Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Am J Physiol Lung Cell Mol Physiol ; 322(1): L149-L161, 2022 01 01.
Article in English | MEDLINE | ID: mdl-35015568

ABSTRACT

Disruption of the lung endothelial barrier is a hallmark of acute respiratory distress syndrome (ARDS), for which no effective pharmacologic treatments exist. Prior work has demonstrated that FTY720 S-phosphonate (Tys), an analog of sphingosine-1-phosphate (S1P) and FTY720, exhibits potent endothelial cell (EC) barrier protective properties. In this study, we investigated the in vitro and in vivo efficacy of Tys against methicillin-resistant Staphylococcus aureus (MRSA), a frequent bacterial cause of ARDS. Tys-protected human lung EC from barrier disruption induced by heat-killed MRSA (HK-MRSA) or staphylococcal α-toxin and attenuated MRSA-induced cytoskeletal changes associated with barrier disruption, including actin stress fiber formation and loss of peripheral VE-cadherin and cortactin. Tys-inhibited Rho and myosin light chain (MLC) activation after MRSA and blocked MRSA-induced NF-κB activation and release of the proinflammatory cytokines, IL-6 and IL-8. In vivo, intratracheal administration of live MRSA in mice caused significant vascular leakage and leukocyte infiltration into the alveolar space. Pre- or posttreatment with Tys attenuated MRSA-induced lung permeability and levels of alveolar neutrophils. Posttreatment with Tys significantly reduced levels of bronchoalveolar lavage (BAL) VCAM-1 and plasma IL-6 and KC induced by MRSA. Dynamic intravital imaging of mouse lungs demonstrated Tys attenuation of HK-MRSA-induced interstitial edema and neutrophil infiltration into lung tissue. Tys did not directly inhibit MRSA growth or viability in vitro. In conclusion, Tys inhibits lung EC barrier disruption and proinflammatory signaling induced by MRSA in vitro and attenuates acute lung injury induced by MRSA in vivo. These results support the potential utility of Tys as a novel ARDS therapeutic strategy.


Subject(s)
Acute Lung Injury/microbiology , Acute Lung Injury/pathology , Cell Membrane Permeability , Endothelial Cells/microbiology , Fingolimod Hydrochloride/analogs & derivatives , Methicillin-Resistant Staphylococcus aureus/physiology , Organophosphonates/pharmacology , Animals , Antigens, CD/metabolism , Cadherins/metabolism , Cell Membrane Permeability/drug effects , Cytoprotection/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Enzyme Activation/drug effects , Fingolimod Hydrochloride/pharmacology , Humans , Inflammation/pathology , Mice , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Signal Transduction/drug effects , rhoA GTP-Binding Protein/metabolism
2.
Neuropharmacology ; 186: 108464, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33460688

ABSTRACT

The sphingosine 1-phosphate (S1P) receptor 1 (S1P1) has emerged as a therapeutic target for the treatment of multiple sclerosis (MS). Fingolimod (FTY720) is the first functional antagonist of S1P1 that has been approved for oral treatment of MS. Previously, we have developed novel butterfly derivatives of FTY720 that acted similar to FTY720 in reducing disease symptoms in a mouse model of experimental autoimmune encephalomyelitis (EAE). In this study, we have synthesized a piperidine derivative of the oxazolo-oxazole compounds, denoted ST-1505, and its ring-opened analogue ST-1478, and characterised their in-vitro and in-vivo functions. Notably, the 3-piperidinopropyloxy moiety resembles a structural motif of pitolisant, a drug with histamine H3R antagonistic/inverse agonist activity approved for the treatment of narcolepsy. Both novel compounds exerted H3R affinities, and in addition, ST-1505 was characterised as a dual S1P1+3 agonist, whereas ST-1478 was a dual S1P1+5 agonist. Both multitargeting compounds were also active in mice and reduced the lymphocyte numbers as well as diminished disease symptoms in the mouse model of MS. The effect of ST-1478 was dependent on SK-2 activity suggesting that it is a prodrug like FTY720, but with a more selective S1P receptor activation profile, whereas ST-1505 is a fully active drug even in the absence of SK-2. In summary, these data suggest that the well soluble piperidine derivatives ST-1505 and ST-1478 hold promise as novel drugs for the treatment of MS and other autoimmune or inflammatory diseases, and by their H3R antagonist potency, they might additionally improve cognitive impairment during disease.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Fingolimod Hydrochloride/administration & dosage , Histamine H3 Antagonists/administration & dosage , Multiple Sclerosis/prevention & control , Neuroprotective Agents/administration & dosage , Sphingosine-1-Phosphate Receptors/agonists , Animals , CHO Cells , Cricetinae , Cricetulus , Encephalomyelitis, Autoimmune, Experimental/metabolism , Female , Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/chemistry , Histamine H3 Antagonists/chemistry , Histamine H3 Antagonists/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multiple Sclerosis/metabolism , Neuroprotective Agents/chemistry , Protein Structure, Secondary , Sphingosine-1-Phosphate Receptors/metabolism
3.
Arch Pharm Res ; 43(10): 1046-1055, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33111965

ABSTRACT

Fingolimod has been evaluated for use as an anticancer agent. However, many steps are required to synthesize fingolimod because of its intricate structure. A fingolimod analogue, N-(4-(2-((4-methoxybenzyl)amino)ethyl)phenyl)heptanamide (MPH), also has anti-cancer effects and is easier to synthesize but is poorly soluble in water. To compensate for its poor water solubility, MPH-loaded polymeric micelles were prepared by thin film hydration method using various polymers and the physicochemical properties of the MPH-loaded micelles such as particle size, drug-loading (DL, %), and encapsulation efficiency (EE, %) were evaluated. A storage stability test was conducted to select the final formulation and the release profile of the MPH-loaded micelles was confirmed by in vitro release assay. MPH-loaded mPEG-b-PLA micelles were selected for further testing based on their stability and physicochemical properties; they were stable for stable for 14 days at 4 °C and 25 °C and for 7 days at 37 °C. They showed anti-cancer efficacy against both A549 and U87 cancer cells. Encapsulation of MPH in polymeric micelles did not decrease the in vitro cytotoxicity of MPH. The findings of this study lay the groundwork for future formulations that enable the effective and stable delivery of poorly water-soluble agents.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Drug Carriers/chemistry , Fingolimod Hydrochloride/pharmacokinetics , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Cell Line, Tumor , Drug Liberation , Drug Screening Assays, Antitumor , Drug Stability , Fingolimod Hydrochloride/administration & dosage , Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/chemistry , Humans , Micelles , Particle Size , Polymers/chemistry , Solubility , Water/chemistry
4.
Int J Mol Sci ; 21(18)2020 Sep 04.
Article in English | MEDLINE | ID: mdl-32899717

ABSTRACT

Multiple sclerosis (MS) is a chronic, inflammatory, autoimmune disease of the central nervous system (CNS) which is associated with lower life expectancy and disability. The experimental antigen-induced encephalomyelitis (EAE) in mice is a useful animal model of MS, which allows exploring the etiopathogenetic mechanisms and testing novel potential therapeutic drugs. A new therapeutic paradigm for the treatment of MS was introduced in 2010 through the sphingosine 1-phosphate (S1P) analogue fingolimod (FTY720, Gilenya®), which acts as a functional S1P1 antagonist on T lymphocytes to deplete these cells from the blood. In this study, we synthesized two novel structures, ST-1893 and ST-1894, which are derived from fingolimod and chemically feature a morpholine ring in the polar head group. These compounds showed a selective S1P1 activation profile and a sustained S1P1 internalization in cultures of S1P1-overexpressing Chinese hamster ovary (CHO)-K1 cells, consistent with a functional antagonism. In vivo, both compounds induced a profound lymphopenia in mice. Finally, these substances showed efficacy in the EAE model, where they reduced clinical symptoms of the disease, and, on the molecular level, they reduced the T-cell infiltration and several inflammatory mediators in the brain and spinal cord. In summary, these data suggest that S1P1-selective compounds may have an advantage over fingolimod and siponimod, not only in MS but also in other autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/metabolism , Fingolimod Hydrochloride/pharmacology , Morpholinos/pharmacology , Animals , CHO Cells , Central Nervous System/drug effects , Cricetulus , Disease Models, Animal , Encephalomyelitis/drug therapy , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Fingolimod Hydrochloride/analogs & derivatives , Immunosuppressive Agents/therapeutic use , Ligands , Lymphopenia/drug therapy , Lysophospholipids/metabolism , Mice , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors/drug effects , Sphingosine-1-Phosphate Receptors/metabolism , Spinal Cord/drug effects , T-Lymphocytes/drug effects
5.
J Parkinsons Dis ; 10(1): 185-192, 2020.
Article in English | MEDLINE | ID: mdl-31561385

ABSTRACT

BACKGROUND: Parkinson's disease (PD) and multiple system atrophy (MSA) patients often suffer from gastrointestinal (GI) dysfunction and GI dysbiosis (microbial imbalance). GI dysfunction also occurs in mouse models of PD and MSA. OBJECTIVES: To assess gut dysfunction and dysbiosis in PD subjects as compared to controls, identify potential shared microbial taxa in humans and mouse models of PD and MSA, and to assess the effects of potential therapies on mouse GI microbiota. METHODS: In this human pilot study, GI function was assessed by fecal consistency/frequency measured using the Bristol Stool Form Scale and GI transit time assessed using Sitzmarks pills and abdominal radiology. Human and mouse microbiota were analyzed by extracting fecal genomic DNA followed by 16S rRNA sequencing. RESULTS: In our PD patients genera Akkermansia significantly increased while a trend toward increased Bifidobacterium and decreased Prevotella was observed. Families Bacteroidaceae and Lachnospiraceae and genera Prevotella and Bacteroides were detected in both humans and PD mice, suggesting potential shared biomarkers. In mice treated with the approved multiple sclerosis drug, FTY720, or with our FTY720-Mitoxy-derivative, we saw that FTY720 had little effect while FTY720-Mitoxy increased beneficial Ruminococcus and decreased Rickenellaceae family. CONCLUSION: Akkermansia and Prevotellaceae data reported by others were replicated in our human pilot study suggesting the use of those taxa as potential biomarkers for PD diagnosis. The effect of FTY720-Mitoxy on taxa Rikenellaceae and Ruminococcus and the relevance of S24-7 await further evaluation. It also remains to be determined if mouse microbiota have predictive power for human subjects.


Subject(s)
Dysbiosis/microbiology , Fingolimod Hydrochloride/pharmacology , Gastrointestinal Microbiome , Immunosuppressive Agents/pharmacology , Microbiota , Multiple System Atrophy/microbiology , Parkinson Disease/microbiology , Adult , Aged , Aged, 80 and over , Animals , Constipation/physiopathology , Disease Models, Animal , Female , Fingolimod Hydrochloride/administration & dosage , Fingolimod Hydrochloride/analogs & derivatives , Gastrointestinal Microbiome/drug effects , Gastrointestinal Motility/physiology , Humans , Immunosuppressive Agents/administration & dosage , Male , Mice , Mice, Transgenic , Middle Aged , Pilot Projects , RNA, Ribosomal, 16S
6.
Cancer Lett ; 468: 1-13, 2020 01 01.
Article in English | MEDLINE | ID: mdl-31593801

ABSTRACT

Acute myeloid leukemia (AML) is an aggressive disease associated with very poor prognosis. Most patients are older than 60 years, and in this group only 5-15% of cases survive over 5 years. Therefore, it is urgent to develop more effective targeted therapies. Inactivation of protein phosphatase 2 A (PP2A) is a recurrent event in AML, and overexpression of its endogenous inhibitor SET is detected in ~30% of patients. The PP2A activating drug FTY720 has potent anti-leukemic effects; nevertheless, FTY720 induces cardiotoxicity at the anti-neoplastic dose. Here, we have developed a series of non-phosphorylable FTY720 analogues as a new therapeutic strategy for AML. Our results show that the lead compound CM-1231 re-activates PP2A by targeting SET-PP2A interaction, inhibiting cell proliferation and promoting apoptosis in AML cell lines and primary patient samples. Notably, CM-1231 did not induce cardiac toxicity, unlike FTY720, in zebrafish models, and reduced the invasion and aggressiveness of AML cells more than FTY720 in zebrafish xenograft models. In conclusion, CM-1231 is safer and more effective than FTY720; therefore, this compound could represent a novel and promising approach for treating AML patients with SET overexpression.


Subject(s)
Cardiotoxicity/prevention & control , DNA-Binding Proteins/metabolism , Fingolimod Hydrochloride/administration & dosage , Histone Chaperones/metabolism , Leukemia, Myeloid, Acute/drug therapy , Protein Phosphatase 2/metabolism , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/drug effects , Cardiotoxicity/etiology , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/toxicity , Heart Rate/drug effects , Humans , Leukemia, Myeloid, Acute/pathology , Male , Middle Aged , Protein Binding/drug effects , Toxicity Tests, Acute , Xenograft Model Antitumor Assays , Zebrafish
7.
Exp Neurol ; 325: 113120, 2020 03.
Article in English | MEDLINE | ID: mdl-31751571

ABSTRACT

Multiple system atrophy (MSA) is a fatal disorder with no effective treatment. MSA pathology is characterized by α-synuclein (aSyn) accumulation in oligodendrocytes, the myelinating glial cells of the central nervous system (CNS). aSyn accumulation in oligodendrocytes forms the pathognomonic glial cytoplasmic inclusions (GCIs) of MSA. MSA aSyn pathology is also associated with motor and autonomic dysfunction, including an impaired ability to sweat. MSA patients have abnormal CNS expression of glial-cell-line-derived neurotrophic factor (GDNF) and brain-derived neurotrophic factor (BDNF). Our prior studies using the parent compound FTY720, a food and drug administration (FDA) approved immunosuppressive for multiple sclerosis, reveal that FTY720 protects parkinsonian mice by increasing BDNF. Our FTY720-derivative, FTY720-Mitoxy, is known to increase expression of oligodendrocyte BDNF, GDNF, and nerve growth factor (NGF) but does not reduce levels of circulating lymphocytes as it is not phosphorylated so cannot modulate sphingosine 1 phosphate receptors (S1PRs). To preclinically assess FTY720-Mitoxy for MSA, we used mice expressing human aSyn in oligodendrocytes under a 2,' 3'-cyclic nucleotide 3'-phosphodiesterase (CNP) promoter. CNP-aSyn transgenic (Tg) mice develop motor dysfunction between 7 and 9 mo, and progressive GCI pathology. Using liquid chromatography-mass spectrometry (LC-MS/MS) and enzymatic assays, we confirmed that FTY720-Mitoxy was stable and active. Vehicle or FTY720-Mitoxy (1.1 mg/kg/day) was delivered to wild type (WT) or Tg littermates from 8.5-11.5 mo by osmotic pump. We behaviorally assessed their movement by rotarod and sweat production by starch­iodine test. Postmortem tissues were evaluated by qPCR for BDNF, GDNF, NGF and GDNF-receptor RET mRNA and for aSyn, BDNF, GDNF, and Iba1 protein by immunoblot. MicroRNAs (miRNAs) were also assessed by qPCR. FTY720-Mitoxy normalized movement, sweat function and soleus muscle mass in 11.5 mo Tg MSA mice. FTY720-Mitoxy also increased levels of brain GDNF and reduced brain miR-96-5p, a miRNA that acts to decrease GDNF expression. Moreover, FTY720-Mitoxy blocked aSyn pathology measured by sequential protein extraction and immunoblot, and microglial activation assessed by immunohistochemistry and immunoblot. In the 3-nitropropionic acid (3NP) toxin model of MSA, FTY720-Mitoxy protected movement and mitochondria in WT and CNP-aSyn Tg littermates. Our data confirm potent in vivo protection by FTY720-Mitoxy, supporting its further evaluation as a potential therapy for MSA and related synucleinopathies.


Subject(s)
Fingolimod Hydrochloride/analogs & derivatives , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Multiple System Atrophy/pathology , Neuroprotective Agents/pharmacology , Animals , Behavior, Animal/drug effects , Disease Models, Animal , Female , Fingolimod Hydrochloride/pharmacology , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/drug effects , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Transgenic , MicroRNAs/drug effects , MicroRNAs/metabolism , Multiple System Atrophy/metabolism , Proto-Oncogene Proteins c-ret/biosynthesis , Proto-Oncogene Proteins c-ret/drug effects , alpha-Synuclein/genetics
8.
Neuropharmacology ; 158: 107701, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31291595

ABSTRACT

Multiple system atrophy (MSA) is a fatal demyelinating disorder lacking any disease-modifying therapies. MSA pathology stems from aggregated α-synuclein (aSyn) accumulation in glial cytosolic inclusions of oligodendroglial cell (OLGs), the myelinating cells of brain. In MSA brains and in MSA animal models with aSyn accumulation in OLGs, aberrant expression of brain-derived neurotrophic factor (BDNF) and glial-cell-line-derived neurotrophic factor (GDNF) occur. Nerve growth factor (NGF) expression can also be altered in neurodegenerative diseases. It is unclear if oxidative stress impacts the viability of aSyn-accumulating OLG cells. Here, we show that OLN-93 cells stably expressing human wild type aSyn or the MSA-associated-aSyn-mutants G51D or A53E, are more vulnerable to oxidative stress. In dose response studies we found that OLN-93 cells treated 48 h with 160 nM FTY720 or our new non-immunosuppressive FTY720-C2 or FTY720-Mitoxy derivatives sustained normal viability. Also, FTY720, FTY720-C2, and FTY720-Mitoxy all stimulated NGF expression at 24 h. However only FTY720-Mitoxy also increased BDNF and GDNF mRNA at 24 h, an effect paralleled by increases in histone 3 acetylation and ERK1/2 phosphorylation. Myelin associated glycoprotein (MAG) levels were also increased in OLN-93 cells after 48 h treatment with FTY720-Mitoxy. FTY720, FTY720-C2, and FTY720-Mitoxy all prevented oxidative-stress-associated-cell-death of OLN-93 cells that lack any aSyn expression. However, only FTY720-Mitoxy protected MSA-like aSyn-expressing-OLN-93-cells against oxidative-cell-death. These data identify potent protective effects for FTY720-Mitoxy with regard to trophic factors as well as MAG expression by OLG cells. Testing of FTY720-Mitoxy in mice is thus a judicious next step for neuropharmacological preclinical development.


Subject(s)
Ceramides/pharmacology , Fingolimod Hydrochloride/analogs & derivatives , Multiple System Atrophy/metabolism , Oligodendroglia/drug effects , Oxidative Stress/drug effects , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , alpha-Synuclein/drug effects , Animals , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/metabolism , Cell Line , Fingolimod Hydrochloride/pharmacology , Glial Cell Line-Derived Neurotrophic Factor/drug effects , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Myelin-Associated Glycoprotein/drug effects , Myelin-Associated Glycoprotein/metabolism , Nerve Growth Factor/drug effects , Nerve Growth Factor/metabolism , Oligodendroglia/metabolism , Rats , alpha-Synuclein/metabolism
9.
Pharmazie ; 74(2): 107-110, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30782260

ABSTRACT

SYL-927 is a selective sphingosine-1-phosphate receptor 1 (S1P1) agonist for autoimmune diseases. It undergoes phosphorylation to the active SYL-927-P in vivo, which activates S1P1 on lymphocytes, causing lymphopenia by retention of lymphocytes in the lymph nodes. The aim of this study was to identify the involvement of blood cells in the phosphorylation of SYL-927. In addition, pharmacokinetics of SYL-927 and SYL-927-P in blood and plasma were compared in rats. The results demonstrated that SYL-927 can be converted to SYL-927-P in rat blood, but not in rat plasma. However, both rat blood and plasma are capable of dephosphorylating SYL-927-P to SYL-927. SYL-927-P generation and release were observed after incubating SYL-927 with rat and human erythrocytes and platelets. The addition of sphingosine kinases (SPHKs) inhibitors N,N-dimethylsphingosine (DMS) and FTY720 significantly inhibited SYL-927-P generation, indicating the involvement of SPHKs. In addition, SYL-927 and SYL-927-P levels in blood were significantly higher than those in plasma after oral administration of SYL-927 in rats, suggesting the blood cells for the production of SYL-927-P. In summary, the blood cells such as erythrocytes and platelets contribute to the generation and release of SYL-927-P, which is important for maintaining plasma active phosphate levels for prolonged effects.


Subject(s)
Blood Platelets/metabolism , Erythrocytes/metabolism , Fingolimod Hydrochloride/analogs & derivatives , Immunosuppressive Agents/blood , Receptors, Lysosphingolipid/agonists , Administration, Oral , Animals , Fingolimod Hydrochloride/administration & dosage , Fingolimod Hydrochloride/blood , Humans , Immunosuppressive Agents/administration & dosage , Phosphorylation , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/blood , Rats , Sphingosine/administration & dosage , Sphingosine/analogs & derivatives , Sphingosine/blood
10.
J Neurol Sci ; 399: 6-14, 2019 Apr 15.
Article in English | MEDLINE | ID: mdl-30738334

ABSTRACT

Subarachnoid hemorrhage (SAH) results in neurological damage, acute cardiac damage and has a high mortality rate. Immunoresponse in the acute phase after SAH plays a key role in mediating vasospasm, edema, inflammation and neuronal damage. The S1P/S1PR pathway impacts multiple cellular functions, exerts anti-inflammatory and anti-apoptotic effects, promotes remyelination, and improves outcome in several central nervous system (CNS) diseases. RP001 hydrochloride is a novel S1PR agonist, which sequesters lymphocytes within their secondary tissues and prevents infiltration of immune cells into the CNS thereby reducing immune response. In this study, we investigated whether RP001 attenuates neuronal injury after SAH by reducing inflammation. S1PRs, specifically S1PR1, 3 not only exerts anti-inflammatory effects, but also decreases heart rate and induces atrioventricular conduction abnormalities. Therefore, we also tested whether RP001 treatment of SAH regulates cardiac functional outcome. Male adult C57BL/6 mice were subjected to SAH, and neurological function tests, echocardiography, and immunohistochemical analysis were performed. SAH induces neurological deficits and acute cardiac dysfunction compared to sham control mice. Treatment of SAH with a low-dose of RP001 induces better neurological outcome and cardiac function compared to a high-dose of RP001. Low-dose-RP001 treatment significantly decreases apoptosis, white matter damage, blood brain barrier permeability, microglial/astrocyte activation, macrophage chemokine protein-1, matrix metalloproteinase-9 and NADPH oxidase-2 expression in the brain compared to SAH control mice. Our findings indicate that low-dose of RP001 alleviates neurological damage after SAH, in part by decreasing neuroinflammation.


Subject(s)
Brain/drug effects , Neurons/drug effects , Recovery of Function/drug effects , Sphingosine 1 Phosphate Receptor Modulators/therapeutic use , Sphingosine-1-Phosphate Receptors/agonists , Subarachnoid Hemorrhage/drug therapy , Animals , Apoptosis/drug effects , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/metabolism , Disease Models, Animal , Fingolimod Hydrochloride/analogs & derivatives , Heart Rate/drug effects , Inflammation/drug therapy , Male , Mice , Neurons/metabolism , Signal Transduction/drug effects , Sphingosine 1 Phosphate Receptor Modulators/pharmacology , Subarachnoid Hemorrhage/metabolism
11.
Neurosci Lett ; 690: 178-180, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30359694

ABSTRACT

In searching for Parkinson's disease (PD) pharmacotherapies we began studying FTY720, a food and drug administration (FDA) approved drug. We also created derivatives, FTY720-C2 and FTY720-Mitoxy, and began assessing them. Here we treated dopaminergic MN9D cells with FTY720s then measured microRNA (miRNA) levels by PCR arrays. We discovered that all three FTY720s increased miR376b-3p, while FTY720-C2 also increased miR-128-3p, miR-146b-5p, miR-7a-5p, and miR-9-5p, and FTY720-Mitoxy also increased miR-30d-5p. Investigations revealed that some miRNAs downregulate alpha-synuclein, while others reduce apoptosis, suggesting that FTY720s may act to reduce synucleinopathy and dopaminergic neuron loss in PD and related disorders.


Subject(s)
Ceramides/pharmacology , Dopaminergic Neurons/metabolism , Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/pharmacology , MicroRNAs/metabolism , Neuroprotective Agents/metabolism , Up-Regulation/drug effects , Animals , Cells, Cultured , Mice
12.
Molecules ; 23(11)2018 Oct 24.
Article in English | MEDLINE | ID: mdl-30355990

ABSTRACT

FTY720 inhibits various cancers through PP2A activation. The structure of FTY720 is also used as a basic structure for the design of sphingosine kinase (SK) inhibitors. We have synthesized derivatives using an amide chain in FTY720 with a phenyl backbone, and then compounds were screened by an MTT cell viability assay. The PP2A activity of compound 7 was examined. The phosphorylation levels of AKT and ERK, downstream targets of PP2A, in the presence of compound 7, were determined. Compound 7 may exhibit anticancer effects through PP2A activation rather than the mechanism by inhibition of SK1 in cancer cells. In the docking study of compound 7 and PP2A, the amide chain of compound 7 showed an interaction with Asn61 that was different from FTY720, which is expected to affect the activity of the compound.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacology , Fingolimod Hydrochloride/chemical synthesis , Fingolimod Hydrochloride/pharmacology , Protein Phosphatase 2/antagonists & inhibitors , Cell Line, Tumor , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Fingolimod Hydrochloride/analogs & derivatives , Humans , Inhibitory Concentration 50 , Models, Molecular , Molecular Conformation , Molecular Structure , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Structure-Activity Relationship
13.
Biopharm Drug Dispos ; 39(9): 431-436, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30362120

ABSTRACT

SYL-927, a novel and selective S1P1 agonist, is transferred to its active phosphate for the regulation of lymphocyte recirculation. This in vitro metabolism study is to elucidate the P450-mediated oxidation pathway of SYL-927 in human liver microsomes (HLMs). The results demonstrated that the ω-1 hydroxylated metabolite SYL-927-M was formed after incubation of SYL-927 with HLMs. Recombinant human CYP1A1 and CYP2J2 can efficiently catalyse SYL-927-M formation, followed by markedly less substrate conversion with CYP1A2, CYP2C19 and CYP2D6. Inhibition studies with chemical inhibitors and antibodies suggested that arachidonic acid, the substrate of CYP2J2, and CYP2J2-specific antibody effectively inhibited the formation of SYL-927-M in HLMs whereas no significant inhibition was observed with the inhibitors for CYP1A1, CYP1A2, CYP2C9, CYP2C19, CYP2D6, CYP2E1 and CYP3A4, demonstrating that CYP2J2 was primarily responsible for the formation of SYL-927-M.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Fingolimod Hydrochloride/analogs & derivatives , Microsomes, Liver/enzymology , Oxazoles/metabolism , Sphingosine-1-Phosphate Receptors/agonists , Cytochrome P-450 CYP2J2 , Humans , Hydroxylation , Oxazoles/pharmacology
14.
Eur J Med Chem ; 159: 217-242, 2018 Nov 05.
Article in English | MEDLINE | ID: mdl-30292898

ABSTRACT

A series of compounds containing pyrrolidine and pyrrolizidine cores with appended hydrophobic substituents were prepared as constrained analogs of FTY720 and phytosphingosine. The effect of these compounds on the viability of cancer cells, on downregulation of the nutrient transport systems, and on their ability to cause vacuolation was studied. An attempt to inhibit HDACs with some phosphate esters of our analogs was thwarted by our failure to reproduce the reported inhibitory action of FTY720-phosphate.


Subject(s)
Antineoplastic Agents/pharmacology , Epigenesis, Genetic/drug effects , Fingolimod Hydrochloride/pharmacology , Sphingosine/analogs & derivatives , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/chemistry , Mice , Molecular Structure , Sphingosine/chemical synthesis , Sphingosine/chemistry , Sphingosine/pharmacology , Structure-Activity Relationship
15.
Chem Commun (Camb) ; 54(85): 12002-12005, 2018 Oct 23.
Article in English | MEDLINE | ID: mdl-30221278

ABSTRACT

The direct, catalytic vicinal difluorination of terminal alkenes via an I(i)/I(iii) manifold was exploited to install a chiral, hybrid bioisostere of the CF3 and Et groups (BITE) in Gilenya®; the first orally available drug for the clinical management of Multiple Sclerosis (MS). This subtle fluorination pattern allows lipophilicity (log D) to be tempered compared to the corresponding CF3 and Et derivatives (CH2CH3 > CH2CF3 > CHFCH2F).


Subject(s)
Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/chemistry , Immunosuppressive Agents/chemistry , Animals , Drug Stability , Fingolimod Hydrochloride/chemical synthesis , Halogenation , Hepatocytes/drug effects , Humans , Immunosuppressive Agents/chemical synthesis , Microsomes, Liver/metabolism , Multiple Sclerosis/drug therapy , Rats
16.
Bioorg Med Chem ; 24(10): 2273-9, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27068143

ABSTRACT

SYL927 and SYL930 are selective S1P1 agonists under preclinical development. However, during their pharmacokinetic studies we detected two metabolites in rat blood that were tentatively identified as monohydroxylated metabolites of SYL927 and SYL930 based on LC-MS/MS data. In this study, we designed and synthesized possible monohydroxylated products 6a-e and used them as references to confirm the structures of the two metabolites detected by LC-MS/MS. We also evaluated the in vitro and in vivo biological activities of these two metabolites.


Subject(s)
Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/pharmacology , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Receptors, Lysosphingolipid/agonists , Animals , Chromatography, Liquid , Fingolimod Hydrochloride/administration & dosage , Hydroxylation , Immunosuppressive Agents/administration & dosage , Lymphocyte Count , Lymphocytes/drug effects , Rats, Sprague-Dawley , Receptors, Lysosphingolipid/immunology , Tandem Mass Spectrometry
17.
Chem Phys Lipids ; 194: 85-93, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26496151

ABSTRACT

Effective therapeutic agents are lacking for the prevention and reversal of vascular leak, a frequent pathophysiologic result of inflammatory processes such as acute respiratory distress syndrome (ARDS) and sepsis. We previously demonstrated the potent barrier-enhancing effects of related compounds sphingosine 1-phosphate (S1P), the pharmaceutical agent FTY720, and its analog (S)-FTY720 phosphonate (Tys) in models of inflammatory lung injury. In this study, we characterize additional novel FTY720 analogs for their potential to reduce vascular leak as well as utilize them as tools to better understand the mechanisms by which this class of agents modulates permeability. Transendothelial resistance (TER) and labeled dextran studies demonstrate that (R)-methoxy-FTY720 ((R)-OMe-FTY), (R)/(S)-fluoro-FTY720 (FTY-F), and ß-glucuronide-FTY720 (FTY-G) compounds display in vitro barrier-enhancing properties comparable or superior to FTY720 and S1P. In contrast, the (S)-methoxy-FTY720 ((S)-OMe-FTY) analog disrupts lung endothelial cell (EC) barrier integrity in TER studies in association with actin stress fiber formation and robust intracellular calcium release, but independent of myosin light chain or ERK phosphorylation. Additional mechanistic studies with (R)-OMe-FTY, FTY-F, and FTY-G suggest that lung EC barrier enhancement is mediated through lipid raft signaling, Gi-linked receptor coupling to downstream tyrosine phosphorylation events, and S1PR1-dependent receptor ligation. These results provide important mechanistic insights into modulation of pulmonary vascular barrier function by FTY720-related compounds and highlight common signaling events that may assist the development of novel therapeutic tools in the prevention or reversal of the pulmonary vascular leak that characterizes ARDS.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/drug effects , Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/pharmacology , Pulmonary Artery/cytology , Pulmonary Artery/drug effects , Cells, Cultured , Endothelial Cells/metabolism , Fingolimod Hydrochloride/chemistry , Humans , Permeability/drug effects , Pulmonary Artery/metabolism , Structure-Activity Relationship
18.
ACS Chem Biol ; 11(2): 409-14, 2016 Feb 19.
Article in English | MEDLINE | ID: mdl-26653336

ABSTRACT

FTY720 sequesters lymphocytes in secondary lymphoid organs through effects on sphingosine-1-phosphate (S1P) receptors. However, at higher doses than are required for immunosuppression, FTY720 also functions as an anticancer agent in multiple animal models. Our published work indicates that the anticancer effects of FTY720 do not depend on actions at S1P receptors but instead stem from FTY720s ability to restrict access to extracellular nutrients by down-regulating nutrient transporter proteins. This result was significant because S1P receptor activation is responsible for FTY720s dose-limiting toxicity, bradycardia, that prevents its use in cancer patients. Here, we describe diastereomeric and enantiomeric 3- and 4-C-aryl 2-hydroxymethyl pyrrolidines that are more active than the previously known analogues. Of importance is that these compounds fail to activate S1P1 or S1P3 receptors in vivo but retain inhibitory effects on nutrient transporter proteins and anticancer activity in solid tumor xenograft models. Our studies reaffirm that the anticancer activity of FTY720 does not depend upon S1P receptor activation and uphold the promise of using S1P receptor-inactive azacyclic FTY720 analogues in human cancer patients.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/therapeutic use , Neoplasms/drug therapy , Pyrrolidines/chemistry , Pyrrolidines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Carrier Proteins/metabolism , Cell Line, Tumor , Fingolimod Hydrochloride/pharmacology , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Mice, Nude , Neoplasms/metabolism , Neoplasms/pathology , Pyrrolidines/pharmacology , Receptors, Lysosphingolipid/metabolism
19.
Chem Phys Lipids ; 191: 16-24, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26272033

ABSTRACT

Effective therapeutic agents are lacking for the prevention and reversal of vascular leak, a frequent pathophysiologic result of inflammatory processes such as acute respiratory distress syndrome (ARDS) and sepsis. We previously demonstrated the potent barrier-enhancing effects of related compounds sphingosine 1-phosphate (S1P), the pharmaceutical agent FTY720, and its analog (S)-FTY720 phosphonate (Tys) in models of inflammatory lung injury. In this study, we characterize additional novel FTY720 analogs for their potential to reduce vascular leak as well as utilize them as tools to better understand the mechanisms by which this class of agents modulates permeability. Transendothelial resistance (TER) and labeled dextran studies demonstrate that (R)-methoxy-FTY720 ((R)-OMe-FTY), (R)/(S)-fluoro-FTY720 (FTY-F), and ß-glucuronide-FTY720 (FTY-G) compounds display in vitro barrier-enhancing properties comparable or superior to FTY720 and S1P. In contrast, the (S)-methoxy-FTY720 ((S)-OMe-FTY) analog disrupts lung endothelial cell (EC) barrier integrity in TER studies in association with actin stress fiber formation and robust intracellular calcium release, but independent of myosin light chain or ERK phosphorylation. Additional mechanistic studies with (R)-OMe-FTY, FTY-F, and FTY-G suggest that lung EC barrier enhancement is mediated through lipid raft signaling, Gi-linked receptor coupling to downstream tyrosine phosphorylation events, and S1PR1-dependent receptor ligation. These results provide important mechanistic insights into modulation of pulmonary vascular barrier function by FTY720-related compounds and highlight common signaling events that may assist the development of novel therapeutic tools in the prevention or reversal of the pulmonary vascular leak that characterizes ARDS.


Subject(s)
Fingolimod Hydrochloride/analogs & derivatives , Calcium/metabolism , Cell Line , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fingolimod Hydrochloride/pharmacology , Fluorides/chemistry , Glucuronides/chemistry , Humans , Lysophospholipids/metabolism , Microscopy, Fluorescence , Permeability/drug effects , Phosphorylation , Pulmonary Artery/cytology , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Sphingosine/metabolism
20.
Microvasc Res ; 99: 102-9, 2015 May.
Article in English | MEDLINE | ID: mdl-25862132

ABSTRACT

RATIONALE: Modulation of pulmonary vascular barrier function is an important clinical goal given the devastating effects of vascular leak in acute lung injury (ALI). We previously demonstrated that FTY720 S-phosphonate (Tys), an analog of sphingosine 1-phosphate (S1P) and FTY720, has more potent pulmonary barrier protective effects than these agents in vitro and in mouse models of ALI. Tys preserves expression of the barrier-promoting S1P1 receptor (S1PR1), whereas S1P and FTY720 induce its ubiquitination and degradation. Here we further characterize the novel barrier promoting effects of Tys in cultured human pulmonary endothelial cells (EC). METHODS/RESULTS: In human lung EC, Tys significantly increased peripheral redistribution of adherens junction proteins VE-cadherin and ß-catenin and tight junction protein ZO-1. Inhibition of VE-cadherin with blocking antibody significantly attenuated Tys-induced transendothelial resistance (TER) elevation, while ZO-1 siRNA partially inhibited this elevation. Tys significantly increased focal adhesion formation and phosphorylation of focal adhesion kinase (FAK). Pharmacologic inhibition of FAK significantly attenuated Tys-induced TER elevation. Tys significantly increased phosphorylation and peripheral redistribution of the actin-binding protein, cortactin, while cortactin siRNA partially attenuated Tys-induced TER elevation. Although Tys significantly increased phosphorylation of Akt and GSK3ß, neither PI3 kinase nor GSK3ß inhibition altered Tys-induced TER elevation. Tys significantly increased Rac1 activity, while inhibition of Rac1 activity significantly attenuated Tys-induced VE-cadherin redistribution and TER elevation. CONCLUSION: Junctional complex, focal adhesion rearrangement and Rac1 activation play critical roles in Tys-mediated barrier protection in pulmonary EC. These results provide mechanistic insights into the effects of this potential ALI therapy.


Subject(s)
Endothelial Cells/drug effects , Fingolimod Hydrochloride/analogs & derivatives , Fingolimod Hydrochloride/therapeutic use , Lung/blood supply , Organophosphonates/therapeutic use , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Antigens, CD/metabolism , Antigens, Nuclear/metabolism , Cadherins/metabolism , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Focal Adhesions/metabolism , Humans , Immunosuppressive Agents/therapeutic use , Lung/drug effects , Lysophospholipids/chemistry , Microscopy, Fluorescence , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA, Small Interfering/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/analogs & derivatives , Sphingosine/chemistry , Transcription Factors/metabolism , Zonula Occludens-1 Protein/metabolism , rac1 GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...