1
|
Kaur P, Karuppuchamy T, Chilukuri A, Kim M, Urrete J, Shen Z, Saxon L, Lundborg LR, Mikulski Z, Jedlicka P, Rivera-Nieves J. S1P Lyase Inhibition Increased Intestinal S1P, Disrupted the Intestinal Barrier and Aggravated DSS-Induced Colitis. Inflamm Bowel Dis 2025:izaf030. [PMID: 39960746 DOI: 10.1093/ibd/izaf030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Indexed: 04/02/2025]
Abstract
BACKGROUND Sphingosine-1-phospate (S1P) receptor agonists (eg, ozanimod) desensitize migrating lymphocytes by irreversibly binding to S1P receptors (S1PR) and triggering their proteasomal degradation. Desensitized lymphocytes cannot sense S1P, therefore, halting lymphocyte recirculation. The S1P lyase (SPL) irreversibly degrades S1P and its inhibition disrupts the S1P gradient. We previously found that systemic SPL inhibitors induce central immunosuppression. Here, we examined whether SPL inhibition may attenuate colitis without systemic immunotoxicity. METHODS We first analyzed SPL expression and localization in mice using qRT-PCR and immunohistochemistry. SPL inhibitors 4-deoxypyridoxine hydrochloride (DOP) and 2-acetyl-4-(tetrahydroxybutyl) imidazole (THI) were used to inhibit SPL systemically, whereas a conditional intestinal epithelial cell (IEC)-specific SPL-deficient mouse was used to evaluate the effects of IEC-specific SPL inhibition on survival, disease activity, histological severity of dextran sulfate sodium-induced colitis, S1P levels, and intestinal permeability. RESULTS Sgpl1 mRNA transcripts and protein were ubiquitously expressed in gastrointestinal (GI) tract leukocytes and IEC. Systemic SPL inhibitors did not induce colitis by themselves but depleted CD4+ and CD8+ T cells from blood. However, contrary to its therapeutic effects on ileitis, systemic inhibition reduced survival, accelerated weight loss, worsened histopathological inflammation indices, and tissue damage. We then examined the effects of IEC-specific inhibition on peripheral cell counts and severity of colitis. We found that while it spared peripheral immunity, it similarly hastened colitis. Finally, we examined whether colitis acceleration was due to epithelial barrier compromise after disruption of the S1P gradient. We found that not only systemic but also IEC-specific SPL inhibition increased local S1P levels and led to IEC barrier compromise. CONCLUSION Homeostatic intestinal S1P levels are critical for the regulation of IEC barrier function. Further studies using adaptive immunity-based inflammatory bowel diseases (IBD) models are required to assess the translational value of IEC-specific SPL inhibition as a therapeutic target for human IBD.
Collapse
Affiliation(s)
- Prabhdeep Kaur
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
| | - Thangaraj Karuppuchamy
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA 92161, USA
| | - Amruth Chilukuri
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
| | - Margaret Kim
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA 92161, USA
| | - Josef Urrete
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
| | - Zining Shen
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
| | - Leo Saxon
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA 92161, USA
| | - Luke R Lundborg
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA 92161, USA
| | - Zbigniew Mikulski
- Histology and Microscopy Core, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Paul Jedlicka
- Department of Pathology, University of Colorado, Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jesús Rivera-Nieves
- Division of Gastroenterology, Department of Medicine, University of California San Diego, La Jolla, CA 92093-0063, USA
- Gastroenterology Section, San Diego VA Medical Center, La Jolla Village Drive, San Diego, CA 92161, USA
| |
Collapse
|
2
|
Zhang K, Zhang R, Zhang Y, Zhang M, Su H, Zhao F, Wang D, Cao G, Zhang Y. Regulation of LPS-Induced Inflammatory Responses in Bovine Mammary Epithelial Cells via TLR4-Mediated NF-κB and MAPK Signaling Pathways by Lactoferrin. Life (Basel) 2025; 15:69. [PMID: 39860009 PMCID: PMC11767191 DOI: 10.3390/life15010069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/05/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Lactoferrin (LF), a member of the transferrin family, is widely present in mammalian milk and other secretions, exhibiting anti-inflammatory, antibacterial, and anti-infective properties. Although the biological functions of LF have been extensively studied, there are few reports on its effects and molecular mechanisms concerning bovine mastitis caused by bacterial infection. This study used bovine mammary epithelial cells (BMECs) cultured in vitro as the research model. An inflammatory injury model was established by stimulating BMECs with LPS to investigate whether LF at different concentrations (10, 50, 100, and 200 μg·mL-1) could inhibit the inflammatory response before and after the onset of inflammation. The expression of inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α at both the gene and protein levels was detected using RT-qPCR and ELISA. Western blotting was employed to evaluate the phosphorylation levels in the inflammatory signaling pathways MAPK/P38/ERK and NF-κB/P65, while RT-qPCR was used to examine the impact on TLR4 receptor gene expression. The results display that pretreatment with LF prior to LPS-induced inflammation in BMECs reduced the expression of inflammatory cytokines IL-1β, IL-6, IL-8, and TNF-α at both the gene and protein levels (p < 0.05). LF also inhibited the phosphorylation of NF-κB/P65 and MAPK/P38/ERK signaling pathways and downregulated TLR4 receptor gene expression (p < 0.05). However, when LF was added after the onset of LPS-induced inflammation, inflammatory cytokine expression and phosphorylation levels in the NF-κB/P65 and MAPK/P38/ERK pathways remained elevated, along with high expression of the TLR4 receptor gene (p < 0.05). These findings show that LF can antagonize LPS-induced inflammatory responses in BMECs and reduce cytokine expression, exhibiting anti-inflammatory effects when administered before inflammation. Conversely, when LF is added post-inflammation, it appears to enhance cytokine expression, potentially promoting the recruitment of more cells or factors to resolve inflammation rapidly. Both effects are mediated through the TLR4 receptor and the NF-κB/P65 and MAPK/P38/ERK signaling pathways.
Collapse
Affiliation(s)
- Kai Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China; (K.Z.); (R.Z.); (Y.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Ruizhen Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China; (K.Z.); (R.Z.); (Y.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Yuanyuan Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China; (K.Z.); (R.Z.); (Y.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Min Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China; (K.Z.); (R.Z.); (Y.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Hong Su
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China; (K.Z.); (R.Z.); (Y.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Feifei Zhao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China; (K.Z.); (R.Z.); (Y.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Daqing Wang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China; (K.Z.); (R.Z.); (Y.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| | - Guifang Cao
- College of Life Sciences, Inner Mongolia University, Hohhot 010011, China
| | - Yong Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010010, China; (K.Z.); (R.Z.); (Y.Z.); (M.Z.); (H.S.); (F.Z.); (D.W.)
- Animal Embryo and Developmental Engineering Key Laboratory of Higher Education, Institutions of Inner Mongolia Autonomous Region, Hohhot 010011, China
- Inner Mongolia Autonomous Region Key Laboratory of Basic Veterinary Medicine, Hohhot 010011, China
| |
Collapse
|
3
|
Yilmaz A, Koca M, Ercan S, Acar OO, Boga M, Sen A, Kurt A. Amelioration potential of synthetic oxime chemical cores against multiple sclerosis and Alzheimer's diseases: Evaluation in aspects of in silico and in vitro experiments. J Mol Struct 2024; 1318:139193. [DOI: 10.1016/j.molstruc.2024.139193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
|
4
|
George N, Xiao J. Inhibiting sphingosine 1-phosphate lyase: From efficacy to mechanism. Neurobiol Dis 2024; 199:106585. [PMID: 38955289 DOI: 10.1016/j.nbd.2024.106585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 06/28/2024] [Accepted: 06/28/2024] [Indexed: 07/04/2024] Open
Abstract
Sphingosine-1 phosphate (S1P) is a lipid metabolite regulating diverse biological processes, including proliferation, differentiation, migration, and apoptosis, highlighting its physiological and therapeutic significance. Current S1P-based therapeutic approaches primarily focus on modulating the downstream signalling via targeting S1P receptors, however, this is challenged by incomplete receptor internalisation. Sphingosine-1-phosphate lyase (SPL) is a highly conserved enzyme that "gatekeeps" the final step of S1P degradation. Cognisant of the complex ligand and receptor interaction and dynamic metabolic networks, the selective modulation of SPL activity presents a new opportunity to regulate S1P biosynthesis and reveal its role in various systems. Over the past decade, an evolving effort has been made to identify new molecules that could block SPL activity in vitro or in vivo. This review focuses on summarising the current understanding of the reported SPL inhibitors identified through various screening approaches, discussing their efficacy in diverse model systems and the possible mechanism of action. Whilst effective modulation of S1P levels via inhibiting SPL is feasible, the specificity of those inhibitors remains inconclusive, presenting a clear challenge for future implications. Yet, none of the currently available SPL inhibitors is proven effective in elevating S1P levels within the central nervous system. This review article embraces future research focusing on investigating selective SPL inhibitors with high potency and possibly blood-brain-barrier permeability, which would aid the development of new S1P-based therapeutics for neurological disorders.
Collapse
Affiliation(s)
- Nelson George
- Department of Health Sciences and Biostatistics, School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia
| | - Junhua Xiao
- Department of Health Sciences and Biostatistics, School of Health Sciences, Swinburne University of Technology, Hawthorn, Victoria, Australia.
| |
Collapse
|
5
|
Ziegler AC, Haider RS, Hoffmann C, Gräler MH. S1PR3 agonism and S1P lyase inhibition rescue mice in the severe state of experimental sepsis. Biomed Pharmacother 2024; 174:116575. [PMID: 38599060 DOI: 10.1016/j.biopha.2024.116575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 04/03/2024] [Accepted: 04/05/2024] [Indexed: 04/12/2024] Open
Abstract
Sepsis is characterized as life-threatening organ dysfunction caused by a dysregulated host response to an infection. Despite numerous clinical trials that addressed this syndrome, there is still no causative treatment available to dampen its severity. Curtailing the infection at an early stage with anti-infectives is the only effective treatment regime besides intensive care. In search for additional treatment options, we recently discovered the inhibition of the sphingosine 1-phosphate (S1P) lyase and subsequent activation of the S1P receptor type 3 (S1PR3) in pre-conditioning experiments as promising targets for sepsis prevention. Here, we demonstrate that treatment of septic mice with the direct S1P lyase inhibitor C31 or the S1PR3 agonist CYM5541 in the advanced phase of sepsis resulted in a significantly increased survival rate. A single dose of each compound led to a rapid decline of sepsis severity in treated mice and coincided with decreased cytokine release and increased lung barrier function with unaltered bacterial load. The survival benefit of both compounds was completely lost in S1PR3 deficient mice. Treatment of the murine macrophage cell line J774.1 with either C31 or CYM5541 resulted in decreased protein kinase B (Akt) and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) phosphorylation without alteration of the mitogen-activated protein kinase (MAPK) p38 and p44/42 phosphorylation. Thus, activation of S1PR3 in the acute phase of sepsis by direct agonism or S1P lyase inhibition dampened Akt and JNK phosphorylation, resulting in decreased cytokine release, improved lung barrier stability, rapid decline of sepsis severity and better survival in mice.
Collapse
Affiliation(s)
- Anke C Ziegler
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Str. 2. Jena D-07745, Germany
| | - Raphael S Haider
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena D-07745, Germany; Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, UK; Centre of Membrane Protein and Receptors, Universities of Birmingham and Nottingham, Midlands NG2 7AG, UK
| | - Carsten Hoffmann
- Institut für Molekulare Zellbiologie, CMB - Center for Molecular Biomedicine, Universitätsklinikum Jena, Friedrich-Schiller-Universität Jena, Hans-Knöll-Straße 2, Jena D-07745, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Str. 2. Jena D-07745, Germany; Center for Sepsis Control and Care, Jena University Hospital, Jena 07740, Germany.
| |
Collapse
|
6
|
Xiao J. Sphingosine 1-Phosphate Lyase in the Developing and Injured Nervous System: a Dichotomy? Mol Neurobiol 2023; 60:6869-6882. [PMID: 37507574 PMCID: PMC10657793 DOI: 10.1007/s12035-023-03524-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
Sphingosine 1-phosphate lyase (SPL) is the terminal enzyme that controls the degradation of the bioactive lipid sphingosine 1-phosphate (S1P) within an interconnected sphingolipid metabolic network. The unique metabolic position of SPL in maintaining S1P levels implies SPL could be an emerging new therapeutic target. Over the past decade, an evolving effort has been made to unravel the role of SPL in the nervous system; however, to what extent SPL influences the developing and mature nervous system through altering S1P biosynthesis remains opaque. While congenital SPL deletion is associated with deficits in the developing nervous system, the loss of SPL activity in adults appears to be neuroprotective in acquired neurological disorders. The controversial findings concerning SPL's role in the nervous system are further constrained by the current genetic and pharmacological tools. This review attempts to focus on the multi-faceted nature of SPL function in the mammalian nervous systems, implying its dichotomy in the developing and adult central nervous system (CNS). This article also highlights SPL is emerging as a therapeutic molecule that can be selectively targeted to modulate S1P for the treatment of acquired neurodegenerative diseases, raising new questions for future investigation. The development of cell-specific inducible conditional SPL mutants and selective pharmacological tools will allow the precise understanding of SPL's function in the adult CNS, which will aid the development of a new strategy focusing on S1P-based therapies for neuroprotection.
Collapse
Affiliation(s)
- Junhua Xiao
- Department of Health Sciences and Biostatistics, School of Health Sciences, Swinburne University of Technology, John Street, Hawthorn, VIC, 3022, Australia.
| |
Collapse
|
7
|
Kihara Y, Chun J. Molecular and neuroimmune pharmacology of S1P receptor modulators and other disease-modifying therapies for multiple sclerosis. Pharmacol Ther 2023; 246:108432. [PMID: 37149155 DOI: 10.1016/j.pharmthera.2023.108432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
Multiple sclerosis (MS) is a neurological, immune-mediated demyelinating disease that affects people in the prime of life. Environmental, infectious, and genetic factors have been implicated in its etiology, although a definitive cause has yet to be determined. Nevertheless, multiple disease-modifying therapies (DMTs: including interferons, glatiramer acetate, fumarates, cladribine, teriflunomide, fingolimod, siponimod, ozanimod, ponesimod, and monoclonal antibodies targeting ITGA4, CD20, and CD52) have been developed and approved for the treatment of MS. All the DMTs approved to date target immunomodulation as their mechanism of action (MOA); however, the direct effects of some DMTs on the central nervous system (CNS), particularly sphingosine 1-phosphate (S1P) receptor (S1PR) modulators, implicate a parallel MOA that may also reduce neurodegenerative sequelae. This review summarizes the currently approved DMTs for the treatment of MS and provides details and recent advances in the molecular pharmacology, immunopharmacology, and neuropharmacology of S1PR modulators, with a special focus on the CNS-oriented, astrocyte-centric MOA of fingolimod.
Collapse
Affiliation(s)
- Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, United States of America.
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, United States of America
| |
Collapse
|
8
|
Pournasiri Z, Madani A, Nazarpack F, Sayer JA, Chavoshzadeh Z, Nili F, Tran P, Saba JD, Jamee M. Sphingosine phosphate lyase insufficiency syndrome: a systematic review. World J Pediatr 2023; 19:425-437. [PMID: 36371483 DOI: 10.1007/s12519-022-00615-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND Sphingosine-1-phosphate lyase insufficiency syndrome (SPLIS) or nephrotic syndrome type-14 is caused by biallelic mutations in SGPL1. Here, we conducted a systematic review to delineate the characteristics of SPLIS patients. METHODS A literature search was performed in PubMed, Web of Science, and Scopus databases, and eligible studies were included. For all patients, demographic, clinical, laboratory, and molecular data were collected and analyzed. RESULTS Fifty-five SPLIS patients (54.9% male, 45.1% female) were identified in 19 articles. Parental consanguinity and positive family history were reported in 70.9% and 52.7% of patients, respectively. Most patients (54.9%) primarily manifested within the first year of life, nearly half of whom survived, while all patients with a prenatal diagnosis of SPLIS (27.5%) died at a median [interquartile (IQR)] age of 2 (1.4-5.3) months (P = 0.003). The most prevalent clinical feature was endocrinopathies, including primary adrenal insufficiency (PAI) (71.2%) and hypothyroidism (32.7%). Kidney disorders (42, 80.8%) were mainly in the form of steroid-resistant nephrotic syndrome (SRNS) and progressed to end-stage kidney disease (ESKD) in 19 (36.5%) patients at a median (IQR) age of 6 (1.4-42.6) months. Among 30 different mutations in SGPL1, the most common was c.665G > A (p.Arg222Gln) in 11 (20%) patients. Twenty-six (49.1%) patients with available outcome were deceased at a median (IQR) age of 5 (1.5-30.5) months, mostly following ESKD (23%) or sepsis/septic shock (23%). CONCLUSION In patients with PAI and/or SRNS, SGPL1 should be added to diagnostic genetic panels, which can provide an earlier diagnosis of SPLIS and prevention of ESKD and other life-threatening complications.
Collapse
Affiliation(s)
- Zahra Pournasiri
- Pediatric Nephrology Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Madani
- Department of Pediatric Nephrology, Children's Medical Center, Pediatric Chronic Kidney Disease Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Nazarpack
- Pediatric Nephrology Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - John A Sayer
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Central Parkway, Newcastle upon Tyne, NE1 3BZ, UK
- Renal Services, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE7 7DN, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, NE45PL, Tyne and Wear, UK
| | - Zahra Chavoshzadeh
- Immunology and Allergy Department, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, 15514-15468, Iran
| | - Fatemeh Nili
- Department of Pathology, Imam Khomeini Complex Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Paulina Tran
- Allergy Immunology Division, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, USA
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| | - Julie D Saba
- Division of Hematology/Oncology, Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Mahnaz Jamee
- Pediatric Nephrology Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Immunology and Allergy Department, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, 15514-15468, Iran.
| |
Collapse
|
9
|
Brommage R, Liu J, Powell DR. Skeletal phenotypes in secreted frizzled-related protein 4 gene knockout mice mimic skeletal architectural abnormalities in subjects with Pyle's disease from SFRP4 mutations. Bone Res 2023; 11:9. [PMID: 36808149 PMCID: PMC9941579 DOI: 10.1038/s41413-022-00242-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/26/2022] [Accepted: 11/03/2022] [Indexed: 02/22/2023] Open
Abstract
Mutations in SFRP4 cause Pyle's bone disease with wide metaphyses and increased skeletal fragility. The WNT signaling pathway plays important roles in determining skeletal architecture and SFRP4 is a secreted Frizzled decoy receptor that inhibits WNT signaling. Seven cohorts of male and female Sfrp4 gene knockout mice, examined through 2 years of age, had a normal lifespan but showed cortical and trabecular bone phenotypes. Mimicking human Erlenmeyer flask deformities, bone cross-sectional areas were elevated 2-fold in the distal femur and proximal tibia but only 30% in femur and tibia shafts. Reduced cortical bone thickness was observed in the vertebral body, midshaft femur and distal tibia. Elevated trabecular bone mass and numbers were observed in the vertebral body, distal femur metaphysis and proximal tibia metaphysis. Midshaft femurs retained extensive trabecular bone through 2 years of age. Vertebral bodies had increased compressive strength, but femur shafts had reduced bending strength. Trabecular, but not cortical, bone parameters in heterozygous Sfrp4 mice were modestly affected. Ovariectomy resulted in similar declines in both cortical and trabecular bone mass in wild-type and Sfrp4 KO mice. SFRP4 is critical for metaphyseal bone modeling involved in determining bone width. Sfrp4 KO mice show similar skeletal architecture and bone fragility deficits observed in patients with Pyle's disease with SFRP4 mutations.
Collapse
Affiliation(s)
- Robert Brommage
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
- BoneGenomics, The Woodlands, TX, USA.
| | - Jeff Liu
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
- Biogen, Cambridge, MA, USA.
| | - David R Powell
- Department of Metabolism Research, Lexicon Pharmaceuticals, The Woodlands, TX, 77381, USA.
| |
Collapse
|
10
|
Tran P, Jamee M, Pournasiri Z, Chavoshzadeh Z, Sullivan KE. SGPL1 Deficiency: Nephrotic Syndrome with Lymphopenia. J Clin Immunol 2023; 43:72-75. [PMID: 36050428 DOI: 10.1007/s10875-022-01348-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/11/2022] [Indexed: 01/18/2023]
Affiliation(s)
- Paulina Tran
- Department of Pediatrics, Allergy Immunology Division, Children's Hospital of Philadelphia, Philadelphia, USA. .,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA.
| | - Mahnaz Jamee
- Pediatric Nephrology Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Immunology and Allergy Department, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Pournasiri
- Pediatric Nephrology Research Center, Research Institute for Children's Health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Chavoshzadeh
- Immunology and Allergy Department, Mofid Children's Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kathleen E Sullivan
- Department of Pediatrics, Allergy Immunology Division, Children's Hospital of Philadelphia, Philadelphia, USA.,Perelman School of Medicine at the University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
11
|
Yang S, He Y, Zhou J, Yuan H, Qiu L. Steroid-resistant nephrotic syndrome associated with certain SGPL1 variants in a family: Case report and literature review. Front Pediatr 2023; 11:1079758. [PMID: 36873630 PMCID: PMC9978203 DOI: 10.3389/fped.2023.1079758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/16/2023] [Indexed: 02/18/2023] Open
Abstract
OBJECTIVES Steroid-resistant nephrotic syndrome (SRNS) is a clinical syndrome characterized by the lack of response to standard steroid therapy, usually progressing to end-stage renal disease. We reported two cases of female identical twins with SRNS caused by SGPL1 variants in one family, reviewed the relevant literature, and summarized their clinical phenotypes, pathological types, and genotypic characteristics. METHODS Two cases of nephrotic syndrome caused by SGPL1 variants were admitted to Tongji Hospital, affiliated with Tongji Medical College of Huazhong University of Science and Technology. Their clinical data were retrospectively collected, and the peripheral blood genomic DNA was captured and sequenced by whole exome sequencing. Related literature published in PubMed, CNKI, and Wan fang databases was reviewed. RESULTS We described two Chinese identical twin girls with isolated SRNS due to compound heterozygous variants in the SGPL1 (intron4 c.261 + 1G > A and intron12 c.1298 + 6T > C). The patients were followed up for 60.0 months and 53.0 months, respectively, having no extra-renal manifestations. They all died due to renal failure. A total of 31 children with SGPL1 variants causing nephrotic syndrome (including the reported two cases) were identified through a literature review. CONCLUSIONS These two female identical twins were the first reported cases of isolated SRNS caused by SGPL1 variants. Almost all homozygous and compound heterozygous variants of SGPL1 had extra-renal manifestations, but compound heterozygous variants in the intron of SGPL1 may have no obvious extra-renal manifestations. Additionally, a negative genetic testing result does not completely rule out genetic SRNS because the Human Gene Mutation Database or ClinVar is constantly being updated.
Collapse
Affiliation(s)
- Siying Yang
- The Nephrology Division of Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yonghua He
- The Nephrology Division of Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jianhua Zhou
- The Nephrology Division of Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiqing Yuan
- The Nephrology Division of Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liru Qiu
- The Nephrology Division of Department of Pediatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
12
|
Keul P, Peters S, von Wnuck Lipinski K, Schröder NH, Nowak MK, Duse DA, Polzin A, Weske S, Gräler MH, Levkau B. Sphingosine-1-Phosphate (S1P) Lyase Inhibition Aggravates Atherosclerosis and Induces Plaque Rupture in ApoE−/− Mice. Int J Mol Sci 2022; 23:ijms23179606. [PMID: 36077004 PMCID: PMC9455951 DOI: 10.3390/ijms23179606] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 08/09/2022] [Accepted: 08/20/2022] [Indexed: 11/17/2022] Open
Abstract
Altered plasma sphingosine-1-phosphate (S1P) concentrations are associated with clinical manifestations of atherosclerosis. However, whether long-term elevation of endogenous S1P is pro- or anti-atherogenic remains unclear. Here, we addressed the impact of permanently high S1P levels on atherosclerosis in cholesterol-fed apolipoprotein E-deficient (ApoE−/−) mice over 12 weeks. This was achieved by pharmacological inhibition of the S1P-degrading enzyme S1P lyase with 4-deoxypyridoxine (DOP). DOP treatment dramatically accelerated atherosclerosis development, propagated predominantly unstable plaque phenotypes, and resulted in frequent plaque rupture with atherothrombosis. Macrophages from S1P lyase-inhibited or genetically deficient mice had a defect in cholesterol efflux to apolipoprotein A-I that was accompanied by profoundly downregulated cholesterol transporters ATP-binding cassette transporters ABCA1 and ABCG1. This was dependent on S1P signaling through S1PR3 and resulted in dramatically enhanced atherosclerosis in ApoE−/−/S1PR3−/− mice, where DOP treatment had no additional effect. Thus, high endogenous S1P levels promote atherosclerosis, compromise cholesterol efflux, and cause genuine plaque rupture.
Collapse
Affiliation(s)
- Petra Keul
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Susann Peters
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Karin von Wnuck Lipinski
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Nathalie H. Schröder
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Melissa K. Nowak
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Dragos A. Duse
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Amin Polzin
- Division of Cardiology, Pulmonology, and Vascular Medicine, Heinrich Heine University Medical Center Düsseldorf, 40225 Düsseldorf, Germany
| | - Sarah Weske
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Markus H. Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care and Center for Molecular Biomedicine, University Hospital Jena, 07743 Jena, Germany
| | - Bodo Levkau
- Institute for Molecular Medicine III, University Hospital Düsseldorf, University of Düsseldorf, 40225 Düsseldorf, Germany
- Correspondence: ; Tel.: +49-211-88-12611
| |
Collapse
|
13
|
Hu Y, Dai K. Sphingosine 1-Phosphate Metabolism and Signaling. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1372:67-76. [PMID: 35503175 DOI: 10.1007/978-981-19-0394-6_6] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a well-defined bioactive lipid molecule derived from membrane sphingolipid metabolism. In the past decades, a series of key enzymes involved in generation of S1P have been identified and characterized in detail, as well as enzymes degrading S1P. S1P requires transporter to cross the plasma membrane and carrier to deliver to its cognate receptors and therefore transduces signaling in autocrine, paracrine, or endocrine fashions. The essential roles in regulation of development, metabolism, inflammation, and many other aspects of life are mainly executed when S1P binds to receptors provoking the downstream signaling cascades in distinct cells. This chapter will review the synthesis, degradation, transportation, and signaling of S1P and try to provide a comprehensive view of the biology of S1P, evoking new enthusiasms and ideas into the field of the fascinating S1P.
Collapse
Affiliation(s)
- Yan Hu
- Department of Psychiatry, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Kezhi Dai
- Department of Psychiatry, School of Mental Health, Wenzhou Medical University, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
14
|
|
15
|
Qi T, Li L, Weidong T. The Role of Sphingolipid Metabolism in Bone Remodeling. Front Cell Dev Biol 2021; 9:752540. [PMID: 34912800 PMCID: PMC8666436 DOI: 10.3389/fcell.2021.752540] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/11/2021] [Indexed: 02/05/2023] Open
Abstract
Emerging studies of bioactive lipids have made many exciting discoveries in recent years. Sphingolipids and their metabolites perform a wide variety of cellular functions beyond energy metabolism. Emerging evidence based on genetically manipulated mouse models and molecular biology allows us to obtain new insights into the role sphingolipid played on skeletal remodeling. This review summarizes studies or understandings of the crosstalk between sphingomyelin, ceramide, and sphingosine-1-phosphate (S1P) of sphingolipids family and the cells, especially osteoblasts and osteoclasts of the bone through which bone is remodeled during life constantly. This review also shows agonists and antagonists of S1P as possible therapeutic options and opportunities on bone diseases.
Collapse
Affiliation(s)
- Tang Qi
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Liao Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tian Weidong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Engineering Research Center of Oral Translational Medicine, Ministry of Education, National Engineering Laboratory for Oral Regenerative Medicine, West China Hospital of Stomatology, West China School of Public Health, West China Fourth Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
16
|
Spohner AK, Jakobi K, Trautmann S, Thomas D, Schumacher F, Kleuser B, Lütjohann D, El-Hindi K, Grösch S, Pfeilschifter J, Saba JD, Meyer zu Heringdorf D. Mouse Liver Compensates Loss of Sgpl1 by Secretion of Sphingolipids into Blood and Bile. Int J Mol Sci 2021; 22:10617. [PMID: 34638955 PMCID: PMC8508615 DOI: 10.3390/ijms221910617] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 12/23/2022] Open
Abstract
Sphingosine 1 phosphate (S1P) lyase (Sgpl1) catalyses the irreversible cleavage of S1P and thereby the last step of sphingolipid degradation. Loss of Sgpl1 in humans and mice leads to accumulation of sphingolipids and multiple organ injuries. Here, we addressed the role of hepatocyte Sgpl1 for regulation of sphingolipid homoeostasis by generating mice with hepatocyte-specific deletion of Sgpl1 (Sgpl1HepKO mice). Sgpl1HepKO mice had normal body weight, liver weight, liver structure and liver enzymes both at the age of 8 weeks and 8 months. S1P, sphingosine and ceramides, but not glucosylceramides or sphingomyelin, were elevated by ~1.5-2-fold in liver, and this phenotype did not progress with age. Several ceramides were elevated in plasma, while plasma S1P was normal. Interestingly, S1P and glucosylceramides, but not ceramides, were elevated in bile of Sgpl1HepKO mice. Furthermore, liver cholesterol was elevated, while LDL cholesterol decreased in 8-month-old mice. In agreement, the LDL receptor was upregulated, suggesting enhanced uptake of LDL cholesterol. Expression of peroxisome proliferator-activated receptor-γ, liver X receptor and fatty acid synthase was unaltered. These data show that mouse hepatocytes largely compensate the loss of Sgpl1 by secretion of accumulating sphingolipids in a specific manner into blood and bile, so that they can be excreted or degraded elsewhere.
Collapse
Affiliation(s)
- Anna Katharina Spohner
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (A.K.S.); (K.J.); (J.P.)
| | - Katja Jakobi
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (A.K.S.); (K.J.); (J.P.)
| | - Sandra Trautmann
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theo-dor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (D.T.); (K.E.-H.); (S.G.)
| | - Dominique Thomas
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theo-dor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (D.T.); (K.E.-H.); (S.G.)
| | - Fabian Schumacher
- Institut für Pharmazie, Pharmakologie und Toxikologie, Freie Universität Berlin, Königin-Luise-Straße 2-4, 14195 Berlin, Germany; (F.S.); (B.K.)
| | - Burkhard Kleuser
- Institut für Pharmazie, Pharmakologie und Toxikologie, Freie Universität Berlin, Königin-Luise-Straße 2-4, 14195 Berlin, Germany; (F.S.); (B.K.)
| | - Dieter Lütjohann
- Institut für Klinische Chemie und Pharmakologie, Universitätsklinikum Bonn, Sigmund-Freud-Straße 25, 53127 Bonn, Germany;
| | - Khadija El-Hindi
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theo-dor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (D.T.); (K.E.-H.); (S.G.)
| | - Sabine Grösch
- Institut für Klinische Pharmakologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theo-dor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (S.T.); (D.T.); (K.E.-H.); (S.G.)
| | - Josef Pfeilschifter
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (A.K.S.); (K.J.); (J.P.)
| | - Julie D. Saba
- Department of Pediatrics, Division of Hematology/Oncology, University of California, 505 Parnassus Ave, San Francisco, CA 94143, USA;
| | - Dagmar Meyer zu Heringdorf
- Institut für Allgemeine Pharmakologie und Toxikologie, Universitätsklinikum, Goethe-Universität Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany; (A.K.S.); (K.J.); (J.P.)
| |
Collapse
|
17
|
Cui M, Göbel V, Zhang H. Uncovering the 'sphinx' of sphingosine 1-phosphate signalling: from cellular events to organ morphogenesis. Biol Rev Camb Philos Soc 2021; 97:251-272. [PMID: 34585505 PMCID: PMC9292677 DOI: 10.1111/brv.12798] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 11/02/2022]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive sphingolipid metabolite, functioning as a signalling molecule in diverse cellular processes. Over the past few decades, studies of S1P signalling have revealed that the physiological activity of S1P largely depends on S1P metabolizing enzymes, transporters and receptors on the plasma membrane, as well as on the intracellular proteins that S1P binds directly to. In addition to its roles in cancer signalling, immunity and inflammation, a large body of evidence has identified a close link of S1P signalling with organ morphogenesis. Here we discuss the vital role of S1P signalling in orchestrating various cellular events during organ morphogenesis through analysing each component along the extracellular and intracellular S1P signalling axes. For each component, we review advances in our understanding of S1P signalling and function from the upstream regulators to the downstream effectors and from cellular behaviours to tissue organization, primarily in the context of morphogenetic mechanisms. S1P-mediated vesicular trafficking is also discussed as a function independent of its signalling function. A picture emerges that reveals a multifaceted role of S1P-dependent pathways in the development and maintenance of organ structure and function.
Collapse
Affiliation(s)
- Mengqiao Cui
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China
| | - Verena Göbel
- Mucosal Immunology and Biology Research Center, Department of Pediatrics, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, U.S.A
| | - Hongjie Zhang
- Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, China.,MoE Frontiers Science Center for Precision Oncology, University of Macau, Taipa, Macau SAR, 999078, China
| |
Collapse
|
18
|
Sphingolipids in Hematopoiesis: Exploring Their Role in Lineage Commitment. Cells 2021; 10:cells10102507. [PMID: 34685487 PMCID: PMC8534120 DOI: 10.3390/cells10102507] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/30/2021] [Accepted: 09/18/2021] [Indexed: 11/17/2022] Open
Abstract
Sphingolipids, associated enzymes, and the sphingolipid pathway are implicated in complex, multifaceted roles impacting several cell functions, such as cellular homeostasis, apoptosis, cell differentiation, and more through intrinsic and autocrine/paracrine mechanisms. Given this broad range of functions, it comes as no surprise that a large body of evidence points to important functions of sphingolipids in hematopoiesis. As the understanding of the processes that regulate hematopoiesis and of the specific characteristics that define each type of hematopoietic cells is being continuously refined, the understanding of the roles of sphingolipid metabolism in hematopoietic lineage commitment is also evolving. Recent findings indicate that sphingolipid alterations can modulate lineage commitment from stem cells all the way to megakaryocytic, erythroid, myeloid, and lymphoid cells. For instance, recent evidence points to the ability of de novo sphingolipids to regulate the stemness of hematopoietic stem cells while a substantial body of literature implicates various sphingolipids in specialized terminal differentiation, such as thrombopoiesis. This review provides a comprehensive discussion focused on the mechanisms that link sphingolipids to the commitment of hematopoietic cells to the different lineages, also highlighting yet to be resolved questions.
Collapse
|
19
|
Saba JD, Keller N, Wang JY, Tang F, Slavin A, Shen Y. Genotype/Phenotype Interactions and First Steps Toward Targeted Therapy for Sphingosine Phosphate Lyase Insufficiency Syndrome. Cell Biochem Biophys 2021; 79:547-559. [PMID: 34133011 DOI: 10.1007/s12013-021-01013-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/07/2021] [Indexed: 10/21/2022]
Abstract
Sphingosine-1-phosphate lyase insufficiency syndrome (SPLIS) is a rare metabolic disorder caused by a deficiency in sphingosine-1-phosphate lyase (SPL), the final enzyme in the sphingolipid degradative pathway. Inactivating mutations of SGPL1-the gene encoding SPL-lead to a deficiency of its downstream products, and buildup of sphingolipid intermediates, including its bioactive substrate, sphingosine-1-phosphate (S1P), the latter causing lymphopenia, a hallmark of the disease. Other manifestations of SPLIS include nephrotic syndrome, neuronal defects, and adrenal insufficiency, but their pathogenesis remains unknown. In this report, we describe the correlation between SGPL1 genotypes, age at diagnosis, and patient outcome. Vitamin B6 serves as a cofactor for SPL. B6 supplementation may aid some SPLIS patients by overcoming poor binding kinetics and promoting proper folding and stability of mutant SPL proteins. However, this approach remains limited to patients with a susceptible allele. Gene therapy represents a potential targeted therapy for SPLIS patients harboring B6-unresponsive missense mutations, truncations, deletions, and splice-site mutations. When Sgpl1 knockout (SPLKO) mice that model SPLIS were treated with adeno-associated virus (AAV)-mediated SGPL1 gene therapy, they showed profound improvement in survival and kidney and neurological function compared to untreated SPLKO mice. Thus, gene therapy appears promising as a universal, potentially curative treatment for SPLIS.
Collapse
Affiliation(s)
- Julie D Saba
- UCSF Department of Pediatrics, San Francisco, CA, USA.
| | - Nancy Keller
- UCSF Department of Pediatrics, San Francisco, CA, USA
| | - Jen-Yeu Wang
- UCSF Department of Pediatrics, San Francisco, CA, USA
| | - Felicia Tang
- UCSF Department of Pediatrics, San Francisco, CA, USA
| | - Avi Slavin
- UCSF Department of Pediatrics, San Francisco, CA, USA
| | - Yizhuo Shen
- UCSF Department of Pediatrics, San Francisco, CA, USA
| |
Collapse
|
20
|
Zhao P, Tassew GB, Lee JY, Oskouian B, Muñoz DP, Hodgin JB, Watson GL, Tang F, Wang JY, Luo J, Yang Y, King S, Krauss RM, Keller N, Saba JD. Efficacy of AAV9-mediated SGPL1 gene transfer in a mouse model of S1P lyase insufficiency syndrome. JCI Insight 2021; 6:145936. [PMID: 33755599 PMCID: PMC8119223 DOI: 10.1172/jci.insight.145936] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 03/17/2021] [Indexed: 12/26/2022] Open
Abstract
Sphingosine-1-phosphate lyase insufficiency syndrome (SPLIS) is a rare metabolic disorder caused by inactivating mutations in sphingosine-1-phosphate lyase 1 (SGPL1), which is required for the final step of sphingolipid metabolism. SPLIS features include steroid-resistant nephrotic syndrome and impairment of neurological, endocrine, and hematopoietic systems. Many affected individuals die within the first 2 years. No targeted therapy for SPLIS is available. We hypothesized that SGPL1 gene replacement would address the root cause of SPLIS, thereby serving as a universal treatment for the condition. As proof of concept, we evaluated the efficacy of adeno-associated virus 9–mediated transfer of human SGPL1 (AAV-SPL) given to newborn Sgpl1-KO mice that model SPLIS and die in the first weeks of life. Treatment dramatically prolonged survival and prevented nephrosis, neurodevelopmental delay, anemia, and hypercholesterolemia. STAT3 pathway activation and elevated proinflammatory and profibrogenic cytokines observed in KO kidneys were attenuated by treatment. Plasma and tissue sphingolipids were reduced in treated compared with untreated KO pups. SGPL1 expression and activity were measurable for at least 40 weeks. In summary, early AAV-SPL treatment prevents nephrosis, lipidosis, and neurological impairment in a mouse model of SPLIS. Our results suggest that SGPL1 gene replacement holds promise as a durable and universal targeted treatment for SPLIS.
Collapse
Affiliation(s)
- Piming Zhao
- Department of Pediatrics, UCSF, San Francisco, California, USA.,Cure Genetics, Suzhou, China
| | | | - Joanna Y Lee
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Babak Oskouian
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Denise P Muñoz
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Jeffrey B Hodgin
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Gordon L Watson
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Felicia Tang
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Jen-Yeu Wang
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Jinghui Luo
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Yingbao Yang
- Department of Pathology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sarah King
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Ronald M Krauss
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Nancy Keller
- Department of Pediatrics, UCSF, San Francisco, California, USA
| | - Julie D Saba
- Department of Pediatrics, UCSF, San Francisco, California, USA
| |
Collapse
|
21
|
Ziegler AC, Müller T, Gräler MH. Sphingosine 1-phosphate in sepsis and beyond: Its role in disease tolerance and host defense and the impact of carrier molecules. Cell Signal 2020; 78:109849. [PMID: 33249088 DOI: 10.1016/j.cellsig.2020.109849] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/29/2022]
Abstract
Sphingosine 1-phosphate (S1P) is an important immune modulator responsible for physiological cellular responses like lymphocyte development and function, positioning and emigration of T and B cells and cytokine secretion. Recent reports indicate that S1P does not only regulate immunity, but can also protect the function of organs by inducing disease tolerance. S1P also influences the replication of certain pathogens, and sphingolipids are also involved in pathogen recognition and killing. Certain carrier molecules for S1P like serum albumin and high density lipoproteins contribute to the regulation of S1P effects. They are able to associate with S1P and modulate its signaling properties. Similar to S1P, both carrier molecules are also decreased in sepsis patients and likely contribute to sepsis pathology and severity. In this review, we will introduce the concept of disease tolerance and the involvement of S1P. We will also discuss the contribution of S1P and its precursor sphingosine to host defense mechanisms against pathogens. Finally, we will summarize current data demonstrating the influence of carrier molecules for differential S1P signaling. The presented data may lead to new strategies for the prevention and containment of sepsis.
Collapse
Affiliation(s)
- Anke C Ziegler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07740 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany
| | - Tina Müller
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07740 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, 07740 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany.
| |
Collapse
|
22
|
Müller T, Gräler MH. Development and validation of a QTrap method for sensitive quantification of sphingosine 1-phosphate. Biomed Chromatogr 2020; 35:e5004. [PMID: 33063871 DOI: 10.1002/bmc.5004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/30/2020] [Accepted: 10/06/2020] [Indexed: 01/20/2023]
Abstract
Sphingosine 1-phosphate (S1P) is a bioactive phospholipid and ligand for five G protein-coupled cell-surface receptors designated S1PR1-5. The determination of low levels of S1P remains a challenge and usually requires sophisticated analytical instrumentation and methodology. This report describes a technique using the linear ion trap mode of a basic QTrap triple-quadrupole mass spectrometer. S1P was extracted from acidified biological samples using a modified Folch extraction procedure. After the addition of C17-sphingosine as an internal standard, a step gradient LC method was used to separate the analytes on a reversed-phase C18 MultoHigh analytical column. After the internal standard C17-sphingosine was detected by multiple reaction monitoring (MRM), the detection mode was switched to enhanced product ion (EPI) mode for the detection of S1P. The mode was switched back to MRM again for the detection of other analytes. Using this QTrap method, we reached a limit of detection of 1 nM and a limit of quantification of 3 nM for S1P, which was up to 30 times more sensitive than the MRM mode with the same instrument. Intra-day precision ranged between -3.8 and 6.3%, and inter-day precision was between -13.8 and 3.3%, depending on the spiked S1P concentration.
Collapse
Affiliation(s)
- Tina Müller
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany.,Center for Molecular Biomedicine, Jena University Hospital, Jena, Germany.,Center for Sepsis Control and Care, Jena University Hospital, Jena, Germany
| |
Collapse
|
23
|
Anwar M, Mehta D. Post-translational modifications of S1PR1 and endothelial barrier regulation. Biochim Biophys Acta Mol Cell Biol Lipids 2020; 1865:158760. [PMID: 32585303 PMCID: PMC7409382 DOI: 10.1016/j.bbalip.2020.158760] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/09/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022]
Abstract
Sphingosine-1-phosphate receptor-1 (S1PR1), a G-protein coupled receptor that is expressed in endothelium and activated upon ligation by the bioactive lipid sphingosine-1-phosphate (S1P), is an important vascular-barrier protective mechanism at the level of adherens junctions (AJ). Loss of endothelial barrier function is a central factor in the pathogenesis of various inflammatory conditions characterized by protein-rich lung edema formation, such as acute respiratory distress syndrome (ARDS). While several S1PR1 agonists are available, the challenge of arresting the progression of protein-rich edema formation remains to be met. In this review, we discuss the role of S1PRs, especially S1PR1, in regulating endothelial barrier function. We review recent findings showing that replenishment of the pool of cell-surface S1PR1 may be crucial to the effectiveness of S1P in repairing the endothelial barrier. In this context, we discuss the S1P generating machinery and mechanisms that regulate S1PR1 at the cell surface and their impact on endothelial barrier function.
Collapse
Affiliation(s)
- Mumtaz Anwar
- Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago Chicago, IL 60612, United States of America
| | - Dolly Mehta
- Department of Pharmacology and Center for Lung and Vascular Biology, University of Illinois at Chicago Chicago, IL 60612, United States of America.
| |
Collapse
|
24
|
Zhao P, Liu ID, Hodgin JB, Benke PI, Selva J, Torta F, Wenk MR, Endrizzi JA, West O, Ou W, Tang E, Goh DLM, Tay SKH, Yap HK, Loh A, Weaver N, Sullivan B, Larson A, Cooper MA, Alhasan K, Alangari AA, Salim S, Gumus E, Chen K, Zenker M, Hildebrandt F, Saba JD. Responsiveness of sphingosine phosphate lyase insufficiency syndrome to vitamin B6 cofactor supplementation. J Inherit Metab Dis 2020; 43:1131-1142. [PMID: 32233035 PMCID: PMC8072405 DOI: 10.1002/jimd.12238] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/26/2022]
Abstract
Sphingosine-1-phosphate (S1P) lyase is a vitamin B6-dependent enzyme that degrades sphingosine-1-phosphate in the final step of sphingolipid metabolism. In 2017, a new inherited disorder was described caused by mutations in SGPL1, which encodes sphingosine phosphate lyase (SPL). This condition is referred to as SPL insufficiency syndrome (SPLIS) or alternatively as nephrotic syndrome type 14 (NPHS14). Patients with SPLIS exhibit lymphopenia, nephrosis, adrenal insufficiency, and/or neurological defects. No targeted therapy for SPLIS has been reported. Vitamin B6 supplementation has therapeutic activity in some genetic diseases involving B6-dependent enzymes, a finding ascribed largely to the vitamin's chaperone function. We investigated whether B6 supplementation might have activity in SPLIS patients. We retrospectively monitored responses of disease biomarkers in patients supplemented with B6 and measured SPL activity and sphingolipids in B6-treated patient-derived fibroblasts. In two patients, disease biomarkers responded to B6 supplementation. S1P abundance and activity levels increased and sphingolipids decreased in response to B6. One responsive patient is homozygous for an SPL R222Q variant present in almost 30% of SPLIS patients. Molecular modeling suggests the variant distorts the dimer interface which could be overcome by cofactor supplementation. We demonstrate the first potential targeted therapy for SPLIS and suggest that 30% of SPLIS patients might respond to cofactor supplementation.
Collapse
Affiliation(s)
- Piming Zhao
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Isaac D. Liu
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Jeffrey B. Hodgin
- Department of Pathology, University of Michigan Hospitals and Health Center, Ann Arbor, Michigan
| | - Peter I. Benke
- SLING, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jeremy Selva
- SLING, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Federico Torta
- SLING, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Markus R. Wenk
- SLING, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - James A. Endrizzi
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Olivia West
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Weixing Ou
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Emily Tang
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, California
| | - Denise Li-Meng Goh
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Stacey Kiat-Hong Tay
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Hui-Kim Yap
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Alwin Loh
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University Health System, Singapore, Singapore
| | - Nicole Weaver
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
| | - Bonnie Sullivan
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio
- Division of Clinical Genetics, Children’s Mercy Kansas City, Kansas City, Missouri
- Department of Pediatrics, University of Missouri, Kansas City, Missouri
| | - Austin Larson
- Department of Pediatrics, University of Colorado School of Medicine, Denver, Colorado
| | - Megan A. Cooper
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Khalid Alhasan
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah A. Alangari
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Suha Salim
- Department of Pediatrics, College of Medicine, King Saud University, Riyadh, Saudi Arabia
| | - Evren Gumus
- Department of Medicine, Harran University, Sanliurfa, Turkey
| | - Karin Chen
- Department of Pediatrics, Division of Allergy and Immunology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Martin Zenker
- Institute of Human Genetics, Otto von Guericke University, Magdeburg, Germany
| | | | - Julie D. Saba
- Department of Pediatrics, Division of Hematology/Oncology, University of California, San Francisco, California
| |
Collapse
|
25
|
Weigel C, Hüttner SS, Ludwig K, Krieg N, Hofmann S, Schröder NH, Robbe L, Kluge S, Nierhaus A, Winkler MS, Rubio I, von Maltzahn J, Spiegel S, Gräler MH. S1P lyase inhibition protects against sepsis by promoting disease tolerance via the S1P/S1PR3 axis. EBioMedicine 2020; 58:102898. [PMID: 32711251 PMCID: PMC7381498 DOI: 10.1016/j.ebiom.2020.102898] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 06/19/2020] [Accepted: 07/01/2020] [Indexed: 12/29/2022] Open
Abstract
Background One-third of all deaths in hospitals are caused by sepsis. Despite its demonstrated prevalence and high case fatality rate, antibiotics remain the only target-oriented treatment option currently available. Starting from results showing that low-dose anthracyclines protect against sepsis in mice, we sought to find new causative treatment options to improve sepsis outcomes. Methods Sepsis was induced in mice, and different treatment options were evaluated regarding cytokine and biomarker expression, lung epithelial cell permeability, autophagy induction, and survival benefit. Results were validated in cell culture experiments and correlated with patient samples. Findings Effective low-dose epirubicin treatment resulted in substantial downregulation of the sphingosine 1-phosphate (S1P) degrading enzyme S1P lyase (SPL). Consequent accumulation and secretion of S1P in lung parenchyma cells stimulated the S1P-receptor type 3 (S1PR3) and mitogen-activated protein kinases p38 and ERK, reducing tissue damage via increased disease tolerance. The protective effects of SPL inhibition were absent in S1PR3 deficient mice. Sepsis patients showed increased expression of SPL, stable expression of S1PR3, and increased levels of mucin-1 and surfactant protein D as indicators of lung damage. Interpretation Our work highlights a tissue-protective effect of SPL inhibition in sepsis due to activation of the S1P/S1PR3 axis and implies that SPL inhibitors and S1PR3 agonists might be potential therapeutics to protect against sepsis by increasing disease tolerance against infections. Funding This study was supported by the Center for Sepsis Control and Care (CSCC), the German Research Foundation (DFG), RTG 1715 (to M. H. G. and I. R.) and the National Institutes of Health, Grant R01GM043880 (to S. S.).
Collapse
Affiliation(s)
- Cynthia Weigel
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Sören S Hüttner
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Kristin Ludwig
- Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Institute of Molecular Cell Biology, Jena University Hospital, 07745 Jena, Germany
| | - Nadine Krieg
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany
| | - Susann Hofmann
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany
| | - Nathalie H Schröder
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany
| | - Linda Robbe
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Axel Nierhaus
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Martin S Winkler
- Department of Anesthesiology and Intensive Care Medicine, University Medical Center Göttingen, 37075 Göttingen, Germany
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany
| | - Julia von Maltzahn
- Leibniz Institute on Aging - Fritz Lipmann Institute (FLI), 07745 Jena, Germany
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, USA
| | - Markus H Gräler
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany; Center for Molecular Biomedicine, Jena University Hospital, 07745 Jena, Germany; Center for Sepsis Control and Care, Jena University Hospital, 07740 Jena, Germany.
| |
Collapse
|
26
|
Dawoody Nejad L, Stumpe M, Rauch M, Hemphill A, Schneiter R, Bütikofer P, Serricchio M. Mitochondrial sphingosine-1-phosphate lyase is essential for phosphatidylethanolamine synthesis and survival of Trypanosoma brucei. Sci Rep 2020; 10:8268. [PMID: 32427974 PMCID: PMC7237492 DOI: 10.1038/s41598-020-65248-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 04/29/2020] [Indexed: 01/18/2023] Open
Abstract
Sphingosine-1-phosphate is a signaling molecule involved in the control of cell migration, differentiation, survival and other physiological processes. This sphingolipid metabolite can be degraded by the action of sphingosine-1-phosphate lyase (SPL) to form hexadecenal and ethanolamine phosphate. The importance of SPL-mediated ethanolamine phosphate formation has been characterized in only few cell types. We show that in the protozoan parasite Trypanosoma brucei, expression of TbSpl is essential for cell survival. Ablation of TbSpl expression increased sphingosine-1-phosphate levels and reduced de novo formation and steady-state levels of the glycerophospholipid phosphatidylethanolamine (PE). Growth of TbSpl-depleted parasites could be in part rescued by ethanolamine supplementation to the growth medium, indicating that the main function of TbSpl is to provide ethanolamine phosphate for PE synthesis. In contrast to most cell types analyzed, where SPL localizes to the endoplasmic reticulum, we found by high-resolution microscopy that TbSpl is a mitochondrial protein. In spite of its mitochondrial localization, TbSpl depletion had no apparent effect on mitochondrial morphology but resulted in aggregation of acidocalcisomes. Our results link mitochondria to sphingolipid metabolism and suggest possible roles for PE in acidocalcisome function.
Collapse
Affiliation(s)
- Ladan Dawoody Nejad
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland.,Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Michael Stumpe
- Division of Biochemistry, Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Monika Rauch
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland
| | - Andrew Hemphill
- Institute of Parasitology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Roger Schneiter
- Division of Biochemistry, Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Peter Bütikofer
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland.
| | - Mauro Serricchio
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, Switzerland.
| |
Collapse
|
27
|
Abstract
The signaling lipid sphingosine 1-phosphate (S1P) plays critical roles in an immune response. Drugs targeting S1P signaling have been remarkably successful in treatment of multiple sclerosis, and they have shown promise in clinical trials for colitis and psoriasis. One mechanism of these drugs is to block lymphocyte exit from lymph nodes, where lymphocytes are initially activated, into circulation, from which lymphocytes can reach sites of inflammation. Indeed, S1P can be considered a circulation marker, signaling to immune cells to help them find blood and lymphatic vessels, and to endothelial cells to stabilize the vasculature. That said, S1P plays pleiotropic roles in the immune response, and it will be important to build an integrated view of how S1P shapes inflammation. S1P can function so effectively because its distribution is exquisitely tightly controlled. Here we review how S1P gradients regulate immune cell exit from tissues, with particular attention to key outstanding questions in the field.
Collapse
Affiliation(s)
- Audrey A.L. Baeyens
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA;,
| | - Susan R. Schwab
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA;,
| |
Collapse
|
28
|
Dixit D, Okuniewska M, Schwab SR. Secrets and lyase: Control of sphingosine 1-phosphate distribution. Immunol Rev 2020; 289:173-185. [PMID: 30977198 DOI: 10.1111/imr.12760] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 03/05/2019] [Accepted: 03/09/2019] [Indexed: 12/26/2022]
Abstract
The signaling lipid sphingosine 1-phosphate (S1P) plays key roles in many physiological processes. In the immune system, S1P's best-described function is to draw cells out of tissues into circulation. Here, we will review models of S1P distribution in the thymus, lymph nodes, spleen, and nonlymphoid tissues. These models have been challenging to construct, because of the lack of tools to map lipid gradients. Nonetheless, evidence to date suggests that S1P distribution is exquisitely tightly controlled, and that concentrations of signaling-available S1P cannot be predicted by standard rules of thumb. The fine regulation of S1P gradients may explain how S1P can simultaneously direct multiple cell movements both between tissues and circulation and within tissues. It may also make it feasible to develop drugs that enable spatially specific modulation of S1P signaling.
Collapse
Affiliation(s)
- Dhaval Dixit
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York
| | - Martyna Okuniewska
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York
| | - Susan R Schwab
- Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York City, New York
| |
Collapse
|
29
|
Heckt T, Brylka LJ, Neven M, Amling M, Schinke T. Deficiency of sphingosine-1-phosphate receptor 3 does not affect the skeletal phenotype of mice lacking sphingosine-1-phosphate lyase. PLoS One 2019; 14:e0219734. [PMID: 31314788 PMCID: PMC6636735 DOI: 10.1371/journal.pone.0219734] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/01/2019] [Indexed: 01/17/2023] Open
Abstract
Albeit osteoporosis is one of the most prevalent disorders in the aged population, treatment options stimulating the activity of bone-forming osteoblasts are still limited. We and others have previously identified sphingosine-1-phosphate (S1P) as a bone remodeling coupling factor, which is released by bone-resorbing osteoclasts to stimulate bone formation. Moreover, S1pr3, encoding one of the five known S1P receptors (S1P3), was found differentially expressed in osteoblasts, and S1P3 deficiency corrected the moderate high bone mass phenotype of a mouse model (deficient for the calcitonin receptor) with increased S1P release from osteoclasts. In the present study we addressed the question, if S1P3 deficiency would also influence the skeletal phenotype of mice lacking S1P-lyase (encoded by Sgpl1), which display markedly increased S1P levels due to insufficient degradation. Consistent with previous reports, the majority of Sgpl1-deficient mice died before or shortly after weaning, and this lethality was not influenced by additional S1P3 deficiency. At 3 weeks of age, Sgpl1-deficient mice displayed increased trabecular bone mass, which was associated with enhanced osteoclastogenesis and bone resorption, but also with increased bone formation. Most importantly however, none of the skeletal parameters assessed by μCT, histomorphometry and serum analyses were significantly influenced by additional S1P3 deficiency. Taken together, our findings fully support the concept that S1P is a potent osteoanabolic molecule, although S1P3 is not the sole receptor mediating this influence. Since S1P receptors are considered excellent drug targets, it is now required to screen for the impact of other family members on bone formation.
Collapse
Affiliation(s)
- Timo Heckt
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Laura J. Brylka
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Mona Neven
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg Eppendorf, Hamburg, Germany
- * E-mail:
| |
Collapse
|
30
|
Brommage R, Powell DR, Vogel P. Predicting human disease mutations and identifying drug targets from mouse gene knockout phenotyping campaigns. Dis Model Mech 2019; 12:dmm038224. [PMID: 31064765 PMCID: PMC6550044 DOI: 10.1242/dmm.038224] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Two large-scale mouse gene knockout phenotyping campaigns have provided extensive data on the functions of thousands of mammalian genes. The ongoing International Mouse Phenotyping Consortium (IMPC), with the goal of examining all ∼20,000 mouse genes, has examined 5115 genes since 2011, and phenotypic data from several analyses are available on the IMPC website (www.mousephenotype.org). Mutant mice having at least one human genetic disease-associated phenotype are available for 185 IMPC genes. Lexicon Pharmaceuticals' Genome5000™ campaign performed similar analyses between 2000 and the end of 2008 focusing on the druggable genome, including enzymes, receptors, transporters, channels and secreted proteins. Mutants (4654 genes, with 3762 viable adult homozygous lines) with therapeutically interesting phenotypes were studied extensively. Importantly, phenotypes for 29 Lexicon mouse gene knockouts were published prior to observations of similar phenotypes resulting from homologous mutations in human genetic disorders. Knockout mouse phenotypes for an additional 30 genes mimicked previously published human genetic disorders. Several of these models have helped develop effective treatments for human diseases. For example, studying Tph1 knockout mice (lacking peripheral serotonin) aided the development of telotristat ethyl, an approved treatment for carcinoid syndrome. Sglt1 (also known as Slc5a1) and Sglt2 (also known as Slc5a2) knockout mice were employed to develop sotagliflozin, a dual SGLT1/SGLT2 inhibitor having success in clinical trials for diabetes. Clinical trials evaluating inhibitors of AAK1 (neuropathic pain) and SGLT1 (diabetes) are underway. The research community can take advantage of these unbiased analyses of gene function in mice, including the minimally studied 'ignorome' genes.
Collapse
Affiliation(s)
- Robert Brommage
- Department of Metabolism Research, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381, USA
| | - David R Powell
- Department of Metabolism Research, Lexicon Pharmaceuticals, 8800 Technology Forest Place, The Woodlands, TX 77381, USA
| | - Peter Vogel
- St. Jude Children's Research Hospital, Pathology, MS 250, Room C5036A, 262 Danny Thomas Place, Memphis, TN 38105, USA
| |
Collapse
|
31
|
Maharaj A, Maudhoo A, Chan LF, Novoselova T, Prasad R, Metherell LA, Guasti L. Isolated glucocorticoid deficiency: Genetic causes and animal models. J Steroid Biochem Mol Biol 2019; 189:73-80. [PMID: 30817990 DOI: 10.1016/j.jsbmb.2019.02.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Revised: 01/04/2019] [Accepted: 02/25/2019] [Indexed: 12/27/2022]
Abstract
Hereditary adrenocorticotropin (ACTH) resistance syndromes encompass the genetically heterogeneous isolated or Familial Glucocorticoid Deficiency (FGD) and the distinct clinical entity known as Triple A syndrome. The molecular basis of adrenal resistance to ACTH includes defects in ligand binding, MC2R/MRAP receptor trafficking, cellular redox balance, cholesterol synthesis and sphingolipid metabolism. Biochemically, this manifests as ACTH excess in the setting of hypocortisolaemia. Triple A syndrome is an inherited condition involving a tetrad of adrenal insufficiency, achalasia, alacrima and neuropathy. FGD is an autosomal recessive condition characterized by the presence of isolated glucocorticoid deficiency, classically in the setting of preserved mineralocorticoid secretion. Primarily there are three established subtypes of the disease: FGD 1, FGD2 and FGD3 corresponding to mutations in the Melanocortin 2 receptor MC2R (25%), Melanocortin 2 receptor accessory protein MRAP (20%), and Steroidogenic acute regulatory protein STAR (5-10%) respectively. Together, mutations in these 3 genes account for approximately half of cases. Whole exome sequencing in patients negative for MC2R, MRAP and STAR mutations, identified mutations in minichromosome maintenance 4 MCM4, nicotinamide nucleotide transhydrogenase NNT, thioredoxin reductase 2 TXNRD2, cytochrome p450scc CYP11A1, and sphingosine 1-phosphate lyase SGPL1 accounting for a further 10% of FGD. These novel genes have linked replicative and oxidative stress and altered redox potential as a mechanism of adrenocortical damage. However, a genetic diagnosis is still unclear in about 40% of cases. We describe here an updated list of FGD genes and provide a description of relevant mouse models that, despite some being flawed, have been precious allies in the understanding of FGD pathobiology.
Collapse
Affiliation(s)
- Avinaash Maharaj
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Ashwini Maudhoo
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Li F Chan
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Tatiana Novoselova
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Rathi Prasad
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Louise A Metherell
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary, University of London, Charterhouse Square, London, United Kingdom.
| |
Collapse
|
32
|
Snider JM, Luberto C, Hannun YA. Approaches for probing and evaluating mammalian sphingolipid metabolism. Anal Biochem 2019; 575:70-86. [PMID: 30917945 DOI: 10.1016/j.ab.2019.03.014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 03/22/2019] [Accepted: 03/22/2019] [Indexed: 01/02/2023]
Abstract
Sphingolipid metabolism plays a critical role in regulating processes that control cellular fate. This dynamic pathway can generate and degrade the central players: ceramide, sphingosine and sphingosine-1-phosphate in almost any membrane in the cell, adding an unexpected level of complexity in deciphering signaling events. While in vitro assays have been developed for most enzymes in SL metabolism, these assays are setup for optimal activity conditions and can fail to take into account regulatory components such as compartmentalization, substrate limitations, and binding partners that can affect cellular enzymatic activity. Therefore, many in-cell assays have been developed to derive results that are authentic to the cellular situation which may give context to alteration in SL mass. This review will discuss approaches for utilizing probes for mammalian in-cell assays to interrogate most enzymatic steps central to SL metabolism. The use of inhibitors in conjunction with these probes can verify the specificity of cellular assays as well as provide valuable insight into flux in the SL network. The use of inhibitors specific to each of the central sphingolipid enzymes are also discussed to assist researchers in further interrogation of these pathways.
Collapse
Affiliation(s)
- Justin M Snider
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; The Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA
| | - Chiara Luberto
- The Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Department of Physiology and Biophysics, Stony Brook University, Stony Brook, NY, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA; The Stony Brook Cancer Center, Stony Brook University, Stony Brook, NY, USA; Departments of Biochemistry, Pathology and Pharmacology, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
33
|
Cancer-induced inflammation and inflammation-induced cancer in colon: a role for S1P lyase. Oncogene 2019; 38:4788-4803. [PMID: 30816345 DOI: 10.1038/s41388-019-0758-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 12/14/2022]
Abstract
A role of sphingolipids for inflammatory bowel disease and cancer is evident. However, the relative and separate contribution of sphingolipid deterioration in inflammation versus carcinogenesis for the pathophysiology of colitis-associated colon cancer (CAC) was unknown and therefore examined in this study. We performed isogenic bone marrow transplantation of inducible sphingosine-1-phosphate (S1P) lyase knockout mice to specifically modulate sphingolipids and associated genes and proteins in a compartment-specific way in a DSS/AOM mediated CAC model. 3D organoid cultures were used in vitro. S1P lyase (SGPL1) knockout in either immune cells or tissue, caused local sphingolipid accumulation leading to a dichotomic development of CAC: Immune cell SGPL1 knockout (I-SGPL-/-) augmented massive immune cell infiltration initiating colitis with lesions and calprotectin increase. Pathological crypt remodeling plus extracellular S1P-signaling caused delayed tumor formation characterized by S1P receptor 1, STAT3 mRNA increase, as well as programmed cell death ligand 1 expression, accompanied by a putatively counter regulatory STAT1S727 phosphorylation. In contrast, tissue SGPL1 knockout (T-SGPL-/-) provoked immediate occurrence of epithelial-driven tumors with upregulated sphingosine kinase 1, S1P receptor 2 and epidermal growth factor receptor. Here, progressing carcinogenesis was accompanied by an IL-12 to IL-23 shift with a consecutive development of a Th2/GATA3-driven, tumor-favoring microenvironment. Moreover, the knockout models showed distinct lymphopenia and neutrophilia, different from the full SGPL1 knockout. This study shows that depending on the initiating cellular S1P source, the pathophysiology of inflammation-induced cancer versus cancer-induced inflammation develops through separate, discernible molecular steps.
Collapse
|
34
|
Baker JE, Boudreau RM, Seitz AP, Caldwell CC, Gulbins E, Edwards MJ. Sphingolipids and Innate Immunity: A New Approach to Infection in the Post-Antibiotic Era? Surg Infect (Larchmt) 2018; 19:792-803. [DOI: 10.1089/sur.2018.187] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Jennifer E. Baker
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Ryan M. Boudreau
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Aaron P. Seitz
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| | - Charles C. Caldwell
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
- Division of Research, Shriners Hospital for Children, Cincinnati, Ohio
| | - Erich Gulbins
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
- Department of Molecular Biology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Michael J. Edwards
- Division of Research, Department of Surgery, University of Cincinnati, Cincinnati, Ohio
| |
Collapse
|
35
|
Rida R, Kreydiyyeh S. FTY720P inhibits the Na +/K + ATPase in Caco-2 cells via S1PR2: PGE2 and NO are along the signaling pathway. Life Sci 2018; 215:198-206. [PMID: 30439376 DOI: 10.1016/j.lfs.2018.11.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 11/10/2018] [Accepted: 11/11/2018] [Indexed: 01/08/2023]
Abstract
AIMS Sphingosine-1-phosphate (S1P) has been implicated lately in inflammatory bowel disease which has diarrhea as one of its symptoms. Diarrhea is due to altered water movements as a result of altered electrolyte transport, and in particular sodium. Sodium movements are geared by the sodium gradient established by the Na+/K+ ATPase. The aim of this work was to investigate if S1P can modulate the activity of the ATPase, using Caco-2 cells as a model and the S1P analogue, FTY720P. MATERIALS AND METHODS The activity of the ATPase was assayed by measuring the amount of inorganic phosphate liberated in presence and absence of ouabain. Protein expression of the various S1P receptors was studied by western blot analysis. KEY FINDINGS Caco-2 cells were found to express mainly S1PR2 and S1PR3. FTY720P (7.5 nM) reduced significantly the activity of the Na+/K+ ATPase when applied for 15 min. This inhibitory effect disappeared in presence of JTE-013, a specific blocker of S1PR2, and indomethacin, an inhibitor of cyclooxygenase enzymes, and was mimicked by CYM5520, a S1PR2 agonist and by exogenous PGE2. The inhibitory effect of PGE2 did not appear when EP3 receptors were blocked or when a nitric oxide scavenger was added. RpcAMP, a PKA inhibitor, reduced the activity of the Na+/K+ ATPase, while dbcAMP, a PKA activator was without any effect and when added, abrogated the effect of PGE2. SIGNIFICANCE It was concluded that FTY720P inhibits the Na+/K+ ATPase via activation of S1PR2 and generation of PGE2 nitric oxide.
Collapse
Affiliation(s)
- Reem Rida
- Department of Biology, Faculty of Arts & Sciences, American University of Beirut, Beirut, Lebanon
| | - Sawsan Kreydiyyeh
- Department of Biology, Faculty of Arts & Sciences, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
36
|
Choi YJ, Saba JD. Sphingosine phosphate lyase insufficiency syndrome (SPLIS): A novel inborn error of sphingolipid metabolism. Adv Biol Regul 2018; 71:128-140. [PMID: 30274713 DOI: 10.1016/j.jbior.2018.09.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/12/2018] [Accepted: 09/12/2018] [Indexed: 02/06/2023]
Abstract
Sphingosine-1-phosphate lyase (SPL) is an intracellular enzyme that controls the final step in the sphingolipid degradative pathway, the only biochemical pathway for removal of sphingolipids. Specifically, SPL catalyzes the cleavage of sphingosine 1-phosphate (S1P) at the C2-3 carbon bond, resulting in its irreversible degradation to phosphoethanolamine (PE) and hexadecenal. The substrate of the reaction, S1P, is a bioactive sphingolipid metabolite that signals through a family of five G protein-coupled S1P receptors (S1PRs) to mediate biological activities including cell migration, cell survival/death/proliferation and cell extrusion, thereby contributing to development, physiological functions and - when improperly regulated - the pathophysiology of disease. In 2017, several groups including ours reported a novel childhood syndrome that featured a wide range of presentations including fetal hydrops, steroid-resistant nephrotic syndrome (SRNS), primary adrenal insufficiency (PAI), rapid or insidious neurological deterioration, immunodeficiency, acanthosis and endocrine abnormalities. In all cases, the disease was attributed to recessive mutations in the human SPL gene, SGPL1. We now refer to this condition as SPL Insufficiency Syndrome, or SPLIS. Some features of this new sphingolipidosis were predicted by the reported phenotypes of Sgpl1 homozygous null mice that serve as vertebrate SPLIS disease models. However, other SPLIS features reveal previously unrecognized roles for SPL in human physiology. In this review, we briefly summarize the biochemistry, functions and regulation of SPL, the main clinical and biochemical features of SPLIS and what is known about the pathophysiology of this condition from murine and cell models. Lastly, we consider potential therapeutic strategies for the treatment of SPLIS patients.
Collapse
Affiliation(s)
- Youn-Jeong Choi
- UCSF Benioff Children's Hospital Oakland, Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA, 94609, USA
| | - Julie D Saba
- UCSF Benioff Children's Hospital Oakland, Children's Hospital Oakland Research Institute, 5700 Martin Luther King Jr. Way, Oakland, CA, 94609, USA.
| |
Collapse
|
37
|
Don-Doncow N, Zhang Y, Matuskova H, Meissner A. The emerging alliance of sphingosine-1-phosphate signalling and immune cells: from basic mechanisms to implications in hypertension. Br J Pharmacol 2018; 176:1989-2001. [PMID: 29856066 DOI: 10.1111/bph.14381] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 05/24/2018] [Accepted: 05/28/2018] [Indexed: 01/19/2023] Open
Abstract
The immune system plays a considerable role in hypertension. In particular, T-lymphocytes are recognized as important players in its pathogenesis. Despite substantial experimental efforts, the molecular mechanisms underlying the nature of T-cell activation contributing to an onset of hypertension or disease perpetuation are still elusive. Amongst other cell types, lymphocytes express distinct profiles of GPCRs for sphingosine-1-phosphate (S1P) - a bioactive phospholipid that is involved in many critical cell processes and most importantly majorly regulates T-cell development, lymphocyte recirculation, tissue-homing patterns and chemotactic responses. Recent findings have revealed a key role for S1P chemotaxis and T-cell mobilization for the onset of experimental hypertension, and elevated circulating S1P levels have been linked to several inflammation-associated diseases including hypertension in patients. In this article, we review the recent progress towards understanding how S1P and its receptors regulate immune cell trafficking and function and its potential relevance for the pathophysiology of hypertension. LINKED ARTICLES: This article is part of a themed section on Immune Targets in Hypertension. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.12/issuetoc.
Collapse
Affiliation(s)
| | - Yun Zhang
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden
| | - Hana Matuskova
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden.,Department of Neurology, University Hospital Bonn, Bonn, Germany
| | - Anja Meissner
- Department of Experimental Medical Sciences, Lund University, Lund, Sweden.,Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
38
|
Weske S, Vaidya M, Reese A, von Wnuck Lipinski K, Keul P, Bayer JK, Fischer JW, Flögel U, Nelsen J, Epple M, Scatena M, Schwedhelm E, Dörr M, Völzke H, Moritz E, Hannemann A, Rauch BH, Gräler MH, Heusch G, Levkau B. Targeting sphingosine-1-phosphate lyase as an anabolic therapy for bone loss. Nat Med 2018; 24:667-678. [PMID: 29662200 DOI: 10.1038/s41591-018-0005-y] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 02/13/2018] [Indexed: 11/09/2022]
Abstract
Sphingosine-1-phosphate (S1P) signaling influences bone metabolism, but its therapeutic potential in bone disorders has remained unexplored. We show that raising S1P levels in adult mice through conditionally deleting or pharmacologically inhibiting S1P lyase, the sole enzyme responsible for irreversibly degrading S1P, markedly increased bone formation, mass and strength and substantially decreased white adipose tissue. S1P signaling through S1P2 potently stimulated osteoblastogenesis at the expense of adipogenesis by inversely regulating osterix and PPAR-γ, and it simultaneously inhibited osteoclastogenesis by inducing osteoprotegerin through newly discovered p38-GSK3β-β-catenin and WNT5A-LRP5 pathways. Accordingly, S1P2-deficient mice were osteopenic and obese. In ovariectomy-induced osteopenia, S1P lyase inhibition was as effective as intermittent parathyroid hormone (iPTH) treatment in increasing bone mass and was superior to iPTH in enhancing bone strength. Furthermore, lyase inhibition in mice successfully corrected severe genetic osteoporosis caused by osteoprotegerin deficiency. Human data from 4,091 participants of the SHIP-Trend population-based study revealed a positive association between serum levels of S1P and bone formation markers, but not resorption markers. Furthermore, serum S1P levels were positively associated with serum calcium , negatively with PTH , and curvilinearly with body mass index. Bone stiffness, as determined through quantitative ultrasound, was inversely related to levels of both S1P and the bone formation marker PINP, suggesting that S1P stimulates osteoanabolic activity to counteract decreasing bone quality. S1P-based drugs should be considered as a promising therapeutic avenue for the treatment of osteoporotic diseases.
Collapse
Affiliation(s)
- Sarah Weske
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mithila Vaidya
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Alina Reese
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Karin von Wnuck Lipinski
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Petra Keul
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Julia K Bayer
- Institute of Pharmacology and Clinical Pharmacology, University of Düsseldorf, Düsseldorf, Germany
| | - Jens W Fischer
- Institute of Pharmacology and Clinical Pharmacology, University of Düsseldorf, Düsseldorf, Germany
| | - Ulrich Flögel
- Institute of Molecular Cardiology, University of Düsseldorf, Düsseldorf, Germany
| | - Jens Nelsen
- Institute of Inorganic Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Matthias Epple
- Institute of Inorganic Chemistry, University of Duisburg-Essen, Essen, Germany
| | - Marta Scatena
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Edzard Schwedhelm
- Institute of Clinical Pharmacology and Toxicology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.,German Centre for Cardiovascular Research (DZHK), partner site Hamburg, Hamburg, Germany
| | - Marcus Dörr
- DZHK, partner site Greifswald, Greifswald, Germany.,Department of Internal Medicine B, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Eileen Moritz
- DZHK, partner site Greifswald, Greifswald, Germany.,Institute of Pharmacology, Department of General Pharmacology, University Medicine Greifswald, Greifswald, Germany
| | - Anke Hannemann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Bernhard H Rauch
- DZHK, partner site Greifswald, Greifswald, Germany.,Institute of Pharmacology, Department of General Pharmacology, University Medicine Greifswald, Greifswald, Germany
| | - Markus H Gräler
- Institute of Pharmacology, Department of General Pharmacology, University Medicine Greifswald, Greifswald, Germany.,Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care, and Center for Molecular Biomedicine, University Hospital Jena, Jena, Germany
| | - Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bodo Levkau
- Institute for Pathophysiology, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
39
|
Yu FPS, Islam D, Sikora J, Dworski S, Gurka J, López-Vásquez L, Liu M, Kuebler WM, Levade T, Zhang H, Medin JA. Chronic lung injury and impaired pulmonary function in a mouse model of acid ceramidase deficiency. Am J Physiol Lung Cell Mol Physiol 2018; 314:L406-L420. [PMID: 29167126 PMCID: PMC5900354 DOI: 10.1152/ajplung.00223.2017] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 11/16/2017] [Accepted: 11/16/2017] [Indexed: 11/22/2022] Open
Abstract
Farber disease (FD) is a debilitating lysosomal storage disorder (LSD) caused by a deficiency of acid ceramidase (ACDase) activity due to mutations in the gene ASAH1. Patients with ACDase deficiency may develop a spectrum of clinical phenotypes. Severe cases of FD are frequently associated with neurological involvement, failure to thrive, and respiratory complications. Mice homozygous ( Asah1P361R/P361R) for an orthologous patient mutation in Asah1 recapitulate human FD. In this study, we show significant impairment in lung function, including low compliance and increased airway resistance in a mouse model of ACDase deficiency. Impaired lung mechanics in Farber mice resulted in decreased blood oxygenation and increased red blood cell production. Inflammatory cells were recruited to both perivascular and peribronchial areas of the lung. We observed large vacuolated foamy histiocytes that were full of storage material. An increase in vascular permeability led to protein leakage, edema, and impacted surfactant homeostasis in the lungs of Asah1P361R/P361R mice. Bronchial alveolar lavage fluid (BALF) extraction and analysis revealed accumulation of a highly turbid lipoprotein-like substance that was composed in part of surfactants, phospholipids, and ceramides. The phospholipid composition of BALF from Asah1P361R/P361R mice was severely altered, with an increase in both phosphatidylethanolamine (PE) and sphingomyelin (SM). Ceramides were also found at significantly higher levels in both BALF and lung tissue from Asah1P361R/P361R mice when compared with levels from wild-type animals. We demonstrate that a deficiency in ACDase leads to sphingolipid and phospholipid imbalance, chronic lung injury caused by significant inflammation, and increased vascular permeability, leading to impaired lung function.
Collapse
Affiliation(s)
- Fabian P S Yu
- Institute of Medical Science, University of Toronto , Toronto, Ontario , Canada
| | - Diana Islam
- Institute of Medical Science, University of Toronto , Toronto, Ontario , Canada
| | - Jakub Sikora
- Research Unit for Rare Diseases, Department of Pediatrics and Adolescent Medicine, Charles University, First Faculty of Medicine , Prague , Czech Republic
- Institute of Pathology, Charles University, First Faculty of Medicine and General University Hospital , Prague , Czech Republic
| | - Shaalee Dworski
- Institute of Medical Science, University of Toronto , Toronto, Ontario , Canada
| | - Jiří Gurka
- Department of Cardiology, Institute for Clinical and Experimental Medicine , Prague , Czech Republic
| | - Lucía López-Vásquez
- Institute of Medical Science, University of Toronto , Toronto, Ontario , Canada
| | - Mingyao Liu
- Institute of Medical Science, University of Toronto , Toronto, Ontario , Canada
- Department of Physiology, University of Toronto , Toronto Ontario , Canada
- University Health Network , Toronto, Ontario , Canada
| | - Wolfgang M Kuebler
- Institute of Medical Science, University of Toronto , Toronto, Ontario , Canada
- Department of Physiology, University of Toronto , Toronto Ontario , Canada
- Keenan Research Centre for Biomedical Science, Saint Michael's Hospital , Toronto, Ontario , Canada
| | - Thierry Levade
- Laboratoire de Biochimie Métabolique, Institut Fédératif de Biologie, CHU Purpan, and INSERM UMR1037 CRCT, Université de Toulouse , Toulouse , France
| | - Haibo Zhang
- Institute of Medical Science, University of Toronto , Toronto, Ontario , Canada
- Department of Physiology, University of Toronto , Toronto Ontario , Canada
- Keenan Research Centre for Biomedical Science, Saint Michael's Hospital , Toronto, Ontario , Canada
- Department of Anesthesia, University of Toronto , Toronto, Ontario , Canada
| | - Jeffrey A Medin
- Institute of Medical Science, University of Toronto , Toronto, Ontario , Canada
- University Health Network , Toronto, Ontario , Canada
- Departments of Pediatrics and Biochemistry, Medical College of Wisconsin , Milwaukee, Wisconsin
| |
Collapse
|
40
|
Fang C, Bian G, Ren P, Xiang J, Song J, Yu C, Zhang Q, Liu L, Chen K, Liu F, Zhang K, Wu C, Sun R, Hu D, Ju G, Wang J. S1P transporter SPNS2 regulates proper postnatal retinal morphogenesis. FASEB J 2018; 32:3597-3613. [DOI: 10.1096/fj.201701116r] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Chao Fang
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Ganlan Bian
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Pan Ren
- Department of Plastic SurgeryTangdu HospitalXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Jie Xiang
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Jun Song
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Caiyong Yu
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Qian Zhang
- Department of NeurologyHainan Branch of Chinese People's Liberation Army General HospitalSanyaChina
| | - Ling Liu
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Kun Chen
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Fangfang Liu
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Kun Zhang
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Chunfeng Wu
- BIOS LaboratoryBIOS Bioscience and Technology Limited CompanyGuangzhouChina
| | - Ruixia Sun
- BIOS LaboratoryBIOS Bioscience and Technology Limited CompanyGuangzhouChina
| | - Dan Hu
- Department of OphthalmologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Gong Ju
- Department of NeurobiologyXijing HospitalFourth Military Medical UniversityXi'anChina
| | - Jian Wang
- BIOS LaboratoryBIOS Bioscience and Technology Limited CompanyGuangzhouChina
- Institutes for Life Sciences and School of MedicineSouth China University of TechnologyGuangzhouChina
| |
Collapse
|
41
|
Deletion of MCP-1 Impedes Pathogenesis of Acid Ceramidase Deficiency. Sci Rep 2018; 8:1808. [PMID: 29379059 PMCID: PMC5789088 DOI: 10.1038/s41598-018-20052-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/10/2018] [Indexed: 12/22/2022] Open
Abstract
Farber Disease (FD) is an ultra-rare Lysosomal Storage Disorder caused by deficient acid ceramidase (ACDase) activity. Patients with ACDase deficiency manifest a spectrum of symptoms including formation of nodules, painful joints, and a hoarse voice. Classic FD patients will develop histiocytes in organs and die in childhood. Monocyte chemotactic protein (MCP-1; CCL2) is significantly elevated in both FD patients and a mouse model we previously generated. Here, to further study MCP-1 in FD, we created an ACDase;MCP-1 double mutant mouse. We show that deletion of MCP-1 reduced leukocytosis, delayed weight loss, and improved lifespan. Reduced inflammation and fibrosis were observed in livers from double mutant animals. Bronchial alveolar lavage fluid analyses revealed a reduction in cellular infiltrates and protein accumulation. Furthermore, reduced sphingolipid accumulation was observed in the lung and liver but not in the brain. The neurological and hematopoietic defects observed in FD mice were maintained. A compensatory cytokine response was found in the double mutants, however, that may contribute to continued signs of inflammation and injury. Taken together, targeting a reduction of MCP-1 opens the door to a better understanding of the mechanistic consequences of ceramide accumulation and may even delay the progression of FD in some organ systems.
Collapse
|
42
|
Yi JK, Xu R, Jeong E, Mileva I, Truman JP, Lin CL, Wang K, Snider J, Wen S, Obeid LM, Hannun YA, Mao C. Aging-related elevation of sphingoid bases shortens yeast chronological life span by compromising mitochondrial function. Oncotarget 2018; 7:21124-44. [PMID: 27008706 PMCID: PMC5008273 DOI: 10.18632/oncotarget.8195] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/04/2016] [Indexed: 01/08/2023] Open
Abstract
Sphingoid bases (SBs) as bioactive sphingolipids, have been implicated in aging in yeast. However, we know neither how SBs are regulated during yeast aging nor how they, in turn, regulate it. Herein, we demonstrate that the yeast alkaline ceramidases (YPC1 and YDC1) and SB kinases (LCB4 and LCB5) cooperate in regulating SBs during the aging process and that SBs shortens chronological life span (CLS) by compromising mitochondrial functions. With a lipidomics approach, we found that SBs were increased in a time-dependent manner during yeast aging. We also demonstrated that among the enzymes known for being responsible for the metabolism of SBs, YPC1 was upregulated whereas LCB4/5 were downregulated in the course of aging. This inverse regulation of YPC1 and LCB4/5 led to the aging-related upregulation of SBs in yeast and a reduction in CLS. With the proteomics-based approach (SILAC), we revealed that increased SBs altered the levels of proteins related to mitochondria. Further mechanistic studies demonstrated that increased SBs inhibited mitochondrial fusion and caused fragmentation, resulting in decreases in mtDNA copy numbers, ATP levels, mitochondrial membrane potentials, and oxygen consumption. Taken together, these results suggest that increased SBs mediate the aging process by impairing mitochondrial structural integrity and functions.
Collapse
Affiliation(s)
- Jae Kyo Yi
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA.,Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Ruijuan Xu
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Eunmi Jeong
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Izolda Mileva
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | | | - Chih-Li Lin
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA.,Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Kai Wang
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Justin Snider
- Graduate Program in Molecular and Cellular Biology, Stony Brook University, Stony Brook, NY, USA.,Department of Medicine, Stony Brook University, Stony Brook, NY, USA
| | - Sally Wen
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Lina M Obeid
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA.,Northport Veterans Affairs Medical Center, Northport, NY, USA
| | - Yusuf A Hannun
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| | - Cungui Mao
- Department of Medicine, Stony Brook University, Stony Brook, NY, USA.,Stony Brook Cancer Center, Stony Brook, NY, USA
| |
Collapse
|
43
|
Li F, Xu R, Low BE, Lin CL, Garcia-Barros M, Schrandt J, Mileva I, Snider A, Luo CK, Jiang XC, Li MS, Hannun YA, Obeid LM, Wiles MV, Mao C. Alkaline ceramidase 2 is essential for the homeostasis of plasma sphingoid bases and their phosphates. FASEB J 2018; 32:3058-3069. [PMID: 29401619 DOI: 10.1096/fj.201700445rr] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Sphingosine-1-phosphate (S1P) plays important roles in cardiovascular development and immunity. S1P is abundant in plasma because erythrocytes-the major source of S1P-lack any S1P-degrading activity; however, much remains unclear about the source of the plasma S1P precursor, sphingosine (SPH), derived mainly from the hydrolysis of ceramides by the action of ceramidases that are encoded by 5 distinct genes, acid ceramidase 1 ( ASAH1)/ Asah1, ASAH2/ Asah2, alkaline ceramidase 1 ( ACER1)/ Acer1, ACER2/ Acer2, and ACER3/ Acer3, in humans/mice. Previous studies have reported that knocking out Asah1 or Asah2 failed to reduce plasma SPH and S1P levels in mice. In this study, we show that knocking out Acer1 or Acer3 also failed to reduce the blood levels of SPH or S1P in mice. In contrast, knocking out Acer2 from either whole-body or the hematopoietic lineage markedly decreased the blood levels of SPH and S1P in mice. Of interest, knocking out Acer2 from whole-body or the hematopoietic lineage also markedly decreased the levels of dihydrosphingosine (dhSPH) and dihydrosphingosine-1-phosphate (dhS1P) in blood. Taken together, these results suggest that ACER2 plays a key role in the maintenance of high plasma levels of sphingoid base-1-phosphates-S1P and dhS1P-by controlling the generation of sphingoid bases-SPH and dhSPH-in hematopoietic cells.-Li, F., Xu, R., Low, B. E., Lin, C.-L., Garcia-Barros, M., Schrandt, J., Mileva, I., Snider, A., Luo, C. K., Jiang, X.-C., Li, M.-S., Hannun, Y. A., Obeid, L. M., Wiles, M. V., Mao, C. Alkaline ceramidase 2 is essential for the homeostasis of plasma sphingoid bases and their phosphates.
Collapse
Affiliation(s)
- Fang Li
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Ruijuan Xu
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Benjamin E Low
- Technology Evaluation and Development, The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Chih-Li Lin
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Monica Garcia-Barros
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Jennifer Schrandt
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Izolda Mileva
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Ashley Snider
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA.,Northport Veterans Administration Medical Center, Northport, New York, USA
| | - Catherine K Luo
- Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Xian-Cheng Jiang
- Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, New York, USA
| | - Ming-Song Li
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yusuf A Hannun
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA
| | - Lina M Obeid
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA.,Northport Veterans Administration Medical Center, Northport, New York, USA
| | - Michael V Wiles
- Technology Evaluation and Development, The Jackson Laboratory, Bar Harbor, Maine, USA
| | - Cungui Mao
- Department of Medicine, State University of New York at Stony Brook, Stony Brook, New York, USA.,Cancer Center, State University of New York at Stony Brook, Stony Brook, New York, USA
| |
Collapse
|
44
|
Huwiler A, Pfeilschifter J. Sphingolipid signaling in renal fibrosis. Matrix Biol 2018; 68-69:230-247. [PMID: 29343457 DOI: 10.1016/j.matbio.2018.01.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 01/08/2018] [Accepted: 01/08/2018] [Indexed: 12/28/2022]
Abstract
Over the last decade, various sphingolipid subspecies have gained increasing attention as important signaling molecules that regulate a multitude of physiological and pathophysiological processes including inflammation and tissue remodeling. These mediators include ceramide, sphingosine 1-phosphate (S1P), the cerebroside glucosylceramide, lactosylceramide, and the gangliosides GM3 and Gb3. These lipids have been shown to accumulate in various chronic kidney diseases that typically end in renal fibrosis and ultimately renal failure. This review will summarize the effects and contributions of those enzymes that regulate the generation and interconversion of these lipids, notably the acid sphingomyelinase, the acid sphingomyelinase-like protein SMPDL3B, the sphingosine kinases, the S1P lyase, the glucosylceramide synthase, the GM3 synthase, and the α-galactosidase A, to renal fibrotic diseases. Strategies of manipulating these enzymes for therapeutic purposes and the impact of existing drugs on renal pathologies will be discussed.
Collapse
Affiliation(s)
- Andrea Huwiler
- Institute of Pharmacology, University of Bern, Inselspital INO-F, CH-3010 Bern, Switzerland.
| | - Josef Pfeilschifter
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe- University, Frankfurt am Main, Germany
| |
Collapse
|
45
|
S1P Lyase Regulation of Thymic Egress and Oncogenic Inflammatory Signaling. Mediators Inflamm 2017; 2017:7685142. [PMID: 29333002 PMCID: PMC5733215 DOI: 10.1155/2017/7685142] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 09/13/2017] [Indexed: 12/17/2022] Open
Abstract
Sphingosine-1-phosphate (S1P) is a potent lipid signaling molecule that regulates pleiotropic biological functions including cell migration, survival, angiogenesis, immune cell trafficking, inflammation, and carcinogenesis. It acts as a ligand for a family of cell surface receptors. S1P concentrations are high in blood and lymph but low in tissues, especially the thymus and lymphoid organs. S1P chemotactic gradients are essential for lymphocyte egress and other aspects of physiological cell trafficking. S1P is irreversibly degraded by S1P lyase (SPL). SPL regulates lymphocyte trafficking, inflammation and other physiological and pathological processes. For example, SPL located in thymic dendritic cells acts as a metabolic gatekeeper that controls the normal egress of mature T lymphocytes from the thymus into the circulation, whereas SPL deficiency in gut epithelial cells promotes colitis and colitis-associated carcinogenesis (CAC). Recently, we identified a complex syndrome comprised of nephrosis, adrenal insufficiency, and immunological defects caused by inherited mutations in human SGPL1, the gene encoding SPL. In the present article, we review current evidence supporting the role of SPL in thymic egress, inflammation, and cancer. Lastly, we summarize recent progress in understanding other SPL functions, its role in inherited disease, and SPL targeting for therapeutic purposes.
Collapse
|
46
|
Abou Daher A, El Jalkh T, Eid AA, Fornoni A, Marples B, Zeidan YH. Translational Aspects of Sphingolipid Metabolism in Renal Disorders. Int J Mol Sci 2017; 18:E2528. [PMID: 29186855 PMCID: PMC5751131 DOI: 10.3390/ijms18122528] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 11/16/2017] [Accepted: 11/17/2017] [Indexed: 12/13/2022] Open
Abstract
Sphingolipids, long thought to be passive components of biological membranes with merely a structural role, have proved throughout the past decade to be major players in the pathogenesis of many human diseases. The study and characterization of several genetic disorders like Fabry's and Tay Sachs, where sphingolipid metabolism is disrupted, leading to a systemic array of clinical symptoms, have indeed helped elucidate and appreciate the importance of sphingolipids and their metabolites as active signaling molecules. In addition to being involved in dynamic cellular processes like apoptosis, senescence and differentiation, sphingolipids are implicated in critical physiological functions such as immune responses and pathophysiological conditions like inflammation and insulin resistance. Interestingly, the kidneys are among the most sensitive organ systems to sphingolipid alterations, rendering these molecules and the enzymes involved in their metabolism, promising therapeutic targets for numerous nephropathic complications that stand behind podocyte injury and renal failure.
Collapse
Affiliation(s)
- Alaa Abou Daher
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Tatiana El Jalkh
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Assaad A Eid
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon.
| | - Alessia Fornoni
- Department of Medicine, Peggy and Harold Katz Family Drug Discovery Center, University of Miami, Miami, FL 33136, USA.
| | - Brian Marples
- Department of Radiation Oncology, Miller School of Medicine/Sylvester Cancer Center, University of Miami, Miami, FL 33136, USA.
| | - Youssef H Zeidan
- Department of Anatomy, Cell Biology and Physiology, Faculty of Medicine, American University of Beirut, Beirut 1107 2020, Lebanon.
- Department of Radiation Oncology, American University of Beirut Medical Center, Beirut 1107 2020, Lebanon.
| |
Collapse
|
47
|
Wollny T, Wątek M, Durnaś B, Niemirowicz K, Piktel E, Żendzian-Piotrowska M, Góźdź S, Bucki R. Sphingosine-1-Phosphate Metabolism and Its Role in the Development of Inflammatory Bowel Disease. Int J Mol Sci 2017; 18:ijms18040741. [PMID: 28362332 PMCID: PMC5412326 DOI: 10.3390/ijms18040741] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/12/2022] Open
Abstract
Beyond their role as structural molecules, sphingolipids are involved in many important cellular processes including cell proliferation, apoptosis, inflammation, and migration. Altered sphingolipid metabolism is observed in many pathological conditions including gastrointestinal diseases. Inflammatory bowel disease (IBD) represents a state of complex, unpredictable, and destructive inflammation of unknown origin within the gastrointestinal tract. The mechanisms explaining the pathophysiology of IBD involve signal transduction pathways regulating gastro-intestinal system’s immunity. Progressive intestinal tissue destruction observed in chronic inflammation may be associated with an increased risk of colon cancer. Sphingosine-1-phosphate (S1P), a sphingolipid metabolite, functions as a cofactor in inflammatory signaling and becomes a target in the treatment of IBD, which might prevent its conversion to cancer. This paper summarizes new findings indicating the impact of (S1P) on IBD development and IBD-associated carcinogenesis.
Collapse
Affiliation(s)
- Tomasz Wollny
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734 Kielce, Poland.
| | - Marzena Wątek
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734 Kielce, Poland.
- Department of Microbiology and Immunology, The Faculty of Health Sciences of the Jan Kochanowski University in Kielce, Aleja IX Wieków Kielc, 25-317 Kielce, Poland.
| | - Bonita Durnaś
- Department of Microbiology and Immunology, The Faculty of Health Sciences of the Jan Kochanowski University in Kielce, Aleja IX Wieków Kielc, 25-317 Kielce, Poland.
| | - Katarzyna Niemirowicz
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland.
| | - Ewelina Piktel
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland.
| | | | - Stanisław Góźdź
- Holy Cross Oncology Center of Kielce, Artwińskiego 3, 25-734 Kielce, Poland.
- Department of Microbiology and Immunology, The Faculty of Health Sciences of the Jan Kochanowski University in Kielce, Aleja IX Wieków Kielc, 25-317 Kielce, Poland.
| | - Robert Bucki
- Department of Microbiological and Nanobiomedical Engineering, Medical University of Białystok, 15-222 Białystok, Poland.
| |
Collapse
|
48
|
Phytosphingosine degradation pathway includes fatty acid α-oxidation reactions in the endoplasmic reticulum. Proc Natl Acad Sci U S A 2017; 114:E2616-E2623. [PMID: 28289220 DOI: 10.1073/pnas.1700138114] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Although normal fatty acids (FAs) are degraded via β-oxidation, unusual FAs such as 2-hydroxy (2-OH) FAs and 3-methyl-branched FAs are degraded via α-oxidation. Phytosphingosine (PHS) is one of the long-chain bases (the sphingolipid components) and exists in specific tissues, including the epidermis and small intestine in mammals. In the degradation pathway, PHS is converted to 2-OH palmitic acid and then to pentadecanoic acid (C15:0-COOH) via FA α-oxidation. However, the detailed reactions and genes involved in the α-oxidation reactions of the PHS degradation pathway have yet to be determined. In the present study, we reveal the entire PHS degradation pathway: PHS is converted to C15:0-COOH via six reactions [phosphorylation, cleavage, oxidation, CoA addition, cleavage (C1 removal), and oxidation], in which the last three reactions correspond to the α-oxidation. The aldehyde dehydrogenase ALDH3A2 catalyzes both the first and second oxidation reactions (fatty aldehydes to FAs). In Aldh3a2-deficient cells, the unmetabolized fatty aldehydes are reduced to fatty alcohols and are incorporated into ether-linked glycerolipids. We also identify HACL2 (2-hydroxyacyl-CoA lyase 2) [previous name, ILVBL; ilvB (bacterial acetolactate synthase)-like] as the major 2-OH acyl-CoA lyase involved in the cleavage (C1 removal) reaction in the FA α-oxidation of the PHS degradation pathway. HACL2 is localized in the endoplasmic reticulum. Thus, in addition to the already-known FA α-oxidation in the peroxisomes, we have revealed the existence of FA α-oxidation in the endoplasmic reticulum in mammals.
Collapse
|
49
|
Characterization of cholesterol homeostasis in sphingosine-1-phosphate lyase-deficient fibroblasts reveals a Niemann-Pick disease type C-like phenotype with enhanced lysosomal Ca 2+ storage. Sci Rep 2017; 7:43575. [PMID: 28262793 PMCID: PMC5337937 DOI: 10.1038/srep43575] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 01/25/2017] [Indexed: 02/08/2023] Open
Abstract
Sphingosine-1-phosphate (S1P) lyase irreversibly cleaves S1P, thereby catalysing the ultimate step of sphingolipid degradation. We show here that embryonic fibroblasts from S1P lyase-deficient mice (Sgpl1−/−-MEFs), in which S1P and sphingosine accumulate, have features of Niemann-Pick disease type C (NPC) cells. In the presence of serum, overall cholesterol content was elevated in Sgpl1−/−-MEFs, due to upregulation of the LDL receptor and enhanced cholesterol uptake. Despite this, activation of sterol regulatory element-binding protein-2 was increased in Sgpl1−/−-MEFs, indicating a local lack of cholesterol at the ER. Indeed, free cholesterol was retained in NPC1-containing vesicles, which is a hallmark of NPC. Furthermore, upregulation of amyloid precursor protein in Sgpl1−/−-MEFs was mimicked by an NPC1 inhibitor in Sgpl1+/+-MEFs and reduced by overexpression of NPC1. Lysosomal pH was not altered by S1P lyase deficiency, similar to NPC. Interestingly, lysosomal Ca2+ content and bafilomycin A1-induced [Ca2+]i increases were enhanced in Sgpl1−/−-MEFs, contrary to NPC. These results show that both a primary defect in cholesterol trafficking and S1P lyase deficiency cause overlapping phenotypic alterations, and challenge the present view on the role of sphingosine in lysosomal Ca2+ homeostasis.
Collapse
|
50
|
Prasad R, Hadjidemetriou I, Maharaj A, Meimaridou E, Buonocore F, Saleem M, Hurcombe J, Bierzynska A, Barbagelata E, Bergadá I, Cassinelli H, Das U, GOSgene, Krone R, Hacihamdioglu B, Sari E, Yesilkaya E, Storr HL, Clemente M, Fernandez-Cancio M, Camats N, Ram N, Achermann JC, Van Veldhoven PP, Guasti L, Braslavsky D, Guran T, Metherell LA. Sphingosine-1-phosphate lyase mutations cause primary adrenal insufficiency and steroid-resistant nephrotic syndrome. J Clin Invest 2017; 127:942-953. [PMID: 28165343 PMCID: PMC5330744 DOI: 10.1172/jci90171] [Citation(s) in RCA: 116] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Accepted: 12/12/2016] [Indexed: 12/26/2022] Open
Abstract
Primary adrenal insufficiency is life threatening and can present alone or in combination with other comorbidities. Here, we have described a primary adrenal insufficiency syndrome and steroid-resistant nephrotic syndrome caused by loss-of-function mutations in sphingosine-1-phosphate lyase (SGPL1). SGPL1 executes the final decisive step of the sphingolipid breakdown pathway, mediating the irreversible cleavage of the lipid-signaling molecule sphingosine-1-phosphate (S1P). Mutations in other upstream components of the pathway lead to harmful accumulation of lysosomal sphingolipid species, which are associated with a series of conditions known as the sphingolipidoses. In this work, we have identified 4 different homozygous mutations, c.665G>A (p.R222Q), c.1633_1635delTTC (p.F545del), c.261+1G>A (p.S65Rfs*6), and c.7dupA (p.S3Kfs*11), in 5 families with the condition. In total, 8 patients were investigated, some of whom also manifested other features, including ichthyosis, primary hypothyroidism, neurological symptoms, and cryptorchidism. Sgpl1-/- mice recapitulated the main characteristics of the human disease with abnormal adrenal and renal morphology. Sgpl1-/- mice displayed disrupted adrenocortical zonation and defective expression of steroidogenic enzymes as well as renal histology in keeping with a glomerular phenotype. In summary, we have identified SGPL1 mutations in humans that perhaps represent a distinct multisystemic disorder of sphingolipid metabolism.
Collapse
Affiliation(s)
- Rathi Prasad
- Centre for Endocrinology, William Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Irene Hadjidemetriou
- Centre for Endocrinology, William Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Avinaash Maharaj
- Centre for Endocrinology, William Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Eirini Meimaridou
- Centre for Endocrinology, William Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Federica Buonocore
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Moin Saleem
- Children’s and Academic Renal Unit, University of Bristol, Bristol, United Kingdom
| | - Jenny Hurcombe
- Children’s and Academic Renal Unit, University of Bristol, Bristol, United Kingdom
| | - Agnieszka Bierzynska
- Children’s and Academic Renal Unit, University of Bristol, Bristol, United Kingdom
| | - Eliana Barbagelata
- Servicio de Nefrología, Hospital de Niños “Ricardo Gutiérrez,” Buenos Aires, Argentina
| | - Ignacio Bergadá
- Centro de Investigaciones Endocrinológicas “Dr. Cesar Bergadá” (CEDIE) – CONICET – FEI – División de Endocrinología, Hospital de Niños “Ricardo Gutiérrez,” Buenos Aires, Argentina
| | - Hamilton Cassinelli
- Centro de Investigaciones Endocrinológicas “Dr. Cesar Bergadá” (CEDIE) – CONICET – FEI – División de Endocrinología, Hospital de Niños “Ricardo Gutiérrez,” Buenos Aires, Argentina
| | - Urmi Das
- Alderhey Children’s Hospital NHS Foundation Trust, Eaton Road, Liverpool, United Kingdom
| | - GOSgene
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
- The center is detailed in the Supplemental Acknowledgments
| | - Ruth Krone
- Birmingham Children’s Hospital, Birmingham, United Kingdom
| | - Bulent Hacihamdioglu
- Health Sciences University, Suleymaniye Maternity and Children’s Training and Research Hospital, Department of Paediatric Endocrinology and Diabetes, Istanbul, Turkey
| | - Erkan Sari
- Gulhane Military Medical School Department of Paediatric Endocrinology and Diabetes, Ankara, Turkey
| | - Ediz Yesilkaya
- Gulhane Military Medical School Department of Paediatric Endocrinology and Diabetes, Ankara, Turkey
| | - Helen L. Storr
- Centre for Endocrinology, William Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Maria Clemente
- Growth and Development Research Unit, Vall d’Hebron Research Institute (VHIR), Hospital Vall d’Hebron, CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Monica Fernandez-Cancio
- Growth and Development Research Unit, Vall d’Hebron Research Institute (VHIR), Hospital Vall d’Hebron, CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Nuria Camats
- Growth and Development Research Unit, Vall d’Hebron Research Institute (VHIR), Hospital Vall d’Hebron, CIBERER, Instituto de Salud Carlos III, Barcelona, Spain
| | - Nanik Ram
- Department of Medicine, Aga Khan University Hospital, Karachi, Pakistan
| | - John C. Achermann
- Genetics and Genomic Medicine, UCL Great Ormond Street Institute of Child Health, University College London, London, United Kingdom
| | - Paul P. Van Veldhoven
- Laboratory of Lipid Biochemistry and Protein Interactions (LIPIT), Campus Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Leonardo Guasti
- Centre for Endocrinology, William Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| | - Debora Braslavsky
- Centro de Investigaciones Endocrinológicas “Dr. Cesar Bergadá” (CEDIE) – CONICET – FEI – División de Endocrinología, Hospital de Niños “Ricardo Gutiérrez,” Buenos Aires, Argentina
| | - Tulay Guran
- Marmara University, Department of Paediatric Endocrinology and Diabetes, Istanbul, Turkey
| | - Louise A. Metherell
- Centre for Endocrinology, William Harvey Research Institute, John Vane Science Centre, Queen Mary, University of London, Charterhouse Square, London, United Kingdom
| |
Collapse
|