27 results on '"Kaushansky, Alexis"'
Search Results
2. Malaria parasites target the hepatocyte receptor EphA2 for successful host infection.
- Author
-
Kaushansky, Alexis, Douglass, Alyse N., Arang, Nadia, Vigdorovich, Vladimir, Dambrauskas, Nicholas, Kain, Heather S., Austin, Laura S., Sather, D. Noah, and Kappe, Stefan H. I.
- Subjects
- *
MALARIA , *LIVER cells , *PLASMODIUM , *SPOROZOITES , *ANOPHELES , *PROTOZOAN diseases - Abstract
The invasion of a suitable host hepatocyte by mosquito-transmitted Plasmodium sporozoites is an essential early step in successful malaria parasite infection. Yet precisely how sporozoites target their host cell and facilitate productive infection remains largely unknown. We found that the hepatocyte EphA2 receptor was critical for establishing a permissive intracellular replication compartment, the parasitophorous vacuole. Sporozoites productively infected hepatocytes with high EphA2 expression, and the deletion of EphA2 protected mice from liver infection. Lack of host EphA2 phenocopied the lack of the sporozoite proteins P52 and P36. Our data suggest that P36 engages EphA2, which is likely to be a key step in establishing the permissive replication compartment. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
3. Selection and refinement: the malaria parasite's infection and exploitation of host hepatocytes.
- Author
-
Kaushansky, Alexis and Kappe, Stefan HI
- Subjects
- *
PLASMODIUM , *APICOMPLEXA , *SPOROZOITES , *LIVER cells , *HOST-parasite relationships - Abstract
Plasmodium parasites belong to the Apicomplexan phylum, which consists mostly of obligate intracellular pathogens that vary dramatically in host cell tropism. Plasmodium sporozoites are highly hepatophilic. The specific molecular mechanisms, which facilitate sporozoite selection and successful infection of hepatocytes, remain poorly defined. Here, we discuss the parasite and host factors which are critical to hepatocyte infection. We derive a model where sporozoites initially select host cells that constitute a permissive environment and then further refine the chosen hepatocyte during liver stage development, ensuring life cycle progression. While many unknowns of pre-erythrocytic infection remain, advancing models and technologies that enable analysis of human malaria parasites and of single infected cells will catalyze a comprehensive understanding of the interaction between the malaria parasite and its hepatocyte host. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
4. Susceptibility to P lasmodium liver stage infection is altered by hepatocyte polyploidy.
- Author
-
Austin, Laura S., Kaushansky, Alexis, and Kappe, Stefan H. I.
- Subjects
- *
DISEASE susceptibility , *PLASMODIUM , *POLYPLOIDY , *LIVER cells , *PARASITIC diseases , *LIVER diseases , *MAMMAL diseases - Abstract
P lasmodium parasites infect hepatocytes of their mammalian hosts and undergo obligate liver stage development. The specific host cell attributes that are important for liver infection remain largely unknown. Several host signalling pathways are perturbed in infected hepatocytes, some of which are important in the generation of hepatocyte polyploidy. To test the functional consequence of polyploidy on liver infection, we infected hepatocytes with the rodent malaria parasite P lasmodium yoelii both in vitro and in vivo and examined the ploidy of infected and uninfected hepatocytes by flow cytometry. In both hepatoma cell lines and in the mouse liver, the fraction of polyploid cells was higher in the infected cell population than in the uninfected cell population. When the data were reanalysed by comparing the extent of P lasmodium infection within each ploidy subset, we found that infection rates were elevated in more highly polyploid cells and lower in diploid cells. Furthermore, we found that the parasite's preference for host cells with high ploidy is conserved among rodent malaria species and the human malaria parasite P lasmodium falciparum. This parasite preference for host cells of high ploidy cannot be explained by differences in hepatocyte size or DNA replication. We conclude that P lasmodium preferentially infects and develops in polyploid hepatocytes. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
5. Of men in mice: the success and promise of humanized mouse models for human malaria parasite infections.
- Author
-
Kaushansky, Alexis, Mikolajczak, Sebastian A., Vignali, Marissa, and Kappe, Stefan H. I.
- Subjects
- *
PLASMODIUM , *ANTIPARASITIC agents , *PARASITIC diseases , *DRUG development , *LABORATORY mice ,RISK of malaria - Abstract
Forty percent of people worldwide are at risk of malaria infection, and despite control efforts it remains the most deadly parasitic disease. Unfortunately, rapid discovery and development of new interventions for malaria are hindered by the lack of small animal models that support the complex life cycles of the main parasite species infecting humans. Such tools must accommodate human parasite tropism for human tissue. Mouse models with human tissue developed to date have already enhanced our knowledge of human parasites, and are useful tools for assessing anti-parasitic interventions. Although these systems are imperfect, their continued refinement will likely broaden their utility. Some of the malaria parasite's interactions with human hepatocytes and human erythrocytes can already be modelled with available humanized mouse systems. However, interactions with other relevant human tissues such as the skin and immune system, as well as most transitions between life cycle stages in vivo will require refinement of existing humanized mouse models. Here, we review the recent successes achieved in modelling human malaria parasite biology in humanized mice, and discuss how these models have potential to become a valuable part of the toolbox used for understanding the biology of, and development of interventions to, malaria. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
6. Development of a quantitative flow cytometry-based assay to assess infection by Plasmodium falciparum sporozoites
- Author
-
Kaushansky, Alexis, Rezakhani, Nastaran, Mann, Henning, and Kappe, Stefan H.I.
- Subjects
- *
PLASMODIUM falciparum , *SPOROZOITES , *ETIOLOGY of diseases , *FLOW cytometry , *CLINICAL trials , *IMMUNOGLOBULINS ,MALARIA transmission - Abstract
Abstract: The human malaria parasite Plasmodium falciparum causes the most deadly parasitic disease worldwide, necessitating the development of interventions that block infection. Yet, preclinical assays to measure inhibition of infection date from the 1980s and are based on microscopy. Here, we describe the development of a simple flow cytometric assay that can be used to quantitatively assess P. falciparum sporozoite infection in vitro in low and medium throughput. We demonstrate the utility of this assay for assessing both drug inhibition of infection and measuring efficacy of antibodies in blocking parasite infection. This methodology will aid in assessing functional antibody responses to vaccination and novel drugs that prevent mosquito-to-man transmission of malaria. [Copyright &y& Elsevier]
- Published
- 2012
- Full Text
- View/download PDF
7. Tensin2 is a novel mediator in thrombopoietin (TPO)-induced cellular proliferation by promoting Akt signaling.
- Author
-
Jung, Andre Scott, Kaushansky, Alexis, MacBeath, Gavin, and Kaushansky, Kenneth
- Published
- 2011
- Full Text
- View/download PDF
8. System-wide Investigation of ErbB4 Reveals 19 Sites of Tyr Phosphorylation that Are Unusually Selective in Their Recruitment Properties
- Author
-
Kaushansky, Alexis, Gordus, Andrew, Budnik, Bogdan A., Lane, William S., Rush, John, and MacBeath, Gavin
- Subjects
- *
HUMAN genetics , *PHOSPHORYLATION , *PROTEIN-tyrosine kinases , *TANDEM mass spectrometry , *PROTEIN microarrays , *ONCOGENES - Abstract
Summary: The first three members of the ErbB family of receptor tyrosine kinases activate a wide variety of signaling pathways and are frequently misregulated in cancer. Much less is known about ErbB4. Here we use tandem mass spectrometry to identify 19 sites of tyrosine phosphorylation on ErbB4, and protein microarrays to quantify biophysical interactions between these sites and virtually every SH2 and PTB domain encoded in the human genome. Our unbiased approach highlighted several previously unrecognized interactions and led to the finding that ErbB4 can recruit and activate STAT1. At a systems level, we found that ErbB4 is much more selective than the other ErbB receptors. This suggests that ErbB4 may enable ErbB2 and ErbB3 to signal independently of EGFR under normal conditions, and provides a possible explanation for the protective properties of ErbB4 in cancer. [Copyright &y& Elsevier]
- Published
- 2008
- Full Text
- View/download PDF
9. The crucial role of hepatocyte growth factor receptor during liver-stage infection is not conserved among Plasmodium species.
- Author
-
Kaushansky, Alexis and Kappe, Stefan H I
- Subjects
- *
LETTERS to the editor , *HEPATOCYTE growth factor , *LIVER diseases - Abstract
A letter to the editor is presented regarding a study on the crucial role played by hepatocyte growth factor (HGF) receptor in liver-stage infection.
- Published
- 2011
- Full Text
- View/download PDF
10. Viral protein engagement of GBF1 induces host cell vulnerability through synthetic lethality.
- Author
-
Navare, Arti T., Mast, Fred D., Olivier, Jean Paul, Bertomeu, Thierry, Neal, Maxwell L., Carpp, Lindsay N., Kaushansky, Alexis, Coulombe-Huntington, Jasmin, Tyers, Mike, and Aitchison, John D.
- Subjects
- *
RNA virus infections , *SYNTHETIC proteins , *PROTEIN-protein interactions - Abstract
Viruses co-opt host proteins to carry out their lifecycle. Repurposed host proteins may thus become functionally compromised; a situation analogous to a loss-of-function mutation. We term such host proteins as viral-induced hypomorphs. Cells bearing cancer driver loss-of-function mutations have successfully been targeted with drugs perturbing proteins encoded by the synthetic lethal (SL) partners of cancer-specific mutations. Similarly, SL interactions of viral-induced hypomorphs can potentially be targeted as host-based antiviral therapeutics. Here, we use GBF1, which supports the infection of many RNA viruses, as a proof-of-concept. GBF1 becomes a hypomorph upon interaction with the poliovirus protein 3A. Screening for SL partners of GBF1 revealed ARF1 as the top hit, disruption of which selectively killed cells that synthesize 3A alone or in the context of a poliovirus replicon. Thus, viral protein interactions can induce hypomorphs that render host cells selectively vulnerable to perturbations that leave uninfected cells otherwise unscathed. Exploiting viral-induced vulnerabilities could lead to broad-spectrum antivirals for many viruses, including SARS-CoV-2. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
11. Germinal center activity and B cell maturation are associated with protective antibody responses against Plasmodium pre-erythrocytic infection.
- Author
-
Visweswaran, Ganesh Ram R., Vijayan, Kamalakannan, Chandrasekaran, Ramyavardhanee, Trakhimets, Olesya, Brown, Samantha L., Vigdorovich, Vladimir, Yang, Ashton, Raappana, Andrew, Watson, Alex, Selman, William, Zuck, Meghan, Dambrauskas, Nicholas, Kaushansky, Alexis, and Sather, D. Noah
- Subjects
- *
ANTIBODY formation , *GERMINAL centers , *IMMUNOLOGIC memory , *PATHOLOGY , *CIRCUMSPOROZOITE protein , *IMMUNOGLOBULIN G , *B cells - Abstract
Blocking Plasmodium, the causative agent of malaria, at the asymptomatic pre-erythrocytic stage would abrogate disease pathology and prevent transmission. However, the lack of well-defined features within vaccine-elicited antibody responses that correlate with protection represents a major roadblock to improving on current generation vaccines. We vaccinated mice (BALB/cJ and C57BL/6J) with Py circumsporozoite protein (CSP), the major surface antigen on the sporozoite, and evaluated vaccine-elicited humoral immunity and identified immunological factors associated with protection after mosquito bite challenge. Vaccination achieved 60% sterile protection and otherwise delayed blood stage patency in BALB/cJ mice. In contrast, all C57BL/6J mice were infected similar to controls. Protection was mediated by antibodies and could be passively transferred from immunized BALB/cJ mice into naïve C57BL/6J. Dissection of the underlying immunological features of protection revealed early deficits in antibody titers and polyclonal avidity in C57BL/6J mice. Additionally, PyCSP-vaccination in BALB/cJ induced a significantly higher proportion of antigen-specific B-cells and class-switched memory B-cell (MBCs) populations than in C57BL/6J mice. Strikingly, C57BL/6J mice also had markedly fewer CSP-specific germinal center experienced B cells and class-switched MBCs compared to BALB/cJ mice. Analysis of the IgG γ chain repertoires by next generation sequencing in PyCSP-specific memory B-cell repertoires also revealed higher somatic hypermutation rates in BALB/cJ mice than in C57BL/6J mice. These findings indicate that the development of protective antibody responses in BALB/cJ mice in response to vaccination with PyCSP was associated with increased germinal center activity and somatic mutation compared to C57BL/6J mice, highlighting the key role B cell maturation may have in the development of vaccine-elicited protective antibodies against CSP. Author summary: Identifying specific features of vaccine-elicited antibody responses that are associated with protection from malaria infection is a key step toward the development of a safe and effective vaccine. Here we compared antibody and B cell responses in two mouse strains that exhibited a differential ability to generate antibodies that protect from infection challenge. We found that protection was due to the presence of vaccine-elicited antibodies and could be transferred between strains, and that the ability of antibodies to neutralize the parasite was directly linked to the strength (affinity) with which it binds CSP. Thus, we sought to understand if there were differences in the two strains in the process of B cell maturation that leads to generation of high affinity, protective antibody responses after vaccination. Overall, our comparative analysis indicates that germinal center (GC) activity, a key process in B cell maturation, was significantly diminished in the non-protected strain. Further, we observed evidence of higher levels of somatic mutation, which is a result of germinal center activity, in protected mice. Thus, our results indicate that the ability to generate protective antibody responses was linked to enhanced B cell maturation in the protected strain, providing a key clue to the type of responses that should be generated by future vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
12. Crippling life support for SARS-CoV-2 and other viruses through synthetic lethality.
- Author
-
Mast, Fred D., Navare, Arti T., van der Sloot, Almer M., Coulombe-Huntington, Jasmin, Rout, Michael P., Baliga, Nitin S., Kaushansky, Alexis, Chait, Brian T., Aderem, Alan, Rice, Charles M., Sali, Andrej, Tyers, Mike, and Aitchison, John D.
- Subjects
- *
SARS-CoV-2 , *VIRUS diseases , *SYNTHETIC biology , *CHICKEN diseases , *COMMUNICABLE diseases - Abstract
With the rapid global spread of SARS-CoV-2, we have become acutely aware of the inadequacies of our ability to respond to viral epidemics. Although disrupting the viral life cycle is critical for limiting viral spread and disease, it has proven challenging to develop targeted and selective therapeutics. Synthetic lethality offers a promising but largely unexploited strategy against infectious viral disease; as viruses infect cells, they abnormally alter the cell state, unwittingly exposing new vulnerabilities in the infected cell. Therefore, we propose that effective therapies can be developed to selectively target the virally reconfigured host cell networks that accompany altered cellular states to cripple the host cell that has been converted into a virus factory, thus disrupting the viral life cycle. [ABSTRACT FROM AUTHOR]
- Published
- 2020
- Full Text
- View/download PDF
13. Spatial presentation of biological molecules to cells by localized diffusive transfer.
- Author
-
Regier, Mary C., Olszewski, Emily, Carter, Christoph C., Aitchison, John D., Kaushansky, Alexis, Davis, Jennifer, Berthier, Erwin, Beebe, David J., and Stevens, Kelly R.
- Subjects
- *
BIOMOLECULES , *DIFFUSION kinetics , *CELL aggregation , *FLUID flow , *ZIKA virus , *GENETIC recombination - Abstract
Cellular decisions in human development, homeostasis, regeneration, and disease are coordinated in large part by signals that are spatially localized in tissues. These signals are often soluble, such that biomolecules produced by one cell diffuse to receiving cells. To recapitulate soluble factor patterning in vitro, several microscale strategies have been developed. However, these techniques often introduce new variables into cell culture experiments (e.g., fluid flow) or are limited in their ability to pattern diverse solutes in a user-defined manner. To address these challenges, we developed an adaptable method that facilitates spatial presentation of biomolecules across cells in traditional open cultures in vitro. This technique employs device inserts that are placed in standard culture wells, which support localized diffusive pattern transmission through microscale spaces between device features and adherent cells. Devices can be removed and cultures can be returned to standard media following patterning. We use this method to spatially control cell labeling with pattern features ranging in scale from several hundred microns to millimeters and with sequential application of multiple patterns. To better understand the method we investigate relationships between pattern fidelity, device geometry, and consumption and diffusion kinetics using finite element modeling. We then apply the method to spatially defining reporter cell heterogeneity by patterning a small molecule modulator of genetic recombination with the requisite sustained exposure. Finally, we demonstrate use of this method for patterning larger and more slowly diffusing particles by creating focal sites of gene delivery and infection with adenoviral, lentiviral, and Zika virus particles. Thus, our method leverages devices that interface with standard culture vessels to pattern diverse diffusible factors, geometries, exposure dynamics, and recipient cell types, making it well poised for adoption by researchers across various fields of biological research. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
14. Opportunities for Host-targeted Therapies for Malaria.
- Author
-
Glennon, Elizabeth K.K., Dankwa, Selasi, Smith, Joseph D., and Kaushansky, Alexis
- Subjects
- *
MALARIA , *ARTEMISININ , *PATHOGENIC microorganisms , *POLYPHARMACY , *THERAPEUTICS - Abstract
Despite the recent successes of artemisinin-based antimalarial drugs, many still die from severe malaria, and eradication efforts are hindered by the limited drugs currently available to target transmissible gametocyte parasites and liver-resident dormant Plasmodium vivax hypnozoites. Host-targeted therapy is a new direction for infectious disease drug development and aims to interfere with host molecules, pathways, or networks that are required for infection or that contribute to disease. Recent advances in our understanding of host pathways involved in parasite development and pathogenic mechanisms in severe malaria could facilitate the development of host-targeted interventions against Plasmodium infection and malaria disease. This review discusses new opportunities for host-targeted therapeutics for malaria and the potential to harness drug polypharmacology to simultaneously target multiple host pathways using a single drug intervention. Highlights Host-targeted therapy is a strategy for eliminating or reducing symptoms of infectious disease, and it holds promise for application to malaria. New insights into the malaria parasite–host interaction may lead to novel interventions to target liver-dormant forms called hypnozoites or to counteract the complications associated with cerebral malaria. Systematic approaches that exploit the polyphamacology of drugs developed for other indications could facilitate repurposing drugs that impact one or more points in the malaria parasite life cycle and also address severe complications of the disease. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
15. <italic>Plasmodium yoelii</italic> S4/CelTOS is important for sporozoite gliding motility and cell traversal.
- Author
-
Steel, Ryan W. J., Pei, Ying, Camargo, Nelly, Kaushansky, Alexis, Dankwa, Dorender A., Martinson, Thomas, Nguyen, Thao, Betz, Will, Cardamone, Hayley, Vigdorovich, Vladimir, Dambrauskas, Nicholas, Carbonetti, Sara, Vaughan, Ashley M., Sather, D. Noah, and Kappe, Stefan H. I.
- Subjects
- *
MEROZOITES , *SPOROZOITES , *PROTOZOAN spores , *PLASMODIUM yoelii , *PHENOTYPIC plasticity - Abstract
Abstract: Gliding motility and cell traversal by the Plasmodium ookinete and sporozoite invasive stages allow penetration of cellular barriers to establish infection of the mosquito vector and mammalian host, respectively. Motility and traversal are not observed in red cell infectious merozoites, and we have previously classified genes that are expressed in sporozoites but not merozoites (S genes) in order to identify proteins involved in these processes. The S4 gene has been described as criticaly involved in Cell Traversal for Ookinetes and Sporozoites (CelTOS), yet knockout parasites (s4/celtos¯) do not generate robust salivary gland sporozoite numbers, precluding a thorough analysis of S4/CelTOS function during host infection. We show here that a failure of oocysts to develop or survive in the midgut contributes to the poor mosquito infection by Plasmodium yoelii (Py) s4/celtos¯ rodent malaria parasites. We rescued this phenotype by expressing S4/CelTOS under the ookinete‐specific circumsporozoite protein and thrombospondin‐related anonymous protein‐related protein (CTRP) promoter (S4/CelTOSCTRP), generating robust numbers of salivary gland sporozoites lacking S4/CelTOS that were suitable for phenotypic analysis. Py S4/CelTOSCTRP sporozoites showed reduced infectivity in BALB/c mice when compared to wild‐type sporozoites, although they appeared more infectious than sporozoites deficient in the related traversal protein PLP1/SPECT2 (Py plp1/spect2¯). Using
in vitro assays, we substantiate the role of S4/CelTOS in sporozoite cell traversal, but also uncover a previously unappreciated role for this protein for sporozoite gliding motility. [ABSTRACT FROM AUTHOR]- Published
- 2018
- Full Text
- View/download PDF
16. A genome-wide CRISPR-Cas9 screen identifies CENPJ as a host regulator of altered microtubule organization during Plasmodium liver infection.
- Author
-
Vijayan, Kamalakannan, Arang, Nadia, Wei, Ling, Morrison, Robert, Geiger, Rechel, Parks, K. Rachael, Lewis, Adam J., Mast, Fred D., Douglass, Alyse N., Kain, Heather S., Aitchison, John D., Johnson, Jarrod S., Aderem, Alan, and Kaushansky, Alexis
- Subjects
- *
PARASITES , *CRISPRS , *MEDICAL screening , *PLASMODIUM , *LIVER , *MICROTUBULES , *CENTROMERE , *MEROZOITES - Abstract
Prior to initiating symptomatic malaria, a single Plasmodium sporozoite infects a hepatocyte and develops into thousands of merozoites, in part by scavenging host resources, likely delivered by vesicles. Here, we demonstrate that host microtubules (MTs) dynamically reorganize around the developing liver stage (LS) parasite to facilitate vesicular transport to the parasite. Using a genome-wide CRISPR-Cas9 screen, we identified host regulators of cytoskeleton organization, vesicle trafficking, and ER/Golgi stress that regulate LS development. Foci of γ-tubulin localized to the parasite periphery; depletion of centromere protein J (CENPJ), a novel regulator identified in the screen, exacerbated this re-localization and increased infection. We demonstrate that the Golgi acts as a non-centrosomal MT organizing center (ncMTOC) by positioning γ-tubulin and stimulating MT nucleation at parasite periphery. Together, these data support a model where the Plasmodium LS recruits host Golgi to form MT-mediated conduits along which host organelles are recruited to PVM and support parasite development. [Display omitted] • A genome-wide CRISPR screen identifies host factors of Plasmodium liver infection • Plasmodium liver stages reorganize the host microtubule (MT) network • Host Golgi acts as non-centrosomal MT organizing complex (MTOC) at parasite periphery • Golgi-mediated MTOC repositioning regulates host vesicular trafficking to the parasite To identify host factors required for Plasmodium liver infection, Vijayan et al. conducted a genome-wide CRISPR knockout screen in hepatocytes. They demonstrate that liver stage parasites reorganize host microtubules by repositioning the microtubule organizing center at the parasite periphery in a Golgi-dependent fashion. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
17. Host-based Prophylaxis Successfully Targets Liver Stage Malaria Parasites.
- Author
-
Douglass, Alyse N, Kain, Heather S, Abdullahi, Marian, Arang, Nadia, Austin, Laura S, Mikolajczak, Sebastian A, Billman, Zachary P, Hume, Jen C C, Murphy, Sean C, Kappe, Stefan H I, and Kaushansky, Alexis
- Subjects
- *
PLASMODIUM , *PROTOZOAN diseases , *BILIARY tract , *MALARIA , *AVIAN malaria - Abstract
Eliminating malaria parasites during the asymptomatic but obligate liver stages (LSs) of infection would stop disease and subsequent transmission. Unfortunately, only a single licensed drug that targets all LSs, Primaquine, is available. Targeting host proteins might significantly expand the repertoire of prophylactic drugs against malaria. Here, we demonstrate that both Bcl-2 inhibitors and P53 agonists dramatically reduce LS burden in a mouse malaria model in vitro and in vivo by altering the activity of key hepatocyte factors on which the parasite relies. Bcl-2 inhibitors act primarily by inducing apoptosis in infected hepatocytes, whereas P53 agonists eliminate parasites in an apoptosis-independent fashion. In combination, Bcl-2 inhibitors and P53 agonists act synergistically to delay, and in some cases completely prevent, the onset of blood stage disease. Both families of drugs are highly effective at doses that do not cause substantial hepatocyte cell death in vitro or liver damage in vivo. P53 agonists and Bcl-2 inhibitors were also effective when administered to humanized mice infected with Plasmodium falciparum. Our data demonstrate that host-based prophylaxis could be developed into an effective intervention strategy that eliminates LS parasites before the onset of clinical disease and thus opens a new avenue to prevent malaria. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
18. Phosphorylated c-Mpl tyrosine 591 regulates thrombopoietin-induced signaling.
- Author
-
Sangkhae, Veena, Saur, Sebastian Jonas, Kaushansky, Alexis, Kaushansky, Kenneth, and Hitchcock, Ian Stuart
- Subjects
- *
PHOSPHORYLATION , *TYROSINE , *THROMBOPOIETIN , *CELLULAR signal transduction , *BLOOD platelets , *CELL proliferation , *CELL differentiation , *CELL receptors - Abstract
Thrombopoietin (TPO) is the primary regulator of platelet production, affecting cell survival, proliferation, and differentiation through binding to and stimulation of the cell surface receptor the cellular myeloproliferative leukemia virus oncogene (c-Mpl). Activating mutations in c-Mpl constitutively stimulate downstream signaling pathways, leading to aberrant hematopoiesis, and contribute to development of myeloproliferative neoplasms. Several studies have mapped the tyrosine residues within the cytoplasmic domain of c-Mpl that mediate these cellular signals; however, secondary signaling pathways are incompletely understood. In this study, we focused on c-Mpl tyrosine 591 (Y591). We found Y591 of wild-type c-Mpl to be phosphorylated in the presence of TPO. Additionally, eliminating Y591 phosphorylation by mutation to Phe resulted in decreased total receptor phosphorylation. Using a Src homology 2/phosphotyrosine-binding (SH2/PTB) domain binding microarray, we identified novel c-Mpl binding partners for phosphorylated Y591, including Src homology region 2 domain-containing phosphatase-1 (SHP-1), spleen tyrosine kinase (SYK) and Bruton's tyrosine kinase (BTK). The functional significance of binding partners was determined through small interfering RNA treatment of Ba/F3-Mpl cells, confirming that the increase in pERK1/2 resulting from removal of Y591 may be mediated by spleen tyrosine kinase. These findings identify a novel negative regulatory pathway that controls TPO-mediated signaling, advancing our understanding of the mechanisms required for successful maintenance of hematopoietic stem cells and megakaryocyte development. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
19. A Next-generation Genetically Attenuated Plasmodium falciparum Parasite Created by Triple Gene Deletion.
- Author
-
Mikolajczak, Sebastian A, Lakshmanan, Viswanathan, Fishbaugher, Matthew, Camargo, Nelly, Harupa, Anke, Kaushansky, Alexis, Douglass, Alyse N, Baldwin, Michael, Healer, Julie, O'Neill, Matthew, Phuong, Thuan, Cowman, Alan, and Kappe, Stefan H I
- Subjects
- *
PLASMODIUM falciparum genetics , *DELETION mutation , *MALARIA prevention , *MOSQUITO vectors , *SPOROZOITES - Abstract
Immunization with live-attenuated Plasmodium sporozoites completely protects against malaria infection. Genetic engineering offers a versatile platform to create live-attenuated sporozoite vaccine candidates. We previously generated a genetically attenuated parasite (GAP) by deleting the P52 and P36 genes in the NF54 wild-type (WT) strain of Plasmodium falciparum (Pf p52−/p36− GAP). Preclinical assessment of p52−/p36− GAP in a humanized mouse model indicated an early and severe liver stage growth defect. However, human exposure to >200 Pf p52−/p36− GAP-infected mosquito bites in a safety trial resulted in peripheral parasitemia in one of six volunteers, revealing that this GAP was incompletely attenuated. We have now created a triple gene deleted GAP by additionally removing the SAP1 gene (Pf p52−/p36−/sap1− GAP) and employed flippase (FLP)/flippase recognition target (FRT) recombination for drug selectable marker cassette removal. This next-generation GAP was indistinguishable from WT parasites in blood stage and mosquito stage development. Using an improved humanized mouse model transplanted with human hepatocytes and human red blood cells, we show that despite a high-dose sporozoite challenge, Pf p52−/p36−/sap1− GAP did not transition to blood stage infection and appeared to be completely attenuated. Thus, clinical testing of Pf p52−/p36−/sap1− GAP assessing safety, immunogenicity, and efficacy against sporozoite challenge is warranted. [ABSTRACT FROM AUTHOR]
- Published
- 2014
- Full Text
- View/download PDF
20. Immunization with genetically attenuated P. falciparum parasites induces long-lived antibodies that efficiently block hepatocyte invasion by sporozoites.
- Author
-
Finney, Olivia C., Keitany, Gladys J., Smithers, Hannah, Kaushansky, Alexis, Kappe, Stefan, and Wang, Ruobing
- Subjects
- *
IMMUNIZATION , *PLASMODIUM falciparum , *IMMUNOGLOBULINS , *LIVER cells , *SPOROZOITES , *MICROBIAL invasiveness , *MALARIA vaccines , *CLINICAL trials - Abstract
Abstract: Whole-parasite malaria vaccines have shown promise in clinical trials. We recently reported the first human trial of a malaria vaccine based on Plasmodium falciparum genetically attenuated parasites (PfGAP). Herein we report for the first time that PfGAP induces prolonged functional humoral responses in humans. Six volunteers were exposed to 5 bites of PfGAP-infected mosquitoes followed by approximately 200 bites. Plasma collected from all volunteers 3 months after the last exposure efficiently inhibits invasion of hepatocytes by P. falciparum sporozoites. The level of inhibition observed is comparable to that attained using plasma collected after 4–5 intravenously administrations of high numbers of irradiated sporozoites, validating the potential of PfGAP malaria vaccines. Our data highlight the role of antibody responses in pre-erythrocytic stages of human malaria, and suggests that to be protective, malaria vaccines might need to elicit long-lasting functional antibodies in addition to cellular responses. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
21. Complete Plasmodium falciparum liver-stage development in liver-chimeric mice.
- Author
-
Vaughan AM, Mikolajczak SA, Wilson EM, Grompe M, Kaushansky A, Camargo N, Bial J, Ploss A, Kappe SH, Vaughan, Ashley M, Mikolajczak, Sebastian A, Wilson, Elizabeth M, Grompe, Markus, Kaushansky, Alexis, Camargo, Nelly, Bial, John, Ploss, Alexander, and Kappe, Stefan H I
- Abstract
Plasmodium falciparum, which causes the most lethal form of human malaria, replicates in the host liver during the initial stage of infection. However, in vivo malaria liver-stage (LS) studies in humans are virtually impossible, and in vitro models of LS development do not reconstitute relevant parasite growth conditions. To overcome these obstacles, we have adopted a robust mouse model for the study of P. falciparum LS in vivo: the immunocompromised and fumarylacetoacetate hydrolase-deficient mouse (Fah-/-, Rag2-/-, Il2rg-/-, termed the FRG mouse) engrafted with human hepatocytes (FRG huHep). FRG huHep mice supported vigorous, quantifiable P. falciparum LS development that culminated in complete maturation of LS at approximately 7 days after infection, providing a relevant model for LS development in humans. The infections allowed observations of previously unknown expression of proteins in LS, including P. falciparum translocon of exported proteins 150 (PTEX150) and exported protein-2 (EXP-2), components of a known parasite protein export machinery. LS schizonts exhibited exoerythrocytic merozoite formation and merosome release. Furthermore, FRG mice backcrossed to the NOD background and repopulated with huHeps and human red blood cells supported reproducible transition from LS infection to blood-stage infection. Thus, these mice constitute reliable models to study human LS directly in vivo and demonstrate utility for studies of LS-to-blood-stage transition of a human malaria parasite. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
22. Complete Plasmodium falciparum liver stage development in liver-chimeric mice.
- Author
-
Vaughan, Ashley M., Mikolajczak, Sebastian A., Wilson, Elizabeth M., Grompe, Markus, Kaushansky, Alexis, Camargo, Nelly, Bial, John, Ploss, Alexander, and Kappe, Stefan H. I.
- Subjects
- *
PLASMODIUM falciparum , *LIVER development , *CHIMERIC proteins , *MALARIA , *ERYTHROCYTES , *LIVER diseases , *LABORATORY mice - Abstract
Plasmodium falciparum, which causes the most lethal form of human malaria, replicates in the host liver during the initial stage of infection. However, in vivo malaria liver-stage (LS) studies in humans are virtually impossible, and in vitro models of LS development do not reconstitute relevant parasite growth conditions. To overcome these obstacles, we have adopted a robust mouse model for the study of P. falciparum LS in vivo: the immunocompromised and fumarylacetoacetate hydrolase-deficient mouse (Fah-/-, Rag2-/-, Il2rg-/-, termed the FRG mouse) engrafted with human hepatocytes (FRG huHep). FRG huHep mice supported vigorous, quantifiable P. falciparum LS development that culminated in complete maturation of LS at approximately 7 days after infection, providing a relevant model for LS development in humans. The infections allowed observations of previously unknown expression of proteins in LS, including P. falciparum translocon of exported proteins 150 (PTEX150) and exported protein-2 (EXP-2), components of a known parasite protein export machinery. LS schizonts exhibited exoerythrocytic merozoite formation and merosome release. Furthermore, FRG mice backcrossed to the NOD background and repopulated with huHeps and human red blood cells supported reproducible transition from LS infection to blood-stage infection. Thus, these mice constitute reliable models to study human LS directly in vivo and demonstrate utility for studies of LS-to-blood-stage transition of a human malaria parasite. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
23. Tyrosine-Phosphorylated Caveolin-1 Blocks Bacterial Uptake by Inducing Vav2-RhoA-Mediated Cytoskeletal Rearrangements.
- Author
-
Boettcher, Jan Peter, Kirchner, Marieluise, Churin, Yuri, Kaushansky, Alexis, Pompaiah, Malvika, Thorn, Hans, Brinkmann, Volker, MacBeath, Gavin, and Meyer, Thomas F.
- Subjects
- *
BACTERIAL adhesion , *CELL communication , *CYTOSKELETAL proteins , *PROTEIN-tyrosine phosphatase , *ESCHERICHIA coli physiology , *BACTERIAL disease prevention - Abstract
Certain bacterial adhesins appear to promote a pathogen's extracellular lifestyle rather than its entry into host cells. However, little is known about the stimuli elicited upon such pathogen host-cell interactions. Here, we report that type IV pili (Tfp)-producing Neisseria gonorrhoeae (P+GC) induces an immediate recruitment of caveolin-1 (Cav1) in the host cell, which subsequently prevents bacterial internalization by triggering cytoskeletal rearrangements via downstream phosphotyrosine signaling. A broad and unbiased analysis of potential interaction partners for tyrosine-phosphorylated Cav1 revealed a direct interaction with the Rho-family guanine nucleotide exchange factor Vav2. Both Vav2 and its substrate, the small GTPase RhoA, were found to play a direct role in the Cav1-mediated prevention of bacterial uptake. Our findings, which have been extended to enteropathogenic Escherichia coli, highlight how Tfp-producing bacteria avoid host cell uptake. Further, our data establish a mechanistic link between Cav1 phosphorylation and pathogen-induced cytoskeleton reorganization and advance our understanding of caveolin function. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
24. Tarp regulates early Chlamydia-induced host cell survival through interactions with the human adaptor protein SHC1.
- Author
-
Mehlitz, Adrian, Banhart, Sebastian, Mäurer, André P., Kaushansky, Alexis, Gordus, Andrew G., Zielecki, Julia, MacBeath, Gavin, and Meyer, Thomas F.
- Subjects
- *
PHOSPHOPROTEINS , *CELL communication , *CELLULAR control mechanisms , *PROTEINS , *CHLAMYDIA , *TYROSINE - Abstract
Many bacterial pathogens translocate effector proteins into host cells to manipulate host cell functions. Here, we used a protein microarray comprising virtually all human SRC homology 2 (SH2) and phosphotyrosine binding domains to comprehensively and quantitatively assess interactions between host cell proteins and the early phase Chlamydia trachomatis effector protein translocated actin-recruiting phosphoprotein (Tarp), which is rapidly tyrosine phosphorylated upon host cell entry. We discovered numerous novel interactions between human SH2 domains and phosphopeptides derived from Tarp. The adaptor protein SHC1 was among Tarp's strongest interaction partners. Transcriptome analysis of SHC1-dependent gene regulation during infection indicated that SHC1 regulates apoptosis- and growth-related genes. SHC1 knockdown sensitized infected host cells to tumor necrosis factor-induced apoptosis. Collectively, our findings reveal a critical role for SHC1 in early C. trachomatis-induced cell survival and suggest that Tarp functions as a multivalent phosphorylation-dependent signaling hub that is important during the early phase of chlamydial infection. [ABSTRACT FROM AUTHOR]
- Published
- 2010
- Full Text
- View/download PDF
25. Exploiting polypharmacology to dissect host kinases and kinase inhibitors that modulate endothelial barrier integrity.
- Author
-
Dankwa, Selasi, Dols, Mary-Margaret, Wei, Ling, Glennon, Elizabeth K.K., Kain, Heather S., Kaushansky, Alexis, and Smith, Joseph D.
- Subjects
- *
KINASE inhibitors , *DRUG repositioning , *MACHINE learning , *PHENOTYPES , *DASATINIB - Abstract
Kinase inhibitors are promising drugs to stabilize the endothelial barrier following inflammatory damage. However, our limited knowledge of how kinase signaling activates barrier-restorative pathways and the complexity of multi-target drugs have hindered drug discovery and repurposing efforts. Here, we apply a kinase regression approach that exploits drug polypharmacology to investigate endothelial barrier regulation. A screen of 28 kinase inhibitors identified multiple inhibitors that promote endothelial barrier integrity and revealed divergent barrier phenotypes for BCR-ABL drugs. Target deconvolution predicted 50 barrier-regulating kinases from diverse kinase families. Using gene knockdowns, we identified kinases with a role in endothelial barrier regulation and dissected different mechanisms of action of barrier-protective kinase inhibitors. These results demonstrate the importance of polypharmacology in the endothelial barrier phenotype of kinase inhibitors and provide promising new leads for barrier-strengthening therapies. [Display omitted] • BCR-ABL drugs bosutinib, dasatinib, and imatinib have distinct barrier effects • Polypharmacology influences the endothelial barrier activity of kinase inhibitors • Kinase inhibitors can amplify or inhibit thrombin-mediated barrier disruption • Several kinases in diverse pathways modulate endothelial barrier properties Dankwa et al. combine kinase inhibitor screens with a machine learning model to demonstrate that kinase inhibitors that target multiple kinases can strengthen and protect the endothelial barrier from proinflammatory, barrier-disruptive agents in vitro. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
26. A Molecular Signature in Blood Reveals a Role for p53 in Regulating Malaria-Induced Inflammation.
- Author
-
Tran, Tuan M., Guha, Rajan, Portugal, Silvia, Skinner, Jeff, Ongoiba, Aissata, Bhardwaj, Jyoti, Jones, Marcus, Moebius, Jacqueline, Venepally, Pratap, Doumbo, Safiatou, DeRiso, Elizabeth A., Li, Shanping, Vijayan, Kamalakannan, Anzick, Sarah L., Hart, Geoffrey T., O'Connell, Elise M., Doumbo, Ogobara K., Kaushansky, Alexis, Alter, Galit, and Felgner, Phillip L.
- Subjects
- *
MALARIA , *FC receptors , *IMMUNOGLOBULIN G , *P53 protein , *PARASITEMIA , *FEVER , *T helper cells , *PROTEIN expression - Abstract
Immunity that controls parasitemia and inflammation during Plasmodium falciparum (Pf) malaria can be acquired with repeated infections. A limited understanding of this complex immune response impedes the development of vaccines and adjunctive therapies. We conducted a prospective systems biology study of children who differed in their ability to control parasitemia and fever following Pf infection. By integrating whole-blood transcriptomics, flow-cytometric analysis, and plasma cytokine and antibody profiles, we demonstrate that a pre-infection signature of B cell enrichment, upregulation of T helper type 1 (Th1) and Th2 cell-associated pathways, including interferon responses, and p53 activation associated with control of malarial fever and coordinated with Pf -specific immunoglobulin G (IgG) and Fc receptor activation to control parasitemia. Our hypothesis-generating approach identified host molecules that may contribute to differential clinical outcomes during Pf infection. As a proof of concept, we have shown that enhanced p53 expression in monocytes attenuated Plasmodium -induced inflammation and predicted protection from fever. • A 3-year study identifies children who control malaria fever and parasitemia • Systems analysis before and during infection in immune versus susceptible children • Cellular and humoral immune profiles associate with control of fever and parasitemia • p53 in monocytes attenuates malaria inflammation and predicts protection from fever The mechanisms that protect from febrile malaria remain unclear. Tran et al. applied a systems-based approach to a longitudinal pediatric study to identify immune signatures that associate with control of malaria fever and parasitemia, revealing that p53 upregulation in monocytes attenuates malaria-induced inflammation and predicts protection from fever. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
27. Linear combinations of docking affinities explain quantitative differences in RTK signaling.
- Author
-
Gordus, Andrew, Krall, Jordan A, Beyer, Elsa M, Kaushansky, Alexis, Wolf‐Yadlin, Alejandro, Sevecka, Mark, Chang, Bryan H, Rush, John, and MacBeath, Gavin
- Subjects
- *
PROTEIN-tyrosine kinases , *PROTEINS , *PHOSPHORYLATION , *MATHEMATICAL models , *CYTOLOGY - Abstract
Receptor tyrosine kinases (RTKs) process extracellular cues by activating a broad array of signaling proteins. Paradoxically, they often use the same proteins to elicit diverse and even opposing phenotypic responses. Binary, ‘on–off’ wiring diagrams are therefore inadequate to explain their differences. Here, we show that when six diverse RTKs are placed in the same cellular background, they activate many of the same proteins, but to different quantitative degrees. Additionally, we find that the relative phosphorylation levels of upstream signaling proteins can be accurately predicted using linear models that rely on combinations of receptor-docking affinities and that the docking sites for phosphoinositide 3-kinase (PI3K) and Shc1 provide much of the predictive information. In contrast, we find that the phosphorylation levels of downstream proteins cannot be predicted using linear models. Taken together, these results show that information processing by RTKs can be segmented into discrete upstream and downstream steps, suggesting that the challenging task of constructing mathematical models of RTK signaling can be parsed into separate and more manageable layers. [ABSTRACT FROM AUTHOR]
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.