Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(1): 101300, 2024 01 16.
Article in English | MEDLINE | ID: mdl-38118442

ABSTRACT

Personalized treatment of complex diseases has been mostly predicated on biomarker identification of one drug-disease combination at a time. Here, we use a computational approach termed Disruption Networks to generate a data type, contextualized by cell-centered individual-level networks, that captures biology otherwise overlooked when performing standard statistics. This data type extends beyond the "feature level space", to the "relations space", by quantifying individual-level breaking or rewiring of cross-feature relations. Applying Disruption Networks to dissect high-dimensional blood data, we discover and validate that the RAC1-PAK1 axis is predictive of anti-TNF response in inflammatory bowel disease. Intermediate monocytes, which correlate with the inflammatory state, play a key role in the RAC1-PAK1 responses, supporting their modulation as a therapeutic target. This axis also predicts response in rheumatoid arthritis, validated in three public cohorts. Our findings support blood-based drug response diagnostics across immune-mediated diseases, implicating common mechanisms of non-response.


Subject(s)
Arthritis, Rheumatoid , Inflammatory Bowel Diseases , Humans , Infliximab/therapeutic use , Tumor Necrosis Factor Inhibitors/therapeutic use , Tumor Necrosis Factor-alpha , Arthritis, Rheumatoid/drug therapy , Inflammatory Bowel Diseases/drug therapy
2.
Nat Commun ; 14(1): 6840, 2023 10 27.
Article in English | MEDLINE | ID: mdl-37891175

ABSTRACT

Diseases change over time, both phenotypically and in their underlying molecular processes. Though understanding disease progression dynamics is critical for diagnostics and treatment, capturing these dynamics is difficult due to their complexity and the high heterogeneity in disease development between individuals. We present TimeAx, an algorithm which builds a comparative framework for capturing disease dynamics using high-dimensional, short time-series data. We demonstrate the utility of TimeAx by studying disease progression dynamics for multiple diseases and data types. Notably, for urothelial bladder cancer tumorigenesis, we identify a stromal pro-invasion point on the disease progression axis, characterized by massive immune cell infiltration to the tumor microenvironment and increased mortality. Moreover, the continuous TimeAx model differentiates between early and late tumors within the same tumor subtype, uncovering molecular transitions and potential targetable pathways. Overall, we present a powerful approach for studying disease progression dynamics-providing improved molecular interpretability and clinical benefits for patient stratification and outcome prediction.


Subject(s)
Carcinoma, Transitional Cell , Urinary Bladder Neoplasms , Humans , Urinary Bladder Neoplasms/diagnosis , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/pathology , Carcinoma, Transitional Cell/pathology , Disease Progression , Tumor Microenvironment
3.
Autoimmun Rev ; 22(6): 103314, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36918090

ABSTRACT

Systemic sclerosis (SSc) is a rare and chronic autoimmune disease characterized by a pathogenic triad of immune dysregulation, vasculopathy, and progressive fibrosis. Clinical tools commonly used to assess patients, including the modified Rodnan skin score, difference between limited or diffuse forms of skin involvement, presence of lung, heart or kidney involvement, or of various autoantibodies, are important prognostic factors, but still fail to reflect the large heterogeneity of the disease. SSc treatment options are diverse, ranging from conventional drugs to autologous hematopoietic stem cell transplantation, and predicting response is challenging. Genome-wide technologies, such as high throughput microarray analyses and RNA sequencing, allow accurate, unbiased, and broad assessment of alterations in expression levels of multiple genes. In recent years, many studies have shown robust changes in the gene expression profiles of SSc patients compared to healthy controls, mainly in skin tissues and peripheral blood cells. The objective analysis of molecular patterns in SSc is a powerful tool that can further classify SSc patients with similar clinical phenotypes and help predict response to therapy. In this review, we describe the journey from the first discovery of differentially expressed genes to the identification of enriched pathways and intrinsic subsets identified in SSc, using machine learning algorithms. Finally, we discuss the use of these new tools to predict the efficacy of various treatments, including stem cell transplantation. We suggest that the use of RNA gene expression-based classifications according to molecular subsets may bring us one step closer to precision medicine in Systemic Sclerosis.


Subject(s)
Autoimmune Diseases , Scleroderma, Systemic , Humans , Precision Medicine , Scleroderma, Systemic/therapy , Scleroderma, Systemic/drug therapy , Fibrosis , Autoantibodies/therapeutic use
4.
Cell Rep Med ; 3(6): 100652, 2022 06 21.
Article in English | MEDLINE | ID: mdl-35675822

ABSTRACT

Disease recovery dynamics are often difficult to assess, as patients display heterogeneous recovery courses. To model recovery dynamics, exemplified by severe COVID-19, we apply a computational scheme on longitudinally sampled blood transcriptomes, generating recovery states, which we then link to cellular and molecular mechanisms, presenting a framework for studying the kinetics of recovery compared with non-recovery over time and long-term effects of the disease. Specifically, a decrease in mature neutrophils is the strongest cellular effect during recovery, with direct implications on disease outcome. Furthermore, we present strong indications for global regulatory changes in gene programs, decoupled from cell compositional changes, including an early rise in T cell activation and differentiation, resulting in immune rebalancing between interferon and NF-κB activity and restoration of cell homeostasis. Overall, we present a clinically relevant computational framework for modeling disease recovery, paving the way for future studies of the recovery dynamics in other diseases and tissues.


Subject(s)
COVID-19 , NF-kappa B , Cell Differentiation , Humans , Interferons/metabolism , NF-kappa B/genetics , Neutrophils/metabolism , Signal Transduction
5.
Med ; 3(7): 468-480.e5, 2022 07 08.
Article in English | MEDLINE | ID: mdl-35716665

ABSTRACT

BACKGROUND: Much remains unknown regarding the response of the immune system to severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) vaccination. METHODS: We employed circulating cell-free DNA (cfDNA) to assess the turnover of specific immune cell types following administration of the Pfizer/BioNTech vaccine. FINDINGS: The levels of B cell cfDNA after the primary dose correlated with development of neutralizing antibodies and memory B cells after the booster, revealing a link between early B cell turnover-potentially reflecting affinity maturation-and later development of effective humoral response. We also observed co-elevation of B cell, T cell, and monocyte cfDNA after the booster, underscoring the involvement of innate immune cell turnover in the development of humoral and cellular adaptive immunity. Actual cell counts remained largely stable following vaccination, other than a previously demonstrated temporary reduction in neutrophil and lymphocyte counts. CONCLUSIONS: Immune cfDNA dynamics reveal the crucial role of the primary SARS-CoV-2 vaccine in shaping responses of the immune system following the booster vaccine. FUNDING: This work was supported by a generous gift from Shlomo Kramer. Supported by grants from Human Islet Research Network (HIRN UC4DK116274 and UC4DK104216 to R.S. and Y.D.), Ernest and Bonnie Beutler Research Program of Excellence in Genomic Medicine, The Alex U Soyka Pancreatic Cancer Fund, The Israel Science Foundation, the Waldholtz/Pakula family, the Robert M. and Marilyn Sternberg Family Charitable Foundation, the Helmsley Charitable Trust, Grail, and the DON Foundation (to Y.D.). Y.D. holds the Walter and Greta Stiel Chair and Research Grant in Heart Studies. I.F.-F. received a fellowship from the Glassman Hebrew University Diabetes Center.


Subject(s)
BNT162 Vaccine , COVID-19 , Cell-Free Nucleic Acids , SARS-CoV-2 , Adult , Aged , Antibodies, Neutralizing/genetics , Antibodies, Neutralizing/immunology , Antibodies, Viral/genetics , Antibodies, Viral/immunology , BNT162 Vaccine/administration & dosage , COVID-19/immunology , COVID-19/prevention & control , Cell-Free Nucleic Acids/genetics , Cell-Free Nucleic Acids/immunology , Female , Humans , Immunization, Secondary , Male , Memory B Cells/immunology , Memory B Cells/metabolism , Middle Aged , SARS-CoV-2/immunology , Young Adult
6.
Methods Mol Biol ; 2184: 77-90, 2020.
Article in English | MEDLINE | ID: mdl-32808219

ABSTRACT

Macrophages play an essential role in diverse biological processes, from the immune response to inflammatory and neurodegenerative disorders, to various cancers. A macrophage subpopulation, known as tumor-associated macrophages (TAMs), has been shown to promote tumorigenesis, metastasis, and immune escape of cancer cells. Some of the pro-tumorigenic effects of TAMs are mediated via the secretion of nano-vesicles (exosomes) from macrophages to neighboring cells. In this chapter, we describe peritoneal macrophage isolation methods, polarization of TAMs, and purification and characterization of macrophage-derived exosomes.


Subject(s)
Exosomes/physiology , Macrophages/physiology , Animals , Carcinogenesis/pathology , Cell Line , Mice , Mice, Inbred C57BL
7.
ACS Nano ; 13(10): 11008-11021, 2019 10 22.
Article in English | MEDLINE | ID: mdl-31503443

ABSTRACT

Overexpressed extracellular matrix (ECM) in pancreatic ductal adenocarcinoma (PDAC) limits drug penetration into the tumor and is associated with poor prognosis. Here, we demonstrate that a pretreatment based on a proteolytic-enzyme nanoparticle system disassembles the dense PDAC collagen stroma and increases drug penetration into the pancreatic tumor. More specifically, the collagozome, a 100 nm liposome encapsulating collagenase, was rationally designed to protect the collagenase from premature deactivation and prolonged its release rate at the target site. Collagen is the main component of the PDAC stroma, reaching 12.8 ± 2.3% vol in diseased mice pancreases, compared to 1.4 ± 0.4% in healthy mice. Upon intravenous injection of the collagozome, ∼1% of the injected dose reached the pancreas over 8 h, reducing the level of fibrotic tissue to 5.6 ± 0.8%. The collagozome pretreatment allowed increased drug penetration into the pancreas and improved PDAC treatment. PDAC tumors, pretreated with the collagozome followed by paclitaxel micelles, were 87% smaller than tumors pretreated with empty liposomes followed by paclitaxel micelles. Interestingly, degrading the ECM did not increase the number of circulating tumor cells or metastasis. This strategy holds promise for degrading the extracellular stroma in other diseases as well, such as liver fibrosis, enhancing tissue permeability before drug administration.


Subject(s)
Adenocarcinoma/drug therapy , Carcinoma, Pancreatic Ductal/drug therapy , Collagenases/pharmacology , Nanoparticles/chemistry , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Membrane Permeability/drug effects , Collagen/chemistry , Collagen/genetics , Collagenases/chemistry , Disease Models, Animal , Extracellular Matrix/drug effects , Extracellular Matrix/genetics , Fibrosis/drug therapy , Fibrosis/pathology , Fibrosis/prevention & control , Humans , Liposomes/chemistry , Liposomes/pharmacology , Mice , Nanoparticles/therapeutic use , Paclitaxel/chemistry , Paclitaxel/pharmacology , Pancreas/drug effects , Pancreas/pathology , Tumor Microenvironment/drug effects
8.
Drug Resist Updat ; 45: 1-12, 2019 07.
Article in English | MEDLINE | ID: mdl-31369918

ABSTRACT

Exosomes are a class of extracellular vesicles ranging in size from 40 to 100 nm, which are secreted by both cancer cells and multiple stromal cells in the tumor microenvironment. Following their secretion, exosomes partake in endocrine, paracrine and autocrine signaling. Internalization of exosomes by tumor cells influences several cellular pathways which alter cancer cell physiology. Tumor-derived exosomes secreted by cancer or stromal cells can also confer anticancer drug-resistant traits upon cancer cells. These exosomes promote chemoresistance by transferring their cargo which includes nucleic acids, proteins, and metabolites to cancer cells or act as a decoy for immunotherapeutic targets. Depletion of exosomes can reverse some of the detrimental effects on tumor metabolism and restore drug sensitivity to chemotherapeutic treatment. Herein we discuss various approaches that have been developed to deplete exosomes for therapeutic purposes. The natural composition, low immunogenicity and cytotoxicity of exosomes, along with their ability to specifically target tumor cells, render them an appealing platform for drug delivery. The ability of exosomes to mediate autocrine and paracrine signaling in target cells, along with their natural structure and low immunogenicity render them an attractive vehicle for the delivery of anticancer drugs to tumors.


Subject(s)
Carcinogenesis/pathology , Drug Resistance, Neoplasm/physiology , Exosomes/pathology , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Carcinogenesis/drug effects , Drug Delivery Systems/methods , Exosomes/drug effects , Humans , Neoplasms/drug therapy , Signal Transduction/drug effects , Signal Transduction/physiology , Tumor Microenvironment/drug effects , Tumor Microenvironment/physiology
9.
Nat Commun ; 10(1): 809, 2019 02 18.
Article in English | MEDLINE | ID: mdl-30778058

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human cancers. It thrives in a nutrient-poor environment; however, the mechanisms by which PDAC cells undergo metabolic reprogramming to adapt to metabolic stress are still poorly understood. Here, we show that microRNA-135 is significantly increased in PDAC patient samples compared to adjacent normal tissue. Mechanistically, miR-135 accumulates specifically in response to glutamine deprivation and requires ROS-dependent activation of mutant p53, which directly promotes miR-135 expression. Functionally, we found miR-135 targets phosphofructokinase-1 (PFK1) and inhibits aerobic glycolysis, thereby promoting the utilization of glucose to support the tricarboxylic acid (TCA) cycle. Consistently, miR-135 silencing sensitizes PDAC cells to glutamine deprivation and represses tumor growth in vivo. Together, these results identify a mechanism used by PDAC cells to survive the nutrient-poor tumor microenvironment, and also provide insight regarding the role of mutant p53 and miRNA in pancreatic cancer cell adaptation to metabolic stresses.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Glycolysis/genetics , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Phosphofructokinase-1, Type C/genetics , Animals , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Survival/genetics , Gene Expression Regulation, Neoplastic , Glutamine/genetics , Glutamine/metabolism , Humans , Male , Mice, Nude , MicroRNAs/metabolism , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phosphofructokinase-1, Type C/metabolism , Stress, Physiological/genetics , Xenograft Model Antitumor Assays
10.
Int J Cancer ; 144(12): 3014-3022, 2019 06 15.
Article in English | MEDLINE | ID: mdl-30515799

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) remains a deadly disease, affecting about 40,000 individuals in the United States annually. We aimed to characterize the role of RET as a co-driver of pancreas tumorigenesis. To assess the role of RET as a co-driver of PDA, we generated a novel triple mutant transgenic mouse based on the cre-activated p53R172H gene and a constitutively active RET M919T mutant (PRC). Survival analysis was performed using Kaplan-Meier analysis. Study of human PDA specimens and Pdx-1-Cre/KrasG12D /p53R172H (KPC) mice revealed that RET is upregulated during pancreas tumorigenesis, from inception through precursor lesions, to invasive cancer. We demonstrated that activation of RET is capable of inducing invasive pancreatic carcinomas in the background of the P53 inactivation mutation. Compared to KPC mice, PRC animals had distinct phenotypes, including longer latency to tumor progression, longer survival, and the presence of multiple macrometastases. Enhanced activation of the MAPK pathway was observed as early as the PanIN 2 stage. Sequencing of the exonic regions of KRAS in PRC-derived PDA cells revealed no evidence of KRAS mutations. RET can be an essential co-driver of pancreatic tumorigenesis in conjugation with KRAS activity. These data suggest that RET may be a potential target in the treatment of PDA.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins c-ret/metabolism , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/pathology , Enzyme Activation , Female , Humans , MAP Kinase Signaling System , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins c-ret/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Up-Regulation
11.
Sci Rep ; 8(1): 17871, 2018 12 14.
Article in English | MEDLINE | ID: mdl-30552383

ABSTRACT

Major complications and mortality from Plasmodium falciparum malaria are associated with cytoadhesion of parasite-infected erythrocytes (IE). The main parasite ligands for cytoadhesion are members of the P. falciparum erythrocyte membrane protein 1 (PfEMP1) family. Interactions of different host receptor-ligand pairs may lead to various pathological outcomes, like placental or cerebral malaria. It has been shown previously that IE can bind integrin αVß3. Using bead-immobilized PfEMP1 constructs, we have identified that the PFL2665c DBLδ1_D4 domain binds to αVß3 and αVß6. A parasite line expressing PFL2665c binds to surface-immobilized αVß3 and αVß6; both are RGD motif-binding integrins. Interactions can be inhibited by cyloRGDFV peptide, an antagonist of RGD-binding integrins. This is a first, to the best of our knowledge, implication of a specific PfEMP1 domain for binding to integrins. These host receptors have important physiological functions in endothelial and immune cells; therefore, these results will contribute to future studies and a better understanding, at the molecular level, of the physiological outcome of interactions between IE and integrin receptors on the surface of host cells.


Subject(s)
Antigens, Neoplasm/metabolism , Cell Adhesion , Erythrocytes/physiology , Erythrocytes/parasitology , Integrin alphaVbeta3/metabolism , Integrins/metabolism , Plasmodium falciparum/growth & development , Protozoan Proteins/metabolism , Humans , Protein Binding
12.
Cancer Res ; 78(18): 5287-5299, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30042153

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is known for its resistance to gemcitabine, which acts to inhibit cell growth by termination of DNA replication. Tumor-associated macrophages (TAM) were recently shown to contribute to gemcitabine resistance; however, the exact mechanism of this process is still unclear. Using a genetic mouse model of PDAC and electron microscopy analysis, we show that TAM communicate with the tumor microenvironment via secretion of approximately 90 nm vesicles, which are selectively internalized by cancer cells. Transfection of artificial dsDNA (barcode fragment) to murine peritoneal macrophages and injection to mice bearing PDAC tumors revealed a 4-log higher concentration of the barcode fragment in primary tumors and in liver metastasis than in normal tissue. These macrophage-derived exosomes (MDE) significantly decreased the sensitivity of PDAC cells to gemcitabine, in vitro and in vivo This effect was mediated by the transfer of miR-365 in MDE. miR-365 impaired activation of gemcitabine by upregulation of the triphospho-nucleotide pool in cancer cells and the induction of the enzyme cytidine deaminase; the latter inactivates gemcitabine. Adoptive transfer of miR-365 in TAM induced gemcitabine resistance in PDAC-bearing mice, whereas immune transfer of the miR-365 antagonist recovered the sensitivity to gemcitabine. Mice deficient of Rab27 a/b genes, which lack exosomal secretion, responded significantly better to gemcitabine than did wildtype. These results identify MDE as key regulators of gemcitabine resistance in PDAC and demonstrate that blocking miR-365 can potentiate gemcitabine response.Significance: Harnessing macrophage-derived exosomes as conveyers of antagomiRs augments the effect of chemotherapy against cancer, opening new therapeutic options against malignancies where resistance to nucleotide analogs remains an obstacle to overcome. Cancer Res; 78(18); 5287-99. ©2018 AACR.


Subject(s)
Adenocarcinoma/therapy , Carcinoma, Pancreatic Ductal/drug therapy , Drug Resistance, Neoplasm , Exosomes/metabolism , MicroRNAs/metabolism , Pancreatic Neoplasms/therapy , Adenocarcinoma/metabolism , Animals , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Gene Transfer Techniques , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pancreatic Neoplasms/metabolism , Tumor Microenvironment , Up-Regulation , rab27 GTP-Binding Proteins/genetics , Gemcitabine
13.
PLoS One ; 11(7): e0159449, 2016.
Article in English | MEDLINE | ID: mdl-27434123

ABSTRACT

Malaria vaccine development has been hampered by the limited availability of antigens identified through conventional discovery approaches, and improvements are needed to enhance the efficacy of the leading vaccine candidate RTS,S that targets the circumsporozoite protein (CSP) of the infective sporozoite. Here we report a transcriptome-based approach to identify novel pre-erythrocytic vaccine antigens that could potentially be used in combination with CSP. We hypothesized that stage-specific upregulated genes would enrich for protective vaccine targets, and used tiling microarray to identify P. falciparum genes transcribed at higher levels during liver stage versus sporozoite or blood stages of development. We prepared DNA vaccines for 21 genes using the predicted orthologues in P. yoelii and P. berghei and tested their efficacy using different delivery methods against pre-erythrocytic malaria in rodent models. In our primary screen using P. yoelii in BALB/c mice, we found that 16 antigens significantly reduced liver stage parasite burden. In our confirmatory screen using P. berghei in C57Bl/6 mice, we confirmed 6 antigens that were protective in both models. Two antigens, when combined with CSP, provided significantly greater protection than CSP alone in both models. Based on the observations reported here, transcriptional patterns of Plasmodium genes can be useful in identifying novel pre-erythrocytic antigens that induce protective immunity alone or in combination with CSP.


Subject(s)
Antibodies, Protozoan/immunology , Malaria Vaccines/immunology , Malaria, Falciparum/drug therapy , Protozoan Proteins/immunology , Animals , Antibodies, Protozoan/therapeutic use , Antigens, Protozoan/immunology , Female , Humans , Malaria Vaccines/genetics , Malaria Vaccines/therapeutic use , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Mice , Mice, Inbred C57BL , Plasmodium falciparum/immunology , Plasmodium falciparum/pathogenicity , Plasmodium yoelii/immunology , Vaccines, DNA/genetics , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use
14.
J Infect Dis ; 214(1): 23-31, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27117511

ABSTRACT

BACKGROUND: Herpes simplex virus type 2 (HSV-2) reactivation is accompanied by a sustained influx of CD4(+) and CD8(+) T cells that persist in genital tissue for extended periods. While CD4(+) T cells have long been recognized as being present in herpetic ulcerations, their role in subclinical reactivation and persistence is less well known, especially the role of CD4(+) regulatory T cells (Tregs). METHODS: We characterized the Treg (CD4(+)Foxp3(+)) population during human HSV-2 reactivation in situ in sequential genital skin biopsy specimens obtained from HSV-2-seropositive subjects at the time of lesion onset up to 8 weeks after healing. RESULTS: High numbers of Tregs infiltrated to the site of viral reactivation and persisted in proximity to conventional CD4(+) T cells (Tconvs) and CD8(+) T cells. Treg density peaked during the lesion stage of the reactivation. The number of Tregs from all time points (lesion, healed, 2 weeks after healing, 4 weeks after healing, and 8 weeks after healing) was significantly higher than in control biopsy specimens from unaffected skin. There was a direct correlation between HSV-2 titer and Treg density. CONCLUSIONS: The association of a high Treg to Tconv ratio with high viral shedding suggests that the balance between regulatory and effector T cells influences human HSV-2 disease.


Subject(s)
Genitalia/innervation , Genitalia/virology , Herpes Genitalis/physiopathology , Herpesvirus 2, Human/physiology , T-Lymphocytes, Regulatory/virology , Virus Activation/physiology , Virus Shedding/physiology , Female , Humans , Male , Washington
15.
Nucleic Acids Res ; 43(15): 7349-59, 2015 Sep 03.
Article in English | MEDLINE | ID: mdl-26130711

ABSTRACT

Meiotic programmed DNA double-strand break (DSB) repair is essential for crossing-over and viable gamete formation and requires removal of Spo11-oligonucleotide complexes from 5' ends (clipping) and their resection to generate invasive 3'-end single-stranded DNA (resection). Ctp1 (Com1, Sae2, CtIP homolog) acting with the Mre11-Rad50-Nbs1 (MRN) complex is required in both steps. We isolated multiple S. pombe ctp1 mutants deficient in clipping but proficient in resection during meiosis. Remarkably, all of the mutations clustered in or near the conserved CxxC or RHR motif in the C-terminal portion. The mutants tested, like ctp1Δ, were clipping-deficient by both genetic and physical assays-. But, unlike ctp1Δ, these mutants were recombination-proficient for Rec12 (Spo11 homolog)-independent break-repair and resection-proficient by physical assay. We conclude that the intracellular Ctp1 C-terminal portion is essential for clipping, while the N-terminal portion is sufficient for DSB end-resection. This conclusion agrees with purified human CtIP resection and endonuclease activities being independent. Our mutants provide intracellular evidence for separable functions of Ctp1. Some mutations truncate Ctp1 in the same region as one of the CtIP mutations linked to the Seckel and Jawad severe developmental syndromes, suggesting that these syndromes are caused by a lack of clipping at DSB ends that require repair.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Endodeoxyribonucleases/metabolism , Meiosis/genetics , Schizosaccharomyces pombe Proteins/metabolism , Camptothecin/toxicity , DNA-Binding Proteins/genetics , Endodeoxyribonucleases/genetics , Methyl Methanesulfonate/toxicity , Mutation , Recombination, Genetic , Schizosaccharomyces pombe Proteins/genetics , Spores, Fungal/genetics
16.
J Infect Dis ; 211(7): 1134-43, 2015 Apr 01.
Article in English | MEDLINE | ID: mdl-25355939

ABSTRACT

BACKGROUND: We developed a 2-step approach to screen molecules that prevent and/or reverse Plasmodium falciparum-infected erythrocyte (IE) binding to host receptors. IE adhesion and sequestration in vasculature causes severe malaria, and therefore antiadhesion therapy might be useful as adjunctive treatment. IE adhesion is mediated by the polymorphic family (approximately 60 members) of P. falciparum EMP1 (PfEMP1) multidomain proteins. METHODS: We constructed sets of PfEMP1 domains that bind ICAM-1, CSA, or CD36, receptors that commonly support IE binding. Combinations of domain-coated beads were assayed by Bio-Plex technology as a high-throughput molecular platform to screen antiadhesion molecules (antibodies and small molecules). Molecules identified as so-called hits in the screen (first step) then could be assayed individually for inhibition of binding of live IE to receptors (second step). RESULTS: In proof-of-principle studies, the antiadhesion activity of several antibodies was concordant in Bio-Plex and live IE assays. Using this 2-step approach, we identified several molecules in a small molecule library of 10 000 compounds that could inhibit and reverse binding of IEs to ICAM-1 and CSA receptors. CONCLUSION: This 2-step screening approach should be efficient for identification of antiadhesion drug candidates for falciparum malaria.


Subject(s)
Cell Adhesion Molecules/metabolism , Erythrocytes/parasitology , Malaria, Falciparum/prevention & control , Plasmodium falciparum/drug effects , Protozoan Proteins/metabolism , CD36 Antigens/metabolism , Cell Adhesion/drug effects , Cell Adhesion/immunology , Cell Line , Erythrocytes/immunology , Erythrocytes/metabolism , High-Throughput Screening Assays , Humans , Intercellular Adhesion Molecule-1/metabolism , Malaria, Falciparum/immunology , Malaria, Falciparum/parasitology , Plasmodium falciparum/immunology , Small Molecule Libraries
17.
Genome Res ; 24(10): 1650-64, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25024163

ABSTRACT

Fission yeast Rec12 (Spo11 homolog) initiates meiotic recombination by forming developmentally programmed DNA double-strand breaks (DSBs). DSB distributions influence patterns of heredity and genome evolution, but the basis of the highly nonrandom choice of Rec12 cleavage sites is poorly understood, largely because available maps are of relatively low resolution and sensitivity. Here, we determined DSBs genome-wide at near-nucleotide resolution by sequencing the oligonucleotides attached to Rec12 following DNA cleavage. The single oligonucleotide size class allowed us to deeply sample all break events. We find strong evidence across the genome for differential DSB repair accounting for crossover invariance (constant cM/kb in spite of DSB hotspots). Surprisingly, about half of all crossovers occur in regions where DSBs occur at low frequency and are widely dispersed in location from cell to cell. These previously undetected, low-level DSBs thus play an outsized and crucial role in meiosis. We further find that the influence of underlying nucleotide sequence and chromosomal architecture differs in multiple ways from that in budding yeast. DSBs are not strongly restricted to nucleosome-depleted regions, as they are in budding yeast, but are nevertheless spatially influenced by chromatin structure. Our analyses demonstrate that evolutionarily fluid factors contribute to crossover initiation and regulation.


Subject(s)
Crossing Over, Genetic , DNA Breaks, Double-Stranded , Meiosis , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/genetics , Chromatin/metabolism , Evolution, Molecular , Genome, Fungal , Models, Genetic , Schizosaccharomyces/cytology , Sequence Analysis, DNA
18.
Expert Rev Vaccines ; 11(10): 1261-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23176657

ABSTRACT

Pre-erythrocytic malaria vaccines target Plasmodium during its sporozoite and liver stages, and can prevent progression to blood-stage disease, which causes a million deaths each year. Whole organism sporozoite vaccines induce sterile immunity in animals and humans and guide subunit vaccine development. A recombinant protein-in-adjuvant pre-erythrocytic vaccine called RTS,S reduces clinical malaria without preventing infection in field studies and additional antigens may be required to achieve sterile immunity. Although few vaccine antigens have progressed to human testing, new insights into parasite biology, expression profiles and immunobiology have offered new targets for intervention. Future advances require human trials of additional antigens, as well as platforms to induce the durable antibody and cellular responses including CD8(+) T cells that contribute to sterile protection.


Subject(s)
Antigens, Protozoan/immunology , Malaria Vaccines/immunology , Plasmodium/immunology , Animals , Antigens, Protozoan/genetics , Humans , Malaria Vaccines/genetics , Plasmodium/genetics , Plasmodium/pathogenicity , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology
19.
J Biol Chem ; 286(47): 40566-74, 2011 Nov 25.
Article in English | MEDLINE | ID: mdl-21984849

ABSTRACT

Kinetoplast DNA (kDNA), the mitochondrial genome of trypanosomatids, consists of several thousand topologically interlocked DNA circles. Mitochondrial histone H1-like proteins were implicated in the condensation of kDNA into a nucleoid structure in the mitochondrial matrix. However, the mechanism that remodels kDNA, promoting its accessibility to the replication machinery, has not yet been described. Analyses, using yeast two hybrid system, co-immunoprecipitation, and protein-protein cross-linking, revealed specific protein-protein interactions between the kDNA replication initiator protein universal minicircle sequence-binding protein (UMSBP) and two mitochondrial histone H1-like proteins. Fluorescence and electron microscopy, as well as biochemical analyses, demonstrated that these protein-protein interactions result in the decondensation of kDNA. UMSBP-mediated decondensation rendered the kDNA network accessible to topological decatenation by topoisomerase II, yielding free kDNA minicircle monomers. Hence, UMSBP has the potential capacity to function in vivo in the activation of the prereplication release of minicircles from the network, a key step in kDNA replication, which precedes and enables its replication initiation. These observations demonstrate the prereplication remodeling of a condensed mitochondrial DNA, which is mediated via specific interactions of histone-like proteins with a replication initiator, rather than through their posttranslational covalent modifications.


Subject(s)
DNA Replication , DNA, Kinetoplast/biosynthesis , DNA-Binding Proteins/metabolism , Genome, Mitochondrial/genetics , Histones/metabolism , Protozoan Proteins/metabolism , Crithidia fasciculata , DNA, Kinetoplast/metabolism , Protein Binding , Substrate Specificity
20.
Mol Cell Biol ; 29(22): 5998-6005, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19752195

ABSTRACT

Programmed DNA double-strand breaks (DSBs) in meiosis are formed by Spo11 (Rec12 in fission yeast), a topoisomerase II-like protein, which becomes covalently attached to DNA 5' ends. For DSB repair through homologous recombination, the protein must be removed from these DNA ends. We show here that Rec12 is endonucleolytically removed from DSB ends attached to a short oligonucleotide (Rec12-oligonucleotide complex), as is Spo11 in budding yeast. Fission yeast, however, has only one size class of Rec12-oligonucleotide complexes, whereas budding yeast has two size classes, suggesting different endonucleolytic regulatory mechanisms. Rec12-oligonucleotide generation strictly requires Ctp1 (Sae2 nuclease homolog), the Rad32 (Mre11) nuclease domain, and Rad50 of the MRN complex. Surprisingly, Nbs1 is not strictly required, indicating separable roles for the MRN subunits. On the basis of these and other data, we propose that Rad32 nuclease has the catalytic site for Rec12-oligonucleotide generation and is activated by Ctp1, which plays an additional role in meiotic recombination.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , DNA-Binding Proteins/metabolism , Meiosis , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces/cytology , Endonucleases/metabolism , Kinetics , Microbial Viability , Models, Biological , Mutation/genetics , Oligonucleotides/metabolism , Recombination, Genetic , Schizosaccharomyces/enzymology , Spores, Fungal/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...