Network


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

Hotspot


Dive into the research topics where Pranoti Sahasrabhojane is active.

Publication


Featured researches published by Pranoti Sahasrabhojane.


Journal of Clinical Investigation | 2011

Naturally occurring single amino acid replacements in a regulatory protein alter streptococcal gene expression and virulence in mice.

Ronan K. Carroll; Samuel A. Shelburne; Randall J. Olsen; Bryce Suber; Pranoti Sahasrabhojane; Muthiah Kumaraswami; Stephen B. Beres; Patrick R. Shea; Anthony R. Flores; James M. Musser

Infection with different strains of the same species of bacteria often results in vastly different clinical outcomes. Despite extensive investigation, the genetic basis of microbial strain-specific virulence remains poorly understood. Recent whole-genome sequencing has revealed that SNPs are the most prevalent form of genetic diversity among different strains of the same species of bacteria. For invasive serotype M3 group A streptococci (GAS) strains, the gene encoding regulator of proteinase B (RopB) has the highest frequency of SNPs. Here, we have determined that ropB polymorphisms alter RopB function and modulate GAS host-pathogen interactions. Sequencing of ropB in 171 invasive serotype M3 GAS strains identified 19 distinct ropB alleles. Inactivation of the ropB gene in strains producing distinct RopB variants had dramatically divergent effects on GAS global gene expression. Additionally, generation of isoallelic GAS strains differing only by a single amino acid in RopB confirmed that variant proteins affected transcript levels of the gene encoding streptococcal proteinase B, a major RopB-regulated virulence factor. Comparison of parental, RopB-inactivated, and RopB isoallelic strains in mouse infection models demonstrated that ropB polymorphisms influence GAS virulence and disease manifestations. These data detail a paradigm in which unbiased, whole-genome sequence analysis of populations of clinical bacterial isolates creates new avenues of productive investigation into the pathogenesis of common human infections.


PLOS Pathogens | 2010

A Combination of Independent Transcriptional Regulators Shapes Bacterial Virulence Gene Expression during Infection

Samuel A. Shelburne; Randall J. Olsen; Bryce Suber; Pranoti Sahasrabhojane; Paul Sumby; Richard G. Brennan; James M. Musser

Transcriptional regulatory networks are fundamental to how microbes alter gene expression in response to environmental stimuli, thereby playing a critical role in bacterial pathogenesis. However, understanding how bacterial transcriptional regulatory networks function during host-pathogen interaction is limited. Recent studies in group A Streptococcus (GAS) suggested that the transcriptional regulator catabolite control protein A (CcpA) influences many of the same genes as the control of virulence (CovRS) two-component gene regulatory system. To provide new information about the CcpA and CovRS networks, we compared the CcpA and CovR transcriptomes in a serotype M1 GAS strain. The transcript levels of several of the same genes encoding virulence factors and proteins involved in basic metabolic processes were affected in both ΔccpA and ΔcovR isogenic mutant strains. Recombinant CcpA and CovR bound with high-affinity to the promoter regions of several co-regulated genes, including those encoding proteins involved in carbohydrate and amino acid metabolism. Compared to the wild-type parental strain, ΔccpA and ΔcovRΔccpA isogenic mutant strains were significantly less virulent in a mouse myositis model. Inactivation of CcpA and CovR alone and in combination led to significant alterations in the transcript levels of several key GAS virulence factor encoding genes during infection. Importantly, the transcript level alterations in the ΔccpA and ΔcovRΔccpA isogenic mutant strains observed during infection were distinct from those occurring during growth in laboratory medium. These data provide new knowledge regarding the molecular mechanisms by which pathogenic bacteria respond to environmental signals to regulate virulence factor production and basic metabolic processes during infection.


Emerging Infectious Diseases | 2014

Streptococcus mitis strains causing severe clinical disease in cancer patients.

Samuel A. Shelburne; Pranoti Sahasrabhojane; Miguel Saldaña; Hui Yao; Xiaoping Su; Nicola Horstmann; Erika Thompson; Anthony R. Flores

The genetically diverse viridans group streptococci (VGS) are increasingly recognized as the cause of a variety of human diseases. We used a recently developed multilocus sequence analysis scheme to define the species of 118 unique VGS strains causing bacteremia in patients with cancer; Streptococcus mitis (68 patients) and S. oralis (22 patients) were the most frequently identified strains. Compared with patients infected with non–S. mitis strains, patients infected with S. mitis strains were more likely to have moderate or severe clinical disease (e.g., VGS shock syndrome). Combined with the sequence data, whole-genome analyses showed that S. mitis strains may more precisely be considered as >2 species. Furthermore, we found that multiple S. mitis strains induced disease in neutropenic mice in a dose-dependent fashion. Our data define the prominent clinical effect of the group of organisms currently classified as S. mitis and lay the groundwork for increased understanding of this understudied pathogen.


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

Sequence type 1 group B Streptococcus, an emerging cause of invasive disease in adults, evolves by small genetic changes

Anthony R. Flores; Jessica Galloway-Peña; Pranoti Sahasrabhojane; Miguel Saldaña; Hui Yao; Xiaoping Su; Nadim J. Ajami; Michael Holder; Joseph F. Petrosino; Erika Thompson; Immaculada Margarit Y Ros; Roberto Rosini; Guido Grandi; Nicola Horstmann; Sarah Teatero; Allison McGeer; Nahuel Fittipaldi; Rino Rappuoli; Carol J. Baker; Samuel A. Shelburne

Significance Serotype V group B Streptococcus (GBS) infection rates in humans have steadily increased during the past several decades. We determined that 92% of bloodstream infections caused by serotype V GBS in Houston and Toronto are caused by genetically related strains called sequence type (ST) 1. Whole-genome analysis of 202 serotype V ST-1 strains revealed the molecular relationship among these strains and that they are closely related to a bovine strain. Moreover, we found that a subset of GBS genes is under selective evolutionary pressure, indicating that proteins produced by these genes likely contribute to GBS host–pathogen interaction. These data will assist in understanding how bacteria adapt to cause disease in humans, thereby potentially informing new preventive and therapeutic strategies. The molecular mechanisms underlying pathogen emergence in humans is a critical but poorly understood area of microbiologic investigation. Serotype V group B Streptococcus (GBS) was first isolated from humans in 1975, and rates of invasive serotype V GBS disease significantly increased starting in the early 1990s. We found that 210 of 229 serotype V GBS strains (92%) isolated from the bloodstream of nonpregnant adults in the United States and Canada between 1992 and 2013 were multilocus sequence type (ST) 1. Elucidation of the complete genome of a 1992 ST-1 strain revealed that this strain had the highest homology with a GBS strain causing cow mastitis and that the 1992 ST-1 strain differed from serotype V strains isolated in the late 1970s by acquisition of cell surface proteins and antimicrobial resistance determinants. Whole-genome comparison of 202 invasive ST-1 strains detected significant recombination in only eight strains. The remaining 194 strains differed by an average of 97 SNPs. Phylogenetic analysis revealed a temporally dependent mode of genetic diversification consistent with the emergence in the 1990s of ST-1 GBS as major agents of human disease. Thirty-one loci were identified as being under positive selective pressure, and mutations at loci encoding polysaccharide capsule production proteins, regulators of pilus expression, and two-component gene regulatory systems were shown to affect the bacterial phenotype. These data reveal that phenotypic diversity among ST-1 GBS is mainly driven by small genetic changes rather than extensive recombination, thereby extending knowledge into how pathogens adapt to humans.


PLOS Pathogens | 2011

Distinct single amino acid replacements in the control of virulence regulator protein differentially impact streptococcal pathogenesis.

Nicola Horstmann; Pranoti Sahasrabhojane; Bryce Suber; Muthiah Kumaraswami; Randall J. Olsen; Anthony R. Flores; James M. Musser; Richard G. Brennan; Samuel A. Shelburne

Sequencing of invasive strains of group A streptococci (GAS) has revealed a diverse array of single nucleotide polymorphisms in the gene encoding the control of virulence regulator (CovR) protein. However, there is limited information regarding the molecular mechanisms by which CovR single amino acid replacements impact GAS pathogenesis. The crystal structure of the CovR C-terminal DNA-binding domain was determined to 1.50 Å resolution and revealed a three-stranded β-sheet followed by a winged helix-turn-helix DNA binding motif. Modeling of the CovR protein-DNA complex indicated that CovR single amino acid replacements observed in clinical GAS isolates could directly alter protein-DNA interaction and impact protein structure. Isoallelic GAS strains that varied by a single amino acid replacement in the CovR DNA binding domain had significantly different transcriptomes compared to wild-type and to each other. Similarly, distinct recombinant CovR variants had differential binding affinity for DNA from the promoter regions of several virulence factor-encoding genes. Finally, mice that were challenged with GAS CovR isoallelic strains had significantly different survival times, which correlated with the transcriptome and protein-DNA binding studies. Taken together, these data provide structural and functional insights into the critical and distinct effects of variation in the CovR protein on GAS pathogenesis.


PLOS Pathogens | 2014

Dual-Site Phosphorylation of the Control of Virulence Regulator Impacts Group A Streptococcal Global Gene Expression and Pathogenesis

Nicola Horstmann; Miguel Saldaña; Pranoti Sahasrabhojane; Hui Yao; Xiaoping Su; Erika Thompson; Antonius Koller; Samuel A. Shelburne

Phosphorylation relays are a major mechanism by which bacteria alter transcription in response to environmental signals, but understanding of the functional consequences of bacterial response regulator phosphorylation is limited. We sought to characterize how phosphorylation of the control of virulence regulator (CovR) protein from the major human pathogen group A Streptococcus (GAS) influences GAS global gene expression and pathogenesis. CovR mainly serves to repress GAS virulence factor-encoding genes and has been shown to homodimerize following phosphorylation on aspartate-53 (D53) in vitro. We discovered that CovR is phosphorylated in vivo and that such phosphorylation is partially heat-stable, suggesting additional phosphorylation at non-aspartate residues. Using mass spectroscopy along with targeted mutagenesis, we identified threonine-65 (T65) as an additional CovR phosphorylation site under control of the serine/threonine kinase (Stk). Phosphorylation on T65, as mimicked by the recombinant CovR T65E variant, abolished in vitro CovR D53 phosphorylation. Similarly, isoallelic GAS strains that were either unable to be phosphorylated at D53 (CovR-D53A) or had functional constitutive phosphorylation at T65 (CovR-T65E) had essentially an identical gene repression profile to each other and to a CovR-inactivated strain. However, the CovR-D53A and CovR-T65E isoallelic strains retained the ability to positively influence gene expression that was abolished in the CovR-inactivated strain. Consistent with these observations, the CovR-D53A and CovR-T65E strains were hypervirulent compared to the CovR-inactivated strain in a mouse model of invasive GAS disease. Surprisingly, an isoalleic strain unable to be phosphorylated at CovR T65 (CovR-T65A) was hypervirulent compared to the wild-type strain, as auto-regulation of covR gene expression resulted in lower covR gene transcript and CovR protein levels in the CovR-T65A strain. Taken together, these data establish that CovR is phosphorylated in vivo and elucidate how the complex interplay between CovR D53 activating phosphorylation, T65 inhibiting phosphorylation, and auto-regulation impacts streptococcal host-pathogen interaction.


Molecular Microbiology | 2011

An amino-terminal signal peptide of Vfr protein negatively influences RopB-dependent SpeB expression and attenuates virulence in Streptococcus pyogenes.

Samuel A. Shelburne; Randall J. Olsen; Nishanth Makthal; Nicholas G. Brown; Pranoti Sahasrabhojane; Ebru M. Watkins; Timothy Palzkill; James M. Musser; Muthiah Kumaraswami

Streptococcal pyrogenic exotoxin B (SpeB) is an extracellular cysteine protease that is a critical virulence factor made by the major human pathogen group A Streptococcus (GAS). speB expression is dependent on the regulator of proteinase B (RopB) and is upregulated with increasing cell density and during infection. Because computer modelling suggested significant structural similarity between RopB and peptide‐sensing regulatory proteins made by other Gram‐positive bacteria, we hypothesized that speB expression is influenced by RopB–peptide interactions. Inactivation of the gene (vfr) encoding the virulence factor related (Vfr) protein resulted in increased speB transcript level during the exponential growth phase, whereas provision of only the amino‐terminal region of Vfr comprising the secretion signal sequence in trans restored a wild‐type speB expression profile. Addition of the culture supernatant from a Vfr signal peptide‐expressing GAS strain restored wild‐type speB transcript level to a vfr‐inactivated isogenic mutant strain. A distinct peptide in the Vfr secretion signal sequence specifically bound to recombinant RopB. Finally, overexpression of the Vfr secretion signal sequence significantly decreased speB transcript level and attenuated GAS virulence in two mouse models of invasive infection. Taken together, these data delineate a previously unknown small peptide‐mediated regulatory system that controls GAS virulence factor production.


Clinical Infectious Diseases | 2014

Development and Validation of a Clinical Model to Predict the Presence of β-Lactam Resistance in Viridans Group Streptococci Causing Bacteremia in Neutropenic Cancer Patients

Samuel A. Shelburne; Robert E. Lasky; Pranoti Sahasrabhojane; Jeffrey T. Tarrand; Kenneth V. I. Rolston

BACKGROUND Concern for serious infection due to β-lactam-resistant viridans group streptococci (VGS) is a major factor driving empiric use of an anti-gram-positive antimicrobial in patients with febrile neutropenia. We sought to develop and validate a prediction model for the presence of β-lactam resistance in VGS causing bloodstream infection (BSI) in neutropenic patients. METHODS Data from 569 unique cases of VGS BSI in neutropenic patients from 2000 to 2010 at the MD Anderson Cancer Center were used to develop the clinical prediction model. Validation was done using 163 cases from 2011 to 2013. In vitro activity of β-lactam agents was determined for 2011-2013 VGS bloodstream isolates. RESULTS In vitro resistance to β-lactam agents commonly used in the empiric treatment of febrile neutropenia was observed only for VGS isolates with a penicillin minimum inhibitory concentration (MIC) of ≥ 2 µg/mL. One hundred twenty-nine of 732 patients (17%) were infected with VGS strains with a penicillin MIC ≥ 2 µg/mL. For the derivation and validation cohorts, 98% of patients infected by VGS with a penicillin MIC of ≥ 2 µg/mL had at least 1 of the following risk factors: current use of a β-lactam as antimicrobial prophylaxis, receipt of a β-lactam antimicrobial in the previous 30 days, or nosocomial VGS BSI onset. Limiting empiric anti-gram-positive therapy to neutropenic patients having at least 1 of these 3 risk factors would have reduced such use by 42%. CONCLUSIONS Simple clinical criteria can assist with targeting of anti-gram-positive therapy to febrile neutropenic patients at risk of serious β-lactam-resistant VGS infection.


Cancer | 2016

The role of the gastrointestinal microbiome in infectious complications during induction chemotherapy for acute myeloid leukemia

Jessica Galloway-Peña; Daniel P. Smith; Pranoti Sahasrabhojane; Nadim J. Ajami; W. Duncan Wadsworth; Naval Daver; Roy F. Chemaly; Lisa Marsh; Shashank S. Ghantoji; Naveen Pemmaraju; Guillermo Garcia-Manero; Katayoun Rezvani; Amin M. Alousi; Jennifer A. Wargo; Elizabeth J. Shpall; Phillip Andrew Futreal; Michele Guindani; Joseph F. Petrosino; Dimitrios P. Kontoyiannis; Samuel A. Shelburne

Despite increasing data on the impact of the microbiome on cancer, the dynamics and role of the microbiome in infection during therapy for acute myelogenous leukemia (AML) are unknown. Therefore, the authors sought to determine correlations between microbiome composition and infectious outcomes in patients with AML who were receiving induction chemotherapy (IC).


Infection and Immunity | 2015

Characterization of the Effect of the Histidine Kinase CovS on Response Regulator Phosphorylation in Group A Streptococcus

Nicola Horstmann; Pranoti Sahasrabhojane; Miguel Saldaña; Nadim J. Ajami; Anthony R. Flores; Paul Sumby; Chang Gong Liu; Hui Yao; Xiaoping Su; Erika Thompson; Samuel A. Shelburne

ABSTRACT Two-component gene regulatory systems (TCSs) are a major mechanism by which bacteria respond to environmental stimuli and thus are critical to infectivity. For example, the control of virulence regulator/sensor kinase (CovRS) TCS is central to the virulence of the major human pathogen group A Streptococcus (GAS). Here, we used a combination of quantitative in vivo phosphorylation assays, isoallelic strains that varied by only a single amino acid in CovS, and transcriptome analyses to characterize the impact of CovS on CovR phosphorylation and GAS global gene expression. We discovered that CovS primarily serves to phosphorylate CovR, thereby resulting in the repression of virulence factor-encoding genes. However, a GAS strain selectively deficient in CovS phosphatase activity had a distinct transcriptome relative to that of its parental strain, indicating that both CovS kinase and phosphatase activities influence the CovR phosphorylation status. Surprisingly, compared to a serotype M3 strain, serotype M1 GAS strains had high levels of phosphorylated CovR, low transcript levels of CovR-repressed genes, and strikingly different responses to environmental cues. Moreover, the inactivation of CovS in the serotype M1 background resulted in a greater decrease in phosphorylated CovR levels and a greater increase in the transcript levels of CovR-repressed genes than did CovS inactivation in a serotype M3 strain. These data clarify the influence of CovS on the CovR phosphorylation status and provide insight into why serotype M1 GAS strains have high rates of spontaneous mutations in covS during invasive GAS infection, thus providing a link between TCS molecular function and the epidemiology of deadly bacterial infections.

Collaboration


Dive into the Pranoti Sahasrabhojane's collaboration.

Top Co-Authors

Avatar

Samuel A. Shelburne

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jessica Galloway-Peña

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Anthony R. Flores

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Nicola Horstmann

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Miguel Saldaña

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Erika Thompson

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hui Yao

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

James M. Musser

Houston Methodist Hospital

View shared research outputs
Top Co-Authors

Avatar

Nadim J. Ajami

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Randall J. Olsen

Houston Methodist Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge