Network


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

Hotspot


Dive into the research topics where Sultan Tousif is active.

Publication


Featured researches published by Sultan Tousif.


Journal of Biological Chemistry | 2013

Mycobacterium tuberculosis Controls MicroRNA-99b (miR-99b) Expression in Infected Murine Dendritic Cells to Modulate Host Immunity

Yogesh Singh; Vandana Kaul; Alka Mehra; Samit Chatterjee; Sultan Tousif; Ved Prakash Dwivedi; Mrutyunjay Suar; Luc Van Kaer; William R. Bishai; Gobardhan Das

Background: Modulation of host miRNAs coincides with increased pathogenicity in various infectious diseases. Results: miR-99b is up-regulated in M. tuberculosis-infected dendritic cells, which inhibits production of proinflammatory cytokines. Conclusion: Our findings unfold a novel immune evasion strategy of M. tuberculosis by modulating miRNAs. Significance: Our study opens up the possibility to design vaccines and immunotherapies for tuberculosis by targeting specific miRNAs. Mycobacterium tuberculosis resides and replicates within host phagocytes by modulating host microbicidal responses. In addition, it suppresses the production of host protective cytokines to prevent activation of and antigen presentation by M. tuberculosis-infected cells, causing dysregulation of host protective adaptive immune responses. Many cytokines are regulated by microRNAs (miRNAs), a newly discovered class of small noncoding RNAs, which have been implicated in modulating host immune responses in many bacterial and viral diseases. Here, we show that miRNA-99b (miR-99b), an orphan miRNA, plays a key role in the pathogenesis of M. tuberculosis infection. We found that miR-99b expression was highly up-regulated in M. tuberculosis strain H37Rv-infected dendritic cells (DCs) and macrophages. Blockade of miR-99b expression by antagomirs resulted in significantly reduced bacterial growth in DCs. Interestingly, knockdown of miR-99b in DCs significantly up-regulated proinflammatory cytokines such as IL-6, IL-12, and IL-1β. Furthermore, mRNA and membrane-bound protein data indicated that inhibition of miR-99b augments TNF-α and TNFRSF-4 production. Thus, miR-99b targets TNF-α and TNFRSF-4 receptor genes. Treatment of anti-miR-99b-transfected DCs with anti-TNF-α antibody resulted in increased bacterial burden. Thus, our findings unveil a novel host evasion mechanism adopted by M. tuberculosis via miR-99b, which may open up new avenues for designing miRNA-based vaccines and therapies.


PLOS ONE | 2011

T Cells from Programmed Death-1 Deficient Mice Respond Poorly to Mycobacterium tuberculosis Infection

Sultan Tousif; Yogesh Singh; Durbaka V. R. Prasad; Pawan Sharma; Luc Van Kaer; Gobardhan Das

Background Programmed Death-1 (PD-1; CD279) receptor molecule is widely believed to be a negative regulator predominantly expressed by exhausted/activated mouse T cells. Upon interaction with its ligands, PD-L1 and PD-L2, PD-1 inhibits activation of T cells and cytokine production, which has been documented in various viral and fungal infections as well as in vitro studies. Therefore, inhibition of T cell responses by PD-1 resulted in disease resistance in a variety of mouse infection models studied heretofore. Methodology/Principal Findings Here, we report that PD-1 deficient (PD-1−/−) mice infected with Mycobacterium tuberculosis (M. tb) H37Rv by the aerosol route have increased susceptibility as compared with their wild type littermates. Surprisingly, M. tb antigen-specific T cell proliferation was dramatically reduced in PD-1 deficient animals compared with wild-type littermates, and this was due to increased numbers of regulatory T cells (Tregs) and recruitment of mesenchymal stem cells. Furthermore, PD-1−/− mice exhibited decreases in the autophagy-induced LC3-B marker protein in macrophages. Conclusions/Significance Our findings suggest that PD-1 does not play an inhibitory role during M. tb infection and instead promotes mycobacterial clearance in mice.


Nature Communications | 2015

Host ICAMs play a role in cell invasion by Mycobacterium tuberculosis and Plasmodium falciparum

Kuhulika Bhalla; Monika Chugh; Sonali Mehrotra; Sumit Rathore; Sultan Tousif; Ved Prakash Dwivedi; Prem Prakash; Sachin K. Samuchiwal; Sushil Kumar; Dhiraj Kumar Singh; Swapnil Ghanwat; Dhiraj Kumar; Gobardhan Das; Asif Mohmmed; Pawan Malhotra; Anand Ranganathan

Intercellular adhesion molecules (ICAMs) belong to the immunoglobulin superfamily and participate in diverse cellular processes including host-pathogen interactions. ICAM-1 is expressed on various cell types including macrophages, whereas ICAM-4 is restricted to red blood cells. Here we report the identification of an 11-kDa synthetic protein, M5, that binds to human ICAM-1 and ICAM-4, as shown by in vitro interaction studies, surface plasmon resonance and immunolocalization. M5 greatly inhibits the invasion of macrophages and erythrocytes by Mycobacterium tuberculosis and Plasmodium falciparum, respectively. Pharmacological and siRNA-mediated inhibition of ICAM-1 expression also results in reduced M. tuberculosis invasion of macrophages. ICAM-4 binds to P. falciparum merozoites, and the addition of recombinant ICAM-4 to parasite cultures blocks invasion of erythrocytes by newly released merozoites. Our results indicate that ICAM-1 and ICAM-4 play roles in host cell invasion by M. tuberculosis and P. falciparum, respectively, either as receptors or as crucial accessory molecules.


European Journal of Immunology | 2013

Mesenchymal stem cells play an important role in host protective immune responses against malaria by modulating regulatory T cells.

Reva S. Thakur; Sultan Tousif; Vikky Awasthi; Anirban Sanyal; P. K. Atul; Parveen Punia; Jyoti Das

Plasmodium spp. parasites, the causative agents of malaria, survive and replicate in human hosts by modulating host protective immune responses. In a rodent model, malaria manifests as a severe splenomegaly, with infiltration of cells and lympho‐proliferation as major contributing factors of the immunopathology. However, the cellular contents and the functions of these cells have not been well studied. Here, we report that Plasmodium berghei infection of mice leads to massive recruitment of mesen‐chymal stem cells (MSCs) in secondary lymphoid organs. Infusion of these cells into naïve mice was able to confer host resistance against malaria. Furthermore, MSCs augmented interleukin (IL)‐12 production but suppressed IL‐10 production in recipient animals. In addition, we observed dramatic reductions of regulatory T (Treg) cells in animals that received MSCs. Taken together, our findings have identified recruitment of MSCs as a novel host protective mechanism adopted by the host to combat malaria by modulating Treg‐cell responses.


Journal of Biological Chemistry | 2012

Transforming growth factor-β protein inversely regulates in vivo differentiation of interleukin-17 (IL-17)-producing CD4+ and CD8+ T cells.

Ved Prakash Dwivedi; Sultan Tousif; Debapriya Bhattacharya; Durbaka Vijay Raghva Prasad; Luc Van Kaer; Jyoti Das; Gobardhan Das

Background: In vitro TGF-β differentiates Th17 and Tc17 cells, but TGF-βRIIDN mice display multiorgan autoimmune disorders. Results: CD4+T cells from TGF-βRIIDN mice are resistant to Th17 cell differentiation, whereas CD8+T cells acquire IL-17-producing phenotype, and IL-17 neutralization or depletion inhibited inflammation in TGF-βRIIDN mice. Conclusion: Tc17 cell differentiations in vivo are distinct from Tc17 cell differentiations in vitro. Significance: The Tc17 cell differentiation program is unique to Th17. TGF-β is a pleiotropic cytokine that predominantly exerts inhibitory functions in the immune system. Unexpectedly, the in vitro differentiation of both Th17 and Tc17 cells requires TGF-β. However, animals that are impaired in TGF-β signaling (TGF-βRIIDN mice) display multiorgan autoimmune disorders. Here we show that CD4+ T cells from TGF-βRIIDN mice are resistant to Th17 cell differentiation and, paradoxically, that CD8+ T cells from these animals spontaneously acquire an IL-17-producing phenotype. Neutralization of IL-17 or depletion of CD8+ T cells dramatically inhibited inflammation in TGF-βRIIDN mice. Therefore, the absence of TGF-β triggers spontaneous differentiation of IL-17-producing CD8+ T cells, suggesting that the in vivo and in vitro conditions that promote the differentiation of IL-17-producing CD8+ T cells are distinct.


Journal of Biological Chemistry | 2014

Isoniazid Induces Apoptosis Of Activated CD4+ T Cells IMPLICATIONS FOR POST-THERAPY TUBERCULOSIS REACTIVATION AND REINFECTION

Sultan Tousif; Dhiraj Kumar Singh; Shaheer Ahmad; Prashini Moodley; Maitree Bhattacharyya; Luc Van Kaer; Gobardhan Das

Background: DOTS-treated patients exhibit vulnerability to reactivation and reinfection with TB, suggesting therapy-related immune impairment. Results: Isoniazid (INH) dramatically reduces Mycobacterium tuberculosis antigen-specific immune responses by inducing apoptosis in activated CD4+ T cells. Conclusion: Animals treated with INH exhibit post-treatment vulnerability to TB reinfection and reactivation. Significance: The immune-impairing effects of antibiotics need to be considered in TB treatment. Tuberculosis (TB) remains the second highest killer from a single infectious disease worldwide. Current therapy of TB is lengthy and consists of multiple expensive antibiotics, in a strategy referred to as Directly Observed Treatment, Short Course (DOTS). Although this therapy is effective, it has serious disadvantages. These therapeutic agents are toxic and are associated with the development of a variety of drug-resistant TB strains. Furthermore, patients treated with DOTS exhibit enhanced post-treatment susceptibility to TB reactivation and reinfection, suggesting therapy-related immune impairment. Here we show that Isoniazid (INH) treatment dramatically reduces Mycobacterium tuberculosis antigen-specific immune responses, induces apoptosis in activated CD4+ T cells, and renders treated animals vulnerable to TB reactivation and reinfection. Consequently, our findings suggest that TB treatment is associated with immune impairment.


Journal of Biological Chemistry | 2012

CD4+ T Cell-derived Novel Peptide Thp5 Induces Interleukin-4 Production in CD4+ T Cells to Direct T Helper 2 Cell Differentiation

Mohd Moin Khan; Samit Chatterjee; Ved Prakash Dwivedi; Nishant Kumar Pandey; Yogesh Singh; Sultan Tousif; Neel Sarovar Bhavesh; Luc Van Kaer; Jyoti Das; Gobardhan Das

Background: CD4+ T cells produce IL-4 that drives Th2 cell differentiation. Early production of IL-4 in naïve T cells leads to Th2 cell differentiation. Results: Thp5, a novel peptide, regulates IL-4 production in early activated CD4+ T cells. Conclusion: Early activated CD4+ T cells produce Thp5, which plays a critical role in the differentiation of Th2 cells. Significance: Thp5 acts as an important determinant of Th2 cell differentiation during early T cell activation. The differentiation of naïve CD4+ T cells into T helper 2 (Th2) cells requires production of the cytokine IL-4 in the local microenvironment. It is evident that naïve/quiescently activated CD4+ T cells produce the IL-4 that drives Th2 cell differentiation. Because early production of IL-4 in naïve T cells leads to preferential Th2 cell differentiation, this process needs to be tightly regulated so as to avoid catastrophic and misdirected Th2 cell differentiation. Here, we show that Thp5, a novel peptide with structural similarity to vasoactive intestinal peptide, regulates production of early IL-4 in newly activated CD4+ T cells. Induction of IL-4 in CD4+ T cells by Thp5 is independent of the transcription factor STAT6 but dependent on ERK1/2 signaling. Furthermore, cytokines (IL-12 and TGF-β) that promote the differentiation of Th1 or Th17 cells inhibit Thp5 induction, thus suppressing Th2 cell differentiation. We further showed that Thp5 enhances Th2 responses and exacerbates allergic airway inflammation in mice. Taken together, our findings reveal that early activated CD4+ T cells produce Thp5, which plays a critical role as a molecular switch in the differentiation of Th cells, biasing the response toward the Th2 cell phenotype.


BMC Infectious Diseases | 2014

A peptide fragment from the human COX3 protein disrupts association of Mycobacterium tuberculosis virulence proteins ESAT-6 and CFP10, inhibits mycobacterial growth and mounts protective immune response

Sachin K. Samuchiwal; Sultan Tousif; Dhiraj Kumar Singh; Arun Kumar; Anamika Ghosh; Kuhulika Bhalla; Prem Prakash; Sushil Kumar; Maitree Bhattacharyya; Prashini Moodley; Gobardhan Das; Anand Ranganathan

BackgroundTuberculosis (TB) is one of the most prevalent infectious diseases affecting millions worldwide. The currently available anti-TB drugs and vaccines have proved insufficient to contain this scourge, necessitating an urgent need for identification of novel drug targets and therapeutic strategies. The disruption of crucial protein-protein interactions, especially those that are responsible for virulence in Mycobacterium tuberculosis – for example the ESAT-6:CFP10 complex – are a worthy pursuit in this direction.MethodsWe therefore sought to improvise a method to attenuate M. tuberculosis while retaining the latter’s antigenic properties. We screened peptide libraries for potent ESAT-6 binders capable of dissociating CFP10 from ESAT-6. We assessed the disruption by a peptide named HCL2, of the ESAT-6:CFP10 complex and studied its effects on mycobacterial survival and virulence.ResultsWe found that HCL2, derived from the human cytochrome c oxidase subunit 3 (COX3) protein, disrupts ESAT-6:CFP10 complex, binds ESAT-6 potently, disintegrates bacterial cell wall and inhibits extracellular as well as intracellular mycobacterial growth. In addition, an HCL2 expressing M. tuberculosis strain induces both Th1 and Th17 host protective responses.ConclusionsDisruption of ESAT-6:CFP10 association could, therefore, be an alternate method for attenuating M. tuberculosis, and a possible route towards future vaccine generation.


PLOS ONE | 2013

Expression of the ARPC4 Subunit of Human Arp2/3 Severely Affects Mycobacterium tuberculosis Growth and Suppresses Immunogenic Response in Murine Macrophages

Anamika Ghosh; Sultan Tousif; Debapriya Bhattacharya; Sachin K. Samuchiwal; Kuhulika Bhalla; Megha Tharad; Sushil Kumar; Prem Prakash; Purnima S. Kumar; Gobardhan Das; Anand Ranganathan

Background The search for molecules against Mycobacterium tuberculosis is urgent. The mechanisms facilitating the intra-macrophage survival of Mycobacterium tuberculosis are as yet not entirely understood. However, there is evidence showing the involvement of host cell cytoskeleton in every step of establishment and persistence of mycobacterial infection. Methodology/Principal Findings Here we show that expression of ARPC4, a subunit of the Actin related protein 2/3 (Arp2/3) protein complex, severely affects the pathogen’s growth. TEM studies display shedding of the mycobacterial outer-coat. Furthermore, in infected macrophages, mycobacteria expressing ARPC4 were cleared off at a much faster rate, and were unable to mount a pro-inflammatory cytokine response. The translocation of ARPC4-expressing mycobacteria to the lysosome of the infected macrophage was also impaired. Additionally, the ARPC4 subunit was shown to interact with Rv1626, an essential secretory mycobacterial protein. Real-time PCR analysis showed that upon expression of ARPC4 in mycobacteria, Rv1626 expression is downregulated as much as six-fold. Rv1626 was found to also interact with mammalian cytoskeleton protein, Arp2/3, and enhance the rate of actin polymerization. Conclusions/Significance With crystal structures for Rv1626 and ARPC4 subunit already known, our finding lays out the effect of a novel molecule on mycobacteria, and represents a viable starting point for developing potent peptidomimetics.


Frontiers in Immunology | 2017

Nanoparticle-Formulated Curcumin Prevents Posttherapeutic Disease Reactivation and Reinfection with Mycobacterium tuberculosis following Isoniazid Therapy

Sultan Tousif; Dhiraj Kumar Singh; Sitabja Mukherjee; Shaheer Ahmad; Rakesh Arya; Ranjan Kumar Nanda; Anand Ranganathan; Maitree Bhattacharyya; Luc Van Kaer; Santosh K. Kar; Gobardhan Das

Curcumin, the bioactive component of turmeric also known as “Indian Yellow Gold,” exhibits therapeutic efficacy against several chronic inflammatory and infectious diseases. Even though considered as a wonder drug pertaining to a myriad of reported benefits, the translational potential of curcumin is limited by its low systemic bioavailability due to its poor intestinal absorption, rapid metabolism, and rapid systemic elimination. Therefore, the translational potential of this compound is specifically challenged by bioavailability issues, and several laboratories are making efforts to improve its bioavailability. We developed a simple one-step process to generate curcumin nanoparticles of ~200 nm in size, which yielded a fivefold enhanced bioavailability in mice over regular curcumin. Curcumin nanoparticles drastically reduced hepatotoxicity induced by antitubercular antibiotics during treatment in mice. Most interestingly, co-treatment of nanoparticle-formulated curcumin along with antitubercular antibiotics dramatically reduced the risk for disease reactivation and reinfection, which is the major shortfall of current antibiotic treatment adopted by Directly Observed Treatment Short-course. Furthermore, nanoparticle-formulated curcumin significantly reduced the time needed for antibiotic therapy to obtain sterile immunity, thereby reducing the possibility of generating drug-resistant variants of the organisms. Therefore, adjunct therapy of nano-formulated curcumin with enhanced bioavailability may be beneficial to treatment of tuberculosis and possibly other diseases.

Collaboration


Dive into the Sultan Tousif's collaboration.

Top Co-Authors

Avatar

Gobardhan Das

Jawaharlal Nehru University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anand Ranganathan

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Dhiraj Kumar Singh

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Kuhulika Bhalla

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Ved Prakash Dwivedi

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Prem Prakash

Central Drug Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yogesh Singh

University of Tübingen

View shared research outputs
Researchain Logo
Decentralizing Knowledge