Network


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

Hotspot


Dive into the research topics where Jurandir J. Dalle Lucca is active.

Publication


Featured researches published by Jurandir J. Dalle Lucca.


Clinical Immunology | 2009

IL-17 producing CD4+ T cells mediate accelerated ischemia/reperfusion-induced injury in autoimmunity-prone mice.

Colin Edgerton; José C. Crispín; Chantal Moratz; Estelle Bettelli; Mohamed Oukka; Milomir Simovic; Athina Zacharia; Ryan Egan; Jie Chen; Jurandir J. Dalle Lucca; Yuang Taung Juang; George C. Tsokos

Elements of the innate and adaptive immune response have been implicated in the development of tissue damage after ischemic reperfusion (I/R). Here we demonstrate that T cells infiltrate the intestine of C57BL/6 mice subjected to intestinal I/R during the first hour of reperfusion. The intensity of the T cell infiltration was higher in B6.MRL/lpr mice subjected to intestinal I/R and reflected more severe tissue damage than that observed in control mice. Depletion of T cells limited I/R damage in B6.MRL/lpr mice, whereas repletion of B6.MRL/lpr lymph node-derived T cells into the I/R-resistant Rag-1(-/-) mouse reconstituted tissue injury. The tissue-infiltrating T cells were found to produce IL-17. Finally, IL-23 deficient mice, which are known not to produce IL-17, displayed significantly less intestinal damage when subjected to I/R. Our data assign T cells a major role in intestinal I/R damage by virtue of producing the pro-inflammatory cytokine IL-17.


Clinical Immunology | 2011

Immunopathogenesis of ischemia/reperfusion-associated tissue damage.

Antonis Ioannou; Jurandir J. Dalle Lucca; George C. Tsokos

Ischemia/reperfusion (IR) instigates a complex array of inflammatory events which result in damage to the local tissue. IR-related organ damage occurs invariably in several clinical conditions including trauma, organ transplantation, autoimmune diseases and revascularization procedures. We critically review available pre-clinical experimental information on the role of immune response in the expression of tissue damage following IR. Distinct elements of the innate and adaptive immune response are involved in the expression of tissue injury. Interventions such as prevention of binding of natural antibody to antigen expressed on the surface of ischemia-conditioned cells, inhibition of the ensuing complement activation, modulation of Toll-like receptors, B or T cell depletion and blockade of inflammatory cytokines and chemokines limit IR injury in preclinical studies. Clinical trials that will determine the therapeutic value of each approach is needed.


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.


Journal of the Neurological Sciences | 2012

Blast-induced moderate neurotrauma (BINT) elicits early complement activation and tumor necrosis factor alpha (TNFα) release in a rat brain

Jurandir J. Dalle Lucca; Mikulas Chavko; Michael A. Dubick; Saleena Adeeb; Michael Falabella; Jessica L. Slack; Richard M. McCarron; Yansong Li

Blast-induced neurotrauma (BINT) is a major medical concern yet its etiology is largely undefined. Complement activation may play a role in the development of secondary injury following traumatic brain injury; however, its role in BINT is still undefined. The present study was designed to characterize the complement system and adaptive immune-inflammatory responses in a rat model of moderate BINT. Anesthetized rats were exposed to a moderate blast (120 kPa) using an air-driven shock tube. Brain tissue injury, systemic and local complement, cerebral edema, inflammatory cell infiltration, and pro-inflammatory cytokine production were measured at 0.5, 3, 48, 72, 120, and 168 h. Injury to brain tissue was evaluated by histological evaluation. Systemic complement was measured via ELSIA. The remaining measurements were determined by immunohistoflourescent staining. Moderate blast triggers moderate brain injuries, elevated levels of local brain C3/C5b-9 and systemic C5b-9, increased leukocyte infiltration, unregulated tumor necrosis factor alpha (TNFα), and aquaporin-4 in rat brain cortex at 3- and 48-hour post blast. Early immune-inflammatory response to BINT involves complement and TNFα, which correlates with hippocampus and cerebral cortex damage. Complement and TNFα activation may be a novel therapeutic target for reducing the damaging effects of BINT inflammation.


Journal of Neuroinflammation | 2010

Decay accelerating factor (CD55) protects neuronal cells from chemical hypoxia-induced injury

Ying Wang; Yansong Li; Shawn L Dalle Lucca; Milomir Simovic; George C. Tsokos; Jurandir J. Dalle Lucca

BackgroundActivated complement system is known to mediate neuroinflammation and neurodegeneration following exposure to hypoxic-ischemic insults. Therefore, inhibition of the complement activation cascade may represent a potential therapeutic strategy for the management of ischemic brain injury. Decay-accelerating factor (DAF, also known as CD55) inhibits complement activation by suppressing the function of C3/C5 convertases, thereby limiting local generation or deposition of C3a/C5a and membrane attack complex (MAC or C5b-9) production. The present study investigates the ability of DAF to protect primary cultured neuronal cells subjected to sodium cyanide (NaCN)-induced hypoxia from degeneration and apoptosis.MethodsCultured primary cortical neurons from embryonic Sprague-Dawley rats were assigned one of four groups: control, DAF treatment alone, hypoxic, or hypoxic treated with DAF. Hypoxic cultures were exposed to NaCN for 1 hour, rinsed, followed by 24 hour exposure to 200 ng/ml of recombinant human DAF in normal medium. Human DAF was used in the present study and it has been shown to effectively regulate complement activation in rats. Neuronal cell function, morphology and viability were investigated by measuring plateau depolarization potential, counting the number dendritic spines, and observing TUNEL and MTT assays. Complement C3, C3a, C3a receptor (R) production, C3a-C3aR interaction and MAC formation were assessed along with the generation of activated caspase-9, activated caspase-3, and activated Src.ResultsWhen compared to controls, hypoxic cells had fewer dendritic spines, reduced plateau depolarization accompanied by increased apoptotic activity and accumulation of MAC, as well as up-regulation of C3, C3a and C3aR, enhancement of C3a-C3aR engagement, and elevated caspase and Src activity. Treatment of hypoxic cells with 200 ng/ml of recombinant human DAF resulted in attenuation of neuronal apoptosis and exerted significant protection against neuronal dendritic spine loss and plateau depolarization reduction. Furthermore, treatment with DAF resulted in decreased accumulation of C3a, MAC, C3a-C3aR interaction, caspase-9, activated caspase-3, and pTyr416-Src (activated Src) tyrosine kinase.ConclusionDAF was found to reduce neuronal cell death and apoptosis in NaCN induced hypoxia. This effect is attributed to the ability of DAF to limit complement activation and inhibit the activity of Src and caspases 9 and 3. This study supports the inhibiting of complement as a neuroprotective strategy against CNS ischemia/reperfusion injury.


American Journal of Physiology-gastrointestinal and Liver Physiology | 2010

Spleen tyrosine kinase inhibition prevents tissue damage after ischemia-reperfusion

Omer Nuri Pamuk; Peter H. Lapchak; Poonam Rani; Polly Pine; Jurandir J. Dalle Lucca; George C. Tsokos

Reperfusion injury to tissue following an ischemic event occurs as a consequence of an acute inflammatory response that can cause significant morbidity and mortality. Components of both the innate (complement, immunoglobulin, monocytes, and neutrophils) and adaptive (B and T lymphocytes) immune systems have been demonstrated to mediate tissue injury. Spleen tyrosine kinase (Syk) is responsible for membrane-mediated signaling in various cell types including B lymphocytes, macrophages, and T cells. We investigated the ability of a small drug Syk inhibitor, R788, to protect mice against mesenteric ischemia-reperfusion (I/R)-induced local (intestine) and remote lung injury. Mice were fed with chow containing a Syk inhibitor for 6 days before the performance of intestinal I/R, which resulted in silencing of the expression of the active phosphorylated Syk. Syk inhibition significantly suppressed both local and remote lung injury. The beneficial effect was associated with reduced IgM and complement 3 deposition in the tissues and significant reduction of polymorphonuclear cell infiltration. Our data place Syk upstream of events leading to the binding of natural antibodies to the ischemia-conditioned tissues and urge the consideration of the use of Syk inhibitors in the prevention or improvement of tissue injury of organs exposed to ischemia or hypoperfusion.


Journal of Surgical Research | 2011

A Novel Inhibitor of the Alternative Pathway of Complement Attenuates Intestinal Ischemia/Reperfusion-Induced Injury

Jie Chen; José C. Crispín; Jurandir J. Dalle Lucca; George C. Tsokos

Complement activation has been demonstrated to contribute significantly to the expression of IR-induced tissue damage. Each of the three complement pathways, classic, alternative, and lectin, has been implicated in the instigation of tissue pathology. In this study, we used a selective inhibitor of the alternative pathway, that is, a soluble form of complement receptor of the immunoglobulin superfamily (CRIg-Fc) to determine whether it can prevent IR tissue injury. We demonstrate that treatment of C57B1/6 mice prior to mesenteric IR prevents local (intestinal) and remote (lung) injury by limiting deposition of complement and entry of polymorphonuclear cells to the sites of injury. Our results show that CRIg-Fc represents a candidate to limit IR injury as it occurs in various clinical conditions.


Journal of Autoimmunity | 2009

B cells contribute to ischemia/reperfusion-mediated tissue injury

Jie Chen; José C. Crispín; Thomas F. Tedder; Jurandir J. Dalle Lucca; George C. Tsokos

Multiple elements are known to participate in ischemia/reperfusion (I/R)-mediated tissue injury. Amongst them, B cells have been shown to contribute by the production of antibodies that bind to ischemic cells and fix complement. It is currently unknown whether B cells participate through antibody-independent mechanisms in the pathogenesis of I/R. In a mesenteric I/R model we found that B cells infiltrate the injured intestine of normal and autoimmune mice 2h after reperfusion is established. B cell depletion protected mice from the development of I/R-mediated intestinal damage. The protection conferred by B cell depletion was significantly greater in MRL/lpr mice. Finally, we show that ischemic tissue expressed the B cell-attractant CXCL13 and infiltrating B cells expressed the corresponding receptor CXCR5. Our data grant B cells an antibody-independent role in the pathogenesis of intestinal I/R and suggest that B cells accumulate in the injured tissue in response to the chemokine CXCL13.


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.


Shock | 2012

Effects of C1 inhibitor on tissue damage in a porcine model of controlled hemorrhage.

Jurandir J. Dalle Lucca; Yansong Li; Milomir Simovic; Anthony E. Pusateri; Michael Falabella; Michael A. Dubick; George C. Tsokos

ABSTRACT Activation of the complement system has been associated with tissue injury after hemorrhage and resuscitation in animals. We investigated whether administration of recombinant human C1-esterase inhibitor (rhC1-INH), a regulator of complement and contact activation systems, reduces tissue damage and cytokine release and improves metabolic acidosis in a porcine model of hemorrhagic shock. Male Yorkshire swine were assigned to experimental groups and subjected to controlled, isobaric hemorrhage to a target mean arterial pressure of 35 mmHg. Hypotension was maintained for 20 min followed by a bolus intravenous injection of rhC1-INH or vehicle; animals were then observed for 3 h. Blood chemistry and physiologic parameters were recorded. Lung and small intestine tissue samples were subjected to histopathologic evaluation and immunohistochemistry to determine the extent of injury and deposition of complement proteins. Cytokine levels and quantitative assessment of renal and hepatic function were measured via enzyme-linked immunosorbent assay and chemistry analyzer, respectively. Pharmacokinetics of rhC1-INH revealed dose proportionality for maximum concentration, half-life, and the time span in which the functional C1-INH level was greater than 1 IU/mL. Recombinant human C1-INH significantly reduced renal, intestinal, and lung tissue damage in a dose-dependent manner (100 and 250 IU/kg). In addition, rhC1-INH (250 IU/kg) markedly improved hemorrhage-induced metabolic acidosis and circulating tumor necrosis factor &agr;. The tissue-protective effects of rhC1-INH appear to be related to its ability to reduce tissue complement activation and deposition. Recombinant human C1-INH decreased tissue complement activation and deposition in hemorrhaged animals, improved metabolic acidosis, reduced circulating tumor necrosis factor &agr;, and attenuated tissue damage in this model. The observed beneficial effects of rhC1-INH treatment on tissue injury 20 min into severe hypotension present an attractive model of low-volume resuscitation, particularly in situations with a restrictive medical logistical footprint.

Collaboration


Dive into the Jurandir J. Dalle Lucca's collaboration.

Top Co-Authors

Avatar

George C. Tsokos

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar

Lakshmi Kannan

Beth Israel Deaconess Medical Center

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

Yansong Li

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Milomir Simovic

Walter Reed Army Institute of Research

View shared research outputs
Top Co-Authors

Avatar

Maria Tsokos

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

Michael Falabella

Walter Reed Army Institute of Research

View shared research outputs
Researchain Logo
Decentralizing Knowledge