Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
2.
Article in English | MEDLINE | ID: mdl-30249602

ABSTRACT

The α7 nicotinic acetylcholine receptor (α7nAChR) is central to the anti-inflammatory function of the vagus nerve in a physiological mechanism termed the inflammatory reflex. Studies on the inflammatory reflex have been instrumental for the current development of the field of bioelectronic medicine. An independent investigation of the biological role of αB-crystallin (HspB5), the most abundant gene transcript present in active multiple sclerosis lesions in human brains, also led to α7nAChR. Induction of experimental autoimmune encephalomyelitis (EAE) in HspB5-/- mice results in greater paralytic signs, increased levels of proinflammatory cytokines, and T-lymphocyte activation relative to wild-type animals. Administration of HspB5 was therapeutic in animal models of multiple sclerosis, retinal and cardiac ischemia, and stroke. Structure-activity studies established that residues 73-92 were as potent as the parent protein, but only when it formed amyloid fibrils. Amyloid fibrils and small heat shock proteins (sHsps) selectively bound α7nAChR on peritoneal macrophages (MΦs) and B lymphocytes, converting the MΦs to an immune suppressive phenotype and mobilizing the migration of both cell types from the peritoneum to secondary lymph organs. Here, we review multiple aspects of this work, which may be of interest for developing future therapeutic approaches for multiple sclerosis and other disorders.


Subject(s)
Amyloid/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Heat-Shock Proteins, Small/immunology , Macrophages, Peritoneal/immunology , Nicotine/immunology , alpha7 Nicotinic Acetylcholine Receptor/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cytokines/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immune Tolerance , Lymphocyte Activation , Macrophages, Peritoneal/pathology , Mice , Mice, Knockout
3.
Proc Natl Acad Sci U S A ; 115(27): 7081-7086, 2018 07 03.
Article in English | MEDLINE | ID: mdl-29915045

ABSTRACT

Although certain dogma portrays amyloid fibrils as drivers of neurodegenerative disease and neuroinflammation, we have found, paradoxically, that amyloid fibrils and small heat shock proteins (sHsps) are therapeutic in experimental autoimmune encephalomyelitis (EAE). They reduce clinical paralysis and induce immunosuppressive pathways, diminishing inflammation. A key question was the identification of the target for these molecules. When sHsps and amyloid fibrils were chemically cross-linked to immune cells, a limited number of proteins were precipitated, including the α7 nicotinic acetylcholine receptor (α7 NAChR). The α7 NAChR is noteworthy among the over 20 known receptors for amyloid fibrils, because it plays a central role in a well-defined immune-suppressive pathway. Competitive binding between amyloid fibrils and α-bungarotoxin to peritoneal macrophages (MΦs) confirmed the involvement of α7 NAChR. The mechanism of immune suppression was explored, and, similar to nicotine, amyloid fibrils inhibited LPS induction of a common set of inflammatory cytokines while inducing Stat3 signaling and autophagy. Consistent with this, previous studies have established that nicotine, sHsps, and amyloid fibrils all were effective therapeutics in EAE. Interestingly, B lymphocytes were needed for the therapeutic effect. These results suggest that agonists of α7 NAChR might have therapeutic benefit for a variety of inflammatory diseases.


Subject(s)
Amyloid/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Heat-Shock Proteins/immunology , Macrophages, Peritoneal/immunology , Nicotine/immunology , alpha7 Nicotinic Acetylcholine Receptor/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Immune Tolerance , Macrophages, Peritoneal/pathology , Mice
4.
J Neuroimmunol ; 298: 42-50, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27609274

ABSTRACT

Amyloid hexapeptide molecules are effective in the treatment of the murine model of neuroinflammation, known as experimental autoimmune encephalomyelitis (EAE). Efficacy however differs between two inbred mouse strains, C57BL/6J (B6) and C57BL/10SnJ (B10). Amyloid hexapeptide treatments improved the clinical outcomes of B6, but not B10 mice, indicating that genetic background influences therapeutic efficacy. Moreover, although previous studies indicated that prion protein deficiency results in more severe EAE in B6 mice, we observed no such effect in B10 mice. In addition, we found that amyloid hexapeptide treatments of B10 and B6 mice elicited differential IL4 responses. Thus, the modulatory potential of prion protein and related treatments with other amyloid hexapeptides in EAE depends on mouse strain.


Subject(s)
Amyloid beta-Peptides/therapeutic use , Anti-Inflammatory Agents/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/genetics , Genetic Background , Peptide Fragments/therapeutic use , Prion Proteins/genetics , Amyloid beta-Peptides/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/etiology , Encephalomyelitis, Autoimmune, Experimental/pathology , Eosinophils/pathology , Eosinophils/ultrastructure , Female , Gene Expression Regulation/immunology , Interleukin-4/metabolism , Lymphocytes/metabolism , Lymphocytes/pathology , Lymphocytes/ultrastructure , Mice , Mice, Inbred Strains , Mice, Transgenic , Monocytes/pathology , Monocytes/ultrastructure , Myelin-Oligodendrocyte Glycoprotein/toxicity , Peptide Fragments/toxicity , Prion Proteins/metabolism , Species Specificity , tau Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 112(49): 15016-23, 2015 Dec 08.
Article in English | MEDLINE | ID: mdl-26621719

ABSTRACT

Amyloid fibrils composed of peptides as short as six amino acids are therapeutic in experimental autoimmune encephalomyelitis (EAE), reducing paralysis and inflammation, while inducing several pathways of immune suppression. Intraperitoneal injection of fibrils selectively activates B-1a lymphocytes and two populations of resident macrophages (MΦs), increasing IL-10 production, and triggering their exodus from the peritoneum. The importance of IL-10-producing B-1a cells in this effective therapy was established in loss-of-function experiments where neither B-cell-deficient (µMT) nor IL10(-/-) mice with EAE responded to the fibrils. In gain-of-function experiments, B-1a cells, adoptively transferred to µMT mice with EAE, restored their therapeutic efficacy when Amylin 28-33 was administered. Stimulation of adoptively transferred bioluminescent MΦs and B-1a cells by amyloid fibrils resulted in rapid (within 60 min of injection) trafficking of both cell types to draining lymph nodes. Analysis of gene expression indicated that the fibrils activated the CD40/B-cell receptor pathway in B-1a cells and induced a set of immune-suppressive cell-surface proteins, including BTLA, IRF4, and Siglec G. Collectively, these data indicate that the fibrils activate B-1a cells and F4/80(+) MΦs, resulting in their migration to the lymph nodes, where IL-10 and cell-surface receptors associated with immune-suppression limit antigen presentation and T-cell activation. These mechanisms culminate in reduction of paralytic signs of EAE.


Subject(s)
Amyloid/pharmacology , B-Lymphocytes/immunology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Lymphocyte Activation/drug effects , Adoptive Transfer , Amyloid/metabolism , Amyloid/therapeutic use , Animals , Encephalomyelitis, Autoimmune, Experimental/immunology , Endocytosis , Female , Interleukin-10/physiology , Islet Amyloid Polypeptide/pharmacology , Mice , Mice, Inbred C57BL
6.
Curr Top Behav Neurosci ; 26: 221-32, 2015.
Article in English | MEDLINE | ID: mdl-25981913

ABSTRACT

Amyloidogenic proteins have long been linked to neurodegenerative diseases. However, amyloid fibrils composed of six amino acids are protective in an animal model of multiple sclerosis (MS), experimental autoimmune encephalomyelitis (EAE). The reduction of pro-inflammatory cytokines, decrease in the number of inflammatory foci in the parenchyma and meninges of the brain and spinal cord, and amelioration of the neurological signs of EAE when amyloid fibril-forming hexapeptides are administered reveal that some fibrils provide benefit. The therapeutic activity of the amyloid fibrils arise from diverse pathways that include binding of pro-inflammatory mediators in the plasma, reduction of IL-6, TNF-α, and IFN-γ levels, and induction of type 1 interferon (IFN). Type 1 IFN has been used widely as a therapeutic agent for the treatment of MS and has been shown to be therapeutic in EAE with adoptive transfer of Th1 lymphocytes. However, type 1 IFN is known to exacerbate EAE with adoptive transfer of Th17 lymphocytes. Indeed, the amyloid fibril-forming peptide Tau 623-628 was therapeutic in Th1 adoptively transferred EAE, but ineffective in Th17 adoptively transferred EAE. However, the therapeutic effect of Tau 623-628 was restored in IFN-α/ß receptor (IFNAR) knockout mice, indicating that other immune pathways independent of type 1 IFN induction play a role in the amelioration of EAE. Moreover, Amylin 28-33, a polar, non-ionizable peptide that does not form fibrils as rapidly as Tau 623-628, induces a small fraction of type 1 IFN compared to Tau 623-628 and is therapeutic in Th17 EAE. The diverse immunological pathways modulated by the self-assembling hexapeptides are under investigation with a goal to develop novel, safe, and potent therapeutics for neuroinflammation.


Subject(s)
Amyloid/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Peptides/therapeutic use , Amyloid/chemistry , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Humans , Mice , Molecular Chaperones , Peptides/chemistry
7.
J Exp Med ; 211(9): 1847-56, 2014 Aug 25.
Article in English | MEDLINE | ID: mdl-25073790

ABSTRACT

Amyloid fibrils composed of peptides as short as six amino acids are effective therapeutics for experimental autoimmune encephalomyelitis (EAE). Immunosuppression arises from at least two pathways: (1) expression of type 1 IFN by pDCs, which were induced by neutrophil extracellular traps arising from the endocytosis of the fibrils; and (2) the reduced expression of IFN-γ, TNF, and IL-6. The two independent pathways stimulated by the fibrils can act in concert to be immunosuppressive in Th1 indications, or in opposition, resulting in inflammation when Th17 T lymphocytes are predominant. The generation of type 1 IFN can be minimized by using polar, nonionizable, amyloidogenic peptides, which are effective in both Th1 and Th17 polarized EAE.


Subject(s)
Amyloid/immunology , Amyloid/therapeutic use , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/therapy , Peptide Fragments/immunology , Peptide Fragments/therapeutic use , Adoptive Transfer , Adult , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Female , Gene Expression , Humans , Immunosuppressive Agents/therapeutic use , Interferon Type I/metabolism , Interferon-gamma/metabolism , Interleukin-6/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Th1 Cells/immunology , Th17 Cells/immunology , Tumor Necrosis Factor-alpha/metabolism , tau Proteins/immunology , tau Proteins/therapeutic use
8.
J Clin Immunol ; 34 Suppl 1: S61-3, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24711007

ABSTRACT

Amyloid forming molecules are generally considered harmful. In Alzheimer's Disease two amyloid molecules Aß A4 and tau vie for consideration as the main pathogenic culprit. But molecules obey the laws of chemistry and defy the way we categorize them as humans with our well-known proclivities to bias in our reasoning. We have been exploring the brains of multiple sclerosis patients to identify molecules that are associated with protection from inflammation and degeneration. In 2001 we noted that aB crystallin (cryab) was the most abundant transcript found in MS lesions, but not in healthy brains. Cryab can reverse paralysis and attenuate inflammation in several models of inflammation including experimental autoimmune encephalomyelitis (EAE), and various models of ischemia. Cryab is an amyloid forming molecule. We have identified a core structure common to many amyloids including amyloid protein Aß A4, tau, amylin, prion protein, serum amyloid protein P, and cryab. The core hexapeptide structure is highly immune suppressive and can reverse paralysis in EAE when administered systemically. Administration of this amyloid forming hexapeptide quickly lowers inflammatory cytokines in plasma like IL-6 and IL-2. The hexapeptide bind a set of proinflammatory mediators in plasma, including acute phase reactants and complement components. The beneficial properties of amyloid forming hexapeptides provide a potential new therapeutic direction. These experiments indicate that amyloid forming molecules have Janus faces, providing unexpected benefit for neuroinflammatory conditions.


Subject(s)
Alzheimer Disease/immunology , Amyloidogenic Proteins/immunology , Brain/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Animals , Humans , Interleukin-6/immunology , Mice , Neurogenic Inflammation/immunology
9.
Mult Scler ; 19(14): 1867-77, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23736536

ABSTRACT

BACKGROUND: Suppression of activation of pathogenic CD4(+) T cells is a potential therapeutic intervention in multiple sclerosis (MS). We previously showed that a small heat shock protein, CRYAB, reduced T cell proliferation, pro-inflammatory cytokine production and clinical signs of experimental allergic encephalomyelitis, a model of MS. OBJECTIVE: We assessed whether the ability of CRYAB to reduce the activation of T cells translated to the human disease. METHODS: CD4(+) T cells from healthy controls and volunteers with MS were activated in vitro in the presence or absence of a CRYAB peptide (residues 73-92). Parameters of activation (proliferation rate, cytokine secretion) and tolerance (anergy, activation-induced cell death, microRNAs) were evaluated. RESULTS: The secretion of pro-inflammatory cytokines by CD4(+) T cells was decreased in the presence of CRYAB in a subset of relapsing-remitting multiple sclerosis (RRMS) participants with mild disease severity while no changes were observed in healthy controls. Further, there was a correlation for higher levels of miR181a microRNA, a marker upregulated in tolerant CD8(+) T cells, in CD4(+) T cells of MS patients that displayed suppressed cytokine production (responders). CONCLUSION: CRYAB may be capable of suppressing the activation of CD4(+) T cells from a subset of RRMS patients who appear to have less disability but similar age and disease duration.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Lymphocyte Activation , Multiple Sclerosis, Relapsing-Remitting/metabolism , Peptide Fragments/metabolism , alpha-Crystallin B Chain/metabolism , Adult , CD4-Positive T-Lymphocytes/immunology , Case-Control Studies , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Female , Humans , Immune Tolerance , Inflammation Mediators/metabolism , Male , MicroRNAs/metabolism , Middle Aged , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Multiple Sclerosis, Relapsing-Remitting/genetics , Multiple Sclerosis, Relapsing-Remitting/immunology , Severity of Illness Index , Time Factors , Young Adult
10.
Sci Transl Med ; 5(179): 179ra42, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23552370

ABSTRACT

The amyloid-forming proteins tau, αB crystallin, and amyloid P protein are all found in lesions of multiple sclerosis (MS). Our previous work established that amyloidogenic peptides from the small heat shock protein αB crystallin (HspB5) and from amyloid ß fibrils, characteristic of Alzheimer's disease, were therapeutic in experimental autoimmune encephalomyelitis (EAE), reflecting aspects of the pathology of MS. To understand the molecular basis for the therapeutic effect, we showed a set of amyloidogenic peptides composed of six amino acids, including those from tau, amyloid ß A4, major prion protein (PrP), HspB5, amylin, serum amyloid P, and insulin B chain, to be anti-inflammatory and capable of reducing serological levels of interleukin-6 and attenuating paralysis in EAE. The chaperone function of the fibrils correlates with the therapeutic outcome. Fibrils composed of tau 623-628 precipitated 49 plasma proteins, including apolipoprotein B-100, clusterin, transthyretin, and complement C3, supporting the hypothesis that the fibrils are active biological agents. Amyloid fibrils thus may provide benefit in MS and other neuroinflammatory disorders.


Subject(s)
Amyloid/chemistry , Inflammation/drug therapy , Inflammation/pathology , Nervous System/pathology , Peptides/therapeutic use , Protein Multimerization , Amino Acid Sequence , Animals , Benzothiazoles , Biotinylation/drug effects , Blood Proteins/metabolism , Chemical Precipitation , Encephalomyelitis, Autoimmune, Experimental/blood , Encephalomyelitis, Autoimmune, Experimental/complications , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Hydrogen-Ion Concentration , Inflammation/blood , Inflammation/complications , Interleukin-6/blood , Mice , Mice, Inbred C57BL , Molecular Chaperones/metabolism , Molecular Sequence Data , Nervous System/drug effects , Paralysis/blood , Paralysis/complications , Paralysis/drug therapy , Peptides/chemistry , Peptides/pharmacology , Protein Multimerization/drug effects , Thiazoles/metabolism
11.
Mult Scler ; 19(1): 5-14, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23303879

ABSTRACT

Four questions were posed about multiple sclerosis (MS) at the 2011 Charcot Lecture, Oct. 22, 2011. 1. The Male/Female Disparity: Why are women developing MS so much more frequently than men? 2. Neuronal and Glial Protection: Are there guardian molecules that protect the nervous system in MS? 3. Predictive Medicine: With all the approved drugs, how can we rationally decide which one to use? 4. The Precise Scalpel vs. the Big Hammer for Therapy: Is antigen-specific therapy for demyelinating disease possible? To emphasize how our views on the pathogenesis and treatment of MS are evolving, and given the location of the talk in Amsterdam, Piet Mondrian's progressive interpretations of trees serve as a heuristic.


Subject(s)
Multiple Sclerosis , Humans
12.
J Biol Chem ; 287(43): 36423-34, 2012 Oct 19.
Article in English | MEDLINE | ID: mdl-22955287

ABSTRACT

To determine whether the therapeutic activity of αB crystallin, small heat shock protein B5 (HspB5), was shared with other human sHsps, a set of seven human family members, a mutant of HspB5 G120 known to exhibit reduced chaperone activity, and a mycobacterial sHsp were expressed and purified from bacteria. Each of the recombinant proteins was shown to be a functional chaperone, capable of inhibiting aggregation of denatured insulin with varying efficiency. When injected into mice at the peak of disease, they were all effective in reducing the paralysis in experimental autoimmune encephalomyelitis. Additional structure activity correlations between chaperone activity and therapeutic function were established when linear regions within HspB5 were examined. A single region, corresponding to residues 73-92 of HspB5, forms amyloid fibrils, exhibited chaperone activity, and was an effective therapeutic for encephalomyelitis. The linkage of the three activities was further established by demonstrating individual substitutions of critical hydrophobic amino acids in the peptide resulted in the loss of all of the functions.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Paralysis/prevention & control , alpha-Crystallin B Chain/pharmacology , Amino Acid Substitution , Animals , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Mice , Mutation, Missense , Paralysis/genetics , Paralysis/metabolism , Paralysis/pathology , alpha-Crystallin B Chain/genetics
13.
J Biol Chem ; 287(13): 9708-9721, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22308023

ABSTRACT

The therapeutic benefit of the small heat shock protein αB-crystallin (HspB5) in animal models of multiple sclerosis and ischemia is proposed to arise from its increased capacity to bind proinflammatory proteins at the elevated temperatures within inflammatory foci. By mass spectral analysis, a common set of ∼70 ligands was precipitated by HspB5 from plasma from patients with multiple sclerosis, rheumatoid arthritis, and amyloidosis and mice with experimental allergic encephalomyelitis. These proteins were distinguished from other precipitated molecules because they were enriched in the precipitate as compared with their plasma concentrations, and they exhibited temperature-dependent binding. More than half of these ligands were acute phase proteins or members of the complement or coagulation cascades. Consistent with this proposal, plasma levels of HspB5 were increased in patients with multiple sclerosis as compared with normal individuals. The combination of the thermal sensitivity of the HspB5 combined with the high local concentration of these ligands at the site of inflammation is proposed to explain the paradox of how a protein believed to exhibit nonspecific binding can bind with some relative apparent selectivity to proinflammatory proteins and thereby modulate inflammation.


Subject(s)
Blood Proteins/immunology , Molecular Chaperones/pharmacology , Multiple Sclerosis/blood , alpha-Crystallin B Chain/pharmacology , Amyloidosis/blood , Amyloidosis/drug therapy , Animals , Arthritis, Rheumatoid/blood , Arthritis, Rheumatoid/drug therapy , Encephalomyelitis, Autoimmune, Experimental , Female , Humans , Inflammation/blood , Inflammation/drug therapy , Mice , Mice, Inbred BALB C , Molecular Chaperones/blood , Multiple Sclerosis/drug therapy , Protein Binding , alpha-Crystallin B Chain/blood
14.
Proc Natl Acad Sci U S A ; 108(32): 13287-92, 2011 Aug 09.
Article in English | MEDLINE | ID: mdl-21828004

ABSTRACT

Tissue plasminogen activator is the only treatment option for stroke victims; however, it has to be administered within 4.5 h after symptom onset, making its use very limited. This report describes a unique target for effective treatment of stroke, even 12 h after onset, by the administration of αB-crystallin (Cryab), an endogenous immunomodulatory neuroprotectant. In Cryab(-/-) mice, there was increased lesion size and diminished neurologic function after stroke compared with wild-type mice. Increased plasma Cryab was detected after experimental stroke in mice and after stroke in human patients. Administration of Cryab even 12 h after experimental stroke reduced both stroke volume and inflammatory cytokines associated with stroke pathology. Cryab is an endogenous anti-inflammatory and neuroprotectant molecule produced after stroke, whose beneficial properties can be augmented when administered therapeutically after stroke.


Subject(s)
Stroke/drug therapy , Stroke/immunology , alpha-Crystallin B Chain/therapeutic use , Animals , Brain/drug effects , Brain/immunology , Brain/pathology , Humans , Immune System/drug effects , Immune System/immunology , Mice , Stroke/blood , Stroke/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , alpha-Crystallin B Chain/administration & dosage , alpha-Crystallin B Chain/blood , alpha-Crystallin B Chain/pharmacology
15.
J Immunol ; 186(7): 4263-8, 2011 Apr 01.
Article in English | MEDLINE | ID: mdl-21357544

ABSTRACT

For 15 y, α B-crystallin (heat shock protein [Hsp] B5) has been labeled an autoantigen in multiple sclerosis (MS) based on humoral and cellular responses found in humans and animal models. However, there have been several scientific inconsistencies with this assignment, ranging from studies demonstrating small differences in anticrystallin responses between patients and healthy individuals to the inability of crystallin-specific T cells to induce symptoms of experimental allergic encephalomyelitis in animal models. Experiments in this article demonstrate that the putative anti-HspB5 Abs from 23 MS patients cross-react with 7 other members of the human small Hsp family and were equally present in normal plasma. Biolayer interferometry demonstrates that the binding was temperature dependent, and that the calculated K(a) increased as the concentration of the sHsp decreased. These two patterns are characteristic of multiple binding sites with varying affinities, the composition of which changes with temperature, supporting the hypothesis that HspB5 bound the Ab and not the reverse. HspB5 also precipitated Ig heavy and L chains from sera from patients with MS. These results establish that small Hsps bind Igs with high affinity and refute much of the serological data used to assign α B-crystallin as an autoantigen.


Subject(s)
Heat-Shock Proteins, Small/metabolism , Molecular Chaperones/metabolism , alpha-Crystallin B Chain/metabolism , Animals , Autoantigens/immunology , Autoantigens/metabolism , Binding Sites, Antibody , Disease Models, Animal , Heat-Shock Proteins, Small/chemistry , Humans , Immunoglobulin Fc Fragments/chemistry , Immunoglobulin Fc Fragments/metabolism , Mice , Molecular Chaperones/chemistry , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , alpha-Crystallin B Chain/chemistry
16.
Methods Mol Biol ; 683: 487-504, 2011.
Article in English | MEDLINE | ID: mdl-21053152

ABSTRACT

A major challenge confronting the further advancement of using molecular transporters conjugated to small molecular weight therapeutics in the clinic is the development of linkers that would allow for the controllable release of a free drug/probe only after cell entry. Development of assays that would allow for the rapid real-time quantification of transporter conjugate uptake and cargo release in cells and animals would greatly help in their development. In this chapter, we describe a imaging method that quantitatively measures transporter conjugate uptake and cargo release in real-time in both cell culture and animal models.


Subject(s)
Drug Carriers/chemistry , Drug Carriers/metabolism , Firefly Luciferin/chemistry , Firefly Luciferin/metabolism , Molecular Imaging/methods , Peptides/chemistry , Administration, Topical , Animals , Biological Transport , Calibration , Cell Line, Tumor , Cyclization , Drug Carriers/administration & dosage , Firefly Luciferin/administration & dosage , Hydrolysis , Injections, Intradermal , Intracellular Space/metabolism , Kinetics , Luciferases/genetics , Luciferases/metabolism , Luminescent Measurements , Membrane Potentials , Mice , Mice, Transgenic , Oxidation-Reduction , Polylysine/chemistry
17.
Proc Natl Acad Sci U S A ; 104(25): 10340-5, 2007 Jun 19.
Article in English | MEDLINE | ID: mdl-17563383

ABSTRACT

Many therapeutic leads fail to advance clinically because of bioavailability, selectivity, and formulation problems. Molecular transporters can be used to address these problems. Molecular transporter conjugates of otherwise poorly soluble or poorly bioavailable drugs or probes exhibit excellent solubility in water and biological fluids and at the same time an enhanced ability to enter tissues and cells and with modification to do so selectively. For many conjugates, however, it is necessary to release the drug/probe cargo from the transporter after uptake to achieve activity. Here, we describe an imaging method that provides quantification of transporter conjugate uptake and cargo release in real-time in animal models. This method uses transgenic (luciferase) reporter mice and whole-body imaging, allowing noninvasive quantification of transporter conjugate uptake and probe (luciferin) release in real time. This process effectively emulates drug-conjugate delivery, drug release, and drug turnover by an intracellular target, providing a facile method to evaluate comparative uptake of new transporters and efficacy and selectivity of linker release as required for fundamental studies and therapeutic applications.


Subject(s)
Firefly Luciferin/metabolism , Firefly Luciferin/pharmacokinetics , Genes, Reporter , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Animals , Biological Transport, Active , Disulfides/chemistry , Dose-Response Relationship, Drug , Firefly Luciferin/chemistry , Injections, Intradermal , Luminescence , Luminescent Measurements , Membrane Transport Proteins/chemistry , Mice , Mice, Transgenic , Molecular Structure , Whole Body Imaging
19.
J Am Chem Soc ; 128(20): 6526-7, 2006 May 24.
Article in English | MEDLINE | ID: mdl-16704230

ABSTRACT

The design, synthesis, and evaluation of conjugates of arginine-rich transporters and luciferin are described that release luciferin only after entry into cells that are stably transfected with luciferase. Each molecule of free luciferin that is released after entry generates a photon that can be measured allowing for real-time quantification of uptake and release in cells. The process provides a method to assay uptake and release of free luciferin as a function of variations in the releasable linker and in the transporter.


Subject(s)
Biological Transport/physiology , Firefly Luciferin/analogs & derivatives , 2,2'-Dipyridyl/analogs & derivatives , 2,2'-Dipyridyl/chemistry , Animals , Cell Line, Tumor , Cells/metabolism , Disulfides/chemistry , Disulfides/metabolism , Luciferases/metabolism , Luminescent Measurements , Male , Prostatic Neoplasms/metabolism , Transfection
20.
Adv Drug Deliv Rev ; 57(4): 495-504, 2005 Feb 28.
Article in English | MEDLINE | ID: mdl-15722160

ABSTRACT

A mechanistic hypothesis is presented for how water-soluble guanidinium-rich transporters attached to small cargoes (MW ca. <3000) can migrate across the non-polar lipid membrane of a cell and enter the cytosol. Positively charged and water-soluble, arginine oligomers can associate with negatively charged, bidentate hydrogen bond acceptor groups of endogenous membrane constituents, leading to the formation of membrane-soluble ion pair complexes. The resultant less polar, ion pair complexes partition into the lipid bilayer and migrate in a direction, and with a rate, influenced by the membrane potential. The complex dissociates on the inner leaf of the membrane and the transporter conjugate enters the cytosol. This mechanism could also be involved in the translocation of guanidinium-rich molecules that are endocytosed due to their size or the conditions of the assay, across the endosomal membrane.


Subject(s)
Endocytosis/physiology , Guanidine/metabolism , Hydrogen Bonding , Membrane Transport Proteins/metabolism , Biological Transport , Guanidine/chemistry , Humans , Membrane Potentials , Membrane Transport Proteins/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...