibroblasts in basal conditions. The absence of K-Ras was accompanied by reduced Ras activation and ERK phosphorylation, and increased AKT phosphorylation, but no differences were observed in TGF-ß1-induced Smad signaling. The MEK inhibitor U0126 decreased cell proliferation independently of the presence of K-ras but reduced migration and ECM proteins expression only in wild-type fibroblasts, while the PI3K-AKT inhibitor LY294002 decreased cell proliferation, migration, and ECM synthesis in both types of fibroblasts. Thus, our data unveil that K-Ras and its downstream effector pathways distinctively regulate key biological processes in the development of fibrosis. Moreover, we show that K-Ras may be a crucial mediator in TGF-ß1-mediated effects in this cell type. J. Cell. Physiol. 231: 2224-2235, 2016. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Movement , Cell Proliferation , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Animals , Butadienes/pharmacology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Extracellular Matrix Proteins/metabolism , Fibronectins/metabolism , Mice , Mice, Knockout , Nitriles/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins p21(ras)/deficiency , Transforming Growth Factor beta1/metabolism
20.
Pharmacol Ther ; 156: 44-58, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26493350

ABSTRACT

The understanding of renal fibrosis in chronic kidney disease (CKD) remains as a challenge. More than 10% of the population of developed countries suffer from CKD. Proliferation and activation of myofibroblasts and accumulation of extracellular matrix proteins are the main features of kidney fibrosis, a process in which a large number of cytokines are involved. Targeting cytokines responsible for kidney fibrosis development might be an important strategy to face the problem of CKD. The increasing knowledge of the signaling pathway network of the transforming growth factor beta (TGF-ß) superfamily members, such as the profibrotic cytokine TGF-ß1 or the bone morphogenetic proteins (BMPs), and their involvement in the regulation of kidney fibrosis, has stimulated numerous research teams to look for potential strategies to inhibit profibrotic cytokines or to enhance the anti-fibrotic actions of other cytokines. The consequence of all these studies is a better understanding of all these canonical (Smad-mediated) and non-canonical signaling pathways. In addition, the different receptors involved for signaling of each cytokine, the different combinations of type I-type II receptors, and the presence and function of co-receptors that can influence the biological response have been also described. However, are these studies leading to suitable strategies to block the appearance and progression of kidney fibrosis? In this review, we offer a critical perspective analyzing the achievements using the most important strategies developed up till now: TGF-ß antibodies, chemical inhibitors of TGF-ß receptors, miRNAs and signaling pathways and BMP agonists with a potential role as therapeutic molecules against kidney fibrosis.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Kidney/pathology , Receptors, Transforming Growth Factor beta/metabolism , Renal Insufficiency, Chronic/pathology , Transforming Growth Factor beta1/metabolism , Bone Morphogenetic Proteins/agonists , Bone Morphogenetic Proteins/antagonists & inhibitors , Cytokines/metabolism , Fibrosis , Humans , MicroRNAs , Receptors, Transforming Growth Factor beta/antagonists & inhibitors , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...