Paraoxonase-1 and Chemokine Ligand 2: History
Please note this is an old version of this entry, which may differ significantly from the current revision.
Subjects: Allergy
Contributor:

Infectious and many non-infectious diseases share common molecular mechanisms. Among them, oxidative stress and the subsequent inflammatory reaction are of particular note. Metabolic disorders induced by external agents, be they bacterial or viral pathogens, excessive calorie intake, poor-quality nutrients, or environmental factors produce an imbalance between the production of free radicals and endogenous antioxidant systems; the consequence being the oxidation of lipids, proteins, and nucleic acids. Oxidation and inflammation are closely related, and whether oxidative stress and inflammation represent the causes or consequences of cellular pathology, both produce metabolic alterations that influence the pathogenesis of the disease. In this entry, authors highlight two key molecules in the regulation of these processes: Paraoxonase-1 (PON1) and chemokine (C-C motif) ligand 2 (CCL2). 

  • cancer
  • cardiovascular disease
  • chemokine (C-C motif) ligand 2
  • fatty liver
  • infection
  • inflammation
  • metabolism
  • obesity
  • paraoxonase-1

1. Oxidation, Inflammation and Disease

Tissues produce reactive oxygen species (ROS) as a metabolic by-product in response to environmental factors such as an imbalanced diet or an infectious process. ROS react with lipids, proteins, and nucleic acids and result in alterations in cell structure and function [1]. The organism has enzymatic and non-enzymatic antioxidants to block the harmful effects of ROS. However, these protective systems can be overwhelmed in disease states. An inflammatory reaction is generated when oxidative equilibrium is disrupted. For example, in infectious diseases, ROS production by host macrophages is an important part of the defense mechanism against infecting bacteria or viruses, and the imbalances produced can trigger an inflammatory reaction [2]. In turn, inflammation can lead to a further increase in oxidative stress and thus enter a vicious cycle that can aggravate the disease [3][4]. Independently of whether oxidative stress and inflammation represent the causes or the consequences of cellular alterations, an overwhelming amount of evidence indicates that both processes contribute to the pathogenesis of many diseases. The role played by macrophages and their polarization need to be considered in detail in the setting of chronic inflammatory diseases [5]. These diseases are associated with an increase in M1, or “classically activated” macrophages, and a decrease in M2, or “alternatively activated” macrophages [6][7][8]. Chemokines are involved in macrophage polarization, while directing the traffic of immune cells to sites of inflammation and activating the production and secretion of inflammatory cytokines [9]. Monocytes migrate to the site of inflammation and differentiate into macrophages when the chemokine (C-C motif) receptor (CCR2) interacts with the chemokine (C-C motif) ligand 2 (CCL2): a key process in the development of inflammatory diseases [10]. Recent studies indicate that other chemokines and chemokine receptors also play important roles. For example, the CCL5/CCR5 complex has been related to cancer and infection [11][12][13][14].

2. The Protective Role of Paraoxonases on CCL2 Expression, Mitochondrial Function, and Metabolism

Several lines of research suggest that the PON family of enzymes play a prominent role in the protection of cells against mitochondrial dysfunction and metabolic alterations. Most studies in this field have been conducted on PON2 because the general consensus is that this is the major intracellular enzyme. For example, PON2 has been reported to reduce the unfolded protein response (UPR) accompanying oxidative stress and UPR-derived caspase activation in human vascular cells [15][16][17]. In addition, the expression of genes related to endoplasmic reticulum stress was increased in macrophages from apolipoprotein E and PON2 double deficient mice, compared to those that were only apolipoprotein E -deficient [18]. These authors observed that treatment of macrophages from apolipoprotein E and PON2 double deficient mice with an inducer of endoplasmic reticulum stress resulted in mitochondrial dysfunction, increased oxidative stress, and increased cell apoptosis. Further studies in several experimental models have added more evidence that PON2 protects mitochondrial function and prevents apoptosis [19][20][21].
From the above-mentioned data, it is inferred that PON2 does indeed play a protective role in mitochondrial function, but this is not so clear regarding PON1. Data suggest that this enzyme also protects cells from oxidation, and that this effect involves the inhibition of CCL2 synthesis. Studies from our research group have shown that PON1 deficient mice fed an atherogenic diet had increased hepatic fat depots and a marked depression of the tricarboxylic acid (TCA) cycle. In addition, the hepatic concentrations of several markers of oxidative stress and CCL2 expression were increased [22]. Further experimental data [23] showed that dietary fat caused liver steatosis, oxidative stress, and the accumulation of pro-inflammatory macrophages in the livers of LDL-receptor and PON1 double deficient mice, together with alterations in energy metabolism, in the methionine cycle, in the glutathione reduction pathway, and autophagy. Conversely, when we established a line of LDL-receptor, PON1, and CCL2 triple deficient mice, we observed that the deletion of this chemokine normalized the metabolic disturbances and increased lysosome-associated membrane protein 2 expression, which suggests enhanced chaperone-mediated autophagy. In humans, studies have observed that individuals with obesity have impaired PON1 activity and impaired mitochondrial function [24][25]. Our group has a special interest in evaluating the hepatic alterations in patients with morbid obesity treated with bariatric surgery and, as well, in observing the metabolic effects of the treatment. To date, results have shown that one-year post-surgery, the hepatic histology of all patients was improved, especially in those who had severe steatohepatitis, bridging fibrosis, and/or cirrhosis. Additionally, we observed pre-surgery differences in plasma and liver markers of oxidative stress and inflammation (including CCL2 and PON1), which were corrected one-year post-surgery [26]. In addition, patients with steatohepatitis presented pre-surgery alterations in energy metabolism, especially in plasma concentrations of α-ketoglutarate and oxaloacetate, which reverted one-year post-surgery [27]. Overall, these results suggest an entanglement of PON1 and CCL2 in the regulation of metabolism and mitochondrial function in the liver of experimental animals, and in humans (Figure 1).
Figure 1. Oxidation, inflammation, and disturbances in energy metabolism are closely related. To date, the evidence reported suggests that excessive production of reactive oxygen species (ROS) would inhibit paraoxonase-1 (PON1) activity in high-density lipoprotein (HDL) particles and in the mitochondrial membranes of somatic cells. At the same time, it would stimulate the synthesis of chemokine (C-C motif) ligand 2 (CCL2) through several pathways, notably that of pathogen-associated molecular patterns/damage-associated molecular patterns/pattern-recognition receptors (PAMP/DAMP/PRR). The decrease in PON1 activity and the increase in CCl2 would cause alterations in mitochondrial metabolism and an inhibition of autophagy. At the same time, CCL2 would interact with its receptor (CRR2) and present on monocytes, promoting their migration to sites of injury, their differentiation to macrophages, and their synthesis of new ROS, producing a vicious circle that would trigger and aggravate the disease.

3. Mechanism of Action of CCL2 in the Immune Response and Inflammation and Its Relationship with Multiple Metabolic Alterations

One of the proposed mechanisms by which oxidative stress would enhance the inflammatory response is the induction and assembly of multiprotein complexes called inflammasomes. ROS activate the NOD-like receptor Pyrin domain 3 (NLRP3) in macrophages, triggering the formation of inflammasomes and an immune reaction that involves the synthesis of pro-inflammatory chemokines, from which CCL2 is, probably, the most representative [28][29][30][31]. This chemokine is upregulated following tissue injury and is expressed by both inflammatory and stromal cells. CCL2 has been reported to promote endoplasmic reticulum (ER) stress and autophagy and to regulate NF-ĸB expression by catalyzing de-ubiquitination [32]. The main pathway triggering inflammation is, probably, the activation of pattern-recognition receptors (PRRs), which recognize pathogen-associated molecular patterns (PAMPs), synthesized as a response to pathogens, or damage-associated molecular patterns (DAMPs), which are products of damaged cells [33][34][35]. The binding of PAMP/DAMP to a PRR leads to NF-ĸB activation and the production of adhesion molecules and chemokines that lead to infiltration of immune cells into the sites of tissue damage [36]. Other alternative pathways also result in similar outcomes, particularly the phosphoinositide 3-kinase-related signaling pathway, the mitogen-activated protein kinase pathway, and the Janus kinase/signal transducers and activators of transcription signaling pathway [37][38][39]. These changes induce the UPR, essentially by three ER-related transmembrane proteins, i.e., the inositol-requiring enzyme 1, the protein kinase RNA-like endoplasmic reticulum kinase, and the activating transcription factor 6 [40][41][42][43]. CCL2 and other chemokines, together with oxidative stress, trigger ER stress. In addition, the UPR may regulate inflammation through several pathways, such as the regulation of oxidative stress or the upregulation of CCR2 expression [44]; the UPR links ER stress with cell death and autophagy [45]. When cell damage is moderate, autophagy helps cells survive the injury, allowing them to heal and thus preventing cell death by removing toxic protein aggregates. However, when cell damage is high, the result is a non-apoptotic form of cell death that can be detrimental. The role of autophagy in the maintenance of mitochondrial integrity seems to be paramount [46]. Mitophagy increases cell lifespan, while repression of autophagy reduces lifespan. Several studies have linked mitochondrial dysfunction, autophagy, and age-related diseases with the activity of the inflammasomes [47][48][49][50]. Taken together, these results define a clear relationship between oxidative stress, chemokines, and mitochondrial impairment, resulting in metabolic alterations and their involvement in diseases.
Activation of the immune response and chronic inflammation has been associated with aging and age-related diseases [51][52][53]. Senescent cells secrete chemokines, which influence the trafficking of immune cells [54][55]. Epidemiological studies have suggested that CCL2 levels are increased in older individuals, independently of metabolic alterations. Moreover, in vitro studies have shown that chemokines appear to confer senescence to neighboring normal cells in an autocrine and paracrine fashion [56][57][58]. A recent study by our research group in mice with accelerated aging is a good example of such relationships [59]. We crossbred mice that overexpressed CCL2 with progeroid mice bearing a mutation in the lamin A gene. Wild-type animals and progeroid mice not overexpressing CCL2 were used as controls. We observed that progeroid mice lost weight (relative to the wild-type animals) and developed lordokyphosis and lipodystrophy. The lifespan was significantly reduced in both strains of progeroid mice, but this reduction was higher in those overexpressing CCL2. These mice also presented specific characteristics of metabolic dysregulation in skeletal muscle, including alterations in the glucose and TCA cycles, and in one-carbon metabolism. These data suggest that mitochondrial metabolites play major roles in pathological aging. Consequently, we investigated the mitochondrial respiratory complexes in skeletal muscle, and observed that the expressions of complexes I and V were lower in mice overexpressing CCL2. In addition, the protein concentrations of translocase of outer membrane 20 (TOM20) and mitofusin 2 (MFN2) in the muscles of the progeroid mice were decreased, indicating alterations in the correct formation of the mitochondrial network. We also observed an increase in p53, which would indicate the triggering of aging through a p53-mediated transcriptional program involving the mechanistic target of rapamycin. Indeed, we found inhibition of phosphorylation of phosphoinositide 3-kinase, indicating a mechanistic target of rapamycin inhibition. Finally, the higher microtubule-associated proteins 1A/1B light chain 3B (LC3) II/I ratio, and lower lysosome-associated membrane protein 2 (LAMP2A) and sequestosome 1 (p62) expressions suggested the involvement of chaperone-mediated autophagy as a consequence of the CCL2 overexpression in the progeroid animals.
Following on from the actions described above, in many infectious and non-infectious diseases, the adaptive immunity deteriorates, whereas the innate immunity is more responsive to stimuli; the consequence is the development of an inflammatory reaction. Several studies have linked the activation of NLRP3, which is dependent on increased generation of free radical species by mitochondria, with metabolic disturbances [60][61][62]. Hence, it is of note that awareness of the origin of free radicals and the putative mechanisms of prevention (i.e., PON1) is critical when establishing possible therapeutic interventions in order to preempt an inflammatory reaction. Within this context, the interaction of PON1 with CCL2 can play a key role.

This entry is adapted from the peer-reviewed paper 10.3390/biom11070971

References

  1. Pizzino, G.; Irrera, N.; Cucinotta, M.; Pallio, G.; Mannino, F.; Arcoraci, V.; Squadrito, F.; Altavilla, D.; Bitto, A. Oxidative stress: Harms and benefits for human health. Oxid. Med. Cell. Longev. 2017, 2017, 8416763.
  2. Masud, S.; Torraca, V.; Meijer, A.H. Modeling infectious diseases in the context of a developing immune system. Curr. Top. Dev. Biol. 2017, 124, 277–329.
  3. He, J.; Liu, J.; Huang, Y.; Tang, X.; Xiao, H.; Hu, Z. Oxidative Stress, inflammation, and autophagy: Potential targets of mesenchymal stem cells-based therapies in ischemic stroke. Front. Neurosci. 2021, 15, 641157.
  4. Kibel, A.; Lukinac, A.M.; Dambic, V.; Juric, I.; Relatic, K.S. Oxidative stress in ischemic heart disease. Oxid. Med. Cell. Longev. 2020, 2020, 6627144.
  5. Atri, C.; Guerfali, F.Z.; Laouini, D. Role of human macrophage polarization in inflammation during infectious diseases. Int. J. Mol. Sci. 2018, 19, 1801.
  6. Parisi, L.; Gini, E.; Baci, D.; Tremolati, M.; Fanuli, M.; Bassani, B.; Farronato, G.; Bruno, A.; Mortara, L. Macrophage polarization in chronic inflammatory diseases: Killers or builders? J. Immunol. Res. 2018, 2018, 8917804.
  7. Poltavets, A.S.; Vishnyakova, P.A.; Elchaninov, A.V.; Sukhikh, G.T.; Fatkhudinov, T.K. Macrophage modification strategies for efficient cell therapy. Cells 2020, 9, 1535.
  8. Viola, A.; Munari, F.; Sánchez-Rodríguez, R.; Scolaro, T.; Castegna, A. The metabolic signature of macrophage responses. Front. Immunol. 2019, 10, 1462.
  9. Turner, M.D.; Nedjai, B.; Hurst, T.; Pennington, D.J. Cytokines and chemokines: At the crossroads of cell signalling and inflammatory disease. Biochim. Biophys. Acta 2014, 1843, 2563–2582.
  10. Gschwandtner, M.; Derler, R.; Midwood, K.S. More than just attractive: How CCL2 influences myeloid cell behavior beyond chemotaxis. Front. Immunol. 2019, 10, 2759.
  11. Huang, R.; Guo, L.; Gao, M.; Li, J.; Xiang, S. Research trends and regulation of CCL5 in prostate cancer. Onco. Targets Ther. 2021, 14, 1417–1427.
  12. Agresti, N.; Lalezari, J.P.; Amodeo, P.P.; Mody, K.; Mosher, S.F.; Seethamraju, H.; Kelly, S.A.; Pourhassan, N.Z.; Sudduth, C.D.; Bovinet, C.; et al. Disruption of CCR5 signaling to treat COVID-19-associated cytokine storm: Case series of four critically ill patients treated with leronlimab. J. Transl. Autoimmun. 2021, 4, 100083.
  13. Yao, X.; Matosevic, S. Chemokine networks modulating natural killer cell trafficking to solid tumors. Cytokine Growth Factor Rev. 2021, 36–45.
  14. Necula, D.; Riviere-Cazaux, C.; Shen, Y.; Zhou, M. Insight into the roles of CCR5 in learning and memory in normal and disordered states. Brain Behav. Immun. 2021, 92, 1–9.
  15. Manco, G.; Porzio, E.; Carusone, T.M. Human paraoxonase-2 (PON2): Protein functions and modulation. Antioxidants 2021, 10, 256.
  16. Horke, S.; Witte, I.; Wilgenbus, P.; Krüger, M.; Strand, D.; Förstermann, U. Paraoxonase-2 reduces oxidative stress in vascular cells and decreases endoplasmic reticulum stress-induced caspase activation. Circulation 2007, 115, 2055–2064.
  17. Horke, S.; Witte, I.; Wilgenbus, P.; Altenhöfer, S.; Krüger, M.; Li, H.; Förstermann, U. Protective effect of paraoxonase-2 against endoplasmic reticulum stress-induced apoptosis is lost upon disturbance of calcium homoeostasis. Biochem. J. 2008, 416, 395–405.
  18. Devarajan, A.; Grijalva, V.R.; Bourquard, N.; Meriwether, D.; Imaizumi, S.; Shin, B.C.; Devaskar, S.U.; Reddy, S.T. Macrophage paraoxonase 2 regulates calcium homeostasis and cell survival under endoplasmic reticulum stress conditions and is sufficient to prevent the development of aggravated atherosclerosis in paraoxonase 2 deficiency/apoE-/- mice on a Western diet. Mol. Genet. Metab. 2012, 107, 416–427.
  19. Sulaiman, D.; Li, J.; Devarajan, A.; Cunningham, C.M.; Li, M.; Fishbein, G.A.; Fogelman, A.M.; Eghbali, M.; Reddy, S.T. Paraoxonase 2 protects against acute myocardial ischemia-reperfusion injury by modulating mitochondrial function and oxidative stress via the PI3K/Akt/GSK-3β RISK pathway. J. Mol. Cell. Cardiol. 2019, 129, 154–164.
  20. Tao, S.; Niu, L.; Cai, L.; Geng, Y.; Hua, C.; Ni, Y.; Zhao, R. N-(3-oxododecanoyl)-l-homoserine lactone modulates mitochondrial function and suppresses proliferation in intestinal goblet cells. Life Sci. 2018, 201, 81–88.
  21. Tao, S.; Luo, Y.; Bin, H.; Liu, J.; Qian, X.; Ni, Y.; Zhao, R. Paraoxonase 2 modulates a proapoptotic function in LS174T cells in response to quorum sensing molecule N-(3-oxododecanoyl)-L-homoserine lactone. Sci. Rep. 2016, 6, 28778.
  22. García-Heredia, A.; Kensicki, E.; Mohney, R.P.; Rull, A.; Triguero, I.; Marsillach, J.; Tormos, C.; Mackness, B.; Mackness, M.; Shih, D.M.; et al. Paraoxonase-1 deficiency is associated with severe liver steatosis in mice fed a high-fat high-cholesterol diet: A metabolomic approach. J. Proteome Res. 2013, 12, 1946–1955.
  23. Luciano-Mateo, F.; Cabré, N.; Fernández-Arroyo, S.; Baiges-Gaya, G.; Hernández- Aguilera, A.; Rodríguez-Tomàs, E.; Mercado-Gómez, M.; Menendez, J.A.; Camps, J.; Joven, J. Chemokine (C-C motif) ligand 2 gene ablation protects low-density lipoprotein and paraoxonase-1 double deficient mice from liver injury, oxidative stress and inflammation. Biochim. Biophys. Acta Mold Basis Dis. 2019, 1865, 1555–1566.
  24. Meneses, M.J.; Silvestre, R.; Sousa-Lima, I.; Macedo, M.P. Paraoxonase-1 as a regulator of glucose and lipid homeostasis: Impact on the onset and progression of metabolic disorders. Int. J. Mol. Sci. 2019, 20, 4049.
  25. Calvo, N.; Beltrán-Debón, R.; Rodríguez-Gallego, E.; Hernández-Aguilera, A.; Guirro, M.; Mariné-Casadó, R.; Millá, L.; Alegret, J.M.; Sabench, F.; del Castillo, D.; et al. Liver fat deposition and mitochondrial dysfunction in morbid obesity: An approach combining metabolomics with liver imaging and histology. World J. Gastroenterol. 2015, 21, 7529–7544.
  26. Cabré, N.; Luciano-Mateo, F.; Fernández-Arroyo, S.; Baiges-Gayà, G.; Hernández-Aguilera, A.; Fibla, M.; Fernández-Julià, R.; París, M.; Sabench, F.; Castillo, D.D.; et al. Laparoscopic sleeve gastrectomy reverses non-alcoholic fatty liver disease modulating oxidative stress and inflammation. Metabolism 2019, 99, 81–89.
  27. Cabré, N.; Luciano-Mateo, F.; Baiges-Gayà, G.; Fernández-Arroyo, S.; Rodríguez-Tomàs, E.; Hernández-Aguilera, A.; París, M.; Sabench, F.; Del Castillo, D.; López-Miranda, J.; et al. Plasma metabolic alterations in patients with severe obesity and non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 2020, 51, 374–387.
  28. Kurihara, C.; Lecuona, E.; Wu, Q.; Yang, W.; Núñez-Santana, F.L.; Akbarpour, M.; Liu, X.; Ren, Z.; Li, W.; Querrey, M.; et al. Crosstalk between nonclassical monocytes and alveolar macrophages mediates transplant ischemia-reperfusion injury through classical monocyte recruitment. JCI Insight 2021, 6, 147282.
  29. Mohammed, S.; Nicklas, E.H.; Thadathil, N.; Selvarani, R.; Royce, G.H.; Kinter, M.; Richardson, A.; Deepa, S.S. Role of necroptosis in chronic hepatic inflammation and fibrosis in a mouse model of increased oxidative stress. Free Radic. Biol. Med. 2021, 164, 315–328.
  30. Zhu, F.; Willette-Brown, J.; Zhang, J.; Ferre, E.M.N.; Sun, Z.; Wu, X.; Lionakis, M.S.; Hu, Y. NLRP3 inhibition ameliorates severe cutaneous autoimmune manifestations in a mouse model of autoimmune polyendocrinopathy-candidiasis-ectodermal dystrophy-like disease. J. Investig. Dermatol. 2020, 141, 1404–1415.
  31. McKnight, A.H.; Katzenberger, D.R.; Britnell, S.R. Colchicine in acute coronary syndrome: A systematic review. Ann. Pharmacother. 2021, 55, 187–197.
  32. Kolattukudy, P.E.; Niu, J. Inflammation, endoplasmic reticulum stress, autophagy, and the monocyte chemoattractant protein-1/CCR2 pathway. Circ. Res. 2012, 110, 174–189.
  33. Pandey, E.; Nour, A.S.; Harris, E.N. Prominent receptors of liver sinusoidal endothelial cells in liver homeostasis and disease. Front. Physiol. 2020, 11, 873.
  34. Andersson, U.; Ottestad, W.; Tracey, K.J. Extracellular HMGB1: A therapeutic target in severe pulmonary inflammation including COVID-19? Mol. Med. 2020, 26, 42.
  35. Shamilov, R.; Ackley, T.W.; Aneskievich, B.J. Enhanced wound healing- and inflammasome-associated gene expression in TNFAIP3-interacting protein 1-(TNIP1-) deficient HaCaT keratinocytes parallels reduced reepithelialization. Mediat. Inflamm. 2020, 2020, 5919150.
  36. Relja, B.; Land, W.G. Damage-associated molecular patterns in trauma. Eur. J. Trauma. Emerg. Surg. 2020, 46, 751–775.
  37. Afrose, S.S.; Junaid, M.; Akter, Y.; Tania, M.; Zheng, M.; Khan, M.A. Targeting kinases with thymoquinone: A molecular approach to cancer therapeutics. Drug Discov. Today. 2020, 25, 2294–2306.
  38. Dantonio, P.M.; Klein, M.O.; Freire, M.R.V.B.; Araujo, C.N.; Chiacetti, A.C.; Correa, R.G. Exploring major signaling cascades in melanomagenesis: A rationale route for targetted skin cancer therapy. Biosci. Rep. 2018, 38, BSR20180511.
  39. Stawski, L.; Trojanowska, M. Oncostatin M and its role in fibrosis. Connect. Tissue Res. 2019, 60, 40–49.
  40. Slaine, P.D.; Kleer, M.; Duguay, B.A.; Pringle, E.S.; Kadijk, E.; Ying, S.; Balgi, A.; Roberge, M.; McCormick, C.; Khaperskyy, D.A. Thiopurines activate an antiviral unfolded protein response that blocks influenza A virus glycoprotein accumulation. J. Virol. 2021, 95, e00453-21.
  41. Féral, K.; Jaud, M.; Philippe, C.; Di Bella, D.; Pyronnet, S.; Rouault-Pierre, K.; Mazzolini, L.; Touriol, C. ER Stress and unfolded protein response in leukemia: Friend, foe, or both? Biomolecules 2021, 11, 199.
  42. Huang, J.; Pan, H.; Wang, J.; Wang, T.; Huo, X.; Ma, Y.; Lu, Z.; Sun, B.; Jiang, H. Unfolded protein response in colorectal cancer. Cell. Biosci. 2021, 11, 26.
  43. Robinson, C.M.; Talty, A.; Logue, S.E.; Mnich, K.; Gorman, A.M.; Samali, A. An emerging role for the unfolded protein response in pancreatic cancer. Cancers 2021, 13, 261.
  44. Morris, G.; Puri, B.K.; Walder, K.; Berk, M.; Stubbs, B.; Maes, M.; Carvalho, A.F. The endoplasmic reticulum stress response in neuroprogressive diseases: Emerging pathophysiological role and translational implications. Mol. Neurobiol. 2018, 55, 8765–8787.
  45. Rashid, H.O.; Yadav, R.K.; Kim, H.R.; Chae, H.J. ER stress: Autophagy induction, inhibition and selection. Autophagy 2015, 11, 1956–1977.
  46. Dymkowska, D. The involvement of autophagy in the maintenance of endothelial homeostasis: The role of mitochondria. Mitochondrion 2021, 57, 131–147.
  47. Picca, A.; Calvani, R.; Coelho-Junior, H.J.; Marzetti, E. Cell death and inflammation: The role of mitochondria in health and disease. Cells 2021, 10, 537.
  48. Su, Y.J.; Wang, P.W.; Weng, S.W. The role of mitochondria in immune-cell-mediated tissue regeneration and ageing. Int. J. Mol Sci. 2021, 22, 2668.
  49. Jang, J.Y.; Blum, A.; Liu, J.; Finkel, T. The role of mitochondria in aging. J. Clin. Investig. 2018, 128, 3662–3670.
  50. Meyers, A.K.; Zhu, X. The NLRP3 Inflammasome: Metabolic regulation and contribution to inflammaging. Cells 2020, 9, 1808.
  51. López-Otín, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217.
  52. Fulop, T.; Witkowski, J.M.; Olivieri, F.; Larbi, A. The integration of inflammaging in age-related diseases. Semin. Immunol. 2018, 40, 17–35.
  53. Hotamisligil, G.S. Endoplasmic reticulum stress and the inflammatory basis of metabolic disease. Cell 2010, 140, 900–917.
  54. Berent-Maoz, B.; Montecino-Rodriguez, E.; Signer, R.A.; Dorshkind, K. Fibroblast growth factor-7 partially reverses murine thymocyte progenitor aging by repression of Ink4a. Blood 2012, 119, 5715–5721.
  55. Sierra-Filardi, E.; Nieto, C.; Domínguez-Soto, A.; Barroso, R.; Sánchez-Mateos, P.; Puig-Kroger, A.; López-Bravo, M.; Joven, J.; Ardavín, C.; Rodríguez-Fernández, J.L.; et al. CCL2 shapes macrophage polarization by GM-CSF and M-CSF: Identification of CCL2/CCR2-dependent gene expression profile. J. Immunol. 2014, 192, 3858–3867.
  56. Yousefzadeh, M.J.; Schafer, M.J.; Noren Hooten, N.; Atkinson, E.J.; Evans, M.K.; Baker, D.J.; Quarles, E.K.; Robbins, P.D.; Ladiges, W.C.; LeBrasseur, N.K.; et al. Circulating levels of monocyte chemoattractant protein-1 as a potential measure of biological age in mice and frailty in humans. Aging Cell 2018, 17, e12706.
  57. Acosta, J.C.; Banito, A.; Wuestefeld, T.; Georgilis, A.; Janich, P.; Morton, J.P.; Athineos, D.; Kang, T.W.; Lasitschka, F.; Andrulis, M.; et al. A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat. Cell. Biol. 2013, 15, 978–990.
  58. Lee, J.Y.; Yu, K.R.; Lee, B.C.; Kang, I.; Kim, J.J.; Jung, E.J.; Kim, H.S.; Seo, Y.; Choi, S.W.; Kang, K.S. GATA4-dependent regulation of the secretory phenotype via MCP-1 underlies lamin a-mediated human mesenchymal stem cell aging. Exp. Mol. Med. 2018, 50, 1–12.
  59. Luciano-Mateo, F.; Cabré, N.; Baiges-Gaya, G.; Fernández-Arroyo, S.; Hernández-Aguilera, A.; Rodríguez-Tomàs, E.; Arenas, M.; Camps, J.; Menéndez, J.A.; Joven, J. Systemic overexpression of C-C motif chemokine ligand 2 promotes metabolic dysregulation and premature death in mice with accelerated aging. Aging 2020, 12, 20001–20023.
  60. Hoyt, L.R.; Randall, M.J.; Ather, J.L.; DePuccio, D.P.; Landry, C.C.; Qian, X.; Janssen-Heininger, Y.M.; van der Vliet, A.; Dixon, A.E.; Amiel, E.; et al. Mitochondrial ROS induced by chronic ethanol exposure promote hyper-activation of the NLRP3 inflammasome. Redox. Biol. 2017, 12, 883–896.
  61. Lavallard, V.; Cottet-Dumoulin, D.; Wassmer, C.H.; Rouget, C.; Parnaud, G.; Brioudes, E.; Lebreton, F.; Bellofatto, K.; Berishvili, E.; Berney, T.; et al. NLRP3 inflammasome is activated in rat pancreatic islets by transplantation and hypoxia. Sci. Rep. 2020, 10, 7011.
  62. Fusco, R.; Gugliandolo, E.; Siracusa, R.; Scuto, M.; Cordaro, M.; D’Amico, R.; Evangelista, M.; Peli, A.; Peritore, A.F.; Impellizzeri, D.; et al. Formyl peptide receptor 1 signaling in acute inflammation and neural differentiation induced by traumatic brain injury. Biology 2020, 9, 238.
More
This entry is offline, you can click here to edit this entry!