Network


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

Hotspot


Dive into the research topics where Miriam Braunstein is active.

Publication


Featured researches published by Miriam Braunstein.


Journal of Clinical Investigation | 2007

Enhanced priming of adaptive immunity by a proapoptotic mutant of Mycobacterium tuberculosis

Joseph Hinchey; Sunhee Lee; Bo Y. Jeon; Randall J. Basaraba; Manjunatha M. Venkataswamy; Bing Chen; John Chan; Miriam Braunstein; Ian M. Orme; Steven C. Derrick; Sheldon L. Morris; William R. Jacobs; Steven A. Porcelli

The inhibition of apoptosis of infected host cells is a well-known but poorly understood function of pathogenic mycobacteria. We show that inactivation of the secA2 gene in Mycobacterium tuberculosis, which encodes a component of a virulence-associated protein secretion system, enhanced the apoptosis of infected macrophages by diminishing secretion of mycobacterial superoxide dismutase. Deletion of secA2 markedly increased priming of antigen-specific CD8(+) T cells in vivo, and vaccination of mice and guinea pigs with a secA2 mutant significantly increased resistance to M. tuberculosis challenge compared with standard M. bovis bacille Calmette-Guérin vaccination. Our results define a mechanism for a key immune evasion strategy of M. tuberculosis and provide what we believe to be a novel approach for improving mycobacterial vaccines.


Journal of Biological Chemistry | 2005

The CATERPILLER Protein Monarch-1 Is an Antagonist of Toll-like Receptor-, Tumor Necrosis Factor α-, and Mycobacterium tuberculosis-induced Pro-inflammatory Signals

Kristi L. Williams; John D. Lich; Joseph A. Duncan; William Reed; Prasad Rallabhandi; Chris B. Moore; Sherry Kurtz; V. McNeil Coffield; Mary Ann Accavitti-Loper; Lishan Su; Stefanie N. Vogel; Miriam Braunstein; Jenny P.-Y. Ting

The CATERPILLER (CLR, also NOD and NLR) proteins share structural similarities with the nucleotide binding domain (NBD)-leucine-rich repeat (LRR) superfamily of plant disease-resistance (R) proteins and are emerging as important immune regulators in animals. CLR proteins contain NBD-LRR motifs and are linked to a limited number of distinct N-terminal domains including transactivation, CARD (caspase activation and recruitment), and pyrin domains (PyD). The CLR gene, Monarch-1/Pypaf7, is expressed by resting primary myeloid/monocytic cells, and its expression in these cells is reduced by Toll-like receptor (TLR) agonists tumor necrosis factor (TNF) α and Mycobacterium tuberculosis. Monarch-1 reduces NFκB activation by TLR-signaling molecules MyD88, IRAK-1 (type I interleukin-1 receptor-associated protein kinase), and TRAF6 (TNF receptor (TNFR)-associated factor) as well as TNFR signaling molecules TRAF2 and RIP1 but not the downstream NFκB subunit p65. This indicates that Monarch-1 is a negative regulator of both TLR and TNFR pathways. Reducing Monarch-1 expression with small interference RNA in myeloid/monocytic cells caused a dramatic increase in NFκB activation and cytokine expression in response to TLR2/TLR4 agonists, TNFα, or M. tuberculosis infection, suggesting that Monarch-1 is a negative regulator of inflammation. Because Monarch-1 is the first CLR protein that interferes with both TLR2 and TLR4 activation, the mechanism of this interference is significant. We find that Monarch-1 associates with IRAK-1 but not MyD88, resulting in the blockage of IRAK-1 hyperphosphorylation. Mutants containing the NBD-LRR or PyD-NBD also blocked IRAK-1 activation. This is the first example of a CLR protein that antagonizes inflammatory responses initiated by TLR agonists via interference with IRAK-1 activation.


Proceedings of the National Academy of Sciences of the United States of America | 2003

Crystal structure of Mycobacterium tuberculosis SecA, a preprotein translocating ATPase

Vivek Sharma; Arulandu Arockiasamy; Donald R. Ronning; Christos G. Savva; Andreas Holzenburg; Miriam Braunstein; William R. Jacobs; James C. Sacchettini

In bacteria, the majority of exported proteins are translocated by the Sec system, which recognizes the signal sequence of a preprotein and uses ATP and the proton motive force to mediate protein translocation across the cytoplasmic membrane. SecA is an essential protein component of this system, containing the molecular motor that facilitates translocation. Here we report the three-dimensional structure of the SecA protein of Mycobacterium tuberculosis. Each subunit of the homodimer contains a “motor” domain and a translocation domain. The structure predicts that SecA can interact with the SecYEG pore and function as a molecular ratchet that uses ATP hydrolysis for physical movement of the preprotein. Knowledge of this structure provides a framework for further elucidation of the translocation process.


Journal of Bacteriology | 2001

Two Nonredundant SecA Homologues Function in Mycobacteria

Miriam Braunstein; Amanda M. Brown; Sherry Kurtz; William R. Jacobs

The proper extracytoplasmic localization of proteins is an important aspect of mycobacterial physiology and the pathogenesis of Mycobacterium tuberculosis. The protein export systems of mycobacteria have remained unexplored. The Sec-dependent protein export pathway has been well characterized in Escherichia coli and is responsible for transport across the cytoplasmic membrane of proteins containing signal sequences at their amino termini. SecA is a central component of this pathway, and it is highly conserved throughout bacteria. Here we report on an unusual property of mycobacterial protein export--the presence of two homologues of SecA (SecA1 and SecA2). Using an allelic-exchange strategy in Mycobacterium smegmatis, we demonstrate that secA1 is an essential gene. In contrast, secA2 can be deleted and is the first example of a nonessential secA homologue. The essential nature of secA1, which is consistent with the conserved Sec pathway, leads us to believe that secA1 represents the equivalent of E. coli secA. The results of a phenotypic analysis of a Delta secA2 mutant of M. smegmatis are presented here and also indicate a role for SecA2 in protein export. Based on our study, it appears that SecA2 can assist SecA1 in the export of some proteins via the Sec pathway. However, SecA2 is not the functional equivalent of SecA1. This finding, in combination with the fact that SecA2 is highly conserved throughout mycobacteria, suggests a second role for SecA2. The possibility exists that another role for SecA2 is to export a specific subset of proteins.


Molecular Microbiology | 2008

A new twist on an old pathway – accessory secretion systems

Nathan W. Rigel; Miriam Braunstein

The export of proteins from their site of synthesis in the cytoplasm across the inner membrane is an important aspect of bacterial physiology. Because the location of extracytoplasmic proteins is ideal for host–pathogen interactions, protein export is also important to bacterial virulence. In bacteria, there are conserved protein export systems that are responsible for the majority of protein export: the general secretion (Sec) pathway and the twin‐arginine translocation pathway. In some bacteria, there are also specialized export systems dedicated to exporting specific subsets of proteins. In this review, we discuss a specialized export system that exists in some Gram‐positive bacteria and mycobacteria – the accessory Sec system. The common element to the accessory Sec system is an accessory SecA protein called SecA2. Here we present our current understanding of accessory Sec systems in Streptococcus gordonii, Streptococcus parasanguinis, Mycobacterium smegmatis, Mycobacterium tuberculosis and Listeria monocytogenes, making an effort to highlight apparent similarities and differences between the systems. We also review the data showing that accessory Sec systems can contribute to bacterial virulence.


Infection and Immunity | 2006

The SecA2 secretion factor of Mycobacterium tuberculosis promotes growth in macrophages and inhibits the host immune response.

Sherry Kurtz; Karen P. McKinnon; Marschall S. Runge; Jenny P.-Y. Ting; Miriam Braunstein

ABSTRACT The SecA protein is present in all bacteria, and it is a central component of the general Sec-dependent protein export pathway. An unusual property of Mycobacterium tuberculosis is the presence of two SecA proteins: SecA1, the essential “housekeeping” SecA, and SecA2, the accessory secretion factor. Here, we report that a ΔsecA2 mutant of M. tuberculosis was defective for growth in the early stages of low-dose aerosol infection of C57BL/6 mice, a time during which the bacillus is primarily replicating in macrophages. Consistent with this in vivo phenotype, we found that the ΔsecA2 mutant was defective for growth in macrophages from C57BL/6 mice. The ΔsecA2 mutant was also attenuated for growth in macrophages from phox−/− mice and from NOS2−/− mice. These mice are defective in the reactive oxygen intermediate (ROI)-generating phagocyte oxidase and the reactive nitrogen intermediate (RNI)-generating inducible nitric oxide synthase, respectively. This indicated a role for SecA2 in the intracellular growth of M. tuberculosis that is independent of protecting against these ROIs or RNIs. Macrophages infected with the ΔsecA2 mutant produced higher levels of tumor necrosis factor alpha, interleukin-6, RNI, and gamma interferon-induced major histocompatibility complex class II. This demonstrated a function for M. tuberculosis SecA2 in suppressing macrophage immune responses, which could explain the role of SecA2 in intracellular growth. Our results provide another example of a relationship between M. tuberculosis virulence and inhibition of the host immune response.


Methods in Enzymology | 2002

Genetic methods for deciphering virulence determinants of Mycobacterium tuberculosis.

Miriam Braunstein; Stoyan S. Bardarov; William R. Jacobs

Publisher Summary This chapter describes the methodologies for directed allelic exchange and transposon mutagenesis, employed by laboratory to engineer mutant strains of Mycobacterium. tuberculosis . Allelic exchange protocols based on plasmid transformation or a recently developed mycobacteriophage delivery system are presented. The chapter also describes the use of the mycobacteriophage delivery system for transposon mutagenesis of Mycobacterium. tuberculosis . The ability to apply molecular genetics to the study of M. tuberculosis only recently became a reality. Efficient methods for DNA transfer, utilizing transformation by electroporation and phage transduction, are now available for the production of defined mutations in M. tuberculosis. The development of these techniques IS impeded by the intrinsic difficulties of working with M. tuberculosis that include slow growth (a generation time of 18-24 hr), biosafety considerations, and a tendency to grow in clumps (making isolation of individual clones problematic).


Nature Reviews Microbiology | 2012

Emerging themes in SecA2-mediated protein export

Meghan E. Feltcher; Miriam Braunstein

The conserved general secretion (Sec) pathway carries out most protein export in bacteria and is powered by the essential ATPase SecA. Interestingly, mycobacteria and some Gram-positive bacteria possess two SecA proteins: SecA1 and SecA2. In these species, SecA1 is responsible for exporting most proteins, whereas SecA2 exports only a subset of substrates and is implicated in virulence. However, despite the impressive body of knowledge about the canonical SecA1, less is known concerning SecA2 function. Here, we review our current understanding of the different types of SecA2 systems and outline future directions for their study.


Journal of Bacteriology | 2007

Identification of two Mycobacterium smegmatis lipoproteins exported by a SecA2-dependent pathway

Henry S. Gibbons; Frank Wolschendorf; Michelle Y. Abshire; Michael Niederweis; Miriam Braunstein

The SecA2 protein is part of a specialized protein export system of mycobacteria. We set out to identify proteins exported to the bacterial cell envelope by the mycobacterial SecA2 system. By comparing the protein profiles of cell wall and membrane fractions from wild-type and DeltasecA2 mutant Mycobacterium smegmatis, we identified the Msmeg1712 and Msmeg1704 proteins as SecA2-dependent cell envelope proteins. These are the first endogenous M. smegmatis proteins identified as dependent on SecA2 for export. Both proteins are homologous to periplasmic sugar-binding proteins of other bacteria, and both contain functional amino-terminal signal sequences with lipobox motifs. These two proteins appeared to be genuine lipoproteins as shown by Triton X-114 fractionation and sensitivity to globomycin, an inhibitor of lipoprotein signal peptidase. The role of SecA2 in the export of these proteins was specific; not all mycobacterial lipoproteins required SecA2 for efficient localization or processing. Finally, Msmeg1704 was recognized by the SecA2 pathway of Mycobacterium tuberculosis, as indicated by the appearance of an export intermediate when the protein was expressed in a DeltasecA2 mutant of M. tuberculosis. Taken together, these results indicate that a select subset of envelope proteins containing amino-terminal signal sequences can be substrates of the mycobacterial SecA2 pathway and that some determinants for SecA2-dependent export are conserved between M. smegmatis and M. tuberculosis.


Infection and Immunity | 2012

The Mycobacterium tuberculosis SecA2 system subverts phagosome maturation to promote growth in macrophages

Jonathan Tabb Sullivan; Ellen F. Young; Jessica R. McCann; Miriam Braunstein

ABSTRACT The ability of Mycobacterium tuberculosis to grow in macrophages is critical to the virulence of this important pathogen. One way M. tuberculosis is thought to maintain a hospitable niche in macrophages is by arresting the normal process of phagosomes maturing into acidified phagolysosomes. The process of phagosome maturation arrest by M. tuberculosis is not fully understood, and there has remained a need to firmly establish a requirement for phagosome maturation arrest for M. tuberculosis growth in macrophages. Other intracellular pathogens that control the phagosomal environment use specialized protein export systems to deliver effectors of phagosome trafficking to the host cell. In M. tuberculosis, the accessory SecA2 system is a specialized protein export system that is required for intracellular growth in macrophages. In studying the importance of the SecA2 system in macrophages, we discovered that SecA2 is required for phagosome maturation arrest. Shortly after infection, phagosomes containing a ΔsecA2 mutant of M. tuberculosis were more acidified and showed greater association with markers of late endosomes than phagosomes containing wild-type M. tuberculosis. We further showed that inhibitors of phagosome acidification rescued the intracellular growth defect of the ΔsecA2 mutant, which demonstrated that the phagosome maturation arrest defect of the ΔsecA2 mutant is responsible for the intracellular growth defect. This study demonstrates the importance of phagosome maturation arrest for M. tuberculosis growth in macrophages, and it suggests there are effectors of phagosome maturation that are exported into the host environment by the accessory SecA2 system.

Collaboration


Dive into the Miriam Braunstein's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ellen F. Young

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jennifer D. Hayden

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jessica R. McCann

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Meghan E. Feltcher

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Henry S. Gibbons

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Jonathan Tabb Sullivan

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

Nathan W. Rigel

University of North Carolina at Chapel Hill

View shared research outputs
Top Co-Authors

Avatar

William R. Jacobs

University of North Carolina at Chapel Hill

View shared research outputs
Researchain Logo
Decentralizing Knowledge