Network


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

Hotspot


Dive into the research topics where Frances E. Lund is active.

Publication


Featured researches published by Frances E. Lund.


Immunity | 2016

T Follicular Helper Cell Plasticity Shapes Pathogenic T Helper 2 Cell-Mediated Immunity to Inhaled House Dust Mite

Andre Ballesteros-Tato; Troy D. Randall; Frances E. Lund; Rosanne Spolski; Warren J. Leonard; Beatriz León

Exposure to environmental antigens, such as house dust mite (HDM), often leads to T helper 2 (Th2) cell-driven allergic responses. However, the mechanisms underlying the development of these responses are incompletely understood. We found that the initial exposure to HDM did not lead to Th2 cell development but instead promoted the formation of interleukin-4 (IL-4)-committed T follicular helper (Tfh) cells. Following challenge exposure to HDM, Tfh cells differentiated into IL-4 and IL-13 double-producing Th2 cells that accumulated in the lung and recruited eosinophils. B cells were required to expand IL-4-committed Tfh cells during the sensitization phase, but did not directly contribute to disease. Impairment of Tfh cell responses during the sensitization phase or Tfh cell depletion prevented Th2 cell-mediated responses following challenge. Thus, our data demonstrate that Tfh cells are precursors of HDM-specific Th2 cells and reveal an unexpected role of B cells and Tfh cells in the pathogenesis of allergic asthma.


Journal of Experimental Medicine | 2017

Blimp-1-dependent and -independent natural antibody production by B-1 and B-1-derived plasma cells

Hannah P. Savage; Vanessa Yenson; Sanjam S. Sawhney; Betty J. Mousseau; Frances E. Lund; Nicole Baumgarth

Natural antibodies contribute to tissue homeostasis and protect against infections. They are secreted constitutively without external antigenic stimulation. The differentiation state and regulatory pathways that enable continuous natural antibody production by B-1 cells, the main cellular source in mice, remain incompletely understood. Here we demonstrate that natural IgM-secreting B-1 cells in the spleen and bone marrow are heterogeneous, consisting of (a) terminally differentiated B-1–derived plasma cells expressing the transcriptional regulator of differentiation, Blimp-1, (b) Blimp-1+, and (c) Blimp-1neg phenotypic B-1 cells. Blimp-1neg IgM-secreting B-1 cells are not simply intermediates of cellular differentiation. Instead, they secrete similar amounts of IgM in wild-type and Blimp-1–deficient (PRDM-1&Dgr;Ex1A) mice. Blimp-1neg B-1 cells are also a major source of IgG3. Consequently, deletion of Blimp-1 changes neither serum IgG3 levels nor the amount of IgG3 secreted per cell. Thus, the pool of natural antibody-secreting B-1 cells is heterogeneous and contains a distinct subset of cells that do not use Blimp-1 for initiation or maximal antibody secretion.


Annals of Neurology | 2015

Alzheimer's disease pathology is attenuated in a CD38-deficient mouse model.

Eran Blacher; Tulin Dadali; Alina Bespalko; Viola J. Haupenthal; Marcus O. W. Grimm; Tobias Hartmann; Frances E. Lund; Reuven Stein; Ayelet Levy

Alzheimers disease (AD)‐associated dementia is due to tissue damage caused by amyloid β (Aβ) deposition within the brain and by accompanying neuroinflammation. The nicotinamide adenine dinucleotide (NAD) glycohydrolase CD38, which is expressed by neurons, astrocytes, and microglial cells, regulates inflammatory and repair processes in the brain and other tissues by degrading NAD and repressing the activity of other NAD‐consuming enzymes and by producing NAD‐derived metabolites that regulate calcium signaling and migration of inflammatory cells. Given the role of CD38 in neuroinflammation and repair, we examined the effect of CD38 deletion on AD pathology.


Journal of Immunology | 2014

Dendritic Cells and B Cells: Unexpected Partners in Th2 Development

Beatriz León; Andre Ballesteros-Tato; Frances E. Lund

Although we have known for decades that B cells contribute to immune responses by secreting Ab, it is now clear that they are more than simply factories for Ig production, and they also play key roles as modulators of T cell–dependent immunity. Indeed, the evidence showing that Ag-presenting and cytokine-producing B cells can alter the magnitude and quality of CD4 T cell responses continues to grow. In this article, we review the data showing that B cells, working in partnership with dendritic cells, regulate the development of Th2 cells and the subsequent allergic response.


Journal of Immunology | 2015

B Lymphocytes Are Required during the Early Priming of CD4+ T Cells for Clearance of Pneumocystis Infection in Mice

Michael M. Opata; Melissa Hollifield; Frances E. Lund; Troy D. Randall; Robert Dunn; Beth A. Garvy; David J. Feola

B cells play a critical role in the clearance of Pneumocystis. In addition to production of Pneumocystis-specific Abs, B cells are required during the priming phase for CD4+ T cells to expand normally and generate memory. Clearance of Pneumocystis was found to be dependent on Ag specific B cells and on the ability of B cells to secrete Pneumocystis-specific Ab, as mice with B cells defective in these functions or with a restricted BCR were unable to control Pneumocystis infection. Because Pneumocystis-specific antiserum was only able to partially protect B cell–deficient mice from infection, we hypothesized that optimal T cell priming requires fully functional B cells. Using adoptive transfer and B cell depletion strategies, we determined that optimal priming of CD4+ T cells requires B cells during the first 2–3 d of infection and that this was independent of the production of Ab. T cells that were removed from Pneumocystis-infected mice during the priming phase were fully functional and able to clear Pneumocystis infection upon adoptive transfer into Rag1−/− hosts, but this effect was ablated in mice that lacked fully functional B cells. Our results indicate that T cell priming requires a complete environment of Ag presentation and activation signals to become fully functional in this model of Pneumocystis infection.


Journal of Biological Chemistry | 2013

Cluster of differentiation 38 (CD38) mediates bile-acid induced acinar cell injury and pancreatitis through cyclic ADP ribose and intracellular calcium release

Abrahim I. Orabi; Kamaldeen A. Muili; Tanveer A. Javed; Shunqian Jin; Thottala Jayaraman; Frances E. Lund; Sohail Z. Husain

Background: Bile acids cause ryanodine receptor (RyR) Ca2+ release and lead to injury in pancreatic acinar cells, yet the mechanism is unknown. Results: Inhibition of the RyR activator cADPR reduces bile acid-induced acinar cell Ca2+ release, cell injury, and pancreatitis. Conclusion: CD38-cADPR facilitates bile-induced Ca2+ release, cell injury, and pancreatitis. Significance: The CD38-cADPR pathway may serve as a target for the treatment of some forms of pancreatitis. Aberrant Ca2+ signals within pancreatic acinar cells are an early and critical feature in acute pancreatitis, yet it is unclear how these signals are generated. An important mediator of the aberrant Ca2+ signals due to bile acid exposure is the intracellular Ca2+ channel ryanodine receptor. One putative activator of the ryanodine receptor is the nucleotide second messenger cyclic ADP-ribose (cADPR), which is generated by an ectoenzyme ADP-ribosyl cyclase, CD38. In this study, we examined the role of CD38 and cADPR in acinar cell Ca2+ signals and acinar injury due to bile acids using pharmacologic inhibitors of CD38 and cADPR as well as mice deficient in Cd38 (Cd38−/−). Cytosolic Ca2+ signals were imaged using live time-lapse confocal microscopy in freshly isolated mouse acinar cells during perifusion with the bile acid taurolithocholic acid 3-sulfate (TLCS; 500 μm). To focus on intracellular Ca2+ release and to specifically exclude Ca2+ influx, cells were perifused in Ca2+-free medium. Cell injury was assessed by lactate dehydrogenase leakage and propidium iodide uptake. Pretreatment with either nicotinamide (20 mm) or the cADPR antagonist 8-Br-cADPR (30 μm) abrogated TLCS-induced Ca2+ signals and cell injury. TLCS-induced Ca2+ release and cell injury were reduced by 30 and 95%, respectively, in Cd38-deficient acinar cells compared with wild-type cells (p < 0.05). Cd38-deficient mice were protected against a model of bile acid infusion pancreatitis. In summary, these data indicate that CD38-cADPR mediates bile acid-induced pancreatitis and acinar cell injury through aberrant intracellular Ca2+ signaling.


American Journal of Physiology-lung Cellular and Molecular Physiology | 2015

Airway responsiveness in CD38-deficient mice in allergic airway disease: studies with bone marrow chimeras

A. Guedes; Joseph A. Jude; Jaime Paulin; Laura Rivero-Nava; Hirohito Kita; Frances E. Lund; Mathur S. Kannan

CD38 is a cell-surface protein involved in calcium signaling and contractility of airway smooth muscle. It has a role in normal airway responsiveness and in airway hyperresponsiveness (AHR) developed following airway exposure to IL-13 and TNF-α but appears not to be critical to airway inflammation in response to the cytokines. CD38 is also involved in T cell-mediated immune response to protein antigens. In this study, we assessed the contribution of CD38 to AHR and inflammation to two distinct allergens, ovalbumin and the epidemiologically relevant environmental fungus Alternaria. We also generated bone marrow chimeras to assess whether Cd38(+/+) inflammatory cells would restore AHR in the CD38-deficient (Cd38(-/-)) hosts following ovalbumin challenge. Results show that wild-type (WT) mice develop greater AHR to inhaled methacholine than Cd38(-/-) mice following challenge with either allergen, with comparable airway inflammation. Reciprocal bone marrow transfers did not change the native airway phenotypic differences between WT and Cd38(-/-) mice, indicating that the lower airway reactivity of Cd38(-/-) mice stems from Cd38(-/-) lung parenchymal cells. Following bone marrow transfer from either source and ovalbumin challenge, the phenotype of Cd38(-/-) hosts was partially reversed, whereas the airway phenotype of the WT hosts was preserved. Airway inflammation was similar in Cd38(-/-) and WT chimeras. These results indicate that loss of CD38 on hematopoietic cells is not sufficient to prevent AHR and that the magnitude of airway inflammation is not the predominant underlying determinant of AHR in mice.


Immunity | 2018

Distinct Effector B Cells Induced by Unregulated Toll-like Receptor 7 Contribute to Pathogenic Responses in Systemic Lupus Erythematosus

Scott A. Jenks; Kevin S. Cashman; Esther Zumaquero; Urko M. Marigorta; Aakash V. Patel; Xiaoqian Wang; Deepak Tomar; Matthew Woodruff; Zoe Simon; Regina Bugrovsky; Emily L. Blalock; Christopher D. Scharer; Christopher Tipton; Chungwen Wei; S. Sam Lim; Michelle Petri; Timothy B. Niewold; Jennifer H. Anolik; Greg Gibson; F. Eun-Hyung Lee; Jeremy M. Boss; Frances E. Lund; Ignacio Sanz

Graphical Abstract Figure. No caption available. SUMMARY Systemic Lupus Erythematosus (SLE) is characterized by B cells lacking IgD and CD27 (double negative; DN). We show that DN cell expansions reflected a subset of CXCR5‐ CD11c+ cells (DN2) representing pre‐plasma cells (PC). DN2 cells predominated in African‐American patients with active disease and nephritis, anti‐Smith and anti‐RNA autoantibodies. They expressed a T‐bet transcriptional network; increased Toll‐like receptor‐7 (TLR7); lacked the negative TLR regulator TRAF5; and were hyper‐responsive to TLR7. DN2 cells shared with activated naive cells (aNAV), phenotypic and functional features, and similar transcriptomes. Their PC differentiation and autoantibody production was driven by TLR7 in an interleukin‐21 (IL‐21)‐mediated fashion. An in vivo developmental link between aNAV, DN2 cells, and PC was demonstrated by clonal sharing. This study defines a distinct differentiation fate of autoreactive naive B cells into PC precursors with hyper‐responsiveness to innate stimuli, as well as establishes prominence of extra‐follicular B cell activation in SLE, and identifies therapeutic targets. HIGHLIGHTSAutoreactive CD27‐ IgD‐ CXCR5‐ CD11c+ (DN2) B cells expand in lupus patientsDN2 cells derive from naive cells and are poised to generate plasmablastsDN2 B cells are hyper‐responsive to Toll‐like receptor‐7 signalingThe properties of SLE DN2 B cells stem from distinct transcriptional networks &NA; The role of extrafollicular B cells in human systemic lupus is unknown. Jenks et al. define the main components of this pathway and its prominence in severe disease. Its activation is mediated by hyper‐responsiveness to Toll‐like receptor‐7 and leads to the generation of autoreactive antibody‐secreting plasmablasts.


Nature Communications | 2018

Factors of the bone marrow microniche that support human plasma cell survival and immunoglobulin secretion

Doan C. Nguyen; Swetha Garimalla; Haopeng Xiao; Shuya Kyu; Igor Albizua; Jacques Galipeau; Kuang-Yueh Chiang; Edmund K. Waller; Ronghu Wu; Greg Gibson; James R. Roberson; Frances E. Lund; Troy D. Randall; Iñaki Sanz; F. Eun-Hyung Lee

Human antibody-secreting cells (ASC) in peripheral blood are found after vaccination or infection but rapidly apoptose unless they migrate to the bone marrow (BM). Yet, elements of the BM microenvironment required to sustain long-lived plasma cells (LLPC) remain elusive. Here, we identify BM factors that maintain human ASC > 50 days in vitro. The critical components of the cell-free in vitro BM mimic consist of products from primary BM mesenchymal stromal cells (MSC), a proliferation-inducing ligand (APRIL), and hypoxic conditions. Comparative analysis of protein–protein interactions between BM-MSC proteomics with differential RNA transcriptomics of blood ASC and BM LLPC identify two major survival factors, fibronectin and YWHAZ. The MSC secretome proteins and hypoxic conditions play a role in LLPC survival utilizing mechanisms that downregulate mTORC1 signaling and upregulate hypoxia signatures. In summary, we identify elements of the BM survival niche critical for maturation of blood ASC to BM LLPC.Antibody-secreting cells (ASC) such as plasma cells must migrate to the bone marrow to survive, but microniche elements that promote survival are unknown. Here the authors define specific factors from the microniche that can maintain ASC in vitro for over 50 days, involving MSC secretome proteins, APRIL, and hypoxic conditions.


Carcinogenesis | 2018

CD38 knockout suppresses tumorigenesis in mice and clonogenic growth of human lung cancer cells

Xiangning Bu; Jiro Kato; Julie A. Hong; Maria J. Merino; David S. Schrump; Frances E. Lund; Joel Moss

The ectodomain of the plasma membrane ectoenzyme CD38 functions as both an NAD glycohydrolase and an ADP-ribosyl cyclase by catalyzing, respectively, the conversion of NAD to nicotinamide and ADP-ribose or cyclic ADP-ribose. CD38 is attracting particular attention in cancer therapy. An anti-CD38 monoclonal antibody (daratumumab) was approved for treatment of patients with multiple myeloma. However, the role of CD38 in non-hematological malignancies has not been explored. Previously, we reported that ADP-ribose-acceptor hydrolase (ARH)-1 deficiency in mice was associated with tumor development. In the present study, we found that in wild-type and ARH1-deficient mice deletion of the CD38 gene reduced tumor formation. Significant reductions in tumor number were observed in lymphomas, adenocarcinomas and hemangio/histolytic sarcomas. Consistent with a role for CD38 in tumorigenesis, CRISPR/Cas9-based knockout of CD38 in A549 human adenocarcinoma cells inhibited anchorage-independent cell growth, cell invasion and xenograft growth in nude mice. CD38 mRNA and protein expression were evaluated in human lung cancer cell lines and in human lung cancer specimens. CD38 overexpression in tumor cells was identified in 11 of 27 patient samples. In addition, some human lung cancer cell lines had dramatically higher CD38 mRNA and protein expression than normal cells. Consistent with these observations, search of the Oncomine database showed that some human lung adenocarcinomas had higher CD38 mRNA levels compared to normal lung tissues. In total, our data are consistent with the conclusion that CD38 plays a role in murine and human lung tumorigenesis and that anti-CD38 treatment may have therapeutic potential in lung cancer.

Collaboration


Dive into the Frances E. Lund's collaboration.

Top Co-Authors

Avatar

Davide Botta

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Troy D. Randall

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Andre Ballesteros-Tato

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Beatriz León

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Betty J. Mousseau

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Sara L Stone

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

Tulin Dadali

University of Alabama at Birmingham

View shared research outputs
Top Co-Authors

Avatar

A. Guedes

University of Minnesota

View shared research outputs
Top Co-Authors

Avatar

Anna Manouvakhova

Southern Research Institute

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge