Network


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

Hotspot


Dive into the research topics where Sudha Kumari is active.

Publication


Featured researches published by Sudha Kumari.


Nature Medicine | 2016

Eradication of large established tumors in mice by combination immunotherapy that engages innate and adaptive immune responses

Kelly D. Moynihan; Cary Francis Opel; Gregory L. Szeto; Alice Tzeng; Eric F. Zhu; Jesse M. Engreitz; Robert T. Williams; Kavya Rakhra; Michael H Zhang; Adrienne Rothschilds; Sudha Kumari; Ryan L. Kelly; Byron Hua Kwan; Wuhbet Abraham; Kevin Hu; Naveen K. Mehta; Monique J. Kauke; Heikyung Suh; Jennifer R. Cochran; Douglas A. Lauffenburger; K. Dane Wittrup; Darrell J. Irvine

Checkpoint blockade with antibodies specific for cytotoxic T lymphocyte–associated protein (CTLA)-4 or programmed cell death 1 (PDCD1; also known as PD-1) elicits durable tumor regression in metastatic cancer, but these dramatic responses are confined to a minority of patients. This suboptimal outcome is probably due in part to the complex network of immunosuppressive pathways present in advanced tumors, which are unlikely to be overcome by intervention at a single signaling checkpoint. Here we describe a combination immunotherapy that recruits a variety of innate and adaptive immune cells to eliminate large tumor burdens in syngeneic tumor models and a genetically engineered mouse model of melanoma; to our knowledge tumors of this size have not previously been curable by treatments relying on endogenous immunity. Maximal antitumor efficacy required four components: a tumor-antigen-targeting antibody, a recombinant interleukin-2 with an extended half-life, anti-PD-1 and a powerful T cell vaccine. Depletion experiments revealed that CD8+ T cells, cross-presenting dendritic cells and several other innate immune cell subsets were required for tumor regression. Effective treatment induced infiltration of immune cells and production of inflammatory cytokines in the tumor, enhanced antibody-mediated tumor antigen uptake and promoted antigen spreading. These results demonstrate the capacity of an elicited endogenous immune response to destroy large, established tumors and elucidate essential characteristics of combination immunotherapies that are capable of curing a majority of tumors in experimental settings typically viewed as intractable.


eLife | 2015

Actin foci facilitate activation of the phospholipase C-γ in primary T lymphocytes via the WASP pathway

Sudha Kumari; David Depoil; Roberta Martinelli; Edward Judokusumo; Guillaume Carmona; Frank B. Gertler; Lance C. Kam; Christopher V. Carman; Janis K. Burkhardt; Darrell J. Irvine; Michael L. Dustin

Wiscott Aldrich Syndrome protein (WASP) deficiency results in defects in calcium ion signaling, cytoskeletal regulation, gene transcription and overall T cell activation. The activation of WASP constitutes a key pathway for actin filament nucleation. Yet, when WASP function is eliminated there is negligible effect on actin polymerization at the immunological synapse, leading to gaps in our understanding of the events connecting WASP and calcium ion signaling. Here, we identify a fraction of total synaptic F-actin selectively generated by WASP in the form of distinct F-actin ‘foci’. These foci are polymerized de novo as a result of the T cell receptor (TCR) proximal tyrosine kinase cascade, and facilitate distal signaling events including PLCγ1 activation and subsequent cytoplasmic calcium ion elevation. We conclude that WASP generates a dynamic F-actin architecture in the context of the immunological synapse, which then amplifies the downstream signals required for an optimal immune response. DOI: http://dx.doi.org/10.7554/eLife.04953.001


Proceedings of the National Academy of Sciences of the United States of America | 2016

Sustained antigen availability during germinal center initiation enhances antibody responses to vaccination

Hok Hei Tam; Mariane B. Melo; Myungsun Kang; Jeisa M. Pelet; Vera M. Ruda; Maria Hottelet Foley; Joyce K. Hu; Sudha Kumari; Jordan Crampton; Alexis D. Baldeon; Rogier W. Sanders; John P. Moore; Shane Crotty; Robert Langer; Daniel G. Anderson; Arup K. Chakraborty; Darrell J. Irvine

Significance We explored the effect of nontraditional vaccine dosing profiles on antibody titers of vaccines and discovered that certain dosing profiles demonstrate >10-fold higher antibody production than the traditional single-dose prime–boost method. We also present a computational model that captures the experimental results and provides a mechanistic understanding of the biology behind the effectiveness of our strategy. This work has clinical significance in vaccine design because it is a simple method to increase the efficacy of subunit vaccines, which may lead to the development of efficacious vaccines for diseases such as HIV. Natural infections expose the immune system to escalating antigen and inflammation over days to weeks, whereas nonlive vaccines are single bolus events. We explored whether the immune system responds optimally to antigen kinetics most similar to replicating infections, rather than a bolus dose. Using HIV antigens, we found that administering a given total dose of antigen and adjuvant over 1–2 wk through repeated injections or osmotic pumps enhanced humoral responses, with exponentially increasing (exp-inc) dosing profiles eliciting >10-fold increases in antibody production relative to bolus vaccination post prime. Computational modeling of the germinal center response suggested that antigen availability as higher-affinity antibodies evolve enhances antigen capture in lymph nodes. Consistent with these predictions, we found that exp-inc dosing led to prolonged antigen retention in lymph nodes and increased Tfh cell and germinal center B-cell numbers. Thus, regulating the antigen and adjuvant kinetics may enable increased vaccine potency.


Journal of Clinical Investigation | 2016

A DOCK8-WIP-WASp complex links T cell receptors to the actin cytoskeleton

Erin Janssen; Mira Tohmé; Mona Hedayat; Marion Leick; Sudha Kumari; Narayanaswamy Ramesh; Michel J. Massaad; Sumana Ullas; Veronica Azcutia; Christopher C. Goodnow; Katrina L. Randall; Qi Qiao; Hao Wu; Waleed Al-Herz; Dianne Cox; John H. Hartwig; Darrell J. Irvine; Francis W. Luscinskas; Raif S. Geha

Wiskott-Aldrich syndrome (WAS) is associated with mutations in the WAS protein (WASp), which plays a critical role in the initiation of T cell receptor-driven (TCR-driven) actin polymerization. The clinical phenotype of WAS includes susceptibility to infection, allergy, autoimmunity, and malignancy and overlaps with the symptoms of dedicator of cytokinesis 8 (DOCK8) deficiency, suggesting that the 2 syndromes share common pathogenic mechanisms. Here, we demonstrated that the WASp-interacting protein (WIP) bridges DOCK8 to WASp and actin in T cells. We determined that the guanine nucleotide exchange factor activity of DOCK8 is essential for the integrity of the subcortical actin cytoskeleton as well as for TCR-driven WASp activation, F-actin assembly, immune synapse formation, actin foci formation, mechanotransduction, T cell transendothelial migration, and homing to lymph nodes, all of which also depend on WASp. These results indicate that DOCK8 and WASp are in the same signaling pathway that links TCRs to the actin cytoskeleton in TCR-driven actin assembly. Further, they provide an explanation for similarities in the clinical phenotypes of WAS and DOCK8 deficiency.


ACS Nano | 2017

Enhancing Adoptive Cell Therapy of Cancer through Targeted Delivery of Small-Molecule Immunomodulators to Internalizing or Noninternalizing Receptors

Yiran Zheng; Li Tang; Llian Mabardi; Sudha Kumari; Darrell J. Irvine

Adoptive cell therapy (ACT) has achieved striking efficacy in B-cell leukemias, but less success treating other cancers, in part due to the rapid loss of ACT T-cell effector function in vivo due to immunosuppression in solid tumors. Transforming growth factor-β (TGF-β) signaling is an important mechanism of immune suppression in the tumor microenvironment, but systemic inhibition of TGF-β is toxic. Here we evaluated the potential of targeting a small molecule inhibitor of TGF-β to ACT T-cells using PEGylated immunoliposomes. Liposomes were prepared that released TGF-β inhibitor over ∼3 days in vitro. We compared the impact of targeting these drug-loaded vesicles to T-cells via an internalizing receptor (CD90) or noninternalizing receptor (CD45). When lymphocytes were preloaded with immunoliposomes in vitro prior to adoptive therapy, vesicles targeted to both CD45 and CD90 promoted enhanced T-cell expression of granzymes relative to free systemic drug administration, but only targeting to CD45 enhanced accumulation of granzyme-expressing T-cells in tumors, which correlated with the greatest enhancement of T-cell antitumor activity. By contrast, when administered i.v. to target T-cells in vivo, only targeting of a CD90 isoform expressed exclusively by the donor T-cells led to greater tumor regression over equivalent doses of free systemic drug. These results suggest that in vivo, targeting of receptors uniquely expressed by donor T-cells is of paramount importance for maximal efficacy. This immunoliposome strategy should be broadly applicable to target exogenous or endogenous T-cells and defines parameters to optimize delivery of supporting (or suppressive) drugs to these important immune effectors.


Immunology and Cell Biology | 2016

Actin polymerization-dependent activation of Cas-L promotes immunological synapse stability.

Luís C Santos; David A. Blair; Sudha Kumari; Michael Cammer; Thomas Iskratsch; Olivier Herbin; Konstantina Alexandropoulos; Michael L. Dustin; Michael P. Sheetz

The immunological synapse formed between a T‐cell and an antigen‐presenting cell is important for cell–cell communication during T‐cell‐mediated immune responses. Immunological synapse formation begins with stimulation of the T‐cell receptor (TCR). TCR microclusters are assembled and transported to the center of the immunological synapse in an actin polymerization‐dependent process. However, the physical link between TCR and actin remains elusive. Here we show that lymphocyte‐specific Crk‐associated substrate (Cas‐L), a member of a force sensing protein family, is required for transport of TCR microclusters and for establishing synapse stability. We found that Cas‐L is phosphorylated at TCR microclusters in an actin polymerization‐dependent fashion. Furthermore, Cas‐L participates in a positive feedback loop leading to amplification of Ca2+ signaling, inside–out integrin activation, and actomyosin contraction. We propose a new role for Cas‐L in T‐cell activation as a mechanical transducer linking TCR microclusters to the underlying actin network and coordinating multiple actin‐dependent structures in the immunological synapse. Our studies highlight the importance of mechanotransduction processes in T‐cell‐mediated immune responses.


JCI insight | 2017

DOCK8 enforces immunological tolerance by promoting IL-2 signaling and immune synapse formation in Tregs

Erin Janssen; Sudha Kumari; Mira Tohmé; Sumana Ullas; Victor Barrera; Jeroen M.J. Tas; Marcela Castillo-Rama; Roderick T. Bronson; Shariq M. Usmani; Darrell J. Irvine; Thorsten R. Mempel; Raif S. Geha

Patients deficient in the guanine nucleotide exchange factor DOCK8 have decreased numbers and impaired in vitro function of Tregs and make autoantibodies, but they seldom develop autoimmunity. We show that, similarly, Dock8-/- mice have decreased numbers and impaired in vitro function of Tregs but do not develop autoimmunity. In contrast, mice with selective DOCK8 deficiency in Tregs develop lymphoproliferation, autoantibodies, and gastrointestinal inflammation, despite a normal percentage and in vitro function of Tregs, suggesting that deficient T effector cell function might protect DOCK8-deficient patients from autoimmunity. We demonstrate that DOCK8 associates with STAT5 and is important for IL-2-driven STAT5 phosphorylation in Tregs. DOCK8 localizes within the lamellar actin ring of the Treg immune synapse (IS). Dock8-/- Tregs have abnormal TCR-driven actin dynamics, decreased adhesiveness, an altered gene expression profile, an unstable IS with decreased recruitment of signaling molecules, and impaired transendocytosis of the costimulatory molecule CD86. These data suggest that DOCK8 enforces immunological tolerance by promoting IL-2 signaling, TCR-driven actin dynamics, and the IS in Tregs.


Current Biology | 2015

Immunology: Dendritic Cells Pull the T Cell's Strings.

Sudha Kumari; Michael L. Dustin

Cells communicate by sensing diffusible or surface-associated chemical signals, with the surface-associated molecules also providing mechanical cues. New work now shows that both chemical and mechanical signals are critical for the communication between T cells and antigen-presenting dendritic cells in the initiation and maintenance of immune responses.


bioRxiv | 2018

Cytoskeletal tension actively sustains the T cell immunological synapse

Sudha Kumari; Michael Mak; Yehchuin Poh; Mira Tohme; Nicki Watson; Mariane B. Melo; Erin Janssen; Michael L. Dustin; Raif S. Geha; Darrell J. Irvine

Immunological synapses formed between T cells and cognate antigen presenting cells (APCs) mediate exchange of biochemical information between T cells and APCs, and the duration of such synapses is a critical determinant of T cell activation and effector functions. While the process of synapse formation has been well-characterized, the cellular processes that regulate synaptic lifetime remain unknown. Here we identify an antigen-triggered cytoskeletal mechanism that actively promotes T cell synapse stability by generating mechanical tension in the plane of the synapse. Cytoskeletal tension is generated by focal nucleation of actin via Wiskott-Aldrich syndrome Protein (WASP) and contraction of the resultant actin filaments by myosin II. Once T cells are activated, WASP is degraded, leading to cytoskeletal re-arrangement, tension decay and consequently, synaptic disengagement. The study identifies a mechanical program within T cells that regulates the lifetime of the T cell-APC synapse.When migratory T cells encounter antigen presenting cells (APCs), they arrest and form radially symmetric, stable intercellular junctions termed immunological synapses which facilitate exchange of crucial biochemical information and are critical for T cell immunity. While the cellular processes underlying synapse formation have been well-characterized, those that maintain the symmetry, and thereby the stability of the synapse remain unknown. Here we identify an antigen-triggered mechanism that actively promotes T cell synapse symmetry by generating cytoskeletal tension in the plane of the synapse through focal nucleation of actin via Wiskott -Aldrich syndrome Protein (WASP), and contraction of the resultant actin filaments by myosin II. Following T cell activation, WASP is degraded, leading to cytoskeletal rearrangement and tension decay, which result in synapse breaking. Thus, our study identifies and characterizes a mechanical program within otherwise highly motile T cells that sustains the symmetry and stability of the T cell-APC synaptic contact.


Journal of Clinical Investigation | 2018

The vimentin intermediate filament network restrains regulatory T cell suppression of graft-versus-host disease

Cameron McDonald-Hyman; J Muller; Michael Loschi; Govindarajan Thangavelu; Asim Saha; Sudha Kumari; Dawn K. Reichenbach; Michelle J. Smith; Guoan Zhang; Brent H. Koehn; Jiqiang Lin; Jason S. Mitchell; Brian T. Fife; Angela Panoskaltsis-Mortari; Colby J. Feser; Andrew Kemal Kirchmeier; Mark J. Osborn; Keli L. Hippen; Ameeta Kelekar; Jonathan S. Serody; Laurence A. Turka; David H. Munn; Hongbo Chi; Thomas A. Neubert; Michael L. Dustin; Bruce R. Blazar

Regulatory T cells (Tregs) are critical for maintaining immune homeostasis. However, current Treg immunotherapies do not optimally treat inflammatory diseases in patients. Understanding the cellular processes that control Treg function may allow for the augmentation of therapeutic efficacy. In contrast to activated conventional T cells, in which protein kinase C-&thgr; (PKC-&thgr;) localizes to the contact point between T cells and antigen-presenting cells, in human and mouse Tregs, PKC-&thgr; localizes to the opposite end of the cell in the distal pole complex (DPC). Here, using a phosphoproteomic screen, we identified the intermediate filament vimentin as a PKC-&thgr; phospho target and show that vimentin forms a DPC superstructure on which PKC-&thgr; accumulates. Treatment of mouse Tregs with either a clinically relevant PKC-&thgr; inhibitor or vimentin siRNA disrupted vimentin and enhanced Treg metabolic and suppressive activity. Moreover, vimentin-disrupted mouse Tregs were significantly better than controls at suppressing alloreactive T cell priming in graft-versus-host disease (GVHD) and GVHD lethality, using a complete MHC-mismatch mouse model of acute GVHD (C57BL/6 donor into BALB/c host). Interestingly, vimentin disruption augmented the suppressor function of PKC-&thgr;–deficient mouse Tregs. This suggests that enhanced Treg activity after PKC-&thgr; inhibition is secondary to effects on vimentin, not just PKC-&thgr; kinase activity inhibition. Our data demonstrate that vimentin is a key metabolic and functional controller of Treg activity and provide proof of principle that disruption of vimentin is a feasible, translationally relevant method to enhance Treg potency.

Collaboration


Dive into the Sudha Kumari's collaboration.

Top Co-Authors

Avatar

Darrell J. Irvine

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erin Janssen

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Kelly D. Moynihan

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Raif S. Geha

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Adrienne Rothschilds

Massachusetts Institute of Technology

View shared research outputs
Top Co-Authors

Avatar

Alice Tzeng

Massachusetts Institute of Technology

View shared research outputs
Researchain Logo
Decentralizing Knowledge