Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38645053

ABSTRACT

In the last decade cellular senescence, a hallmark of aging, has come into focus for pharmacologically targeting aging processes. Senolytics are one of these interventive strategies that have advanced into clinical trials, creating an unmet need for minimally invasive biomarkers of senescent cell load to identify patients at need for senotherapy. We created a landscape of miRNA and mRNA expression in five human cell types induced to senescence in-vitro and provide proof-of-principle evidence that miRNA expression can track senescence burden dynamically in-vivo using transgenic p21 high senescent cell clearance in HFD fed mice. Finally, we profiled miRNA expression in seven different tissues, total plasma, and plasma derived EVs of young and 25 months old mice. In a systematic analysis, we identified 22 candidate senomiRs with potential to serve as circulating biomarkers of senescence not only in rodents, but also in upcoming human clinical senolytic trials.

2.
Nat Cancer ; 5(3): 448-462, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38267628

ABSTRACT

Chemotherapy often generates intratumoral senescent cancer cells that strongly modify the tumor microenvironment, favoring immunosuppression and tumor growth. We discovered, through an unbiased proteomics screen, that the immune checkpoint inhibitor programmed cell death 1 ligand 2 (PD-L2) is highly upregulated upon induction of senescence in different types of cancer cells. PD-L2 is not required for cells to undergo senescence, but it is critical for senescent cells to evade the immune system and persist intratumorally. Indeed, after chemotherapy, PD-L2-deficient senescent cancer cells are rapidly eliminated and tumors do not produce the senescence-associated chemokines CXCL1 and CXCL2. Accordingly, PD-L2-deficient pancreatic tumors fail to recruit myeloid-derived suppressor cells and undergo regression driven by CD8 T cells after chemotherapy. Finally, antibody-mediated blockade of PD-L2 strongly synergizes with chemotherapy causing remission of mammary tumors in mice. The combination of chemotherapy with anti-PD-L2 provides a therapeutic strategy that exploits vulnerabilities arising from therapy-induced senescence.


Subject(s)
Pancreatic Neoplasms , Animals , Mice , Pancreatic Neoplasms/metabolism , CD8-Positive T-Lymphocytes/pathology , Immune Tolerance , Immunosuppression Therapy , Cellular Senescence , Tumor Microenvironment
3.
Nat Metab ; 5(12): 2111-2130, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38097808

ABSTRACT

Fibrogenesis is part of a normal protective response to tissue injury that can become irreversible and progressive, leading to fatal diseases. Senescent cells are a main driver of fibrotic diseases through their secretome, known as senescence-associated secretory phenotype (SASP). Here, we report that cellular senescence, and multiple types of fibrotic diseases in mice and humans are characterized by the accumulation of iron. We show that vascular and hemolytic injuries are efficient in triggering iron accumulation, which in turn can cause senescence and promote fibrosis. Notably, we find that senescent cells persistently accumulate iron, even when the surge of extracellular iron has subdued. Indeed, under normal conditions of extracellular iron, cells exposed to different types of senescence-inducing insults accumulate abundant ferritin-bound iron, mostly within lysosomes, and present high levels of labile iron, which fuels the generation of reactive oxygen species and the SASP. Finally, we demonstrate that detection of iron by magnetic resonance imaging might allow non-invasive assessment of fibrotic burden in the kidneys of mice and in patients with renal fibrosis. Our findings suggest that iron accumulation plays a central role in senescence and fibrosis, even when the initiating events may be independent of iron, and identify iron metabolism as a potential therapeutic target for senescence-associated diseases.


Subject(s)
Cellular Senescence , Senescence-Associated Secretory Phenotype , Humans , Iron , Kidney , Fibrosis
4.
Nat Med ; 28(8): 1556-1568, 2022 08.
Article in English | MEDLINE | ID: mdl-35953721

ABSTRACT

Interlinked and fundamental aging processes appear to be a root-cause contributor to many disorders and diseases. One such process is cellular senescence, which entails a state of cell cycle arrest in response to damaging stimuli. Senescent cells can arise throughout the lifespan and, if persistent, can have deleterious effects on tissue function due to the many proteins they secrete. In preclinical models, interventions targeting those senescent cells that are persistent and cause tissue damage have been shown to delay, prevent or alleviate multiple disorders. In line with this, the discovery of small-molecule senolytic drugs that selectively clear senescent cells has led to promising strategies for preventing or treating multiple diseases and age-related conditions in humans. In this Review, we outline the rationale for senescent cells as a therapeutic target for disorders across the lifespan and discuss the most promising strategies-including recent and ongoing clinical trials-for translating small-molecule senolytics and other senescence-targeting interventions into clinical use.


Subject(s)
Aging , Senotherapeutics , Aging/physiology , Cell Cycle Checkpoints , Cellular Senescence , Humans
5.
Handb Exp Pharmacol ; 274: 165-180, 2022.
Article in English | MEDLINE | ID: mdl-34697668

ABSTRACT

Obesity is a major risk factor for the development of comorbidities such as type 2 diabetes, neurodegenerative disorders, osteoarthritis, cancer, cardiovascular and renal diseases. The onset of obesity is linked to an increase of senescent cells within adipose tissue and other organs. Cellular senescence is a stress response that has been shown to be causally linked to aging and development of various age-related diseases such as obesity. The senescence-associated-secretory phenotype of senescent cells creates a chronic inflammatory milieu that leads to local and systemic dysfunction. The elimination of senescent cells using pharmacological approaches (i.e., senolytics) has been shown to delay, prevent, or alleviate obesity-related organ dysfunction.


Subject(s)
Diabetes Mellitus, Type 2 , Senotherapeutics , Cellular Senescence/physiology , Humans , Obesity/drug therapy
7.
PLoS One ; 16(8): e0254697, 2021.
Article in English | MEDLINE | ID: mdl-34424918

ABSTRACT

The PAF complex (PAFC) coordinates transcription elongation and mRNA processing and its CDC73/parafibromin subunit functions as a tumour suppressor. The NF2/Merlin tumour suppressor functions both at the cell cortex and nucleus and is a key mediator of contact inhibition but the molecular mechanisms remain unclear. In this study we have used affinity proteomics to identify novel Merlin interacting proteins and show that Merlin forms a complex with multiple proteins involved in RNA processing including the PAFC and the CHD1 chromatin remodeller. Tumour-derived inactivating mutations in both Merlin and the CDC73 PAFC subunit mutually disrupt their interaction and growth suppression by Merlin requires CDC73. Merlin interacts with the PAFC in a cell density-dependent manner and we identify a role for FAT cadherins in regulating the Merlin-PAFC interaction. Our results suggest that in addition to its function within the Hippo pathway, Merlin is part of a tumour suppressor network regulated by cell-cell adhesion which coordinates post-initiation steps of the transcription cycle of genes mediating contact inhibition.


Subject(s)
Cell Adhesion/genetics , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Neoplasms/genetics , Neurofibromin 2/genetics , Tumor Suppressor Proteins/genetics , Cell Proliferation/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly/genetics , Contact Inhibition/genetics , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Neoplasms/pathology , Protein Binding/genetics , Protein Interaction Maps/genetics , Signal Transduction/genetics
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.
AAPS J ; 20(1): 9, 2017 11 30.
Article in English | MEDLINE | ID: mdl-29192345

ABSTRACT

Combining the multikinase inhibitor sorafenib with the platinum-based chemotherapy of solid tumors was expected to improve treatment outcome. However, in many clinical trials, no benefit from sorafenib addition to the platinum-containing regimen could be demonstrated. Moreover, in some studies, decreased survival of ovarian cancer patients as well as non-small cell lung cancer patients with squamous cell histology was observed. The aim of this study was to investigate the cellular mechanisms of the pharmacological interaction between platinum drugs and sorafenib in different cancer cell lines. The interaction was characterized by combination index analysis, platinum accumulation and DNA platination were determined using flameless atomic absorption spectrometry, and protein expression was assessed with Western blot. In the sensitive A2780 ovarian carcinoma and H520 squamous cell lung carcinoma cell lines, sorafenib induced downregulation of Na+,K+-ATPase. In A2780 cells, the kinase inhibitor also decreased the expression of copper transporter 1 (CTR1). As a result, sorafenib treatment led to a diminished cellular accumulation of cisplatin and carboplatin and to a decrease in DNA platination in these cell lines. This was not the case in the cisplatin-resistant A2780cis ovarian carcinoma and H522 lung adenocarcinoma cell lines featuring lower basal expression of the above-mentioned transporters. In all cell lines studied, an antagonistic interaction between platinum drugs and sorafenib was found. Our results suggest that sorafenib impairs cisplatin and carboplatin uptake through downregulation of CTR1 and/or Na+,K+-ATPase resulting in reduction of DNA platination. This effect is not observed in cancer cells with defects in platinum accumulation.


Subject(s)
Antineoplastic Agents/pharmacology , Carboplatin/pharmacokinetics , Cation Transport Proteins/physiology , Cisplatin/pharmacokinetics , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Sodium-Potassium-Exchanging ATPase/physiology , Adenosine Triphosphate/metabolism , Cation Transport Proteins/antagonists & inhibitors , Cell Line, Tumor , Copper Transporter 1 , DNA/metabolism , Drug Interactions , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Niacinamide/pharmacology , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Sorafenib
10.
J Am Chem Soc ; 139(26): 8808-8811, 2017 07 05.
Article in English | MEDLINE | ID: mdl-28625064

ABSTRACT

A naphthalimide-based two-photon probe (AHGa) for the detection of cell senescence is designed. The probe contains a naphthalimide core, an l-histidine methyl ester linker, and an acetylated galactose bonded to one of the aromatic nitrogen atoms of the l-histidine through a hydrolyzable N-glycosidic bond. Probe AHGa is transformed into AH in senescent cells resulting in an enhanced fluorescent emission intensity. In vivo detection of senescence is validated in mice bearing tumor xenografts treated with senescence-inducing chemotherapy.


Subject(s)
Fluorescent Dyes/chemistry , Naphthalimides/chemistry , Neoplasms/drug therapy , Photons , Animals , Cellular Senescence/drug effects , Humans , Mice , Reference Standards , Xenograft Model Antitumor Assays
11.
PLoS One ; 11(12): e0165214, 2016.
Article in English | MEDLINE | ID: mdl-27959931

ABSTRACT

BACKGROUND: There is great interest in repurposing the commonly prescribed anti-diabetic drug metformin for cancer therapy. Intracellular uptake and retention of metformin is affected by the expression of organic cation transporters (OCT) 1-3 and by multidrug and toxic compound extrusion (MATE) 1-2. Inside cells, metformin inhibits mitochondrial function, which leads to reduced oxygen consumption and inhibition of proliferation. Reduced oxygen consumption can lead to improved tumor oxygenation and radiation response. PURPOSE: Here we sought to determine if there is an association between the effects of metformin on inhibiting oxygen consumption, proliferation and expression of OCTs and MATEs in a panel of 19 cancer cell lines. RESULTS: There was relatively large variability in the anti-proliferative response of different cell lines to metformin, with a subset of cell lines being very resistant. In contrast, all cell lines demonstrated sensitivity to the inhibition of oxygen consumption by metformin, with relatively small variation. The expression of OCT1 correlated with expression of both OCT2 and OCT3. OCT1 and OCT2 were relatively uniformly expressed, whereas expression of OCT3, MATE1 and MATE2 showed substantial variation across lines. There were statistically significant associations between resistance to inhibition of proliferation and MATE2 expression, as well as between sensitivity to inhibition of oxygen consumption and OCT3 expression. One cell line (LNCaP) with high OCT3 and low MATE2 expression in concert, had substantially higher intracellular metformin concentration than other cell lines, and was exquisitely sensitive to both anti-proliferative and anti-respiratory effects. In all other cell lines, the concentration of metformin required to inhibit oxygen consumption acutely in vitro was substantially higher than that achieved in the plasma of diabetic patients. However, administering anti-diabetic doses of metformin to tumor-bearing mice resulted in intratumoral accumulation of metformin and reduced hypoxic tumor fractions. CONCLUSIONS: All cancer cells are susceptible to inhibition of oxygen consumption by metformin, which results in reduced hypoxic tumor fractions beneficial for the response to radiotherapy. High MATE2 expression may result in resistance to the anti-proliferative effect of metformin and should be considered as a negative predictive biomarker in clinical trials.


Subject(s)
Antineoplastic Agents/pharmacology , Biomarkers, Tumor/metabolism , Drug Resistance, Neoplasm , Hypoglycemic Agents/pharmacology , Metformin/pharmacology , Organic Cation Transport Proteins/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Cell Proliferation/drug effects , Cells, Cultured , HCT116 Cells , HeLa Cells , Humans , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/metabolism , Octamer Transcription Factors/genetics , Octamer Transcription Factors/metabolism , Organic Cation Transport Proteins/genetics , Oxygen Consumption/drug effects
12.
Clin Cancer Res ; 19(24): 6741-50, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24141625

ABSTRACT

PURPOSE: Tumor hypoxia is a negative prognostic factor in multiple cancers, due in part to its role in causing resistance to radiotherapy. Hypoxia arises in tumor regions distal to blood vessels as oxygen is consumed by more proximal tumor cells. Reducing the rate of oxygen consumption is therefore a potential strategy to reduce tumor hypoxia. We hypothesized that the anti-diabetic drug metformin, which reduces oxygen consumption through inhibition of mitochondrial complex I, would improve radiation response by increasing tumor oxygenation. EXPERIMENTAL DESIGN: Tumor hypoxia was measured in xenografts before and after metformin treatment using 2-nitroimidazole hypoxia markers quantified by immunohistochemistry (IHC), flow cytometry, and positron emission tomography (PET) imaging. Radiation response was determined by tumor growth delay and clonogenic survival in xenografts with and without administration of metformin. The impact of metformin use on outcome was assessed in 504 patients with localized prostate cancer treated with curative-intent, image-guided radiotherapy (IGRT) from 1996 to 2012. Three-year biochemical relapse-free rates were assessed using the Kaplan-Meier method. RESULTS: Metformin treatment significantly improved tumor oxygenation in two xenograft models as measured by IHC, flow cytometry, and PET imaging. Metformin also led to improved radiotherapy responses when mice were administered metformin immediately before irradiation. Clinically, metformin use was associated with an independent and significant decrease in early biochemical relapse rates (P = 0.0106). CONCLUSION: Our data demonstrate that metformin can improve tumor oxygenation and response to radiotherapy. Our study suggests that metformin may represent an effective and inexpensive means to improve radiotherapy outcome with an optimal therapeutic ratio.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/radiotherapy , Metformin/administration & dosage , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/radiotherapy , Animals , Cell Hypoxia/drug effects , Colorectal Neoplasms/pathology , Electron Transport Complex I/drug effects , HCT116 Cells , Humans , Male , Mice , Oxygen/metabolism , Oxygen Consumption/drug effects , Positron-Emission Tomography , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...