Network


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

Hotspot


Dive into the research topics where Lakshmi Kannan is active.

Publication


Featured researches published by Lakshmi Kannan.


Cell Reports | 2013

Human Complement Receptor Type 1/CD35 Is an Epstein-Barr Virus Receptor

Javier Gordon Ogembo; Lakshmi Kannan; Ionita Ghiran; Anne Nicholson-Weller; Robert W. Finberg; George C. Tsokos; Joyce D. Fingeroth

Epstein-Barr virus (EBV) attachment to primary B cells initiates virus entry. Although CD21 is the only known receptor for EBVgp350/220, a recent report documents EBV-infected B cells from a patient genetically deficient in CD21. On normal resting B cells, CD21 forms two membrane complexes: one with CD19 and another with CD35. Whereas the CD21/CD19 complex is widely retained on immortalized and B cell tumor lines, the related complement-regulatory protein CD35 is lost. To determine the role(s) of CD35 in initial infection, we transduced a CD21-negative pre-B cell and myeloid leukemia line with CD35, CD21, or both. Cells expressing CD35 alone bound gp350/220 and became latently infected when the fusion receptor HLA II was coexpressed. Temporal, biophysical, and structural characteristics of CD35-mediated infection were distinct from CD21. Identification of CD35 as an EBV receptor uncovers a salient role in primary infection, addresses unsettled questions of virus tropism, and underscores the importance of EBVgp350/220 for vaccine development.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2011

Depletion of gut commensal bacteria attenuates intestinal ischemia/reperfusion injury

Kazuhisa Yoshiya; Peter H. Lapchak; To-Ha Thai; Lakshmi Kannan; Poonam Rani; Jurandir J. Dalle Lucca; George C. Tsokos

Gut commensal bacteria play important roles in the development and homeostasis of intestinal immunity. However, the role of gut commensals in intestinal ischemia/reperfusion (I/R) injury is unclear. To determine the roles of gut commensal bacteria in intestinal IR injury, we depleted gut microbiota with a broad-spectrum antibiotic cocktail and performed mesenteric I/R (M I/R). First, we confirmed that antibiotic treatment completely depleted gut commensal bacteria and diminished the size of secondary lymphoid tissues such as the Peyers patches. We next found that antibiotic treatment attenuated intestinal injury following M I/R. Depletion of gut commensal bacteria reduced the expression of Toll-like receptor (TLR)2 and TLR4 in the intestine. Both are well-known receptors for gram-positive and -negative bacteria. Decreased expression of TLR2 and TLR4 led to the reduction of inflammatory mediators, such as TNF, IL-6, and cyclooxygenase-2. Intestinal I/R injury is initiated when natural antibodies recognize neo-antigens that are revealed on ischemic cells and activate the complement pathway. Thus we evaluated complement and immunoglobulin (Ig) deposition in the damaged intestine and found that antibiotic treatment decreased the deposition of both C3 and IgM. Interestingly, we also found that the deposition of IgA also increased in the intestine following M I/R compared with control mice and that antibiotic treatment decreased the deposition of IgA in the damaged intestine. These results suggest that depletion of gut commensal bacteria decreases B cells, Igs, and TLR expression in the intestine, inhibits complement activation, and attenuates intestinal inflammation and injury following M I/R.


PLOS ONE | 2012

The Role of Platelet Factor 4 in Local and Remote Tissue Damage in a Mouse Model of Mesenteric Ischemia/ Reperfusion Injury

Peter H. Lapchak; Antonis Ioannou; Poonam Rani; Linda A. Lieberman; Kazuhisa Yoshiya; Lakshmi Kannan; Jurandir J. Dalle Lucca; M. Anna Kowalska; George C. Tsokos

The robust inflammatory response that occurs during ischemia reperfusion (IR) injury recruits factors from both the innate and adaptive immune systems. However the contribution of platelets and their products such as Platelet Factor 4 (PF4; CXCL4), during the pathogenesis of IR injury has not been thoroughly investigated. We show that a deficiency in PF4 protects mice from local and remote tissue damage after 30 minutes of mesenteric ischemia and 3 hours of reperfusion in PF4-/- mice compared to control B6 mice. This protection was independent from Ig or complement deposition in the tissues. However, neutrophil and monocyte infiltration were decreased in the lungs of PF4-/- mice compared with B6 control mice. Platelet-depleted B6 mice transfused with platelets from PF4-/- mice displayed reduced tissue damage compared with controls. In contrast, transfusion of B6 platelets into platelet depleted PF4-/- mice reconstituted damage in both intestine and lung tissues. We also show that PF4 may modulate the release of IgA. Interestingly, we show that PF4 expression on intestinal epithelial cells is increased after IR at both the mRNA and protein levels. In conclusion, these findings demonstrate that may PF4 represent an important mediator of local and remote tissue damage.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2012

Platelets orchestrate remote tissue damage after mesenteric ischemia-reperfusion

Peter H. Lapchak; Lakshmi Kannan; Antonis Ioannou; Poonam Rani; Peter Karian; Jurandir J. Dalle Lucca; George C. Tsokos

Ischemia-reperfusion (I/R) injury is a leading cause of morbidity and mortality. A functional role for platelets in tissue damage after mesenteric I/R is largely unknown. The hypothesis that mesenteric I/R local and remote injury are platelet dependent was tested. Using a murine mesenteric I/R model, we demonstrate that platelets orchestrate remote lung tissue damage that follows mesenteric I/R injury and also contribute, albeit to a lesser degree, to local villi damage. While lung damage is delayed compared with villi damage, it increased over time and was characterized by accumulation of platelets in the pulmonary vasculature early, followed by alveolar capillaries and extravasation into the pulmonary space. Both villi and lung tissues displayed complement deposition. We demonstrate that villi and lung damage are reduced in mice made platelet deficient before I/R injury and that platelet transfusion into previously platelet-depleted mice before I/R increased both villi and lung tissue damage. Increased C3 deposition accompanied platelet sequestration in the lung, which was mostly absent in platelet-depleted mice. In contrast, C3 deposition was only minimally reduced on villi of platelet-depleted mice. Our findings position platelets alongside complement as a significant early upstream component that orchestrates remote lung tissue damage after mesenteric I/R and strongly suggest that reperfusion injury mitigating modalities should consider the contribution of platelets.


Journal of Immunology | 2016

Intracellular Activation of Complement 3 Is Responsible for Intestinal Tissue Damage during Mesenteric Ischemia

Abhigyan Satyam; Lakshmi Kannan; Matsumoto N; Mayya Geha; Peter H. Lapchak; Robin E. Bosse; Guo-Ping Shi; Dalle Lucca Jj; Maria Tsokos; George C. Tsokos

Intestinal ischemia followed by reperfusion leads to local and remote organ injury attributed to inflammatory response during the reperfusion phase. The extent to which ischemia contributes to ischemia/reperfusion injury has not been thoroughly studied. After careful evaluation of intestinal tissue following 30 min of ischemia, we noticed significant local mucosal injury in wild-type mice. This injury was drastically reduced in C3-deficient mice, suggesting C3 involvement. Depletion of circulating complement with cobra venom factor eliminated, as expected, injury recorded at the end of the reperfusion phase but failed to eliminate injury that occurred during the ischemic phase. Immunohistochemical studies showed that tissue damage during ischemia was associated with increased expression of C3/C3 fragments primarily in the intestinal epithelial cells, suggesting local involvement of complement. In vitro studies using Caco2 intestinal epithelial cells showed that in the presence of LPS or exposure to hypoxic conditions the cells produce higher C3 mRNA as well as C3a fragment. Caco2 cells were also noted to produce cathepsins B and L, and inhibition of cathepsins suppressed the release of C3a. Finally, we found that mice treated with a cathepsin inhibitor and cathepsin B–deficient mice suffer limited intestinal injury during the ischemic phase. To our knowledge, our findings demonstrate for the first time that significant intestinal injury occurs during ischemia prior to reperfusion and that this is due to activation of C3 within the intestinal epithelial cells in a cathepsin-dependent manner. Modulation of cathepsin activity may prevent injury of organs exposed to ischemia.


Proceedings of the National Academy of Sciences of the United States of America | 2013

R-spondin3 prevents mesenteric ischemia/reperfusion-induced tissue damage by tightening endothelium and preventing vascular leakage

Lakshmi Kannan; Katalin Kis-Toth; Kazuhisa Yoshiya; To-Ha Thai; Seema Sehrawat; Tanya N. Mayadas; Jurandir J. Dalle Lucca; George C. Tsokos

Inflammation and vascular injury triggered by ischemia/reperfusion (I/R) represent a leading cause of morbidity and mortality in a number of clinical settings. Wnt and its homolog partners R-spondins, in addition to regulating embryonic development have recently been demonstrated to serve as wound-healing agents in inflammation-associated conditions. Here we ask whether R-spondins could prevent inflammation-associated tissue damage in ischemic disorders and thus investigate the role of R-spondin3 (R-spo3) in a mouse model of mesenteric I/R. We demonstrate that R-spo3 ameliorates mesenteric I/R-induced local intestinal as well as remote lung damage by suppressing local and systemic cytokine response and deposition of IgM and complement in intestinal tissues. We also show that decreased inflammatory response is accompanied by tightening of endothelial cell junctions and reduction in vascular leakage. We conclude that R-spo3 stabilizes endothelial junctions and inhibits vascular leakage during I/R and thereby mitigates the inflammatory events and associated tissue damage. Our findings uniquely demonstrate a protective effect of R-spo3 in I/R-related tissue injury and suggest a mechanism by which it may have these effects.


Arthritis & Rheumatism | 2016

Selective Loss of Signaling Lymphocytic Activation Molecule Family Member 4–Positive CD8+ T Cells Contributes to the Decreased Cytotoxic Cell Activity in Systemic Lupus Erythematosus

Katalin Kis-Toth; Denis Comte; Maria P. Karampetsou; Vasileios C. Kyttaris; Lakshmi Kannan; Cox Terhorst; George C. Tsokos

Engagement of signaling lymphocytic activation molecule family member 4 (SLAMF4; CD244, 2B4) by its ligand SLAMF2 (CD48) modulates the function and expansion of both natural killer cells and a subset of cytotoxic CD8+ T cells. Because the cytotoxicity of CD8+ T lymphocytes isolated from patients with systemic lupus erythematosus (SLE) is known to be impaired, the aim of this study was to assess whether the expression and function of the checkpoint regulator SLAMF4 are altered on CD8+ T cells from patients with SLE.


Arthritis & Rheumatism | 2015

The selective loss of SLAMF4+ CD8+ T cells contributes to the decreased cytotoxic cell activity in systemic lupus erythematosus

Katalin Kis-Toth; Denis Comte; Maria P. Karampetsou; Vasileios C. Kyttaris; Lakshmi Kannan; Cox Terhorst; George C. Tsokos

Engagement of signaling lymphocytic activation molecule family member 4 (SLAMF4; CD244, 2B4) by its ligand SLAMF2 (CD48) modulates the function and expansion of both natural killer cells and a subset of cytotoxic CD8+ T cells. Because the cytotoxicity of CD8+ T lymphocytes isolated from patients with systemic lupus erythematosus (SLE) is known to be impaired, the aim of this study was to assess whether the expression and function of the checkpoint regulator SLAMF4 are altered on CD8+ T cells from patients with SLE.


Autoimmunity | 2013

Platelets, complement and tissue inflammation.

Antonis Ioannou; Lakshmi Kannan; George C. Tsokos

The release of immunoregulatory and inflammatory molecules following platelet activation has been invariably associated with the expression of tissue injury in several clinical conditions including trauma, organ transplantation, inflammatory bowel diseases and autoimmune diseases. We present a thorough review of the available information on the role of platelets and their interaction with complement cascade on the expression of tissue inflammation and organ damage. We propose that in autoimmune diseases and conditions associated with ischemia/reperfusion, platelets are decorated with complement, become activated and lodge tissues inappropriately to spread the inflammatory process. Interventions such as limiting complement decoration and suppression of signaling processes leading to platelet activation should be met with clinical benefit.


Critical Care Medicine | 2014

C4d Deposits on the Surface of RBCs in Trauma Patients and Interferes With Their Function

Takashi Muroya; Lakshmi Kannan; Ionita Ghiran; Sergey S. Shevkoplyas; Ziv Paz; Maria Tsokos; Jurandir J. Dalle Lucca; Nathan I. Shapiro; George C. Tsokos

Objective:Complement system is activated in patients with trauma. Although complement activation is presumed to contribute to organ damage and constitutional symptoms, little is known about the involved mechanisms. Because complement components may deposit on RBCs, we asked whether complement deposits on the surface of RBC in trauma and whether such deposition alters RBC function. Design:A prospective experimental study. Setting:Research laboratory. Subjects:Blood samples collected from 42 trauma patients and 21 healthy donors. Intervention:None. Measurements and Main Results:RBC and sera were collected from trauma patients and control donors. RBCs from trauma patients (n = 40) were found to display significantly higher amounts of C4d on their surface by flow cytometry compared with RBCs from control (n = 17) (p < 0.01). Increased amounts of iC3b were found in trauma sera (n = 27) (vs 12 controls, p < 0.01) by enzyme-linked immunosorbent assay. Incubation of RBC from universal donors (type O, Rh negative) with trauma sera (n = 10) promoted C4d deposition on their surface (vs six controls, p< 0.05). Complement-decorated RBC (n = 6) displayed limited their deformability (vs six controls, p < 0.05) in two-dimensional microchannel arrays. Incubation of RBC with trauma sera (n = 10) promoted the phosphorylation of band 3, a cytoskeletal protein important for the function of the RBC membrane (vs eight controls, p < 0.05), and also accelerated calcium influx (n = 9) and enhanced nitric oxide production (n = 12) (vs four and eight controls respectively, p < 0.05) in flow cytometry. Conclusions:Our study found the presence of extensive complement activation in trauma patients and presents new evidence in support of the hypothesis that complement activation products deposit on the surface of RBC. Such deposition could limit RBC deformability and promote the production of nitric oxide. Our findings suggest that RBC in trauma patients malfunctions, which may explain organ damage and constitutional symptoms that is not accounted for otherwise by previously known pathophysiologic mechanisms.

Collaboration


Dive into the Lakshmi Kannan's collaboration.

Top Co-Authors

Avatar

George C. Tsokos

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jurandir J. Dalle Lucca

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Peter H. Lapchak

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Antonis Ioannou

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Poonam Rani

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Kazuhisa Yoshiya

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Katalin Kis-Toth

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

To-Ha Thai

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Ionita Ghiran

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Maria Tsokos

Beth Israel Deaconess Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge