Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
PLoS Pathog ; 17(9): e1008768, 2021 09.
Article in English | MEDLINE | ID: mdl-34559857

ABSTRACT

Trypanosome Lytic Factor (TLF) is a primate-specific high-density lipoprotein (HDL) complex that, through the cation channel-forming protein apolipoprotein L-1 (APOL1), provides innate immunity to select kinetoplastid parasites. The immunoprotective effects of TLF have been extensively investigated in the context of its interaction with the extracellular protozoan Trypanosoma brucei brucei, to which it confers sterile immunity. We previously showed that TLF could act against an intracellular pathogen Leishmania, and here we dissected the role of TLF and its synergy with host-immune cells. Leishmania major is transmitted by Phlebotomine sand flies, which deposit the parasite intradermally into mammalian hosts, where neutrophils are the predominant phagocytes recruited to the site of infection. Once in the host, the parasites are phagocytosed and shed their surface glycoconjugates during differentiation to the mammalian-resident amastigote stage. Our data show that mice producing TLF have reduced parasite burdens when infected intradermally with metacyclic promastigotes of L. major, the infective, fly-transmitted stage. This TLF-mediated reduction in parasite burden was lost in neutrophil-depleted mice, suggesting that early recruitment of neutrophils is required for TLF-mediated killing of L. major. In vitro we find that only metacyclic promastigotes co-incubated with TLF in an acidic milieu were lysed. However, amastigotes were not killed by TLF at any pH. These findings correlated with binding experiments, revealing that labeled TLF binds specifically to the surface of metacyclic promastigotes, but not to amastigotes. Metacyclic promastigotes of L. major deficient in the synthesis of surface glycoconjugates LPG and/or PPG (lpg1- and lpg5A-/lpg5B- respectively) whose absence mimics the amastigote surface, were resistant to TLF-mediated lysis. We propose that TLF binds to the outer surface glycoconjugates of metacyclic promastigotes, whereupon it kills the parasite in the acidic phagosome of phagocytes. We hypothesize that resistance to TLF requires shedding of the surface glycoconjugates, which occurs several hours after phagocytosis by immune cells, creating a relatively short-lived but effective window for TLF to act against Leishmania.


Subject(s)
Host-Parasite Interactions/physiology , Immunity, Innate , Leishmaniasis, Cutaneous , Lipoproteins, HDL/metabolism , Animals , Humans , Leishmania major , Leishmaniasis, Cutaneous/immunology , Leishmaniasis, Cutaneous/metabolism , Leishmaniasis, Cutaneous/pathology , Lipoproteins, HDL/immunology , Mice
2.
J Biol Chem ; 295(38): 13138-13149, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32727852

ABSTRACT

The human innate immunity factor apolipoprotein L-I (APOL1) protects against infection by several protozoan parasites, including Trypanosoma brucei brucei Endocytosis and acidification of high-density lipoprotein-associated APOL1 in trypanosome endosomes leads to eventual lysis of the parasite due to increased plasma membrane cation permeability, followed by colloid-osmotic swelling. It was previously shown that recombinant APOL1 inserts into planar lipid bilayers at acidic pH to form pH-gated nonselective cation channels that are opened upon pH neutralization. This corresponds to the pH changes encountered during endocytic recycling, suggesting APOL1 forms a cytotoxic cation channel in the parasite plasma membrane. Currently, the mechanism and domains required for channel formation have yet to be elucidated, although a predicted helix-loop-helix (H-L-H) was suggested to form pores by virtue of its similarity to bacterial pore-forming colicins. Here, we compare recombinant human and baboon APOL1 orthologs, along with interspecies chimeras and individual amino acid substitutions, to identify regions required for channel formation and pH gating in planar lipid bilayers. We found that whereas neutralization of glutamates within the H-L-H may be important for pH-dependent channel formation, there was no evidence of H-L-H involvement in either pH gating or ion selectivity. In contrast, we found two residues in the C-terminal domain, tyrosine 351 and glutamate 355, that influence pH gating properties, as well as a single residue, aspartate 348, that determines both cation selectivity and pH gating. These data point to the predicted transmembrane region closest to the APOL1 C terminus as the pore-lining segment of this novel channel-forming protein.


Subject(s)
Apolipoprotein L1/chemistry , Immunity, Innate , Animals , Apolipoprotein L1/genetics , Apolipoprotein L1/immunology , Helix-Loop-Helix Motifs , Humans , Hydrogen-Ion Concentration , Papio hamadryas , Trypanosoma brucei brucei/immunology
3.
J Cell Biochem ; 118(2): 237-251, 2017 02.
Article in English | MEDLINE | ID: mdl-27302072

ABSTRACT

Mammalian NUMB is alternatively spliced generating four isoforms NUMB1-NUMB4 that can function as tumor suppressors. NUMB1-NUMB4 proteins, which normally determine how different cell types develop, are reduced in 21% of primary breast tumors. Our previous work has, however, indicated that two novel NUMB isoforms, NUMB5 and NUMB6 have the pro-oncogenic functions. Herein, we address a novel function of human NUMB isoform 6 (NUMB6) in promoting cancer cell migration and invasion. We found that NUMB6 induced expression of embryonic transcription factor Slug, which in turn actively repressed E-cadherin, prompting cells to undergo epithelial-mesenchymal transition (EMT). Low-metastatic breast cancer cells DB-7 stably expressing NUMB6, lost their epithelial phenotype, exhibited migratory and pro-invasive behavior, and ultimately elevated expression of mesenchymal markers. Among these markers, increased vimentin, ß-catenin, and fibronectin expression elicited metalloproteinase 9 (MMP9) production. Our results revealed that NUMB6-DB-7 cells have significantly increased level of Akt1 and Akt2 phosphorylation. Therefore, antagonizing Akt signaling using a chemical inhibitor LY294002, we found that NUMB6-induced Slug expression was reduced, and ultimately accompanied with decreased cell migration and invasion. In summary, this study identified a novel molecular determinant of breast cancer progression, uncovering a potential oncogenic role for the NUMB6 protein in cancer cell migration and invasion, coupled to the maintenance of mesenchymal-like cells. J. Cell. Biochem. 118: 237-251, 2017. © 2016 Wiley Periodicals, Inc.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Neoplastic Stem Cells/metabolism , Nerve Tissue Proteins/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Membrane Proteins/genetics , Neoplasm Invasiveness , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/genetics , Protein Isoforms/genetics , Protein Isoforms/metabolism
4.
Toxicol Lett ; 396: 1-10, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38588756

ABSTRACT

The surge in opioid-related deaths, driven predominantly by fentanyl and its synthetic derivatives, has become a critical public health concern, which is particularly evident in the United States. While the situation is less severe in Europe, the European Monitoring Centre for Drugs and Drug Addiction reports a rise in drug overdose deaths, with emerging concerns about the impact of fentanyl-related molecules. Synthetic opioids, initially designed for medical use, have infiltrated illicit markets due to their low production costs and high potency, with carfentanil posing additional threats, including potential chemical weaponization. Existing overdose mitigation heavily relies on naloxone, requiring timely intervention and caregiver presence, while therapeutic prevention strategies face many access challenges. To provide an additional treatment option, we propose the use of a fentanyl-specific monoclonal antibody (mAb), as a non-opioid method of prophylaxis against fentanyl and carfentanil. This mAb shows protection from opioid effects in a pre-clinical murine model. mAbs could emerge as a versatile countermeasure in civilian and biodefense settings, offering a novel approach to combat opioid-associated mortality.


Subject(s)
Analgesics, Opioid , Antibodies, Monoclonal , Fentanyl , Fentanyl/analogs & derivatives , Fentanyl/immunology , Animals , Mice , Humans
5.
PLoS Negl Trop Dis ; 17(9): e0011621, 2023 09.
Article in English | MEDLINE | ID: mdl-37656766

ABSTRACT

Long-term immune evasion by the African trypanosome is achieved through repetitive cycles of surface protein replacement with antigenically distinct versions of the dense Variant Surface Glycoprotein (VSG) coat. Thousands of VSG genes and pseudo-genes exist in the parasite genome that, together with genetic recombination mechanisms, allow for essentially unlimited immune escape from the adaptive immune system of the host. The diversity space of the "VSGnome" at the protein level was thought to be limited to a few related folds whose structures were determined more than 30 years ago. However, recent progress has shown that the VSGs possess significantly more architectural variation than had been appreciated. Here we combine experimental X-ray crystallography (presenting structures of N-terminal domains of coat proteins VSG11, VSG21, VSG545, VSG558, and VSG615) with deep-learning prediction using Alphafold to produce models of hundreds of VSG proteins. We classify the VSGnome into groups based on protein architecture and oligomerization state, contextualize recent bioinformatics clustering schemes, and extensively map VSG-diversity space. We demonstrate that in addition to the structural variability and post-translational modifications observed thus far, VSGs are also characterized by variations in oligomerization state and possess inherent flexibility and alternative conformations, lending additional variability to what is exposed to the immune system. Finally, these additional experimental structures and the hundreds of Alphafold predictions confirm that the molecular surfaces of the VSGs remain distinct from variant to variant, supporting the hypothesis that protein surface diversity is central to the process of antigenic variation used by this organism during infection.


Subject(s)
Antigenic Variation , Membrane Glycoproteins , Protozoan Proteins , Trypanosoma , Membrane Proteins
6.
Cell Rep ; 42(3): 112262, 2023 03 28.
Article in English | MEDLINE | ID: mdl-36943866

ABSTRACT

The African trypanosome survives the immune response of its mammalian host by antigenic variation of its major surface antigen (the variant surface glycoprotein or VSG). Here we describe the antibody repertoires elicited by different VSGs. We show that the repertoires are highly restricted and are directed predominantly to distinct epitopes on the surface of the VSGs. They are also highly discriminatory; minor alterations within these exposed epitopes confer antigenically distinct properties to these VSGs and elicit different repertoires. We propose that the patterned and repetitive nature of the VSG coat focuses host immunity to a restricted set of immunodominant epitopes per VSG, eliciting a highly stereotyped response, minimizing cross-reactivity between different VSGs and facilitating prolonged immune evasion through epitope variation.


Subject(s)
Trypanosoma brucei brucei , Trypanosoma , Animals , Immunodominant Epitopes , Immune Evasion , Variant Surface Glycoproteins, Trypanosoma , Antigenic Variation , Epitopes , Mammals
7.
Cell Rep ; 42(2): 112049, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36719797

ABSTRACT

Poorly immunogenic small molecules pose challenges for the production of clinically efficacious vaccines and antibodies. To address this, we generate an immunization platform derived from the immunogenic surface coat of the African trypanosome. Through sortase-based conjugation of the target molecules to the variant surface glycoprotein (VSG) of the trypanosome surface coat, we develop VSG-immunogen array by sortase tagging (VAST). VAST elicits antigen-specific memory B cells and antibodies in a murine model after deploying the poorly immunogenic molecule fentanyl as a proof of concept. We also develop a single-cell RNA sequencing (RNA-seq)-based computational method that synergizes with VAST to specifically identify memory B cell-encoded antibodies. All computationally selected antibodies bind to fentanyl with picomolar affinity. Moreover, these antibodies protect mice from fentanyl effects after passive immunization, demonstrating the ability of these two coupled technologies to elicit therapeutic antibodies to challenging immunogens.


Subject(s)
Trypanosoma brucei brucei , Trypanosoma , Trypanosomiasis, African , Animals , Mice , Trypanosoma brucei brucei/genetics , Trypanosomiasis, African/drug therapy , Analgesics, Opioid , Fentanyl/pharmacology , Fentanyl/therapeutic use , Variant Surface Glycoproteins, Trypanosoma , Immunotherapy
8.
Nat Commun ; 13(1): 3308, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35676259

ABSTRACT

During the ongoing COVID-19 pandemic, PCR testing and antigen tests have proven critical for helping to stem the spread of its causative agent, SARS-CoV-2. However, these methods suffer from either general applicability and/or sensitivity. Moreover, the emergence of variant strains creates the need for flexibility to correctly and efficiently diagnose the presence of substrains. To address these needs we developed the diagnostic test ADESSO (Accurate Detection of Evolving SARS-CoV-2 through SHERLOCK (Specific High Sensitivity Enzymatic Reporter UnLOCKing) Optimization) which employs Cas13 to diagnose patients in 1 h without sophisticated equipment. Using an extensive panel of clinical samples, we demonstrate that ADESSO correctly identifies infected individuals at a sensitivity and specificity comparable to RT-qPCR on extracted RNA and higher than antigen tests for unextracted samples. Altogether, ADESSO is a fast, sensitive and cheap method that can be applied in a point of care setting to diagnose COVID-19 and can be quickly adjusted to detect new variants.


Subject(s)
COVID-19 , COVID-19/diagnosis , COVID-19 Testing , Humans , Pandemics , RNA, Viral/analysis , RNA, Viral/genetics , SARS-CoV-2 , Sensitivity and Specificity
9.
J Cell Physiol ; 226(11): 3064-75, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21302306

ABSTRACT

FGF1, a widely expressed proangiogenic factor involved in tissue repair and carcinogenesis, is released from cells through a non-classical pathway independent of endoplasmic reticulum and Golgi. Although several proteins participating in FGF1 export were identified, genetic mechanisms regulating this process remained obscure. We found that FGF1 export and expression are regulated through Notch signaling mediated by transcription factor CBF1 and its partner MAML. The expression of a dominant negative (dn) form of CBF1 in 3T3 cells induces transcription of FGF1 and sphingosine kinase 1 (SphK1), which is a component of FGF1 export pathway. dnCBF1 expression stimulates the stress-independent release of transduced FGF1 from NIH 3T3 cells and endogenous FGF1 from A375 melanoma cells. NIH 3T3 cells transfected with dnCBF1 form colonies in soft agar and produce rapidly growing highly angiogenic tumors in nude mice. The transformed phenotype of dnCBF1 transfected cells is efficiently blocked by dn forms of FGF receptor 1 and S100A13, which is a component of FGF1 export pathway. FGF1 export and acceleration of cell growth induced by dnCBF1 depend on SphK1. Similar to dnCBF1, dnMAML transfection induces FGF1 expression and release, and accelerates cell proliferation. The latter effect is strongly decreased in FGF1 null cells. We suggest that the regulation of FGF1 expression and release by CBF1-mediated Notch signaling can play an important role in tumor formation.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Fibroblast Growth Factor 1/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Receptors, Notch/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic/genetics , Humans , Melanoma/drug therapy , Melanoma/metabolism , Mice , Mice, Nude , NIH 3T3 Cells , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Nuclear Proteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , S100 Proteins/pharmacology , Signal Transduction/drug effects , Transcription Factors/metabolism , Transfection
10.
Nat Cell Biol ; 4(8): 547-55, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12105416

ABSTRACT

beta-Arrestins are important in chemoattractant receptor-induced granule release, a process that may involve Ral-dependent regulation of the actin cytoskeleton. We have identified the Ral GDP dissociation stimulator (Ral-GDS) as a beta-arrestin-binding protein by yeast two-hybrid screening and co-immunoprecipitation from human polymorphonuclear neutrophilic leukocytes (PMNs). Under basal conditions, Ral-GDS is localized to the cytosol and remains inactive in a complex formed with beta-arrestins. In response to formyl-Met-Leu-Phe (fMLP) receptor stimulation, beta-arrestin Ral-GDS protein complexes dissociate and Ral-GDS translocates with beta-arrestin from the cytosol to the plasma membrane, resulting in the Ras-independent activation of the Ral effector pathway required for cytoskeletal rearrangement. The subsequent re-association of beta-arrestin Ral-GDS complexes is associated with the inactivation of Ral signalling. Thus, beta-arrestins regulate multiple steps in the Ral-dependent processes that result in chemoattractant-induced cytoskeletal reorganization.


Subject(s)
Arrestins/metabolism , Cytoskeleton/metabolism , ral GTP-Binding Proteins/metabolism , ral Guanine Nucleotide Exchange Factor/metabolism , Animals , Arrestins/chemistry , Biological Transport, Active , COS Cells , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cytoskeleton/drug effects , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Humans , Macromolecular Substances , Models, Biological , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophils/drug effects , Neutrophils/metabolism , Rats , Receptors, Formyl Peptide , Receptors, Immunologic/metabolism , Receptors, Peptide/metabolism , Signal Transduction , Two-Hybrid System Techniques , beta-Arrestins , ral GTP-Binding Proteins/chemistry , ral Guanine Nucleotide Exchange Factor/chemistry
11.
BMC Biol ; 8: 7, 2010 Jan 25.
Article in English | MEDLINE | ID: mdl-20100315

ABSTRACT

BACKGROUND: Previous studies have linked neurotrophin receptor-interacting MAGE protein to the bone morphogenic protein signaling pathway and its effect on p38 mediated apoptosis of neural progenitor cells via the XIAP-Tak1-Tab1 complex. Its effect on NF-kappaB has yet to be explored. RESULTS: Herein we report that NRAGE, via the same XIAP-Tak1-Tab1 complex, is required for the phosphorylation of IKK -alpha/beta and subsequent transcriptional activation of the p65 subunit of NF-kappaB. Ablation of endogenous NRAGE by siRNA inhibited NF-kappaB pathway activation, while ablation of Tak1 and Tab1 by morpholino inhibited overexpression of NRAGE from activating NF-kappaB. Finally, cytokine profiling of an NRAGE over-expressing stable line revealed the expression of macrophage migration inhibitory factor. CONCLUSION: Modulation of NRAGE expression revealed novel roles in regulating NF-kappaB activity in the non-canonical bone morphogenic protein signaling pathway. The expression of macrophage migration inhibitory factor by bone morphogenic protein -4 reveals novel crosstalk between an immune cytokine and a developmental pathway.


Subject(s)
Antigens, Neoplasm/metabolism , Bone Morphogenetic Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/drug effects , Transcription Factor RelA/metabolism , Animals , Antigens, Neoplasm/genetics , Blotting, Western , Bone Morphogenetic Protein 4/pharmacology , Bone Morphogenetic Proteins/genetics , Cell Line , Cell Line, Tumor , Humans , I-kappa B Kinase/metabolism , Immunohistochemistry , Immunoprecipitation , Kidney/metabolism , Macrophage Migration-Inhibitory Factors/genetics , Macrophage Migration-Inhibitory Factors/metabolism , Mice , Mice, Transgenic , Neoplasm Proteins/genetics , Phosphorylation/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/physiology , Signal Transduction/genetics , Transcription Factor RelA/genetics
12.
PLoS Negl Trop Dis ; 15(9): e0009814, 2021 09.
Article in English | MEDLINE | ID: mdl-34587165

ABSTRACT

Anemia caused by trypanosome infection is poorly understood. Autoimmunity during Trypanosoma brucei infection was proposed to have a role during anemia, but the mechanisms involved during this pathology have not been elucidated. In mouse models and human patients infected with malaria parasites, atypical B-cells promote anemia through the secretion of autoimmune anti-phosphatidylserine (anti-PS) antibodies that bind to uninfected erythrocytes and facilitate their clearance. Using mouse models of two trypanosome infections, Trypanosoma brucei and Trypanosoma cruzi, we assessed levels of autoantibodies and anemia. Our results indicate that acute T. brucei infection, but not T. cruzi, leads to early increased levels of plasma autoantibodies against different auto antigens tested (PS, DNA and erythrocyte lysate) and expansion of atypical B cells (ABCs) that secrete these autoantibodies. In vitro studies confirmed that a lysate of T. brucei, but not T. cruzi, could directly promote the expansion of these ABCs. PS exposure on erythrocyte plasma membrane seems to be an important contributor to anemia by delaying erythrocyte recovery since treatment with an agent that prevents binding to it (Annexin V) ameliorated anemia in T. brucei-infected mice. Analysis of the plasma of patients with human African trypanosomiasis (HAT) revealed high levels of anti-PS antibodies that correlated with anemia. Altogether these results suggest a relation between autoimmunity against PS and anemia in both mice and patients infected with T. brucei.


Subject(s)
Anemia/etiology , Autoimmunity , Phosphatidylserines/immunology , Trypanosomiasis, African/immunology , Adolescent , Adult , Animals , Autoantibodies/immunology , Erythrocytes/immunology , Female , Humans , Male , Mice , Middle Aged , Trypanosoma , Trypanosoma brucei brucei/immunology , Trypanosomiasis, African/complications , Young Adult
13.
Nat Microbiol ; 6(3): 392-400, 2021 03.
Article in English | MEDLINE | ID: mdl-33462435

ABSTRACT

Suramin has been a primary early-stage treatment for African trypanosomiasis for nearly 100 yr. Recent studies revealed that trypanosome strains that express the variant surface glycoprotein (VSG) VSGsur possess heightened resistance to suramin. Here, we show that VSGsur binds tightly to suramin but other VSGs do not. By solving high-resolution crystal structures of VSGsur and VSG13, we also demonstrate that these VSGs define a structurally divergent subgroup of the coat proteins. The co-crystal structure of VSGsur with suramin reveals that the chemically symmetric drug binds within a large cavity in the VSG homodimer asymmetrically, primarily through contacts of its central benzene rings. Structure-based, loss-of-contact mutations in VSGsur significantly decrease the affinity to suramin and lead to a loss of the resistance phenotype. Altogether, these data show that the resistance phenotype is dependent on the binding of suramin to VSGsur, establishing that the VSG proteins can possess functionality beyond their role in antigenic variation.


Subject(s)
Drug Resistance/immunology , Suramin/metabolism , Trypanosoma brucei rhodesiense/immunology , Variant Surface Glycoproteins, Trypanosoma/chemistry , Variant Surface Glycoproteins, Trypanosoma/metabolism , Antigenic Variation/drug effects , Antigenic Variation/immunology , Binding Sites , Crystallography, X-Ray , Drug Resistance/genetics , Endocytosis/genetics , Immune Evasion , Mutation , Protein Binding , Protein Conformation , Suramin/toxicity , Trypanocidal Agents/metabolism , Trypanocidal Agents/toxicity , Trypanosoma brucei rhodesiense/chemistry , Trypanosoma brucei rhodesiense/drug effects , Trypanosoma brucei rhodesiense/metabolism , Trypanosomiasis, African/parasitology , Variant Surface Glycoproteins, Trypanosoma/genetics
14.
Apoptosis ; 15(1): 63-70, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19937275

ABSTRACT

Bone morphogenetic signaling (BMP) is a key pathway during neurogenesis and depends on many downstream intermediators to carry out its signaling. One such signaling pathway utilizes neurotrophin receptor-interacting MAGE protein (NRAGE), a member of the melanoma-associated antigen (MAGE) family, to upregulate p38 mitogen activated protein kinase (p38(MAPK)) in response to cellular stress and activate caspases which are critical in leading cells to death. NRAGE consists of two conserved MAGE homology domains separated by a unique hexapeptide repeat domain. Although we have previously implicated NRAGE in inducing apoptosis in neural progenitors and P19 cells, a model system for neural progenitors, its domains have yet to be explored in determining which one may be responsible for setting up the signaling for apoptosis. Here, we overexpressed a series of deletion mutations in P19 cells to show that only those with at least half of the repeat domain, activated p38(MAPK) and underwent apoptosis offering intriguing incite into NRAGE's contribution in BMP apoptotic signaling.


Subject(s)
Apoptosis , Bone Morphogenetic Proteins/metabolism , Neoplasm Proteins/chemistry , Signal Transduction , Animals , Bone Morphogenetic Proteins/genetics , Cell Line , Cell Survival , Female , Humans , Mice , Mice, Inbred ICR , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Protein Structure, Tertiary , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Methods Mol Biol ; 2116: 463-483, 2020.
Article in English | MEDLINE | ID: mdl-32221937

ABSTRACT

Interest in trypanosome lytic factors (TLFs) and apolipoprotein L1, the ion channel-forming protein component of TLFs, has increased tenfold since 2010. This is due to the association of African variants of APOL1 with kidney disease such that interest has reached circles beyond parasitology. We have extensive experience purifying and working with these proteins and protein complexes. Herein we describe our detailed purification protocols to aid the new burgeoning field by providing an opportunity for consistency in reagents used across laboratories. We emphasize that it is imperative to maintain APOL1 protein intact (~42 kDa) to analyze the active ion channel-forming component/protein.


Subject(s)
Apolipoprotein L1/isolation & purification , Lipoproteins, HDL/isolation & purification , Trypanosomiasis, African/blood , Apolipoprotein L1/blood , Apolipoprotein L1/chemistry , Apolipoprotein L1/metabolism , Humans , Kidney Diseases/blood , Kidney Diseases/immunology , Lipoproteins, HDL/chemistry , Lipoproteins, HDL/metabolism , Molecular Weight , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism , Trypanosoma/immunology , Trypanosomiasis, African/complications , Trypanosomiasis, African/immunology , Trypanosomiasis, African/parasitology
16.
Cell Host Microbe ; 28(1): 79-88.e4, 2020 07 08.
Article in English | MEDLINE | ID: mdl-32416060

ABSTRACT

Trypanosomiasis is a devastating neglected tropical disease affecting livestock and humans. Humans are susceptible to two Trypanosoma brucei subspecies but protected from other trypanosomes by circulating high-density lipoprotein (HDL) complexes called trypanosome lytic factors (TLFs) 1 and 2. TLFs contain apolipoprotein L-1 contributing to lysis and haptoglobin-related protein (HPR), which can function as a ligand for a parasite receptor. TLF2 also uniquely contains non-covalently associated immunoglobin M (IgM) antibodies, the role and origin of which remain unclear. Here, we show that these TLF2-associated IgMs interact with both HPR and alternate trypanosome surface proteins, including variant surface glycoprotein, likely facilitating complex biogenesis and TLF uptake into parasites. TLF2-IgMs are germline antibodies that, while present at basal concentrations in healthy individuals, are elicited by trypanosome infection in both murine models and human sleeping sickness patients. These data suggest that poly- and self-reactive germline antibodies such as TLF2-associated IgMs play a role in antimicrobial immunity.


Subject(s)
Antibodies, Protozoan/immunology , Antigens, Neoplasm/immunology , Apolipoprotein L1/immunology , Haptoglobins/immunology , Immunoglobulin M/immunology , Lipoproteins, HDL/immunology , Trypanosomiasis, African/immunology , Adolescent , Adult , Aged , Animals , Cell Line , Child , Female , Germ Cells/immunology , Host-Parasite Interactions , Humans , Male , Membrane Glycoproteins/immunology , Mice , Mice, Knockout , Middle Aged , Models, Animal , Parasites , Trypanosoma brucei brucei , Young Adult
17.
Biotechniques ; 44(4): 547-9, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18476819

ABSTRACT

Cellular interactions in development of the kidney are used as a model of reciprocal inductive events between epithelium and mesenchyme. Time- and labor-intensive methods have been developed to study this phenomenon. For example, in mice, the targeted disruption of genes in vivo has been used to modify the genetic program directing kidney development. However, gene targeting is a resource-intensive approach and alternative strategies for gene and protein modification in the kidney need to be developed. Herein, we have developed an efficient system for the delivery of antisense morpholino to alter normal protein expression. We describe the use of Endo-Porter to effectively deliver morpholinos to all parts and regions of the kidney explant. Also, we definitively show via confocal microscopy and Western blot analysis that the use of Endo-Porter in delivering antisense morpholinos is robust throughout the entire kidney explant, providing efficient suppression of protein expression. This method saves time and cost when compared with targeted disruption and is an improvement upon previous kidney organ culture methods.


Subject(s)
Drug Delivery Systems , Kidney/cytology , Peptides , Animals , Fluorescein/metabolism , Fluorescent Dyes/metabolism , Mice , Mice, Inbred ICR , Models, Biological , Neoplasm Proteins/antagonists & inhibitors , Oligonucleotides, Antisense/chemistry , Organ Culture Techniques , RNA, Messenger/metabolism , Time Factors
18.
Stem Cells ; 25(12): 3133-42, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17761753

ABSTRACT

The activation of Notch signaling in neural crest stem cells (NCSCs) results in the rapid loss of neurogenic potential and differentiation into glia. We now show that the attenuation of endogenous Notch signaling within expanding NCSC clones by the Notch ligand soluble Jagged1 (sJ1), maintains NCSCs in a clonal self-renewing state in vitro without affecting their sensitivity to instructive differentiation signals observed previously during NCSC self-renewal. sJ1 functions as a competitive inhibitor of Notch signaling to modulate endogenous cell-cell communication to levels sufficient to inhibit neural differentiation but insufficient to instruct gliogenic differentiation. Attenuated Notch signaling promotes the induction and nonclassic release of fibroblast growth factor 1 (FGF1). The functions of sJ1 and FGF1 signaling are complementary, as abrogation of FGF signaling diminishes the ability of sJ1 to promote NCSC expansion, yet the secondary NCSCs maintain the dosage sensitivity of the founder. These results validate and build upon previous studies on the role of Notch signaling in stem cell self-renewal and suggest that the differentiation bias or self-renewal potential of NCSCs is intrinsically linked to the level of endogenous Notch signaling. This should provide a unique opportunity for the expansion of NCSCs ex vivo without altering their differentiation bias for clinical cell replacement or transplant strategies in tissue repair. Disclosure of potential conflicts of interest is found at the end of this article.


Subject(s)
Calcium-Binding Proteins/physiology , Cell Differentiation/physiology , Intercellular Signaling Peptides and Proteins/physiology , Membrane Proteins/physiology , Neural Crest/cytology , Neural Crest/physiology , Stem Cells/cytology , Stem Cells/physiology , Animals , Calcium-Binding Proteins/genetics , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Clone Cells/cytology , Clone Cells/physiology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Jagged-1 Protein , Membrane Proteins/genetics , Neural Inhibition/genetics , Neural Inhibition/physiology , Neurons/cytology , Neurons/physiology , Rats , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch1/physiology , Serrate-Jagged Proteins , Signal Transduction/genetics , Signal Transduction/physiology , Solubility , Stem Cells/metabolism
19.
Mol Cell Biol ; 25(17): 7711-24, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16107717

ABSTRACT

Understanding the molecular events that govern neural progenitor lineage commitment, mitotic arrest, and differentiation into functional progeny are germane to our understanding of neocortical development. Members of the family of bone morphogenetic proteins (BMPs) play pivotal roles in regulating neural differentiation and apoptosis during neurogenesis through combined actions involving Smad and TAK1 activation. We demonstrate that BMP signaling is required for the induction of apoptosis of neural progenitors and that NRAGE is a mandatory component of the signaling cascade. NRAGE possesses the ability to bind and function with the TAK1-TAB1-XIAP complex facilitating the activation of p38. Disruption of NRAGE or any other member of the noncanonical signaling cascaded is sufficient to block p38 activation and thus the proapoptotic signals generated through BMP exposure. The function of NRAGE is independent of Smad signaling, but the introduction of a dominant-negative Smad5 also rescues neural progenitor apoptosis, suggesting that both canonical and noncanonical pathways can converge and regulate BMP-mediated apoptosis. Collectively, these results establish NRAGE as an integral component in BMP signaling and clarify its role during neural progenitor development.


Subject(s)
Apoptosis/drug effects , Bone Morphogenetic Proteins/pharmacology , Neoplasm Proteins/metabolism , Neurons/cytology , Signal Transduction/drug effects , Stem Cells/cytology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Bone Morphogenetic Protein 4 , Caspase 3 , Caspases/metabolism , Cell Cycle , Cell Differentiation , Cells, Cultured , Enzyme Activation , MAP Kinase Signaling System/drug effects , Mice , Neurons/metabolism , Stem Cells/metabolism , Tretinoin/pharmacology
20.
Nat Microbiol ; 3(8): 932-938, 2018 08.
Article in English | MEDLINE | ID: mdl-29988048

ABSTRACT

The African trypanosome Trypanosoma brucei spp. is a paradigm for antigenic variation, the orchestrated alteration of cell surface molecules to evade host immunity. The parasite elicits robust antibody-mediated immune responses to its variant surface glycoprotein (VSG) coat, but evades immune clearance by repeatedly accessing a large genetic VSG repertoire and 'switching' to antigenically distinct VSGs. This persistent immune evasion has been ascribed exclusively to amino-acid variance on the VSG surface presented by a conserved underlying protein architecture. We establish here that this model does not account for the scope of VSG structural and biochemical diversity. The 1.4-Å-resolution crystal structure of the variant VSG3 manifests divergence in the tertiary fold and oligomeric state. The structure also reveals an O-linked carbohydrate on the top surface of VSG3. Mass spectrometric analysis indicates that this O-glycosylation site is heterogeneously occupied in VSG3 by zero to three hexose residues and is also present in other VSGs. We demonstrate that this O-glycosylation increases parasite virulence by impairing the generation of protective immunity. These data alter the paradigm of antigenic variation by the African trypanosome, expanding VSG variability beyond amino-acid sequence to include surface post-translational modifications with immunomodulatory impact.


Subject(s)
Antibodies, Protozoan/metabolism , Trypanosoma brucei brucei/pathogenicity , Variant Surface Glycoproteins, Trypanosoma/chemistry , Variant Surface Glycoproteins, Trypanosoma/genetics , Binding Sites , Crystallography, X-Ray , Genetic Variation , Glycosylation , Models, Molecular , Protein Conformation , Protein Domains , Trypanosoma brucei brucei/immunology , Variant Surface Glycoproteins, Trypanosoma/immunology
SELECTION OF CITATIONS
SEARCH DETAIL