Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Stuart M. Cahalan is active.

Publication


Featured researches published by Stuart M. Cahalan.


Science | 2012

Crystal structure of a lipid G protein-coupled receptor.

Michael A. Hanson; Christopher B. Roth; Euijung Jo; Mark T. Griffith; Fiona Scott; G Reinhart; H Desale; B Clemons; Stuart M. Cahalan; S.C Schuerer; Mg Sanna; Gye Won Han; Peter Kuhn; Hugh Rosen; Raymond C. Stevens

A Lipid-Sensing GPCR Sphingosine 1-phosphate (S1P) is a sphingolipid that binds to the G protein–coupled receptor subtype 1 (S1P1) to activate signaling pathways involved in regulation of the vascular and immune systems. Hanson et al. (p. 851) determined the crystal structure of S1PR in complex with an antagonist sphingolipid mimic. Ligand access to the receptor from the extracellular milieu is occluded, and a gap between helices I and VII may provide ligand access from within the membrane. The structural information, together with mutagenesis and structure activity relationship data, provides insight into the molecular recognition events that modulate signaling. A channel in a lipid-dependent G protein–coupled receptor allows a ligand to access its binding site from within the plasma membrane. The lyso-phospholipid sphingosine 1-phosphate modulates lymphocyte trafficking, endothelial development and integrity, heart rate, and vascular tone and maturation by activating G protein–coupled sphingosine 1-phosphate receptors. Here, we present the crystal structure of the sphingosine 1-phosphate receptor 1 fused to T4-lysozyme (S1P1-T4L) in complex with an antagonist sphingolipid mimic. Extracellular access to the binding pocket is occluded by the amino terminus and extracellular loops of the receptor. Access is gained by ligands entering laterally between helices I and VII within the transmembrane region of the receptor. This structure, along with mutagenesis, agonist structure-activity relationship data, and modeling, provides a detailed view of the molecular recognition and requirement for hydrophobic volume that activates S1P1, resulting in the modulation of immune and stromal cell responses.


Cell | 2011

Endothelial cells are central orchestrators of cytokine amplification during influenza virus infection

John R. Teijaro; Kevin B. Walsh; Stuart M. Cahalan; Daniel M. Fremgen; Edward Roberts; Fiona Scott; Esther Martinborough; Robert Peach; Michael B. A. Oldstone; Hugh Rosen

Summary Cytokine storm during viral infection is a prospective predictor of morbidity and mortality, yet the cellular sources remain undefined. Here, using genetic and chemical tools to probe functions of the S1P1 receptor, we elucidate cellular and signaling mechanisms that are important in initiating cytokine storm. Whereas S1P1 receptor is expressed on endothelial cells and lymphocytes within lung tissue, S1P1 agonism suppresses cytokines and innate immune cell recruitment in wild-type and lymphocyte-deficient mice, identifying endothelial cells as central regulators of cytokine storm. Furthermore, our data reveal immune cell infiltration and cytokine production as distinct events that are both orchestrated by endothelial cells. Moreover, we demonstrate that suppression of early innate immune responses through S1P1 signaling results in reduced mortality during infection with a human pathogenic strain of influenza virus. Modulation of endothelium with a specific agonist suggests that diseases in which amplification of cytokine storm is a significant pathological component could be chemically tractable.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Piezo1, a mechanically activated ion channel, is required for vascular development in mice

Sanjeev S. Ranade; Zhaozhu Qiu; Seung Hyun Woo; Sung Sik Hur; Swetha E. Murthy; Stuart M. Cahalan; Jie Xu; Jayanti Mathur; Michael Bandell; Bertrand Coste; Yi Shuan J Li; Shu Chien; Ardem Patapoutian

Significance Ion channels that are activated by mechanical force have been implicated in numerous physiological systems. In mammals, the identity of these channels remains poorly understood. We recently described Piezos as evolutionarily conserved mechanically activated ion channels and showed that Piezo2 is required for activation of touch receptors in the skin. Here we show that Piezo1 is a critical component of endothelial cell mechanotransduction and is required for embryonic development. Piezo1 is expressed in embryonic endothelial cells and is activated by fluid shear stress. Loss of Piezo1 affects the ability of endothelial cells to alter their alignment when subjected to shear stress. These results suggest a potential role for Piezo1 in mechanotransduction in adult cardiovascular function and disease. Mechanosensation is perhaps the last sensory modality not understood at the molecular level. Ion channels that sense mechanical force are postulated to play critical roles in a variety of biological processes including sensing touch/pain (somatosensation), sound (hearing), and shear stress (cardiovascular physiology); however, the identity of these ion channels has remained elusive. We previously identified Piezo1 and Piezo2 as mechanically activated cation channels that are expressed in many mechanosensitive cell types. Here, we show that Piezo1 is expressed in endothelial cells of developing blood vessels in mice. Piezo1-deficient embryos die at midgestation with defects in vascular remodeling, a process critically influenced by blood flow. We demonstrate that Piezo1 is activated by shear stress, the major type of mechanical force experienced by endothelial cells in response to blood flow. Furthermore, loss of Piezo1 in endothelial cells leads to deficits in stress fiber and cellular orientation in response to shear stress, linking Piezo1 mechanotransduction to regulation of cell morphology. These findings highlight an essential role of mammalian Piezo1 in vascular development during embryonic development.


Nature Communications | 2013

Dehydrated hereditary stomatocytosis linked to gain-of-function mutations in mechanically activated PIEZO1 ion channels.

Juliette Albuisson; Swetha E. Murthy; Michael Bandell; Bertrand Coste; Hélène Louis-Dit-Picard; Jayanti Mathur; Madeleine Fénéant-Thibault; Gérard Tertian; Jean-Pierre de Jaureguiberry; Pierre-Yves Syfuss; Stuart M. Cahalan; Loïc Garçon; Fabienne Toutain; Pierre Simon Rohrlich; Jean Delaunay; Véronique Picard; Xavier Jeunemaitre; Ardem Patapoutian

Dehydrated hereditary stomatocytosis (DHS) is a genetic condition with defective red blood cell (RBC) membrane properties that causes an imbalance in intracellular cation concentrations. Recently, two missense mutations inthe mechanically activated PIEZO1(FAM38A) ion channel were associated with DHS. However, it is not known how these mutations affect PIEZO1 function. Here, by combining linkage analysis and whole-exome sequencing in a large pedigree and Sanger sequencing in two additional kindreds and 11 unrelated DHS cases, we identifythree novel missense mutations and one recurrent duplication in PIEZO1, demonstrating that it is the major gene for DHS. All the DHS-associated mutations locate at C-terminal half of PIEZO1. Remarkably, we find that all PIEZO1 mutations give rise to mechanically activated currents that inactivate more slowly than wild-type currents. This gain-of-function PIEZO1 phenotype provides insight that helps to explain the increased permeability of cations in RBCs of DHS patients. Our findings also suggest a new role for mechanotransduction in RBC biology and pathophysiology.


Immunological Reviews | 2008

Modulating tone: the overture of S1P receptor immunotherapeutics

Hugh Rosen; Pedro J. Gonzalez-Cabrera; David Marsolais; Stuart M. Cahalan; Anthony S. Don; M. Germana Sanna

Summary: Modulation of complex functions within the immune system has proven to be surprisingly sensitive to alterations in the lysophospholipid sphingosine 1‐phosphate (S1P) receptor‐ligand rheostat. This has become increasingly evident from both chemical and genetic manipulation of the S1P system, with pharmacological effects upon lymphoid cells, dendritic cell function, as well as vascular interfaces. The integrated immune system, perhaps as a result of its relatively recent evolutionary ontogeny, has selected for a number of critical control points regulated by five distinct high affinity G‐protein‐coupled receptor subtypes with a shared ligand, with receptors distributed on lymphocytes, dendritic cells, and endothelium. All of these cellular components of the axis are capable of modulating immune responses in vivo, with the impact on the immune response being very different from classical immunosuppressants, by virtue of selective spatial and temporal sparing of humoral and myeloid elements of host defense. Pharmacological subversion of the S1P rheostat is proving to be clinically efficacious in multiple sclerosis, and both the scope and limitations of therapeutic modulation of the S1P axis in immunotherapy are becoming clearer as understanding of the integrated chemical physiology of the S1P system emerges.


Cell | 2016

LRRC8 Proteins Form Volume-Regulated Anion Channels that Sense Ionic Strength

Ruhma Syeda; Zhaozhu Qiu; Adrienne E. Dubin; Swetha E. Murthy; Maria N. Florendo; Daniel E. Mason; Jayanti Mathur; Stuart M. Cahalan; Eric C. Peters; Mauricio Montal; Ardem Patapoutian

The volume-regulated anion channel (VRAC) is activated when a cell swells, and it plays a central role in maintaining cell volume in response to osmotic challenges. SWELL1 (LRRC8A) was recently identified as an essential component of VRAC. However, the identity of the pore-forming subunits of VRAC and how the channel is gated by cell swelling are unknown. Here, we show that SWELL1 and up to four other LRRC8 subunits assemble into heterogeneous complexes of ∼800 kDa. When reconstituted into bilayers, LRRC8 complexes are sufficient to form anion channels activated by osmolality gradients. In bilayers, as well as in cells, the single-channel conductance of the complexes depends on the LRRC8 composition. Finally, low ionic strength (Γ) in the absence of an osmotic gradient activates the complexes in bilayers. These data demonstrate that LRRC8 proteins together constitute the VRAC pore and that hypotonic stress can activate VRAC through a decrease in cytoplasmic Γ.


Molecular Pharmacology | 2012

S1P1 Receptor Modulation with Cyclical Recovery from Lymphopenia Ameliorates Mouse Model of Multiple Sclerosis

Pedro J. Gonzalez-Cabrera; Stuart M. Cahalan; Nhan Nguyen; Gor Sarkisyan; Nora Leaf; Michael D. Cameron; Tomoyuki Kago; Hugh Rosen

Multiple sclerosis (MS) therapies modulate T-cell autoimmunity in the central nervous system (CNS) but may exacerbate latent infections. Fingolimod, a nonselective sphingosine-1-phosphate (S1P) receptor agonist that induces sustained lymphopenia and accumulates in the CNS, represents a new treatment modality for MS. We hypothesized that sustained lymphopenia would not be required for efficacy and that a selective, CNS-penetrant, peripherally short-acting, S1P1 agonist would show full efficacy in a mouse MS model. Using daily treatment with 10 mg/kg 2-(4-(5-(3,4-diethoxyphenyl)-1,2,4-oxadiazol-3-yl)-2,3-dihydro-1H-inden-1-yl amino)ethanol (CYM-5442) at the onset of clinical signs in myelin oligodendrocyte glycoprotein MOG35–55- induced experimental allergic encephalomyelitis (EAE), we assessed clinical scores, CNS cellular infiltration, demyelination, and gliosis for 12 days with CYM-5442, vehicle, or fingolimod. CYM-5442 levels in CNS and plasma were determined at experiment termination, and blood lymphopenia was measured 3 and 24 h after the last injection. Plasma levels of cytokines were assayed at the end of the protocol. Changes in S1P1-enhanced green fluorescent protein expression on neurons and astrocytes during active EAE and upon CYM-5442 treatment were quantified with flow cytometry and Western blotting by using native-locus enhanced green fluorescent protein-tagged S1P1 mice. S1P1 agonism alone reduced pathological features as did fingolimod (maximally lymphopenic throughout), despite full reversal of lymphopenia within each dosing interval. CYM-5442 levels in CNS but not in plasma were sustained. Neuronal and astrocytic S1P1 expression in EAE was suppressed by CYM-5442 treatment, relative to vehicle, and levels of key cytokines, such as interleukin 17A, were also significantly reduced in drug-treated mice. S1P1-selective agonists that induce reversible lymphopenia while persisting in the CNS may be effective MS treatments.


Nature Chemical Biology | 2011

Actions of a picomolar short-acting S1P1 agonist in S1P1-eGFP knock-in mice

Stuart M. Cahalan; Pedro J. Gonzalez-Cabrera; Gor Sarkisyan; Nhan Nguyen; Marie Therese Schaeffer; Liming Huang; Adam Richard Yeager; Bryan Clemons; Fiona Scott; Hugh Rosen

Sphingosine 1-Phosphate Receptor 1 (S1P1) plays a critical role in lymphocyte recirculation and is a clinical target for treatment of multiple sclerosis. By generating a short-duration S1P1 agonist and mice where fluorescently tagged S1P1 replaces wild-type receptor, we elucidate physiological and agonist-perturbed changes in expression of S1P1 at a subcellular level in vivo. We demonstrate differential downregulation of S1P1 on lymphocytes and endothelia following agonist treatment.


Molecular Pharmacology | 2013

Sphingosine 1-Phosphate Receptor 1 (S1P1) Upregulation and Amelioration of Experimental Autoimmune Encephalomyelitis by an S1P1 Antagonist

Stuart M. Cahalan; Pedro J. Gonzalez-Cabrera; Nhan Nguyen; Miguel Guerrero; Elizabeth A. George Cisar; Nora Leaf; Steven J. Brown; Edward Roberts; Hugh Rosen

Sphingosine 1-phosphate receptor 1 (S1P1) is a G protein–coupled receptor that is critical for proper lymphocyte development and recirculation. Agonists to S1P1 are currently in use clinically for the treatment of multiple sclerosis, and these drugs may act on both S1P1 expressed on lymphocytes and S1P1 expressed within the central nervous system. Agonists to S1P1 and deficiency in S1P1 both cause lymphocyte sequestration in the lymph nodes. In the present study, we show that S1P1 antagonism induces lymphocyte sequestration in the lymph nodes similar to that observed with S1P1 agonists while upregulating S1P1 on lymphocytes and endothelial cells. Additionally, we show that S1P1 antagonism reverses experimental autoimmune encephalomyelitis in mice without acting on S1P1 expressed within the central nervous system, demonstrating that lymphocyte sequestration via S1P1 antagonism is sufficient to alleviate autoimmune pathology.


Molecular Pharmacology | 2015

Bitopic Sphingosine 1-Phosphate Receptor 3 (S1P3) Antagonist Rescue from Complete Heart Block: Pharmacological and Genetic Evidence for Direct S1P3 Regulation of Mouse Cardiac Conduction.

Mg Sanna; Kevin P. Vincent; E Repetto; Nhan Nguyen; Steven J. Brown; L Abgaryan; Sean Riley; Nora Leaf; Stuart M. Cahalan; William B. Kiosses; Y Kohno; Joan Heller Brown; Andrew D. McCulloch; Hugh Rosen; Pedro J. Gonzalez-Cabrera

The molecular pharmacology of the G protein–coupled receptors for sphingosine 1-phosphate (S1P) provides important insight into established and new therapeutic targets. A new, potent bitopic S1P3 antagonist, SPM-354, with in vivo activity, has been used, together with S1P3-knockin and S1P3-knockout mice to define the spatial and functional properties of S1P3 in regulating cardiac conduction. We show that S1P3 is a key direct regulator of cardiac rhythm both in vivo and in isolated perfused hearts. 2-Amino-2-[2-(4-octylphenyl)ethyl]propane-1,3-diol in vivo and S1P in isolated hearts induced a spectrum of cardiac effects, ranging from sinus bradycardia to complete heart block, as measured by a surface electrocardiogram in anesthetized mice and in volume-conducted Langendorff preparations. The agonist effects on complete heart block are absent in S1P3-knockout mice and are reversed in wild-type mice with SPM-354, as characterized and described here. Homologous knockin of S1P3-mCherry is fully functional pharmacologically and is strongly expressed by immunohistochemistry confocal microscopy in Hyperpolarization Activated Cyclic Nucleotide Gated Potassium Channel 4 (HCN4)-positive atrioventricular node and His-Purkinje fibers, with relative less expression in the HCN4-positive sinoatrial node. In Langendorff studies, at constant pressure, SPM-354 restored sinus rhythm in S1P-induced complete heart block and fully reversed S1P-mediated bradycardia. S1P3 distribution and function in the mouse ventricular cardiac conduction system suggest a direct mechanism for heart block risk that should be further studied in humans. A richer understanding of receptor and ligand usage in the pacemaker cells of the cardiac system is likely to be useful in understanding ventricular conduction in health, disease, and pharmacology.

Collaboration


Dive into the Stuart M. Cahalan's collaboration.

Top Co-Authors

Avatar

Hugh Rosen

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Ardem Patapoutian

Howard Hughes Medical Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jayanti Mathur

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Michael Bandell

Genomics Institute of the Novartis Research Foundation

View shared research outputs
Top Co-Authors

Avatar

Adrienne E. Dubin

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Nhan Nguyen

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Nora Leaf

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Bertrand Coste

Aix-Marseille University

View shared research outputs
Researchain Logo
Decentralizing Knowledge