Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters










Publication year range
2.
Methods Cell Biol ; 185: xvii-xxiv, 2024.
Article in English | MEDLINE | ID: mdl-38556455
3.
Methods Cell Biol ; 176: xvii-xix, 2023.
Article in English | MEDLINE | ID: mdl-37164545
4.
Methods Cell Biol ; 175: 1-15, 2023.
Article in English | MEDLINE | ID: mdl-36967137

ABSTRACT

Primary cilia (PC) are sensory organelles that function as cellular antennas, transmitting signals between the extracellular and intracellular spaces in many vertebrate tissues. The cell generates and assembles PC through a highly regulated process called ciliogenesis. This complex process is involved in several physiological functions, including embryonic development, locomotion, cell cycle regulation or energetic homeostasis control. In general, when a cell finishes its cell division, the oldest centriole usually migrates to the plasma membrane and becomes a basal body that gives rise to the formation of a cilium. For this reason, the presence of cilia is incompatible with cell division, so when a cell is going to divide, the cilium and the basal body disappear. Ciliogenesis is triggered by various stimuli, all of them related to cell cycle blockade. This cell cycle, and ciliogenesis induction, can be observed by: (1) the influence of growth factors (lack of serum and consequent inability to promote cell cycle exit and increase the proportion of cells in G0); (2) pharmacological cell cycle inhibitors (staurosporine or etoposide); or (3) physiological cell cycle inhibition (excessive contact between neighboring cells). Evaluation of ciliogenesis induction is vitally important for the study of diseases related to ciliary dysfunction, called ciliopathies. That is why the use of correct protocols for inducing cilia formation and an accurate posterior visualization of the cilia after performing said protocols are essential parts in the study of these diseases. To facilitate this task, here we described detailed protocols to induce ciliogenesis in vitro and visualize PC by immunofluorescence microscopy in cultured cells.


Subject(s)
Cilia , Organelles , Cilia/metabolism , Cells, Cultured , Cell Division , Cell Cycle , Centrioles/physiology
5.
Methods Cell Biol ; 175: xv-xviii, 2023.
Article in English | MEDLINE | ID: mdl-36967149

Subject(s)
Cilia
6.
Int Rev Cell Mol Biol ; 373: 107-123, 2022.
Article in English | MEDLINE | ID: mdl-36283764

ABSTRACT

The therapeutic outcome of multiple anticancer regimens relies upon a fine balance between tumor intrinsic and host-related factors. In this context, qualitative changes in dietary composition as well as alterations in total calorie supply influence essential aspects of cancer biology, spanning from tumor initiation to metastatic spreading. On the one hand, circumstances of nutritional imbalance or excessive calorie intake promote oncogenesis, accelerate tumor progression, and hamper the efficacy of anticancer treatments. On the other hand, approaches based on bulk (e.g., fasting, fasting mimicking diets) or selective (e.g., amino acids) shortage of nutrients are currently in the spotlight for their ability to potentiate the effect of anticancer drugs. While the chemosensitizing effect of fasting has long been attributed to the overdemanding metabolic requirements of neoplastic cells, recent findings suggest that caloric restriction improves the efficacy of chemotherapy and immunotherapy by boosting anticancer immunosurveillance. Here, we provide a critical overview of current preclinical and clinical studies that address the impact of nutritional interventions on the response to cancer therapy, laying particular emphasis on fasting-related interventions.


Subject(s)
Antineoplastic Agents , Neoplasms , Humans , Fasting/physiology , Caloric Restriction , Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Amino Acids
7.
Int J Mol Sci ; 23(15)2022 Jul 22.
Article in English | MEDLINE | ID: mdl-35897655

ABSTRACT

A proteomic approach was used to characterize potential mediators involved in the improvement in cardiac fibrosis observed with the administration of the mitochondrial antioxidant MitoQ in obese rats. Male Wistar rats were fed a standard diet (3.5% fat; CT) or a high-fat diet (35% fat; HFD) and treated with vehicle or MitoQ (200 µM) in drinking water for 7 weeks. Obesity modulated the expression of 33 proteins as compared with controls of the more than 1000 proteins identified. These include proteins related to endoplasmic reticulum (ER) stress and oxidative stress. Proteomic analyses revealed that HFD animals presented with an increase in cardiac transthyretin (TTR) protein levels, an effect that was prevented by MitoQ treatment in obese animals. This was confirmed by plasma levels, which were associated with those of cardiac levels of both binding immunoglobulin protein (BiP), a marker of ER stress, and fibrosis. TTR stimulated collagen I production and BiP in cardiac fibroblasts. This upregulation was prevented by the presence of MitoQ. In summary, the results suggest a role of TTR in cardiac fibrosis development associated with obesity and the beneficial effects of treatment with mitochondrial antioxidants.


Subject(s)
Prealbumin , Ubiquinone , Animals , Antioxidants/metabolism , Antioxidants/pharmacology , Diet, High-Fat/adverse effects , Fibrosis , Male , Obesity/complications , Obesity/metabolism , Oxidative Stress , Prealbumin/metabolism , Proteomics , Rats , Rats, Wistar , Ubiquinone/metabolism , Ubiquinone/pharmacology
9.
Cell Death Dis ; 13(4): 356, 2022 04 18.
Article in English | MEDLINE | ID: mdl-35436993

ABSTRACT

Acyl-coenzyme-A-binding protein (ACBP), also known as a diazepam-binding inhibitor (DBI), is a potent stimulator of appetite and lipogenesis. Bioinformatic analyses combined with systematic screens revealed that peroxisome proliferator-activated receptor gamma (PPARγ) is the transcription factor that best explains the ACBP/DBI upregulation in metabolically active organs including the liver and adipose tissue. The PPARγ agonist rosiglitazone-induced ACBP/DBI upregulation, as well as weight gain, that could be prevented by knockout of Acbp/Dbi in mice. Moreover, liver-specific knockdown of Pparg prevented the high-fat diet (HFD)-induced upregulation of circulating ACBP/DBI levels and reduced body weight gain. Conversely, knockout of Acbp/Dbi prevented the HFD-induced upregulation of PPARγ. Notably, a single amino acid substitution (F77I) in the γ2 subunit of gamma-aminobutyric acid A receptor (GABAAR), which abolishes ACBP/DBI binding to this receptor, prevented the HFD-induced weight gain, as well as the HFD-induced upregulation of ACBP/DBI, GABAAR γ2, and PPARγ. Based on these results, we postulate the existence of an obesogenic feedforward loop relying on ACBP/DBI, GABAAR, and PPARγ. Interruption of this vicious cycle, at any level, indistinguishably mitigates HFD-induced weight gain, hepatosteatosis, and hyperglycemia.


Subject(s)
Diazepam Binding Inhibitor , Receptors, GABA-A , Animals , Carrier Proteins , Coenzyme A/metabolism , Diazepam Binding Inhibitor/genetics , Diazepam Binding Inhibitor/metabolism , Mice , PPAR gamma/genetics , PPAR gamma/metabolism , Receptors, GABA/metabolism , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Weight Gain , gamma-Aminobutyric Acid
10.
Int J Mol Sci ; 23(7)2022 Mar 23.
Article in English | MEDLINE | ID: mdl-35408859

ABSTRACT

Amyloidoses are characterized by the accumulation and aggregation of misfolded proteins into fibrils in different organs, leading to cell death and consequent organ dysfunction. The specific substitution of Leu 75 for Pro in Apolipoprotein A-I protein sequence (ApoA-I; L75P-ApoA-I) results in late onset amyloidosis, where deposition of extracellular protein aggregates damages the normal functions of the liver. In this work, we describe that the autophagic process is inhibited in the presence of the L75P-ApoA-I amyloidogenic variant in stably transfected human hepatocyte carcinoma cells. The L75P-ApoA-I amyloidogenic variant alters the redox status of the cells, resulting into excessive mitochondrial stress and consequent cell death. Moreover, L75P-ApoA-I induces an impairment of the autophagic flux. Pharmacological induction of autophagy or transfection-enforced overexpression of the pro-autophagic transcription factor EB (TFEB) restores proficient proteostasis and reduces oxidative stress in these experimental settings, suggesting that pharmacological stimulation of autophagy could be a promising target to alleviate ApoA-I amyloidosis.


Subject(s)
Amyloidosis , Immunoglobulin Light-chain Amyloidosis , Amyloidosis/genetics , Apolipoprotein A-I/genetics , Apolipoprotein A-I/metabolism , Autophagy/genetics , Humans , Protein Aggregates
11.
STAR Protoc ; 3(1): 101095, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35059656

ABSTRACT

Keyhole limpet hemocyanin (KLH) is a glycosylated multi-subunit metalloprotein that elicits a strong nonspecific immune activation, thus inducing both cellular and humoral immune responses. The exceptional immunogenicity of this protein can be leveraged to vaccinate mice against self-antigens that otherwise would not induce an autoimmune response. This protocol describes the covalent conjugation of KLH with acyl-coenzyme A-binding protein (ACBP), the autovaccination of mice with ACBP-KLH conjugate together with a potent adjuvant, and the detection of the produced anti-ACBP autoantibodies. For complete details on the use and execution of this profile, please refer to Bravo-San Pedro et al. (2019c).


Subject(s)
Carrier Proteins , Immunization , Animals , Hemocyanins , Mice , Peptides , Vaccination
12.
Cell Death Dis ; 12(11): 1039, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34725331

ABSTRACT

Pro-apoptotic multi-domain proteins of the BCL2 family such as BAX and BAK are well known for their important role in the induction of mitochondrial outer membrane permeabilization (MOMP), which is the rate-limiting step of the intrinsic pathway of apoptosis. Human or mouse cells lacking both BAX and BAK (due to a double knockout, DKO) are notoriously resistant to MOMP and cell death induction. Here we report the surprising finding that BAX/BAK DKO cells proliferate less than control cells expressing both BAX and BAK (or either BAX or BAK) when they are driven into tetraploidy by transient exposure to the microtubule inhibitor nocodazole. Mechanistically, in contrast to their BAX/BAK-sufficient controls, tetraploid DKO cells activate a senescent program, as indicated by the overexpression of several cyclin-dependent kinase inhibitors and the activation of ß-galactosidase. Moreover, DKO cells manifest alterations in ionomycin-mobilizable endoplasmic reticulum (ER) Ca2+ stores and store-operated Ca2+ entry that are affected by tetraploidization. DKO cells manifested reduced expression of endogenous sarcoplasmic/endoplasmic reticulum Ca2+ ATPase 2a (Serca2a) and transfection-enforced reintroduction of Serca2a, or reintroduction of an ER-targeted variant of BAK into DKO cells reestablished the same pattern of Ca2+ fluxes as observed in BAX/BAK-sufficient control cells. Serca2a reexpression and ER-targeted BAK also abolished the tetraploidy-induced senescence of DKO cells, placing ER Ca2+ fluxes downstream of the regulation of senescence by BAX/BAK. In conclusion, it appears that BAX/BAK prevent the induction of a tetraploidization-associated senescence program. Speculatively, this may contribute to the low incidence of cancers in BAX/BAK DKO mice and explain why human cancers rarely lose the expression of both BAX and BAK.


Subject(s)
Tetraploidy , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Calcium/metabolism , Calcium Signaling , Cell Line , Cellular Senescence , Clone Cells , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Humans , Mice, Inbred C57BL , Mice, Knockout , Microtubules/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , bcl-2 Homologous Antagonist-Killer Protein/deficiency , bcl-2-Associated X Protein/deficiency
13.
EMBO J ; 40(19): e108863, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34459017

ABSTRACT

Autophagy is a core molecular pathway for the preservation of cellular and organismal homeostasis. Pharmacological and genetic interventions impairing autophagy responses promote or aggravate disease in a plethora of experimental models. Consistently, mutations in autophagy-related processes cause severe human pathologies. Here, we review and discuss preclinical data linking autophagy dysfunction to the pathogenesis of major human disorders including cancer as well as cardiovascular, neurodegenerative, metabolic, pulmonary, renal, infectious, musculoskeletal, and ocular disorders.


Subject(s)
Autophagy , Disease Susceptibility , Animals , Autophagy/drug effects , Autophagy/genetics , Autophagy/immunology , Biomarkers , Gene Expression Regulation , Genetic Predisposition to Disease , Homeostasis , Host-Pathogen Interactions , Humans , Organ Specificity , Signal Transduction
14.
Methods Cell Biol ; 164: 39-46, 2021.
Article in English | MEDLINE | ID: mdl-34225917

ABSTRACT

The activation of autophagy has long been recognized as a central mechanism of healthspan and lifespan regulation at the organismal level, thus spurring major interest in identifying pharmacological or lifestyle interventions able to ignite the autophagic reaction in vivo. Consistently, there is growing need for the implementation in the preclinical practice of an "autophagometer," to be intended as a simple and non-invasive method to measure the autophagic flux in living organisms. Using fasting as the prototypical trigger of autophagy, we describe here a system (based on a leupeptin-based assay and video-flow cytometric detection of LC3B puncta) to quantitate autophagy in circulating leukocytes in mouse. We suggest that this method can be reliably used in the experimental routine to validate the pro-autophagy action of candidate drugs in vivo.


Subject(s)
Autophagy , Leukocytes , Animals , Flow Cytometry , Mice , Microtubule-Associated Proteins
15.
Methods Cell Biol ; 165: 111-122, 2021.
Article in English | MEDLINE | ID: mdl-34311860

ABSTRACT

Acyl-CoA binding protein (ACBP), also called diazepam-binding inhibitor (DBI), is a ubiquitous protein that can be secreted from cells by an unconventional pathway. Depending on its levels and on its subcellular localization, ACBP/DBI can regulate lipid metabolism. Several studies have shown that ACBP/DBI is secreted by an autophagy-dependent mechanism, positioning this catabolic pathway as the mechanism that controls lipid metabolism through the intracellular modulation of the levels of this protein. Autophagy is activated, among other stimuli, when cells have increased energy requirements; this causes a drop in the intracellular ACBP/DBI levels due to its release into the extracellular space and triggers an increase in the lipid catabolism. Conversely, when autophagy is inhibited, during pathological (obesity) or physiological (after-meal) situations, the intracellular levels of ACBP/DBI increase resulting in the activation of lipid anabolism, this effect has been demonstrated to be the link between obesity and autophagy inhibition. Here, we detail three different protocols for the detection of the ACBP/DBI levels by immunofluorescence, image flow cytometry or immunoblot techniques, which allow the quantification of ACBP/DBI levels and, indirectly, its autophagy-dependent release.


Subject(s)
Diazepam Binding Inhibitor , Obesity , Autophagy , Diazepam Binding Inhibitor/metabolism , Humans , Lipid Metabolism
16.
Methods Mol Biol ; 2267: 227-239, 2021.
Article in English | MEDLINE | ID: mdl-33786796

ABSTRACT

Mitotic catastrophe (MC) is a cell death modality induced by DNA damage that involves the activation of cell cycle checkpoints such as the "DNA structure checkpoint" and "spindle assembly checkpoint" (SAC) leading to aberrant mitosis. Depending on the signal, MC can drive the cell to death or to senescence. The suppression of MC favors aneuploidy. Several cancer therapies, included microtubular poisons and radiations, trigger MC. The clonogenic assay has been used to study the capacity of single cells to proliferate and to generate macroscopic colonies and to evaluate the efficacy of anticancer drugs. Nevertheless, this method cannot analyze MC events. Here, we report an improved technique based on the use of human colon cancer HCT116 stable expressing histone H2B-GFP and DsRed-centrin proteins, allowing to determine the capacity of cells to proliferate, and to determine changes in the nucleus and centrosomes.


Subject(s)
Cell Death , Cell Proliferation , Mitosis , Tumor Stem Cell Assay/methods , Antimitotic Agents/toxicity , Antineoplastic Agents/toxicity , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HCT116 Cells , Histones/genetics , Histones/metabolism , Humans
17.
Antioxidants (Basel) ; 10(1)2021 Jan 12.
Article in English | MEDLINE | ID: mdl-33445755

ABSTRACT

Reactive oxygen species (ROS) operate as key regulators of cellular homeostasis within a physiological range of concentrations, yet they turn into cytotoxic entities when their levels exceed a threshold limit. Accordingly, ROS are an important etiological cue for obesity, which in turn represents a major risk factor for multiple diseases, including diabetes, cardiovascular disorders, non-alcoholic fatty liver disease, and cancer. Therefore, the implementation of novel therapeutic strategies to improve the obese phenotype by targeting oxidative stress is of great interest for the scientific community. To this end, it is of high importance to shed light on the mechanisms through which cells curtail ROS production or limit their toxic effects, in order to harness them in anti-obesity therapy. In this review, we specifically discuss the role of autophagy in redox biology, focusing on its implication in the pathogenesis of obesity. Because autophagy is specifically triggered in response to redox imbalance as a quintessential cytoprotective mechanism, maneuvers based on the activation of autophagy hold promises of efficacy for the prevention and treatment of obesity and obesity-related morbidities.

18.
Adv Drug Deliv Rev ; 169: 40-50, 2021 02.
Article in English | MEDLINE | ID: mdl-33301821

ABSTRACT

Autophagy is quintessential for the maintenance of cellular homeostasis in all eukaryotic cells, explaining why both normal and malignant cells benefit from proficient autophagic responses. Moreover, autophagy is intimately involved in the immunological control of malignant transformation, tumor progression and response to therapy. However, the net effect of autophagy activation or inhibition on the natural growth or therapeutic response of tumors evolving in immunocompetent hosts exhibits a considerable degree of context dependency. Here, we discuss the complex cross-talk between autophagy and immuno-oncology as delineated by genetic and pharmacological approaches in mouse models of cancer.


Subject(s)
Autophagy/immunology , Neoplasms/immunology , Animals , Carcinogenesis , Disease Progression , Humans , Neoplasms/therapy
19.
Nat Commun ; 11(1): 2401, 2020 05 14.
Article in English | MEDLINE | ID: mdl-32409639

ABSTRACT

The molecular connections between homeostatic systems that maintain both genome integrity and proteostasis are poorly understood. Here we identify the selective activation of the unfolded protein response transducer IRE1α under genotoxic stress to modulate repair programs and sustain cell survival. DNA damage engages IRE1α signaling in the absence of an endoplasmic reticulum (ER) stress signature, leading to the exclusive activation of regulated IRE1α-dependent decay (RIDD) without activating its canonical output mediated by the transcription factor XBP1. IRE1α endoribonuclease activity controls the stability of mRNAs involved in the DNA damage response, impacting DNA repair, cell cycle arrest and apoptosis. The activation of the c-Abl kinase by DNA damage triggers the oligomerization of IRE1α to catalyze RIDD. The protective role of IRE1α under genotoxic stress is conserved in fly and mouse. Altogether, our results uncover an important intersection between the molecular pathways that sustain genome stability and proteostasis.


Subject(s)
Cell Survival/genetics , DNA Repair , Drosophila Proteins/metabolism , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , RNA Stability/genetics , Animals , DNA Damage , Drosophila Proteins/genetics , Drosophila melanogaster , Endoribonucleases/genetics , Female , Fibroblasts , Genomic Instability , HEK293 Cells , Humans , Mice , Mice, Knockout , Protein Multimerization , Protein Serine-Threonine Kinases/genetics , Proteostasis/genetics , Proto-Oncogene Proteins c-abl/metabolism , RNA, Messenger/metabolism
20.
Int J Cancer ; 146(1): 10-17, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31396957

ABSTRACT

In contrast to prior belief, cancer cells require oxidative phosphorylation (OXPHOS) to strive, and exacerbated OXPHOS dependency frequently characterizes cancer stem cells, as well as primary or acquired resistance against chemotherapy or tyrosine kinase inhibitors. A growing arsenal of therapeutic agents is being designed to suppress the transfer of mitochondria from stromal to malignant cells, to interfere with mitochondrial biogenesis, to directly inhibit respiratory chain complexes, or to disrupt mitochondrial function in other ways. For the experimental treatment of cancers, OXPHOS inhibitors can be advantageously combined with tyrosine kinase inhibitors, as well as with other strategies to inhibit glycolysis, thereby causing a lethal energy crisis. Unfortunately, most of the preclinical data arguing in favor of OXPHOS inhibition have been obtained in xenograft models, in which human cancer cells are implanted in immunodeficient mice. Future studies on OXPHOS inhibitors should elaborate optimal treatment schedules and combination regimens that stimulate-or at least are compatible with-anticancer immune responses for long-term tumor control.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Oxidative Phosphorylation , Protein Kinase Inhibitors/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis , Glycolysis , Humans , Mice , Neoplasms/enzymology , Neoplasms/metabolism , Organelle Biogenesis , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...