Acessibilidade / Reportar erro

Lysosomes, Lysosomal Storage Diseases, and Inflammation

Abstract

Lysosomes were originally described in the early 1950s by de Duve who was also the first to recognize the importance of these organelles in human disease. We know now that lysosomes are involved in numerous biological processes, and abnormalities in lysosomal function may result in a broad range of diseases. This review will briefly discuss the role of lysosomes in inflammation and how disruption of normal lysosomal function in the lysosomal storage diseases (LSDs) leads to abnormalities in inflammation and immunity.

Keywords
lysosomal storage disorders; inflammation

Lysosomes were originally described in the early 1950s by de Duve who was also the first to recognize the importance of these organelles in human disease.11 de Duve, C . The significance of lysosomes in pathology and medicine. Proc Inst Med Chic. 1966;26(4):73–76.,22 de Duve, C. The role of lysosomes in the pathogeny of disease. Scand J Rheumatol Suppl. 1975;12:63–66. We know now that lysosomes are involved in numerous biological processes, and abnormalities in lysosomal function may result in a broad range of diseases. This review will briefly discuss the role of lysosomes in inflammation and how disruption of normal lysosomal function in the lysosomal storage diseases (LSDs) leads to abnormalities in inflammation and immunity.

General Role of Lysosomes in Inflammation

Lysosomes and Autophagy

The primary function of lysosomes is to degrade macromolecules through a series of enzymatic reactions that are activated at acidic pH. This process is required to degrade molecules that are internalized by cells (eg, by the process of endocytosis) or endogenous molecules that are components of normal cell structures (eg, cell membranes, cytoskeleton, nucleic acids). This latter process is referred to as autophagy or autophagocytosis and is one of the key housekeeping functions of lysosomes (for a recent review, see Huber and Teis, 2016).33 Huber, LA, Teis, D. Lysosomal signaling in control of degradation pathways. Curr Opin Cell Biol. 2016;39:8–14. Autophagy is required for the normal homeostasis of cells and is activated in conditions of cellular stress, such as starvation or exposure to toxic molecules. Defective autophagy is found in many human diseases, including cancers, autoimmune diseases, and LSDs.

Autophagy plays a key role in normal host defense mechanisms, and abnormalities in autophagy can lead to numerous inflammatory changes.44 Netea-Maier, RT, Plantinga, TS, van de Veerkdonk, FL, Smit, JW, Netea, MG. Modulation of inflammation by autophagy: consequences for human disease. Autophagy. 2016;12(2):245–260. In healthy cells, autophagy is required to eliminate invading pathogens, to control adaptive immunity through regulation of antigen presentation, and to modulate the inflammatory response. For example, engagement of Toll-like receptor 4 (TLR4) by lipopolysaccharide on gram-negative bacteria results in ubiquitination of beclin-1 and activation of the serine/threonine protein kinase, ULK1.55 Shi, CS, Kehrl, JH. Traf6 and A20 differentially regulate TLR4-induced autophagy by affecting the ubiquitination of Beclin 1. Autophagy. 2010;6(7):986–987. Both pathways lead to activation of autophagy. Engagement of other TLRs also induces autophagy as does activation of NOD-like receptors on dendritic cells. This latter process is required to direct proper antigen handling and presentation.66 Cooney, R, Baker, J, Brain, O. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med. 2010;16(1):90–97. In addition, several proinflammatory cytokines, including tumor necrosis factor (TNF) and interleukin (IL-1β), induce autophagy to improve infection control. In contrast, anti-inflammatory cytokines such as IL-4 and IL-13 antagonize autophagy induction.77 Mostowy, S, Sancho-Shimizu, V, Hamon, MA. p62 and NDP52 proteins target intracytosolic Shigella and Listeria to different autophagy pathways. J Biol Chem. 2011;286(30):26987–269895.,88 Hartman, ML, Kornfeld, H. Interactions between na?¯ve and infected macrophages reduce Mycobacterium tuberculosis viability. PLoS One. 2011;6(11):e27972. Type I and II interferons also largely regulate host cell defense against viruses through the activation of autophagy.99 Singh, SB, Ornatowski, W, Vergne, I. Human IRGM regulates autophagy and cell-autonomous immunity functions through mitochondria. Nat Cell Biol. 2010;12(12):1154–1165.

Perhaps the best studied relationship between autophagy and inflammation is IL-1β release by inflammasomes.1010 Abdelaziz, DH, Khalil, H, Cormet-Boyaka, E, Amer, AO. The cooperation between the autophagy machinery and the inflammasome to implement an appropriate innate immune response: do they regulate each other? Immunol Rev. 2015;265(1):194–204. Inflammasomes are multiprotein complexes containing proteases (mostly caspase 1) required to activate pro-IL-1β and IL-18 into active cytokines.1111 Maltez, VI, Miao, EA. Reassessing the evolutionary importance of inflammasomes. J Immunol. 2016;196(3):956–962. One of the first investigations to link autophagy with inflammation used macrophages from Atg1611 knockout mice that are defective in autophagy.1212 Saitoh, T, Fujita, N, Jang, MH. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008;456(7219):264–268. These mice exhibit increased production of active IL-1β after stimulation with lipopolysaccharides (LPS). Additional studies confirmed these findings and showed that autophagy has an important anti-inflammatory effect in healthy cells and that defective autophagy activates inflammation. Although the mechanisms responsible for this anti-inflammatory effect of autophagy are not fully known, several hypotheses have been put forward. For example, it has been suggested that defective autophagy leads to the accumulation of mitochondria in cells and elevated release of inflammasome activators through the production of reactive oxidative species (ROS) and mitochondrial DNA and/or that autophagy may be required to remove aggregated inflammasome structures, thereby reducing proinflammatory responses.1313 van de Veerdonk, FL, Dinarello, CA. Deficient autophagy unravels the ROS paradox in chronic granulomatous disease. Autophagy. 2014;10(6):1141–1142.,1414 Shi, CS, Shenderov, K, Huang, NN. Activation of autophagy by inflammatory signals limits IL-1? production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol. 2012;13(3):255–263. Aside from the important inhibitory effect of autophagy on inflammasome activation in healthy cells, autophagy also inhibits IL-1β production through non–caspase-1 mechanisms and reduces nuclear factor kappa-light-chain-enhancer of activated B cells (NFKB) activation as well.1515 Cri?an, TO., Plantinga, TS., van de Veerdonk, FL. Inflammasome-independent modulation of cytokine response by autophagy in human cells. PLoS One. 2011;6(4):e18666. It further amplifies the TLR response to improve intracellular delivery of microbial ligands, leading to the production of type I interferon. Thus, autophagy has broad and important effects on host cell immune responses and inflammation, and in the absence of normal lysosomal function and autophagy, dysregulation of these pathways becomes evident.

Lysosomes and Autoimmunity

Further insights regarding the role of lysosomes and inflammation have come from the study of autoimmune diseases. Autoimmune diseases are a diverse group of disorders where organisms produce autoantigens that the immune system cannot distinguish from normal antigens. Recent evidence suggests that lysosomal enzyme activities may contribute to the generation of autoantigens. For example, the lysosomal enzyme α-galactosidase A (defective in the LSD, Fabry disease) normally degrades lipid antigens to prevent their accumulation and activation of CD1d-restricted natural killer T (NKT) cells. Deficiency of this enzyme in Fabry disease causes aberrant accumulation of lipid antigens and activation of immature NKT cells, resulting in autoimmunity.1616 Mauhin, W., Lidove, O., Masat, E. Innate and adaptive immune response in Fabry disease. JIMD Rep. 2015;22:1–10.

The first major autoinflammatory disease in which lysosome was suggested to play a role was Crohn disease (for review see Spalinger et al).1717 Spalinger, MR, Rogler, G, Scharl, M. Crohn’s disease: loss of tolerance or a disorder of autophagy? Dig Dis. 2014;32(4):370–377. Crohn disease is a chronic inflammatory disorder of the intestine caused by a combination of factors including defective host response, altered mucosal barriers, and exaggerated cytokine production. Genetic variants of several important autophagy genes have been associated with susceptibility and clinical severity of Crohn disease. Variants of ATG16L1, in particular, have been linked to defective bacterial defense, aberrant antigen presentation, and increased production of proinflammatory cytokines, including IL-1β and IL-18.1818 Murthy, A., Li, Y., Peng, I. A Crohn’s disease variant in Atg16l1 enhances its degradation by caspase 3. Nature. 2014;506(7489):456–462.,1919 Lassen, KG., Kuballa, P., Conway, KL. Activation of autophagy by inflammatory signals limits IL-1? production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol. 2012;13(3):255–263. Other important insights regarding the relationship of autophagy, lysosomes, and human disease arise from studies of chronic granulomatosus diseases (CGDs). Chronic granulomatosus disease is a primary immunodeficiency characterized by defective ROS production, defects in host defense, and hyperinflammatory responses leading to colitis in a significant number of patients.2020 Holmes, B, Page, AR, Good, RA. Studies of the metabolic activity of leukocytes from patients with a genetic abnormality of phagocytic function. J Clin Invest. 1967;46(9):1422–1432. Defective autophagy has also been demonstrated in patients with CGD, leading to enhanced inflammasome activation and release of IL-1β, which can be treated using the IL-1 receptor antagonist, IL1RN/IL1RA.1313 van de Veerdonk, FL, Dinarello, CA. Deficient autophagy unravels the ROS paradox in chronic granulomatous disease. Autophagy. 2014;10(6):1141–1142. Other epidemiology studies have shown associations of autophagy genes with other autoimmune conditions as well, including systemic lupus erythematosus and multiple sclerosis.2121 Gateva, V, Sandling, JK, Hom, G. A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nat Genet. 2009;41(11):1228–1233. Thus, healthy lysosomes are required to maintain a normal autoimmune response, and lysosomal dysfunction may lead to a range of autoimmune-related pathologies.

Lysosomes, Sphingolipid Metabolism, and Inflammation

Another important, recent link between inflammation and lysosomes has come from the study of bioactive sphingolipids, and in particular ceramide and its derivative, sphingosine-1-phosphate (S1P; for review, see Gomez-Muñoz et al).2222 Gomez-Mu?±oz, A., Presa, N., Gomez-Larrauri, A., Rivera, IG., Trueba, M., Ordoñez, M. Control of inflammatory responses by ceramide, sphingosine 1-phosphate and ceramide 1-phosphate. Prog Lipid Res. 2016;61:51–62. Lysosomes are a major cellular compartment involved in the breakdown and metabolism of sphingolipids, and defects in lysosomal function leads to dysregulation of these lipids and activation of inflammatory pathways.2323 Iwabuchi, K., Nakayama, H., Oizumi, A. Role of ceramide from glycosphingolipids and its metabolites in immunological and inflammatory responses in humans. Mediators Inflamm. 2015;2015:120748. Ceramides are a diverse group of sphingolipids consisting of an 18-carbon sphingoid base backbone, sphingosine, linked via an amide bond to a long-chain fatty acid. The fatty acids present in ceramides may vary in chain length and the degree of saturation, providing structural and functional diversity to this lipid class. Ceramides are important regulators of many biological processes, including apoptosis, cell adhesion, inflammation, and infection. They are degraded through the activity of a class of enzymes known as ceramidases, releasing sphingosine and free fatty acids.2424 Mao, C., Obeid, LM. Ceramidases: regulators of cellular responses mediated by ceramide, sphingosine, and sphingosine-1-phosphate. Biochim Biophys Acta. 2008;1781(9):424–434. Sphingosine may be further hydrolyzed to S1P by the activity of sphingosine kinases. Sphingosine-1-phosphate similarly has numerous biological roles, including the induction of cell proliferation and inflammation.

Lysosomes are a key site of ceramide production through the breakdown of sphingomyelin and/or glycosphingolipids. The lysosomal enzyme, acid ceramidase, is then required to hydrolyze ceramides into sphingosine, thereby maintaining the levels of these 2 important signaling molecules. Sphingosine is converted into S1P in the cytosol, where it may then be released into the circulation. Defective lysosomal function, as in the LSDs, disrupts the normal metabolism of sphingolipids, particularly, ceramides, sphingosine, and S1P, thereby dysregulating many important signaling pathways.

One of the best examples linking abnormal sphingolipid metabolism with inflammation comes from studies of cystic fibrosis (CF). Seitz et al were the first to show that lung cells and tissues from patients and mice with CF had elevated ceramides and that inhibition of ceramide production (through inhibition of the lysosomal enzyme acid sphingomyelinase [ASM], which degrades sphingomyelin into ceramide) reduced inflammation and infection.2525 Seitz, AP, Grassmé, H, Edwards, MJ, Pewzner-Jung, Y, Gulbins, E. Ceramide and sphingosine in pulmonary infections. Biol Chem. 2015;396(6-7):611–620. Acid sphingomyelinase is activated in CF and many other diseases, producing excess amounts of ceramides. This, in turn, alters the biophysical properties of the cell membranes, inducing abnormal cell signaling. Others have shown that several proinflammatory cytokines, including TNF-ñ, interferons, and IL-1β, activate ASM activity, contributing to the elevated ceramides.2626 Chakravarthy, H, Navitskaya, S, O’Reilly, S. Role of acid sphingomyelinase in shifting the balance between pro-inflammatory and reparative bone marrow cells in diabetic retinopathy. Stem Cells. 2016;34(4):972–983. Other inflammatory lung conditions, including asthma and chronic obstructive lung disease, have also been linked to excess ceramides.2727 Caretti, A., Torelli, R, Perdoni. Inhibition of ceramide de novo synthesis by myriocin produces the double effect of reducing pathological inflammation and exerting antifungal activity against A. fumigatus airways infection. Biochim Biophys Acta. 2016;1860(6):1089–1097.,2828 Petrache, I., Berdyshev, EV. Ceramide signaling and metabolism in pathophysiological states of the lung. Annu Rev Physiol. 2016;78:463–480. Most recently, Pewzner-Jung et al showed that sphingosine, the product of acid ceramidase activity, is required to prevent bacterial infections and that patients with CF had reduced acid ceramidase activity.2929 Pewzner-Jung, Y, Tavakoli Tabazavareh, S, Grassmé, H. Sphingoid long chain bases prevent lung infection by Pseudomonas aeruginosa. EMBO Mol Med. 2014;6(9):1205–12014. Inhalation of recombinant acid ceramidase into the lungs of CF mice reduced ceramide and elevated sphingosine and protected these mice from infection with Pseudomonas aeruginosa.

Thus, lysosomes play a diverse and important role in immunity and inflammation, in part through the regulation of autophagy, control of inflammasome release of cytokines, and the regulation of sphingolipid metabolism. In general, healthy lysosomes are required to exhibit a normal host response to infection and to maintain a normal inflammatory response. Defective lysosomes lead to abnormal autophagy, activation of inflammation, and reduced infection control.

Lysosomal Storage Diseases and Inflammation

The LSDs represent a group of more than 60 disorders due to inherited deficiencies of lysosomal enzymes or transport proteins.3030 Vitner, EB, Farfel-Becker, T, Eilam, R, Biton, I, Futerman, AH. Contribution of brain inflammation to neuronal cell death in neuronopathic forms of Gaucher’s disease. Brain. 2012;135(pt 6):1724–1735. All are inherited as autosomal recessive traits except for two, Fabry disease and Hunter disease (mucopolysaccharidosis [MPS] II), which are X-linked.3131 Parkinson-Lawrence, EJ, Shandala, T, Prodoehl, M, Plew, R, Borlace, GN, Brooks, DA. Lysosomal storage diseases: revealing lysosomal function and physiology. Physiology. 2010;25(2):102–115. The result of these deficiencies is the accumulation of undegraded macromolecules, initially within lysosomes but spreading over time to other cellular compartments. The pattern of macromolecule accumulation varies with the disease. In general, it initiates with a specific enzyme substrate, but as the disease progresses and the lysosomal dysfunction continues, secondary deficiencies of other enzymes occur, leading to a complex pattern of macromolecule storage. A common, initial response by the cell to the LSD abnormalities is the production of more lysosomes. However, since these newly formed lysosomes also have inherent abnormalities, they become dysfunctional as well. Eventually, the lysosomal system shuts down, leading to abnormalities in endocytosis, autophagy, inflammation, and ultimately causing cell death.

One of the best examples linking LSDs with inflammation is Gaucher disease (acid β-glucosidase [glucocerebrosidase] deficiency). The accumulating substrate, glucosylceramide, appears predominantly in macrophages as does its deacylated product, glucosylsphingosine. The accumulation of these lipids leads to macrophage activation, inducing the release of various cytokines, including chitotriosidase, TNF-ñ, IL-1β, and others.3232 Barak, V, Acker, M, Nisman, B. Cytokines in Gaucher’s disease. Eur Cytokine Netw. 1999;10(2):205–210.3434 Hollak, CE., Evers, L., Aerts, JM., van Oers, MH. Elevated levels of M-CSF, sCD14 and IL8 in type 1 Gaucher disease. Blood Cells Mol Dis. 1997;23(2):201–212. This, in turn, sends out a call signal to recruit additional immune cells, including macrophages and neutrophils, to the sites of pathology. However, since these cells are themselves dysfunctional, they simply serve to amplify the pathology and disease burden in patients, rather than correct it.

In addition to macrophage activation and cytokine release, polyclonal and monoclonal gammopathies also occur in some patients with Gaucher disease.3535 Linari, S, Castaman, G. Hematological manifestations and complications of Gaucher disease. Expert Rev Hematol. 2016;9(1):51–58. Monoclonal gammopathy is a risk factor for certain myeloid cancers, which have been reported to occur at a higher than expected frequency in patients with Gaucher disease.3636 Machaczka, M, Lerner, R, Klimkowska, M, Hägglund, H. Treatment of multiple myeloma in patients with Gaucher disease. Am J Hematol. 2009;84(10):694–696. Interleukin 6 is elevated in patients with Gaucher disease as well and is associated with the expansion of myeloid cells. Also, IL-10 is elevated and may contribute to the increased occurrence of autoantibodies and B-cell lymphomas. It is unclear how accumulation of glucosylceramide and/or glucosylsphingosine stimulates these events, but an attractive hypothesis has to do with the sphingolipid pathway discussed earlier. Both accumulating lipids in Gaucher disease are precursors of ceramide and sphingosine (and ultimately S1P) and could influence these inflammatory pathways through the production of these downstream metabolites.

Also of note, Aflaki et al have recently shown that impaired autophagy in Gaucher disease macrophages leads to inflammasome activation and elevated secretion of IL-1β. These investigators further demonstrated that treatment with a molecular chaperone (NCGC758) that enhances the function of mutant glucocerebrosidase reversed these defects, confirming the relationship between lysosomal dysfunction and inflammasome activation.3737 Aflaki, E, Moaven, N, Borger, DK. Lysosomal storage and impaired autophagy lead to inflammasome activation in Gaucher macrophages. Aging Cell. 2016;15(1):77–88.

Others have studied the specific role of neuroinflammation in Gaucher disease and found highly elevated levels of IL-1β, TNF-ñ macrophage colony-stimulating factor, and transforming growth factor β in the brains of a neuropathic Gaucher disease mouse model.3030 Vitner, EB, Farfel-Becker, T, Eilam, R, Biton, I, Futerman, AH. Contribution of brain inflammation to neuronal cell death in neuronopathic forms of Gaucher’s disease. Brain. 2012;135(pt 6):1724–1735. Activation of microglia in the Gaucher disease brain has also been demonstrated. Kartha et al have also shown that patients with Gaucher type 1 disease exhibit increased systemic oxidative stress in addition to inflammation.3838 Kartha, RV, Elnashar, G, Hovde, L, Mishra, U, Utz, J, Cloyd, JC. Characterization of oxidative stress and inflammation in type 1 Gaucher disease-method evaluation and preliminary analysis. Mol Genet Metab. 2016;117(2):S65. Thus, inflammation is a major contributing factor to the multiorgan system pathology in Gaucher disease, and targeting inflammation could potentially provide therapeutic benefits in patients with Gaucher disease.

Another important LSD in which inflammation and an abnormal immune response play a major role is Fabry disease, due to the deficiency of the enzyme ñ-galactosidase A.1616 Mauhin, W., Lidove, O., Masat, E. Innate and adaptive immune response in Fabry disease. JIMD Rep. 2015;22:1–10. The primary substrate accumulating in this disorder is the glycosphingolipid, globotriaosylceramide (Gb3). In Fabry disease knockout mice, age progressive increases in Gb3 led to a progressive decrease in invariant (type 1) NKT (iNKT) cells, principally in the spleen but other organs as well.3939 Macedo, MF, Quinta, R, Pereira, CS, Sa Miranda, MC. Enzyme replacement therapy partially prevents invariant Natural Killer T cell deficiency in the Fabry disease mouse model. Mol Genet Metab. 2012;106(1):83–91. Pereira et al also reported a decrease in CD4-positive iNKT cells, as well as a reduction in IL-4 production in patients with Fabry disease.4040 Pereira, CS, Azevedo, O, Maia, ML, Dias, AF, Sa-Miranda, C, Macedo, MF. Invariant natural killer T cells are phenotypically and functionally altered in Fabry disease. Mol Genet Metab. 2013;108(4):241–248. Rozenfeld et al showed a low level of Cd1d and hypothesized that this could be due to increased expression of major histocompatibility complex (MHC) class II molecules in monocytes.4141 Rozenfeld, P, Agriello, E, De Francesco, N, Martinez, P, Fossati, C. Leukocyte perturbation associated with Fabry disease. J Inherit Metab Dis. 2009;32(suppl 1):S67–S77. Thus, it appears that glycosphingolipid storage in patients with Fabry disease, and Gb3 specifically, leads to dysregulation of iNKT distribution and the Cd1 pathway in general.

The proinflammatory feature of Fabry disease also has been well described, with an increase in inflammatory cytokines including IL-6, IL-1β, and TNF-ñ among others.4242 De Francesco, PN, Mucci, JM, Ceci, R, Fossati, CA, Rozenfeld, PA. Fabry disease peripheral blood immune cells release inflammatory cytokines: role of globotriaosylceramide. Mol Genet Metab. 2013;109(1):93–99. This elevation appears to be related to the TLR4 pathway, since the elevated cytokine release in Fabry cells was reduced using TLR4-blocking agents. The TLR4 pathway is activated in many LSDs, and TLR4 has been shown to bind glycosaminoglycans (GAGs) that accumulate in the MPS (see below), as well as glycosphingolipids such as the gangliosides GM1 and GM2.4343 Jou, I, Lee, JH, Park, SY, Yoon, HJ, Joe, EH, Park, EJ. Gangliosides trigger inflammatory responses via TLR4 in brain glia. Am J Pathol. 2006;168(5):1619–1630. In addition, Segura et al have shown increased endothelial inflammatory profiles in serum from patients with early-stage Fabry disease.4444 Segura, T, Ayo-Martín, O, Gómez-Fernandez, I, Andrés, C, Barba, MA, Vivancos, J. Cerebral hemodynamics and endothelial function in patients with Fabry disease. BMC Neurol. 2013;13:170. These patients had elevated levels of endothelium-released biomarkers (TNF-ñ, C-reactive protein, and soluble vascular cell adhesion molecule 1), indicative of the systemic endothelial dysfunction that is believed to be the cause of the thrombogenicity in Fabry disease. Biancini et al also investigated proinflammatory cytokines, oxidative stress parameters, and Gb3 levels in patients with Fabry disease receiving enzyme replacement therapy (ERT).4545 Biancini, GB, Vanzin, CS, Rodrigues, DB. Globotriaosylceramide is correlated with oxidative stress and inflammation in Fabry patients treated with enzyme replacement therapy. Biochim Biophys Acta. 2012;1822(2):226–232. Their data suggest that Gb3 is directly correlated with the induction of elevated IL-6, TNF-ñ, and oxidative stress in patients. Also, as with other LSDs, lysosomal dysfunction in Fabry disease leads to defects in autophagy, which stimulates numerous inflammatory pathways (as described earlier).4646 Liebau, MC, Braun, F, H?¶pker, K. Dysregulated autophagy contributes to podocyte damage in Fabry’s disease. PLoS One. 2013;8(5):e63506.

Another intriguing but less studied LSD in which inflammation appears to play an important role is Farber disease, due to the deficiency of the enzyme acid ceramidase and the resultant accumulation of the sphingolipid, ceramide.4747 Frohbergh, M, He, X, Schuchman, EH. The molecular medicine of acid ceramidase. Biol Chem. 2015;396(6-7):759–765. As noted earlier, ceramide is an important signaling lipid that has potent proapoptotic and proinflammatory characteristics. As with other LSDs, Farber disease presents with a wide spectrum of clinical findings, but all patients develop subcutaneous nodules that are composed of lipid-filled immune cells, principally macrophages and neutrophils. These nodules are mostly evident at cartilaginous sites but have been found in the spleen and other organs as well. Alayoubi et al constructed a mouse knock-in model of Farber disease and found the very early elevation in several proinflammatory cytokines, principally monocyte chemokine protein 1 (MCP-1).4848 Alayoubi, AM., Wang, JC., Au, BC. Systemic ceramide accumulation leads to severe and varied pathological consequences. EMBO Mol Med. 2013;5(6):827–842. They went on to hypothesize that the elevation in MCP-1, likely due the accumulation of ceramides, is an initiating event leading to the formation of subcutaneous nodules and other pathologies occurring in patients with Farber disease and that therapies that reduce MCP-1 would be beneficial in these patients.

The first systematic studies demonstrating the importance of inflammation in the LSDs were carried out by Richard Proia at the National Institutes of Health (NIH). Working with mouse models of GM1 and GM2 gangliosidosis, Proia and colleagues found widespread microglial activation in the brains of these animals that preceded neuronal cell death.4949 Wada, R., Tifft, CJ., Proia, RL. Microglial activation precedes acute neurodegeneration in Sandhoff disease and is suppressed by bone marrow transplantation. Proc Natl Acad Sci U S A. 2000;97(20):10954–10959. Bone marrow transplantation (BMT) reduced inflammation and neuronal cell death, indicating a direct link between inflammation and neurodegeneration. Wu and Proia also conducted a study in Sandhoff disease mice where they bred these animals to mice lacking macrophage inflammatory protein 1ñ (MIP-1ñ).5050 Wu, YP, Proia, RL. Deletion of macrophage-inflammatory protein 1 alpha retards neurodegeneration in Sandhoff disease mice. Proc Natl Acad Sci U S A. 2004;101(22):8425–8430. Macrophage inflammatory protein 1ñ release is elevated in these mice, and double-mutant mice lacking this chemokine showed improved neurological status and a longer lifespan. Groh et al have investigated the role of the inflammation-related cell adhesion molecule sialoadhesin (Sn) in Ppt12/2 and Cln32/2 mice, models of the most common form of neuronal ceroid lipofuscinoses, CLN3 disease.5151 Groh, J, Ribechini, E, Stadler, D, Schilling, T, Lutz, MB, Martini, R. Sialoadhesin promotes neuroinflammation-related disease progression in two mouse models of CLN disease. Glia. 2016;64(5):792–809. Microglia/macrophages in the CNS of both models showed an upregulation of Sn and markers for proinflammatory M1 polarization and antigen presentation.

Based on the early work of Proia, others have further investigated the use of anti-inflammatory therapies in patients with LSD, cells, and/or animals.5252 Bosch, ME., Kielian, T. Neuroinflammatory paradigms in lysosomal storage diseases. Front Neurosci. 2015;9:417. Although most of the clinical studies have been isolated reports, in general some benefit has been observed. For example, a recent report revealed the therapeutic potential of Resveratrol in Gaucher disease cells and patients.5353 Seo, CH., Kim, JB. Therapeutic potential of resveratrol in type I Gaucher disease. Phytother Res. 2015;29(6):835–839.,5454 Yoo, S., Kim, JB. Anti-apoptotic and beneficial metabolic activities of resveratrol in type II Gaucher disease. Biol Pharm Bull. 2015;38(6):913–918. Resveratrol is a natural polyphenol with beneficial anti-inflammatory, antioxidant, and neuroprotective effects. Also, Miglustat, an approved therapy for some patients with Gaucher disease, has been shown to work, at least in part, by mitigating inflammation in bronchial epithelial cells.5555 Dechecchi, MC., Nicolis, E., Norez, C. Anti-inflammatory effect of miglustat in bronchial epithelial cells. J Cyst Fibros. 2008;7(6):555–565. In an interesting study, Williams et al showed the synergistic benefit of Miglustat, curcumin, and the nonsteroidal anti-inflammatory ibuprofen in a mouse model of Niemann-Pick disease type C.5656 Williams, IM., Wallom, KL., Smith, DA., Al Eisa, N., Smith, C., Platt, FM. Improved neuroprotection using miglustat, curcumin and ibuprofen as a triple combination therapy in Niemann-Pick disease type C1 mice. Neurobiol Dis. 2014;67:9–17. Stein et al also used the nonsteroidal anti-inflammatory drug, simvastatin, in a mouse model of metachromatic leukodystrophy (MLD) and showed reduced neuroinflammation and retarded demyelination of the spinal cord (but not of the brain).5757 Stein, A., Stroobants, S., Gieselmann, V., D’Hooge, R., Matzner, U. Anti-inflammatory therapy with simvastatin improves neuroinflammation and CNS function in a mouse model of metachromatic leukodystrophy. Mol Ther. 2015;23(7):1160–1168. This MLD mouse model is analogous to the human condition and is characterized by a sustained elevation in MIP-1ñ leading to an upregulation of MIP-1β, MCP-1, and several ILs (1β, 6, 9, 13), ultimately resulting in demyelination. Globoid cell leukodystrophy (GLD) is another example of an LSD, which is characterized by neuroinflammation. The murine model of GLD, twitcher, exhibits dysregulation of the Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling pathway and an abnormal inflammatory response. Rafi et al showed a reduction in inflammation and therapeutic benefit in twitcher mice by combining BMT with CNS-directed gene therapy.5858 Rafi, MA., Rao, HZ., Luzi, P., Wenger, DA. Long-term improvements in lifespan and pathology in CNS and PNS after BMT plus one intravenous injection of AAVrh10-GALC in Twitcher mice. Mol Ther. 2015;23(11):1681–1690. Christina et al have also shown a reduction in systemic and neuroinflammation in the twitcher mice by targeting the JAK/STAT pathway with AZD 1480, an inhibitor of JAK1/2, which has been successfully used in multiple sclerosis mice.5959 Christina, R, Mikulka, Hongwei, Q. Reducing neuroinflammation in globoid cell leukodystrophy by targeting the JAK/STAT pathway. Mol Genet Metab. 2016;117(2):S80.

Neuroinflammation and oxidative stress are also major components of the neuropathology present in several MPS diseases, including Sanfilippo (MPS III) and Sly (MPS VII) diseases.6060 Richard, M, Arfi, A, Rhinn, H, Gandolphe, C, Scherman, D. Identification of new markers for neurodegeneration process in the mouse model of Sly disease as revealed by expression profiling of selected genes. J Neurosci Res. 2008;86(15):3285–3294.,6161 Arfi, A, Richard, M, Gandolphe, C, Bonnefont-Rousselot, D, Thérond, P, Scherman, D. Neuroinflammatory and oxidative stress phenomena in MPS IIIA mouse model: the positive effect of long-term aspirin treatment. Mol Genet Metab. 2011;103(1):18–25. For example, Ausseil et al showed that MIP-1ñ messenger RNA was increased 10-fold in the brain of MPS IIIB mice when compared to normal.6262 Ausseil, J., Desmaris, N., Bigou, S. Early neurodegeneration progresses independently of microglial activation by heparan sulfate in the brain of mucopolysaccharidosis IIIB mice. PLoS One. 2008;3(5):e2296. In addition, serum MIP-ñ was elevated in these mice beginning at 6 weeks of age and increased as the disease progressed. In these neurological forms of MPS, microglia activation via inflammation and oxidative stress contributes to cell dysfunction or apoptosis. Arfi et al also suggested that treatment with nonsteroidal anti-inflammatory drugs such as aspirin could be beneficial both neurologically and systemically by reducing inflammation when used in conjunction with substrate reducing therapies.6161 Arfi, A, Richard, M, Gandolphe, C, Bonnefont-Rousselot, D, Thérond, P, Scherman, D. Neuroinflammatory and oxidative stress phenomena in MPS IIIA mouse model: the positive effect of long-term aspirin treatment. Mol Genet Metab. 2011;103(1):18–25. Simonaro et al have further shown that subcutaneous pentosan polysulfate (PPS) injection (see below) can reduce systemic inflammation, hyperactivity, P-tau formation, and the formation of activated microglia in MPS IIIA mice.6363 Simonaro, CM., Guo, N., Schuchman, EH. Pentosan polysulfate and neuroinflammation in mice with mucopolysaccharidosis type IIIA mice. Mol Genet Metab. 2016;117(2):S106. Thus, inflammation has been shown to be a critical pathogenic factor in many LSDs, and anti-inflammatory drugs should be considered alone or as combination therapies with ERTs and gene therapies that target the primary enzyme defect.

Inflammation and the MPS Disorders

Perhaps the best studied use of anti-inflammatory therapies in the LSDs is within the MPS subgroup of disorders. The MPS comprises a group of 11 LSDs due to deficiencies of enzymes that degrade GAGs. Since GAGs are major constituents of connective tissues, these disorders primarily affect the skeletal system (cartilage and bone), skin, and other connective tissue systems. ERTs are available for 4 of these disorders and under development for several others. However, because the recombinant enzymes do not reach many of the connective tissue compartments, these therapies have limited effects in these tissues.

In 2005, Simonaro and colleagues showed that GAG storage in MPS leads to cytokine elevation in MPS cells and animals.6464 Simonaro, CM, D’Angelo, M, Haskins, ME, Schuchman, EH. Joint and bone disease in mucopolysaccharidoses VI and VII: identification of new therapeutic targets and biomarkers using animal models. Pediatr Res. 2005;57(5 pt 1):701–707. Among these cytokines, TNF-ñ and IL-1β were the most predominant. Although the elevation in proinflammatory cytokines could be due to the general lysosomal dysfunction and mechanisms discussed above (eg, defective autophagy), in the case of the MPS diseases, GAG storage seemed to have an exacerbating and important additional effect. These studies also showed that in addition to proinflammatory cytokines, there was a very significant elevation in inflammatory proteases, such as the metalloproteinases, leading to degradation of extracellular matrix components, as well as increased apoptosis of connective tissue cells.

A further study investigated the mechanism of inflammation in MPS and found that GAGs bind TLR4 receptors and activate the TLR4 pathway, leading to TNF-ñ and other cytokine release.6565 Simonaro, CM, Ge, Y, He, X, Jepsen, KJ, Schuchman, EH. Involvement of the toll-like receptor 4 pathway and the use of anti-TNF-alpha agents for treatment in the mucopolysaccharidoses. Proc Natl Acad Sci U S A. 2010;107(1):222–227. Several different GAGs were evaluated (eg, dermatan, heparin, and chondroitin sulfate), each of which activated TLR4 to varying degrees. Of interest, in this study, ceramide levels were also found to be elevated in MPS cells (chondrocytes), likely contributing to the proinflammatory and proapoptotic cartilage phenotype. In contrast, S1P levels were elevated in synovial fibroblasts, perhaps contributing to the synovial hyperplasia that is observed in patients. These changes in sphingolipid levels are likely due to the general lysosomal dysfunction discussed above.

To demonstrate the importance of the TLR4 pathway in MPS, a study was carried out where mice with MPS type VII (β-glucuronidase deficiency) were bred to mice lacking TLR4. Remarkably, even in the absence of any other therapy, these MPS mice had a substantially improved phenotype, including enhanced bone length, reduced cartilage degradation, and improved tracheal morphology. These investigators also evaluated the use of a TNF-ñ antagonist in rats with MPS type VI and found reduced levels of inflammatory cytokines and enhanced cartilage integrity.

The abovementioned studies highlighted the importance of inflammation in MPS and indicated that the TLR4 pathway was commonly activated in these disorders. However, while TNF-ñ antagonists were effective, they remain extremely expensive and, in general, are potent immunosuppressants that pose some risk to patients. This led to the evaluation of another drug called PPS. Pentosan polysulfate is a low-molecular-weight, carbohydrate polymer with mild antithrombotic and strong anti-inflammatory properties. It has been studied for over 40 years and is currently available as an oral tablet in the United States to treat interstitial cystitis (Elmiron) and as an injectable form in some European countries to treat deep vein thrombosis (SP54). Animal studies have shown it has potent anti-inflammatory effects in models of diabetes, arthritis, and others, and it is also used in veterinary medicine to treat inflammatory arthritis (Cartrophen).

In 2 studies, PPS was evaluated in MPS-type VI rats. It was found to markedly reduce inflammation, reduce chondrocyte apoptosis, improve tracheal morphology, and improve motility in these animals. Moreover, and unexpectedly, it was also found to significantly reduce GAG levels in the treated animals. Although the mechanism(s) for this latter finding is not known, it has been hypothesized that it may be due to the structural similarities between PPS and GAGs, leading to inhibition of the GAG synthetic pathway (ie, acting as a “substrate-reducing drug”?). Thus, inflammation plays an important and common role in the MPS disorders, and anti-inflammatory therapies such as PPS may have very positive effects, alone or in combination with ERTs.

Recently, Polgreen et al also showed that inflammation mediated by TNF-ñ is responsible for pain, decreased physical activity, and short stature in MPS I, II, and VI children, despite having received BMT, ERT, or both.6666 Polgreen, LE, Vehe, RK, Rudser, K. Elevated TNF-?± is associated with pain and physical disability in mucopolysaccharidosis types I, II, and VI. Mol Genet Metab. 2016;117(4):427–430. They also concluded that in this patient population, resistance to human growth hormone therapy might be due to TNF-ñ inhibition of the hormone. Donida et al have also reported elevated plasma levels of IL-6 and pro-oxidant states in MPS four A (IVA) patients undergoing ERT.6767 Donida, B, Marchetti, DP, Biancini, GB. Oxidative stress and inflammation in mucopolysaccharidosis type IVA patients treated with enzyme replacement therapy. Biochim Biophys Acta. 2015;1852(5):1012–10 These studies confirm the persistence of inflammation despite therapies and warrant the use of anti-inflammatory therapies as adjuncts to alleviate pain and improve quality of life.

Most recently, PPS has been evaluated in 2 small clinical studies (adult MPS type I and type II).6868 Hennermann, JB, Goekce, S, Solymon, A, Mengel, E, Schuchman, EH, Simonaro, CM. Treatment with pentosan polysulphate in patients with mucopolysaccharidosis type I: results from an open label, randomized, monocentric phase 2 study. Mol Genet Metab. 2016;117(2):S56.,6969 Orii, K, Tomatsu, S, Suzuki, Y. Safety study of sodium pentosan polysulfate for adult patients with mucopolysaccharidosis type II. Mol Genet Metab. 2016;117(2):S56. These studies were carried out for 12 and 24 weeks, respectively, and PPS was well tolerated by all patients. Hennermann et al showed an improvement in range of motion in the joints of 3 of 4 patients with MPS-I, with significant decreases in pain score and GAG excretion.6868 Hennermann, JB, Goekce, S, Solymon, A, Mengel, E, Schuchman, EH, Simonaro, CM. Treatment with pentosan polysulphate in patients with mucopolysaccharidosis type I: results from an open label, randomized, monocentric phase 2 study. Mol Genet Metab. 2016;117(2):S56. Orii et al also recorded an improvement in range of motion in patients with MPS-II and a reduction in serum inflammatory cytokines.6969 Orii, K, Tomatsu, S, Suzuki, Y. Safety study of sodium pentosan polysulfate for adult patients with mucopolysaccharidosis type II. Mol Genet Metab. 2016;117(2):S56.

In conclusion, storage of macromolecules in the LSDs activates numerous inflammatory pathways, resulting in both local (eg, brain) and systemic inflammation. Inflammation in the LSDs may be directly due to substrate storage (eg, GAGs in MPS) or indirectly due to the general lysosomal dysfunction that occurs (eg, defective autophagy). Inflammation persists in LSD animal models and patients even after treatment by ERT or BMT, indicating that adjunct anti-inflammatory therapies might be beneficial in these disorders. Such therapies might also be useful in patients for which ERT or BMT is not available or as “bridging”? drugs for patients identified through newborn screening programs that cannot immediately receive these therapies.

Funding

  • The author(s) received no financial support for the research, authorship, and/or publication of this article.

References

  • 1
    de Duve, C . The significance of lysosomes in pathology and medicine. Proc Inst Med Chic. 1966;26(4):73–76.
  • 2
    de Duve, C. The role of lysosomes in the pathogeny of disease. Scand J Rheumatol Suppl. 1975;12:63–66.
  • 3
    Huber, LA, Teis, D. Lysosomal signaling in control of degradation pathways. Curr Opin Cell Biol. 2016;39:8–14.
  • 4
    Netea-Maier, RT, Plantinga, TS, van de Veerkdonk, FL, Smit, JW, Netea, MG. Modulation of inflammation by autophagy: consequences for human disease. Autophagy. 2016;12(2):245–260.
  • 5
    Shi, CS, Kehrl, JH. Traf6 and A20 differentially regulate TLR4-induced autophagy by affecting the ubiquitination of Beclin 1. Autophagy. 2010;6(7):986–987.
  • 6
    Cooney, R, Baker, J, Brain, O. NOD2 stimulation induces autophagy in dendritic cells influencing bacterial handling and antigen presentation. Nat Med. 2010;16(1):90–97.
  • 7
    Mostowy, S, Sancho-Shimizu, V, Hamon, MA. p62 and NDP52 proteins target intracytosolic Shigella and Listeria to different autophagy pathways. J Biol Chem. 2011;286(30):26987–269895.
  • 8
    Hartman, ML, Kornfeld, H. Interactions between na?¯ve and infected macrophages reduce Mycobacterium tuberculosis viability. PLoS One. 2011;6(11):e27972.
  • 9
    Singh, SB, Ornatowski, W, Vergne, I. Human IRGM regulates autophagy and cell-autonomous immunity functions through mitochondria. Nat Cell Biol. 2010;12(12):1154–1165.
  • 10
    Abdelaziz, DH, Khalil, H, Cormet-Boyaka, E, Amer, AO. The cooperation between the autophagy machinery and the inflammasome to implement an appropriate innate immune response: do they regulate each other? Immunol Rev. 2015;265(1):194–204.
  • 11
    Maltez, VI, Miao, EA. Reassessing the evolutionary importance of inflammasomes. J Immunol. 2016;196(3):956–962.
  • 12
    Saitoh, T, Fujita, N, Jang, MH. Loss of the autophagy protein Atg16L1 enhances endotoxin-induced IL-1beta production. Nature. 2008;456(7219):264–268.
  • 13
    van de Veerdonk, FL, Dinarello, CA. Deficient autophagy unravels the ROS paradox in chronic granulomatous disease. Autophagy. 2014;10(6):1141–1142.
  • 14
    Shi, CS, Shenderov, K, Huang, NN. Activation of autophagy by inflammatory signals limits IL-1? production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol. 2012;13(3):255–263.
  • 15
    Cri?an, TO., Plantinga, TS., van de Veerdonk, FL. Inflammasome-independent modulation of cytokine response by autophagy in human cells. PLoS One. 2011;6(4):e18666.
  • 16
    Mauhin, W., Lidove, O., Masat, E. Innate and adaptive immune response in Fabry disease. JIMD Rep. 2015;22:1–10.
  • 17
    Spalinger, MR, Rogler, G, Scharl, M. Crohn’s disease: loss of tolerance or a disorder of autophagy? Dig Dis. 2014;32(4):370–377.
  • 18
    Murthy, A., Li, Y., Peng, I. A Crohn’s disease variant in Atg16l1 enhances its degradation by caspase 3. Nature. 2014;506(7489):456–462.
  • 19
    Lassen, KG., Kuballa, P., Conway, KL. Activation of autophagy by inflammatory signals limits IL-1? production by targeting ubiquitinated inflammasomes for destruction. Nat Immunol. 2012;13(3):255–263.
  • 20
    Holmes, B, Page, AR, Good, RA. Studies of the metabolic activity of leukocytes from patients with a genetic abnormality of phagocytic function. J Clin Invest. 1967;46(9):1422–1432.
  • 21
    Gateva, V, Sandling, JK, Hom, G. A large-scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nat Genet. 2009;41(11):1228–1233.
  • 22
    Gomez-Mu?±oz, A., Presa, N., Gomez-Larrauri, A., Rivera, IG., Trueba, M., Ordoñez, M. Control of inflammatory responses by ceramide, sphingosine 1-phosphate and ceramide 1-phosphate. Prog Lipid Res. 2016;61:51–62.
  • 23
    Iwabuchi, K., Nakayama, H., Oizumi, A. Role of ceramide from glycosphingolipids and its metabolites in immunological and inflammatory responses in humans. Mediators Inflamm. 2015;2015:120748.
  • 24
    Mao, C., Obeid, LM. Ceramidases: regulators of cellular responses mediated by ceramide, sphingosine, and sphingosine-1-phosphate. Biochim Biophys Acta. 2008;1781(9):424–434.
  • 25
    Seitz, AP, Grassmé, H, Edwards, MJ, Pewzner-Jung, Y, Gulbins, E. Ceramide and sphingosine in pulmonary infections. Biol Chem. 2015;396(6-7):611–620.
  • 26
    Chakravarthy, H, Navitskaya, S, O’Reilly, S. Role of acid sphingomyelinase in shifting the balance between pro-inflammatory and reparative bone marrow cells in diabetic retinopathy. Stem Cells. 2016;34(4):972–983.
  • 27
    Caretti, A., Torelli, R, Perdoni. Inhibition of ceramide de novo synthesis by myriocin produces the double effect of reducing pathological inflammation and exerting antifungal activity against A. fumigatus airways infection. Biochim Biophys Acta. 2016;1860(6):1089–1097.
  • 28
    Petrache, I., Berdyshev, EV. Ceramide signaling and metabolism in pathophysiological states of the lung. Annu Rev Physiol. 2016;78:463–480.
  • 29
    Pewzner-Jung, Y, Tavakoli Tabazavareh, S, Grassmé, H. Sphingoid long chain bases prevent lung infection by Pseudomonas aeruginosa EMBO Mol Med. 2014;6(9):1205–12014.
  • 30
    Vitner, EB, Farfel-Becker, T, Eilam, R, Biton, I, Futerman, AH. Contribution of brain inflammation to neuronal cell death in neuronopathic forms of Gaucher’s disease. Brain. 2012;135(pt 6):1724–1735.
  • 31
    Parkinson-Lawrence, EJ, Shandala, T, Prodoehl, M, Plew, R, Borlace, GN, Brooks, DA. Lysosomal storage diseases: revealing lysosomal function and physiology. Physiology. 2010;25(2):102–115.
  • 32
    Barak, V, Acker, M, Nisman, B. Cytokines in Gaucher’s disease. Eur Cytokine Netw. 1999;10(2):205–210.
  • 33
    Allen, MJ, Myer, BJ, Khokher, AM, Rushton, N, Cox, TM. Pro-inflammatory cytokines and the pathogenesis of Gaucher’s disease: increased release of interleukin-6 and interleukin-10. QJM. 1997;90(1):19–25.
  • 34
    Hollak, CE., Evers, L., Aerts, JM., van Oers, MH. Elevated levels of M-CSF, sCD14 and IL8 in type 1 Gaucher disease. Blood Cells Mol Dis. 1997;23(2):201–212.
  • 35
    Linari, S, Castaman, G. Hematological manifestations and complications of Gaucher disease. Expert Rev Hematol. 2016;9(1):51–58.
  • 36
    Machaczka, M, Lerner, R, Klimkowska, M, Hägglund, H. Treatment of multiple myeloma in patients with Gaucher disease. Am J Hematol. 2009;84(10):694–696.
  • 37
    Aflaki, E, Moaven, N, Borger, DK. Lysosomal storage and impaired autophagy lead to inflammasome activation in Gaucher macrophages. Aging Cell. 2016;15(1):77–88.
  • 38
    Kartha, RV, Elnashar, G, Hovde, L, Mishra, U, Utz, J, Cloyd, JC. Characterization of oxidative stress and inflammation in type 1 Gaucher disease-method evaluation and preliminary analysis. Mol Genet Metab. 2016;117(2):S65.
  • 39
    Macedo, MF, Quinta, R, Pereira, CS, Sa Miranda, MC. Enzyme replacement therapy partially prevents invariant Natural Killer T cell deficiency in the Fabry disease mouse model. Mol Genet Metab. 2012;106(1):83–91.
  • 40
    Pereira, CS, Azevedo, O, Maia, ML, Dias, AF, Sa-Miranda, C, Macedo, MF. Invariant natural killer T cells are phenotypically and functionally altered in Fabry disease. Mol Genet Metab. 2013;108(4):241–248.
  • 41
    Rozenfeld, P, Agriello, E, De Francesco, N, Martinez, P, Fossati, C. Leukocyte perturbation associated with Fabry disease. J Inherit Metab Dis. 2009;32(suppl 1):S67–S77.
  • 42
    De Francesco, PN, Mucci, JM, Ceci, R, Fossati, CA, Rozenfeld, PA. Fabry disease peripheral blood immune cells release inflammatory cytokines: role of globotriaosylceramide. Mol Genet Metab. 2013;109(1):93–99.
  • 43
    Jou, I, Lee, JH, Park, SY, Yoon, HJ, Joe, EH, Park, EJ. Gangliosides trigger inflammatory responses via TLR4 in brain glia. Am J Pathol. 2006;168(5):1619–1630.
  • 44
    Segura, T, Ayo-Martín, O, Gómez-Fernandez, I, Andrés, C, Barba, MA, Vivancos, J. Cerebral hemodynamics and endothelial function in patients with Fabry disease. BMC Neurol. 2013;13:170.
  • 45
    Biancini, GB, Vanzin, CS, Rodrigues, DB. Globotriaosylceramide is correlated with oxidative stress and inflammation in Fabry patients treated with enzyme replacement therapy. Biochim Biophys Acta. 2012;1822(2):226–232.
  • 46
    Liebau, MC, Braun, F, H?¶pker, K. Dysregulated autophagy contributes to podocyte damage in Fabry’s disease. PLoS One. 2013;8(5):e63506.
  • 47
    Frohbergh, M, He, X, Schuchman, EH. The molecular medicine of acid ceramidase. Biol Chem. 2015;396(6-7):759–765.
  • 48
    Alayoubi, AM., Wang, JC., Au, BC. Systemic ceramide accumulation leads to severe and varied pathological consequences. EMBO Mol Med. 2013;5(6):827–842.
  • 49
    Wada, R., Tifft, CJ., Proia, RL. Microglial activation precedes acute neurodegeneration in Sandhoff disease and is suppressed by bone marrow transplantation. Proc Natl Acad Sci U S A. 2000;97(20):10954–10959.
  • 50
    Wu, YP, Proia, RL. Deletion of macrophage-inflammatory protein 1 alpha retards neurodegeneration in Sandhoff disease mice. Proc Natl Acad Sci U S A. 2004;101(22):8425–8430.
  • 51
    Groh, J, Ribechini, E, Stadler, D, Schilling, T, Lutz, MB, Martini, R. Sialoadhesin promotes neuroinflammation-related disease progression in two mouse models of CLN disease. Glia. 2016;64(5):792–809.
  • 52
    Bosch, ME., Kielian, T. Neuroinflammatory paradigms in lysosomal storage diseases. Front Neurosci. 2015;9:417.
  • 53
    Seo, CH., Kim, JB. Therapeutic potential of resveratrol in type I Gaucher disease. Phytother Res. 2015;29(6):835–839.
  • 54
    Yoo, S., Kim, JB. Anti-apoptotic and beneficial metabolic activities of resveratrol in type II Gaucher disease. Biol Pharm Bull. 2015;38(6):913–918.
  • 55
    Dechecchi, MC., Nicolis, E., Norez, C. Anti-inflammatory effect of miglustat in bronchial epithelial cells. J Cyst Fibros. 2008;7(6):555–565.
  • 56
    Williams, IM., Wallom, KL., Smith, DA., Al Eisa, N., Smith, C., Platt, FM. Improved neuroprotection using miglustat, curcumin and ibuprofen as a triple combination therapy in Niemann-Pick disease type C1 mice. Neurobiol Dis. 2014;67:9–17.
  • 57
    Stein, A., Stroobants, S., Gieselmann, V., D’Hooge, R., Matzner, U. Anti-inflammatory therapy with simvastatin improves neuroinflammation and CNS function in a mouse model of metachromatic leukodystrophy. Mol Ther. 2015;23(7):1160–1168.
  • 58
    Rafi, MA., Rao, HZ., Luzi, P., Wenger, DA. Long-term improvements in lifespan and pathology in CNS and PNS after BMT plus one intravenous injection of AAVrh10-GALC in Twitcher mice. Mol Ther. 2015;23(11):1681–1690.
  • 59
    Christina, R, Mikulka, Hongwei, Q. Reducing neuroinflammation in globoid cell leukodystrophy by targeting the JAK/STAT pathway. Mol Genet Metab. 2016;117(2):S80.
  • 60
    Richard, M, Arfi, A, Rhinn, H, Gandolphe, C, Scherman, D. Identification of new markers for neurodegeneration process in the mouse model of Sly disease as revealed by expression profiling of selected genes. J Neurosci Res. 2008;86(15):3285–3294.
  • 61
    Arfi, A, Richard, M, Gandolphe, C, Bonnefont-Rousselot, D, Thérond, P, Scherman, D. Neuroinflammatory and oxidative stress phenomena in MPS IIIA mouse model: the positive effect of long-term aspirin treatment. Mol Genet Metab. 2011;103(1):18–25.
  • 62
    Ausseil, J., Desmaris, N., Bigou, S. Early neurodegeneration progresses independently of microglial activation by heparan sulfate in the brain of mucopolysaccharidosis IIIB mice. PLoS One. 2008;3(5):e2296.
  • 63
    Simonaro, CM., Guo, N., Schuchman, EH. Pentosan polysulfate and neuroinflammation in mice with mucopolysaccharidosis type IIIA mice. Mol Genet Metab. 2016;117(2):S106.
  • 64
    Simonaro, CM, D’Angelo, M, Haskins, ME, Schuchman, EH. Joint and bone disease in mucopolysaccharidoses VI and VII: identification of new therapeutic targets and biomarkers using animal models. Pediatr Res. 2005;57(5 pt 1):701–707.
  • 65
    Simonaro, CM, Ge, Y, He, X, Jepsen, KJ, Schuchman, EH. Involvement of the toll-like receptor 4 pathway and the use of anti-TNF-alpha agents for treatment in the mucopolysaccharidoses. Proc Natl Acad Sci U S A. 2010;107(1):222–227.
  • 66
    Polgreen, LE, Vehe, RK, Rudser, K. Elevated TNF-?± is associated with pain and physical disability in mucopolysaccharidosis types I, II, and VI. Mol Genet Metab. 2016;117(4):427–430.
  • 67
    Donida, B, Marchetti, DP, Biancini, GB. Oxidative stress and inflammation in mucopolysaccharidosis type IVA patients treated with enzyme replacement therapy. Biochim Biophys Acta. 2015;1852(5):1012–10
  • 68
    Hennermann, JB, Goekce, S, Solymon, A, Mengel, E, Schuchman, EH, Simonaro, CM. Treatment with pentosan polysulphate in patients with mucopolysaccharidosis type I: results from an open label, randomized, monocentric phase 2 study. Mol Genet Metab. 2016;117(2):S56.
  • 69
    Orii, K, Tomatsu, S, Suzuki, Y. Safety study of sodium pentosan polysulfate for adult patients with mucopolysaccharidosis type II. Mol Genet Metab. 2016;117(2):S56.

Publication Dates

  • Publication in this collection
    30 May 2019
  • Date of issue
    2016

History

  • Received
    18 Apr 2016
  • Accepted
    25 Apr 2016
Latin American Society Inborn Errors and Neonatal Screening (SLEIMPN); Instituto Genética para Todos (IGPT) Rua Ramiro Barcelos, 2350, CEP: 90035-903, Porto Alegre, RS - Brasil, Tel.: 55-51-3359-6338, Fax: 55-51-3359-8010 - Porto Alegre - RS - Brazil
E-mail: rgiugliani@hcpa.edu.br