Network


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

Hotspot


Dive into the research topics where Sandra Lopez-Verges is active.

Publication


Featured researches published by Sandra Lopez-Verges.


Proceedings of the National Academy of Sciences of the United States of America | 2011

Expansion of a unique CD57+NKG2Chi natural killer cell subset during acute human cytomegalovirus infection

Sandra Lopez-Verges; Jeffrey M. Milush; Brian S. Schwartz; Marcelo J. Pando; Jessica Jarjoura; Vanessa A. York; Jeffrey P. Houchins; Steve Miller; Sang-Mo Kang; Phillip J. Norris; Douglas F. Nixon; Lewis L. Lanier

During human CMV infection, there is a preferential expansion of natural killer (NK) cells expressing the activating CD94–NKG2C receptor complex, implicating this receptor in the recognition of CMV-infected cells. We hypothesized that NK cells expanded in response to pathogens will be marked by expression of CD57, a carbohydrate antigen expressed on highly mature cells within the CD56dimCD16+ NK cell compartment. Here we demonstrate the preferential expansion of a unique subset of NK cells coexpressing the activating CD94–NKG2C receptor and CD57 in CMV+ donors. These CD57+NKG2Chi NK cells degranulated in response to stimulation through their NKG2C receptor. Furthermore, CD57+NKG2Chi NK cells preferentially lack expression of the inhibitory NKG2A receptor and the inhibitory KIR3DL1 receptor in individuals expressing its HLA-Bw4 ligand. Moreover, in solid-organ transplant recipients with active CMV infection, the percentage of CD57+NKG2Chi NK cells in the total NK cell population preferentially increased. During acute CMV infection, the NKG2C+ NK cells proliferated, became NKG2Chi, and finally acquired CD57. Thus, we propose that CD57 might provide a marker of “memory” NK cells that have been expanded in response to infection.


Blood | 2010

CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset

Sandra Lopez-Verges; Jeffrey M. Milush; Suchitra Pandey; Vanessa A. York; Janice Arakawa-Hoyt; Hanspeter Pircher; Phillip J. Norris; Douglas F. Nixon; Lewis L. Lanier

Natural killer (NK) cells are innate immune lymphocytes that express a heterogeneous repertoire of germline-encoded receptors and undergo a distinct pattern of maturation. CD57 is a marker of terminal differentiation on human CD8(+) T cells. Very few newborn or fetal NK cells express CD57; however, the frequency of CD57-bearing NK cells increases with age. We assessed the transcriptional, phenotypic, and functional differences between CD57(+) and CD57(-) NK cells within the CD56(dim) mature NK subset. CD57(+) NK cells express a repertoire of NK-cell receptors, suggestive of a more mature phenotype, and proliferate less when stimulated with target cells and/or cytokines. By contrast, a higher frequency of CD57(+) NK cells produced interferon-γ and demonstrated more potent lytic activity when these cells were stimulated through the activating receptor CD16; however, they are less responsive to stimulation by interleukin-12 and interleukin-18. Finally, CD57 expression is induced on CD57(-)CD56(dim) NK cells after activation by interleukin-2. A combination of a mature phenotype, a higher cytotoxic capacity, a higher sensitivity to stimulation via CD16, with a decreased responsiveness to cytokines, and a decreased capacity to proliferate suggest that CD57(+) NK cells are highly mature and might be terminally differentiated.


Blood | 2012

Tim-3 marks human natural killer cell maturation and suppresses cell-mediated cytotoxicity

Lishomwa C. Ndhlovu; Sandra Lopez-Verges; Jason D. Barbour; Richard Bradley Jones; Aashish R. Jha; Brian R. Long; Schoeffler Ec; Tsuyoshi Fujita; Douglas F. Nixon; Lewis L. Lanier

Natural killer (NK) cells are innate lymphocytes that play an important role against viral infections and cancer. This effect is achieved through a complex mosaic of inhibitory and activating receptors expressed by NK cells that ultimately determine the magnitude of the NK-cell response. The T-cell immunoglobulin- and mucin domain-containing (Tim)-3 receptor was initially identified as a T-helper 1-specific type I membrane protein involved in regulating T-cell responses. Human NK cells transcribe the highest amounts of Tim-3 among lymphocytes. Tim-3 protein is expressed on essentially all mature CD56(dim)CD16(+) NK cells and is expressed heterogeneously in the immature CD56(bright)CD16(-) NK-cell subset in blood from healthy adults and in cord blood. Tim-3 expression was induced on CD56(bright)CD16(-) NK cells after stimulation with IL-15 or IL-12 and IL-18 in vitro, suggesting that Tim-3 is a maturation marker on NK cells. Whereas Tim-3 has been used to identify dysfunctional T cells, NK cells expressing high amounts of Tim-3 are fully responsive with respect to cytokine production and cytotoxicity. However, when Tim-3 was cross-linked with antibodies it suppressed NK cell-mediated cytotoxicity. These findings suggest that NK-cell responses may be negatively regulated when NK cells encounter target cells expressing cognate ligands of Tim-3.


Journal of Immunology | 2011

NK Cells and Immune “Memory”

Joseph C. Sun; Sandra Lopez-Verges; Charles C. Kim; Joseph L. DeRisi; Lewis L. Lanier

Immunological memory is a hallmark of the adaptive immune system. However, the ability to remember and respond more robustly against a second encounter with the same pathogen has been described in organisms lacking T and B cells. Recently, NK cells have been shown to mediate Ag-specific recall responses in several different model systems. Although NK cells do not rearrange the genes encoding their activating receptors, NK cells experience a selective education process during development, undergo a clonal-like expansion during virus infection, generate long-lived progeny (i.e., memory cells), and mediate more efficacious secondary responses against previously encountered pathogens—all characteristics previously ascribed only to T and B cells in mammals. This review describes past findings leading up to these new discoveries, summarizes the evidence for and characteristics of NK cell memory, and discusses the attempts and future challenges to identify these long-lived memory NK cell populations in humans.


Retrovirology | 2013

CD56negCD16+ NK cells are activated mature NK cells with impaired effector function during HIV-1 infection.

Jeffrey M. Milush; Sandra Lopez-Verges; Vanessa A. York; Steven G. Deeks; Jeffrey N. Martin; Frederick Hecht; Lewis L. Lanier; Douglas F. Nixon

BackgroundA subset of CD3negCD56negCD16+ Natural Killer (NK) cells is highly expanded during chronic HIV-1 infection. The role of this subset in HIV-1 pathogenesis remains unclear. The lack of NK cell lineage-specific markers has complicated the study of minor NK cell subpopulations.ResultsUsing CD7 as an additional NK cell marker, we found that CD3negCD56negCD16+ cells are a heterogeneous population comprised of CD7+ NK cells and CD7neg non-classical myeloid cells. CD7+CD56negCD16+ NK cells are significantly expanded in HIV-1 infection. CD7+CD56negCD16+ NK cells are mature and express KIRs, the C-type lectin-like receptors NKG2A and NKG2C, and natural cytotoxicity receptors similar to CD7+CD56+CD16+ NK cells. CD7+CD56neg NK cells in healthy donors produced minimal IFNγ following K562 target cell or IL-12 plus IL-18 stimulation; however, they degranulated in response to K562 stimulation similar to CD7+CD56+ NK cells. HIV-1 infection resulted in reduced IFNγ secretion following K562 or cytokine stimulation by both NK cell subsets compared to healthy donors. Decreased granzyme B and perforin expression and increased expression of CD107a in the absence of stimulation, particularly in HIV-1-infected subjects, suggest that CD7+CD56negCD16+ NK cells may have recently engaged target cells. Furthermore, CD7+CD56negCD16+ NK cells have significantly increased expression of CD95, a marker of NK cell activation.ConclusionsTaken together, CD7+CD56negCD16+ NK cells are activated, mature NK cells that may have recently engaged target cells.


Cancer immunology research | 2014

CALGB 150905 (Alliance): Rituximab Broadens the Antilymphoma Response by Activating Unlicensed NK Cells

Juan Du; Sandra Lopez-Verges; Brandelyn N. Pitcher; Jeffrey L. Johnson; Sin-Ho Jung; Lili Zhou; Katharine C. Hsu; Myron S. Czuczman; Bruce D. Cheson; Lawrence D. Kaplan; Lewis L. Lanier; Jeffrey M. Venstrom

Du and colleagues report that a “missing ligand” genotype predictive of unlicensed NK cells was associated with higher progression-free survival in 101 follicular lymphoma patients treated with rituximab-containing mAb combinations, and that rituximab triggered responses in vitro from healthy-donor unlicensed NK cells. Natural killer (NK) cells contribute to clinical responses in patients treated with rituximab, but the rules determining NK-cell responsiveness to mAb therapies are poorly defined. A deeper understanding of the mechanisms responsible for antibody-dependent cellular cytotoxicity (ADCC) could yield useful biomarkers for predicting clinical responses in patients. Unlicensed NK cells, defined as NK cells lacking expression of an inhibitory KIR for self-HLA class I ligands, are hyporesponsive in steady state, but are potent effectors in inflammatory conditions. We hypothesized that antitumor antibodies such as rituximab can overcome NK-cell dependence on licensing, making unlicensed NK cells important for clinical responses. Here, we examined the influences of variations in KIR and HLA class I alleles on in vitro responses to rituximab. We tested the clinical significance in a cohort of patients with follicular lymphoma treated with rituximab-containing mAb combinations, and show that rituximab triggers responses from all NK-cell populations regardless of licensing. Neither IL2 nor accessory cells are required for activating unlicensed NK cells, but both can augment rituximab-mediated ADCC. Moreover, in 101 patients with follicular lymphoma treated with rituximab-containing mAb combinations, a “missing ligand” genotype (predictive of unlicensed NK cells) is associated with a higher rate of progression-free survival. Our data suggest that the clinical efficacy of rituximab may be driven, in part, by its ability to broaden the NK-cell repertoire to include previously hyporesponsive, unlicensed NK cells. A “missing ligand” KIR and HLA class I genotype may be predictive of this benefit and useful for personalizing treatment decisions in lymphomas and other tumors. Cancer Immunol Res; 2(9); 878–89. ©2014 AACR.


Nature Medicine | 2017

A distinct innate lymphoid cell population regulates tumor-associated T cells

Crome Sq; Linh T. Nguyen; Sandra Lopez-Verges; Yang Sy; Martin B; Yam Jy; Johnson Dj; Nie J; Pniak M; Yen Ph; Milea A; Sowamber R; Katz; Marcus Q. Bernardini; Blaise Clarke; Patricia Shaw; Philipp A. Lang; Hal K. Berman; Trevor J. Pugh; Lewis L. Lanier; Pamela S. Ohashi

Antitumor T cells are subject to multiple mechanisms of negative regulation. Recent findings that innate lymphoid cells (ILCs) regulate adaptive T cell responses led us to examine the regulatory potential of ILCs in the context of cancer. We identified a unique ILC population that inhibits tumor-infiltrating lymphocytes (TILs) from high-grade serous tumors, defined their suppressive capacity in vitro, and performed a comprehensive analysis of their phenotype. Notably, the presence of this CD56+CD3− population in TIL cultures was associated with reduced T cell numbers, and further functional studies demonstrated that this population suppressed TIL expansion and altered TIL cytokine production. Transcriptome analysis and phenotypic characterization determined that regulatory CD56+CD3− cells exhibit low cytotoxic activity, produce IL-22, and have an expression profile that overlaps with those of natural killer (NK) cells and other ILCs. NKp46 was highly expressed by these cells, and addition of anti-NKp46 antibodies to TIL cultures abrogated the ability of these regulatory ILCs to suppress T cell expansion. Notably, the presence of these regulatory ILCs in TIL cultures corresponded with a striking reduction in the time to disease recurrence. These studies demonstrate that a previously uncharacterized ILC population regulates the activity and expansion of tumor-associated T cells.


Frontiers in Immunology | 2014

NK Cells during Dengue Disease and Their Recognition of Dengue Virus-Infected cells

Davis Beltrán; Sandra Lopez-Verges

The innate immune response, in addition to the B- and T-cell response, plays a role in protection against dengue virus (DENV) infection and the degree of disease severity. Early activation of natural killer (NK) cells and type-I interferon-dependent immunity may be important in limiting viral replication during the early stages of DENV infection and thus reducing subsequent pathogenesis. NK cells may also produce cytokines that reduce inflammation and tissue injury. On the other hand, NK cells are also capable of inducing liver injury at early-time points of DENV infection. In vitro, NK cells can kill antibody-coated DENV-infected cells through antibody-dependent cell-mediated cytotoxicity. In addition, NK cells may directly recognize DENV-infected cells through their activating receptors, although the increase in HLA class I expression may allow infected cells to escape the NK response. Recently, genome-wide association studies have shown an association between MICB and MICA, which encode ligands of the activating NK receptor NKG2D, and dengue disease outcome. This review focuses on recognition of DENV-infected cells by NK cells and on the regulation of expression of NK cell ligands by DENV.


Frontiers in Immunology | 2017

Editorial: NK Cells in Human Diseases: Friends or Foes?

Vincent Vieillard; Bree Foley; Sandra Lopez-Verges

Natural killer (NK) cells are lymphocytes from the innate immune system that play a protective role against tumors and viral infections. Activated human NK cells respond by killing infected or tumor cells and can shape the adaptive response by producing cytokines and chemokines and by interacting with dendritic cells and other immune cells, and also have memory-like features (1). For these reasons, NK cells are principally considered as protective cells. However, recent discoveries have found that NK cells can have a negative role due to overproduction of proinflammatory cytokines, destruction of host cells in autoimmune diseases, in graft-versus-host diseases, or in regulating other immune cells that provide protective immunity, e.g., in some cancers where NK cells counteract tumor-specific infiltrating T cells. The aim of this research topic is to analyze the different situations in which NK cells could have a beneficial or a deleterious role and what are the signals and mechanisms that could explain these differences in behavior. In this era of immunetherapy, understanding of NK-cell biology and its relation to human disease is crucial for rational design of treatments that are efficient but also are safe.


Blood | 2012

Cytomegalovirus reactivation after allogeneic transplantation promotes a lasting increase in educated NKG2C natural killer cells with potent function

Bree Foley; Sarah Cooley; Michael R. Verneris; Michelle Pitt; Julie Curtsinger; Xianghua Luo; Sandra Lopez-Verges; Lewis L. Lanier; Daniel J. Weisdorf; Jeffrey S. Miller

Collaboration


Dive into the Sandra Lopez-Verges's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Douglas F. Nixon

George Washington University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sang-Mo Kang

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Phillip J. Norris

Systems Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge