1
|
Figueira MI, Carvalho TMA, Macário-Monteiro J, Cardoso HJ, Correia S, Vaz CV, Duarte AP, Socorro S. The Pros and Cons of Estrogens in Prostate Cancer: An Update with a Focus on Phytoestrogens. Biomedicines 2024; 12:1636. [PMID: 39200101 PMCID: PMC11351860 DOI: 10.3390/biomedicines12081636] [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: 06/26/2024] [Revised: 07/14/2024] [Accepted: 07/20/2024] [Indexed: 09/01/2024] Open
Abstract
The role of estrogens in prostate cancer (PCa) is shrouded in mystery, with its actions going from angelic to devilish. The findings by Huggins and Hodges establishing PCa as a hormone-sensitive cancer have provided the basis for using estrogens in therapy. However, despite the clinical efficacy in suppressing tumor growth and the panoply of experimental evidence describing its anticarcinogenic effects, estrogens were abolished from PCa treatment because of the adverse secondary effects. Notwithstanding, research work over the years has continued investigating the effects of estrogens, reporting their pros and cons in prostate carcinogenesis. In contrast with the beneficial therapeutic effects, many reports have implicated estrogens in the disruption of prostate cell fate and tissue homeostasis. On the other hand, epidemiological data demonstrating the lower incidence of PCa in Eastern countries associated with a higher consumption of phytoestrogens support the beneficial role of estrogens in counteracting cancer development. Many studies have investigated the effects of phytoestrogens and the underlying mechanisms of action, which may contribute to developing safe estrogen-based anti-PCa therapies. This review compiles the existing data on the anti- and protumorigenic actions of estrogens and summarizes the anticancer effects of several phytoestrogens, highlighting their promising features in PCa treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Sílvia Socorro
- CICS-UBI, Centro de Investigação em Ciências da Saúde, Universidade da Beira Interior, 6200-506 Covilhã, Portugal; (M.I.F.)
| |
Collapse
|
2
|
Resop RS, Salvatore B, Kim SJ, Gordon BR, Blom B, Vatakis DN, Uittenbogaart CH. HIV-1 Infection Results in Sphingosine-1-Phosphate Receptor 1 Dysregulation in the Human Thymus. Int J Mol Sci 2023; 24:13865. [PMID: 37762169 PMCID: PMC10531245 DOI: 10.3390/ijms241813865] [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: 07/28/2023] [Revised: 08/31/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Regeneration of functional naïve T lymphocytes following the onset of human immunodeficiency virus (HIV) infection remains a crucial issue for people living with HIV (PLWH), even when adhering to antiretroviral therapy (ART). Thus far, reports on the impact of HIV-1 infection on the entry of thymic precursors and the egress of functional naïve T lymphocytes to and from the thymus are limited. We examined the impact of HIV-1 on Sphingosine-1-phosphate (S1P) signaling, which governs the egress of functional naïve thymocytes from the thymus to the periphery. Using in vitro experiments with primary human thymocytes and in vivo and ex vivo studies with humanized mice, we show that HIV-1 infection results in upregulation of the expression of S1P receptor 1 (S1PR1) in the human thymus. Intriguingly, this upregulation occurs during intrathymic infection (direct infection of the human thymic implant) as well as systemic infection in humanized mice. Moreover, considering the dysregulation of pro- and anti-inflammatory cytokines in infected thymi, the increased expression of S1PR1 in response to in vitro exposure to Interferon-Beta (IFN-β) and Tumor Necrosis Factor-Alpha (TNF-α) indicates that cytokine dysregulation following HIV infection may contribute to upregulation of S1PR1. Finally, an increased presence of CD3hiCD69- (fully mature) as well as CD3hiCD69+ (less mature) T cells in the spleen during HIV infection in humanized mice, combined with earlier expression of S1PR1 during thymocyte development, suggests that upregulation of S1PR1 may translate to increased or accelerated egress from the thymus. The egress of thymocytes that are not functionally mature from the thymus to peripheral blood and lymphoid organs may have implications for the immune function of PLWH.
Collapse
Affiliation(s)
- Rachel S. Resop
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (R.S.R.); (B.S.); (S.J.K.)
- UCLA AIDS Institute and Center for AIDS Research, University of California, Los Angeles, CA 90095, USA;
| | - Bradley Salvatore
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (R.S.R.); (B.S.); (S.J.K.)
| | - Shawn J. Kim
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (R.S.R.); (B.S.); (S.J.K.)
| | - Brent R. Gordon
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (R.S.R.); (B.S.); (S.J.K.)
| | - Bianca Blom
- Amsterdam University Medical Centers, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands;
| | - Dimitrios N. Vatakis
- UCLA AIDS Institute and Center for AIDS Research, University of California, Los Angeles, CA 90095, USA;
- Department of Medicine, Division of Hematology-Oncology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| | - Christel H. Uittenbogaart
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA 90095, USA; (R.S.R.); (B.S.); (S.J.K.)
- UCLA AIDS Institute and Center for AIDS Research, University of California, Los Angeles, CA 90095, USA;
- Department of Pediatrics, David Geffen School of Medicine at UCLA, University of California, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
3
|
Gray N, Limberg MM, Wiebe D, Weihrauch T, Langner A, Brandt N, Bräuer AU, Raap U. Differential Upregulation and Functional Activity of S1PR1 in Human Peripheral Blood Basophils of Atopic Patients. Int J Mol Sci 2022; 23:16117. [PMID: 36555755 PMCID: PMC9785255 DOI: 10.3390/ijms232416117] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Basophils are key effector cells in atopic diseases, and the signaling sphingolipid Sphigosine-1-phosphate (S1P) is emerging as an important mediator in these conditions. The possible interaction of S1P and basophils and the resulting biological effects have not yet been studied. We hypothesize that S1P influences the function of basophils in atopy and aim to elucidate the modes of interaction. S1P receptor (S1PR) expression in human peripheral blood basophils from atopic and non-atopic patients was assessed through qRT-PCR and flow cytometry analysis. Functional effects of S1P were assessed through a basophil activation test (BAT), calcium flux, apoptosis, and chemotaxis assays. Immunofluorescence staining was performed to visualize intracellular S1P. Human basophils express S1PR1, S1PR2, S1PR3, and S1PR4 on the mRNA level. 0.1 µM S1P have anti-apoptotic, while 10 µM exhibits apoptotic effects on basophils. Basophils from atopic patients show less chemotactic activity in response to S1P than those from healthy donors. Protein expression of S1PR1 is downregulated in atopic patients, and basophils in lesional AD skin possess intracellular S1P. These findings suggest that the interaction of S1P and basophils might be an important factor in the pathophysiology of atopy.
Collapse
Affiliation(s)
- Natalie Gray
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Division of Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Maren M. Limberg
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Daniela Wiebe
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Tobias Weihrauch
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Anna Langner
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Nicola Brandt
- Division of Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Anja U. Bräuer
- Division of Anatomy, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
| | - Ulrike Raap
- Division of Experimental Allergy and Immunodermatology, School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, 26129 Oldenburg, Germany
- University Clinic of Dermatology and Allergy, University of Oldenburg, 26133 Oldenburg, Germany
| |
Collapse
|
4
|
Birchenall KA, Welsh GI, López Bernal A. The feto-placental metabolome of spontaneous labour is not reproduced following induction of labour. Placenta 2022; 129:111-118. [PMID: 36288646 DOI: 10.1016/j.placenta.2022.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 10/04/2022] [Accepted: 10/09/2022] [Indexed: 12/14/2022]
Abstract
INTRODUCTION The mechanism for human labour remains poorly understood, limiting our ability to manage complications including spontaneous preterm birth and induction of labour (IOL). The study of fetal signals poses specific challenges. Metabolomic analysis of maternal blood, the cord artery (CA), and cord vein (CV), allows simultaneous interrogation of multiple metabolic pathways associated with different modes of labour onset and birth. METHODS Global mass spectrometry metabolomics analysis was performed on serial samples collected from participants during pregnancy, in latent phase of labour, and following birth (CA, CV, and intervillous (IV) blood), from those who spontaneously laboured and birthed vaginally (SL group), had IOL and birthed vaginally (IOL group), or birthed via elective caesarean section (no labour; ECS group). RESULTS There were clear differences in fetal and maternal steroid, arachidonate and sphingosine pathways between the SL and IOL groups, despite similar uterine contractions and vaginal birth. The CA/CV ratio for key steroids of the IOL group were more alike the ECS group than the SL group, including progesterone (CA/CV ratio for: SL group=3.5; IOL group=0.5; and ECS group=0.5), and oestriol (CA/CV ratio for: SL group=4.3; IOL group=0.4; and for ECS group=0.2). There were no such changes in the maternal samples. DISCUSSION These findings indicate that IOL does not reproduce the pathways activated in spontaneous labour. The decreased placental progesterone production observed with spontaneous labour may represent a local intrauterine progesterone withdrawal, which, together with other signals, would activate parturition pathways involving arachidonate and sphingosine metabolism.
Collapse
Affiliation(s)
- Katherine Alice Birchenall
- Department of Obstetrics and Gynaecology, St Michael's Hospital, Southwell Street, Bristol, BS2 8EG, UK; Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | - Gavin Iain Welsh
- Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| | - Andrés López Bernal
- Department of Obstetrics and Gynaecology, St Michael's Hospital, Southwell Street, Bristol, BS2 8EG, UK; Translational Health Sciences, University of Bristol, Dorothy Hodgkin Building, Whitson Street, Bristol, BS1 3NY, UK.
| |
Collapse
|
5
|
Abdel Baky NA, Al-Najjar AH, Elariny HA, Sallam AS, Mohammed AA. Pramipexole and Lactoferrin ameliorate Cyclophosphamide-Induced haemorrhagic cystitis via targeting Sphk1/S1P/MAPK, TLR-4/NF-κB, and NLRP3/caspase-1/IL-1β signalling pathways and modulating the Nrf2/HO-1 pathway. Int Immunopharmacol 2022; 112:109282. [DOI: 10.1016/j.intimp.2022.109282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 08/30/2022] [Accepted: 09/22/2022] [Indexed: 11/28/2022]
|
6
|
Youssef NS, Elzatony AS, Abdel Baky NA. Diacerein attenuate LPS-induced acute lung injury via inhibiting ER stress and apoptosis: Impact on the crosstalk between SphK1/S1P, TLR4/NFκB/STAT3, and NLRP3/IL-1β signaling pathways. Life Sci 2022; 308:120915. [PMID: 36055546 DOI: 10.1016/j.lfs.2022.120915] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/16/2022] [Accepted: 08/24/2022] [Indexed: 10/31/2022]
Abstract
AIMS Acute lung injury (ALI) is a life-threatening clinical problem with high mortality rate and limited treatments or preventive options that represents a major challenge for clinicians. Diacerein (DIA) is a multi-target anthraquinone derivative with potent anti-inflammatory action. The aim of this study is to assess the protective effect of DIA and its potential molecular targets against lipopolysaccharide (LPS)-induced ALI in rats. MATERIALS AND METHODS Adult male Sprague-Dawley rats were orally administrated DIA (50 mg/kg) for 5 consecutive days followed by a single intraperitoneal injection of LPS (5mg/kg). KEY FINDINGS DIA mitigated oxidative lung injury in LPS-challenged rats via significantly decreasing lung wet/dry (W/D) ratio, inflammatory cells infiltration, and lipid peroxidation, with concomitant elevation in enzymatic and non-enzymatic antioxidant levels in lung tissue. Likewise, DIA alleviated endoplasmic reticulum stress and markedly halted inflammation triggered by LPS challenge in pulmonary tissue by suppressing NLRP3/IL-1β and TLR4/NF-κB signaling with parallel decrease in proinflammatory cytokine levels. Interestingly, DIA down regulated Sphk1/S1P axis, reduced GSK-3β and STAT3 proteins expression, and markedly decreased caspase-3 besides increasing Bcl-2 levels in lung tissue of LPS-challenged animals. These biochemical findings was simultaneously associated with marked improvement in histological alterations of lung tissue. SIGNIFICANCE These findings verify the protective effect of DIA against LPS-induced ALI through targeting oxidative stress, endoplasmic reticulum stress, and apoptosis. Importantly, DIA halted the hyperinflammatory state triggered by LPS via multi-faceted inhibitory effect on different signaling pathways, hence DIA could potentially reduce mortality in patients with ALI.
Collapse
Affiliation(s)
- Nagwa Salah Youssef
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Asmaa Sameer Elzatony
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt
| | - Nayira A Abdel Baky
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al-Azhar University, Cairo, Egypt.
| |
Collapse
|
7
|
Plasma membrane effects of sphingolipid-synthesis inhibition by myriocin in CHO cells: a biophysical and lipidomic study. Sci Rep 2022; 12:955. [PMID: 35046440 PMCID: PMC8770663 DOI: 10.1038/s41598-021-04648-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 11/08/2021] [Indexed: 11/08/2022] Open
Abstract
Suppression of a specific gene effect can be achieved by genetic as well as chemical methods. Each approach may hide unexpected drawbacks, usually in the form of side effects. In the present study, the specific inhibitor myriocin was used to block serine palmitoyltransferase (SPT), the first enzyme in the sphingolipid synthetic pathway, in CHO cells. The subsequent biophysical changes in plasma membranes were measured and compared with results obtained with a genetically modified CHO cell line containing a defective SPT (the LY-B cell line). Similar effects were observed with both approaches: sphingomyelin values were markedly decreased in myriocin-treated CHO cells and, in consequence, their membrane molecular order (measured as laurdan general polarization) and mechanical resistance (AFM-measured breakthrough force values) became lower than in the native, non-treated cells. Cells treated with myriocin reacted homeostatically to maintain membrane order, synthesizing more fully saturated and less polyunsaturated GPL than the non-treated ones, although they achieved it only partially, their plasma membranes remaining slightly more fluid and more penetrable than those from the control cells. The good agreement between results obtained with very different tools, such as genetically modified and chemically treated cells, reinforces the use of both methods and demonstrates that both are adequate for their intended use, i.e. the complete and specific inhibition of sphingolipid synthesis in CHO cells, without apparent unexpected effects.
Collapse
|
8
|
Jin H, Ma H, Gan N, Wang H, Li Y, Wang L, Song L. Non-targeted metabolomic profiling of filamentous cyanobacteria Aphanizomenon flos-aquae exposed to a concentrated culture filtrate of Microcystis aeruginosa. HARMFUL ALGAE 2022; 111:102170. [PMID: 35016758 DOI: 10.1016/j.hal.2021.102170] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/13/2021] [Accepted: 12/20/2021] [Indexed: 06/14/2023]
Abstract
Microcystis and Aphanizomenon are two toxic cyanobacteria genera, which frequently cause blooms in freshwater lakes. In some cases, succession of these two genera was observed in natural water bodies. Among the diverse factors contributing to such succession of dominant cyanobacterial genera, an allelopathic effect was proposed to be involved after the growth inhibitory effect of several Microcystis species on A. flos-aquae was investigated. However, the response of target species exposed to Microcystis are poorly described. In the present study, we used two toxic cyanobacteria strains, Aphanizomenon flos-aquae (Aph1395) and Microcystis aeruginosa strain 905 (Ma905) as research subjects. Aph1395 was inhibited with a necessarily concentrated culture filtrate of Ma905 (MA905-SPE), and the response of the inhibited Aph1395 cells was explored via non-targeted metabolomic profiling. In total, 3735 features were significantly different in the Aph1395 treated with Ma905-SPE vs. those treated with BG11 medium. Among them, the annotations of 146 differential features were considered to be confident via MS/MS spectrum matching analysis. Based on the reported physiological functions of the annotated differential features, we proposed a putative model that in the growth-inhibited Aph1395, a suite of increased or decreased features with activities in apoptosis, growth inhibition, and stress response processes contributed to, or defended against, the allelopathic effect caused by Ma905. Our findings provide insights into the interaction between the bloom forming cyanobacterial species that share the same ecological environment.
Collapse
Affiliation(s)
- Hu Jin
- Center for Microalgal Biotechnology and Biofuels, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P. R. China
| | - Haiyan Ma
- Center for Microalgal Biotechnology and Biofuels, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P. R. China.
| | - Nanqin Gan
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P. R. China
| | - Hongxia Wang
- Center for Microalgal Biotechnology and Biofuels, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P. R. China
| | - Yanhua Li
- Center for Microalgal Biotechnology and Biofuels, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P. R. China
| | - Lan Wang
- Center for Microalgal Biotechnology and Biofuels, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P. R. China
| | - Lirong Song
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, P. R. China
| |
Collapse
|
9
|
Fat of the Gut: Epithelial Phospholipids in Inflammatory Bowel Diseases. Int J Mol Sci 2021; 22:ijms222111682. [PMID: 34769112 PMCID: PMC8584226 DOI: 10.3390/ijms222111682] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/20/2021] [Accepted: 10/27/2021] [Indexed: 12/15/2022] Open
Abstract
Inflammatory bowel diseases (IBD) comprise a distinct set of clinical symptoms resulting from chronic inflammation within the gastrointestinal (GI) tract. Despite the significant progress in understanding the etiology and development of treatment strategies, IBD remain incurable for thousands of patients. Metabolic deregulation is indicative of IBD, including substantial shifts in lipid metabolism. Recent data showed that changes in some phospholipids are very common in IBD patients. For instance, phosphatidylcholine (PC)/phosphatidylethanolamine (PE) and lysophosphatidylcholine (LPC)/PC ratios are associated with the severity of the inflammatory process. Composition of phospholipids also changes upon IBD towards an increase in arachidonic acid and a decrease in linoleic and a-linolenic acid levels. Moreover, an increase in certain phospholipid metabolites, such as lysophosphatidylcholine, sphingosine-1-phosphate and ceramide, can result in enhanced intestinal inflammation, malignancy, apoptosis or necroptosis. Because some phospholipids are associated with pathogenesis of IBD, they may provide a basis for new strategies to treat IBD. Current attempts are aimed at controlling phospholipid and fatty acid levels through the diet or via pharmacological manipulation of lipid metabolism.
Collapse
|
10
|
Liu K, Sun T, Luan Y, Chen Y, Song J, Ling L, Yuan P, Li R, Cui K, Ruan Y, Lan R, Wang T, Wang S, Liu J, Rao K. Berberine ameliorates erectile dysfunction in rats with streptozotocin-induced diabetes mellitus through the attenuation of apoptosis by inhibiting the SPHK1/S1P/S1PR2 and MAPK pathways. Andrology 2021; 10:404-418. [PMID: 34674380 DOI: 10.1111/andr.13119] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 09/28/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND The population with diabetes mellitus-induced erectile dysfunction is increasing rapidly, but current drugs are not effective in treating erectile dysfunction. Studies of the traditional Chinese medicine extract berberine on diabetes and its complications provide us with new ideas. OBJECTIVES To evaluate the therapeutic effect and potential mechanism of berberine on the erectile function of diabetic rats. MATERIALS AND METHODS Fifty male Sprague-Dawley rats were randomly grouped, and 42 rats were injected intraperitoneally with streptozotocin to establish a diabetes model. Erectile dysfunction rats were screened out through the apomorphine test and randomly divided into the diabetes mellitus and berberine groups, and these animals were administered berberine (200 mg/kg/day) and normal saline by gavage for 4 weeks. Primary corpus cavernous smooth muscle cells from healthy rats were cultured and treated with berberine. RESULTS Fasting blood glucose in the diabetes mellitus group was significantly increased, while berberine showed no significant effect on glucose. Erectile function was obviously impaired in the diabetes mellitus group, and berberine administration partially rescued this impairment. The expression of sphingosine kinase 1, S1PR2, and sphingosine-1-phosphate in the diabetes mellitus group was increased. Berberine partially inhibited the expression of sphingosine kinase 1 and S1PR2, but the decrease in sphingosine-1-phosphate was not significant. Moreover, mitogen-activated protein kinase pathway factor expression was upregulated and eNOS activity was decreased in the diabetes mellitus group. Berberine treatment could partially reverse these alterations. Severe fibrosis and apoptosis were detected in diabetic rats, accompanied by higher expression of TGFβ1, collagen I/IV, Bax/Bcl-2, and caspase 3 than in the other groups. However, supplementation with berberine inhibited the expression of these proteins and attenuated fibrosis and apoptosis. CONCLUSIONS Berberine ameliorated erectile dysfunction in rats with diabetes mellitus, possibly by improving endothelial function and inhibiting apoptosis and fibrosis by suppressing the sphingosine kinase 1/sphingosine-1-phosphate/S1PR2 and mitogen-activated protein kinase pathways.
Collapse
Affiliation(s)
- Kang Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Taotao Sun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Luan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yinwei Chen
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jingyu Song
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Le Ling
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Penghui Yuan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rui Li
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kai Cui
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yajun Ruan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ruzhu Lan
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tao Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jihong Liu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ke Rao
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| |
Collapse
|
11
|
Matsuzaki E, Minakami M, Matsumoto N, Anan H. Dental regenerative therapy targeting sphingosine-1-phosphate (S1P) signaling pathway in endodontics. JAPANESE DENTAL SCIENCE REVIEW 2020; 56:127-134. [PMID: 33088365 PMCID: PMC7567953 DOI: 10.1016/j.jdsr.2020.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 08/02/2020] [Accepted: 09/18/2020] [Indexed: 12/31/2022] Open
Abstract
The establishment of regenerative therapy in endodontics targeting the dentin-pulp complex, cementum, periodontal ligament tissue, and alveolar bone will provide valuable information to preserve teeth. It is well known that the application of stem cells such as induced pluripotent stem cells, embryonic stem cells, and somatic stem cells is effective in regenerative medicine. There are many somatic stem cells in teeth and periodontal tissues including dental pulp stem cells (DPSCs), stem cells from the apical papilla, and periodontal ligament stem cells. Particularly, several studies have reported the regeneration of clinical pulp tissue and alveolar bone by DPSCs transplantation. However, further scientific issues for practical implementation remain to be addressed. Sphingosine-1-phosphate (S1P) acts as a bioactive signaling molecule that has multiple biological functions including cellular differentiation, and has been shown to be responsible for bone resorption and formation. Here we discuss a strategy for bone regeneration and a possibility for regenerative endodontics targeting S1P signaling pathway as one of approaches for induction of regeneration by improving the regenerative capacity of endogenous cells. SCIENTIFIC FIELD OF DENTAL SCIENCE Endodontology.
Collapse
Affiliation(s)
- Etsuko Matsuzaki
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
- Oral Medicine Research Center, Fukuoka Dental College, Fukuoka, Japan
| | - Masahiko Minakami
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Noriyoshi Matsumoto
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| | - Hisashi Anan
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, 2-15-1 Tamura, Sawara-ku, Fukuoka 814-0193, Japan
| |
Collapse
|
12
|
Ahmed N, Laghari AH, AlBkhoor B, Tabassum S, Meo SA, Muhammad N, Linardi D, Al-Masri AA, Fumagalli G, Luciani GB, Faggian G, Rungatscher A. Fingolimod Plays Role in Attenuation of Myocardial Injury Related to Experimental Model of Cardiac Arrest and Extracorporeal Life Support Resuscitation. Int J Mol Sci 2019; 20:ijms20246237. [PMID: 31835656 PMCID: PMC6940876 DOI: 10.3390/ijms20246237] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 11/22/2019] [Accepted: 12/05/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Sudden cardiac arrest is a major global health concern, and survival of patients with ischemia–reperfusion injury is a leading cause of myocardial dysfunction. The mechanism of this phenomenon is not well understood because of the complex pathophysiological nature of the disease. Aim of the study was to investigate the cardioprotective role of fingolimod in an in vivo model of cardiac arrest and resuscitation. Methods: In this study, an in vivo rat model of cardiac arrest using extracorporeal membrane oxygenation resuscitation monitored by invasive hemodynamic measurement was developed. At the beginning of extracorporeal life support (ECLS), animals were randomly treated with fingolimod (Group A, n = 30) or saline (Group B, n = 30). Half of the animals in each group (Group A1 and B1, n = 15 each) were sacrificed after 1 h, and the remaining animals (Group A2 and B2) after 24 h of reperfusion. Blood and myocardial tissues were collected for analysis of cardiac features, inflammatory biomarkers, and cell signaling pathways. Results: Treatment with fingolimod resulted in activation of survival pathways resulting into reduced inflammation, myocardial oxidative stress and apoptosis of cardiomyocytes. This led to significant improvement in systolic and diastolic functions of the left ventricle and improved contractility index. Conclusions: Sphingosine1phosphate receptor activation with fingolimod improved cardiac function after cardiac arrest supported with ECLS. Present study findings strongly support a cardioprotective role of fingolimod through sphingosine-1-phosphate receptor activation during reperfusion after circulatory arrest.
Collapse
Affiliation(s)
- Naseer Ahmed
- Department of Biological and Biomedical Sciences, The Aga Khan University, Karachi 74800, Pakistan
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
- Correspondence:
| | - Abid H. Laghari
- Department of Medicine, section of Cardiology, Aga Khan University, Karachi 74800, Pakistan;
| | | | - Sobia Tabassum
- Department of Biological Sciences, International Islamic University, Islamabad 44000, Pakistan;
| | - Sultan Ayoub Meo
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (S.A.M.); (A.A.A.-M.)
| | - Nazeer Muhammad
- COMSATS University Islamabad, Wah Campus, Rawalpindi 47040, Pakistan;
| | - Daniele Linardi
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Abeer A. Al-Masri
- Department of Physiology, College of Medicine, King Saud University, Riyadh 11461, Saudi Arabia; (S.A.M.); (A.A.A.-M.)
| | - Guido Fumagalli
- Department of Diagnostics and Public Health, Section of Pharmacology, University of Verona Medical School, 37134 Verona, Italy;
| | - Giovanni Battista Luciani
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Giuseppe Faggian
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| | - Alessio Rungatscher
- Department of Surgery, Cardiac Surgery Division, University of Verona Medical School, 37129 Verona, Italy; (D.L.); (G.B.L.); (G.F.); (A.R.)
| |
Collapse
|
13
|
Blumenfeld Z. Fertility Preservation Using GnRH Agonists: Rationale, Possible Mechanisms, and Explanation of Controversy. CLINICAL MEDICINE INSIGHTS. REPRODUCTIVE HEALTH 2019; 13:1179558119870163. [PMID: 31488958 PMCID: PMC6710670 DOI: 10.1177/1179558119870163] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022]
Abstract
The only clinically accepted method of fertility preservation in young women facing gonadotoxic chemo- and/or radiotherapy for malignant or autoimmune diseases is cryopreservation of embryos or unfertilized ova, whereas cryopreservation of ovarian tissue for future reimplantation, or in vitro maturation of follicles, and the use of gonadotropin-releasing hormone agonists (GnRHa) are still considered investigational, by several authorities. Whereas previous publications have raised the fear of GnRHa's possible detrimental effects in patients with hormone receptor-positive breast cancers, recent randomized controlled trials (RCTs) have shown that it either improves or does not affect disease-free survival (DFS) in such patients. This review summarizes the pros and cons of GnRHa co-treatment for fertility preservation, suggesting 5 theoretical mechanisms for GnRHa action: (1) simulating the prepubertal hypogonadotropic milieu, (2) direct effect on GnRH receptors, (3) decreased ovarian perfusion, (4) upregulation of an ovarian-protecting molecule such as sphingosine-1-phosphate, and (5) protecting a possible germinative stem cell. We try to explain the reasons for the discrepancy between most publications that support the use of GnRHa for fertility preservation and the minority of publications that did not support its efficiency.
Collapse
Affiliation(s)
- Zeev Blumenfeld
- Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
14
|
Grassi S, Mauri L, Prioni S, Cabitta L, Sonnino S, Prinetti A, Giussani P. Sphingosine 1-Phosphate Receptors and Metabolic Enzymes as Druggable Targets for Brain Diseases. Front Pharmacol 2019; 10:807. [PMID: 31427962 PMCID: PMC6689979 DOI: 10.3389/fphar.2019.00807] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Accepted: 06/21/2019] [Indexed: 12/12/2022] Open
Abstract
The central nervous system is characterized by a high content of sphingolipids and by a high diversity in terms of different structures. Stage- and cell-specific sphingolipid metabolism and expression are crucial for brain development and maintenance toward adult age. On the other hand, deep dysregulation of sphingolipid metabolism, leading to altered sphingolipid pattern, is associated with the majority of neurological and neurodegenerative diseases, even those totally lacking a common etiological background. Thus, sphingolipid metabolism has always been regarded as a promising pharmacological target for the treatment of brain disorders. However, any therapeutic hypothesis applied to complex amphipathic sphingolipids, components of cellular membranes, has so far failed probably because of the high regional complexity and specificity of the different biological roles of these structures. Simpler sphingosine-based lipids, including ceramide and sphingosine 1-phosphate, are important regulators of brain homeostasis, and, thanks to the relative simplicity of their metabolic network, they seem a feasible druggable target for the treatment of brain diseases. The enzymes involved in the control of the levels of bioactive sphingoids, as well as the receptors engaged by these molecules, have increasingly allured pharmacologists and clinicians, and eventually fingolimod, a functional antagonist of sphingosine 1-phosphate receptors with immunomodulatory properties, was approved for the therapy of relapsing-remitting multiple sclerosis. Considering the importance of neuroinflammation in many other brain diseases, we would expect an extension of the use of such analogs for the treatment of other ailments in the future. Nevertheless, many aspects other than neuroinflammation are regulated by bioactive sphingoids in healthy brain and dysregulated in brain disease. In this review, we are addressing the multifaceted possibility to address the metabolism and biology of bioactive sphingosine 1-phosphate as novel targets for the development of therapeutic paradigms and the discovery of new drugs.
Collapse
Affiliation(s)
- Sara Grassi
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Laura Mauri
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Simona Prioni
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Livia Cabitta
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Sandro Sonnino
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Alessandro Prinetti
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Paola Giussani
- Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| |
Collapse
|
15
|
Wang G, Bieberich E. Sphingolipids in neurodegeneration (with focus on ceramide and S1P). Adv Biol Regul 2018; 70:51-64. [PMID: 30287225 PMCID: PMC6251739 DOI: 10.1016/j.jbior.2018.09.013] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 09/20/2018] [Accepted: 09/21/2018] [Indexed: 04/14/2023]
Abstract
For many decades, research on sphingolipids associated with neurodegenerative disease focused on alterations in glycosphingolipids, particularly glycosylceramides (cerebrosides), sulfatides, and gangliosides. This seemed quite natural since many of these glycolipids are constituents of myelin and accumulated in lipid storage diseases (sphingolipidoses) resulting from enzyme deficiencies in glycolipid metabolism. With the advent of recognizing ceramide and its derivative, sphingosine-1-phosphate (S1P), as key players in lipid cell signaling and regulation of cell death and survival, research focus shifted toward these two sphingolipids. Ceramide and S1P are invoked in a plethora of cell biological processes participating in neurodegeneration such as ER stress, autophagy, dysregulation of protein and lipid transport, exosome secretion and neurotoxic protein spreading, neuroinflammation, and mitochondrial dysfunction. Hence, it is timely to discuss various functions of ceramide and S1P in neurodegenerative disease and to define sphingolipid metabolism and cell signaling pathways as potential targets for therapy.
Collapse
Affiliation(s)
- Guanghu Wang
- Department of Physiology, University of Kentucky, Lexington, KY, USA
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
16
|
The Role of Gene Therapy in Premature Ovarian Insufficiency Management. Biomedicines 2018; 6:biomedicines6040102. [PMID: 30388808 PMCID: PMC6316312 DOI: 10.3390/biomedicines6040102] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2018] [Revised: 10/23/2018] [Accepted: 10/26/2018] [Indexed: 01/06/2023] Open
Abstract
Premature ovarian insufficiency (POI) is a highly prevalent disorder, characterized by the development of menopause before the age of 40. Most cases are idiopathic; however, in some women the cause of this condition (e.g.; anticancer treatment, genetic disorders, and enzymatic defects) could be identified. Although hormone-replacement therapy, the principal therapeutic approach for POI, helps alleviate the related symptoms, this does not effectively solve the issue of fertility. Assisted reproductive techniques also lack efficacy in these women. Thus, an effective approach to manage patients with POI is highly warranted. Several mechanisms associated with POI have been identified, including the lack of function of the follicle-stimulating hormone (FSH) receptor, alterations in apoptosis control, mutations in Sal-like 4 genes, and thymulin or basonuclin-1 deficiency. The above mentioned may be good targets for gene therapy in order to correct defects leading to POI. The goal of this review is to summarize current experiences on POI studies that employed gene therapy, and to discuss possible future directions in this field.
Collapse
|
17
|
Elevated intrathymic sphingosine-1-phosphate promotes thymus involution during sepsis. Mol Immunol 2017; 90:255-263. [PMID: 28846923 DOI: 10.1016/j.molimm.2017.08.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/03/2017] [Accepted: 08/14/2017] [Indexed: 12/24/2022]
Abstract
Sepsis mouse models revealed thymus atrophy, characterised by decreased thymus weight and loss of thymocytes due to apoptosis. Mice suffered from lymphopenia, a lack of T cells in the periphery, which attenuates their ability to fight against recurring and secondary infections during sepsis progression. Key players in thymus atrophy are IL-6, which is directly involved in thymus involution, and the sphingosine-1-phosphate - sphingosine-1-phosphate receptor 1 signaling, influencing thymocytes emigration. In healthy individuals a sphingosine-1-phosphate (S1P) gradient from lymphoid organs to the circulatory system serves as signal for mature T cell egress. In the present study we investigated, whether inhibition of S1P generation improves thymus involution. In sepsis, induced by cecal ligation and puncture (CLP), S1P in the thymus increased, while it decreased in serum, thus disrupting the naturally occurring S1P gradient. As a potential source of S1P we identified increased numbers of apoptotic cells in the thymic cortex of septic mice. Pharmacological inhibition of the S1P generating sphingosine kinases, by 4- [[4-(4-Chlorophenyl)-2-thiazolyl]amino]phenol (SK I-II), administered directly following CLP, prevented thymus atrophy. This was reflected by lymphocytosis, diminished apoptosis, decreased IL-6 expression, and an unaltered thymus weight. In addition SK I-II-treatment preserved the S1P balance and prevented S1P-dependent internalization of the sphingosine-1-phosphate receptor 1. Our data suggest that inhibition of sphingosine kinase and thus, S1P generation during sepsis restores thymic T cell egress, which might improve septic outcome.
Collapse
|
18
|
Li S, Chen J, Fang X, Xia X. Sphingosine-1-phosphate activates the AKT pathway to inhibit chemotherapy induced human granulosa cell apoptosis. Gynecol Endocrinol 2017; 33:476-479. [PMID: 28277139 DOI: 10.1080/09513590.2017.1290072] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
UNLABELLED To investigate whether sphingosine-1-phosphate (S1P), an apoptosis-inhibitor would be able to inhibit chemotherapy induced human granulosa cell apoptosis. Cultures of primary granulosa cells were isolated from women undergoing in vitro fertilization (IVF). MTT assay was used to measure the optimum concentration of CTX and S1P acts on human granulosa cells. Granulosa cells were added with pertussis toxin (PTX), the PI3K inhibitor LY294002. Western blot analysis was used to analyze the signaling pathway of proteins and cell apoptosis. We found that S1P (10 mm) statistically significantly decreased granulosa cell apoptosis after cyclophosphamide (CTX) treatment. The decreased cell apoptosis induced by S1P was abolished after treatment with LY294002, PI3K inhibitor. CONCLUSIONS Treatment with S1P can inhibit the CTX-induced granulosa cell apoptosis. The S1P protective effect is mediated by activating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Shuyi Li
- a Department of Reproductive Medicine , Xiangya Hospital, Central South University , Changsha , P.R. China and
| | - Jianling Chen
- b Department of Obstetrics and Gynaecology , The Second Xiangya Hospital, Central South University , Changsha , P.R. China
| | - Xiaoling Fang
- b Department of Obstetrics and Gynaecology , The Second Xiangya Hospital, Central South University , Changsha , P.R. China
| | - Xiaomeng Xia
- b Department of Obstetrics and Gynaecology , The Second Xiangya Hospital, Central South University , Changsha , P.R. China
| |
Collapse
|
19
|
Ahmed N, Linardi D, Decimo I, Mehboob R, Gebrie MA, Innamorati G, Luciani GB, Faggian G, Rungatscher A. Characterization and Expression of Sphingosine 1-Phosphate Receptors in Human and Rat Heart. Front Pharmacol 2017; 8:312. [PMID: 28596734 PMCID: PMC5442178 DOI: 10.3389/fphar.2017.00312] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/11/2017] [Indexed: 12/12/2022] Open
Abstract
Aim: Sphingosine 1-phosphate (S1P), sphingolipid derivatives are known anti-inflammatory, anti-apoptotic, and anti-oxidant agent. S1P have been demonstrated to have a role in the cardiovascular system. The purpose of this study was to understand the precise expression and distribution of S1P receptors (S1PRs) in human and rat cardiovascular tissues to know the significance and possible implementation of our experimental studies in rat models. Methods and Results: In this study, we investigated the localization of S1PRs in human heart samples from cardiac surgery department, University of Verona Hospital and rat samples. Immunohistochemical investigation of paraffin-embedded sections illustrated diffused staining of the myocardial samples from human and rat. The signals of the human heart were similar to those of the rat heart in all chambers of the heart. The immunohistochemical expression levels correlated well with the results of RT-PCR-based analysis and western blotting. We confirmed by all techniques that S1PR1 expressed strongly as compared to S1PR3, and are uniformly distributed in all chambers of the heart with no significant difference in human and rat myocardial tissue. S1PR2 expression was significantly weak while S1PR4 and S1PR5 were not detectable in RT-PCR results in both human and rat heart. Conclusion: These results indicate that experimental studies using S1PR agonists on rat models are more likely to have a potential for translation into clinical studies, and second important information revealed by this study is, S1P receptor agonist can be used for cardioprotection in global ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Naseer Ahmed
- Cardiac Surgery Division, University of Verona Medical SchoolVerona, Italy.,Translational Surgery Lab, University of Verona Medical SchoolVerona, Italy.,Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Daniele Linardi
- Cardiac Surgery Division, University of Verona Medical SchoolVerona, Italy
| | - Ilaria Decimo
- Section of Pharmacology, Department of Diagnostics and Public Health, University of VeronaVerona, Italy
| | - Riffat Mehboob
- Department of Biomedical Sciences, King Edward Medical UniversityLahore, Pakistan
| | - Mebratu A Gebrie
- Cardiac Surgery Division, University of Verona Medical SchoolVerona, Italy
| | - Giulio Innamorati
- Translational Surgery Lab, University of Verona Medical SchoolVerona, Italy
| | - Giovanni B Luciani
- Cardiac Surgery Division, University of Verona Medical SchoolVerona, Italy
| | - Giuseppe Faggian
- Cardiac Surgery Division, University of Verona Medical SchoolVerona, Italy
| | | |
Collapse
|
20
|
Yanagawa D, Ishikawa T, Imai H. Synthesis and degradation of long-chain base phosphates affect fumonisin B 1-induced cell death in Arabidopsis thaliana. JOURNAL OF PLANT RESEARCH 2017; 130:571-585. [PMID: 28303405 DOI: 10.1007/s10265-017-0923-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 12/07/2016] [Indexed: 05/12/2023]
Abstract
Fumonisin B1 (FB1), an inducer of cell death, disrupts sphingolipid metabolism; large accumulations of de novo synthesized free long-chain bases (LCBs) are observed. However, it remains unclear whether tolerance to FB1 toxicity in plants is connected with preventing the accumulation of free LCBs through their phosphorylation. Here a workflow for the extraction, detection and quantification of LCB phosphates (LCBPs) in Arabidopsis thaliana was developed. We studied the effect of expression of genes for three enzymes involved in the synthesis and degradation of LCBPs, LCB kinase (LCBK1), LCBP phosphatase (SPP1) and lyase (DPL1) on FB1-induced cell death. As expected, large accumulations of saturated free LCBs, dihydrosphingosine and phytosphingosine, were observed in the FB1-treated leaves. On the other hand, a high level of sphingenine phosphate was found in the FB1-treated leaves even though free sphingenine was found in low amounts in these leaves. In comparison of WT and spp1 plants, the LCBP/LCB ratio is likely to be correlated with the degree of FB1-induced cell death determined by trypan blue staining. The FB1-treated leaves in dpl1 plants showed severe cell death and the elevation of free LCBs and LCBPs. LCBK1-OX and -KD plants showed resistance and sensitivity to FB1, respectively, whereas free LCB and LCBP levels in FB1-treated LCBK1-OX and -KD plants were moderately different to those in FB1-treated WT plants. Overall, the findings described here suggest that LCBP/LCB homeostasis is an important topic that participates in the tolerance of plant cells to FB1.
Collapse
Affiliation(s)
- Daiki Yanagawa
- Department of Biology, Graduate School of Natural Science, Konan University, Kobe, 658-8501, Japan
- The Institute for Integrative Neurobiology, Konan University, Kobe, 658-8501, Japan
| | - Toshiki Ishikawa
- Graduate School of Science and Engineering, Saitama University, Saitama, 338-8570, Japan
| | - Hiroyuki Imai
- Department of Biology, Graduate School of Natural Science, Konan University, Kobe, 658-8501, Japan.
- The Institute for Integrative Neurobiology, Konan University, Kobe, 658-8501, Japan.
| |
Collapse
|
21
|
STAT1 mediates transmembrane TNF-alpha-induced formation of death-inducing signaling complex and apoptotic signaling via TNFR1. Cell Death Differ 2017; 24:660-671. [PMID: 28186502 DOI: 10.1038/cdd.2016.162] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 11/22/2016] [Accepted: 12/12/2016] [Indexed: 01/14/2023] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) exists in two forms: secretory TNF-α (sTNF-α) and transmembrane TNF-α (tmTNF-α). Although both forms of TNF-α induce tumor cell apoptosis, tmTNF-α is able to kill tumor cells that are resistant to sTNF-α-mediated cytotoxicity, indicating their differences in signal transduction. Here, we demonstrate that internalization of TNFR1 is crucial for sTNF-α- but not for tmTNF-α-induced apoptosis. sTNF-α induces binding of tumor necrosis factor receptor type 1-associated death domain protein (TRADD) to the death domain (DD) of TNFR1 and subsequent activation of nuclear factor kappa B (NF-κB), and the formation of death-inducing signaling complexes (DISCs) in the cytoplasm after internalization. In contrast, tmTNF-α induces DISC formation on the membrane in a DD-independent manner. It leads to the binding of signal transducer and activator of transcription 1 (STAT1) to a region spanning amino acids 319-337 of TNFR1 and induces phosphorylation of serine at 727 of STAT1. The phosphorylation of STAT1 promotes its binding to TRADD, and thus recruits Fas-associated protein with DD (FADD) and caspase 8 to form DISC complexes. This STAT1-dependent signaling results in apoptosis but not NF-κB activation. STAT1-deficiency in U3A cells counteracts tmTNF-α-induced DISC formation and apoptosis. Conversely, reconstitution of STAT1 expression restores tmTNF-α-induced apoptotic signaling in the cell line. Consistently, tmTNF-α suppresses the growth of STAT1-containing HT1080 tumors, but not of STAT1-deficient U3A tumors in vivo. Our data reveal an unappreciated molecular mechanism of tmTNF-α-induced apoptosis and may provide a new clue for cancer therapy.
Collapse
|
22
|
Rhein C, Reichel M, Kramer M, Rotter A, Lenz B, Mühle C, Gulbins E, Kornhuber J. Alternative splicing of SMPD1 coding for acid sphingomyelinase in major depression. J Affect Disord 2017; 209:10-15. [PMID: 27866044 DOI: 10.1016/j.jad.2016.09.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 09/09/2016] [Accepted: 09/22/2016] [Indexed: 01/13/2023]
Abstract
BACKGROUND Major depressive disorder (MDD) is a psychiatric disorder characterized by key symptoms that include depressed mood and a loss of interest and pleasure. A recently developed pathogenic model of MDD involves disturbed neurogenesis in the hippocampus, where the acid sphingomyelinase (ASM)/ceramide system plays an important role and is proposed as a molecular target for antidepressant action. Because alternative splicing of SMPD1 mRNA, coding for ASM, is relevant for the regulation of ASM enzymatic activity, we investigated the frequency of alternatively spliced ASM isoforms in peripheral blood cells of MDD patients versus healthy controls. METHODS Because the full-length transcript variant 1 of SMPD1 (termed ASM-1) is the only known form within the splicing pattern that encodes an enzymatically fully active ASM, we determined a fraction of splice isoforms deviating from ASM-1 using PCR amplification and capillary electrophoresis with laser-induced fluorescence analysis. RESULTS ASM alternative splicing events occurred significantly less frequently in MDD patients compared to healthy subjects. After 5 days of antidepressant treatment, the frequency of alternatively spliced ASM isoforms decreased in those patients who were treated with a functional inhibitor of ASM activity (FIASMA) but remained constant in MDD patients treated with other antidepressant drugs. This effect was more pronounced when healthy male volunteers were treated with the FIASMAs fluoxetine or paroxetine, in contrast to a placebo group. LIMITATIONS Patients were treated with different antidepressant drugs, depending on individual parameters and disease courses. CONCLUSIONS This study shows that the ASM alternative splicing pattern could be a biological target with diagnostic relevance and could serve as a novel biomarker for MDD.
Collapse
Affiliation(s)
- Cosima Rhein
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany; Department of Medicine and Stony Brook Cancer Center, Stony Brook University, Stony Brook, New York, USA.
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Marcel Kramer
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany; Genome Analysis, Leibniz Institute on Aging - Fritz Lipmann Institute, Jena, Germany
| | - Andrea Rotter
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Bernd Lenz
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg (FAU), Schwabachanlage 6, D-91054 Erlangen, Germany
| |
Collapse
|
23
|
Xu Y, Xiao YJ, Baudhuin LM, Schwartz BM. The Role and Clinical Applications of Bioactive Lysolipids in Ovarian Cancer. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/107155760100800101] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Affiliation(s)
- Yan Xu
- Department of Cancer Biology Lerner Research Institute and the Department of Gynecology and Obstetrics Cleveland Clinic Foundation; Department of Chemistry, Cleveland State University, Cleveland, Ohio; Department of Cancer Biology, Cleveland Clinic Foundation, 9500 Euclid Ave., Cleveland, OH 44195
| | | | | | - Benjamin M. Schwartz
- Department of Cancer Biology Lerner Research Institute and the Department of Gynecology and Obstetrics Cleveland Clinic Foundation; Department of Chemistry, Cleveland State University, Cleveland, Ohio
| |
Collapse
|
24
|
Investigation of the Cell Surface Proteome of Human Periodontal Ligament Stem Cells. Stem Cells Int 2016; 2016:1947157. [PMID: 27579043 PMCID: PMC4989088 DOI: 10.1155/2016/1947157] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 07/03/2016] [Indexed: 12/14/2022] Open
Abstract
The present study examined the cell surface proteome of human periodontal ligament stem cells (PDLSC) compared to human fibroblasts. Cell surface proteins were prelabelled with CyDye before processing to extract the membrane lysates, which were separated using 2D electrophoresis. Selected differentially expressed protein “spots” were identified using Mass spectrometry. Four proteins were selected for validation: CD73, CD90, Annexin A2, and sphingosine kinase 1 previously associated with mesenchymal stem cells. Flow cytometric analysis found that CD73 and CD90 were highly expressed by human PDLSC and gingival fibroblasts but not by keratinocytes, indicating that these antigens could be used as potential markers for distinguishing between mesenchymal cells and epithelial cell populations. Annexin A2 was also found to be expressed at low copy number on the cell surface of human PDLSC and gingival fibroblasts, while human keratinocytes lacked any cell surface expression of Annexin A2. In contrast, sphingosine kinase 1 expression was detected in all the cell types examined using immunocytochemical analysis. These proteomic studies form the foundation to further define the cell surface protein expression profile of PDLSC in order to better characterise this cell population and help develop novel strategies for the purification of this stem cell population.
Collapse
|
25
|
Houck JD, Dawson TK, Kennedy AJ, Kharel Y, Naimon ND, Field SD, Lynch KR, Macdonald TL. Structural Requirements and Docking Analysis of Amidine-Based Sphingosine Kinase 1 Inhibitors Containing Oxadiazoles. ACS Med Chem Lett 2016; 7:487-92. [PMID: 27190598 DOI: 10.1021/acsmedchemlett.6b00002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/01/2016] [Indexed: 12/18/2022] Open
Abstract
Sphingosine 1-phosphate (S1P) is a potent growth-signaling lipid that has been implicated in cancer progression, inflammation, sickle cell disease, and fibrosis. Two sphingosine kinases (SphK1 and 2) are the source of S1P; thus, inhibitors of the SphKs have potential as targeted cancer therapies and will help to clarify the roles of S1P and the SphKs in other hyperproliferative diseases. Recently, we reported a series of amidine-based inhibitors with high selectivity for SphK1 and potency in the nanomolar range. However, these inhibitors display a short half-life. With the goal of increasing metabolic stability and maintaining efficacy, we designed an analogous series of molecules containing oxadiazole moieties. Generation of a library of molecules resulted in the identification of the most selective inhibitor of SphK1 reported to date (705-fold selectivity over SphK2), and we found that potency and selectivity vary significantly depending on the particular oxadiazole isomer employed. The best inhibitors were subjected to in silico molecular dynamics docking analysis, which revealed key insights into the binding of amidine-based inhibitors by SphK1. Herein, the design, synthesis, biological evaluation, and docking analysis of these molecules are described.
Collapse
Affiliation(s)
- Joseph D. Houck
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Thomas K. Dawson
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Andrew J. Kennedy
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Yugesh Kharel
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22904, United States
| | - Niels D. Naimon
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Saundra D. Field
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
| | - Kevin R. Lynch
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22904, United States
| | - Timothy L. Macdonald
- Department of Chemistry, University of Virginia, McCormick Road, Charlottesville, Virginia 22904, United States
- Department of Pharmacology, University of Virginia, 1340 Jefferson Park Avenue, Charlottesville, Virginia 22904, United States
| |
Collapse
|
26
|
Melland-Smith M, Ermini L, Chauvin S, Craig-Barnes H, Tagliaferro A, Todros T, Post M, Caniggia I. Disruption of sphingolipid metabolism augments ceramide-induced autophagy in preeclampsia. Autophagy 2016; 11:653-69. [PMID: 25853898 DOI: 10.1080/15548627.2015.1034414] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Bioactive sphingolipids including ceramides are involved in a variety of pathophysiological processes by regulating cell death and survival. The objective of the current study was to examine ceramide metabolism in preeclampsia, a serious disorder of pregnancy characterized by oxidative stress, and increased trophoblast cell death and autophagy. Maternal circulating and placental ceramide levels quantified by tandem mass spectrometry were elevated in pregnancies complicated by preeclampsia. Placental ceramides were elevated due to greater de novo synthesis via high serine palmitoyltransferase activity and reduced lysosomal breakdown via diminished ASAH1 expression caused by TGFB3-induced E2F4 transcriptional repression. SMPD1 activity was reduced; hence, sphingomyelin degradation by SMPD1 did not contribute to elevated ceramide levels in preeclampsia. Oxidative stress triggered similar changes in ceramide levels and acid hydrolase expression in villous explants and trophoblast cells. MALDI-imaging mass spectrometry localized the ceramide increases to the trophophoblast layers and syncytial knots of placentae from pregnancies complicated by preeclampsia. ASAH1 inhibition or ceramide treatment induced autophagy in human trophoblast cells via a shift of the BOK-MCL1 rheostat toward prodeath BOK. Pharmacological inhibition of ASAH1 activity in pregnant mice resulted in increased placental ceramide content, abnormal placentation, reduced fetal growth, and increased autophagy via a similar shift in the BOK-MCL1 system. Our results reveal that oxidative stress-induced reduction of lysosomal hydrolase activities in combination with elevated de novo synthesis leads to ceramide overload, resulting in increased trophoblast cell autophagy, and typifies preeclampsia as a sphingolipid storage disorder.
Collapse
Key Words
- 2-OE, 2-oleoylethanolamine
- 3-KDS, 3-keto dihydrosphingosine
- 3-MA, 3-methyladenine
- ACTB, actin β
- ASAH1, N-acylsphingosine amidohydrolase (acid ceramidase) 1
- BECN1, Beclin 1, autophagy related
- BOK
- BOK, BCL2-related ovarian killer
- BafA1, bafilomycin A1
- CANX, calnexin
- CASP3 (caspase 3, apoptosis-related cysteine peptidase)
- CERs, ceramides
- CT, cytotrophoblast cells
- D-NMAPPD, N-[(1R,2R)-2-hydroxyl-1-(hydroxyL-methyl)-2-(4-nitrophenyl) ethyl]-tetradecanamide
- DHCer, dihydro-ceramide
- E2F4, E2F transcription factor 4, p107/p130-binding
- HIF1A, hypoxia inducible factor 1, α, subunit (basic helix-loop-helix transcription factor)
- LAMP1, lysosomal-associated membrane protein 1
- LC-MS/MS, liquid chromatography-tandem mass spectrometry
- LC3B-II, cleaved and lipidated form of microtubule-associated protein 1 light chain 3 β (MAP1LC3B/LC3B)
- MALDI-MS, matrix-assisted laser desorption/ionization-mass spectrometry
- MCL1
- MCL1, myeloid cell leukemia 1
- PE, preeclampsia
- PTC, preterm control
- S1P, sphingosine-1-phosphate
- SM, sphingomyelin
- SMPD1, sphingomyelin phosphodiesterase 1, acid lysosomal (acid sphingomyelinase)
- SNP, sodium nitroprusside (III)
- SPH, sphingosine
- SPT, serine palmitoyltransferase
- SQSTM1/p62, sequestosome 1
- ST, syncytium/syncytiotrophoblast cells
- Sa, sphinganine
- TC, term control
- TGFB, transforming growth factor β
- autophagy
- oxidative stress
- placenta
- preeclampsia
- siRNA, small-interfering ribonucleic acid
- sphingolipid metabolism
Collapse
Affiliation(s)
- Megan Melland-Smith
- a The Lunenfeld-Tanenbaum Research Institute; Mount Sinai Hospital ; Toronto , ON Canada
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Villasmil ML, Francisco J, Gallo-Ebert C, Donigan M, Liu HY, Brower M, Nickels JT. Ceramide signals for initiation of yeast mating-specific cell cycle arrest. Cell Cycle 2016; 15:441-54. [PMID: 26726837 DOI: 10.1080/15384101.2015.1127475] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Sphingolipids are major constituents of membranes. A number of S. cerevisiae sphingolipid intermediates such as long chains sphingoid bases (LCBs) and ceramides act as signaling molecules regulating cell cycle progression, adaptability to heat stress, and survival in response to starvation. Here we show that S. cerevisiae haploid cells must synthesize ceramide in order to induce mating specific cell cycle arrest. Cells devoid of sphingolipid biosynthesis or defective in ceramide synthesis are sterile and harbor defects in pheromone-induced MAP kinase-dependent transcription. Analyses of G1/S cyclin levels indicate that mutant cells cannot reduce Cln1/2 levels in response to pheromone. FACS analysis indicates a lack of ability to arrest. The addition of LCBs to sphingolipid deficient cells restores MAP kinase-dependent transcription, reduces cyclin levels, and allows for mating, as does the addition of a cell permeable ceramide to cells blocked at ceramide synthesis. Pharmacological studies using the inositolphosphorylceramide synthase inhibitor aureobasidin A indicate that the ability to synthesize and accumulate ceramide alone is sufficient for cell cycle arrest and mating. Studies indicate that ceramide also has a role in PI(4,5)P2 polarization during mating, an event necessary for initiating cell cycle arrest and mating itself. Moreover, our studies suggest a third role for ceramide in localizing the mating-specific Ste5 scaffold to the plasma membrane. Thus, ceramide plays a role 1) in pheromone-induced cell cycle arrest, 2) in activation of MAP kinase-dependent transcription, and 3) in PtdIns(4,5)P2 polarization. All three events are required for differentiation during yeast mating.
Collapse
Affiliation(s)
- Michelle L Villasmil
- a The Institute of Metabolic Disorders, Genesis Biotechnology Group , Hamilton , NJ , USA.,b Cato Research Ltd. , Durham , NC , USA
| | - Jamie Francisco
- a The Institute of Metabolic Disorders, Genesis Biotechnology Group , Hamilton , NJ , USA
| | - Christina Gallo-Ebert
- a The Institute of Metabolic Disorders, Genesis Biotechnology Group , Hamilton , NJ , USA
| | - Melissa Donigan
- a The Institute of Metabolic Disorders, Genesis Biotechnology Group , Hamilton , NJ , USA
| | - Hsing-Yin Liu
- a The Institute of Metabolic Disorders, Genesis Biotechnology Group , Hamilton , NJ , USA
| | - Melody Brower
- a The Institute of Metabolic Disorders, Genesis Biotechnology Group , Hamilton , NJ , USA.,c Synthes, Inc , Paoli , PA , USA
| | - Joseph T Nickels
- a The Institute of Metabolic Disorders, Genesis Biotechnology Group , Hamilton , NJ , USA
| |
Collapse
|
28
|
Zhou YY, Ji XF, Fu JP, Zhu XJ, Li RH, Mu CK, Wang CL, Song WW. Gene Transcriptional and Metabolic Profile Changes in Mimetic Aging Mice Induced by D-Galactose. PLoS One 2015; 10:e0132088. [PMID: 26176541 PMCID: PMC4503422 DOI: 10.1371/journal.pone.0132088] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 06/10/2015] [Indexed: 01/09/2023] Open
Abstract
D-galactose injection has been shown to induce many changes in mice that represent accelerated aging. This mouse model has been widely used for pharmacological studies of anti-aging agents. The underlying mechanism of D-galactose induced aging remains unclear, however, it appears to relate to glucose and 1ipid metabolic disorders. Currently, there has yet to be a study that focuses on investigating gene expression changes in D-galactose aging mice. In this study, integrated analysis of gas chromatography/mass spectrometry-based metabonomics and gene expression profiles was used to investigate the changes in transcriptional and metabolic profiles in mimetic aging mice injected with D-galactose. Our findings demonstrated that 48 mRNAs were differentially expressed between control and D-galactose mice, and 51 potential biomarkers were identified at the metabolic level. The effects of D-galactose on aging could be attributed to glucose and 1ipid metabolic disorders, oxidative damage, accumulation of advanced glycation end products (AGEs), reduction in abnormal substance elimination, cell apoptosis, and insulin resistance.
Collapse
Affiliation(s)
- Yue-Yue Zhou
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, Ningbo University, Ningbo, China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, China
| | - Xiong-Fei Ji
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, Ningbo University, Ningbo, China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, China
| | - Jian-Ping Fu
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, Ningbo University, Ningbo, China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, China
| | - Xiao-Juan Zhu
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, Ningbo University, Ningbo, China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, China
| | - Rong-Hua Li
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, Ningbo University, Ningbo, China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, China
| | - Chang-Kao Mu
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, Ningbo University, Ningbo, China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, China
| | - Chun-Lin Wang
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, Ningbo University, Ningbo, China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, China
- * E-mail: (WWS); (CLW)
| | - Wei-Wei Song
- Key Laboratory of the Ministry of Education for Applied Marine Biotechnology, Ningbo University, Ningbo, China
- Collaborative Innovation Center for Zhejiang Marine High-efficiency and Healthy Aquaculture, Ningbo University, Ningbo, China
- * E-mail: (WWS); (CLW)
| |
Collapse
|
29
|
Resop RS, Uittenbogaart CH. Human T-Cell Development and Thymic Egress: An Infectious Disease Perspective. FORUM ON IMMUNOPATHOLOGICAL DISEASES AND THERAPEUTICS 2015; 6:33-49. [PMID: 28670486 PMCID: PMC5489135 DOI: 10.1615/forumimmundisther.2015014226] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Emigration of mature naïve CD4 SP T cells from the human thymus to the periphery is not fully understood, although elucidation of the mechanisms that govern egress of T cells is crucial to understanding both basic immunology and the immune response in diseases such as HIV infection. Recent work has brought to light the requirement for sphingosine-1-phosphate (S1P) and its receptors in a variety of fields including mature naïve T-cell egress from the thymus of mice. We are examining the expression and function of this novel requisite T-cell egress receptor within the human thymus, characterizing changes observed in the expression and function of this receptor in infectious diseases. To perform this work, we use a variety of humanized murine models reviewed in this article. Future work in the field of T-cell egress, especially as it pertains to S1P receptors, should advance the fields of basic T-cell immunology and immunopathology and open new avenues for exploration into novel therapeutics.
Collapse
Affiliation(s)
- Rachel S. Resop
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- Department of Pediatrics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
| | - Christel H. Uittenbogaart
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- Department of Pediatrics, David Geffen Medical School at UCLA, Los Angeles, CA 90095
- University of California at Los Angeles AIDS Institute, Los Angeles, CA 90095
- Jonsson Comprehensive Cancer Center, David Geffen Medical School at UCLA, Los Angeles, CA 90095
| |
Collapse
|
30
|
Coumestrol suppresses hypoxia inducible factor 1α by inhibiting ROS mediated sphingosine kinase 1 in hypoxic PC-3 prostate cancer cells. Bioorg Med Chem Lett 2014; 24:2560-4. [DOI: 10.1016/j.bmcl.2014.03.084] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Revised: 03/13/2014] [Accepted: 03/26/2014] [Indexed: 11/22/2022]
|
31
|
Tanasescu R, Constantinescu CS. Pharmacokinetic evaluation of fingolimod for the treatment of multiple sclerosis. Expert Opin Drug Metab Toxicol 2014; 10:621-30. [PMID: 24579791 DOI: 10.1517/17425255.2014.894019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Fingolimod is a sphingosine 1-phosphate receptor modulator with a novel mechanism of action and the first oral drug approved for the treatment of relapsing forms of multiple sclerosis (MS). Fingolimod reduces relapses more effectively than intramuscular interferon β1a and delays disability progression. Associated safety risks are bradyarrhythmia and atrioventricular block following the initial dose, requiring monitoring. AREAS COVERED This article examines the characteristics of fingolimod, its pharmacokinetic properties and the efficacy and tolerability in MS. Information on the pharmacology and mechanisms of action is also provided. EXPERT OPINION Fingolimod is an effective therapy for relapsing forms of MS in a convenient oral dose. Fingolimod may target not only inflammation but potentially also neurodegeneration. Antagonizing astrocyte sphingosine signaling may help explain the reduction in cerebral atrophy observed in Phase III trials. Long-term data about the safety of fingolimod are needed.
Collapse
Affiliation(s)
- Radu Tanasescu
- University of Nottingham, Queen's Medical Centre, Academic Division of Clinical Neurology , C Floor, South Block, Nottingham, NG7 2UH , UK +44 115 8754597/98 ; +44 115 823 1443 ;
| | | |
Collapse
|
32
|
Ceccom J, Loukh N, Lauwers-Cances V, Touriol C, Nicaise Y, Gentil C, Uro-Coste E, Pitson S, Maurage CA, Duyckaerts C, Cuvillier O, Delisle MB. Reduced sphingosine kinase-1 and enhanced sphingosine 1-phosphate lyase expression demonstrate deregulated sphingosine 1-phosphate signaling in Alzheimer's disease. Acta Neuropathol Commun 2014; 2:12. [PMID: 24468113 PMCID: PMC3912487 DOI: 10.1186/2051-5960-2-12] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Accepted: 01/18/2014] [Indexed: 12/14/2022] Open
Abstract
Background The accumulation of beta amyloid (Aβ) peptides, a hallmark of Alzheimer’s disease (AD) is related to mechanisms leading to neurodegeneration. Among its pleiotropic cellular effects, Aβ accumulation has been associated with a deregulation of sphingolipid metabolism. Sphingosine 1-phosphate (S1P) derived from sphingosine is emerging as a critical lipid mediator regulating various biological activities including cell proliferation, survival, migration, inflammation, or angiogenesis. S1P tissue level is low and kept under control through equilibrium between its synthesis mostly governed by sphingosine kinase-1 (SphK1) and its degradation by sphingosine 1-phosphate lyase (SPL). We have previously reported that Aβ peptides were able to decrease the activity of SphK1 in cell culture models, an effect that could be blocked by the prosurvival IGF-1/IGF-1R signaling. Results Herein, we report for the first time the expression of both SphK1 and SPL by immunohistochemistry in frontal and entorhinal cortices from 56 human AD brains. Immunohistochemical analysis revealed a decreased expression of SphK1 and an increased expression of SPL both correlated to amyloid deposits in the entorhinal cortex. Otherwise, analysis of brain tissue extracts showed a decrease of SphK1 expression in AD brains whereas SPL expression was increased. The content of IGF-1R, an activator of SphK1, was found decreased in AD brains as well as S1P1, the major receptor for S1P. Conclusions Collectively, these results highlight the importance of S1P in AD suggesting the existence of a global deregulation of S1P signaling in this disease from its synthesis by SphK1 and degradation by SPL to its signaling by the S1P1 receptor.
Collapse
|
33
|
Truman JP, García-Barros M, Obeid LM, Hannun YA. Evolving concepts in cancer therapy through targeting sphingolipid metabolism. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:1174-88. [PMID: 24384461 DOI: 10.1016/j.bbalip.2013.12.013] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2013] [Revised: 12/18/2013] [Accepted: 12/21/2013] [Indexed: 12/29/2022]
Abstract
Traditional methods of cancer treatment are limited in their efficacy due to both inherent and acquired factors. Many different studies have shown that the generation of ceramide in response to cytotoxic therapy is generally an important step leading to cell death. Cancer cells employ different methods to both limit ceramide generation and to remove ceramide in order to become resistant to treatment. Furthermore, sphingosine kinase activity, which phosphorylates sphingosine the product of ceramide hydrolysis, has been linked to multidrug resistance, and can act as a strong survival factor. This review will examine several of the most frequently used cancer therapies and their effect on both ceramide generation and the mechanisms employed to remove it. The development and use of inhibitors of sphingosine kinase will be focused upon as an example of how targeting sphingolipid metabolism may provide an effective means to improve treatment response rates and reduce associated treatment toxicity. This article is part of a Special Issue entitled Tools to study lipid functions.
Collapse
Affiliation(s)
- Jean-Philip Truman
- Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794 Stony Brook, NY, USA.
| | - Mónica García-Barros
- Health Science Center, Stony Brook University, 100 Nicolls Road, T15, 023, 11794 Stony Brook, NY, USA.
| | - Lina M Obeid
- Northport Veterans Affairs Medical Center, Northport, NY 11768, USA; Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 178, 11794 Stony Brook, NY, USA.
| | - Yusuf A Hannun
- Department of Medicine and the Stony Brook Cancer Center, Health Science Center, Stony Brook University, 100 Nicolls Road, L4, 178, 11794 Stony Brook, NY, USA.
| |
Collapse
|
34
|
Gomez-Muñoz A, Gangoiti P, Arana L, Ouro A, Rivera IG, Ordoñez M, Trueba M. New insights on the role of ceramide 1-phosphate in inflammation. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1060-6. [DOI: 10.1016/j.bbalip.2013.02.001] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 01/29/2013] [Accepted: 02/05/2013] [Indexed: 01/08/2023]
|
35
|
Troppens DM, Dmitriev RI, Papkovsky DB, O'Gara F, Morrissey JP. Genome-wide investigation of cellular targets and mode of action of the antifungal bacterial metabolite 2,4-diacetylphloroglucinol in Saccharomyces cerevisiae. FEMS Yeast Res 2013; 13:322-34. [PMID: 23445507 DOI: 10.1111/1567-1364.12037] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 02/01/2013] [Accepted: 02/02/2013] [Indexed: 01/18/2023] Open
Abstract
Saccharomyces cerevisiae is a proven model to investigate the effects of small molecules and drugs on fungal and eukaryotic cells. In this study, the mode of action of an antifungal metabolite, 2,4-diacetylphloroglucinol (DAPG), was determined. Applying a combination of genetic and physiological approaches, it was established that this bacterial metabolite acts as a proton ionophore and dissipates the proton gradient across the mitochondrial membrane. The uncoupling of respiration and ATP synthesis ultimately leads to growth inhibition and is the primary toxic effect of DAPG. A genome-wide screen identified 154 DAPG-tolerant mutants and showed that there are many alterations in cellular metabolism that can confer at least some degree of tolerance to this uncoupler. One mutant, ydc1, was studied in some more detail as it displayed increased tolerance to both DAPG and the uncoupler carbonylcyanide m-chlorophenylhydrazone (CCCP) and appears to be unconnected to other tolerant mutant strains. Deleting YDC1 alters sphingolipid homoeostasis in the cell, and we suggest here that this may be linked to reduced drug sensitivity. Sphingolipids and their derivatives are important eukaryotic signal molecules, and the observation that altering homoeostasis may affect yeast response to metabolic uncoupling agents raises some intriguing questions for future studies.
Collapse
|
36
|
Mühle C, Reichel M, Gulbins E, Kornhuber J. Sphingolipids in psychiatric disorders and pain syndromes. Handb Exp Pharmacol 2013:431-456. [PMID: 23563670 DOI: 10.1007/978-3-7091-1511-4_22] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Despite the high prevalence and devastating impact of psychiatric disorders, little is known about their etiopathology. In this review, we provide an overview on the participation of sphingolipids and enzymes responsible for their metabolism in mechanisms underlying psychiatric disorders. We focus on the pathway from sphingomyelin to proapoptotic ceramide and the subsequent metabolism of ceramide to sphingosine, which is in turn phosphorylated to yield anti-apoptotic sphingosine-1-phosphate (S1P).The sphingomyelinase/ceramide system has been linked to effects of reactive oxygen species and proinflammatory cytokines in the central nervous system as well as to synaptic transmission. Compared to ubiquitously expressed acid sphingomyelinase, acid and neutral ceramidase and neutral sphingomyelinase are highly active in brain regions. Depressed patients show elevated plasma ceramide levels and increased activities of acid sphingomyelinase which is functionally inhibited by many anti-depressive drugs. Exposure to alcohol is associated with an activation of acid and neutral sphingomyelinase observed in cell culture, mouse models and in alcohol-dependent patients and with increased concentrations of ceramide in various organs.Levels of sphingomyelin and ceramide are altered in erythrocytes and post-mortem brain tissues of schizophrenic patients in addition to changes in expression patterns for serine palmitoyltransferase and acid ceramidase leading to impaired myelination. After induction of anxiety-like behavior in animal models, higher serum levels of S1P were reported to lead to neurodegeneration. Correspondingly, S1P infusion appeared to increase anxiety-like behavior. Significantly upregulated levels of the endogenous ceramide catabolite N,N-dimethylsphingosine were observed in rat models of allodynia. Conversely, rats injected intrathecally with N,N-dimethylsphingosine developed mechanical allodynia. Moreover, S1P has been implicated in spinal nociceptive processing.The increasing interest in lipidomics and improved analytical methods led to growing insight into the connection between psychiatric and neurological disorders and sphingolipid metabolism and may once provide new targets and strategies for therapeutic intervention.
Collapse
Affiliation(s)
- C Mühle
- Department of Psychiatry and Psychotherapy, Friedrich-Alexander-University of Erlangen, Erlangen, Germany
| | | | | | | |
Collapse
|
37
|
Post-translational regulation of sphingosine kinases. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:147-56. [DOI: 10.1016/j.bbalip.2012.07.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Revised: 07/04/2012] [Accepted: 07/06/2012] [Indexed: 12/22/2022]
|
38
|
Chan JP, Hu Z, Sieburth D. Recruitment of sphingosine kinase to presynaptic terminals by a conserved muscarinic signaling pathway promotes neurotransmitter release. Genes Dev 2012; 26:1070-85. [PMID: 22588719 DOI: 10.1101/gad.188003.112] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Sphingolipids are potent lipid second messengers that regulate cell differentiation, migration, survival, and secretion, and alterations in sphingolipid signaling have been implicated in a variety of diseases. However, how sphingolipid levels are regulated, particularly in the nervous system, remains poorly understood. Here, we show that the generation of sphingosine-1-phosphate by sphingosine kinase (SphK) promotes neurotransmitter release. Electrophysiological, imaging, and behavioral analyses of Caenorhabditis elegans mutants lacking sphingosine kinase sphk-1 indicate that neuronal development is normal, but there is a significant defect in neurotransmitter release from neuromuscular junctions. SPHK-1 localizes to discrete, nonvesicular regions within presynaptic terminals, and this localization is critical for synaptic function. Muscarinic agonists cause a rapid increase in presynaptic SPHK-1 abundance, whereas reduction of endogenous acetylcholine production results in a rapid decrease in presynaptic SPHK-1 abundance. Muscarinic regulation of presynaptic SPHK-1 abundance is mediated by a conserved presynaptic signaling pathway composed of the muscarinic acetylcholine receptor GAR-3, the heterotrimeric G protein Gαq, and its effector, Trio RhoGEF. SPHK-1 activity is required for the effects of muscarinic signaling on synaptic transmission. This study shows that SPHK-1 promotes neurotransmitter release in vivo and identifies a novel muscarinic pathway that regulates SphK abundance at presynaptic terminals.
Collapse
Affiliation(s)
- Jason P Chan
- Zilkha Neurogenetic Institute, University of Southern California, Los Angeles, California 90033, USA
| | | | | |
Collapse
|
39
|
Fournie P, Galiacy S, Ranty ML, Rico-Lattes I, Malecaze F, Quintyn JC. Sphingosine-1 phosphate prevents ethanol-induced corneal epithelial apoptosis. Indian J Ophthalmol 2012; 60:115-8. [PMID: 22446906 PMCID: PMC3339070 DOI: 10.4103/0301-4738.94052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Apoptosis is a programmed cell death in multicellular organisms, found in a wide variety of conditions, including inflammatory process, everywhere in the body, including the cornea and conjunctiva. Aim: To evaluate the effect of a new topical formulation of sphingosine-1 phosphate on preventing apoptosis of the corneal epithelium. Setting: Medical University. Materials and Methods: We tested several formulations suitable for topical application. Twenty-five rabbits were distributed among five groups. Group 1 comprised the controls. In Group 2, 20% ethanol was applied topically for 20 seconds; in Group 3, 50 μM topical sphingosine-1 phosphate was applied 2 hours prior to 20% ethanol application. In Group 4, 200 μM topical sphingosine-1 phosphate was applied 2 hours before the 20% ethanol application. In Group 5, only 200 μM topical sphingosine-1 phosphate was applied. Apoptosis was evaluated using the terminal deoxynucleotidyl transferase biotin-dUTP Nick End Labeling (TUNEL) assay. Pairwise comparisons were performed using t-tests with Scheffe's correction. Data were analyzed using STATA 9.0 statistical software. Results: A suspension of sphingosine-1 phosphate in the presence of Montanox 80 was stable and could be formulated without sonication. Epithelial apoptosis was detected only in Groups 2 and 3. Conclusion: Sphingosine-1 phosphate can prevent ethanol-induced apoptosis in the corneal epithelium of rabbits.
Collapse
Affiliation(s)
- Pierre Fournie
- EA4555 Génétique des troubles de la réfraction et des anomalies du dévelopement de l'oeil, CHU Purpan, Toulouse, 31000, France
| | | | | | | | | | | |
Collapse
|
40
|
Nakahara T, Iwase A, Nakamura T, Kondo M, Bayasula, Kobayashi H, Takikawa S, Manabe S, Goto M, Kotani T, Kikkawa F. Sphingosine-1-phosphate inhibits H2O2-induced granulosa cell apoptosis via the PI3K/Akt signaling pathway. Fertil Steril 2012; 98:1001-8.e1. [PMID: 22763095 DOI: 10.1016/j.fertnstert.2012.06.008] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2012] [Revised: 06/13/2012] [Accepted: 06/13/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE To investigate the protective effect of sphingosine-1-phosphate (S1P) against H(2)O(2)-induced apoptosis in human granulosa cell cultures with freshly harvested granulosa cells. DESIGN Experimental study. SETTING Academic medical center for reproductive medicine. PATIENT(S) Cultures of primary granulosa cells isolated from women undergoing in vitro fertilization (IVF). INTERVENTION(S) None. MAIN OUTCOME MEASURE(S) Cell apoptosis and Western blot analysis of signaling pathway proteins. RESULT(S) We found that S1P (1 and 10 mM) statistically significantly decreased granulosa cell apoptosis after H(2)O(2) treatment. The decreased cell apoptosis induced by S1P was abolished after treatment with VPC23019, an inhibitor of S1P1 and S1P3 receptors, W146, an inhibitor of S1P1 receptors, and CAY10444, an inhibitor of S1P3 receptors. A Western blot analysis revealed that the level of phospho-Akt increased and peaked at 10 minutes after 10 mM S1P exposure. CONCLUSION(S) Treatment with S1P can inhibit the apoptosis of granulosa cells in response to oxidative stress induced by H(2)O(2). The protective effect of S1P is mediated by activating the PI3K/Akt pathway, and the antiapoptotic effect of S1P is mainly mediated through the S1P1 and S1P3 receptor.
Collapse
Affiliation(s)
- Tatsuo Nakahara
- Department of Obstetrics and Gynecology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Majumdar I, Mastrandrea LD. Serum sphingolipids and inflammatory mediators in adolescents at risk for metabolic syndrome. Endocrine 2012; 41:442-9. [PMID: 22228496 DOI: 10.1007/s12020-011-9589-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 12/20/2011] [Indexed: 11/26/2022]
Abstract
The purpose of this study was to determine low-grade inflammation associated with obesity that is mediated partially by TNF-α, an adipocytokine which stimulates sphingomyelinase activity in adipocytes. Circulating ceramide (Cer) and sphingosine 1-phosphate (S1P) are elevated in genetically obese (ob/ob) mice. We aimed to determine whether serum sphingolipid concentrations correlate with measures of obesity, insulin resistance, and lipid profiles in overweight versus lean adolescents. This cross-sectional study recruited 30 healthy overweight (body mass index, BMI ≥ 85%) and 15 lean (BMI 10-84%) adolescents. Anthropometric measurements and fasting blood samples were collected at one clinic visit. Serum glucose, insulin, and fasting lipid profiles were measured. Serum adipocytokine concentrations were measured by ELISA or colorimetric assay and sphingolipids were measured by HPLC-mass spectrometry. Between group differences in serum sphingolipid concentrations were assessed. Correlations between sphingolipid concentrations and (i) body mass index, (ii) calculated homeostasis model assessment of insulin resistance (HOMA-IR), (iii) adipocytokines, and (iv) lipoproteins were determined. The results showed that significant differences in HOMA-IR (4.5 ± 3.2 vs. 1.2 ± 0.7), free fatty acids (0.8 ± 0.3 mmol/l vs. 0.4 ± 0.3 mmol/l), and adiponectin (6.4 ± 3.8 vs. 12.6 ± 9.9 μg/ml) were seen between groups (overweight vs. lean). There were significant correlations between Cer and TNF-α (r = 0.429), S1P and TNF-α (r = 0.288), Cer and adiponectin (r = 0.321), Cer:S1P and adiponectin (r = 0.324), Cer and HOMA-IR (r = 0.307), and Cer:S1P and LDL cholesterol (r = 0.453); these associations persisted after adjustment for BMI Z-score, sex, and Tanner stage. We concluded that elevated sphingolipid concentrations correlate with TNF-α, adiponectin, lipoprotein profiles, and HOMA-IR. Ceramide is associated with atherogenic lipid profiles and the development of insulin resistance in obese adolescents, similar to adults.
Collapse
Affiliation(s)
- I Majumdar
- Department of Pediatrics, School of Medicine and Biomedical Sciences, University at Buffalo, The State University of New York, Buffalo, USA
| | | |
Collapse
|
42
|
Lee JP, Cha HJ, Lee KS, Lee KK, Son JH, Kim KN, Lee DK, An S. Phytosphingosine-1-phosphate represses the hydrogen peroxide-induced activation of c-Jun N-terminal kinase in human dermal fibroblasts through the phosphatidylinositol 3-kinase/Akt pathway. Arch Dermatol Res 2012; 304:673-8. [PMID: 22566145 DOI: 10.1007/s00403-012-1241-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 04/16/2012] [Accepted: 04/18/2012] [Indexed: 10/28/2022]
Abstract
Dermal fibroblasts are differentiated mesenchymal cells that regulate the extracellular matrix through the production of dermis components. Dermal fibroblasts can be damaged by reactive oxygen species induced by ultraviolet rays and chemicals. In addition to its effects on the dermis, oxidative stress poses a major threat to organisms and is believed to play an essential role in many disease processes. In this study, we show that human dermal fibroblasts (HDFs) express sphingosine-1-phosphate (S1P) receptors S1P(1), S1P(2), and S1P(3). In addition, cell viability of HDFs is increased by phytosphingosine-1-phosphate (PhS1P) via regulation of the Jun N-terminal kinase (JNK)/Akt pathway. Interestingly, regulation of the JNK/Akt pathway by PhS1P attenuated H(2)O(2)-induced cell growth arrest. Together, our data indicate that PhS1P attenuates H(2)O(2)-induced growth arrest through regulation of the signal molecules Akt and JNK, and suggest that PhS1P may have value as an anti-aging material in cosmetics and medicine.
Collapse
Affiliation(s)
- Jeong Pyo Lee
- Coreana Cosmetics Co., Ltd., Seonggeo-eup, Seobuk-gu, Cheonan-si, Chungcheongnam-do, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Rhein C, Tripal P, Seebahn A, Konrad A, Kramer M, Nagel C, Kemper J, Bode J, Mühle C, Gulbins E, Reichel M, Becker CM, Kornhuber J. Functional implications of novel human acid sphingomyelinase splice variants. PLoS One 2012; 7:e35467. [PMID: 22558155 PMCID: PMC3338701 DOI: 10.1371/journal.pone.0035467] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 03/16/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Acid sphingomyelinase (ASM) hydrolyses sphingomyelin and generates the lipid messenger ceramide, which mediates a variety of stress-related cellular processes. The pathological effects of dysregulated ASM activity are evident in several human diseases and indicate an important functional role for ASM regulation. We investigated alternative splicing as a possible mechanism for regulating cellular ASM activity. METHODOLOGY/PRINCIPAL FINDINGS We identified three novel ASM splice variants in human cells, termed ASM-5, -6 and -7, which lack portions of the catalytic- and/or carboxy-terminal domains in comparison to full-length ASM-1. Differential expression patterns in primary blood cells indicated that ASM splicing might be subject to regulatory processes. The newly identified ASM splice variants were catalytically inactive in biochemical in vitro assays, but they decreased the relative cellular ceramide content in overexpression studies and exerted a dominant-negative effect on ASM activity in physiological cell models. CONCLUSIONS/SIGNIFICANCE These findings indicate that alternative splicing of ASM is of functional significance for the cellular stress response, possibly representing a mechanism for maintaining constant levels of cellular ASM enzyme activity.
Collapse
Affiliation(s)
- Cosima Rhein
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Philipp Tripal
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Angela Seebahn
- Institute of Biochemistry, Emil-Fischer-Centre, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Alice Konrad
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Marcel Kramer
- Leibniz Institute for Age Research – Fritz Lipmann Institute and Center for Sepsis Control and Care at Jena University Hospital, Jena, Germany
| | - Christine Nagel
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Jonas Kemper
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Jens Bode
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Christiane Mühle
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Erich Gulbins
- Department of Molecular Biology, University of Duisburg-Essen, Essen, Germany
| | - Martin Reichel
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Cord-Michael Becker
- Institute of Biochemistry, Emil-Fischer-Centre, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| | - Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
44
|
Brogden NK, Mehalick L, Fischer CL, Wertz PW, Brogden KA. The emerging role of peptides and lipids as antimicrobial epidermal barriers and modulators of local inflammation. Skin Pharmacol Physiol 2012; 25:167-81. [PMID: 22538862 DOI: 10.1159/000337927] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 03/01/2012] [Indexed: 12/17/2022]
Abstract
Skin is complex and comprised of distinct layers, each layer with unique architecture and immunologic functions. Cells within these layers produce differing amounts of antimicrobial peptides and lipids (sphingoid bases and sebaceous fatty acids) that limit colonization of commensal and opportunistic microorganisms. Furthermore, antimicrobial peptides and lipids have distinct, concentration-dependent ancillary innate and adaptive immune functions. At 0.1-2.0 μM, antimicrobial peptides induce cell migration and adaptive immune responses to coadministered antigens. At 2.0-6.0 μM, they induce cell proliferation and enhance wound healing. At 6.0-12.0 μM, they can regulate chemokine and cytokine production and at their highest concentrations of 15.0-30.0 μM, antimicrobial peptides can be cytotoxic. At 1-100 nM, lipids enhance cell migration induced by chemokines, suppress apoptosis, and optimize T cell cytotoxicity, and at 0.3-1.0 μM they inhibit cell migration and attenuate chemokine and pro-inflammatory cytokine responses. Recently, many antimicrobial peptides and lipids at 0.1-2.0 μM have been found to attenuate the production of chemokines and pro-inflammatory cytokines to microbial antigens. Together, both the antimicrobial and the anti-inflammatory activities of these peptides and lipids may serve to create a strong, overlapping immunologic barrier that not only controls the concentrations of cutaneous commensal flora but also the extent to which they induce a localized inflammatory response.
Collapse
Affiliation(s)
- N K Brogden
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA.
| | | | | | | | | |
Collapse
|
45
|
Berkey R, Bendigeri D, Xiao S. Sphingolipids and plant defense/disease: the "death" connection and beyond. FRONTIERS IN PLANT SCIENCE 2012; 3:68. [PMID: 22639658 PMCID: PMC3355615 DOI: 10.3389/fpls.2012.00068] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 03/22/2012] [Indexed: 05/19/2023]
Abstract
Sphingolipids comprise a major class of structural materials and lipid signaling molecules in all eukaryotic cells. Over the past two decades, there has been a phenomenal growth in the study of sphingolipids (i.e., sphingobiology) at an average rate of ∼1000 research articles per year. Sphingolipid studies in plants, though accounting for only a small fraction (∼6%) of the total number of publications, have also enjoyed proportionally rapid growth in the past decade. Concomitant with the growth of sphingobiology, there has also been tremendous progress in our understanding of the molecular mechanisms of plant innate immunity. In this review, we (i) cross examine and analyze the major findings that establish and strengthen the intimate connections between sphingolipid metabolism and plant programmed cell death (PCD) associated with plant defense or disease; (ii) highlight and compare key bioactive sphingolipids involved in the regulation of plant PCD and possibly defense; (iii) discuss the potential role of sphingolipids in polarized membrane/protein trafficking and formation of lipid rafts as subdomains of cell membranes in relation to plant defense; and (iv) where possible, attempt to identify potential parallels for immunity-related mechanisms involving sphingolipids across kingdoms.
Collapse
Affiliation(s)
- Robert Berkey
- Institute for Bioscience and Biotechnology Research, University of MarylandRockville, MD, USA
- Department of Plant Sciences and Landscape Architecture, University of MarylandCollege Park, MD, USA
| | - Dipti Bendigeri
- Institute for Bioscience and Biotechnology Research, University of MarylandRockville, MD, USA
- Department of Plant Sciences and Landscape Architecture, University of MarylandCollege Park, MD, USA
| | - Shunyuan Xiao
- Institute for Bioscience and Biotechnology Research, University of MarylandRockville, MD, USA
- Department of Plant Sciences and Landscape Architecture, University of MarylandCollege Park, MD, USA
| |
Collapse
|
46
|
Abstract
The bioactive sphingolipids including, ceramide, sphingosine, and sphingosine-1-phosphate (S1P) have important roles in several types of signaling and regulation of many cellular processes including cell proliferation, apoptosis, senescence, angiogenesis, and transformation. Recent accumulating evidence suggests that ceramide- and S1P-mediated pathways have been implicated in cancer development, progression, and chemotherapy. Ceramide mediates numerous cell-stress responses, such as induction of apoptosis and cell senescence, whereas S1P plays pivotal roles in cell survival, migration, and inflammation. These sphingolipids with opposing roles can be interconverted within cells, suggesting that the balance between them is related to cell fate. Importantly, these sphingolipids are metabolically related through actions of enzymes including ceramidases, ceramide synthases, sphingosine kinases, and S1P phosphatases thereby forming a network of metabolically interrelated bioactive lipid mediators whose importance in normal cellular function and diseases is gaining appreciation. In this review, we summarize involvement of sphingolipids and their related enzymes in pathogenesis and therapy of cancer and discuss future directions of sphingolipid field in cancer research.
Collapse
Affiliation(s)
- Hideki Furuya
- University of Hawaii Cancer Center, 651 Ilalo Street, BSB #222H, Honolulu, HI 96813, USA
| | | | | |
Collapse
|
47
|
Shupik MA, Vanin AF, Alessenko AV. Interaction of the nitric oxide signaling system with the sphingomyelin cycle and peroxidation on transmission of toxic signal of tumor necrosis factor-α in ischemia-reperfusion. BIOCHEMISTRY (MOSCOW) 2012; 76:1197-209. [PMID: 22117546 DOI: 10.1134/s0006297911110010] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
This review discusses the functional role of nitric oxide in ischemia-reperfusion injury and mechanisms of signal transduction of apoptosis, which accompanies ischemic damage to organs and tissues. On induction of apoptosis an interaction is observed of the nitric oxide signaling system with the sphingomyelin cycle, which is a source of a proapoptotic agent ceramide. Evidence is presented of an interaction of the sphingomyelin cycle enzymes and ceramide with nitric oxide and enzymes synthesizing nitric oxide. The role of a proinflammatory cytokine TNF-α in apoptosis and ischemia-reperfusion and mechanisms of its cytotoxic action, which involve nitric oxide, the sphingomyelin cycle, and lipid peroxidation are discussed. A comprehensive study of these signaling systems provides insight into the molecular mechanism of apoptosis during ischemia and allows us to consider new approaches for treatment of diseases associated with the activation of apoptosis.
Collapse
Affiliation(s)
- M A Shupik
- Emanuel Institute of Biochemical Physics, Russian Academy of Sciences, Moscow, Russia
| | | | | |
Collapse
|
48
|
Gault CR, Obeid LM. Still benched on its way to the bedside: sphingosine kinase 1 as an emerging target in cancer chemotherapy. Crit Rev Biochem Mol Biol 2011; 46:342-51. [PMID: 21787121 DOI: 10.3109/10409238.2011.597737] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
For several decades, lipid biologists have investigated how sphingolipids contribute to physiology, cell biology, and cell fate. Foremost among these discoveries is the finding that the bioactive sphingolipids ceramide, sphingosine, and sphingosine-1-phosphate (S1P) have diverse and often opposing effects on cell fate. Interestingly, these bioactive sphingolipids can be interconverted by just a few enzymatic reactions. Therefore, much attention has been paid to the enzymes which govern these reactions with a disproportionate amount of focus on the enzyme sphingosine kinase 1 (SK1). Several studies have found that tissue expression of SK1 correlates with cancer stage, chemotherapy response, and tumor aggressiveness. In addition, overexpression of SK1 in multiple cancer cell lines increases their resistance to chemotherapy, promotes proliferation, allows for anchorage independent growth, and increases local angiogenesis. Inhibition of SK1 using either pharmacological inhibitors or by crossing SK1 null mice has shown promise in many xenograft models of cancer, as well as several genetic and chemically induced mouse models of carcinogenesis. Here, we review the majority of the evidence that suggests SK1 is a promising target for the prevention and/or treatment of various cancers. Also, we strongly advocate for further research into basic mechanisms of bioactive sphingolipid signaling, and an increased focus on the efficacy of SK inhibitors in non-xenograft models of cancer progression.
Collapse
Affiliation(s)
- Christopher R Gault
- Department of Medicine, Medical University of South Carolina, Charleston, SC 29425-7790, USA
| | | |
Collapse
|
49
|
Kornhuber J, Muehlbacher M, Trapp S, Pechmann S, Friedl A, Reichel M, Mühle C, Terfloth L, Groemer TW, Spitzer GM, Liedl KR, Gulbins E, Tripal P. Identification of novel functional inhibitors of acid sphingomyelinase. PLoS One 2011; 6:e23852. [PMID: 21909365 PMCID: PMC3166082 DOI: 10.1371/journal.pone.0023852] [Citation(s) in RCA: 127] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2011] [Accepted: 07/26/2011] [Indexed: 12/19/2022] Open
Abstract
We describe a hitherto unknown feature for 27 small drug-like molecules, namely functional inhibition of acid sphingomyelinase (ASM). These entities named FIASMAs (Functional Inhibitors of Acid SphingoMyelinAse), therefore, can be potentially used to treat diseases associated with enhanced activity of ASM, such as Alzheimer's disease, major depression, radiation- and chemotherapy-induced apoptosis and endotoxic shock syndrome. Residual activity of ASM measured in the presence of 10 µM drug concentration shows a bimodal distribution; thus the tested drugs can be classified into two groups with lower and higher inhibitory activity. All FIASMAs share distinct physicochemical properties in showing lipophilic and weakly basic properties. Hierarchical clustering of Tanimoto coefficients revealed that FIASMAs occur among drugs of various chemical scaffolds. Moreover, FIASMAs more frequently violate Lipinski's Rule-of-Five than compounds without effect on ASM. Inhibition of ASM appears to be associated with good permeability across the blood-brain barrier. In the present investigation, we developed a novel structure-property-activity relationship by using a random forest-based binary classification learner. Virtual screening revealed that only six out of 768 (0.78%) compounds of natural products functionally inhibit ASM, whereas this inhibitory activity occurs in 135 out of 2028 (6.66%) drugs licensed for medical use in humans.
Collapse
Affiliation(s)
- Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University of Erlangen, Erlangen, Germany.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Watters RJ, Liu X, Loughran TP. T-cell and natural killer-cell large granular lymphocyte leukemia neoplasias. Leuk Lymphoma 2011; 52:2217-25. [PMID: 21749307 DOI: 10.3109/10428194.2011.593276] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Large granular lymphocyte (LGL) leukemia is a rare disorder of cytotoxic lymphocytes. LGL cells play an integral role in the immune system and are divided into two major lineages of CD3(-)natural killer (NK) cells and CD3(+) T cells that circulate throughout the blood in search of infected cells, in which they will make contact through a receptor ligand and induce cell death. LGL cells are also programmed to undergo apoptosis after contact with an infected target cell; however, they continue to survive in individuals with LGL leukemia. This unchecked proliferation and cytotoxicity of LGLs in patients results in autoimmunity or malignancy. Rheumatoid arthritis is the most common autoimmune condition seen in individuals with LGL leukemia; however, LGL leukemia is associated with a wide spectrum of other autoimmune diseases. Patients may also suffer from other hematological conditions including hemolytic anemia, pure red cell aplasia, and neutropenia, which lead to recurrent bacterial infections. Currently, the only established treatment involves a low dose of an immunosuppressive regimen with methotrexate, in which 40-50% of patients are either resistant or do not respond. In order to establish new therapeutics it is important to understand the current state of LGL leukemia both in the clinic and in basic research.
Collapse
Affiliation(s)
- Rebecca J Watters
- Penn State Hershey Cancer Institute, Pennsylvania State College of Medicine, Hershey, PA 17033-0850, USA
| | | | | |
Collapse
|