Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38630755

ABSTRACT

BACKGROUND: Transcriptomic subtyping holds promise for personalized therapy in extensive-stage small-cell lung cancer (ES-SCLC). In this study, we aimed to assess intratumoral transcriptomic subtype diversity and to identify biomarkers of long-term chemoimmunotherapy benefit in human ES-SCLC. PATIENTS AND METHODS: We analyzed tumor samples from 58 ES-SCLC patients enrolled in two multicenter single-arm phase IIIb studies evaluating front-line chemoimmunotherapy in Spain: n=32 from the IMfirst trial, and n=26 from the CANTABRICO trial. We utilized the GeoMxTM DSP system to perform multi-region transcriptomic analysis. For subtype classification, we performed hierarchical clustering using the relative expression of ASCL1 (SCLC-A), NEUROD1 (SCLC-N), POU2F3 (SCLC-P), and YAP1 (SCLC-Y). RESULTS: Subtype distribution was similar between both cohorts, except for SCLC-P, not identified in the CANTABRICO_DSP cohort. A total of 44% of the patients in both cohorts had tumors with multiple co-existing transcriptional subtypes. Transcriptional subtypes or subtype heterogeneity were not associated with outcomes. Most potential targets did not show subtype-specific expression. Consistently in both cohorts, tumors from patients with long-term benefit (time to progression ³12 months) contained an IFNg-dominated mRNA profile, including enhanced capacity for antigen presentation. Hypoxia and glycolytic pathways were associated with resistance to chemoimmunotherapy. CONCLUSIONS: This work suggests that intratumoral heterogeneity, inconsistent association with outcome, and unclear subtype-specific target expression might be significant challenges for subtype-based precision oncology in SCLC. Pre-existing IFNg-driven immunity and mitochondrial metabolism seem correlates of long-term efficacy in this study, although the absence of a chemotherapy control arm precludes concluding that these are predictive features specific for immunotherapy.

2.
Clin Cancer Res ; 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38630789

ABSTRACT

Platinum-based chemotherapy plus PD-1 axis blockade is the standard of care in the front-line treatment of extensive-stage small cell lung cancer (ES-SCLC). Despite the robust and consistent increase of long-term survival with PD-1 axis inhibition, the magnitude of the benefit from immunotherapy appears lower as compared to other solid tumors. Several immune evasive mechanisms have been shown to be prominently altered in human SCLC, including, among others, T cell exclusion, downregulation of components of the MHC-class I antigen processing and presentation machinery, or upregulation of macrophage inhibitory checkpoints. New immunotherapies aiming to target some of these dominant immune suppressive features are being intensively evaluated preclinically and clinically in SCLC. They include strategies to enhance the efficacy and/or reverse features that promote intrinsic resistance to PD-1 axis inhibition (e.g., restoring MHC-class I deficiency, targeting DNA damage response [DDR]), and novel immunomodulatory agents beyond T cell checkpoint blockers (e.g., T cell redirecting strategies, antibody drug conjugates [ADCs], or macrophage checkpoint blockers). Among them, DLL3-targeted bi-specific T-cell engagers (BiTEs) are the ones that have shown the most compelling preliminary evidence of clinical efficacy, and hold promise as therapies that might contribute to further improve patient outcomes in this disease. Here, we first provide a brief overview of key tumor microenvironment features of human SCLC. Then, we update the current clinical evidence with immune checkpoint blockade and review other emerging immunotherapy strategies that are gaining increasing attention in SCLC. We finally summarize our future perspective on immunotherapy and precision oncology for this disease.

3.
Nat Rev Clin Oncol ; 20(3): 143-159, 2023 03.
Article in English | MEDLINE | ID: mdl-36639452

ABSTRACT

Non-small-cell lung cancer (NSCLC) has become a paradigm of precision medicine, with the discovery of numerous disease subtypes defined by specific oncogenic driver mutations leading to the development of a range of molecularly targeted therapies. Over the past decade, rapid progress has also been made in the development of immune-checkpoint inhibitors (ICIs), especially antagonistic antibodies targeting the PD-L1-PD-1 axis, for the treatment of NSCLC. Although many of the major oncogenic drivers of NSCLC are associated with intrinsic resistance to ICIs, patients with certain oncogene-driven subtypes of the disease that are highly responsive to specific targeted therapies might also derive benefit from immunotherapy. However, the development of effective immunotherapy approaches for oncogene-addicted NSCLC has been challenged by a lack of predictive biomarkers for patient selection and limited knowledge of how ICIs and oncogene-directed targeted therapies should be combined. Therefore, whether ICIs alone or with chemotherapy or even in combination with molecularly targeted agents would offer comparable benefit in the context of selected oncogenic driver alterations to that observed in the general unselected NSCLC population remains an open question. In this Review, we discuss the effects of oncogenic driver mutations on the efficacy of ICIs and the immune tumour microenvironment as well as the potential vulnerabilities that could be exploited to overcome the challenges of immunotherapy for oncogene-addicted NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Immunotherapy , Lung Neoplasms , Oncogene Addiction , Humans , Antineoplastic Agents/therapeutic use , B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/drug therapy , Oncogenes
5.
J Clin Med ; 11(6)2022 Mar 09.
Article in English | MEDLINE | ID: mdl-35329826

ABSTRACT

Lung cancer is the leading cause of cancer mortality worldwide, with non-small cell lung cancer (NSCLC) being the most prevalent histology. While immunotherapy with checkpoint inhibitors has shown outstanding results in NSCLC, the precise identification of responders remains a major challenge. Most studies attempting to overcome this handicap have focused on adenocarcinomas or squamous cell carcinomas. Among NSCLC subtypes, the molecular and immune characteristics of lung large cell carcinoma (LCC), which represents 10% of NSCLC cases, are not well defined. We hypothesized that specific molecular aberrations may impact the immune microenvironment in LCC and, consequently, the response to immunotherapy. To that end, it is particularly relevant to thoroughly describe the molecular genotype-immunophenotype association in LCC-to identify robust predictive biomarkers and improve potential benefits from immunotherapy. We established a cohort of 18 early-stage, clinically annotated, LCC cases. Their molecular and immune features were comprehensively characterized by genomic and immune-targeted sequencing panels along with immunohistochemistry of immune cell populations. Unbiased clustering defined two novel subgroups of LCC. Pro-immunogenic tumors accumulated certain molecular alterations, showed higher immune infiltration and upregulated genes involved in potentiating immune responses when compared to pro-tumorigenic samples, which favored tumoral progression. This classification identified a set of biomarkers that could potentially predict response to immunotherapy. These results could improve patient selection and expand potential benefits from immunotherapy.

6.
Biomedicines ; 9(8)2021 Jul 29.
Article in English | MEDLINE | ID: mdl-34440118

ABSTRACT

BACKGROUND: Asthma is a chronic lung disease characterized by reversible airflow obstruction, airway hyperresponsiveness (AHR), mucus overproduction and inflammation. Although Insulin-like growth factor 1 receptor (IGF1R) was found to be involved in asthma, its pharmacological inhibition has not previously been investigated in this pathology. We aimed to determine if therapeutic targeting of IGF1R ameliorates allergic airway inflammation in a murine model of asthma. METHODS: C57BL/6J mice were challenged by house dust mite (HDM) extract or PBS for four weeks and therapeutically treated with the IGF1R tyrosine kinase inhibitor (TKI) NVP-ADW742 (NVP) once allergic phenotype was established. RESULTS: Lungs of HDM-challenged mice exhibited a significant increase in phospho-IGF1R levels, incremented AHR, airway remodeling, eosinophilia and allergic inflammation, as well as altered pulmonary surfactant expression, all of being these parameters counteracted by NVP treatment. HDM-challenged lungs also displayed augmented expression of the IGF1R signaling mediator p-ERK1/2, which was greatly reduced upon treatment with NVP. CONCLUSIONS: Our results demonstrate that IGF1R could be considered a potential pharmacological target in murine HDM-induced asthma and a candidate biomarker in allergic asthma.

7.
J Clin Invest ; 129(8): 3293-3309, 2019 05 28.
Article in English | MEDLINE | ID: mdl-31135379

ABSTRACT

Idiopathic Pulmonary Fibrosis (IPF) is a deadly disease with limited therapies. Tissue fibrosis is associated with Type 2 immune response, although the causal contribution of immune cells is not defined. The AP-1 transcription factor Fra-2 is upregulated in IPF lung sections and Fra-2 transgenic mice (Fra-2tg) exhibit spontaneous lung fibrosis. Here we show that Bleomycin-induced lung fibrosis is attenuated upon myeloid-inactivation of Fra-2 and aggravated in Fra-2tg bone marrow chimeras. Type VI collagen (ColVI), a Fra-2 transcriptional target, is up-regulated in three lung fibrosis models, and macrophages promote myofibroblast activation in vitro in a ColVI- and Fra-2-dependent manner. Fra-2 or ColVI inactivation does not affect macrophage recruitment and alternative activation, suggesting that Fra-2/ColVI specifically controls the paracrine pro-fibrotic activity of macrophages. Importantly, ColVI knock-out mice (KO) and ColVI-KO bone marrow chimeras are protected from Bleomycin-induced lung fibrosis. Therapeutic administration of a Fra-2/AP-1 inhibitor reduces ColVI expression and ameliorates fibrosis in Fra-2tg mice and in the Bleomycin model. Finally, Fra-2 and ColVI positively correlate in IPF patient samples and co-localize in lung macrophages. Therefore, the Fra-2/ColVI pro-fibrotic axis is a promising biomarker and therapeutic target for lung fibrosis, and possibly other fibrotic diseases.


Subject(s)
Fos-Related Antigen-2/biosynthesis , Idiopathic Pulmonary Fibrosis/metabolism , Macrophages/immunology , Myofibroblasts/metabolism , Allografts , Animals , Bleomycin/adverse effects , Bleomycin/pharmacology , Bone Marrow/metabolism , Bone Marrow/pathology , Bone Marrow Transplantation , Collagen Type VI/biosynthesis , Collagen Type VI/genetics , Fos-Related Antigen-2/genetics , Humans , Idiopathic Pulmonary Fibrosis/drug therapy , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/pathology , Macrophages/pathology , Mice , Mice, Knockout , Myofibroblasts/pathology , Transplantation Chimera/genetics , Transplantation Chimera/metabolism
8.
EMBO Mol Med ; 10(9)2018 09.
Article in English | MEDLINE | ID: mdl-30012580

ABSTRACT

Senescent cells accumulate in multiple aging-associated diseases, and eliminating these cells has recently emerged as a promising therapeutic approach. Here, we take advantage of the high lysosomal ß-galactosidase activity of senescent cells to design a drug delivery system based on the encapsulation of drugs with galacto-oligosaccharides. We show that gal-encapsulated fluorophores are preferentially released within senescent cells in mice. In a model of chemotherapy-induced senescence, gal-encapsulated cytotoxic drugs target senescent tumor cells and improve tumor xenograft regression in combination with palbociclib. Moreover, in a model of pulmonary fibrosis in mice, gal-encapsulated cytotoxics target senescent cells, reducing collagen deposition and restoring pulmonary function. Finally, gal-encapsulation reduces the toxic side effects of the cytotoxic drugs. Drug delivery into senescent cells opens new diagnostic and therapeutic applications for senescence-associated disorders.


Subject(s)
Cellular Senescence/drug effects , Drug Delivery Systems , Galactose/metabolism , Lysosomes/enzymology , Oligosaccharides/metabolism , beta-Galactosidase/metabolism , Animals , Cell Survival/drug effects , Cytotoxins/administration & dosage , Cytotoxins/pharmacology , Disease Models, Animal , Drug Compounding , Fluorescent Dyes/metabolism , Heterografts , Mice , Neoplasm Transplantation , Neoplasms/drug therapy , Piperazines/administration & dosage , Piperazines/pharmacology , Pyridines/administration & dosage , Pyridines/pharmacology , Staining and Labeling
9.
Blood Purif ; 39(1-3): 200-209, 2015.
Article in English | MEDLINE | ID: mdl-25765663

ABSTRACT

BACKGROUND/AIMS: Peritonitis is a major complication that arises out of peritoneal dialysis (PD), leading to death and loss of mesothelium and peritoneal injury, which may impede PD. We studied the combined impact of inflammatory mediators and PD fluids on mesothelial cell death. METHODS: Cultured human mesothelial cells. RESULTS: Inflammatory cytokines (TNF-α and interferon-γ) cooperate with bioincompatible PD fluids containing high glucose degradation product (GDP) concentrations to promote mesothelial cell death. Thus, the inflammatory cytokine cocktail induced a higher rate of death in cells cultured in high GDP PD fluid than in low GDP PD fluid or cell culture medium (cell death expressed as % hypodiploid cells: TNF-α and interferon-γ in RPMI: 14.15 ± 1.68, TNF-α and interferon-γ in 4.25% low GDP PD fluid 13.16 ± 3.29, TNF-α and interferon-γ in 4.25% high GDP PD fluid 25.88 ± 2.18%, p < 0.05 vs. the other two groups). BclxL BH4 peptides, Apaf-1 inhibition or caspase inhibition failed to protect from apoptosis induced by the combination of inflammatory cytokines and bioincompatible PD fluids, although they protected from other forms of mesothelial cell apoptosis. CONCLUSION: Inflammation cooperates with high GDP PD fluids to promote mesothelial cell death, which is resistant to several therapeutic approaches. This information provides a framework for selection of PD fluid during peritonitis.


Subject(s)
Apoptosis/drug effects , Biocompatible Materials/pharmacology , Epithelial Cells/drug effects , Aged , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Biocompatible Materials/chemistry , Caspases/genetics , Caspases/metabolism , Dialysis Solutions/chemistry , Dialysis Solutions/pharmacology , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelium/drug effects , Epithelium/metabolism , Female , Gene Expression/drug effects , Glucose/pharmacology , Humans , Inflammation , Interferon-gamma/pharmacology , Male , Middle Aged , Models, Biological , Peptides/genetics , Peptides/metabolism , Peritoneal Dialysis , Primary Cell Culture , Tumor Necrosis Factor-alpha/pharmacology , bcl-X Protein/antagonists & inhibitors , bcl-X Protein/genetics , bcl-X Protein/metabolism
10.
J Cell Physiol ; 230(7): 1580-93, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25536182

ABSTRACT

The tubular epithelium may be intrinsically involved in promoting kidney injury by junctional instability, epithelial-mesenchymal transition (EMT) and extracellular matrix remodelling. In this work, we investigated whether the pleiotropic and proinflammatory cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK), could be able to disturb junctional protein expression and to induce EMT of tubular cells. In cultured murine proximal tubular cells TWEAK induced phenotypic changes that were accompanied by F-actin redistribution, loss of epithelial adherent (E-cadherin, Cadherin-16, ß-catenin) and tight junction (ZO-1) proteins, and re-expression of the mesenchymal protein Vimentin. The transcriptional repressors Snail and HNF1ß were also modulated by TWEAK. In a murine model of obstructive renal pathology, TWEAK expression correlated with the appearance of the mesenchymal marker αSMA in kidney tubular cells. Mechanistically, the epithelial changes induced by TWEAK, including loss of epithelial integrity and EMT, via Fn14 were TGF-ß1 independent, but mediated by several intracellular signaling systems, including the canonical NF-κB, ERK activation and the vitamin D receptor modulation. These results highlight potential contributions of TWEAK-induced inflammatory mechanisms that could unveil new pathogenic effects of TWEAK starting tubulointerstitial damage and fibrosis.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Kidney Tubules/cytology , NF-kappa B/metabolism , Tight Junction Proteins/metabolism , Tumor Necrosis Factors/metabolism , Animals , Cadherins/genetics , Cadherins/metabolism , Cell Line , Cytokine TWEAK , Dogs , Epithelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation/physiology , Humans , Mice , Mice, Knockout , NF-kappa B/genetics , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Renal Insufficiency/metabolism , Tight Junction Proteins/genetics , Tumor Necrosis Factors/genetics , Tumor Necrosis Factors/pharmacology
11.
J Nephrol ; 27(6): 607-17, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24756971

ABSTRACT

Vascular calcification in chronic kidney disease (CKD) patients is associated to increased mortality. Osteoprotegerin (OPG) is a soluble tumor necrosis factor (TNF) superfamily receptor that inhibits the actions of the cytokines receptor activator of nuclear factor kappa-B ligand (RANKL) and TNF-related apoptosis-inducing ligand (TRAIL) by preventing their binding to signaling receptors in the cell membrane. OPG-deficient mice display vascular calcification while OPG prevented calcification of cultured vascular smooth muscle cells and protected kidney cells from TRAIL-induced death. OPG may be a biomarker in patients with kidney disease. Circulating OPG is increased in predialysis, dialysis and transplant CKD patients and may predict vascular calcification progression and patient survival. By contrast, circulating OPG is decreased in nephrotic syndrome. In addition, free and exosome-bound urinary OPG is increased in human kidney disease. Increased urinary OPG has been associated with lupus nephritis activity. Despite the association of high OPG levels with disease, experimental functional information available suggests that OPG might be protective in kidney disease and in vascular injury in the context of uremia. Thus, tissue injury results in increased OPG, while OPG may protect from tissue injury. Recombinant OPG was safe in phase I randomized controlled trials. Further research is needed to fully define the therapeutic and biomarker potential of OPG in patients with kidney disease.


Subject(s)
Kidney Diseases/metabolism , Muscle, Smooth, Vascular/metabolism , Osteoprotegerin/metabolism , Signal Transduction , Vascular Calcification/metabolism , Animals , Humans , Kidney Diseases/complications , Kidney Diseases/drug therapy , Muscle, Smooth, Vascular/pathology , Osteoprotegerin/therapeutic use , Recombinant Proteins/therapeutic use , TNF-Related Apoptosis-Inducing Ligand/metabolism , Vascular Calcification/drug therapy , Vascular Calcification/etiology
12.
Nephrol Dial Transplant ; 29 Suppl 1: i54-i62, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24493870

ABSTRACT

Tumour necrosis factor-like weak inducer of apoptosis (TWEAK) activates the fibroblast growth factor-inducible-14 (Fn14) receptor. TWEAK has actions on intrinsic kidney cells and on inflammatory cells of potential pathophysiological relevance. The effects of TWEAK in tubular cells have been explored in most detail. In cultured murine tubular cells TWEAK induces the expression of inflammatory cytokines, downregulates the expression of Klotho, is mitogenic, and in the presence of sensitizing agents promotes apoptosis. Similar actions were observed on glomerular mesangial cells. In vivo TWEAK actions on healthy kidneys mimic cell culture observations. Increased expression of TWEAK and Fn14 was reported in human and experimental acute and chronic kidney injury. The role of TWEAK/Fn14 in kidney injury has been demonstrated in non-inflammatory compensatory renal growth, acute kidney injury and chronic kidney disease of immune and non-immune origin, including hyperlipidaemic nephropathy, lupus nephritis (LN) and anti-GBM nephritis. The nephroprotective effect of TWEAK or Fn14 targeting in immune-mediated kidney injury is the result of protection from TWEAK-induced injury of renal intrinsic cells, not from interference with the immune response. A phase I dose-ranging clinical trial demonstrated the safety of anti-TWEAK antibodies in humans. A phase II randomized placebo-controlled clinical trial exploring the efficacy, safety and tolerability of neutralizing anti-TWEAK antibodies as a tissue protection strategy in LN is ongoing. The eventual success of this trial may expand the range of kidney diseases in which TWEAK targeting should be explored.


Subject(s)
Apoptosis , Kidney Diseases/diagnosis , Translational Research, Biomedical , Tumor Necrosis Factors/metabolism , Animals , Cytokine TWEAK , Disease Progression , Humans , Kidney Diseases/metabolism , Mice
13.
Int Urol Nephrol ; 46(4): 765-76, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24072452

ABSTRACT

Unilateral ureteral obstruction is a popular experimental model of renal injury. However, the study of the kidney response to urinary tract obstruction is only one of several advantages of this model. Unilateral ureteral obstruction causes subacute renal injury characterized by tubular cell injury, interstitial inflammation and fibrosis. For this reason, it serves as a model both of irreversible acute kidney injury and of events taking place during human chronic kidney disease. Being a unilateral disease, it is not useful to study changes in global kidney function, but has the advantage of a low mortality and the availability of an internal control (the non-obstructed kidney). Experimental unilateral ureteral obstruction has illustrated the molecular mechanisms of apoptosis, inflammation and fibrosis, all three key processes in kidney injury of any cause, thus providing information beyond obstruction. Recently this model has supported key concepts on the role in kidney fibrosis of epithelial-mesenchymal transition, tubular epithelial cell G2/M arrest, the anti-aging hormone Klotho and renal innervation. We now review the experimental model and its contribution to identifying novel therapeutic targets in kidney injury and fibrosis, independently of the noxa.


Subject(s)
Acute Kidney Injury/drug therapy , Acute Kidney Injury/metabolism , Disease Models, Animal , Kidney/pathology , Ureteral Obstruction/complications , Acute Kidney Injury/etiology , Acute Kidney Injury/pathology , Animals , Apoptosis , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Humans , Mice , Molecular Targeted Therapy , Nephritis, Interstitial/etiology , Nephritis, Interstitial/metabolism , Rats , Renal Insufficiency, Chronic/etiology
14.
Int Rev Immunol ; 33(1): 9-22, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23802146

ABSTRACT

Recent advances in cell death biology have uncovered an ever increasing range of cell death forms. Macrophages have a bidirectional relationship with cell death that modulates the immune response. Thus, macrophages engulf apoptotic cells and secrete cytokines that may promote cell death in parenchymal cells. Furthermore, the presence of apoptotic or necrotic dead cells in the microenvironment elicits differential macrophage responses. Apoptotic cells elicit anti-inflammatory responses in macrophages. By contrast macrophages may undergo a proinflammatory form of cell death (pyroptosis) in response to damage-associated molecular patterns (DAMPs) released from necrotic cells and also in response to pathogen-associated molecular patterns (PAMPs). Pyroptosis is a recently identified form of cell death that occurs predominantly in subsets of inflammatory macrophages and is associated to the release of interleukin-1ß (IL-1ß) and IL-18. Deregulation of these processes may result in disease. Thus, failure of macrophages to engulf apoptotic cells may be a source of autoantigens in autoimmune diseases, excessive macrophage release of proapoptotic factors or sterile pyroptosis may contribute to tissue injury and failure of pathogen-induced pyroptosis may contribute to pathogen survival. Ongoing research is exploring the therapeutic opportunities resulting this new knowledge.


Subject(s)
Cell Death/physiology , Macrophages/immunology , Animals , Apoptosis , Humans , Macrophages/metabolism , Necrosis
15.
Biochim Biophys Acta ; 1842(2): 293-303, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24287278

ABSTRACT

Diabetic nephropathy (DN) is the most common cause of end-stage renal disease and identification of new therapeutic targets is needed. Nicotinamide phosphoribosyltransferase (NAMPT) is both an extracellular and intracellular protein. Circulating NAMPT is increased in diabetics and in chronic kidney disease patients. The role of NAMPT in renal cell biology is poorly understood. NAMPT mRNA and protein were increased in the kidneys of rats with streptozotocin-induced diabetes. Immunohistochemistry localized NAMPT to glomerular and tubular cells in diabetic rats. The inflammatory cytokine TNFα increased NAMPT mRNA, protein and NAD production in cultured kidney human tubular cells. Exogenous NAMPT increased the mRNA expression of chemokines MCP-1 and RANTES. The NAMPT enzymatic activity inhibitor FK866 prevented these effects. By contrast, FK866 boosted TNFα-induced expression of MCP-1 and RANTES mRNA and endogenous NAMPT targeting by siRNA also had a proinflammatory effect. Furthermore, FK866 promoted tubular cell apoptosis in an inflammatory milieu containing the cytokines TNFα/IFNγ. In an inflammatory environment FK866 promoted tubular cell expression of the lethal cytokine TRAIL. These data are consistent with a role of endogenous NAMPT activity as an adaptive, protective response to an inflammatory milieu that differs from the proinflammatory activity of exogenous NAMPT. Thus, disruption of endogenous NAMPT function in stressed cells promotes tubular cell death and chemokine expression. This information may be relevant for the design of novel therapeutic strategies in DN.


Subject(s)
Apoptosis/genetics , Chemokines/genetics , Epithelial Cells/metabolism , Kidney Tubules, Proximal/cytology , Nicotinamide Phosphoribosyltransferase/genetics , Acrylamides/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokines/metabolism , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/metabolism , Dose-Response Relationship, Drug , Epithelial Cells/drug effects , Gene Expression/drug effects , Humans , Immunohistochemistry , Interleukin-6/genetics , Interleukin-6/metabolism , Kidney/metabolism , Kidney/pathology , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism , Nicotinamide Phosphoribosyltransferase/pharmacology , Piperidines/pharmacology , RNA Interference , Rats , Rats, Inbred WKY , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/pharmacology
16.
PLoS One ; 8(8): e72387, 2013.
Article in English | MEDLINE | ID: mdl-24058411

ABSTRACT

Urinary exosomes have been proposed as potential diagnostic tools. TNF superfamily cytokines and receptors may be present in exosomes and are expressed by proximal tubular cells. We have now studied the expression of selected TNF superfamily proteins in exosome-like vesicles from cultured human proximal tubular cells and human urine and have identified additional proteins in these vesicles by LC-MS/MS proteomics. Human proximal tubular cells constitutively released exosome-like vesicles that did not contain the TNF superfamily cytokines TRAIL or TWEAK. However, exosome-like vesicles contained osteoprotegerin (OPG), a TNF receptor superfamily protein, as assessed by Western blot, ELISA or selected reaction monitoring by nLC-(QQQ)MS/MS. Twenty-one additional proteins were identified in tubular cell exosome-like vesicles, including one (vitamin D binding protein) that had not been previously reported in exosome-like vesicles. Twelve were extracellular matrix proteins, including the basement membrane proteins type IV collagen, nidogen-1, agrin and fibulin-1. Urine from chronic kidney disease patients contained a higher amount of exosomal protein and exosomal OPG than urine from healthy volunteers. Specifically OPG was increased in autosomal dominant polycystic kidney disease urinary exosome-like vesicles and expressed by cystic epithelium in vivo. In conclusion, OPG is present in exosome-like vesicles secreted by proximal tubular epithelial cells and isolated from Chronic Kidney Disease urine.


Subject(s)
Exosomes/chemistry , Kidney Tubules, Proximal/cytology , Osteoprotegerin/analysis , Osteoprotegerin/urine , Renal Insufficiency, Chronic/urine , Adult , Aged , Aged, 80 and over , Amino Acid Sequence , Cell Line , Cytokine TWEAK , Female , Humans , Male , Middle Aged , Molecular Sequence Data , TNF-Related Apoptosis-Inducing Ligand/analysis , Tumor Necrosis Factors/analysis
17.
Biochim Biophys Acta ; 1832(10): 1744-55, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23748045

ABSTRACT

Tumor necrosis factor-like weak inducer of apoptosis (TWEAK) regulates apoptosis, proliferation and inflammation in renal epithelial cells and plays a role in acute kidney injury. However, there is little information on the chronic effects of TWEAK. We hypothesized that TWEAK may influence renal fibrosis and regulate kidney fibroblast biology, in part, through Ras pathway. We studied a chronic model of experimental unilateral ureteral obstruction in wild type and TWEAK deficient mice, and a murine model of systemic TWEAK overexpression. TWEAK actions were also explored in cultured renal and embryonic fibroblasts. TWEAK and TWEAK receptor expression was increased in the obstructed kidneys. The absence of TWEAK decreased early kidney tubular damage, inflammatory infiltrates and myofibroblast number. TWEAK deficient mice had decreased renal fibrosis 21days after obstruction, as assessed by extracellular matrix staining. In mice without prior underlying kidney disease, systemic overexpression of TWEAK induced kidney inflammation and fibrosis. In cultured fibroblasts, TWEAK induced proliferation through activation of the Ras/ERK pathway. TWEAK also activated nuclear factor κB (NFκB)-dependent inflammatory chemokine production in murine renal fibroblasts. In conclusion, lack of TWEAK reduces renal fibrosis in a model of persistent kidney insult and overexpression of TWEAK led to renal fibrosis. TWEAK actions on renal fibroblasts may contribute to the in vivo observations, as TWEAK promotes inflammatory activity and proliferation in fibroblast cultures.


Subject(s)
Cell Proliferation , Fibrosis/physiopathology , Kidney Diseases/physiopathology , Kidney/pathology , Tumor Necrosis Factors/physiology , ras Proteins/physiology , Animals , Cell Line , Cytokine TWEAK , Fibroblasts/pathology , Mice
18.
Photochem Photobiol ; 89(4): 953-60, 2013.
Article in English | MEDLINE | ID: mdl-23360312

ABSTRACT

Metabolic syndrome is characterized by hyperglycemia, hypertension, dyslipidemia and obesity. Diabetes and hypertension are the main causes of chronic end-stage kidney disease in humans. Chronic kidney disease is characterized by kidney inflammation and eventual development of kidney fibrosis. Low-level laser (or light) therapy (LLLT) can be used to relieve pain associated with some inflammatory diseases due to photochemical effects. Despite the known contribution of inflammation to metabolic syndrome and kidney disease, there is scarce information on the potential therapeutic use of LLLT in renal disease. The aim of this randomized, placebo-controlled study was to test the hypothesis that LLLT could modulate chronic kidney injury. Rats with nephropathy, hypertension, hyperlipidemia and type II diabetes (strain ZSF1) were subjected to three different conditions of LLLT or sham treatment for 8 weeks, and then sacrificed 10 weeks later. The main findings of this study are that the LLLT-treated rats had lower blood pressure after treatment and a better preserved glomerular filtration rate with less interstitial fibrosis upon euthanasia at the end of follow-up. This initial proof-of-concept study suggests that LLLT may modulate chronic kidney disease progression, providing a painless, noninvasive, therapeutic strategy, which should be further evaluated.


Subject(s)
Laser Therapy/methods , Metabolic Syndrome/complications , Renal Insufficiency, Chronic/surgery , Animals , Collagen , Fibrosis/surgery , Kidney/pathology , Rats , Renal Insufficiency, Chronic/pathology
19.
PLoS One ; 8(1): e51992, 2013.
Article in English | MEDLINE | ID: mdl-23300960

ABSTRACT

The polyglutamic acid/peptoid 1 (QM56) nanoconjugate inhibits apoptosis by interfering with Apaf-1 binding to procaspase-9. We now describe anti-inflammatory properties of QM56 in mouse kidney and renal cell models.In cultured murine tubular cells, QM56 inhibited the inflammatory response to Tweak, a non-apoptotic stimulus. Tweak induced MCP-1 and Rantes synthesis through JAK2 kinase and NF-κB activation. Similar to JAK2 kinase inhibitors, QM56 inhibited Tweak-induced NF-κB transcriptional activity and chemokine expression, despite failing to inhibit NF-κB-p65 nuclear translocation and NF-κB DNA binding. QM56 prevented JAK2 activation and NF-κB-p65(Ser536) phosphorylation. The anti-inflammatory effect and JAK2 inhibition by QM56 were observed in Apaf-1(-/-) cells. In murine acute kidney injury, QM56 decreased tubular cell apoptosis and kidney inflammation as measured by down-modulations of MCP-1 and Rantes mRNA expression, immune cell infiltration and activation of the JAK2-dependent inflammatory pathway.In conclusion, QM56 has an anti-inflammatory activity which is independent from its role as inhibitor of Apaf-1 and apoptosis and may have potential therapeutic relevance.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/pathology , Kidney Tubules/pathology , Nanomedicine/methods , Peptoids/pharmacology , Polyglutamic Acid/analogs & derivatives , Active Transport, Cell Nucleus , Animals , Apoptosis , Cell Line , Chemokine CCL2/metabolism , Chemokine CCL5/metabolism , Disease Models, Animal , Janus Kinase 2/metabolism , Kidney/metabolism , Kidney/pathology , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Nanoconjugates , Nanoparticles/chemistry , Peptoids/chemistry , Polyglutamic Acid/chemistry , Polyglutamic Acid/pharmacology
20.
Nephrol Dial Transplant ; 27(7): 2720-33, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22302208

ABSTRACT

BACKGROUND: Angiotensin receptor 1 blockers (ARB) are standard nephroprotective drugs in chronic kidney disease. There is less evidence for a nephroprotective effect of HMG-CoA reductase inhibitors (statins) and much less is known about potential benefits of combination therapy. We evaluated the therapeutic potential of a statin alone or in combination with an ARB in experimental chronic kidney disease. METHODS: Subtotally nephrectomized (5/6 Nx) rats were treated early with vehicle, losartan, cerivastatin or losartan/cerivastatin. Expression of messenger RNA (mRNA) was assessed by real-time reverse transcription-polymerase chain reaction. Tissue proteins were localized by immunohistochemistry. Nuclear factor-κB (NF-κB) activation was measured in whole kidneys. RESULTS: In contrast to the sham group, at 6 weeks, vehicle-treated 5/6 Nx rats displayed renal lesions, albuminuria and increased blood pressure, serum creatinine and total kidney NF-κB p65 DNA-binding activity and preproendothelin-1, fibronectin and type I and III collagen mRNA. NF-κB activation correlated with albuminuria and histological renal injury. Losartan or combination therapy preserved renal function, abrogated albuminuria and improved glomerular and interstitial histology. Cerivastatin alone preserved renal function and improved interstitial injury but did not influence albuminuria, glomerular histology or NF-κB activation. Losartan/cerivastatin normalized kidney NF-κB activation and extracellular matrix mRNA expression pattern. The effect of losartan alone on these parameters was less intense. All treatments decreased preproendothelin-1 mRNA and preserved interstitial capillaries. CONCLUSIONS: In a chronic kidney disease model, early treatment with either an ARB or a statin preserved renal function although the mechanisms differed. Combination therapy with an ARB and a statin did not confer clear-cut advantages on biochemical and histological parameters over ARB alone, although it further improved the kidney NF-κB and gene expression profile.


Subject(s)
Angiotensin Receptor Antagonists/therapeutic use , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Losartan/therapeutic use , Nephrectomy/adverse effects , Pyridines/therapeutic use , Angiotensin II Type 1 Receptor Blockers/antagonists & inhibitors , Animals , Blotting, Western , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type III/genetics , Collagen Type III/metabolism , Drug Therapy, Combination , Fibronectins/genetics , Fibronectins/metabolism , Kidney Diseases/pathology , Male , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL
...