Network


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

Hotspot


Dive into the research topics where Michael P. Kurnellas is active.

Publication


Featured researches published by Michael P. Kurnellas.


Journal of Biological Chemistry | 2012

Therapeutic Effects of Systemic Administration of Chaperone αB-Crystallin Associated with Binding Proinflammatory Plasma Proteins

Jonathan B. Rothbard; Michael P. Kurnellas; Sara E. Brownell; Christopher M. Adams; Leon Su; Robert C. Axtell; Rong Chen; C. Garrison Fathman; William H. Robinson; Lawrence Steinman

Background: αB-Crystallin is therapeutic in animal models of multiple sclerosis and ischemia. Results: Crystallin binds ∼70 plasma proteins; over half are members of the acute phase, coagulation, and complement pathways. Conclusion: The heat shock protein can bind with apparent selectivity and modulate inflammation. Significance: The capacity of the heat shock protein to bind a spectrum of ligands represents a unique therapeutic reagent. 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.


Science Translational Medicine | 2013

Amyloid Fibrils Composed of Hexameric Peptides Attenuate Neuroinflammation

Michael P. Kurnellas; Christopher M. Adams; Raymond A. Sobel; Lawrence Steinman; Jonathan B. Rothbard

Amyloid fibrils may provide anti-inflammatory benefit in MS and other neuroinflammatory disorders. MS Had Better Watch Its Six It’s easy to tell the good guys from the bad guys in superhero comics: Have you ever heard a hero give an “evil laugh”? However, in disease pathogenesis, things aren’t so cut and dried. Proteins that are thought to be bad actors can actually play a healing role. Amyloid-forming proteins—such as tau, αB crystallin, and amyloid P—are examples of such complex characters. Although amyloid fibrils are thought to contribute to pathogenesis in Alzheimer’s disease, it’s becoming increasingly clear that that isn’t the whole story. Now, Kurnellas et al. suggest that hexameric amyloid peptides may be broadly anti-inflammatory in the setting of neuroinflammatory disease. The authors examined a series of hexameric amyloid peptides in experimental autoimmune encephalomyelitis (EAE), a mouse model of multiple sclerosis. They found that these peptides reduced the production of the proinflammatory cytokine interleukin-6 (IL-6) as well as attenuated clinical symptoms of EAE such as paralysis. Indeed, amyloid fibrils formed from these hexamers could precipitate proteins out of plasma and correlated with chaperone function, which may contribute to the anti-inflammatory mechanism. 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 β 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 β 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.


Journal of Biological Chemistry | 2012

Chaperone activity of small heat shock proteins underlies therapeutic efficacy in experimental autoimmune encephalomyelitis

Michael P. Kurnellas; Sara E. Brownell; Leon Su; Andrey V. Malkovskiy; Jayakumar Rajadas; Gregory Dolganov; Sidharth Chopra; Gary K. Schoolnik; Raymond A. Sobel; Jonathan Webster; Shalina S. Ousman; Rachel A. Becker; Lawrence Steinman; Jonathan B. Rothbard

Background: The small heat shock protein, HspB5, is therapeutic in experimental autoimmune encephalomyelitis. Results: Eight other human sHsps, a mycobacterial sHsp, and a linear peptide from HspB5 were equally effective therapeutics. Conclusion: All of the therapeutic proteins and peptides were also molecular chaperones. Significance: Correlation between chaperone activity and therapeutic function supports data demonstrating sHsps bind inflammatory mediators in plasma. 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.


Journal of Immunology | 2011

Chaperone Activity of α B-Crystallin Is Responsible for Its Incorrect Assignment as an Autoantigen in Multiple Sclerosis

Jonathan B. Rothbard; Xiaoyan Zhao; Orr Sharpe; Michael Strohman; Michael P. Kurnellas; Elizabeth D. Mellins; William H. Robinson; Lawrence Steinman

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 Ka 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.


Journal of Clinical Immunology | 2014

Janus Faces of Amyloid Proteins in Neuroinflammation

Lawrence Steinman; Jonathan B. Rothbard; Michael P. Kurnellas

Amyloid forming molecules are generally considered harmful. In Alzheimer’s Disease two amyloid molecules Aβ 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 Aβ 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.


Journal of Experimental Medicine | 2014

Mechanisms of action of therapeutic amyloidogenic hexapeptides in amelioration of inflammatory brain disease

Michael P. Kurnellas; Jill Schartner; C. Garrison Fathman; Ann Jagger; Lawrence Steinman; Jonathan B. Rothbard

Immunosuppression from amyloidogenic peptides arises from two pathways, expression of type 1 IFN by pDCs and reduced expression of IFN-γ, TNF, and IL-6, which together modulate the signs of both Th1- and Th17-induced EAE.


Proceedings of the National Academy of Sciences of the United States of America | 2015

Amyloid fibrils activate B-1a lymphocytes to ameliorate inflammatory brain disease

Michael P. Kurnellas; Eliver Eid Bou Ghosn; Jill Schartner; Jeanette Baker; Jesse J. Rothbard; Robert S. Negrin; Leonore A. Herzenberg; C. Garrison Fathman; Lawrence Steinman; Jonathan B. Rothbard

Significance IL-10–secreting B lymphocytes and peritoneal macrophages are activated by immunization with amyloid fibrils composed of short peptides resulting in reduction of paralysis and inflammation in mice with experimental autoimmune encephalomyelitis. B-cell–deficient μMT mice and IL-10 knockout animals were used to establish the critical role of regulatory B cells in the therapeutic mode of action. Reintroduction of B-1a lymphocytes into the μMT animals reconstituted the ability of the fibrils to ameliorate the paralytic signs, leading to the trafficking of both populations of cells from the peritoneum to secondary lymph organs and not to the CNS. The reduction in CNS inflammation, combined with successful intranasal administration, provides support that this strategy could be translated into an effective human therapeutic. 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.


Multiple Sclerosis Journal | 2013

Piet Mondrian's trees and the evolution in understanding multiple sclerosis, Charcot Prize Lecture 2011

Lawrence Steinman; Robert C. Axtell; Donald Barbieri; Roopa Bhat; Sara E. Brownell; Brigit A. de Jong; Shannon E. Dunn; Jacqueline Grant; May H. Han; Peggy P. Ho; Hedwich F. Kuipers; Michael P. Kurnellas; Shalina S. Ousman; Jonathan B. Rothbard

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.


Current topics in behavioral neurosciences | 2015

Self-Assembling Peptides Form Immune Suppressive Amyloid Fibrils Effective in Autoimmune Encephalomyelitis

Michael P. Kurnellas; Jonathan B. Rothbard; Lawrence Steinman

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/β 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.


Journal of Neuroimmunology | 2016

Genetic background modulates outcome of therapeutic amyloid peptides in treatment of neuroinflammation

Allison Kraus; Brent Race; Katie Phillips; Clayton W. Winkler; Greg Saturday; Michael P. Kurnellas; Jonathan B. Rothbard; Bradley R. Groveman; Lawrence Steinman; Byron Caughey

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.

Collaboration


Dive into the Michael P. Kurnellas's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Leon Su

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

William H. Robinson

United States Department of Veterans Affairs

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge