Network


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

Hotspot


Dive into the research topics where Susana L. Silva is active.

Publication


Featured researches published by Susana L. Silva.


Blood | 2011

First use of thymus transplantation therapy for FOXN1 deficiency (nude/SCID): a report of 2 cases.

M. Louise Markert; José G. Marques; Bénédicte Neven; Blythe H. Devlin; Elizabeth A. McCarthy; Ivan K. Chinn; Adriana S. Albuquerque; Susana L. Silva; Claudio Pignata; Geneviève de Saint Basile; Rui M. M. Victorino; Capucine Picard; Marianne Debré; Nizar Mahlaoui; Alain Fischer; Ana E. Sousa

FOXN1 deficiency is a primary immunodeficiency characterized by athymia, alopecia totalis, and nail dystrophy. Two infants with FOXN1 deficiency were transplanted with cultured postnatal thymus tissue. Subject 1 presented with disseminated Bacillus Calmette-Guérin infection and oligoclonal T cells with no naive markers. Subject 2 had respiratory failure, human herpes virus 6 infection, cytopenias, and no circulating T cells. The subjects were given thymus transplants at 14 and 9 months of life, respectively. Subject 1 received immunosuppression before and for 10 months after transplantation. With follow up of 4.9 and 2.9 years, subjects 1 and 2 are well without infectious complications. The pretransplantation mycobacterial disease in subject 1 and cytopenias in subject 2 resolved. Subject 2 developed autoimmune thyroid disease 1.6 years after transplantation. Both subjects developed functional immunity. Subjects 1 and 2 have 1053/mm(3) and 1232/mm(3) CD3(+) cells, 647/mm(3) and 868/mm(3) CD4(+) T cells, 213/mm(3) and 425/mm(3) naive CD4(+) T cells, and 10 200 and 5700 T-cell receptor rearrangement excision circles per 100 000 CD3(+) cells, respectively. They have normal CD4 T-cell receptor β variable repertoires. Both subjects developed antigen-specific proliferative responses and have discontinued immunoglobulin replacement. In summary, thymus transplantation led to T-cell reconstitution and function in these FOXN1 deficient infants.


PLOS ONE | 2011

Primary B-Cell Deficiencies Reveal a Link between Human IL-17-Producing CD4 T-Cell Homeostasis and B-Cell Differentiation

Rita R. Barbosa; Sara Pereira da Silva; Susana L. Silva; Alcinda Campos Melo; Elisa Pedro; Manuel Pereira Barbosa; M. Conceição Pereira-Santos; Rui M. M. Victorino; Ana E. Sousa

IL-17 is a pro-inflammatory cytokine implicated in autoimmune and inflammatory conditions. The development/survival of IL-17-producing CD4 T cells (Th17) share critical cues with B-cell differentiation and the circulating follicular T helper subset was recently shown to be enriched in Th17 cells able to help B-cell differentiation. We investigated a putative link between Th17-cell homeostasis and B cells by studying the Th17-cell compartment in primary B-cell immunodeficiencies. Common Variable Immunodeficiency Disorders (CVID), defined by defects in B-cell differentiation into plasma and memory B cells, are frequently associated with autoimmune and inflammatory manifestations but we found no relationship between these and Th17-cell frequency. In fact, CVID patients showed a decrease in Th17-cell frequency in parallel with the expansion of activated non-differentiated B cells (CD21lowCD38low). Moreover, Congenital Agammaglobulinemia patients, lacking B cells due to impaired early B-cell development, had a severe reduction of circulating Th17 cells. Finally, we found a direct correlation in healthy individuals between circulating Th17-cell frequency and both switched-memory B cells and serum BAFF levels, a crucial cytokine for B-cell survival. Overall, our data support a relationship between Th17-cell homeostasis and B-cell maturation, with implications for the understanding of the pathogenesis of inflammatory/autoimmune diseases and the physiology of B-cell depleting therapies.


PLOS ONE | 2012

Human FOXN1-deficiency is associated with αβ double-negative and FoxP3+ T-cell expansions that are distinctly modulated upon thymic transplantation.

Adriana S. Albuquerque; José G. Marques; Susana L. Silva; Dário Ligeiro; Blythe H. Devlin; Jacques Dutrieux; Rémi Cheynier; Claudio Pignata; Rui M. M. Victorino; M. Louise Markert; Ana E. Sousa

Forkhead box N1 (FOXN1) is a transcription factor crucial for thymic epithelium development and prevention of its involution. Investigation of a patient with a rare homozygous FOXN1 mutation (R255X), leading to alopecia universalis and thymus aplasia, unexpectedly revealed non-maternal circulating T-cells, and, strikingly, large numbers of aberrant double-negative αβ T-cells (CD4negCD8neg, DN) and regulatory-like T-cells. These data raise the possibility that a thymic rudiment persisted, allowing T-cell development, albeit with disturbances in positive/negative selection, as suggested by DN and FoxP3+ cell expansions. Although regulatory-like T-cell numbers normalized following HLA-mismatched thymic transplantation, the αβDN subset persisted 5 years post-transplantation. Involution of thymus allograft likely occurred 3 years post-transplantation based on sj/βTREC ratio, which estimates intrathymic precursor T-cell divisions and, consequently, thymic explant output. Nevertheless, functional immune-competence was sustained, providing new insights for the design of immunological reconstitution strategies based on thymic transplantation, with potential applications in other clinical settings.


Clinical and Experimental Immunology | 2012

Monocyte activation is a feature of common variable immunodeficiency irrespective of plasma lipopolysaccharide levels

Rita R. Barbosa; Sara Pereira da Silva; Susana L. Silva; Rita Tendeiro; Alcinda Campos Melo; Elisa Pedro; Manuel Pereira Barbosa; M. C. P. Santos; Rui M. M. Victorino; Ana E. Sousa

Common variable immunodeficiency disorders (CVID), the most frequent cause of symptomatic primary immunodeficiency, are defined by impaired antibody production. Notwithstanding, T cell activation and granulomatous manifestations represent the main causes of CVID morbidity even in patients receiving immunoglobulin (Ig) G replacement therapy. Additionally, gut pathology is a frequent feature of CVID. In this study, we investigated monocyte imbalances and their possible relationship with increased microbial translocation in CVID patients. Monocyte subsets were defined according to CD14 and CD16 expression levels and evaluated in terms of human leucocyte antigen D‐related (HLA‐DR), CD86 and programmed death‐1 molecule ligand 1 (PD‐L1) expression by flow cytometry, in parallel with the quantification of plasma lipopolysaccharide (LPS) and serum levels of soluble CD14 (sCD14), LPS‐binding protein (LBP) and anti‐LPS antibodies. CVID patients (n = 31) featured significantly increased levels of serum sCD14 and an expansion of CD14brightCD16+ monocytes in direct correlation with T cell and B cell activation, the latter illustrated by the frequency of the CD21lowCD38low subset. Such alterations were not observed in patients lacking B cells due to congenital agammaglobulinaemia (n = 4). Moreover, we found no significant increase in circulating LPS or LBP levels in CVID patients, together with a relative preservation of serum anti‐LPS antibodies, in agreement with their presence in commercial IgG preparations. In conclusion, CVID was associated with monocyte imbalances that correlated directly with T cell activation markers and with B cell imbalances, without an association with plasma LPS levels. The heightened monocyte activated state observed in CVID may represent an important target for complementary therapeutic strategies.


Science immunology | 2017

Human blood Tfr cells are indicators of ongoing humoral activity not fully licensed with suppressive function

Valter R. Fonseca; Ana Agua-Doce; Ana Raquel Maceiras; Wim Pierson; Filipa Ribeiro; Vasco C. Romão; Ana R. Pires; Susana L. Silva; João Eurico Fonseca; Ana E. Sousa; Michelle A. Linterman; Luis Graca

Circulating human T follicular regulatory (Tfr) cells are distinct from tissue Tfr cells. Suppressing Sjögren syndrome T follicular regulatory (Tfr) cells regulate antibody production in the germinal center, yet individuals with the autoimmune disease Sjögren syndrome have increased numbers of circulating Tfr cells compared with healthy individuals. Fonseca et al. compared blood Tfr cells with tissue Tfr cells and found that blood Tfr cells were phenotypically distinct from their tissue counterparts. Moreover, blood Tfr cells did not preferentially suppress humoral responses and had a naïve-like phenotype. These cells were not thymically derived but were generated during germinal center responses, exiting the tissue to enter the blood. These data explain why increased number of blood Tfr cells does not correlate with increased suppression potential and suggest that, instead, increased numbers of blood Tfr cells indicate ongoing humoral activity. Germinal center (GC) responses are controlled by T follicular helper (Tfh) and T follicular regulatory (Tfr) cells and are crucial for the generation of high-affinity antibodies. Although the biology of human circulating and tissue Tfh cells has been established, the relationship between blood and tissue Tfr cells defined as CXCR5+Foxp3+ T cells remains elusive. We found that blood Tfr cells are increased in Sjögren syndrome, an autoimmune disease with ongoing GC reactions, especially in patients with high autoantibody titers, as well as in healthy individuals upon influenza vaccination. Although blood Tfr cells correlated with humoral responses, they lack full B cell–suppressive capacity, despite being able to suppress T cell proliferation. Blood Tfr cells have a naïve-like phenotype, although they are absent from human thymus or cord blood. We found that these cells were generated in peripheral lymphoid tissues before T-B interaction, as they are maintained in B cell–deficient patients. Therefore, blood CXCR5+Foxp3+ T cells in human pathology indicate ongoing humoral activity but are not fully competent circulating Tfr cells.


Journal of Clinical Immunology | 2014

Reduced BAFF-R and increased TACI expression in common variable immunodeficiency.

Rita R. Barbosa; Susana L. Silva; Sara Pereira da Silva; Alcinda Campos Melo; M. Conceição Pereira-Santos; João T. Barata; Lennart Hammarström; Marilia Cascalho; Ana E. Sousa

PurposeB-cell survival and differentiation critically depend on the interaction of BAFF-R and TACI with their ligands, BAFF and APRIL. Mature B-cell defects lead to Common Variable Immunodeficiency (CVID), which is associated with elevated serum levels of BAFF and APRIL. Nevertheless, BAFF-R and TACI expression in CVID and their relationship with ligand availability remain poorly understood.Methods and ResultsWe found that BAFF-R expression was dramatically reduced on B cells of CVID patients, relative to controls. BAFF-R levels inversely correlated with serum BAFF concentration both in CVID and healthy subjects. We also found that recombinant BAFF stimulation reduced BAFF-R expression on B cells without decreasing transcript levels. On the other hand, CVID subjects had increased TACI expression on B cells in direct association with serum BAFF but not APRIL levels. Moreover, splenomegaly was associated with higher TACI expression, suggesting that perturbations of TACI function may underlie lymphoproliferation in CVID.ConclusionsOur results indicate that availability of BAFF determines BAFF-R and TACI expression on B cells, and that BAFF-R expression is controlled by BAFF binding. Identification of the factors governing BAFF-R and TACI is crucial to understanding CVID pathogenesis, and B-cell biology in general, as well as to explore their potential as therapeutic targets.


PLOS ONE | 2017

Autoimmunity and allergy control in adults submitted to complete thymectomy early in infancy

Susana L. Silva; Adriana S. Albuquerque; Andreia Amaral; Quan Zhen Li; Catarina Mota; Rémi Cheynier; Rui M. M. Victorino; M. Conceição Pereira-Santos; Ana E. Sousa

The contribution of the decline in thymic activity for the emergence of autoimmunity is still debatable. Immune-competent adults submitted to complete thymectomy early in life provide a unique model to address this question. We applied here strict criteria to identify adults lacking thymic activity based on sjTREC levels, to exclude thymic rebound and/or ectopic thymuses. In agreement, they featured severe naïve CD4 T-cell depletion and contraction of T-cell receptor diversity. Notwithstanding this, there was neither increased incidence of autoimmune disease in comparison with age-matched controls nor significant changes in their IgG/IgA/IgM/IgE autoreactivity profiles, as assessed through extensive arrays. We reasoned that the observed relative preservation of the regulatory T-cell compartment, including maintenance of naïve regulatory CD4 T-cells, may contribute to limit the emergence of autoimmunity upon thymectomy. Our findings have implications in other clinical settings with impaired thymic activity, and are particularly relevant to studies of autoimmunity in ageing.


Frontiers in Immunology | 2017

IL-7-Induced Proliferation of Human Naive CD4 T-Cells Relies on Continued Thymic Activity

Susana L. Silva; Adriana S. Albuquerque; Paula Matoso; Bénédicte Charmeteau-de-Muylder; Rémi Cheynier; Dário Ligeiro; Miguel Abecasis; Rui Anjos; João T. Barata; Rui M. M. Victorino; Ana E. Sousa

Naive CD4 T-cell maintenance is critical for immune competence. We investigated here the fine-tuning of homeostatic mechanisms of the naive compartment to counteract the loss of de novo CD4 T-cell generation. Adults thymectomized in early childhood during corrective cardiac surgery were grouped based on presence or absence of thymopoiesis and compared with age-matched controls. We found that the preservation of the CD31− subset was independent of the thymus and that its size is tightly controlled by peripheral mechanisms, including prolonged cell survival as attested by Bcl-2 levels. Conversely, a significant contraction of the CD31+ naive subset was observed in the absence of thymic activity. This was associated with impaired responses of purified naive CD4 T-cells to IL-7, namely, in vitro proliferation and upregulation of CD31 expression, which likely potentiated the decline in recent thymic emigrants. Additionally, we found no apparent constraint in the differentiation of naive cells into the memory compartment in individuals completely lacking thymic activity despite upregulation of DUSP6, a phosphatase associated with increased TCR threshold. Of note, thymectomized individuals featuring some degree of thymopoiesis were able to preserve the size and diversity of the naive CD4 compartment, further arguing against complete thymectomy in infancy. Overall, our data suggest that robust peripheral mechanisms ensure the homeostasis of CD31− naive CD4 pool and point to the requirement of continuous thymic activity to the maintenance of IL-7-driven homeostatic proliferation of CD31+ naive CD4 T-cells, which is essential to secure T-cell diversity throughout life.


Oncotarget | 2016

Human naïve regulatory T-cells feature high steady-state turnover and are maintained by IL-7

Susana L. Silva; Adriana S. Albuquerque; Ana Serra-Caetano; Russell B. Foxall; Ana R. Pires; Paula Matoso; Susana M. Fernandes; João Ferreira; Rémi Cheynier; Rui M. M. Victorino; Íris Caramalho; João T. Barata; Ana E. Sousa

Naïve FoxP3-expressing regulatory T-cells (Tregs) are essential to control immune responses via continuous replenishment of the activated-Treg pool with thymus-committed suppressor cells. The mechanisms underlying naïve-Treg maintenance throughout life in face of the age-associated thymic involution remain unclear. We found that in adults thymectomized early in infancy the naïve-Treg pool is remarkably well preserved, in contrast to conventional naïve CD4 T-cells. Naïve-Tregs featured high levels of cycling and pro-survival markers, even in healthy individuals, and contrasted with other circulating naïve/memory CD4 T-cell subsets in terms of their strong γc-cytokine-dependent signaling, particularly in response to IL-7. Accordingly, ex-vivo stimulation of naïve-Tregs with IL-7 induced robust cytokine-dependent signaling, Bcl-2 expression, and phosphatidylinositol 3-kinase (PI3K)-dependent proliferation, whilst preserving naïve phenotype and suppressive capacity. Altogether, our data strongly implicate IL-7 in the thymus-independent long-term survival of functional naïve-Tregs, and highlight the potential of targeting the IL-7 pathway to modulate Tregs in different clinical settings.


Frontiers in Pediatrics | 2016

Establishment and Maintenance of the Human Naïve CD4+ T-Cell Compartment

Susana L. Silva; Ana E. Sousa

The naïve CD4+ T-cell compartment is considered essential to guarantee immune competence throughout life. Its replenishment with naïve cells with broad diverse receptor repertoire, albeit with reduced self-reactivity, is ensured by the thymus. Nevertheless, cumulative data support a major requirement of post-thymic proliferation both for the establishment of the human peripheral naïve compartment during the accelerated somatic growth of childhood, as well as for its lifelong maintenance. Additionally, a dynamic equilibrium is operating at the cell level to fine-tune the T-cell receptor threshold to activation and survival cues, in order to counteract the continuous naïve cell loss by death or conversion into memory/effector cells. The main players in these processes are low-affinity self-peptide/MHC and cytokines, particularly IL-7. Moreover, although naïve CD4+ T-cells are usually seen as a homogeneous population regarding stage of maturation and cell differentiation, increasing evidence points to a variety of phenotypic and functional subsets with distinct homeostatic requirements. The paradigm of cells committed to a distinct lineage in the thymus are the naïve regulatory T-cells, but other functional subpopulations have been identified based on their time span after thymic egress, phenotypic markers, such as CD31, or cytokine production, namely IL-8. Understanding the regulation of these processes is of utmost importance to promote immune reconstitution in several clinical settings, namely transplantation, persistent infections, and aging. In this mini review, we provide an overview of the mechanisms underlying human naïve CD4+ T-cell homeostasis, combining clinical data, experimental studies, and modeling approaches.

Collaboration


Dive into the Susana L. Silva's collaboration.

Top Co-Authors

Avatar

Ana E. Sousa

Instituto de Medicina Molecular

View shared research outputs
Top Co-Authors

Avatar

Rui M. M. Victorino

Instituto de Medicina Molecular

View shared research outputs
Top Co-Authors

Avatar

Adriana S. Albuquerque

Instituto de Medicina Molecular

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sara Pereira da Silva

Instituto de Medicina Molecular

View shared research outputs
Top Co-Authors

Avatar

Carla Costa

Instituto Nacional de Saúde Dr. Ricardo Jorge

View shared research outputs
Top Co-Authors

Avatar

S. Costa

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Alcinda Campos Melo

Instituto de Medicina Molecular

View shared research outputs
Top Co-Authors

Avatar

Amélia Spínola Santos

Rio de Janeiro State University

View shared research outputs
Researchain Logo
Decentralizing Knowledge