ArticlePDF Available

Targeting the Intracellular Environment in Cystic Fibrosis: Restoring Autophagy as a Novel Strategy to Circumvent the CFTR Defect

Frontiers
Frontiers in Pharmacology
Authors:
  • University of Naples Federico II,

Abstract and Figures

Cystic fibrosis (CF) patients harboring the most common deletion mutation of the CF transmembrane conductance regulator (CFTR), F508del, are poor responders to potentiators of CFTR channel activity which can be used to treat a small subset of CF patients who genetically carry plasma membrane (PM)-resident CFTR mutants. The misfolded F508del-CFTR protein is unstable in the PM even if rescued by pharmacological agents that prevent its intracellular retention and degradation. CF is a conformational disease in which defective CFTR induces an impressive derangement of general proteostasis resulting from disabled autophagy. In this review, we discuss how rescuing Beclin 1 (BECN1), a major player of autophagosome formation, either by means of direct gene transfer or indirectly by administration of proteostasis regulators, could stabilize F508del-CFTR at the PM. We focus on the relationship between the improvement of peripheral proteostasis and CFTR PM stability in F508del-CFTR homozygous bronchial epithelia or mouse lungs. Moreover, this article reviews recent pre-clinical evidence indicating that targeting the intracellular environment surrounding the misfolded mutant CFTR instead of protein itself could constitute an attractive therapeutic option to sensitize patients carrying the F508del-CFTR mutation to the beneficial action of CFTR potentiators on lung inflammation.
This content is subject to copyright.
A preview of the PDF is not available
... Emerging evidence indicates that CFTR does not merely function as an anion channel but that it also orchestrates the proteostasis network. Indeed, CFTR operates in a context-specific dynamic system of interactor proteins that is connected to, and influenced by, the proteostasis network [6][7][8]. Functional perturbation of CFTR can result from inherited loss-offunction mutations that cause CF [5] or pharmacological inhibition of CFTR channel activity [8]. In either case, CFTR inhibition leads to a major derangement of cellular proteostasis. ...
... TGM2 undergoes small ubiquitin like-modifier (SUMOylation), a post-translational modification that inhibits TGM2 ubiquitination leading to persistent high TGM2 protein levels and TGM2 activation as the result of permissive elevated Ca2+ levels [11]. Activated TGM2 targets a plethora of substrates, among which the essential autophagy protein Beclin 1 (BECN1) [6][7][8], that is essential for autophagosome formation [2]. BECN1 targeting by TGM2 causes its dislodgement, as well as that of several BECN1 interactors, away from the endoplasmic reticulum, leading to its functional sequestration in intracellular aggregates [6][7][8]. ...
... Activated TGM2 targets a plethora of substrates, among which the essential autophagy protein Beclin 1 (BECN1) [6][7][8], that is essential for autophagosome formation [2]. BECN1 targeting by TGM2 causes its dislodgement, as well as that of several BECN1 interactors, away from the endoplasmic reticulum, leading to its functional sequestration in intracellular aggregates [6][7][8]. Inactivation of the protein complex organized around BECN1 has two major negative effects on cellular proteostasis. First, the functional sequestration of phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3) and of UV radiation resistance-associatedgene (UVRAG), two major components of the BECN1 complex, negatively impacts on intracellular trafficking in CF epithelial cells, as it reduces the availability of phosphatidyl-inositol-3-phosphate (PtdIns3P) at early endosomes and perturbs endosomal fusion/maturation and trafficking [6,12,13]. ...
Article
Full-text available
Cystic Fibrosis (CF) is the most frequent lethal monogenetic disease affecting humans. CF is characterized by mutations in cystic fibrosis transmembrane conductance regulator (CFTR), a chloride channel whose malfunction triggers the activation of transglutaminase-2 (TGM2), as well as the inactivation of the Beclin-1 (BECN1) complex resulting in disabled autophagy. CFTR inhibition, TGM2 activation and BECN1 sequestration engage in an 'infernal trio' that locks the cell in a pro-inflammatory state through anti-homeostatic feedforward loops. Thus, stimulation of CFTR function, TGM2 inhibition and autophagy stimulation can be used to treat CF patients. Several studies indicate that patients with CF have a higher incidence of celiac disease (CD) and that mice bearing genetically determined CFTR defects are particularly sensitive to the enteropathogenic effects of the orally supplied gliadin (a gluten-derived protein). A gluten/gliadin-derived peptide (P31-43) inhibits CFTR in mouse intestinal epithelial cells, causing a local stress response that contributes to the immunopathology of CD. In particular, P31-43-induced CFTR inhibition elicits an epithelial stress response perturbing proteostasis. This event triggers TGM2 activation, BECN1 sequestration and results in molecular crosslinking of CFTR and P31-43 by TGM2. Importantly, stimulation of CFTR function with a pharmacological potentiator (Ivacaftor), which is approved for the treatment of CF, could attenuate the autophagy-inhibition and pro-inflammatory effects of gliadin in preclinical models of CD. Thus, CD shares with CF a common molecular mechanism involving CFTR inhibition that might respond to drugs that intercept the "infernal trio". Here, we highlight how drugs available for CF treatment could be repurposed for the therapy of CD.
... 1). При этом TG2 участвует в сшивании комплекса BECN1 / AMBRA1 [51,81]. Кроме того, известно, что данный фермент, имеющий предполагаемый LIR-мотив, который обеспечивает распознавание LC3 /ATG8, проявляет шаперонную активность, транс портируя грузы на мембрану фагофора [28]. ...
Article
In normal tissue, cellular homeostasis is largely driven by two catabolic pathways: apoptosis and autophagy. Apoptosis, or programmed cell death, is regulated by pro-apoptotic factors, and promotes the removal of problematic cells. Autophagy, which in turn includes three forms: macro-, micro-, and chaperone-mediated autophagy, can promote both cell survival by selectively removing potentially apoptosis-inducing factors and raising the threshold of stress required for the induction of cell death. Recently, evidence has been accumulating suggesting the existence of common molecular pathways between autophagy and apoptosis, as well as the influence of the extracellular matrix on these processes. One of the important enzymes involved in the coordination and regulation of these processes is transglutaminase 2 (TG2). Different types of TG2 activities are involved in maintaining the dynamic balance between extracellular matrix and intracellular autophagy/apoptosis processes, while dysregulation of these processes may contribute to the pathogenesis of various human diseases, including oncogenesis. For example, TG2 can promote the degradation of pro-apoptotic proteins and the survival of renal cell carcinoma cells under nutrient-deficient conditions by modulating the autophagy process. In cells of various tissues deprived of TG2, aggregates of ubiquitinated proteins and damaged mitochondria are observed, which in turn induces proteotoxic stress and cell death. conversely, the transamidase activity of TG2 was observed to inhibit anti-apoptotic signaling in a human leukemic monocytic lymphoma model. In the present review, a number of important functions of TG2 in oncogenesis are described, along with the dual role of TG2 in modulating such opposite processes as cell survival and cell death.
... Alternatively, autophagosomes found in ΔF cells may represent structures used by the cells to supply substrates and energy, a role that cannot be subverted by SARS-CoV-2 in these cells due to the well-known phenomenon of a CFassociated dysregulation of autophagy, due to impaired autophagosome-lysosome fusion and/or degradation [63]. Indeed, CFTR-defective cells are characterized by an increase in the production of reactive oxygen species, leading to an increased activation of transglutaminase-2 that, through inactivating the Beclin1 (BECN1) complex, results in a deficient autophagy process [64] due to sequestration of BECN1 and other interactors into intracellular aggregates [65][66][67]. Moreover, these autophagy-deficient cells showed accumulation of SQSTM1, a protein involved in the formation and elimination of aggregates containing ubiquitinated proteins [68,69]. ...
Article
Full-text available
SARS-CoV-2 replicates in host cell cytoplasm. People with cystic fibrosis, considered at risk of developing severe symptoms of COVID-19, instead, tend to show mild symptoms. We, thus, analyzed at the ultrastructural level the morphological effects of SARS-CoV-2 infection on wild-type (WT) and F508del (ΔF) CFTR-expressing CFBE41o- cells at early and late time points post infection. We also investigated ACE2 expression through immune-electron microscopy. At early times of infection, WT cells exhibited double-membrane vesicles, representing typical replicative structures, with granular and vesicular content, while at late time points, they contained vesicles with viral particles. ∆F cells exhibited double-membrane vesicles with an irregular shape and degenerative changes and at late time of infection, showed vesicles containing viruses lacking a regular structure and a well-organized distribution. ACE2 was expressed at the plasma membrane and present in the cytoplasm only at early times in WT, while it persisted even at late times of infection in ΔF cells. The autophagosome content also differed between the cells: in WT cells, it comprised vesicles associated with virus-containing structures, while in ΔF cells, it comprised ingested material for lysosomal digestion. Our data suggest that CFTR-modified cells infected with SARS-CoV-2 have impaired organization of normo-conformed replicative structures.
... Les flèches vertes représentent nos résultats tandis que les autres interactions représentées compilent des données issues de la littérature 408Par ailleurs, l'activation de mTORC1 est connue pour inhiber la voie de l'autophagie405,411 . Cette dernière est considérée comme une potentielle cible thérapeutique pour les patients atteints de mucoviscidose, dont le défaut de CFTR compromet l'autophagie notamment via la déplétion de la protéine pro-autophagique essentielle Beclin 1 (BECN1)[412][413][414][415] . Sachant que l'autophagie est défectueuse chez les patients et que certaines thérapies visent à l'induire, l'activation de SLC6A14 dans ce cas semble donc contre-indiquée puisqu'il aurait théoriquement tendance à l'inhiber. ...
Thesis
La mucoviscidose est due à des variants pathogènes du gène CFTR et ne dispose d’aucun traitement curatif à ce jour. La principale cause de mortalité des patients atteints de mucoviscidose est l’atteinte pulmonaire, caractérisée par des cycles d’infections et d’inflammation menant, à terme, à une insuffisance respiratoire. La sévérité de la maladie est variable selon les patients porteurs des mêmes variants de CFTR. Cela s’explique en partie par d’autres facteurs génétiques appelés « gènes modificateurs ». Dans le cadre de cette étude, nous nous sommes intéressés au gène SLC6A14, dont certains variants sont associés à une aggravation des atteintes respiratoires de la mucoviscidose. Nous avons pu démontrer que l’allèle G du variant rs3788766, localisé dans la séquence promotrice de SLC6A14, est associé à un déclin de la fonction respiratoire ainsi qu’à une diminution de l’activité du promoteur de SLC6A14. Nous avons montré que SLC6A14 est impliqué dans la voie de signalisation mTOR et favorise la réparation épithéliale bronchique. L’expression et la fonction de SLC6A14 sont modulées en réponse à la cytokine IL-1β et à l’infection par Pseudomonas aeruginosa, contextes inflammatoire et infectieux caractéristiques de la mucoviscidose. L’apport de nouvelles connaissances sur le gène modificateur SLC6A14, qui fait l’objet de recherches depuis peu, est essentiel à une meilleure compréhension de la maladie ainsi qu’au développement de nouvelles pistes thérapeutiques. En ce sens, ce travail apporte un nouvel éclairage sur l’impact du variant rs3788766 et sur les interactions hôte-pathogène existant au sein de l’épithélium bronchique des patients atteints de mucoviscidose.
... Recent work has shown that defective ∆F508-CFTR leads to small ubiquitin like-modifier (SUMO)ylation activation of tissue transglutaminase (TG2), resulting in proteasome degradation and accumulation of SQSTM1 [65]. Correspondingly, the depletion of SQSTM1 can favour the trafficking of ∆F508-CFTR protein to the epithelial cell surface [66], which parallels our findings here. ...
Article
Full-text available
Deletion of phenylalanine 508 (∆F508) of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) anion channel protein is the leading cause of Cystic Fibrosis (CF). Here, we report the analysis of CFTR and ∆F508-CFTR interactomes using BioID (proximity-dependent biotin identification), a technique that can also detect transient associations. We identified 474 high-confidence CFTR proximity-interactors, 57 of which have been previously validated, with the remainder representing novel interaction space. The ∆F508 interactome, comprising 626 proximity-interactors was markedly different from its wild type counterpart, with numerous alterations in protein associations categorized in membrane trafficking and cellular stress functions. Furthermore, analysis of the ∆F508 interactome in cells treated with Orkambi identified several interactions that were altered as a result of this drug therapy. We examined two candidate CFTR proximity interactors, VAPB and NOS1AP, in functional assays designed to assess surface delivery and overall chloride efflux. VAPB depletion impacted both CFTR surface delivery and chloride efflux, whereas NOS1AP depletion only affected the latter. The wild type and ∆F508-CFTR interactomes represent rich datasets that could be further mined to reveal additional candidates for the functional rescue of ∆F508-CFTR. View Full-Text
... Current Research in Cell Biology 1 (2020) 100003 activator of the phosphatidylinositol 3-kinase catalytic subunit type 3 (PIK3C3). Transamidation of BECN1 by TG2 causes autophagy inhibition and p62 accumulation, which in turn regulate aggresome formation (Villella & et al., 2013). This disruption of the autophagic clearance causes the augmentation of inflammatory responses in CFTR-mutant mice (Mayer & et al., 2013). ...
Article
Full-text available
Acute Respiratory Distress Syndrome is a severe disorder affecting thousands of individuals worldwide. The available medical countermeasures do not sufficiently suppress the unacceptable high mortality rates associated with those in need. Thus, intense efforts aim to delineate the function of the lung endothelium, so to deliver new therapeutic approaches against this disease. The present manuscript attempts to shed light on the interrelations between the unfolded protein response and autophagy towards lung disease, to deliver a new line of possible therapeutic approaches against the ferocious Acute Respiratory Distress Syndrome.
Article
Full-text available
SARS-CoV-2, the etiological cause of the COVID-19 pandemic, can cause severe illness in certain at-risk populations, including people with cystic fibrosis (pwCF). Nevertheless, several studies indicated that pwCF do not have higher risks of SARS-CoV-2 infection nor do they demonstrate worse clinical outcomes than those of the general population. Recent in vitro studies indicate cellular and molecular processes to be significant drivers in pwCF lower infection rates and milder symptoms than expected in cases of SARS-CoV-2 infection. These range from cytokine releases to biochemical alterations leading to morphological rearrangements inside the cells associated with CFTR impairment. Based on available data, the reported low incidence of SARS-CoV-2 infection among pwCF is likely a result of several variables linked to CFTR dysfunction, such as thick mucus, IL-6 reduction, altered ACE2 and TMPRSS2 processing and/or functioning, defective anions exchange, and autophagosome formation. An extensive analysis of the relation between SARS-CoV-2 infection and pwCF is essential to elucidate the mechanisms involved in this lower-than-expected infection impact and to possibly suggest potential new antiviral strategies.
Article
Full-text available
Several reports have indicated that SARS-CoV-2 infection displays unexpected mild clinical manifestations in people with cystic fibrosis (pwCF), suggesting that CFTR expression and function may be involved in the SARS-CoV-2 life cycle. To evaluate the possible association of CFTR activity with SARS-CoV-2 replication, we tested the antiviral activity of two well-known CFTR inhibitors (IOWH-032 and PPQ-102) in wild type (WT)-CFTR bronchial cells. SARS-CoV-2 replication was inhibited by IOWH-032 treatment, with an IC50 of 4.52 μM, and by PPQ-102, with an IC50 of 15.92 μM. We confirmed this antiviral effect on primary cells (MucilAirTM wt-CFTR) using 10 μM IOWH-032. According to our results, CFTR inhibition can effectively tackle SARS-CoV-2 infection, suggesting that CFTR expression and function might play an important role in SARS-CoV-2 replication, revealing new perspectives on the mechanisms governing SARS-CoV-2 infection in both normal and CF individuals, as well as leading to potential novel treatments.
Article
Structural lung disease begins very early in children with cystic fibrosis (CF), often in the first three months of life. Inhaled medications represent an attractive therapeutic approach in CF that are routinely used as early intervention strategies. Two aerosolized solutions, hypertonic saline and dornase alfa, have significant potential benefits by improving mucociliary clearance, with minimal associated side-effects. In particular, they favor rehydration of airway surface liquid and cleavage of extracellular DNA in the airways, respectively, consequently reducing rate of pulmonary disease exacerbations. Indirect anti-inflammatory effects have been documented for both drugs, addressing each of the three interrelated elements in the vicious cycle of lung disease in CF: airway obstruction, inflammation and infection. This short review aimed to summarize the main papers that support potential clinical impact of inhaled solutions on pulmonary disease in CF.
Article
High variability in the response rates to treatments can make the interpretation of data from clinical trials very difficult, particularly in rare genetic diseases in which the enrolment of thousands of patients is problematic. Personalized medicine largely depends on the establishment of appropriate early detectors of drug efficacy that may guide the administration (or discontinuation) of specific treatments. Such biomarkers should be capable of predicting the therapeutic response of individual patients and of monitoring early benefits of candidate drugs before late clinical benefits become evident. The identification of these biomarkers implies a rigorous stepwise process of translation from preclinical evaluation in cultured cells, suitable animal models or patient-derived freshly isolated cells to clinical application. In this review, we will discuss how a process of research translation can lead to the implementation of functional and mechanistic disease-relevant biomarkers. Moreover, we will address how preclinical data can be translated into the clinic in a personalized medical approach that can provide the right drug to the right patient within the right timeframe.
Article
Full-text available
Accumulation of unwanted/misfolded proteins in aggregates has been observed in airways of patients with cystic fibrosis (CF), a life-threatening genetic disorder caused by mutations in the gene encoding the cystic fibrosis transmembrane conductance regulator (CFTR). Here we show how the defective CFTR results in defective autophagy and decreases the clearance of aggresomes. Defective CFTR-induced upregulation of reactive oxygen species (ROS) and tissue transglutaminase (TG2) drive the crosslinking of beclin 1, leading to sequestration of phosphatidylinositol-3-kinase (PI(3)K) complex III and accumulation of p62, which regulates aggresome formation. Both CFTR knockdown and the overexpression of green fluorescent protein (GFP)-tagged-CFTR(F508del) induce beclin 1 downregulation and defective autophagy in non-CF airway epithelia through the ROS-TG2 pathway. Restoration of beclin 1 and autophagy by either beclin 1 overexpression, cystamine or antioxidants rescues the localization of the beclin 1 interactome to the endoplasmic reticulum and reverts the CF airway phenotype in vitro, in vivo in Scnn1b-transgenic and Cftr(F508del) homozygous mice, and in human CF nasal biopsies. Restoring beclin 1 or knocking down p62 rescued the trafficking of CFTR(F508del) to the cell surface. These data link the CFTR defect to autophagy deficiency, leading to the accumulation of protein aggregates and to lung inflammation.
Article
Full-text available
Beclin 1, a protein essential for autophagy, binds to hVps34/Class III phosphatidylinositol-3-kinase and UVRAG. Here, we have identified two Beclin 1 associated proteins, Atg14L and Rubicon. Atg14L and UVRAG bind to Beclin 1 in a mutually exclusive manner, whereas Rubicon binds only to a subpopulation of UVRAG complexes; thus, three different Beclin 1 complexes exist. GFP–Atg14L localized to the isolation membrane and autophagosome, as well as to the ER and unknown puncta. Knockout of Atg14L in mouse ES cells caused a defect in autophagosome formation. GFP–Rubicon was localized at the endosome/lysosome. Knockdown of Rubicon caused enhancement of autophagy, especially at the maturation step, as well as enhancement of endocytic trafficking. These data suggest that the Beclin 1–hVps34 complex functions in two different steps of autophagy by altering the subunit composition.
Article
Full-text available
Channel activators (potentiators) of cystic fibrosis (CF) transmembrane conductance regulator (CFTR), can be used for the treatment of the small subset of CF patients that carry plasma membrane-resident CFTR mutants. However, approximately 90% of CF patients carry the misfolded ΔF508-CFTR and are poorly responsive to potentiators, because ΔF508-CFTR is intrinsically unstable at the plasma membrane (PM) even if rescued by pharmacological correctors. We have demonstrated that human and mouse CF airways are autophagy deficient due to functional sequestration of BECN1 and that the tissue transglutaminase-2 inhibitor, cystamine, or antioxidants restore BECN1-dependent autophagy and reduce SQSTM1/p62 levels, thus favoring ΔF508-CFTR trafficking to the epithelial surface. Here, we investigated whether these treatments could facilitate the beneficial action of potentiators on ΔF508-CFTR homozygous airways. Cystamine or the superoxide dismutase (SOD)/catalase-mimetic EUK-134 stabilized ΔF508-CFTR at the plasma membrane of airway epithelial cells and sustained the expression of CFTR at the epithelial surface well beyond drug withdrawal, overexpressing BECN1 and depleting SQSTM1. This facilitates the beneficial action of potentiators in controlling inflammation in ex vivo ΔF508-CFTR homozygous human nasal biopsies and in vivo in mouse ΔF508-CFTR lungs. Direct depletion of Sqstm1 by shRNAs in vivo in ΔF508-CFTR mice synergized with potentiators in sustaining surface CFTR expression and suppressing inflammation. Cystamine pre-treatment restored ΔF508-CFTR response to the CFTR potentiators genistein, Vrx-532 or Vrx-770 in freshly isolated brushed nasal epithelial cells from ΔF508-CFTR homozygous patients. These findings delineate a novel therapeutic strategy for the treatment of CF patients with the ΔF508-CFTR mutation in which patients are first treated with cystamine and subsequently pulsed with CFTR potentiators.
Article
Full-text available
Cystic fibrosis is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene that lead to abnormalities in transepithelial ion transport in the airways of affected patients. Lung disease is the major contributor to morbidity and mortality in patients with cystic fibrosis but recommended therapeutic interventions so far have focussed on symptom control rather than treatment of the underlying causes of the disease. New therapies that are currently in pre-clinical and clinical testing include CFTR pharmacotherapy, drugs targeting other ion channels, and hydrators of the cystic fibrosis airways. The current status of these and other new developments in the treatment of cystic fibrosis are reviewed.
Article
Full-text available
The autophagosome is the central organelle in macroautophagy, a vacuolar lysosomal catabolic pathway that degrades cytoplasmic material to fuel starving cells and eliminates intracellular pathogens. Macroautophagy has important physiological roles during development, ageing and the immune response, and its cytoprotective function is compromised in various diseases. A set of autophagy-related (ATG) proteins is hierarchically recruited to the phagophore, the initial membrane template in the construction of the autophagosome. However, recent findings suggest that macroautophagy can also occur in the absence of some of these key autophagy proteins, through the unconventional biogenesis of canonical autophagosomes. Such alternatives to the evolutionarily conserved scheme might provide additional therapeutic opportunities.
Article
Full-text available
Since the cloning of the cystic fibrosis transmembrane regulator (CFTR) protein in the late 1980s and subsequent significant advances in understanding the pathophysiology of cystic fibrosis (CF) there have been high expectations that transformative, disease-modifying therapies could be developed.1 2 Dysfunction of the CFTR protein results in significant reduction or absence of chloride transport and dysregulation of sodium transport.3–5 Other cationic ions such as bicarbonate may also be transported by CFTR and the interaction with the epithelial sodium channel ENaC still remains to be elucidated.6 7 However, a significant number of in vitro and animal studies strongly support the premise that correction of CFTR function is possible and will make a significant impact on the CF phenotype, particularly in the lung, which accounts for most of the morbidity and mortality from this condition.7 Two approaches to correction of CFTR function are currently being pursued: one based on gene transfer therapy and the other on small molecule modulators. Gene therapy attempts to correct CFTR dysfunction in CF by transfection of airway cells with the wild-type CFTR gene to enable cells to express functional CFTR.8 Progress with this approach has been slow and methodical. Early studies suggest that it is possible to transfect nasal and bronchial epithelial cells and this results in a modest electrophysiological improvement.9 10 To date, there are no clinical studies in humans to determine if sufficient gene transfer can be achieved to result in a clinically meaningful benefit. A two-centre, double-blind, placebo-controlled trial of CF gene therapy is due to commence in the UK in 2012 using a specifically designed plasmid and a GL67 liposome complex. The results of this proof of concept study will help determine if gene therapy has the potential to provide a treatment which will benefit all mutation …
Article
Autophagy is a homeostatic process common to all eukaryotic cells that serves to degrade intracellular components. Amongst three classes of autophagy, macroautophagy is best understood and will be the subject of this review. The function of autophagy is multifaceted and includes removal of long-lived proteins and damaged or unneeded organelles, recycling of intracellular components for nutrients and defense against pathogens. This process has been extensively studied in yeast, and understanding of its functional significance in human disease is increasing as well. This review will explore the basic machinery and regulation of autophagy in mammalian systems, methods employed to measure autophagic activity, and then focus on recent discoveries about the functional significance of autophagy in respiratory diseases, including chronic obstructive pulmonary disease, cystic fibrosis, tuberculosis, idiopathic pulmonary fibrosis, pulmonary arterial hypertension, acute lung injury and lymphangioleiomyomatosis.
Article
Transglutaminase 2 (TG2 or tissue transglutaminase) is a highly complex multifunctional protein that acts as transglutaminase, GTPase/ATPase, protein disulfide isomerase, and protein kinase. Moreover, TG2 has many well-documented nonenzymatic functions that are based on its noncovalent interactions with multiple cellular proteins. A vast array of biochemical activities of TG2 accounts for its involvement in a variety of cellular processes, including adhesion, migration, growth, survival, apoptosis, differentiation, and extracellular matrix organization. In turn, the impact of TG2 on these processes implicates this protein in various physiological responses and pathological states, contributing to wound healing, inflammation, autoimmunity, neurodegeneration, vascular remodeling, tumor growth and metastasis, and tissue fibrosis. TG2 is ubiquitously expressed and is particularly abundant in endothelial cells, fibroblasts, osteoblasts, monocytes/macrophages, and smooth muscle cells. The protein is localized in multiple cellular compartments, including the nucleus, cytosol, mitochondria, endolysosomes, plasma membrane, and cell surface and extracellular matrix, where Ca(2+), nucleotides, nitric oxide, reactive oxygen species, membrane lipids, and distinct protein-protein interactions in the local microenvironment jointly regulate its activities. In this review, we discuss the complex biochemical activities and molecular interactions of TG2 in the context of diverse subcellular compartments and evaluate its wide ranging and cell type-specific biological functions and their regulation.
Article
Cystic fibrosis (CF), the most common lethal genetic disease in the Caucasian population, is caused by loss-of-function mutations of the CF transmembrane conductance regulator (CFTR), a cyclic AMP-regulated plasma membrane chloride channel. The most common mutation, deletion of phenylalanine 508 (ΔF508), impairs CFTR folding and, consequently, its biosynthetic and endocytic processing as well as chloride channel function. Pharmacological treatments may target the ΔF508 CFTR structural defect directly by binding to the mutant protein and/or indirectly by altering cellular protein homeostasis (proteostasis) to promote ΔF508 CFTR plasma membrane targeting and stability. This review discusses recent basic research aimed at elucidating the structural and trafficking defects of ΔF508 CFTR, a prerequisite for the rational design of CF therapy to correct the loss-of-function phenotype.