Kithiganahalli Narayanaswamy Balaji
Indian Institute of Science
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Hotspot
Dive into the research topics where Kithiganahalli Narayanaswamy Balaji is active.
Publication
Featured researches published by Kithiganahalli Narayanaswamy Balaji.
The Lancet Respiratory Medicine | 2014
Stefan H. E. Kaufmann; Christoph Lange; Martin Rao; Kithiganahalli Narayanaswamy Balaji; Michael T. Lotze; Marco Schito; Alimuddin Zumla; Markus Maeurer
Tuberculosis continues to kill 1·4 million people annually. During the past 5 years, an alarming increase in the number of patients with multidrug-resistant tuberculosis and extensively drug-resistant tuberculosis has been noted, particularly in eastern Europe, Asia, and southern Africa. Treatment outcomes with available treatment regimens for drug-resistant tuberculosis are poor. Although substantial progress in drug development for tuberculosis has been made, scientific progress towards development of interventions for prevention and improvement of drug treatment outcomes have lagged behind. Innovative interventions are therefore needed to combat the growing pandemic of multidrug-resistant and extensively drug-resistant tuberculosis. Novel adjunct treatments are needed to accomplish improved cure rates for multidrug-resistant and extensively drug-resistant tuberculosis. A novel, safe, widely applicable, and more effective vaccine against tuberculosis is also desperately sought to achieve disease control. The quest to develop a universally protective vaccine for tuberculosis continues. So far, research and development of tuberculosis vaccines has resulted in almost 20 candidates at different stages of the clinical trial pipeline. Host-directed therapies are now being developed to refocus the anti-Mycobacterium tuberculosis-directed immune responses towards the host; a strategy that could be especially beneficial for patients with multidrug-resistant tuberculosis or extensively drug-resistant tuberculosis. As we are running short of canonical tuberculosis drugs, more attention should be given to host-directed preventive and therapeutic intervention measures.
Infection and Immunity | 2008
Kanhu Charan Mishra; Chantal de Chastellier; Yeddula Narayana; Pablo Bifani; Alistair K. Brown; Gurdyal S. Besra; Vishwa Mohan Katoch; Beenu Joshi; Kithiganahalli Narayanaswamy Balaji; Laurent Kremer
ABSTRACT PE and PPE proteins appear to be important for virulence and immunopathogenicity in mycobacteria, yet the functions of the PE/PPE domains remain an enigma. To decipher the role of these domains, we have characterized the triacylglycerol (TAG) hydrolase LipY from Mycobacterium tuberculosis, which is the only known PE protein expressing an enzymatic activity. The overproduction of LipY in mycobacteria resulted in a significant reduction in the pool of TAGs, consistent with the lipase activity of this enzyme. Unexpectedly, this reduction was more pronounced in mycobacteria overexpressing LipY lacking the PE domain [LipY(ΔPE)], suggesting that the PE domain participates in the modulation of LipY activity. Interestingly, Mycobacterium marinum contains a protein homologous to LipY, termed LipYmar, in which the PE domain is substituted by a PPE domain. As for LipY, overexpression of LipYmar in Mycobacterium smegmatis significantly reduced the TAG pool, and this was further pronounced when the PPE domain of LipYmar was removed. Fractionation studies and Western blot analysis demonstrated that both LipY and LipY(ΔPE) were mainly present in the cell wall, indicating that the PE domain was not required for translocation to this site. Furthermore, electron microscopy immunolabeling of LipY(ΔPE) clearly showed a cell surface localization, thereby suggesting that the lipase may interact with the host immune system. Accordingly, a strong humoral response against LipY and LipY(ΔPE) was observed in tuberculosis patients. Together, our results suggest for the first time that both PE and PPE domains can share similar functional roles and that LipY represents a novel immunodominant antigen.
Journal of Immunology | 2010
Kushagra Bansal; Sri Ramulu Elluru; Yeddula Narayana; Rashmi Chaturvedi; Shripad A. Patil; Srini V. Kaveri; Jagadeesh Bayry; Kithiganahalli Narayanaswamy Balaji
Mycobacterium tuberculosis, the causative agent of pulmonary tuberculosis, infects one-third of the world’s population. Activation of host immune responses for containment of mycobacterial infections involves participation of innate immune cells, such as dendritic cells (DCs). DCs are sentinels of the immune system and are important for eliciting both primary and secondary immune responses to pathogens. In this context, to understand the molecular pathogenesis of tuberculosis and host response to mycobacteria and to conceive prospective vaccine candidates, it is important to understand how cell wall Ags of M. tuberculosis and, in particular, the proline-glutamic acid_polymorphic guanine-cytosine–rich sequence (PE_PGRS) family of proteins modulate DC maturation and function. In this study, we demonstrate that two cell wall-associated/secretory PE_PGRS proteins, PE_PGRS 17 (Rv0978c) and PE_PGRS 11 (Rv0754), recognize TLR2, induce maturation and activation of human DCs, and enhance the ability of DCs to stimulate CD4+ T cells. We further found that PE_PGRS protein-mediated activation of DCs involves participation of ERK1/2, p38 MAPK, and NF-κB signaling pathways. Priming of human DCs with IFN-γ further augmented PE_PGRS 17 or PE_PGRS 11 Ag-induced DC maturation and secretion of key proinflammatory cytokines. Our results suggest that by activating DCs, PE_PGRS proteins, important mycobacterial cell wall Ags, could potentially contribute in the initiation of innate immune responses during tuberculosis infection and hence regulate the clinical course of tuberculosis.
Journal of Biological Chemistry | 2008
Yeddula Narayana; Kithiganahalli Narayanaswamy Balaji
Suppressor of cytokine signaling (SOCS) 3 is a critical negative regulator of cytokine signaling and is induced by Mycobacterium bovis Bacille Calmette-Guérin (M. bovis BCG) in mouse macrophages. However, little is known about the early receptor proximal signaling mechanisms underlying mycobacteria-mediated induction of SOCS3. We demonstrate here for the first time that M. bovis BCG up-regulates NOTCH1 and activates the NOTCH1 signaling pathway, leading to the expression of SOCS3. We show that perturbing Notch signaling in infected macrophages results in the marked reduction in the expression of SOCS3. Furthermore, enforced expression of the Notch1 intracellular domain in RAW 264.7 macrophages induces the expression of SOCS3, which can be further potentiated by M. bovis BCG. The perturbation of Toll-like receptor (TLR) 2 signaling resulted in marked reduction in SOCS3 levels and expression of the NOTCH1 target gene, Hes1. The down-regulation of MyD88 resulted in a significant decrease in SOCS3 expression, implicating the role of the TLR2-MyD88 axis in M. bovis BCG-triggered signaling. However, the SOCS3 inducing ability of M. bovis BCG remains unaltered also upon infection of macrophages from TLR4-defective C3H/HeJ mice. More importantly, signaling perturbation data suggest the involvement of cross-talk among members of the phosphoinositide 3-kinase and mitogen-activated protein kinase cascades with NOTCH1 signaling in SOCS3 expression. Furthermore, SOCS3 expression requires the NOTCH1-mediated recruitment of Suppressor of Hairless (CSL) and nuclear factor-κBtothe Socs3 promoter. Overall, these results implicate NOTCH1 signaling during inducible expression of SOCS3 following infection of macrophages with an intracellular bacillus-like M. bovis BCG.
Molecular and Cellular Biology | 2012
Devram Sampat Ghorpade; Rebecca Leyland; Mariola Kurowska-Stolarska; Shripad A. Patil; Kithiganahalli Narayanaswamy Balaji
ABSTRACT Pathogenic mycobacteria, including Mycobacterium tuberculosis and Mycobacterium bovis, cause significant morbidity and mortality worldwide. However, the vaccine strain Mycobacterium bovis BCG, unlike virulent strains, triggers extensive apoptosis of infected macrophages, a step necessary for the elicitation of robust protective immunity. We here demonstrate that M. bovis BCG triggers Toll-like receptor 2 (TLR2)-dependent microRNA-155 (miR-155) expression, which involves signaling cross talk among phosphatidylinositol 3-kinase (PI3K), protein kinase Cδ (PKCδ), and mitogen-activated protein kinases (MAPKs) and recruitment of NF-κB and c-ETS to miR-155 promoter. Genetic and signaling perturbations presented the evidence that miR-155 regulates PKA signaling by directly targeting a negative regulator of PKA, protein kinase inhibitor alpha (PKI-α). Enhanced activation of PKA signaling resulted in the generation of PKA C-α; phosphorylation of MSK1, cyclic AMP response element binding protein (CREB), and histone H3; and recruitment of phospho-CREB to the apoptotic gene promoters. The miR-155-triggered activation of caspase-3, BAK1, and cytochrome c translocation involved signaling integration of MAPKs and epigenetic or posttranslational modification of histones or CREB. Importantly, M. bovis BCG infection-induced apoptosis was severely compromised in macrophages derived from miR-155 knockout mice. Gain-of-function and loss-of-function studies validated the requirement of miR-155 for M. bovis BCGs ability to trigger apoptosis. Overall, M. bovis BCG-driven miR-155 dictates cell fate decisions of infected macrophages, strongly implicating a novel role for miR-155 in orchestrating cellular reprogramming during immune responses to mycobacterial infection.
Journal of Biological Chemistry | 2010
Kushagra Bansal; Akhauri Yash Sinha; Devram Sampat Ghorpade; Shambhuprasad Kotresh Togarsimalemath; Shripad A. Patil; Srini V. Kaveri; Kithiganahalli Narayanaswamy Balaji; Jagadeesh Bayry
Mycobacterium tuberculosis, an etiological agent of pulmonary tuberculosis, causes significant morbidity and mortality worldwide. Pathogenic mycobacteria survive in the host by subverting host innate immunity. Dendritic cells (DCs) are professional antigen-presenting cells that are vital for eliciting immune responses to infectious agents, including pathogenic mycobacteria. DCs orchestrate distinct Th responses based on the signals they receive. In this perspective, deciphering the interactions of the proline-glutamic acid/proline-proline-glutamic acid (PE/PPE) family of proteins of M. tuberculosis with DCs assumes significant pathophysiological attributes. In this study, we demonstrate that Rv1917c (PPE34), a representative member of the proline-proline-glutamic-major polymorphic tandem repeat family, interacts with TLR2 and triggers functional maturation of human DCs. Signaling perturbations implicated a critical role for integrated cross-talk among PI3K-MAPK and NF-κB signaling cascades in Rv1917c-induced maturation of DCs. However, this maturation of DCs was associated with a secretion of high amounts of anti-inflammatory cytokine IL-10, whereas Th1-polarizing cytokine IL-12 was not induced. Consistent with these results, Rv1917c-matured DCs favored secretion of IL-4, IL-5, and IL-10 from CD4+ T cells and contributed to Th2-skewed cytokine balance ex vivo in healthy individuals and in patients with pulmonary tuberculosis. Interestingly, the Rv1917c-skewed Th2 immune response involved induced expression of cyclooxygenase-2 (COX-2) in DCs. Taken together, these results indicate that Rv1917c facilitates a shift in the ensuing immunity toward the Th2 phenotype and could aid in immune evasion by mycobacteria.
PLOS ONE | 2009
Kushagra Bansal; Nisha Kapoor; Yeddula Narayana; Germain Puzo; Martine Gilleron; Kithiganahalli Narayanaswamy Balaji
Activation of inflammatory immune responses during granuloma formation by the host upon infection of mycobacteria is one of the crucial steps that is often associated with tissue remodeling and breakdown of the extracellular matrix. In these complex processes, cyclooxygenase-2 (COX-2) plays a major role in chronic inflammation and matrix metalloproteinase-9 (MMP-9) significantly in tissue remodeling. In this study, we investigated the molecular mechanisms underlying Phosphatidyl-myo-inositol dimannosides (PIM2), an integral component of the mycobacterial envelope, triggered COX-2 and MMP-9 expression in macrophages. PIM2 triggers the activation of Phosphoinositide-3 Kinase (PI3K) and Notch1 signaling leading to COX-2 and MMP-9 expression in a Toll-like receptor 2 (TLR2)-MyD88 dependent manner. Notch1 signaling perturbations data demonstrate the involvement of the cross-talk with members of PI3K and Mitogen activated protein kinase pathway. Enforced expression of the cleaved Notch1 in macrophages induces the expression of COX-2 and MMP-9. PIM2 triggered significant p65 nuclear factor -κB (NF-κB) nuclear translocation that was dependent on activation of PI3K or Notch1 signaling. Furthermore, COX-2 and MMP-9 expression requires Notch1 mediated recruitment of Suppressor of Hairless (CSL) and NF-κB to respective promoters. Inhibition of PIM2 induced COX-2 resulted in marked reduction in MMP-9 expression clearly implicating the role of COX-2 dependent signaling events in driving the MMP-9 expression. Taken together, these data implicate PI3K and Notch1 signaling as obligatory early proximal signaling events during PIM2 induced COX-2 and MMP-9 expression in macrophages.
Journal of Biological Chemistry | 2010
Rashmi Chaturvedi; Kushagra Bansal; Yeddula Narayana; Nisha Kapoor; Namineni Sukumar; Shambhuprasad Kotresh Togarsimalemath; Nagasuma Chandra; Saurabh Mishra; Parthasarathi Ajitkumar; Beenu Joshi; Vishwa Mohan Katoch; Shripad A. Patil; Kithiganahalli Narayanaswamy Balaji
Mycobacterium tuberculosis utilizes unique strategies to survive amid the hostile environment of infected host cells. Infection-specific expression of a unique mycobacterial cell surface antigen that could modulate key signaling cascades can act as a key survival strategy in curtailing host effector responses like oxidative stress. We demonstrate here that hypothetical PE_PGRS11 ORF encodes a functional phosphoglycerate mutase. The transcriptional analysis revealed that PE_PGRS11 is a hypoxia-responsive gene, and enforced expression of PE_PGRS11 by recombinant adenovirus or Mycobacterium smegmatis imparted resistance to alveolar epithelial cells against oxidative stress. PE_PGRS11-induced resistance to oxidative stress necessitated the modulation of genetic signatures like induced expression of Bcl2 or COX-2. This modulation of specific antiapoptotic molecular signatures involved recognition of PE_PGRS11 by TLR2 and subsequent activation of the PI3K-ERK1/2-NF-κB signaling axis. Furthermore, PE_PGRS11 markedly diminished H2O2-induced p38 MAPK activation. Interestingly, PE_PGRS11 protein was exposed at the mycobacterial cell surface and was involved in survival of mycobacteria under oxidative stress. Furthermore, PE_PGRS11 displayed differential B cell responses during tuberculosis infection. Taken together, our investigation identified PE_PGRS11 as an in vivo expressed immunodominant antigen that plays a crucial role in modulating cellular life span restrictions imposed during oxidative stress by triggering TLR2-dependent expression of COX-2 and Bcl2. These observations clearly provide a mechanistic basis for the rescue of pathogenic Mycobacterium-infected lung epithelial cells from oxidative stress.
Journal of Biological Chemistry | 2013
Devram Sampat Ghorpade; Akhuri Yash Sinha; Sahana Holla; Vikas K. Singh; Kithiganahalli Narayanaswamy Balaji
Background: Genetic variants of NOD2 are linked to inflammatory bowel disease (IBD) etiology. Results: DSS model of colitis in wild-type and inducible nitric-oxide synthase (iNOS) null mice revealed that NOD2-iNOS/NO-responsive microRNA-146a targets NUMB gene facilitating Sonic hedgehog (SHH) signaling. Conclusion: miR-146a-mediated NOD2-SHH signaling regulates gut inflammation. Significance: Identification of novel regulators of IBD provides new insights into pathophysiology and development of new therapy concepts. Inflammatory bowel disease (IBD) is a debilitating chronic inflammatory disorder of the intestine. The interactions between enteric bacteria and genetic susceptibilities are major contributors of IBD etiology. Although genetic variants with loss or gain of NOD2 functions have been linked to IBD susceptibility, the mechanisms coordinating NOD2 downstream signaling, especially in macrophages, during IBD pathogenesis are not precisely identified. Here, studies utilizing the murine dextran sodium sulfate model of colitis revealed the crucial roles for inducible nitric-oxide synthase (iNOS) in regulating pathophysiology of IBDs. Importantly, stimulation of NOD2 failed to activate Sonic hedgehog (SHH) signaling in iNOS null macrophages, implicating NO mediated cross-talk between NOD2 and SHH signaling. NOD2 signaling up-regulated the expression of a NO-responsive microRNA, miR-146a, that targeted NUMB gene and alleviated the suppression of SHH signaling. In vivo and ex vivo studies confirmed the important roles for miR-146a in amplifying inflammatory responses. Collectively, we have identified new roles for miR-146a that established novel cross-talk between NOD2-SHH signaling during gut inflammation. Potential implications of these observations in therapeutics could increase the possibility of defining and developing better regimes to treat IBD pathophysiology.
The Journal of Infectious Diseases | 2012
Jamma Trinath; Mohan S. Maddur; Srini V. Kaveri; Kithiganahalli Narayanaswamy Balaji; Jagadeesh Bayry
To the Editor—CD4CD25FoxP3 regulatory T cells (Tregs) are critical for the maintenance of immune tolerance by suppressing immune cell functions. In the context of infection such as Mycobacterium tuberculosis, Tregs can also suppress efficient protective immune responses against the pathogen and its effective clearance [1]. Results from studies of patients with tuberculosis and experimental models have shown that Tregs are expanded and accumulated at the site of infection. These Tregs efficiently inhibit the arrival of effector T cells in the lungs, production of interferon (IFN)–c and cd T-cell responses to M. tuberculosis [2–4]. However, mechanisms underlying the expansion of Tregs are not clear. Periasamy et al [5] identified that Treg expansion by M. tuberculosis requires expression of the protein programmed death-1 (PD-1, CD279). Inhibition of PD-1 by blocking antibodies or small interfering RNA prevented the Treg expansion. However, the questions that remain unanswered are as follows: does M. tuberculosis trigger dynamic changes in the expression of signaling molecules on innate immune cells that, in turn, facilitate Treg expansion, and if so, which innate pattern recognition receptors are implicated in this process? Because Treg expansion in the periphery requires costimulatory signals and antigen presentation by antigen-presenting cells such as dendritic cells (DCs), we surmised that the Treg expansion observed by the Periasamy et al [5] might reflect the modulation of DCs by M. tuberculosis. After education by M. tuberculosis, these DCs then might provide signals for expansion of Tregs. To address our hypothesis, immature DCs were derived by culturing peripheral blood monocytes (isolated by using CD14 microbeads; Miltenyi Biotec) from healthy blood donors after ethical committee permission, in the presence of granulocyte microphage colonystimulating factor (GM-CSF; 1000 IU/10 cells) and IL-4 (500 IU/10 cells). The 5-day-old immature DCs (0.5x10/ml) were either cultured in the presence of cytokines alone or cytokines plus whole cell lysates (WCLs) of pathogenic M. tuberculosis H37Rv (5 microgram/mL) for 48 h. The DCs were washed thoroughly and were cocultured with 0.2 3 106 CD4 1 T cells at 1:10 ratio for 96 h. CD4 T cells were isolated by using CD4 microbeads. The DCs and CD4 T cells were from unrelated donors, and hence CD3 and CD28 stimulation was not provided. The frequency of CD4CD25FoxP3 Tregs was analyzed using flow cytometry (LSR II, BD Biosciences). Surface staining was performed with fluorochrome-conjugated monoclonal antibodies (MAbs) to CD4 and CD25 (BD Biosciences), and intracellular staining for FoxP3 was performed using fluorochrome-conjugated MAb to FoxP3 and kit from eBioscience. We found that M. tuberculosis–treated DCs significantly expand Tregs (3.5% 6 0.5% in control vs 6.6% 6 0.8% in M. tuberculosis–treated DCs; n 5 5; Figure 1A). The results suggest that the Treg expansion observed by Periasamy et al [5] was indeed attributable to modulation of DC (or innate immune cells in general) functions by M. tuberculosis.