Network


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

Hotspot


Dive into the research topics where Adriana T. Larregina is active.

Publication


Featured researches published by Adriana T. Larregina.


Blood | 2012

Mechanism of transfer of functional microRNAs between mouse dendritic cells via exosomes

Angela Montecalvo; Adriana T. Larregina; William J. Shufesky; Donna B. Stolz; Mara L. Sullivan; Jenny M. Karlsson; Catherine J. Baty; Gregory A. Gibson; Geza Erdos; Zhiliang Wang; Jadranka Milosevic; Olga Tkacheva; Sherrie J. Divito; Rick Jordan; James Lyons-Weiler; Simon C. Watkins; Adrian E. Morelli

Dendritic cells (DCs) are the most potent APCs. Whereas immature DCs down-regulate T-cell responses to induce/maintain immunologic tolerance, mature DCs promote immunity. To amplify their functions, DCs communicate with neighboring DCs through soluble mediators, cell-to-cell contact, and vesicle exchange. Transfer of nanovesicles (< 100 nm) derived from the endocytic pathway (termed exosomes) represents a novel mechanism of DC-to-DC communication. The facts that exosomes contain exosome-shuttle miRNAs and DC functions can be regulated by exogenous miRNAs, suggest that DC-to-DC interactions could be mediated through exosome-shuttle miRNAs, a hypothesis that remains to be tested. Importantly, the mechanism of transfer of exosome-shuttle miRNAs from the exosome lumen to the cytosol of target cells is unknown. Here, we demonstrate that DCs release exosomes with different miRNAs depending on the maturation of the DCs. By visualizing spontaneous transfer of exosomes between DCs, we demonstrate that exosomes fused with the target DCs, the latter followed by release of the exosome content into the DC cytosol. Importantly, exosome-shuttle miRNAs are functional, because they repress target mRNAs of acceptor DCs. Our findings unveil a mechanism of transfer of exosome-shuttle miRNAs between DCs and its role as a means of communication and posttranscriptional regulation between DCs.


Science Translational Medicine | 2012

A Peptide Derived from Endostatin Ameliorates Organ Fibrosis

Yukie Yamaguchi; Takahisa Takihara; Roger Chambers; Kristen L. Veraldi; Adriana T. Larregina; Carol A. Feghali-Bostwick

A naturally occurring peptide from endostatin can inhibit fibrosis in lung and skin, even when it is already established. Putting an End to Fibrotic Organ Failure with Endostatin? Organ fibrosis or “scarring” has the dubious distinction of accounting for nearly half of all deaths in the developed world, taking its toll in disorders ranging from liver cirrhosis to cardiovascular disease. There are no effective treatments for fibrosis, an out-of-control wound-healing process in which excess extracellular matrix components such as collagen and fibronectin replace normal tissue, ultimately resulting in organ failure. Endostatin, a naturally occurring proteolytic fragment of one form of collagen, is elevated in patients with different forms of fibrosis. Previously, endostatin was shown to inhibit the growth of new blood vessels, an effect attributed to its N-terminal region. Because signaling molecules involved in blood vessel formation are also present in fibrotic tissues, Yamaguchi et al. investigated whether endostatin might inhibit fibrosis as well. Indeed, they found that a recombinant form did inhibit dermal fibrosis, in this case induced by the profibrotic factor transforming growth factor–β (TGF-β) in an ex vivo human skin model. So, too, could a smaller peptide, termed E4, which was derived from the C terminus of endostatin. In addition, E4 prevented TGF-β–induced dermal fibrosis in a mouse model—as well as dermal and lung fibrosis induced by a different trigger, bleomycin. E4 also reduced bleomycin-induced cell death in the mouse lung. Furthermore, Yamaguchi et al. found that E4 could reverse ongoing fibrosis, in addition to preventing its initiation, in these models. Finally, the researchers showed that E4 reduced the expression of two key proteins: lysyl oxidase, an enzyme that cross-links collagen and thereby contributes to fibrosis, and the transcription factor Egr-1, which is a central fibrosis regulator. Because E4 can inhibit both skin and lung fibrosis, it may well be able to inhibit fibrosis in other tissues as well. Further research is needed to explore the therapeutic potential of this peptide for preventing or reversing organ fibrosis. Fibroproliferative disorders such as idiopathic pulmonary fibrosis and systemic sclerosis have no effective therapies and result in significant morbidity and mortality due to progressive organ fibrosis. We examined the effect of peptides derived from endostatin on existing fibrosis and fibrosis triggered by two potent mediators, transforming growth factor–β (TGF-β) and bleomycin, in human and mouse tissues in vitro, ex vivo, and in vivo. We identified one peptide, E4, with potent antifibrotic activity. E4 prevented TGF-β–induced dermal fibrosis in vivo in a mouse model, ex vivo in human skin, and in bleomycin-induced dermal and pulmonary fibrosis in vivo, demonstrating that E4 exerts potent antifibrotic effects. In addition, E4 significantly reduced existing fibrosis in these preclinical models. E4 amelioration of fibrosis was accompanied by reduced cell apoptosis and lower levels of lysyl oxidase, an enzyme that cross-links collagen, and Egr-1 (early growth response gene–1), a transcription factor that mediates the effects of several fibrotic triggers. Our findings identify E4 as a peptide with potent antifibrotic activity and a possible therapeutic agent for organ fibrosis.


Arthritis & Rheumatism | 2013

Interleukin‐13–producing CD8+ T cells mediate dermal fibrosis in patients with systemic sclerosis

Patrizia Fuschiotti; Adriana T. Larregina; Johnan Ho; Carol A. Feghali-Bostwick; Thomas A. Medsger

OBJECTIVEnFibrosis is a major contributor to morbidity and mortality in systemic sclerosis (SSc). T cells are the predominant inflammatory infiltrate in affected tissue and are thought to produce cytokines that drive the synthesis of extracellular matrix (ECM) proteins by fibroblasts, resulting in excessive fibrosis. We have previously shown that aberrant interleukin-13 (IL-13) production by peripheral blood effector CD8+ T cells from SSc patients correlates with the extent of skin fibrosis. The present study was undertaken to investigate the role of IL-13 production by CD8+ T cells in dermal fibrosis, an early and specific manifestation of SSc.nnnMETHODSnECM protein production by normal dermal fibroblasts cocultured with SSc CD8+ T cell supernatants was determined by quantitative polymerase chain reaction and Western blotting. Skin-homing receptor expression and IL-13 production by CD8+ T cells in the peripheral blood of SSc patients were measured by flow cytometry. IL-13+ and CD8+ cells in sclerotic skin were identified by immunohistochemistry.nnnRESULTSnIL-13-producing circulating CD8+ T cells from patients with SSc expressed skin-homing receptors and induced a profibrotic phenotype in normal dermal fibroblasts, which was inhibited by an anti-IL-13 antibody. High numbers of CD8+ T cells and IL-13+ cells were found in the skin lesions of SSc patients, particularly during the early inflammatory phase of the disease.nnnCONCLUSIONnThese findings show that IL-13-producing CD8+ T cells are directly involved in modulating dermal fibrosis in SSc. The demonstration that CD8+ T cells homing to the skin early in the course of SSc are associated with accumulation of IL-13 is an important mechanistic contribution to the understanding of the pathogenesis of dermal fibrosis in SSc and may represent a potential target for therapeutic intervention.


American Journal of Transplantation | 2012

Dendritic Cell Therapies in Transplantation Revisited: Deletion of Recipient DCs Deters the Effect of Therapeutic DCs

Zhiliang Wang; Sherrie J. Divito; William J. Shufesky; Tina L. Sumpter; H. Wang; Olga Tkacheva; W. Wang; C. Liu; Adriana T. Larregina; Adrian E. Morelli

A critical goal in transplantation is the achievement of donor‐specific tolerance, minimizing the use of immunosuppressants. Dendritic cells (DCs) are antigen (Ag) presenting cells (APCs) with capability to promote immunity or tolerance. The immune‐regulatory properties of DCs have been exploited for generation of tolerogenic/immunosuppressive (IS) DCs that, when transfer systemically, prolong allograft survival in murine models. Surprisingly, the in vivo mechanisms of therapies based on (donor‐ or recipient‐derived) ISDCs in transplantation remain unknown, given that previous studies investigated their effects in vitro, or ex vivo after transplantation. Since once injected, ISDCs are short‐lived and transfer Ag to recipient APCs, we assessed the role of recipient DCs by depleting them at the time of ISDC‐therapy in a mouse model of cardiac transplantation. The results indicate that, contrary to the accepted paradigm, systemically administered ISDCs reduce the alloresponse and prolong allograft survival, not by themselves, but through conventional DCs (cDCs) of the recipient. These findings raise doubts on the advantages of the currently used ISDC‐therapies, since the immune‐regulatory properties of the injected ISDC do not seem to be functionally relevant in vivo, and the quiescent/pro‐tolerogenic status of cDCs may be compromised in patients with end‐stage diseases that require transplantation.


Journal of Investigative Dermatology | 2013

Human Beta Defensin 3 induces maturation of human langerhans cell like dendritic cells: An antimicrobial peptide that functions as an endogenous adjuvant

Laura K. Ferris; Yvonne K. Mburu; Alicia R. Mathers; Eric Fluharty; Adriana T. Larregina; Robert L. Ferris; Louis D. Falo

Human beta defensins (hBDs) are antimicrobial peptides that play an important role in innate immune responses at epithelial barriers such as the skin. However, the role that hBDs play in initiating cellular immune responses that contribute to antigen-specific adaptive immunity is not well understood. Here we show that one member of the hBD family, hBD3, can induce maturation and T helper type 1 (Th1) skewing function in human Langerhans cell-like DCs (LC-DCs). Specifically, hBD3 potently induces phenotypic maturation of LC-DCs, including increased expression of CCR7 which mediates functional chemotactic responses to CCL19 and CCL21. HBD3-stimulated LC-DCs induce strong proliferation and IFN-γ secretion by naïve human T cells. HBD3 also induces phenotypic maturation of primary human skin-migratory dendritic cells derived from human skin explants. These results suggest an important role for hBD3 in inducing DC activation, migration, and polarization. Thus hBD3 contributes to the integration of innate and adaptive immune responses in the skin and may be a useful adjuvant for skin immunization and an important factor in the pathophysiology of inflammatory skin diseases.


Blood | 2013

Neurokinin-1 receptor agonists bias therapeutic dendritic cells to induce type 1 immunity by licensing host dendritic cells to produce IL-12

Brian M. Janelsins; Tina L. Sumpter; Olga Tkacheva; Darling M. Rojas-Canales; Geza Erdos; Alicia R. Mathers; William J. Shufesky; Walter J. Storkus; Louis D. Falo; Adrian E. Morelli; Adriana T. Larregina

Substance-P and hemokinin-1 are proinflammatory neuropeptides with potential to promote type 1 immunity through agonistic binding to neurokinin-1 receptor (NK1R). Dendritic cells (DCs) are professional antigen-presenting cells that initiate and regulate the outcome of innate and adaptive immune responses. Immunostimulatory DCs are highly desired for the development of positive immunization techniques. DCs express functional NK1R; however, regardless of their potential DC-stimulatory function, the ability of NK1R agonists to promote immunostimulatory DCs remains unexplored. Here, we demonstrate that NK1R signaling activates therapeutic DCs capable of biasing type 1 immunity by inhibition of interleukin-10 (IL-10) synthesis and secretion, without affecting their low levels of IL-12 production. The potent type 1 effector immune response observed following cutaneous administration of NK1R-signaled DCs required their homing in skin-draining lymph nodes (sDLNs) where they induced inflammation and licensed endogenous-conventional sDLN-resident and -recruited inflammatory DCs to secrete IL-12. Our data demonstrate that NK1R signaling promotes immunostimulatory DCs, and provide relevant insight into the mechanisms used by neuromediators to regulate innate and adaptive immune responses.


The Journal of Allergy and Clinical Immunology | 2015

Autocrine hemokinin-1 functions as an endogenous adjuvant for IgE-mediated mast cell inflammatory responses

Tina L. Sumpter; Chin H. Ho; Anna R. Pleet; Olga Tkacheva; William J. Shufesky; Darling M. Rojas-Canales; Adrian E. Morelli; Adriana T. Larregina

BACKGROUNDnEfficient development of atopic diseases requires interactions between allergen and adjuvant to initiate and amplify the underlying inflammatory responses. Substance P (SP) and hemokinin-1 (HK-1) are neuropeptides that signal through the neurokinin-1 receptor (NK1R) to promote inflammation. Mast cells initiate the symptoms and tissue effects of atopic disorders, secreting TNF and IL-6 after FcεRI cross-linking by antigen-IgE complexes (FcεRI-activated mast cells [FcεRI-MCs]). Additionally, MCs express the NK1R, suggesting an adjuvant role for NK1R agonists in FcεRI-MC-mediated pathologies; however, in-depth research addressing this relevant aspect of MC biology is lacking.nnnOBJECTIVEnWe sought to investigate the effect of NK1R signaling and the individual roles of SP and HK-1 as potential adjuvants for FcεRI-MC-mediated allergic disorders.nnnMETHODSnBone marrow-derived mast cells (BMMCs) from C57BL/6 wild-type (WT) or NK1R(-/-) mice were used to investigate the effects of NK1R signaling on FcεRI-MCs. BMMCs generated from Tac1(-/-) mice or after culture with Tac4 small interfering RNA were used to address the adjuvancy of SP and HK-1. WT, NK1R(-/-), and c-Kit(W-sh/W-sh) mice reconstituted with WT or NK1R(-/-) BMMCs were used to evaluate NK1R signaling on FcεRI-MC-mediated passive local and systemic anaphylaxis and on airway inflammation.nnnRESULTSnFcεRI-activated MCs upregulated NK1R and HK-1 transcripts and protein synthesis, without modifying SP expression. In a positive signaling loop HK-1 promoted TNF and IL-6 secretion by MC degranulation and protein synthesis, the latter through the phosphoinositide 3-kinase/Akt/nuclear factor κB pathways. Inxa0vivo NK1R signaling was necessary for the development of passive local and systemic anaphylaxis and airway inflammation.nnnCONCLUSIONSnFcεRI stimulation of MCs promotes autocrine secretion of HK-1, which signals through NK1R to provide adjuvancy for efficient development of FcεRI-MC-mediated disorders.


Journal of Immunology | 2012

Coordinate Stimulation of Macrophages by Microparticles and TLR Ligands Induces Foam Cell Formation

Peter A. Keyel; Olga Tkacheva; Adriana T. Larregina; Russell D. Salter

Aberrant activation of macrophages in arterial walls by oxidized lipoproteins can lead to atherosclerosis. Oxidized lipoproteins convert macrophages to foam cells through lipid uptake and TLR signaling. To investigate the relative contributions of lipid uptake and TLR signaling in foam cell formation, we established an in vitro assay using liposomes of defined lipid compositions. We found that TLRs signaling through Toll/IL-1R domain-containing adapter inducing IFN-β promoted foam cell formation by inducing both NF-κB signaling and type I IFN production, whereas TLRs that do not induce IFN, like TLR2, did not enhance foam cell formation. Addition of IFN-α to TLR2 activator promoted robust foam cell formation. TLR signaling further required peroxisome proliferator-activated receptor α, as inhibition of peroxisome proliferator-activated receptor α blocked foam cell formation. We then investigated the ability of endogenous microparticles (MP) to contribute to foam cell formation. We found that lipid-containing MP promoted foam cell formation, which was enhanced by TLR stimulation or IFN-α. These MP also stimulated foam cell formation in a human skin model. However, these MP suppressed TNF-α production and T cell activation, showing that foam cell formation can occur by immunosuppressive MP. Taken together, the data reveal novel signaling requirements for foam cell formation and suggest that uptake of distinct types of MP in the context of activation of multiple distinct TLR can induce foam cell formation.


Methods of Molecular Biology | 2013

Methods of Analysis of Dendritic Cell-Derived Exosome-Shuttle MicroRNA and Its Horizontal Propagation Between Dendritic Cells

Angela Montecalvo; Adriana T. Larregina; Adrian E. Morelli

Exosomes are extremely small (<100 nm) membrane vesicles, generated in the endocytic compartment that are released to the extracellular milieu by living cells. Although the biological function of exosomes in vivo remains unclear, they seem to function as mechanisms of cell-to-cell communication for horizontal transfer of proteins, antigens, prions, morphogens, mRNA, and noncoding regulatory RNAs, including microRNAs (miRNAs) (also known as exosome-shuttle miRNAs). Dendritic cells (DCs), the most potent professional antigen-presenting leukocytes of the immune system, release relatively high levels of exosomes and also interact with free exosomes present in the extracellular space. Therefore, DCs constitute a good model for the analysis of exosome-shuttle miRNAs and their horizontal propagation between cells. This chapter provides basic protocols for purification of exosomes released by mouse bone marrow-derived DCs, analysis of their miRNA content, and assessment of the function of exosome-shuttle miRNAs, once they are transferred to target/acceptor DCs.


Methods of Molecular Biology | 2014

Methods of purification of CTL-derived exosomes.

Angela Montecalvo; Adriana T. Larregina; Adrian E. Morelli

Exosomes are membrane nanovesicles (approximately <120 nm in size) released by most, if not all, living cells and in particular by leukocytes. They originate within the endocytic compartment by invagination of the endosome membrane. Therefore, they have a different biogenesis and molecular composition than microvesicles (>0.2 μm) shed from the plasma membrane. Although the functions of exosomes in vivo are beginning to be elucidated, increasing evidence suggests that exosomes constitute a mechanism of cell-to-cell communication, transferring antigens, proteins, mRNAs, and noncoding RNAs among cells. Interestingly, effector T cells including cytotoxic T lymphocytes (CTLs) release death-inducing molecules of the TNF superfamily through exosomes contained in their cytotoxic granules. The present chapter provides basic protocols for purification of exosomes secreted by CTLs.

Collaboration


Dive into the Adriana T. Larregina's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Olga Tkacheva

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Carol A. Feghali-Bostwick

Medical University of South Carolina

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Geza Erdos

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Louis D. Falo

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge