Network


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

Hotspot


Dive into the research topics where Barbara Calamini is active.

Publication


Featured researches published by Barbara Calamini.


Nature Chemical Biology | 2012

Small Molecule Proteostasis Regulators for Protein Conformational Diseases

Barbara Calamini; Maria Catarina Silva; Franck Madoux; Darren M. Hutt; Shilpi Khanna; Monica A. Chalfant; S Adrian Saldanha; Peter Hodder; Bradley D. Tait; Dan Garza; William E. Balch; Richard I. Morimoto

Protein homeostasis (proteostasis) is essential for cellular and organismal health. Stress, aging, and the chronic expression of misfolded proteins, however, challenge the proteostasis machinery and the vitality of the cell. Enhanced expression of molecular chaperones, regulated by heat shock transcription factor-1 (HSF-1), has been shown to restore proteostasis in a variety of conformational disease models, suggesting a promising therapeutic approach. We describe the results of a ∼900,000 small molecule screen that identified novel classes of small molecule proteostasis regulators (PRs) that induce HSF-1-dependent chaperone expression and restore protein folding in multiple conformational disease models. The beneficial effects to proteome stability are mediated by HSF-1, DAF-16/FOXO, SKN-1/Nrf-2, and the chaperone machinery through mechanisms that are distinct from current known small molecule activators of the HSR. We suggest that modulation of the proteostasis network by PRs represents a promising therapeutic approach for the treatment of a variety of protein conformational diseases.


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

Protein aggregation can inhibit clathrin-mediated endocytosis by chaperone competition.

Anan Yu; Yoko Shibata; Bijal D. Shah; Barbara Calamini; Donald C. Lo; Richard I. Morimoto

Significance The aggregation of mutant proteins is pathologically implicated in a large number of neuropathies, including Huntington disease and ALS. Although the appearance of protein aggregates is known to sequester other proteins, how this results in the gain-of-function toxicity in these diseases is unclear. Here, we show that the aggregation of disease-associated proteins causes the reversible collapse of clathrin-mediated endocytosis (CME) and inhibits the internalization of membrane receptors that affect neuronal function. CME inhibition occurs through aggregate-mediated sequestration of the molecular chaperone heat shock cognate protein 70, which is essential for CME. We propose that a toxic “tug-of-war” occurs between aggregates and endogenous client proteins for available chaperones, leading to the collapse of multiple cellular processes in neurodegeneration and other protein conformation diseases. Protein conformational diseases exhibit complex pathologies linked to numerous molecular defects. Aggregation of a disease-associated protein causes the misfolding and aggregation of other proteins, but how this interferes with diverse cellular pathways is unclear. Here, we show that aggregation of neurodegenerative disease-related proteins (polyglutamine, huntingtin, ataxin-1, and superoxide dismutase-1) inhibits clathrin-mediated endocytosis (CME) in mammalian cells by aggregate-driven sequestration of the major molecular chaperone heat shock cognate protein 70 (HSC70), which is required to drive multiple steps of CME. CME suppression was also phenocopied by HSC70 RNAi depletion and could be restored by conditionally increasing HSC70 abundance. Aggregation caused dysregulated AMPA receptor internalization and also inhibited CME in primary neurons expressing mutant huntingtin, showing direct relevance of our findings to the pathology in neurodegenerative diseases. We propose that aggregate-associated chaperone competition leads to both gain-of-function and loss-of-function phenotypes as chaperones become functionally depleted from multiple clients, leading to the decline of multiple cellular processes. The inherent properties of chaperones place them at risk, contributing to the complex pathologies of protein conformational diseases.


Current Topics in Medicinal Chemistry | 2013

Protein Homeostasis as a Therapeutic Target for Diseases of Protein Conformation

Barbara Calamini; Richard I. Morimoto

Protein misfolding and aggregation are widely implicated in an increasing number of human diseases providing for new therapeutic opportunities targeting protein homeostasis (proteostasis). The cellular response to proteotoxicity is highly regulated by stress signaling pathways, molecular chaperones, transport and clearance machineries that function as a proteostasis network (PN) to protect the stability and functional properties of the proteome. Consequently, the PN is essential at the cellular and organismal level for development and lifespan. However, when challenged during aging, stress, and disease, the folding and clearance machineries can become compromised leading to both gain-of-function and loss-of-function proteinopathies. Here, we assess the role of small molecules that activate the heat shock response, the unfolded protein response, and clearance mechanisms to increase PN capacity and protect cellular proteostasis against proteotoxicity. We propose that this strategy to enhance cell stress pathways and chaperone activity establishes a cytoprotective state against misfolding and/or aggregation and represents a promising therapeutic avenue to prevent the cellular damage associated with the variety of protein conformational diseases.


Journal of Biological Chemistry | 2013

Heat shock response activation exacerbates inclusion body formation in a cellular model of Huntington disease.

Kirill Bersuker; Mark S. Hipp; Barbara Calamini; Richard I. Morimoto; Ron R. Kopito

Background: Aggregation of mutant huntingtin into inclusion bodies is a pathogenic feature of Huntington disease. Results: Genetic and pharmacological activation of the heat shock stress response (HSR) promotes inclusion body formation. Conclusion: Activation of the HSR contributes to mutant huntingtin aggregation. Significance: HSR activation as a therapeutic strategy may promote rather than suppress aggregation of proteins implicated in neurodegenerative disease. The cellular heat shock response (HSR) protects cells from toxicity associated with defective protein folding, and this pathway is widely viewed as a potential pharmacological target to treat neurodegenerative diseases linked to protein aggregation. Here we show that the HSR is not activated by mutant huntingtin (HTT) even in cells selected for the highest expression levels and for the presence of inclusion bodies containing aggregated protein. Surprisingly, HSR activation by HSF1 overexpression or by administration of a small molecule activator lowers the concentration threshold at which HTT forms inclusion bodies in cells expressing aggregation-prone, polyglutamine-expanded fragments of HTT. These data suggest that the HSR does not mitigate inclusion body formation.


eLife | 2016

Control of the structural landscape and neuronal proteotoxicity of mutant Huntingtin by domains flanking the polyQ tract

Koning Shen; Barbara Calamini; Jonathan A Fauerbach; Boxue Ma; Sarah H. Shahmoradian; Ivana L Serrano Lachapel; Wah Chiu; Donald C. Lo; Judith Frydman

Many neurodegenerative diseases are linked to amyloid aggregation. In Huntington’s disease (HD), neurotoxicity correlates with an increased aggregation propensity of a polyglutamine (polyQ) expansion in exon 1 of mutant huntingtin protein (mHtt). Here we establish how the domains flanking the polyQ tract shape the mHtt conformational landscape in vitro and in neurons. In vitro, the flanking domains have opposing effects on the conformation and stabilities of oligomers and amyloid fibrils. The N-terminal N17 promotes amyloid fibril formation, while the C-terminal Proline Rich Domain destabilizes fibrils and enhances oligomer formation. However, in neurons both domains act synergistically to engage protective chaperone and degradation pathways promoting mHtt proteostasis. Surprisingly, when proteotoxicity was assessed in rat corticostriatal brain slices, either flanking region alone sufficed to generate a neurotoxic conformation, while the polyQ tract alone exhibited minimal toxicity. Linking mHtt structural properties to its neuronal proteostasis should inform new strategies for neuroprotection in polyQ-expansion diseases. DOI: http://dx.doi.org/10.7554/eLife.18065.001


Oncotarget | 2016

IKK/NF-κB signaling contributes to glioblastoma stem cell maintenance

Amanda L. Rinkenbaugh; Patricia C. Cogswell; Barbara Calamini; Denise E. Dunn; Anders Persson; William A. Weiss; Donald C. Lo; Albert S. Baldwin

Glioblastoma multiforme (GBM) carries a poor prognosis and continues to lack effective treatments. Glioblastoma stem cells (GSCs) drive tumor formation, invasion, and drug resistance and, as such, are the focus of studies to identify new therapies for disease control. Here, we identify the involvement of IKK and NF-κB signaling in the maintenance of GSCs. Inhibition of this pathway impairs self-renewal as analyzed in tumorsphere formation and GBM expansion as analyzed in brain slice culture. Interestingly, both the canonical and non-canonical branches of the NF-κB pathway are shown to contribute to this phenotype. One source of NF-κB activation in GBM involves the TGF-β/TAK1 signaling axis. Together, our results demonstrate a role for the NF-κB pathway in GSCs and provide a mechanistic basis for its potential as a therapeutic target in glioblastoma.


Neurotherapeutics | 2013

Experimental Models for Identifying Modifiers of Polyglutamine-Induced Aggregation and Neurodegeneration

Barbara Calamini; Donald C. Lo; Linda S. Kaltenbach

Huntington’s disease (HD) typifies a class of inherited neurodegenerative disorders in which a CAG expansion in a single gene leads to an extended polyglutamine tract and misfolding of the expressed protein, driving cumulative neural dysfunction and degeneration. HD is invariably fatal with symptoms that include progressive neuropsychiatric and cognitive impairments, and eventual motor disability. No curative therapies yet exist for HD and related polyglutamine diseases; therefore, substantial efforts have been made in the drug discovery field to identify potential drug and drug target candidates for disease-modifying treatment. In this context, we review here a range of early-stage screening approaches based in in vitro, cellular, and invertebrate models to identify pharmacological and genetic modifiers of polyglutamine aggregation and induced neurodegeneration. In addition, emerging technologies, including high-content analysis, three-dimensional culture models, and induced pluripotent stem cells are increasingly being incorporated into drug discovery screening pipelines for protein misfolding disorders. Together, these diverse screening strategies are generating novel and exciting new probes for understanding the disease process and for furthering development of therapeutic candidates for eventual testing in the clinical setting.


Human Molecular Genetics | 2017

IKKβ and mutant huntingtin interactions regulate the expression of IL-34: implications for microglial-mediated neurodegeneration in HD

Ali Khoshnan; Adam Sabbaugh; Barbara Calamini; Steven Marinero; Denise E. Dunn; Jung Hyun Yoo; Jan Ko; Donald C. Lo; Paul H. Patterson

Neuronal interleukin-34 (IL-34) promotes the expansion of microglia in the central nervous system-microglial activation and expansion are in turn implicated in the pathogenesis of Huntingtons disease (HD). We thus examined whether the accumulation of an amyloidogenic exon-1 fragment of mutant huntingtin (mHTTx1) modulates the expression of IL-34 in dopaminergic neurons derived from a human embryonic stem cell line. We found that mHTTx1 aggregates induce IL-34 production selectively in post-mitotic neurons. Exposure of neurons to DNA damaging agents or the excitotoxin NMDA elicited similar results suggesting that IL-34 induction may be a general response to neuronal stress including the accumulation of misfolded mHTTx1. We further determined that knockdown or blocking the activity of IκB kinase beta (IKKβ) prevented the aggregation of mHTTx1 and subsequent IL-34 production. While elevated IL-34 itself had no effect on the aggregation or the toxicity of mHTTx1 in neuronal culture, IL-34 expression in a rodent brain slice model with intact neuron-microglial networks exacerbated mHTTx1-induced degeneration of striatal medium-sized spiny neurons. Conversely, an inhibitor of the IL-34 receptor reduced microglial numbers and ameliorated mHTTx1-mediated neurodegeneration. Together, these findings uncover a novel function for IKKβ/mHTTx1 interactions in regulating IL-34 production, and implicate a role for IL-34 in non-cell-autonomous, microglial-dependent neurodegeneration in HD.


Archive | 2009

METHOD OF MODULATING HSF-1

Richard I. Morimoto; Sandy D. Westerheide; Julius Anckar; Lea Sistonen; Barbara Calamini


Biophysical Journal | 2014

Polyglutamine Flanking Regions in Huntingtin Highlight Key Structural Intermediates Relevant for Molecular Chaperone Interaction and Huntington's Disease Pathogenesis

Koning Shen; Barbara Calamini; Donald C. Lo; Judith Frydman

Collaboration


Dive into the Barbara Calamini's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Darren M. Hutt

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Franck Madoux

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Peter Hodder

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

William E. Balch

La Jolla Institute for Allergy and Immunology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Christophe Allais

Scripps Research Institute

View shared research outputs
Top Co-Authors

Avatar

Jill Ferguson

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge