Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
J Neuroinflammation ; 20(1): 194, 2023 Aug 26.
Article En | MEDLINE | ID: mdl-37633912

BACKGROUND: Bruton's tyrosine kinase (BTK) is a key signaling node in B cell receptor (BCR) and Fc receptor (FcR) signaling. BTK inhibitors (BTKi) are an emerging oral treatment option for patients suffering from multiple sclerosis (MS). Remibrutinib (LOU064) is a potent, highly selective covalent BTKi with a promising preclinical and clinical profile for MS and other autoimmune or autoallergic indications. METHODS: The efficacy and mechanism of action of remibrutinib was assessed in two different experimental autoimmune encephalomyelitis (EAE) mouse models for MS. The impact of remibrutinib on B cell-driven EAE pathology was determined after immunization with human myelin oligodendrocyte glycoprotein (HuMOG). The efficacy on myeloid cell and microglia driven neuroinflammation was determined in the RatMOG EAE. In addition, we assessed the relationship of efficacy to BTK occupancy in tissue, ex vivo T cell response, as well as single cell RNA-sequencing (scRNA-seq) in brain and spinal cord tissue. RESULTS: Remibrutinib inhibited B cell-dependent HuMOG EAE in dose-dependent manner and strongly reduced neurological symptoms. At the efficacious oral dose of 30 mg/kg, remibrutinib showed strong BTK occupancy in the peripheral immune organs and in the brain of EAE mice. Ex vivo MOG-specific T cell recall response was reduced, but not polyclonal T cell response, indicating absence of non-specific T cell inhibition. Remibrutinib also inhibited RatMOG EAE, suggesting that myeloid cell and microglia inhibition contribute to its efficacy in EAE. Remibrutinib did not reduce B cells, total Ig levels nor MOG-specific antibody response. In brain and spinal cord tissue a clear anti-inflammatory effect in microglia was detected by scRNA-seq. Finally, remibrutinib showed potent inhibition of in vitro immune complex-driven inflammatory response in human microglia. CONCLUSION: Remibrutinib inhibited EAE models by a two-pronged mechanism based on inhibition of pathogenic B cell autoreactivity, as well as direct anti-inflammatory effects in microglia. Remibrutinib showed efficacy in both models in absence of direct B cell depletion, broad T cell inhibition or reduction of total Ig levels. These findings support the view that remibrutinib may represent a novel treatment option for patients with MS.


Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , Humans , Animals , Mice , Multiple Sclerosis/drug therapy , Neuroinflammatory Diseases , Myeloid Cells , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Agammaglobulinaemia Tyrosine Kinase , Antigen-Antibody Complex , Anti-Inflammatory Agents
2.
Cell Rep Med ; 4(5): 101036, 2023 05 16.
Article En | MEDLINE | ID: mdl-37196635

Genetic and in vivo evidence suggests that aberrant recognition of RNA-containing autoantigens by Toll-like receptors (TLRs) 7 and 8 drives autoimmune diseases. Here we report on the preclinical characterization of MHV370, a selective oral TLR7/8 inhibitor. In vitro, MHV370 inhibits TLR7/8-dependent production of cytokines in human and mouse cells, notably interferon-α, a clinically validated driver of autoimmune diseases. Moreover, MHV370 abrogates B cell, plasmacytoid dendritic cell, monocyte, and neutrophil responses downstream of TLR7/8. In vivo, prophylactic or therapeutic administration of MHV370 blocks secretion of TLR7 responses, including cytokine secretion, B cell activation, and gene expression of, e.g., interferon-stimulated genes. In the NZB/W F1 mouse model of lupus, MHV370 halts disease. Unlike hydroxychloroquine, MHV370 potently blocks interferon responses triggered by specific immune complexes from systemic lupus erythematosus patient sera, suggesting differentiation from clinical standard of care. These data support advancement of MHV370 to an ongoing phase 2 clinical trial.


Autoimmune Diseases , Lupus Erythematosus, Systemic , Humans , Mice , Animals , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 7/therapeutic use , Lupus Erythematosus, Systemic/drug therapy , Lupus Erythematosus, Systemic/metabolism , Hydroxychloroquine/pharmacology , Hydroxychloroquine/therapeutic use , Interferons
3.
J Clin Invest ; 130(9): 4546-4560, 2020 09 01.
Article En | MEDLINE | ID: mdl-32484801

FTY720 is a treatment for relapsing remitting multiple sclerosis (MS). It is an analog of sphingosine-1-phosphate (S1P) and targets S1P receptors 1, 3, 4, and 5. Recent reports indicate an association between long-term exposure to FTY720 and cases of cryptococcal infection. Here, we studied the effect of FTY720 and its derivative, BAF312, which only target S1P receptors 1 and 5, in a mouse model of cryptococcal infection. We found that treatment with FTY720, but not with BAF312, led to decreased survival and increased organ burden in mouse cryptococcal granulomas. Both FTY720 and BAF312 caused a profound CD4+ and CD8+ T cell depletion in blood and lungs but only treatment with FTY720 led to cryptococcal reactivation. Treatment with FTY720, but not with BAF312, was associated with disorganization of macrophages and with M2 polarization at the granuloma site. In a cell system, FTY720 decreased phagocytosis and production of reactive oxygen species by macrophages, a phenotype recapitulated in the S1pr3-/- knockout macrophages. Our results suggest that FTY720 reactivates cryptococcosis from the granuloma through a S1P receptor 3-mediated mechanism and support the rationale for development of more-specific receptor modulators for therapeutic use of MS.


Cryptococcosis/drug therapy , Cryptococcus neoformans/metabolism , Fingolimod Hydrochloride/pharmacology , Granuloma/drug therapy , Macrophages, Peritoneal/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , Animals , Cell Line , Cryptococcosis/metabolism , Cryptococcosis/pathology , Female , Granuloma/metabolism , Granuloma/microbiology , Granuloma/pathology , Humans , Macrophages, Peritoneal/microbiology , Macrophages, Peritoneal/pathology , Male , Mice
4.
J Med Chem ; 63(10): 5102-5118, 2020 05 28.
Article En | MEDLINE | ID: mdl-32083858

Bruton's tyrosine kinase (BTK), a cytoplasmic tyrosine kinase, plays a central role in immunity and is considered an attractive target for treating autoimmune diseases. The use of currently marketed covalent BTK inhibitors is limited to oncology indications based on their suboptimal kinase selectivity. We describe the discovery and preclinical profile of LOU064 (remibrutinib, 25), a potent, highly selective covalent BTK inhibitor. LOU064 exhibits an exquisite kinase selectivity due to binding to an inactive conformation of BTK and has the potential for a best-in-class covalent BTK inhibitor for the treatment of autoimmune diseases. It demonstrates potent in vivo target occupancy with an EC90 of 1.6 mg/kg and dose-dependent efficacy in rat collagen-induced arthritis. LOU064 is currently being tested in phase 2 clinical studies for chronic spontaneous urticaria and Sjoegren's syndrome.


Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/metabolism , Drug Discovery/methods , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Agammaglobulinaemia Tyrosine Kinase/chemistry , Animals , Benzamides/chemistry , Benzamides/metabolism , Benzamides/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Crystallography, X-Ray/methods , Dogs , Dose-Response Relationship, Drug , Female , Humans , Mice , Protein Binding/physiology , Protein Kinase Inhibitors/chemistry , Protein Structure, Secondary , Protein Structure, Tertiary , Rats , Rats, Inbred Lew , Sheep
5.
JCI Insight ; 5(1)2020 01 16.
Article En | MEDLINE | ID: mdl-31821174

Subpial demyelination is a specific hallmark of multiple sclerosis and a correlate of disease progression. Although the mechanism(s) that mediate pathogenesis in the subpial compartment remain unclear, it has been speculated that inflammation in the overlying meninges may be associated with subpial injury. Here we show that adoptive transfer of proteolipid protein-primed Th17 cells into SJL/J recipient mice induces subpial demyelination associated with microglial/macrophage activation, disruption of the glial limitans, and evidence of an oxidative stress response. This pathology was topologically associated with foci of immune cells in the meninges and occurred in the absence of measurable anti-myelin oligodendrocyte glycoprotein IgM or IgG antibodies. To test the role of brain-infiltrating leukocytes on subpial injury, we modulated sphingosine 1-phosphate (S1P) receptor1,5 activity with BAF312 (siponimod) treatment. Administration of BAF312, even after adoptively transferred T cells had entered the brain, significantly ameliorated clinical experimental autoimmune encephalomyelitis and diminished subpial pathology, concomitant with a selective reduction in the capacity of transferred T cells to make Th17 cytokines. We conclude that sustained subpial cortical injury is associated with the capacity for brain-resident T cells to produce Th17 cytokines, and this pathological process occurs in an S1P receptor1,5-dependent manner.


Azetidines/therapeutic use , Benzyl Compounds/therapeutic use , Th17 Cells/drug effects , Adoptive Transfer , Animals , Brain/pathology , Cytidine Deaminase/genetics , Cytokines/immunology , Disease Progression , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Immunoglobulin G , Immunoglobulin M , Inflammation/pathology , Macrophages , Meninges , Mice , Mice, Inbred C57BL , Microglia/pathology , Multiple Sclerosis/immunology , Myelin-Oligodendrocyte Glycoprotein/immunology , Neuroglia , Th17 Cells/immunology
6.
J Immunol ; 194(8): 3723-34, 2015 Apr 15.
Article En | MEDLINE | ID: mdl-25762782

The paracaspase MALT1 plays an important role in immune receptor-driven signaling pathways leading to NF-κB activation. MALT1 promotes signaling by acting as a scaffold, recruiting downstream signaling proteins, as well as by proteolytic cleavage of multiple substrates. However, the relative contributions of these two different activities to T and B cell function are not well understood. To investigate how MALT1 proteolytic activity contributes to overall immune cell regulation, we generated MALT1 protease-deficient mice (Malt1(PD/PD)) and compared their phenotype with that of MALT1 knockout animals (Malt1(-/-)). Malt1(PD/PD) mice displayed defects in multiple cell types including marginal zone B cells, B1 B cells, IL-10-producing B cells, regulatory T cells, and mature T and B cells. In general, immune defects were more pronounced in Malt1(-/-) animals. Both mouse lines showed abrogated B cell responses upon immunization with T-dependent and T-independent Ags. In vitro, inactivation of MALT1 protease activity caused reduced stimulation-induced T cell proliferation, impaired IL-2 and TNF-α production, as well as defective Th17 differentiation. Consequently, Malt1(PD/PD) mice were protected in a Th17-dependent experimental autoimmune encephalomyelitis model. Surprisingly, Malt1(PD/PD) animals developed a multiorgan inflammatory pathology, characterized by Th1 and Th2/0 responses and enhanced IgG1 and IgE levels, which was delayed by wild-type regulatory T cell reconstitution. We therefore propose that the pathology characterizing Malt1(PD/PD) animals arises from an immune imbalance featuring pathogenic Th1- and Th2/0-skewed effector responses and reduced immunosuppressive compartments. These data uncover a previously unappreciated key function of MALT1 protease activity in immune homeostasis and underline its relevance in human health and disease.


B-Lymphocytes, Regulatory/immunology , Caspases/immunology , Cell Differentiation/immunology , Cell Proliferation , Encephalomyelitis, Autoimmune, Experimental/immunology , Neoplasm Proteins/immunology , T-Lymphocytes, Regulatory/immunology , Animals , B-Lymphocytes, Regulatory/pathology , Caspases/genetics , Cell Differentiation/genetics , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immunoglobulin E/genetics , Immunoglobulin E/immunology , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , Interleukin-10/genetics , Interleukin-10/immunology , Interleukin-2/genetics , Interleukin-2/immunology , Mice , Mice, Knockout , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/genetics , T-Lymphocytes, Regulatory/pathology , Th1 Cells/immunology , Th1 Cells/pathology , Th17 Cells/immunology , Th17 Cells/pathology
7.
PLoS One ; 8(3): e59630, 2013.
Article En | MEDLINE | ID: mdl-23544080

BACKGROUND: Sphingosine-1-phosphate (S1P) regulates the egress of T cells from lymphoid organs; levels of S1P in the tissues are controlled by S1P lyase (Sgpl1). Hence, Sgpl1 offers a target to block T cell-dependent inflammatory processes. However, the involvement of Sgpl1 in models of disease has not been fully elucidated yet, since Sgpl1 KO mice have a short life-span. METHODOLOGY: We generated inducible Sgpl1 KO mice featuring partial reduction of Sgpl1 activity and analyzed them with respect to sphingolipid levels, T-cell distribution, and response in models of inflammation. PRINCIPAL FINDINGS: The partially Sgpl1 deficient mice are viable but feature profound reduction of peripheral T cells, similar to the constitutive KO mice. While thymic T cell development in these mice appears normal, mature T cells are retained in thymus and lymph nodes, leading to reduced T cell numbers in spleen and blood, with a skewing towards increased proportions of memory T cells and T regulatory cells. The therapeutic relevance of Sgpl1 is demonstrated by the fact that the inducible KO mice are protected in experimental autoimmune encephalomyelitis (EAE). T cell immigration into the CNS was found to be profoundly reduced. Since S1P levels in the brain of the animals are unchanged, we conclude that protection in EAE is due to the peripheral effect on T cells, leading to reduced CNS immigration, rather than on local effects in the CNS. SIGNIFICANCE: The data suggest Sgpl1 as a novel therapeutic target for the treatment of multiple sclerosis.


Aldehyde-Lyases/deficiency , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/prevention & control , Aldehyde-Lyases/metabolism , Animals , Brain/metabolism , CD4-Positive T-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/complications , Forkhead Transcription Factors/metabolism , Hypersensitivity, Delayed/blood , Hypersensitivity, Delayed/complications , Hypersensitivity, Delayed/immunology , Hypersensitivity, Delayed/pathology , Immunologic Memory/immunology , Integrases/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Count , Mice , Mice, Knockout , Sheep , Sphingolipids/metabolism , Spleen/immunology , Spleen/pathology , Survival Analysis , Thymus Gland/immunology , Thymus Gland/pathology
8.
ACS Med Chem Lett ; 4(3): 333-7, 2013 Mar 14.
Article En | MEDLINE | ID: mdl-24900670

A novel series of alkoxyimino derivatives as S1P1 agonists were discovered through de novo design using FTY720 as the chemical starting point. Extensive structure-activity relationship studies led to the discovery of (E)-1-(4-(1-(((4-cyclohexyl-3-(trifluoromethyl)benzyl)oxy)imino)ethyl)-2-ethylbenzyl)azetidine-3-carboxylic acid (32, BAF312, Siponimod), which has recently completed phase 2 clinical trials in patients with relapsing-remitting multiple sclerosis.

9.
J Immunol ; 189(7): 3700-6, 2012 Oct 01.
Article En | MEDLINE | ID: mdl-22933630

Gradients of the sphingolipid sphingosine-1-phosphate (S1P) are responsible for the egress of lymphocytes from lymph nodes by activating the S1P1 receptor expressed on the surface of lymphocytes. Small molecule drugs that downregulate S1P receptors induce the sequestration of lymphocytes within lymph nodes, thus preventing lymphocytes from accessing sites of inflammation. In particular, FTY720, a pan-S1P receptor agonist, has been efficacious in the treatment of multiple sclerosis as well as its animal model, experimental autoimmune encephalomyelitis (EAE), by virtue of its ability to restrain lymphocytes within the lymph nodes, thus precluding their migration into the CNS. However, multiple leukocyte subsets express S1P receptors of varying types, and although it is beneficial to prevent transmigration of proinflammatory lymphocytes into the CNS, allowing access of regulatory leukocyte subsets to the CNS is desirable. In this study, we show that an S1P1-specific agonist (AUY954) is clinically efficacious in ameliorating pre-established EAE in SJL/J mice. Efficacy of AUY954 correlated with a reduction of lymphocytes in the CNS, but access of plasmacytoid dendritic cells (pDCs) to the CNS was unimpaired, and the presence of pDCs was found to be an important cofactor in mediating the clinical efficacy of AUY954. These results indicate that pDCs are important in quieting autoimmune responses during EAE, and that trafficking inhibitors that are permissive for pDC accumulation in the CNS may be of therapeutic value for the treatment of multiple sclerosis.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Dendritic Cells/drug effects , Dendritic Cells/pathology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/metabolism , Thiophenes/pharmacology , beta-Alanine/analogs & derivatives , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Aggregation/drug effects , Cell Aggregation/immunology , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Female , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Mice , Receptors, Lysosphingolipid/physiology , Sphingosine-1-Phosphate Receptors , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Thiophenes/therapeutic use , beta-Alanine/pharmacology , beta-Alanine/therapeutic use
10.
Bioorg Med Chem Lett ; 20(1): 35-7, 2010 Jan 01.
Article En | MEDLINE | ID: mdl-19954976

High throughput screening and hit to lead optimization led to the identification of 'carene' as a promising scaffold showing selective S1P(1) receptor agonism. In parallel to this work we have established a pharmacophore model for the S1P(1) receptor highlighting the minimal structural requirement necessary for potent receptor agonism.


Monoterpenes/chemistry , Pyrazoles/chemistry , Receptors, Lysosphingolipid/agonists , Bicyclic Monoterpenes , High-Throughput Screening Assays , Hydrogen Bonding , Pyrazoles/chemical synthesis , Pyrazoles/pharmacology , Receptors, Lysosphingolipid/metabolism , Stereoisomerism , Structure-Activity Relationship
11.
J Neuroimmunol ; 216(1-2): 59-65, 2009 Nov 30.
Article En | MEDLINE | ID: mdl-19804913

Experimental autoimmune neuritis (EAN) is a T cell-mediated autoimmune inflammatory demyelinating disease of the peripheral nervous system and an animal model of human inflammatory demyelinating polyradiculoneuropathy. AUY954, which targets selectively the sphingosine-1-phosphate receptor 1 (S1P(1)), is known to sequester lymphocytes into secondary lymphoid tissues. In EAN rats, AUY954 greatly prevented paraparesis if administrated from the day of immunization. T cell, B cell, and macrophage infiltration, inflammatory demyelination, and local expression of interleukine-17 and matrix metalloproteinase-9 in sciatic nerves of EAN rats were significantly decreased by AUY954 treatment. Therefore, S1P(1) modulation might be a potential treatment option for inflammatory neuropathies.


Guillain-Barre Syndrome/drug therapy , Neuritis, Autoimmune, Experimental/drug therapy , Receptors, Lysosphingolipid/drug effects , Thiophenes/pharmacology , beta-Alanine/analogs & derivatives , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Chemotaxis, Leukocyte/drug effects , Chemotaxis, Leukocyte/immunology , Disease Models, Animal , Guillain-Barre Syndrome/immunology , Guillain-Barre Syndrome/metabolism , Immunologic Factors/pharmacology , Immunologic Factors/therapeutic use , Interleukin-17/metabolism , Macrophages/drug effects , Macrophages/immunology , Male , Matrix Metalloproteinase 9/metabolism , Neuritis, Autoimmune, Experimental/immunology , Neuritis, Autoimmune, Experimental/metabolism , Paraparesis/drug therapy , Paraparesis/immunology , Paraparesis/prevention & control , Rats , Rats, Inbred Lew , Receptors, Lysosphingolipid/metabolism , Sciatic Nerve/drug effects , Sciatic Nerve/immunology , Sciatic Nerve/metabolism , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Thiophenes/therapeutic use , Treatment Outcome , beta-Alanine/pharmacology , beta-Alanine/therapeutic use
12.
Int Immunol ; 20(5): 633-44, 2008 May.
Article En | MEDLINE | ID: mdl-18343890

The immunomodulator FTY720 inhibits lymph node (LN) and thymic egress, thereby constraining T cell circulation and reducing peripheral T cell numbers. Here, we analyzed in mouse models the as yet scarcely characterized impact of long-term (up to 6 months) FTY720 exposure on T cell homeostasis and possible consequences for alloreactivity. In green fluorescent protein (GFP) hemopoietic chimeras, the turnover of (initially GFP(-)) peripheral T cell pools was markedly delayed under FTY720, while normal homeostatic differences between CD4 and CD8 T cell sub-populations were retained or amplified further. Homeostatic proliferation was enhanced, and within shrinking T cell pools, the proportions of effector memory phenotype CD4 T cells (CD4T(PEM)) increased in spleens and LNs and of central memory phenotype CD8 T cells (CD8T(PCM)) in LNs. By contrast, the fractions of CD8T(PEM) and CD4T(PCM) remained stably small under FTY720. The enrichment for CD4T(PEM) and CD8T(PCM) correlated with larger proportions of IFNgamma-producing T cells upon nonspecific but not allospecific stimulation. Splenic CD4 T cells from FTY720-treated mice proliferated more strongly upon transfer to semi-allogeneic hosts. However, heart allograft survival was not compromised in FTY720 pre-treated recipients. It correlated with reduced intra-graft CD8 T cells, and the longest surviving transplants contained the highest numbers of CD4 T cells. Thus, continuous FTY720 exposure reveals differential homeostatic responses by memory phenotype CD4 and CD8 T cell sub-populations, and it may enhance alloreactive CD4 T cell proliferation and tissue infiltration without accelerating allograft rejection.


CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Graft Survival , Heart Transplantation/immunology , Immunosuppressive Agents/pharmacology , Propylene Glycols/pharmacology , Sphingosine/analogs & derivatives , Animals , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Female , Fingolimod Hydrochloride , Graft Rejection , Homeostasis/drug effects , Immunologic Memory , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Sphingosine/pharmacology , Spleen/cytology , Spleen/immunology
13.
Transpl Int ; 20(4): 353-64, 2007 Apr.
Article En | MEDLINE | ID: mdl-17326776

Vav proteins mediate T- and B-cell activation by functioning as GTP/GDP exchange factors for small GTPases. We have studied the role of Vav1 and Vav2 in allogeneic T-cell activation, antibody responses and allograft rejection. Alloantigen-induced proliferation of T cells from Vav1- and Vav1/Vav2-knockout (ko) mice was decreased by >90% in a mixed lymphocyte reaction. In whole-blood cultures, Vav deficiency led to markedly impaired T- and B-cell activation. Expansion of Vav1- or Vav1/Vav2-ko T cells (C57BL/6) was reduced after transfer into severe combined immune deficiency/beige recipient mice (BALB/c). After priming with 2,4-dinitrophenyl (DNP)-keyhole limpet hemocyanin, T-cell-dependent anti-DNP IgM and IgG antibody levels were normal in Vav1-ko mice but undetectable in Vav1/Vav2-ko mice. The median survival time of BALB/c cardiac allografts transplanted into C57BL/6 Vav1-ko mice (n = 13) or Vav1/Vav2-ko mice (n = 5) was >100 and >77 days, compared with 8-9 days in the corresponding wild-type mice. Vav1/Vav2-ko mice with <100 days graft survival developed bacterial skin infections and were prematurely killed with beating cardiac allograft. Long-term surviving transplants of single and double ko mice showed mild cellular interstitial rejection and mild to severe vascular remodeling. In conclusion, our studies show for the first time that the absence of Vav1 and Vav1/Vav2 in ko mice strongly reduces alloreactivity and results in long-term allograft survival, whereas antibody responses were only affected in Vav1/Vav2 ko mice.


Graft Survival , Heart Transplantation/immunology , Proto-Oncogene Proteins c-vav/physiology , Animals , Dinitrophenols/immunology , Female , Graft Survival/immunology , Heart Transplantation/pathology , Hemocyanins/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Proto-Oncogene Proteins c-vav/genetics , T-Lymphocytes/immunology , Transplantation, Homologous/immunology
14.
Chem Biol ; 13(11): 1227-34, 2006 Nov.
Article En | MEDLINE | ID: mdl-17114004

FTY720 is an immunomodulator with demonstrated efficacy in a phase II trial of relapsing multiple sclerosis. FTY720-phosphate, the active metabolite generated upon phosphorylation in vivo, acts as a potent agonist on four of the five known sphingosine-1-phosphate (S1P(1)) receptors. AUY954, an aminocarboxylate analog of FTY720, is a low nanomolar, monoselective agonist of the S1P(1) receptor. Due to its selectivity and pharmacokinetic profile, AUY954 is an excellent pharmacological probe of S1P(1)-dependent phenomena. Oral administration of AUY954 induces a profound and reversible reduction of circulating lymphocytes and, in combination with RAD001 (Certican/Everolimus, an mTOR inhibitor), is capable of prolonging the survival of cardiac allografts in a stringent rat transplantation model. This demonstrates that a selective agonist of the S1P(1) receptor is sufficient to achieve efficacy in an animal model of transplantation.


Graft Rejection/prevention & control , Heart Transplantation , Receptors, Lysosphingolipid/agonists , Thiophenes/pharmacology , beta-Alanine/analogs & derivatives , Animals , CHO Cells , Cricetinae , Cricetulus , Everolimus , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Immunosuppressive Agents/pharmacology , Lymphocytes/drug effects , Male , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Inbred Lew , Sirolimus/analogs & derivatives , Sirolimus/pharmacology , Thiophenes/chemical synthesis , Thiophenes/pharmacokinetics , Transplantation, Homologous , beta-Alanine/chemical synthesis , beta-Alanine/pharmacokinetics , beta-Alanine/pharmacology
15.
Bioorg Med Chem Lett ; 14(10): 2451-7, 2004 May 17.
Article En | MEDLINE | ID: mdl-15109631

Combination of structural elements from a potent Y5 antagonist (2) with thiazole fragments that exhibit weak Y5 affinities followed by lead optimisation led to the discovery of (5,6-dihydro-4H-3-thia-1-aza-benzo[e]azulen-2-yl)-piperidin-4-ylmethyl-amino and (4,5-dihydro-6-oxa-3-thia-1-aza-benzo[e]azulen-2-yl)-piperidin-4-ylmethyl-amino derivatives. Both classes of compounds are capable of delivering potent and selective orally and centrally bioavailable NPY Y5 receptor antagonists.


Cycloheptanes/chemical synthesis , Cycloheptanes/pharmacokinetics , Receptors, Neuropeptide Y/antagonists & inhibitors , Administration, Oral , Animals , Azulenes , Biological Availability , Blood , Blood-Brain Barrier , Cycloheptanes/pharmacology , Heterocyclic Compounds, 3-Ring/chemical synthesis , Heterocyclic Compounds, 3-Ring/pharmacokinetics , Heterocyclic Compounds, 3-Ring/pharmacology , Heterocyclic Compounds, 4 or More Rings/chemical synthesis , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/pharmacology , Hypothalamus , Inhibitory Concentration 50 , Rats , Structure-Activity Relationship
16.
Am J Physiol Endocrinol Metab ; 283(3): E604-11, 2002 Sep.
Article En | MEDLINE | ID: mdl-12169455

Suckling-age rats display endogenous circadian rhythmicity of metabolic rate (MR) with energy-saving, torpor-like decreases, which are sympathetically controlled and suppressed by leptin treatment. We investigated whether neonatal monosodium glutamate (MSG) treatment, known to cause arcuate nucleus damage and adult-age obesity, alters energy balance in the first two postnatal weeks. Continuously recorded MR and core temperatures (T(c)) show that MSG treatment disinhibits the periodic, sympathetically controlled, energy-saving drops of T(c) and MR. Increased energy expenditure thus explains reduced body fat at normal lean body mass found in MSG-treated pups artificially nourished identically to controls. In MSG-treated mother-reared pups, lean body mass is additionally reduced, suggesting that MSG also reduces suckling. Plasma leptin levels are similar in controls and MSG-treated pups but higher per unit of fat mass in the latter. We conclude that the postweaning development of MSG obesity and depressed thermogenesis are preceded by an early phase of increased energy expenditure with decreased fat deposition during suckling age and hypothesize cell damage in the arcuate nucleus to be involved in both.


Animals, Suckling/physiology , Body Weight/drug effects , Circadian Rhythm , Energy Metabolism/physiology , Sodium Glutamate/pharmacology , Animal Husbandry/methods , Animals , Animals, Suckling/growth & development , Body Composition/drug effects , Body Temperature/drug effects , Female , Hypothalamus/drug effects , Hypothalamus/pathology , Leptin/blood , Metabolism/drug effects , Mothers , Periodicity , Rats , Rats, Zucker
...