Network


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

Hotspot


Dive into the research topics where Gianluca Carlesso is active.

Publication


Featured researches published by Gianluca Carlesso.


Science Translational Medicine | 2014

Cell Distance Mapping Identifies Functional T Follicular Helper Cells in Inflamed Human Renal Tissue

Vladimir M. Liarski; Natalya V. Kaverina; Anthony Chang; Daniel Brandt; Denisse Yanez; Lauren Talasnik; Gianluca Carlesso; Ronald Herbst; Tammy O. Utset; Christine M. Labno; Yahui Peng; Yulei Jiang; Maryellen L. Giger; Marcus R. Clark

Visualizing and quantifying the spatial relationships between T and B cells identifies adaptive immune cell networks in human inflammation. Putting Human Inflammation on the Map B cells cannot fight infection by antigen stimulation alone—they need help from T cells. In mice, two-photon electron microscopy has demonstrated that T follicular helper (TFH) cells are critical for providing B cell help in germinal centers. However, it has remained unclear whether—and if so, how—TFH cells provide B cell help in humans. Now, Liarski et al. report that cell distance mapping (CDM) can be used to demonstrate cognate TFH-mediated B cell help in the context of human inflammation. CDM is a computational tool that quantifies spatial relationships between different cell types in tissue. The authors used CDM to measure the internuclear distances between TFH and B cells in inflamed human tissues. They were able to discriminate between noncognate and cognate interactions, which are required for providing help. They also characterized cognate-competent TFH cells and found that they expressed Bcl-6 and IL-21. This technique should be generalizable to diverse antigen presentation and immune cell interactions and, if so, should enhance our knowledge of the immune system in situ. T follicular helper (TFH) cells are critical for B cell activation in germinal centers and are often observed in human inflamed tissue. However, it is difficult to know if they contribute in situ to inflammation. Expressed markers define TFH subsets associated with distinct functions in vitro. However, such markers may not reflect in situ function. The delivery of T cell help to B cells requires direct cognate recognition. We hypothesized that by visualizing and quantifying such interactions, we could directly assess TFH cell competency in situ. Therefore, we developed computational tools to quantify spatial relationships between different cell subtypes in tissue [cell distance mapping (CDM)]. Analysis of inflamed human tissues indicated that measurement of internuclear distances between TFH and B cells could be used to discriminate between apparent cognate and noncognate interactions. Furthermore, only cognate-competent TFH cell populations expressed high levels of Bcl-6 and interleukin-21. These data suggest that CDM can be used to identify adaptive immune cell networks driving in situ inflammation. Such knowledge should help identify diseases, and disease subsets, that may benefit from therapeutic targeting of specific T cell–antigen-presenting cell interactions.


Journal of Immunology | 2009

B Cell Receptor-Mediated Sustained c-Rel Activation Facilitates Late Transitional B Cell Survival through Control of B Cell Activating Factor Receptor and NF-κB2

Iris Castro; Jacqueline A. Wright; Bazarragchaa Damdinsuren; Kristen L. Hoek; Gianluca Carlesso; Nicholas P. Shinners; Rachel M. Gerstein; Robert T. Woodland; Ranjan Sen; Wasif N. Khan

Signaling from the BCR and B cell activating factor receptor (BAFF-R or BR3) differentially regulates apoptosis within early transitional (T1) and late transitional (T2; CD21int-T2) B cells during selection processes to generate mature B lymphocytes. However, molecular mechanisms underlying the differential sensitivity of transitional B cells to apoptosis remain unclear. In this study, we demonstrate that BCR signaling induced more long-term c-Rel activation in T2 and mature than in T1 B cells leading to increased expression of anti-apoptotic genes as well as prosurvival BAFF-R and its downstream substrate p100 (NF-κB2). Sustained c-Rel activation required de novo c-Rel gene transcription and translation via Btk-dependent mechanisms. Like T1 cells, mature B cells from Btk- and c-Rel-deficient mice also failed to activate these genes. These findings suggest that the gain of survival potential within transitional B cells is dependent on the ability to produce a long-term c-Rel response, which plays a critical role in T2 B cell survival and differentiation in vivo by inducing anti-apoptotic genes, BAFF-R and NF-κB2, an essential component for BAFF-R survival signaling. Thus, acquisition of resistance to apoptosis during transitional B cell maturation is achieved by integration of BCR and BAFF-R signals.


Immunity | 2017

Follicular Dendritic Cell Activation by TLR Ligands Promotes Autoreactive B Cell Responses.

Abhishek Das; Balthasar A. Heesters; Allison R. Bialas; Joseph O’Flynn; Ian R. Rifkin; Jordi Ochando; Nanette Mittereder; Gianluca Carlesso; Ronald Herbst; Michael C. Carroll

&NA; A hallmark of autoimmunity in murine models of lupus is the formation of germinal centers (GCs) in lymphoid tissues where self‐reactive B cells expand and differentiate. In the host response to foreign antigens, follicular dendritic cells (FDCs) maintain GCs through the uptake and cycling of complement‐opsonized immune complexes. Here, we examined whether FDCs retain self‐antigens and the impact of this process in autoantibody secretion in lupus. We found that FDCs took up and retained self‐immune complexes composed of ribonucleotide proteins, autoantibody, and complement. This uptake, mediated through CD21, triggered endosomal TLR7 and led to the secretion of interferon (IFN) &agr; via an IRF5‐dependent pathway. Blocking of FDC secretion of IFN‐&agr; restored B cell tolerance and reduced the amount of GCs and pathogenic autoantibody. Thus, FDCs are a critical source of the IFN‐&agr; driving autoimmunity in this lupus model. This pathway is conserved in humans, suggesting that it may be a viable therapeutic target in systemic lupus erythematosus. Graphical Abstract Figure. No caption available. HighlightsInternalization of RNP complexes via CD21 triggers TLR7 and IFN‐&agr; in mouse and human FDCsGC maintenance and &agr; anti‐nuclear antibody production are dependent on TLR7 pathway in FDCsLoss of B cell tolerance in RNP‐specific lupus mice is IFNAR dependentFDCs are an essential source of type I IFN in lupus mice &NA; Follicular dendritic cells (FDCs) maintain germinal centers through the uptake and cycling of complement‐opsonized immune complexes. Das et al. show that in a murine model of lupus, uptake of self‐immune complexes by FDCs activates TLR7 and that these stromal cells are a critical source of the IFN‐&agr; driving autoimmunity.


Journal of Immunology | 2009

Absence of mature peripheral B cell populations in mice with concomitant defects in B cell receptor and BAFF-R signaling.

Kristen L. Hoek; Gianluca Carlesso; Emily S. Clark; Wasif N. Khan

Generation of mature B lymphocytes from early (T1) and late transitional (T2) precursors requires cooperative signaling through BCR and B cell-activating factor receptor 3 (BR3). Recent studies have shown that BCR signaling positively regulates NF-κB2, suggesting BCR regulation of BR3 signaling. To investigate the significance of signal integration from BCR and BR3 in B cell development and function, we crossed Btk-deficient mice (btk−/−), which are developmentally blocked between the T2 and the mature follicular B cell stage as a result of a partial defect in BCR signaling, and A/WySnJ mice, which possess a mutant BR3 defective in propagating intracellular signals that results in a severely reduced peripheral B cell compartment, although all B cell subsets are present in relatively normal ratios. A/WySnJ × btk−/− mice display a B cell-autonomous defect, resulting in a developmental block at an earlier stage (T1) than either mutation alone, leading to the loss of mature splenic follicular and marginal zone B cells, as well as the loss of peritoneal B1 and B2 cell populations. The competence of the double mutant T1 B cells to respond to TLR4 and CD40 survival and activation signals is further attenuated compared with single mutations as evidenced by severely reduced humoral immune responses in vivo and proliferation in response to anti-IgM, LPS, and anti-CD40 stimulation in vitro. Thus, BCR and BR3 independently and in concert regulate the survival, differentiation, and function of all B cell populations at and beyond T1, earliest transitional stage.


PLOS Pathogens | 2016

Cytomegalovirus-specific IL-10-producing CD4(+) T cells are governed by Type-I IFN-induced IL-27 and promote virus persistence

Mathew Clement; Morgan Marsden; Maria A. Stacey; Juneid Abdul-Karim; Silvia Gimeno Brias; Diana Filipa Costa Bento; Martin John Scurr; Peter Ghazal; Casey T. Weaver; Gianluca Carlesso; Simon Clare; Simon Arnett Jones; Andrew James Godkin; Gareth Wyn Jones; Ian R. Humphreys

CD4+ T cells support host defence against herpesviruses and other viral pathogens. We identified that CD4+ T cells from systemic and mucosal tissues of hosts infected with the β-herpesviridae human cytomegalovirus (HCMV) or murine cytomegalovirus (MCMV) express the regulatory cytokine interleukin (IL)-10. IL-10+CD4+ T cells co-expressed TH1-associated transcription factors and chemokine receptors. Mice lacking T cell-derived IL-10 elicited enhanced antiviral T cell responses and restricted MCMV persistence in salivary glands and secretion in saliva. Thus, IL-10+CD4+ T cells suppress antiviral immune responses against CMV. Expansion of this T-cell population in the periphery was promoted by IL-27 whereas mucosal IL-10+ T cell responses were ICOS-dependent. Infected Il27rα-deficient mice with reduced peripheral IL-10+CD4+ T cell accumulation displayed robust T cell responses and restricted MCMV persistence and shedding. Temporal inhibition experiments revealed that IL-27R signaling during initial infection was required for the suppression of T cell immunity and control of virus shedding during MCMV persistence. IL-27 production was promoted by type-I IFN, suggesting that β-herpesviridae exploit the immune-regulatory properties of this antiviral pathway to establish chronicity. Further, our data reveal that cytokine signaling events during initial infection profoundly influence virus chronicity.


PLOS ONE | 2014

Germinal Center B Cell Depletion Diminishes CD4+ Follicular T Helper Cells in Autoimmune Mice

Isharat Yusuf; Jessica Stern; Tom M. McCaughtry; Sandra Gallagher; Hong Sun; Changshou Gao; Thomas F. Tedder; Gianluca Carlesso; Laura Carter; Ronald Herbst; Yue Wang

Background Continuous support from follicular CD4+ T helper (Tfh) cells drives germinal center (GC) responses, which last for several weeks to produce high affinity memory B cells and plasma cells. In autoimmune Sle1 and NZB/W F1 mice, elevated numbers of Tfh cells persist, promoting the expansion of self-reactive B cells. Expansion of circulating Tfh like cells have also been described in several autoimmune diseases. Although, the signals required for Tfh differentiation have now been well described, the mechanisms that sustain the maintenance of fully differentiated Tfh are less understood. Recent data demonstrate a role for GC B cells for Tfh maintenance after protein immunization. Methods and Finding Given the pathogenic role Tfh play in autoimmune disease, we explored whether B cells are required for maintenance of autoreactive Tfh. Our data suggest that the number of mature autoreactive Tfh cells is controlled by GC B cells. Depletion of B cells in Sle1 autoimmune mice leads to a dramatic reduction in Tfh cells. In NZB/W F1 autoimmune mice, similar to the SRBC immunization model, GC B cells support the maintenance of mature Tfh, which is dependent mainly on ICOS. The CD28-associated pathway is dispensable for Tfh maintenance in SRBC immunized mice, but is required in the spontaneous NZB/W F1 model. Conclusion These data suggest that mature Tfh cells require signals from GC B cells to sustain their optimal numbers and function in both autoimmune and immunization models. Thus, immunotherapies targeting B cells in autoimmune disease may affect pathogenic Tfh cells.


European Journal of Immunology | 2015

ICOS is required for the generation of both central and effector CD4+ memory T‐cell populations following acute bacterial infection

Clare L. Marriott; Gianluca Carlesso; Ronald Herbst; David R. Withers

Interactions between ICOS and ICOS ligand (ICOSL) are essential for the development of T follicular helper (Tfh) cells and thus the formation and maintenance of GC reactions. Given the conflicting reports on the requirement of other CD4+ T‐cell populations for ICOS signals, we have employed a range of in vivo approaches to dissect requirements for ICOS signals in mice during an endogenous CD4+ T‐cell response and contrasted this with CD28 signals. Genetic absence of ICOSL only modestly reduced the total number of antigen‐specific CD4+ T cells at the peak of the primary response, but resulted in a severely diminished number of both T central memory and T effector memory cells. Treatment with blocking anti‐ICOS mAb during the primary response recapitulated these effects and caused a more substantial reduction than blocking CD28 signals with CTLA4Ig. During the memory phase of the response further signals through ICOS or CD28 were not required for survival. However, upon secondary challenge only Tfh cell expansion remained heavily ICOS‐dependent, while CD28 signals were required for optimal expansion of all subsets. These data demonstrate the importance of ICOS signals specifically for memory CD4+ T‐cell formation, while highlighting the potential of therapeutically targeting this pathway.


Journal of Immunology | 2016

Loss of Immune Tolerance Is Controlled by ICOS in Sle1 Mice

Nanette Mittereder; Ellen Kuta; Geetha Bhat; Karma Dacosta; Lily Cheng; Ronald Herbst; Gianluca Carlesso

ICOS, a member of the CD28 family, represents a key molecule that regulates adaptive responses to foreign Ags. ICOS is prominently expressed on T follicular helper (TFH) cells, a specialized CD4+ T cell subset that orchestrates B cell differentiation within the germinal centers and humoral response. However, the contribution of ICOS and TFH cells to autoantibody profiles under pathological conditions has not been thoroughly investigated. We used the Sle1 lupus-prone mouse model to examine the role of ICOS in the expansion and function of pathogenic TFH cells. Genetic deletion of ICOS impacted the expansion of TFH cells in B6.Sle1 mice and inhibited the differentiation of B lymphocytes into plasma cells. The phenotypic changes observed in B6.Sle1-ICOS–knockout mice were also associated with a significant reduction in class-switched IgG, and anti-nucleosomal IgG-secreting B cells compared with B6.Sle1 animals. The level of vascular cell adhesion protein 1, a molecule that was shown to be elevated in patients with SLE and in lupus models, was also increased in an ICOS-dependent manner in Sle1 mice and correlated with autoantibody levels. The elimination of ICOS-expressing CD4+ T cells in B6.Sle1 mice, using a glyco-engineered anti-ICOS–depleting Ab, resulted in a significant reduction in anti-nucleosomal autoantibodies. Our results indicate that ICOS regulates the ontogeny and homeostasis of B6.Sle1 TFH cells and influences the function of TFH cells during aberrant germinal center B cell responses. Therapies targeting the ICOS signaling pathway may offer new opportunities for the treatment of lupus and other autoimmune diseases.


Science Translational Medicine | 2018

T follicular helper–like cells contribute to skin fibrosis

Devon Taylor; Nanette Mittereder; Ellen Kuta; Tracy Delaney; Timothy Burwell; Karma Dacosta; Weiguang Zhao; Lily Cheng; Charles Brown; Anmarie Boutrin; Xiang Guo; Wendy I. White; Jie Zhu; Huifang Dong; Michael A. Bowen; Jia Lin; Changshou Gao; Li Yu; Madhu Ramaswamy; Marie-Claude Gaudreau; Rob Woods; Ronald Herbst; Gianluca Carlesso

T follicular helper–like cells and IL-21 are drivers of skin fibrosis in systemic sclerosis. Scleroderma’s little helpers Scleroderma, also known as systemic sclerosis, is a devastating disease involving multi-organ fibrosis. Taylor et al. examined immune cells from patients and in the skin of a graft-versus-host disease–based mouse model to elucidate the key players in this disease. They observed that a subset of T follicular helper–like cells expressing inducible costimulator (ICOS) correlated with disease scores. Blocking these cells with anti-ICOS or anti–IL-21, an important T follicular helper cell cytokine, ameliorated disease in the mouse model. Targeting these not-so-helpful T cells could dampen dermal fibrosis and bring relief to scleroderma patients. Systemic sclerosis (SSc) is a debilitating inflammatory and fibrotic disease that affects the skin and internal organs. Although the pathophysiology of SSc remains poorly characterized, mononuclear cells, mainly macrophages and T cells, have been implicated in inflammation and fibrosis. Inducible costimulator (ICOS), which is expressed on a subset of memory T helper (TH) and T follicular helper (TFH) cells, has been shown to be increased in SSc and associated with disease pathology. However, the identity of the relevant ICOS+ T cells and their contribution to inflammation and fibrosis in SSc are still unknown. We show that CD4+ ICOS-expressing T cells with a TFH-like phenotype infiltrate the skin of patients with SSc and are correlated with dermal fibrosis and clinical disease status. ICOS+ TFH-like cells were found to be increased in the skin of graft-versus-host disease (GVHD)–SSc mice and contributed to dermal fibrosis via an interleukin-21– and matrix metalloproteinase 12–dependent mechanism. Administration of an anti-ICOS antibody to GVHD-SSc mice prevented the expansion of ICOS+ TFH-like cells and inhibited inflammation and dermal fibrosis. Interleukin-21 neutralization in GVHD-SSc mice blocked disease pathogenesis by reducing skin fibrosis. These results identify ICOS+ TFH-like profibrotic cells as key drivers of fibrosis in a GVHD-SSc model and suggest that inhibition of these cells could offer therapeutic benefit for SSc.


Reproductive Toxicology | 2017

Effects of ICOS+ T cell depletion via afucosylated monoclonal antibody MEDI-570 on pregnant cynomolgus monkeys and the developing offspring

Simone M. Nicholson; Gianluca Carlesso; Lily Cheng; Halie Cook; Karma Dacosta; Joel Leininger; Kathleen McKeever; Stephen (Weasel) Scott; Devon Taylor; Katie Streicher; Steve Eck; Molly Reed; Raffaella Faggioni; Ronald Herbst; Rakesh Dixit; Patricia C. Ryan

MEDI-570 is a fully human afucosylated monoclonal antibody (MAb) against Inducible T-cell costimulator (ICOS), highly expressed on CD4+ T follicular helper (TFH) cells. Effects of MEDI-570 were evaluated in an enhanced pre-postnatal development toxicity (ePPND) study in cynomolgus monkeys. Administration to pregnant monkeys did not cause any abortifacient effects. Changes in hematology and peripheral blood T lymphocyte subsets in maternal animals and infants and the attenuated infant IgG immune response to keyhole limpet hemocyanin (KLH) were attributed to MEDI-570 pharmacology. Adverse findings included aggressive fibromatosis in one dam and two infant losses in the high dose group with anatomic pathology findings suggestive of atypical lymphoid hyperplasia. The margin of safety relative to the no observed adverse effect level (NOAEL) for the highest planned clinical dose in the Phase 1a study was 7. This study suggests that women of child bearing potential employ effective methods of contraception while being treated with MEDI-570.

Collaboration


Dive into the Gianluca Carlesso's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge