Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
Add more filters

Publication year range
1.
Blood ; 137(5): 690-701, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33232973

ABSTRACT

Transfusion-related acute lung injury (TRALI) is a hazardous transfusion complication with an associated mortality of 5% to 15%. We previously showed that stored (5 days) but not fresh platelets (1 day) cause TRALI via ceramide-mediated endothelial barrier dysfunction. As biological ceramides are hydrophobic, extracellular vesicles (EVs) may be required to shuttle these sphingolipids from platelets to endothelial cells. Adding to complexity, EV formation in turn requires ceramide. We hypothesized that ceramide-dependent EV formation from stored platelets and EV-dependent sphingolipid shuttling induces TRALI. EVs formed during storage of murine platelets were enumerated, characterized for sphingolipids, and applied in a murine TRALI model in vivo and for endothelial barrier assessment in vitro. Five-day EVs were more abundant, had higher long-chain ceramide (C16:0, C18:0, C20:0), and lower sphingosine-1-phosphate (S1P) content than 1-day EVs. Transfusion of 5-day, but not 1-day, EVs induced characteristic signs of lung injury in vivo and endothelial barrier disruption in vitro. Inhibition or supplementation of ceramide-forming sphingomyelinase reduced or enhanced the formation of EVs, respectively, but did not alter the injuriousness per individual EV. Barrier failure was attenuated when EVs were abundant in or supplemented with S1P. Stored human platelet 4-day EVs were more numerous compared with 2-day EVs, contained more long-chain ceramide and less S1P, and caused more endothelial cell barrier leak. Hence, platelet-derived EVs become more numerous and more injurious (more long-chain ceramide, less S1P) during storage. Blockade of sphingomyelinase, EV elimination, or supplementation of S1P during platelet storage may present promising strategies for TRALI prevention.


Subject(s)
Extracellular Vesicles/physiology , Platelet Transfusion/adverse effects , Sphingolipids/metabolism , Transfusion-Related Acute Lung Injury/etiology , Animals , Blood Platelets/ultrastructure , Blood Preservation , Ceramides/metabolism , Endothelial Cells/physiology , Endotoxins/toxicity , Humans , Lysophospholipids/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/physiology , Sphingosine/analogs & derivatives , Sphingosine/physiology , Transfusion-Related Acute Lung Injury/metabolism , Transfusion-Related Acute Lung Injury/prevention & control
2.
Am J Respir Cell Mol Biol ; 64(5): 629-640, 2021 05.
Article in English | MEDLINE | ID: mdl-33662226

ABSTRACT

Deficiency of ASM (acid sphingomyelinase) causes the lysosomal storage Niemann-Pick disease (NPD). Patients with NPD type B may develop progressive interstitial lung disease with frequent respiratory infections. Although several investigations using the ASM-deficient (ASMKO) mouse NPD model revealed inflammation and foamy macrophages, there is little insight into the pathogenesis of NPD-associated lung disease. Using ASMKO mice, we report that ASM deficiency is associated with a complex inflammatory phenotype characterized by marked accumulation of monocyte-derived CD11b+ macrophages and expansion of airspace/alveolar CD11c+ CD11b- macrophages, both with increased size, granularity, and foaminess. Both the alternative and classical pathways were activated, with decreased in situ phagocytosis of opsonized (Fc-coated) targets, preserved clearance of apoptotic cells (efferocytosis), secretion of Th2 cytokines, increased CD11c+/CD11b+ cells, and more than a twofold increase in lung and plasma proinflammatory cytokines. Macrophages, neutrophils, eosinophils, and noninflammatory lung cells of ASMKO lungs also exhibited marked accumulation of chitinase-like protein Ym1/2, which formed large eosinophilic polygonal Charcot-Leyden-like crystals. In addition to providing insight into novel features of lung inflammation that may be associated with NPD, our report provides a novel connection between ASM and the development of crystal-associated lung inflammation with alterations in macrophage biology.


Subject(s)
Glycoproteins/immunology , Lysophospholipase/immunology , Macrophages, Alveolar/immunology , Macrophages/immunology , Niemann-Pick Disease, Type A/immunology , Niemann-Pick Disease, Type B/immunology , Pneumonia/immunology , Sphingomyelin Phosphodiesterase/immunology , Animals , CD11 Antigens/genetics , CD11 Antigens/immunology , CD11b Antigen/genetics , CD11b Antigen/immunology , Cell Size , Chitinases/genetics , Chitinases/immunology , Disease Models, Animal , Eosinophils/immunology , Eosinophils/pathology , Female , Gene Expression , Glycoproteins/genetics , Humans , Lectins/genetics , Lectins/immunology , Lung/immunology , Lung/pathology , Lysophospholipase/genetics , Macrophages/pathology , Macrophages, Alveolar/pathology , Male , Mice , Mice, Knockout , Neutrophils/immunology , Neutrophils/pathology , Niemann-Pick Disease, Type A/enzymology , Niemann-Pick Disease, Type A/genetics , Niemann-Pick Disease, Type A/pathology , Niemann-Pick Disease, Type B/enzymology , Niemann-Pick Disease, Type B/genetics , Niemann-Pick Disease, Type B/pathology , Phagocytosis , Pneumonia/enzymology , Pneumonia/genetics , Pneumonia/pathology , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Th1-Th2 Balance/genetics , beta-N-Acetylhexosaminidases/genetics , beta-N-Acetylhexosaminidases/immunology
3.
Cancer Sci ; 112(9): 3437-3454, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34152672

ABSTRACT

Metastasis is the main cause of death in individuals with cancer. Immune checkpoint blockade (ICB) can potentially reverse CD8+ cytotoxic T lymphocytes (CTLs) dysfunction, leading to significant remission in multiple cancers. However, the mechanism underlying the development of CTL exhaustion during metastatic progression remains unclear. Here, we established an experimental pulmonary metastasis model with melanoma cells and discovered a critical role for melanoma-released exosomes in metastasis. Using genetic knockdown of nSMase2 and Rab27a, 2 key enzymes for exosome secretion, we showed that high levels of effector-like tumor-specific CD8+ T cells with transitory exhaustion, instead of terminal exhaustion, were observed in mice without exosomes; these cells showed limited inhibitory receptors and strong proliferation and cytotoxicity. Mechanistically, the immunosuppression of exosomes depends on exogenous PD-L1, which can be largely rescued by pretreatment with antibody blockade. Notably, we also found that exosomal PD-L1 acts as a promising predictive biomarker for ICB therapies during metastasis. Together, our findings suggest that exosomal PD-L1 may be a potential immunotherapy target, suggesting a new curative therapy for tumor metastasis.


Subject(s)
B7-H1 Antigen/metabolism , Exosomes/metabolism , Immune Tolerance , Lung Neoplasms/blood , Lung Neoplasms/secondary , Melanoma/metabolism , Melanoma/pathology , T-Lymphocytes, Cytotoxic/immunology , Adoptive Transfer/methods , Aged , Animals , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Gene Knockdown Techniques , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Male , Melanoma/immunology , Mice , Mice, Inbred C57BL , Middle Aged , Signal Transduction/drug effects , Signal Transduction/genetics , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Treatment Outcome , rab27 GTP-Binding Proteins/deficiency , rab27 GTP-Binding Proteins/genetics
4.
Int J Mol Sci ; 22(23)2021 Nov 28.
Article in English | MEDLINE | ID: mdl-34884674

ABSTRACT

Acid sphingomyelinase deficiency (ASMD) is a lysosomal storage disease caused by deficient activity of acid sphingomyelinase (ASM) enzyme, leading to the accumulation of varying degrees of sphingomyelin. Lipid storage leads to foam cell infiltration in tissues, and clinical features including hepatosplenomegaly, pulmonary insufficiency and in some cases central nervous system involvement. ASM enzyme replacement therapy is currently in clinical trial being the first treatment addressing the underlying pathology of the disease. Therefore, presently, it is critical to better comprehend ASMD to improve its diagnose and monitoring. Lung disease, including recurrent pulmonary infections, are common in ASMD patients. Along with lung disease, several immune system alterations have been described both in patients and in ASMD animal models, thus highlighting the role of ASM enzyme in the immune system. In this review, we summarized the pivotal roles of ASM in several immune system cells namely on macrophages, Natural Killer (NK) cells, NKT cells, B cells and T cells. In addition, an overview of diagnose, monitoring and treatment of ASMD is provided highlighting the new enzyme replacement therapy available.


Subject(s)
Lysosomal Storage Diseases/immunology , Sphingomyelin Phosphodiesterase/deficiency , Animals , Enzyme Replacement Therapy , Humans , Lung Diseases/enzymology , Lysosomal Storage Diseases/diagnosis , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelin Phosphodiesterase/immunology
5.
Basic Res Cardiol ; 115(6): 64, 2020 10 14.
Article in English | MEDLINE | ID: mdl-33057972

ABSTRACT

By cleaving sphingomyelin into ceramide, which is an essential component of plasma membrane microdomains, acid sphingomyelinase (Asm) pivotally controls cell signaling. To define how the activation of the Asm/ceramide pathway, which occurs within seconds to minutes upon stress stimuli, influences brain ischemia/reperfusion (I/R) injury, we exposed male and female wildtype mice carrying both alleles of Asm's gene sphingomyelinase phosphodiesterase-1 (Smpd1+/+), heterozygously Asm-deficient mice (Smpd1+/-) and homozygously Asm-deficient mice (Smpd1-/-) of different age (8, 12 or 16 weeks) to 30, 60 or 90 min intraluminal middle cerebral artery occlusion (MCAO). For studying the contribution of brain-invading polymorphonuclear neutrophils (PMN) to I/R injury, PMNs were depleted by delivery of a PMN-specific Ly6G antibody. In male and female mice exposed to 30 min, but not 60 or 90 min MCAO, homozygous Smpd1-/- consistently increased I/R injury, blood-brain barrier permeability and brain leukocyte and PMN infiltration, whereas heterozygous Smpd1+/- reduced I/R injury. Increased abundance of the intercellular leukocyte adhesion molecule ICAM-1 was noted on cerebral microvessels of Smpd1-/- mice. PMN depletion by anti-Ly6G delivery prevented the exacerbation of I/R injury in Smpd1-/- compared with wildtype mice and reduced brain leukocyte infiltrates. Our results show that Asm tempers leukocyte entry into the reperfused ischemic brain, thereby attenuating I/R injury.


Subject(s)
Brain/blood supply , Infarction, Middle Cerebral Artery/enzymology , Microvessels/enzymology , Neutrophil Infiltration , Neutrophils/enzymology , Reperfusion Injury/enzymology , Sphingomyelin Phosphodiesterase/deficiency , Animals , Disease Models, Animal , Female , Heterozygote , Homozygote , Infarction, Middle Cerebral Artery/genetics , Infarction, Middle Cerebral Artery/pathology , Intercellular Adhesion Molecule-1/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Microvessels/pathology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Reperfusion Injury/prevention & control , Sphingomyelin Phosphodiesterase/genetics , Time Factors
6.
Am J Med Genet A ; 182(12): 2891-2901, 2020 12.
Article in English | MEDLINE | ID: mdl-32946203

ABSTRACT

The aim of this study was to evaluate the oral, dental, and craniofacial features of individuals affected by the chronic forms of acid sphingomyelinase deficiency (ASMD). This study comprised a sample of adult and pediatric patients (n = 8) with chronic ASMD. The individuals underwent oral examinations to evaluate the occurrence of caries, as well as full-mouth periodontal examinations, to assess the occurrence and severity of periodontal diseases. Panoramic and profile radiographs were obtained to analyze dental conditions and craniofacial parameters. Participants also answered questionnaires to identify systemic impairment, parafunctional habits, and bruxism. Dental anomalies of size, shape, and number were found, with agenesis and microdontia being the predominant findings. The average of caries experience was 11.75 (±8.1). Only one patient had periodontal health and all adult individuals had periodontitis at different stages and degrees. Bruxism was found in 87.5% of the sample. The convex profile and maxillary and mandibular retrusion were the most relevant findings in the cephalometric analysis. It is concluded that individuals with chronic ASMD, in addition to several systemic manifestations, present significant modifications in their oral health, from a greater occurrence of dental anomalies, caries, periodontal disease, in addition to skeletal changes.


Subject(s)
Bruxism/pathology , Craniofacial Abnormalities/pathology , Mouth Diseases/pathology , Niemann-Pick Disease, Type B/complications , Periodontal Diseases/pathology , Sphingomyelin Phosphodiesterase/deficiency , Tooth Abnormalities/pathology , Adolescent , Adult , Bruxism/etiology , Child , Craniofacial Abnormalities/etiology , Female , Humans , Male , Middle Aged , Mouth Diseases/etiology , Niemann-Pick Disease, Type B/enzymology , Periodontal Diseases/etiology , Prognosis , Tooth Abnormalities/etiology , Young Adult
7.
Cell Microbiol ; 21(11): e13065, 2019 11.
Article in English | MEDLINE | ID: mdl-31155842

ABSTRACT

Acid sphingomyelinase (ASM) is a lysosomal enzyme that cleaves the phosphorylcholine head group of sphingomyelin, generating ceramide. Recessive mutations in SMPD1, the gene encoding ASM, cause Niemann-Pick Disease Types A and B. These disorders are attributed not only to lipid accumulation inside lysosomes but also to changes on the outer leaflet of the plasma membrane, highlighting an extracellular role for ASM. Secretion of ASM occurs under physiological conditions, and earlier studies proposed two forms of the enzyme, one resident in lysosomes and another form that would be diverted to the secretory pathway. Such differential intracellular trafficking has been difficult to explain because there is only one SMPD1 transcript that generates an active enzyme, found primarily inside lysosomes. Unexpectedly, studies of cell invasion by the protozoan parasite Trypanosoma cruzi revealed that conventional lysosomes can fuse with the plasma membrane in response to elevations in intracellular Ca2+ , releasing their contents extracellularly. ASM exocytosed from lysosomes remodels the outer leaflet of the plasma membrane, promoting parasite invasion and wound repair. Here, we discuss the possibility that ASM release during lysosomal exocytosis, in response to various forms of stress, may represent a major source of the secretory form of this enzyme.


Subject(s)
Cell Membrane/parasitology , Lysosomes/enzymology , Sphingomyelin Phosphodiesterase/metabolism , Trypanosoma cruzi/pathogenicity , Animals , Bodily Secretions/radiation effects , Calcium/metabolism , Cell Membrane/metabolism , Cell Membrane/pathology , Ceramides/metabolism , Exocytosis , Humans , Lysosomes/metabolism , Niemann-Pick Disease, Type A/enzymology , Niemann-Pick Disease, Type B/enzymology , Protein Transport , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Sphingomyelins/metabolism , Trypanosoma cruzi/metabolism
8.
J Pediatr Hematol Oncol ; 42(6): e499-e502, 2020 08.
Article in English | MEDLINE | ID: mdl-30870388

ABSTRACT

BACKGROUND: Typically, patients with Acid Sphingomyelinase Deficiency (ASMD) because of p.Arg610del mutation, have mild phenotype with normal linear growth. OBSERVATION: We reported the case of 2 Tunisian brothers who have been referred for splenomegaly, polyadenopathies, pubertal, and growth delay. Molecular testing of SMPD1 gene revealed the presence of a homozygous p.Arg610del mutation. Lysosphingomyelin and its isoform-509 were both increased confirming ASMD for both cases. Growth hormone deficiency was highly suspected but growth hormone response after stimulating tests was acceptable for both patients. CONCLUSIONS: There is no correlation between phenotype-genotype in case of p.Arg610del mutation that could be associated to a severe delay of growth.


Subject(s)
Developmental Disabilities/pathology , Homozygote , Mutation , Niemann-Pick Diseases/complications , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Adolescent , Adult , Developmental Disabilities/etiology , Humans , Male , Niemann-Pick Diseases/genetics , Niemann-Pick Diseases/pathology , Phenotype , Prognosis , Siblings , Young Adult
9.
Oral Dis ; 26(3): 637-646, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31883406

ABSTRACT

BACKGROUND: Mutation of the gene for acid sphingomyelinase (ASMase) causes Niemann-Pick disease. However, the effect of ASMase deficiency on periodontal health is unknown. Periodontal disease is a disease resulting from infection and inflammation of periodontal tissue and alveolar bone that support the teeth. The goal of this study was to determine the role of ASMase deficiency in periodontal inflammation and alveolar bone loss. METHODS: We induced periodontitis in wild-type and ASMase-deficient (ASMase-/- ) mice with periodontal lipopolysaccharide (LPS) injection and compared the alveolar bone loss and periodontal inflammation between these mice. RESULTS: Results showed that ASMase deficiency did not significantly change metabolic parameters, but exacerbated LPS-induced alveolar bone loss, osteoclastogenesis, and periodontal tissue inflammation. To understand the mechanisms by which ASMase deficiency aggravates LPS-induced periodontitis, we analyzed sphingolipids in periodontal tissues. Results showed that ASMase deficiency led to increases in not only sphingomyelin, but also ceramide (CER), a bioactive sphingolipid known to promote inflammation. Results further showed that ASMase deficiency increased CER de novo synthesis. CONCLUSION: ASMase deficiency exacerbated LPS-induced alveolar bone loss and periodontal inflammation. ASMase deficiency leads to an unexpected CER increase by stimulating de novo synthesis CER, which is likely to be involved in the ASMase deficiency-exacerbated periodontitis.


Subject(s)
Alveolar Bone Loss/complications , Niemann-Pick Disease, Type A/complications , Periodontitis/complications , Animals , Disease Models, Animal , Lipopolysaccharides , Mice , Mice, Knockout , Periodontitis/chemically induced , Sphingomyelin Phosphodiesterase/deficiency
10.
J Lipid Res ; 60(3): 609-623, 2019 03.
Article in English | MEDLINE | ID: mdl-30662008

ABSTRACT

Traumatic brain injury (TBI) is one of the leading causes of disability worldwide and a prominent risk factor for neurodegenerative diseases. The expansion of nervous tissue damage after the initial trauma involves a multifactorial cascade of events, including excitotoxicity, oxidative stress, inflammation, and deregulation of sphingolipid metabolism that further mitochondrial dysfunction and secondary brain damage. Here, we show that a posttranscriptional activation of an acid sphingomyelinase (ASM), a key enzyme of the sphingolipid recycling pathway, resulted in a selective increase of sphingosine in mitochondria during the first week post-TBI that was accompanied by reduced activity of mitochondrial cytochrome oxidase and activation of the Nod-like receptor protein 3 inflammasome. TBI-induced mitochondrial abnormalities were rescued in the brains of ASM KO mice, which demonstrated improved behavioral deficit recovery compared with WT mice. Furthermore, an elevated autophagy in an ASM-deficient brain at the baseline and during the development of secondary brain injury seems to foster the preservation of mitochondria and brain function after TBI. Of note, ASM deficiency attenuated the early stages of reactive astrogliosis progression in an injured brain. These findings highlight the crucial role of ASM in governing mitochondrial dysfunction and brain-function impairment, emphasizing the importance of sphingolipids in the neuroinflammatory response to TBI.


Subject(s)
Brain Injuries/pathology , Brain Injuries/physiopathology , Gene Knockout Techniques , Mitochondria/pathology , Recovery of Function , Sphingomyelin Phosphodiesterase/deficiency , Animals , Brain Injuries/enzymology , Brain Injuries/genetics , Cognition , Enzyme Activation , Mice , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Sphingomyelin Phosphodiesterase/genetics
11.
Immunology ; 156(4): 373-383, 2019 04.
Article in English | MEDLINE | ID: mdl-30556232

ABSTRACT

Acid sphingomyelinase (ASM) is one of the enzymes that catalyzes the breakdown of sphingomyelin to ceramide and phosphorylcholine. In this study, we aimed at elucidating the role of ASM in allergic asthma. We used an ovalbumin-induced murine model of asthma where we compared wild-type and ASM-deficient mice. In wild-type mice, secretory ASM activity in the bronchoalveolar lavage fluid was increased in the acute ovalbumin model, but not in a tolerogenic model. Furthermore, in the absence of ASM, the serum IgE level was reduced, compared with wild-type mice, while an accumulation of interstitial macrophages and foreign antigen-induced regulatory T cells along with exhausted CD4+ PD1+ T cells was observed in the lungs of ASM-/- mice. In conclusion, in the absence of ASM, we observed an accumulation of immunosuppressive antigen-induced regulatory T cells expressing Foxp3 and CTLA4 in the lung as well as multinucleated interstitial macrophages and exhausted CD4+ PD1+ T cells associated with inhibition of serum IgE in asthma.


Subject(s)
Asthma/enzymology , Asthma/immunology , Sphingomyelin Phosphodiesterase/metabolism , Animals , Asthma/chemically induced , Disease Models, Animal , Female , Immunoglobulin E/blood , Immunoglobulin E/immunology , Immunoglobulin E/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin , Sphingomyelin Phosphodiesterase/deficiency , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 316(4): L679-L690, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30702345

ABSTRACT

Heart failure with reduced ejection fraction (HFREF) increases neutral sphingomyelinase (NSMase) activity and mitochondrial reactive oxygen species (ROS) emission and causes diaphragm weakness. We tested whether a systemic pharmacological NSMase inhibitor or short-hairpin RNA (shRNA) targeting NSMase isoform 3 (NSMase3) would prevent diaphragm abnormalities induced by HFREF caused by myocardial infarction. In the pharmacological intervention, we used intraperitoneal injection of GW4869 or vehicle. In the genetic intervention, we injected adeno-associated virus serotype 9 (AAV9) containing shRNA targeting NSMase3 or a scrambled sequence directly into the diaphragm. We also studied acid sphingomyelinase-knockout mice. GW4869 prevented the increase in diaphragm ceramide content, weakness, and tachypnea caused by HFREF. For example, maximal specific forces (in N/cm2) were vehicle [sham 31 ± 2 and HFREF 26 ± 2 ( P < 0.05)] and GW4869 (sham 31 ± 2 and HFREF 31 ± 1). Respiratory rates were (in breaths/min) vehicle [sham 61 ± 3 and HFREF 84 ± 11 ( P < 0.05)] and GW4869 (sham 66 ± 2 and HFREF 72 ± 2). AAV9-NSMase3 shRNA prevented heightening of diaphragm mitochondrial ROS and weakness [in N/cm2, AAV9-scrambled shRNA: sham 31 ± 2 and HFREF 27 ± 2 ( P < 0.05); AAV9-NSMase3 shRNA: sham 30 ± 1 and HFREF 30 ± 1] but displayed tachypnea. Both wild-type and ASMase-knockout mice with HFREF displayed diaphragm weakness. Our study suggests that activation of NSMase3 causes diaphragm weakness in HFREF, presumably through accumulation of ceramide and elevation in mitochondrial ROS. Our data also reveal a novel inhibitory effect of GW4869 on tachypnea in HFREF likely mediated by changes in neural control of breathing.


Subject(s)
Diaphragm/physiopathology , Heart Failure/physiopathology , Muscle Weakness/prevention & control , RNA, Small Interfering/genetics , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/genetics , Aniline Compounds/pharmacology , Animals , Benzylidene Compounds/pharmacology , Diaphragm/enzymology , Disease Models, Animal , Heart Failure/genetics , Heart Failure/therapy , Humans , Male , Mice , Mice, Knockout , Muscle Weakness/genetics , Muscle Weakness/physiopathology , Rats , Rats, Wistar , Sphingomyelin Phosphodiesterase/deficiency , Stroke Volume/genetics , Stroke Volume/physiology
13.
Neurosignals ; 27(S1): 20-31, 2019.
Article in English | MEDLINE | ID: mdl-31778303

ABSTRACT

BACKGROUND/AIMS: Multiple sclerosis (MS) is one of the most common autoimmune disorders of the central nervous system (CNS) and the leading cause of neurological disability among young adults in the Western world. We have previously shown that the acid sphingomyelinase plays an important role in the pathogenesis of experimental autoimmune encephalomyelitis (EAE), an animal model for multiple sclerosis. METHODS: We induced adoptively transferred EAE in wildtype and acid sphingomyelinase-deficient mice. In addition, we immunized mice with MOGaa35-55 to induce active EAE and treated the mice with amitriptyline, a functional inhibitor of the acid sphingomyelinase. We investigated symptoms of EAE, blood-brain barrier integrity and neuroinflammation. RESULTS: In the model of adoptively transferred EAE we demonstrate that expression of acid sphingomyelinase in the recipients rather than on transferred encephalitogenic T cells contributes to the clinical development of EAE symptoms. To test if pharmacological targeting of acid sphingomyelinase can be explored for the development of novel therapies for MS, we inhibited acid sphingomyelinase with amitriptyline in mice in which EAE was induced by active immunization. We demonstrate that pharmacological inhibition of acid sphingomyelinase using amitriptyline protects against the development of EAE and markedly attenuates the characteristic detrimental neuroinflammatory response. CONCLUSION: The studies identify the acid sphingomyelinase as a novel therapeutic target for treating MS patients.


Subject(s)
Amitriptyline/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/enzymology , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/deficiency , Adrenergic Uptake Inhibitors/pharmacology , Adrenergic Uptake Inhibitors/therapeutic use , Amitriptyline/pharmacology , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Sphingomyelin Phosphodiesterase/genetics
15.
Arterioscler Thromb Vasc Biol ; 38(7): 1479-1492, 2018 07.
Article in English | MEDLINE | ID: mdl-29794115

ABSTRACT

OBJECTIVE: Atherosclerosis is a chronic multifactorial and inflammatory disease of large and medium arteries and the leading cause of cardiovascular diseases worldwide. The aim of this study was to investigate whether and how the nSMase2 (type 2-neutral sphingomyelinase), a key enzyme of sphingolipid metabolism, may contribute to the development of atherosclerotic lesions. APPROACH AND RESULTS: The role of nSMase2 in atherosclerosis was investigated in Apoe-/-;Smpd3fro/fro mice, mutant for nSMase2, and in Apoe-/-;Smpd3+/+ mice intraperitoneally injected with GW4869, a pharmacological nSMase2 inhibitor. The defect or inhibition of nSMase2 resulted in a reduction of atherosclerotic lesions and a decrease in macrophage infiltration and lipid deposition, although cholesterolemia remained unchanged. nSMase2 inhibition decreased the inflammatory response of murine endothelial cells to oxLDL (oxidized low-density lipoprotein), as assessed by the significant reduction of MCP-1 (monocyte chemoattractant protein 1), ICAM-1 (intercellular adhesion molecule-1), and VCAM-1 (vascular cell adhesion molecule-1) mRNA expressions and macrophage recruitment. Likewise, in RAW264.7 or in macrophages isolated from Apoe-/-/Smpd3fro/fro or Apoe-/-/Smpd3+/+ mice stimulated by lipopolysaccharides, nSMase2 inhibition resulted in a decrease in the expression of inflammatory molecules. Mechanistically, the anti-inflammatory response resulting from nSMase2 inhibition involves Nrf2 (nuclear factor [erythroid-derived 2]-like 2 or NF-E2-related factor-2) activation in both endothelial cells and macrophages, as assessed by the lack of protective effect of GW4869 in endothelial cells silenced for Nrf2 by small interfering RNAs, and in lipopolysaccharide-stimulated macrophages issued from Nrf2-KO mice. CONCLUSIONS: The genetic deficiency or inhibition of nSMase2 strongly decreases the development of atherosclerotic lesions in Apoe-/- mice, by reducing inflammatory responses through a mechanism involving the Nrf2 pathway. Inhibitors of nSMase2 may, therefore, constitute a novel approach to slow down atherosclerosis progression.


Subject(s)
Aniline Compounds/pharmacology , Anti-Inflammatory Agents/pharmacology , Aorta/drug effects , Aortic Diseases/prevention & control , Atherosclerosis/prevention & control , Benzylidene Compounds/pharmacology , Enzyme Inhibitors/pharmacology , Inflammation/prevention & control , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Sphingomyelin Phosphodiesterase/deficiency , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/genetics , Aortic Diseases/pathology , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/pathology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Inflammation/enzymology , Inflammation/genetics , Inflammation/pathology , Inflammation Mediators/metabolism , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout, ApoE , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Plaque, Atherosclerotic , RAW 264.7 Cells , Signal Transduction/drug effects , Sphingomyelin Phosphodiesterase/genetics
16.
Int J Mol Sci ; 20(24)2019 Dec 11.
Article in English | MEDLINE | ID: mdl-31835809

ABSTRACT

Farber disease is a rare lysosomal storage disorder resulting from acid ceramidase deficiency and subsequent ceramide accumulation. No treatments for Farber disease are clinically available, and affected patients have a severely shortened lifespan. We have recently reported a novel acid ceramidase deficiency model that mirrors the human disease closely. Acid sphingomyelinase is the enzyme that generates ceramide upstream of acid ceramidase in the lysosomes. Using our acid ceramidase deficiency model, we tested if acid sphingomyelinase could be a potential novel therapeutic target for the treatment of Farber disease. A number of functional acid sphingomyelinase inhibitors are clinically available and have been used for decades to treat major depression. Using these as a therapeutic for Farber disease, thus, has the potential to improve central nervous symptoms of the disease as well, something all other treatment options for Farber disease can't achieve so far. As a proof-of-concept study, we first cross-bred acid ceramidase deficient mice with acid sphingomyelinase deficient mice in order to prevent ceramide accumulation. Double-deficient mice had reduced ceramide accumulation, fewer disease manifestations, and prolonged survival. We next targeted acid sphingomyelinase pharmacologically, to test if these findings would translate to a setting with clinical applicability. Surprisingly, the treatment of acid ceramidase deficient mice with the acid sphingomyelinase inhibitor amitriptyline was toxic to acid ceramidase deficient mice and killed them within a few days of treatment. In conclusion, our study provides the first proof-of-concept that acid sphingomyelinase could be a potential new therapeutic target for Farber disease to reduce disease manifestations and prolong survival. However, we also identified previously unknown toxicity of the functional acid sphingomyelinase inhibitor amitriptyline in the context of Farber disease, strongly cautioning against the use of this substance class for Farber disease patients.


Subject(s)
Farber Lipogranulomatosis/enzymology , Sphingomyelin Phosphodiesterase/deficiency , Acid Ceramidase/metabolism , Amitriptyline/pharmacology , Animals , Ceramides/metabolism , Cytokines/metabolism , Farber Lipogranulomatosis/pathology , Mice, Inbred C57BL , Sphingomyelin Phosphodiesterase/metabolism , Survival Analysis , Weight Gain/drug effects
17.
J Lipid Res ; 59(5): 795-804, 2018 05.
Article in English | MEDLINE | ID: mdl-29567647

ABSTRACT

Sphingolipids are key signaling lipids in cancer. Genome-wide studies have identified neutral SMase-2 (nSMase2), an enzyme generating ceramide from SM, as a potential repressor for hepatocellular carcinoma. However, little is known about the sphingolipids regulated by nSMase2 and their roles in liver tumor development. We discovered growth of spontaneous liver tumors in 27.3% (9 of 33) of aged male nSMase2-deficient (fro/fro) mice. Lipidomics analysis showed a marked increase of SM in the tumor. Unexpectedly, tumor tissues presented with more than a 7-fold increase of C16-ceramide, concurrent with upregulation of ceramide synthase 5. The fro/fro liver tumor, but not adjacent tissue, exhibited substantial accumulation of lipid droplets, suggesting that nSMase2 deficiency is associated with tumor growth and increased neutral lipid generation in the tumor. Tumor tissue expressed significantly increased levels of CD133 and EpCAM mRNA, two markers of liver cancer stem-like cells (CSCs) and higher levels of phosphorylated signal transducer and activator of transcription 3, an essential regulator of stemness. CD133(+) cells showed strong labeling for SM and ceramide. In conclusion, these results suggest that SMase-2 deficiency plays a role in the survival or proliferation of CSCs, leading to spontaneous tumors, which is associated with tumor-specific effects on lipid homeostasis.


Subject(s)
Liver Neoplasms/enzymology , Liver Neoplasms/pathology , Sphingomyelin Phosphodiesterase/deficiency , Animals , Cell Proliferation , Liver Neoplasms/genetics , Male , Mice , Mice, Knockout , Sphingomyelin Phosphodiesterase/genetics
18.
J Lipid Res ; 59(4): 596-606, 2018 04.
Article in English | MEDLINE | ID: mdl-29378782

ABSTRACT

Activation of the lysosomal ceramide-producing enzyme, acid sphingomyelinase (ASM), by various stresses is centrally involved in cell death and has been implicated in autophagy. We set out to investigate the role of the baseline ASM activity in maintaining physiological functions of lysosomes, focusing on the lysosomal nutrient-sensing complex (LYNUS), a lysosomal membrane-anchored multiprotein complex that includes mammalian target of rapamycin (mTOR) and transcription factor EB (TFEB). ASM inhibition with imipramine or sphingomyelin phosphodiesterase 1 (SMPD1) siRNA in human lung cells, or by transgenic Smpd1+/- haploinsufficiency of mouse lungs, markedly reduced mTOR- and P70-S6 kinase (Thr 389)-phosphorylation and modified TFEB in a pattern consistent with its activation. Inhibition of baseline ASM activity significantly increased autophagy with preserved degradative potential. Pulse labeling of sphingolipid metabolites revealed that ASM inhibition markedly decreased sphingosine (Sph) and Sph-1-phosphate (S1P) levels at the level of ceramide hydrolysis. These findings suggest that ASM functions to maintain physiological mTOR signaling and inhibit autophagy and implicate Sph and/or S1P in the control of lysosomal function.


Subject(s)
Autophagy/drug effects , Enzyme Inhibitors/pharmacology , Lysosomes/drug effects , Lysosomes/metabolism , Multiprotein Complexes/antagonists & inhibitors , Signal Transduction/drug effects , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , Animals , Cells, Cultured , Enzyme Inhibitors/chemistry , Humans , Imipramine/chemistry , Imipramine/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Multiprotein Complexes/metabolism , RNA, Small Interfering/chemistry , RNA, Small Interfering/pharmacology , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/metabolism
19.
J Lipid Res ; 59(10): 1841-1850, 2018 10.
Article in English | MEDLINE | ID: mdl-30087205

ABSTRACT

Intestinal alkaline SMase (Alk-SMase) cleaves phosphocholine from SM, platelet-activating factor (PAF), and lysophosphatidylcholine. We recently found that colitis-associated colon cancer was 4- to 5-fold enhanced in Alk-SMase KO mice. Here, we further studied the pathogenesis of colitis induced by dextran sulfate sodium (DSS) in WT and KO mice. Compared with WT mice, KO mice demonstrated greater body weight loss, more severe bloody diarrhea, broader inflammatory cell infiltration, and more serious epithelial injury. Higher levels of PAF and lower levels of interleukin (IL)10 were identified in KO mice 2 days after DSS treatment. A greater and progressive increase of lysophosphatidic acid (LPA) was identified. The change was associated with increased autotaxin expression in both small intestine and colon, which was identified by immunohistochemistry study, Western blot, and sandwich ELISA. The upregulation of autotaxin coincided with an early increase of PAF. IL6 and TNFα were increased in both WT and KO mice. At the later stage (day 8), significant decreases in IL6, IL10, and PAF were identified, and the decreases were greater in KO mice. In conclusion, deficiency of Alk-SMase enhances DSS-induced colitis by mechanisms related to increased autotaxin expression and LPA formation. The early increase of PAF might be a trigger for such reactions.


Subject(s)
Colitis/chemically induced , Colitis/metabolism , Dextran Sulfate/adverse effects , Phosphoric Diester Hydrolases/metabolism , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/genetics , Up-Regulation/drug effects , Animals , Body Weight/drug effects , Body Weight/genetics , Carrier Proteins/biosynthesis , Colitis/enzymology , Colitis/genetics , Colon/drug effects , Colon/metabolism , Colon/pathology , Gene Knockout Techniques , Interleukin-10/metabolism , Interleukin-6/metabolism , Intestine, Small/drug effects , Intestine, Small/metabolism , Intestine, Small/pathology , Lysophospholipids/biosynthesis , Mice , Mice, Inbred C57BL , Tumor Necrosis Factor-alpha/metabolism
20.
Biol Chem ; 399(10): 1175-1182, 2018 09 25.
Article in English | MEDLINE | ID: mdl-29908119

ABSTRACT

Acid sphingomyelinase (ASM) is the rate-limiting enzyme cleaving sphingomyelin into ceramide and phosphorylcholin. CD4+ Foxp3+ regulatory T (Treg) cells depend on CD28 signaling for their survival and function, a receptor that activates the ASM. Both, basal and CD28-induced ASM activities are higher in Treg cells than in conventional CD4+ T (Tconv) cells. In ASM-deficient (Smpd1-/-) as compared to wt mice, membranes of T cells contain 7-10-fold more sphingomyelin and two- to three-fold more ceramide, and are in a state of higher order than membranes of T cells from wt mice, which may facilitate their activation. Indeed, the frequency of Treg cells among CD4+ T cells in ASM-deficient mice and their suppressive activity in vitro are increased. Moreover, in vitro stimulation of ASM-deficient T cells in the presence of TGF-ß and IL-2 leads to higher numbers of induced Treg cells. Pharmacological inhibition of the ASM with a clinically used tricyclic antidepressant such as amitriptyline in mice or in tissue culture of murine or human T cells induces higher frequencies of Treg cells among CD4+ T cells within a few days. This fast alteration of the balance between T cell populations in vitro is due to the elevated cell death of Tconv cells and protection of the CD25high Treg cells by IL-2. Together, these findings suggest that ASM-inhibiting antidepressants, including a fraction of the serotonin re-uptake inhibitors (SSRIs), are moderately immunosuppressive and should be considered for the therapy of inflammatory and autoimmune disorders.


Subject(s)
Amitriptyline/pharmacology , Antidepressive Agents/pharmacology , Forkhead Transcription Factors/metabolism , Immunomodulation/drug effects , Sphingomyelin Phosphodiesterase/antagonists & inhibitors , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/therapy , Humans , Inflammation/therapy , Mice , Sphingomyelin Phosphodiesterase/deficiency , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL