Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
J Virol ; : e0202023, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38884472

ABSTRACT

Human noroviruses (HuNoVs) are a diverse group of RNA viruses that cause endemic and pandemic acute viral gastroenteritis. Previously, we reported that many HuNoV strains require bile or bile acid (BA) to infect human jejunal intestinal enteroid cultures. BA was not essential for the replication of a pandemic-causing GII.4 HuNoV strain. We found the hydrophobic BA glycochenodeoxycholic acid (GCDCA) promotes the replication of the BA-dependent strain GII.3 in jejunal enteroids. Furthermore, we found that inhibition of the G-protein-coupled BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), by JTE-013, reduced GII.3 infection dose-dependently and inhibited GII.3 cellular uptake in enteroids. Herein, we sought to determine whether S1PR2 is required for other BA-dependent HuNoV strains, the BA-independent GII.4, and whether S1PR2 is required for BA-dependent HuNoV infection in HIEs from other small intestinal segments. We found a second S1PR2 inhibitor, GLPG2938, reduces GII.3 infection dose-dependently, and an S1PR2 agonist (CYM-5520) enhances GII.3 replication in the absence of GCDCA. GII.3 replication also is abrogated in the presence of JTE-013 and CYM-5520. JTE-013 inhibition of S1PR2 in jejunal HIEs reduces GI.1, GII.3, and GII.17 (BA-dependent) but not GII.4 Sydney (BA-independent) infection, providing additional evidence of strain-specific differences in HuNoV infection. Finally, GII.3 infection of duodenal, jejunal, and ileal lines derived from the same individual is reduced with S1PR2 inhibition, indicating a common mechanism of BA-dependent infection among multiple segments of the small intestine. Our results support a model where BA-dependent HuNoVs exploit BA effects on S1PR2 to infect the entire small intestine.IMPORTANCEHuman noroviruses (HuNoVs) are important viral human pathogens that cause both outbreaks and sporadic gastroenteritis. These viruses are diverse, and many strains are capable of infecting humans. Our previous studies have identified strain-specific requirements for hydrophobic bile acids (BAs) to infect intestinal epithelial cells. Moreover, we identified a BA receptor, sphingosine-1-phosphate receptor 2 (S1PR2), required for infection by a BA-dependent strain. To better understand how various HuNoV strains enter and infect the small intestine and the role of S1PR2 in HuNoV infection, we evaluated infection by additional HuNoV strains using an expanded repertoire of intestinal enteroid cell lines. We found that multiple BA-dependent strains, but not a BA-independent strain, all require S1PR2 for infection. In addition, BA-dependent infection requires S1PR2 in multiple segments of the small intestine. Together, these results indicate that S1PR2 has value as a potential therapeutic target for BA-dependent HuNoV infection.

2.
bioRxiv ; 2024 May 30.
Article in English | MEDLINE | ID: mdl-38853945

ABSTRACT

Human noroviruses (HuNoVs) are the leading cause of acute gastroenteritis worldwide. Currently, there are no targeted antivirals for the treatment of HuNoV infection. Histo-blood group antigens (HBGAs) on the intestinal epithelium are cellular attachment factors for HuNoVs; molecules that block the binding of HuNoVs to HBGAs thus have the potential to be developed as antivirals. Human milk oligosaccharides (HMOs) are glycans in human milk with structures analogous to HBGAs. HMOs have been shown to act as decoy receptors to prevent the attachment of multiple enteric pathogens to host cells. Previous X-ray crystallography studies have demonstrated the binding of HMO 2'-fucosyllactose (2'FL) in the same pocket as HBGAs for some HuNoV strains. We evaluated the effect of 2'FL on the replication of a globally dominant GII.4 Sydney [P16] HuNoV strain using human intestinal enteroids (HIEs) from adults and children. A significant reduction in GII.4 Sydney [P16] replication was seen in duodenal and jejunal HIEs from multiple adult donors, all segments of the small intestine from an adult organ donor and in two pediatric duodenal HIEs. However, 2'FL did not inhibit HuNoV replication in two infant jejunal HIEs that had significantly lower expression of α1-2-fucosylated glycans. 2'FL can be synthesized in large scale, and safety and tolerance have been assessed previously. Our data suggest that 2'FL has the potential to be developed as a therapeutic for HuNoV gastroenteritis.

3.
bioRxiv ; 2024 May 24.
Article in English | MEDLINE | ID: mdl-38826387

ABSTRACT

Human noroviruses (HuNoVs) are a significant cause of both epidemic and sporadic acute gastroenteritis worldwide. The lack of a reproducible culture system for HuNoVs was a major obstacle in studying virus replication and pathogenesis for almost a half-century. This barrier was overcome with our successful cultivation of multiple HuNoV strains in human intestinal enteroids (HIEs), which has significantly advanced HuNoV research. We previously optimized culture media conditions and generated genetically-modified HIE cultures to enhance HuNoV replication in HIEs. Building upon these achievements, we now present additional advancements to this culture system, which involve testing different media, unique HIE lines, and additional virus strains. HuNoV infectivity was evaluated and compared in new HIE models, including HIEs generated from different intestinal segments of individual adult organ donors, HIEs made from human embryonic stem cell-derived human intestinal organoids that were transplanted into mice (H9tHIEs), genetically-engineered (J4 FUT2 knock-in [ KI ], J2 STAT1 knock-out [ KO ]) HIEs, as well as HIEs derived from a patient with common variable immunodeficiency (CVID) and from infants. Our findings reveal that small intestinal HIEs, but not colonoids, from adults, H9tHIEs, HIEs from a CVID patient, and HIEs from infants support HuNoV replication with segment and strain-specific differences in viral infection. J4 FUT2-KI HIEs exhibit the highest susceptibility to HuNoV infection, allowing the cultivation of a broader range of GI and GII HuNoV strains than previously reported. Overall, these results contribute to a deeper understanding of HuNoVs and highlight the transformative potential of HIE cultures in HuNoV research. Importance: Human noroviruses (HuNoVs) are very contagious and cause significant acute gastroenteritis globally, but studying them has been hindered by the lack of a reproducible culture system for nearly 50 years. This barrier was overcome by successfully cultivating multiple HuNoV strains in human intestinal enteroids (HIEs), advancing HuNoV research. We previously optimized culture conditions and developed genetically modified HIEs to enhance HuNoV replication. In this study, we tested different media, unique HIE lines, and additional virus strains, evaluating HuNoV infectivity in new HIE models. These models include HIEs from various intestinal segments of adult donors, human embryonic stem cell-derived HIEs transplanted into mice (H9tHIEs), genetically-engineered HIEs (J4 FUT2 knock-in [ KI ], J2 STAT1 knock-out [ KO ]), HIEs from a common variable immunodeficiency (CVID) patient, and from infants. Our findings show that adult small intestinal HIEs, H9tHIEs, CVID patient HIEs, and infant HIEs support HuNoV replication with segment and strain-specific differences. J4 FUT2-KI HIEs exhibited the highest susceptibility, allowing cultivation of a broader range of HuNoV strains. These results enhance the understanding of HuNoVs and highlight the transformative potential of HIE cultures in HuNoV research.

4.
Nat Commun ; 14(1): 6516, 2023 10 16.
Article in English | MEDLINE | ID: mdl-37845211

ABSTRACT

Acute gastroenteritis caused by human noroviruses (HuNoVs) is a significant global health and economic burden and is without licensed vaccines or antiviral drugs. The GII.4 HuNoV causes most epidemics worldwide. This virus undergoes epochal evolution with periodic emergence of variants with new antigenic profiles and altered specificity for histo-blood group antigens (HBGA), the determinants of cell attachment and susceptibility, hampering the development of immunotherapeutics. Here, we show that a llama-derived nanobody M4 neutralizes multiple GII.4 variants with high potency in human intestinal enteroids. The crystal structure of M4 complexed with the protruding domain of the GII.4 capsid protein VP1 revealed a conserved epitope, away from the HBGA binding site, fully accessible only when VP1 transitions to a "raised" conformation in the capsid. Together with dynamic light scattering and electron microscopy of the GII.4 VLPs, our studies suggest a mechanism in which M4 accesses the epitope by altering the conformational dynamics of the capsid and triggering its disassembly to neutralize GII.4 infection.


Subject(s)
Blood Group Antigens , Caliciviridae Infections , Norovirus , Humans , Capsid Proteins/chemistry , Capsid/metabolism , Norovirus/genetics , Binding Sites , Epitopes/metabolism , Blood Group Antigens/metabolism
5.
Nat Commun ; 14(1): 1148, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36854760

ABSTRACT

Globally, most cases of gastroenteritis are caused by pandemic GII.4 human norovirus (HuNoV) strains with no approved therapies or vaccines available. The cellular pathways that these strains exploit for cell entry and internalization are unknown. Here, using nontransformed human jejunal enteroids (HIEs) that recapitulate the physiology of the gastrointestinal tract, we show that infectious GII.4 virions and virus-like particles are endocytosed using a unique combination of endosomal acidification-dependent clathrin-independent carriers (CLIC), acid sphingomyelinase (ASM)-mediated lysosomal exocytosis, and membrane wound repair pathways. We found that besides the known interaction of the viral capsid Protruding (P) domain with host glycans, the Shell (S) domain interacts with both galectin-3 (gal-3) and apoptosis-linked gene 2-interacting protein X (ALIX), to orchestrate GII.4 cell entry. Recognition of the viral and cellular determinants regulating HuNoV entry provides insight into the infection process of a non-enveloped virus highlighting unique pathways and targets for developing effective therapeutics.


Subject(s)
Cell Membrane , Norovirus , Virus Internalization , Humans , Clathrin , Norovirus/physiology , Signal Transduction , Cell Membrane/virology
6.
mSphere ; 6(1)2021 01 27.
Article in English | MEDLINE | ID: mdl-33504663

ABSTRACT

Human noroviruses (HuNoVs) are the leading cause of epidemic and sporadic acute gastroenteritis worldwide. We previously demonstrated human intestinal stem cell-derived enteroids (HIEs) support cultivation of several HuNoV strains. However, HIEs did not support virus replication from every HuNoV-positive stool sample, which led us to test and optimize new medium conditions, identify characteristics of stool samples that allow replication, and evaluate consistency of replication over time. Optimization of our HIE-HuNoV culture system has shown the following: (i) a new HIE culture medium made with conditioned medium from a single cell line and commercial media promotes robust replication of HuNoV strains that replicated poorly in HIEs grown in our original culture medium made with conditioned media from 3 separate cell lines; (ii) GI.1, 11 GII genotypes (GII.1, GII.2, GII.3, GII.4, GII.6, GII.7, GII.8, GII.12, GII.13, GII.14, and GII.17), and six GII.4 variants can be cultivated in HIEs; (iii) successful replication is more likely with virus in stools with higher virus titers; (iv) GII.4_Sydney_2012 virus replication was reproducible over 3 years; and (v) HuNoV infection is restricted to the small intestine, based on replication of two viral strains in duodenal and ileal HIEs, but not colonoids, from two susceptible donors. These results improve the HIE culture system for HuNoV replication. Use of HIEs by several laboratories worldwide to study the molecular mechanisms that regulate HuNoV replication confirms the usefulness of this culture system, and our optimized methods for virus replication will advance the development of effective therapies and methods for virus control.IMPORTANCE Human noroviruses (HuNoVs) are highly contagious and cause acute and sporadic diarrheal illness in all age groups. In addition, chronic infections occur in immunocompromised cancer and transplant patients. These viruses are antigenically and genetically diverse, and there are strain-specific differences in binding to cellular attachment factors. In addition, new discoveries are being made on strain-specific differences in virus entry and replication and the epithelial cell response to infection in human intestinal enteroids. Human intestinal enteroids are a biologically relevant model to study HuNoVs; however, not all strains can be cultivated at this time. A complete understanding of HuNoV biology thus requires cultivation conditions that will allow the replication of multiple strains. We report optimization of HuNoV cultivation in human intestinal enteroid cultures to increase the numbers of cultivatable strains and the magnitude of replication, which is critical for testing antivirals, neutralizing antibodies, and methods of virus inactivation.


Subject(s)
Intestinal Mucosa/virology , Norovirus/growth & development , Organoids/virology , Child , Child, Preschool , Culture Media , Humans , Infant , Intestinal Mucosa/cytology , Stem Cells/cytology , Virus Replication/physiology
7.
Proc Natl Acad Sci U S A ; 117(38): 23782-23793, 2020 09 22.
Article in English | MEDLINE | ID: mdl-32907944

ABSTRACT

Human noroviruses (HuNoVs) are the leading cause of viral gastroenteritis worldwide; yet currently, no vaccines or FDA-approved antiviral drugs are available to counter these pathogens. To understand HuNoV biology and the epithelial response to infection, we performed transcriptomic analyses, RT-qPCR, CRISPR-Cas9 modification of human intestinal enteroid (HIE) cultures, and functional studies with two virus strains (a pandemic GII.4 and a bile acid-dependent GII.3 strain). We identified a predominant type III interferon (IFN)-mediated innate response to HuNoV infection. Replication of both strains is sensitive to exogenous addition of IFNs, suggesting the potential of IFNs as therapeutics. To obtain insight into IFN pathway genes that play a role in the antiviral response to HuNoVs, we developed knockout (KO) HIE lines for IFN alpha and lambda receptors and the signaling molecules, MAVS, STAT1, and STAT2 An unexpected differential response of enhanced replication and virus spread was observed for GII.3, but not the globally dominant GII.4 HuNoV in STAT1-knockout HIEs compared to parental HIEs. These results indicate cellular IFN responses restrict GII.3 but not GII.4 replication. The strain-specific sensitivities of innate responses against HuNoV replication provide one explanation for why GII.4 infections are more widespread and highlight strain specificity as an important factor in HuNoV biology. Genetically modified HIEs for innate immune genes are useful tools for studying immune responses to viral or microbial pathogens.


Subject(s)
Caliciviridae Infections , Host-Pathogen Interactions/immunology , Interferons , Intestines , Norovirus , CRISPR-Cas Systems , Caliciviridae Infections/immunology , Caliciviridae Infections/virology , Humans , Interferons/genetics , Interferons/metabolism , Intestines/immunology , Intestines/virology , Models, Biological , Norovirus/genetics , Norovirus/immunology , Norovirus/pathogenicity , Organoids/immunology , Organoids/virology , Sequence Analysis, RNA , Transcriptome/genetics , Virus Replication
8.
mBio ; 11(2)2020 03 17.
Article in English | MEDLINE | ID: mdl-32184242

ABSTRACT

Human noroviruses (HuNoVs) are the leading cause of nonbacterial gastroenteritis worldwide. Histo-blood group antigen (HBGA) expression is an important susceptibility factor for HuNoV infection based on controlled human infection models and epidemiologic studies that show an association of secretor status with infection caused by several genotypes. The fucosyltransferase 2 gene (FUT2) affects HBGA expression in intestinal epithelial cells; secretors express a functional FUT2 enzyme, while nonsecretors lack this enzyme and are highly resistant to infection and gastroenteritis caused by many HuNoV strains. These epidemiologic associations are confirmed by infections in stem cell-derived human intestinal enteroid (HIE) cultures. GII.4 HuNoV does not replicate in HIE cultures derived from nonsecretor individuals, while HIEs from secretors are permissive to infection. However, whether FUT2 expression alone is critical for infection remains unproven, since routinely used secretor-positive transformed cell lines are resistant to HuNoV replication. To evaluate the role of FUT2 in HuNoV replication, we used CRISPR or overexpression to genetically manipulate FUT2 gene function to produce isogenic HIE lines with or without FUT2 expression. We show that FUT2 expression alone affects both HuNoV binding to the HIE cell surface and susceptibility to HuNoV infection. These findings indicate that initial binding to a molecule(s) glycosylated by FUT2 is critical for HuNoV infection and that the HuNoV receptor is present in nonsecretor HIEs. In addition to HuNoV studies, these isogenic HIE lines will be useful tools to study other enteric microbes where infection and/or disease outcome is associated with secretor status.IMPORTANCE Several studies have demonstrated that secretor status is associated with susceptibility to human norovirus (HuNoV) infection; however, previous reports found that FUT2 expression is not sufficient to allow infection with HuNoV in a variety of continuous laboratory cell lines. Which cellular factor(s) regulates susceptibility to HuNoV infection remains unknown. We used genetic manipulation of HIE cultures to show that secretor status determined by FUT2 gene expression is necessary and sufficient to support HuNoV replication based on analyses of isogenic lines that lack or express FUT2. Fucosylation of HBGAs is critical for initial binding and for modification of another putative receptor(s) in HIEs needed for virus uptake or uncoating and necessary for successful infection by GI.1 and several GII HuNoV strains.


Subject(s)
Blood Group Antigens/metabolism , Caliciviridae Infections/genetics , Fucosyltransferases/genetics , Fucosyltransferases/metabolism , Intestine, Small/enzymology , Organoids/virology , Genetic Predisposition to Disease , Humans , Intestine, Small/cytology , Intestine, Small/virology , Norovirus/pathogenicity , Organoids/enzymology , Virus Replication , Galactoside 2-alpha-L-fucosyltransferase
9.
Proc Natl Acad Sci U S A ; 117(3): 1700-1710, 2020 01 21.
Article in English | MEDLINE | ID: mdl-31896578

ABSTRACT

Human noroviruses (HuNoVs) cause sporadic and epidemic outbreaks of gastroenteritis in all age groups worldwide. We previously reported that stem cell-derived human intestinal enteroid (HIE) cultures support replication of multiple HuNoV strains and that some strains (e.g., GII.3) replicate only in the presence of bile. Heat- and trypsin-treatment of bile did not reduce GII.3 replication, indicating a nonproteinaceous component in bile functions as an active factor. Here we show that bile acids (BAs) are critical for GII.3 replication and replication correlates with BA hydrophobicity. Using the highly effective BA, glycochenodeoxycholic acid (GCDCA), we show BAs act during the early stage of infection, BA-dependent replication in HIEs is not mediated by detergent effects or classic farnesoid X receptor or Takeda G protein-coupled receptor 5 signaling but involves another G protein-coupled receptor, sphingosine-1-phosphate receptor 2, and BA treatment of HIEs increases particle uptake. We also demonstrate that GCDCA induces multiple cellular responses that promote GII.3 replication in HIEs, including enhancement of 1) endosomal uptake, 2) endosomal acidification and subsequent activity of endosomal/lysosomal enzyme acid sphingomyelinase (ASM), and 3) ceramide levels on the apical membrane. Inhibitors of endosomal acidification or ASM reduce GII.3 infection and exogenous addition of ceramide alone permits infection. Furthermore, inhibition of lysosomal exocytosis of ASM, which is required for ceramide production at the apical surface, decreases GII.3 infection. Together, our results support a model where GII.3 exploits rapid BA-mediated cellular endolysosomal dynamic changes and cellular ceramide to enter and replicate in jejunal HIEs.


Subject(s)
Bile Acids and Salts/metabolism , Ceramides/metabolism , Intestines/virology , Norovirus/drug effects , Virus Internalization/drug effects , Virus Replication/drug effects , Bile Acids and Salts/pharmacology , Ceramides/pharmacology , Glycochenodeoxycholic Acid , Humans , Receptors, G-Protein-Coupled , Sphingomyelin Phosphodiesterase/metabolism , Sphingosine-1-Phosphate Receptors
10.
J Infect Dis ; 221(5): 739-743, 2020 02 18.
Article in English | MEDLINE | ID: mdl-31613328

ABSTRACT

BACKGROUND: The development of an in vitro cultivation system for human noroviruses allows the measurement of neutralizing antibody levels. METHODS: Serum neutralizing antibody levels were determined using a GII.4/Sydney/2012-like virus in human intestinal enteroids in samples collected before and 4 weeks after administration of an investigational norovirus vaccine and were compared with those measured in histo-blood group antigen (HBGA)-blocking assays. RESULTS: Neutralizing antibody seroresponses were observed in 71% of 24 vaccinated adults, and antibody levels were highly correlated (r = 0.82, P < .001) with those measured by HBGA blocking. CONCLUSIONS: HBGA-blocking antibodies are a surrogate for neutralization in human noroviruses. CLINICAL TRIALS REGISTRATION: NCT02475278.


Subject(s)
Antibodies, Blocking/immunology , Antibodies, Neutralizing/immunology , Antibodies, Viral/immunology , Blood Group Antigens/immunology , Caliciviridae Infections/prevention & control , Norovirus/immunology , Vaccination , Viral Vaccines/immunology , Adolescent , Adult , Antibodies, Viral/analysis , Caliciviridae Infections/virology , Female , Healthy Volunteers , Humans , Male , Middle Aged , Neutralization Tests , Young Adult
11.
Viruses ; 11(7)2019 07 11.
Article in English | MEDLINE | ID: mdl-31336765

ABSTRACT

Noroviruses, in the genus Norovirus, are a significant cause of viral gastroenteritis in humans and animals. For almost 50 years, the lack of a cultivation system for human noroviruses (HuNoVs) was a major barrier to understanding virus biology and the development of effective antiviral strategies. This review presents a historical perspective of the development of a cultivation system for HuNoVs in human intestinal epithelial cell cultures. Successful cultivation was based on the discovery of genetically-encoded host factors required for infection, knowledge of the site of infection in humans, and advances in the cultivation of human intestinal epithelial cells achieved by developmental and stem cell biologists. The human stem cell-derived enteroid cultivation system recapitulates the multicellular, physiologically active human intestinal epithelium, and allows studies of virus-specific replication requirements, evaluation of human host-pathogen interactions, and supports the pre-clinical assessment of methods to prevent and treat HuNoV infections.


Subject(s)
Epithelial Cells/virology , Intestinal Mucosa/virology , Norovirus/growth & development , Stem Cells/virology , Virus Cultivation/methods , Caliciviridae Infections/drug therapy , Caliciviridae Infections/prevention & control , Host-Pathogen Interactions , Humans , Norovirus/physiology , Stem Cells/physiology , Virus Replication
12.
Indian J Nucl Med ; 34(2): 96-98, 2019.
Article in English | MEDLINE | ID: mdl-31040519

ABSTRACT

BACKGROUND: Here, we present a retrospective study conducted from 2009 to 2018, which showed the presence of unilateral uptake of radioactive tracer on 99mTc thyroid scintigraphy scan in 15 patients with Graves' disease. MATERIALS AND METHODS: All these patients had either clinical features of Graves' disease or elevated thyroid hormone levels along with ultrasonographic features, showing either normal thyroid gland or diffuse thyroiditis. The scintigraphic features revealed increased uptake in one lobe of the thyroid gland with the other lobe being normal. RESULTS: Of the 15 patients, 13 were females and two were males. The mean age of the patients was 47 years with standard deviation of 3.4 years (range 26-70 years). Eight of the 15 patients had increased uptake on the right lobe and seven had increased uptake in the left lobe. CONCLUSION: This study shows that there exists an entity called unilateral Graves' disease which should be further evaluated.

13.
Appl Biochem Biotechnol ; 185(3): 778-785, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29330770

ABSTRACT

In a recombinant antibody scFv format, the presence of an unpaired cysteine (Cys) is implicated in reduced soluble expression and inefficient presentation in phage display. Compared to other species, antibodies derived from rabbits are more likely to contain this unpaired Cys residue at position 80 (Cys80), when generated in a scFv format. In a screening campaign to isolate rabbit scFv against cardiac troponin I (cTnI), it was found that, a large proportion of isolated cTnI-specific clones contained unpaired Cys80. To analyze the factors that led to the selection of anti-cTnI Cys80 scFv, after five rounds of biopanning, the biopanning experiments were repeated with a Cys80 scFv (MG4Cys), its alanine variant (MG4Ala), and an irrelevant high expressing scFv control. It was found that the selection and subsequent enrichment of MG4Cys scFv was ousted by the superior expressing variant MG4Ala, indicating that the Cys80 scFv was selected primarily due to its affinity. It is evident that phage-based selection is influenced by specific sequence characteristics affecting the expression as well as the binding specificity and this needs to be taken into account for selection of optimal antibody derivatives.


Subject(s)
Antibody Affinity , Cysteine/metabolism , Single-Chain Antibodies/genetics , Alanine/metabolism , Animals , Antibody Specificity , Bacteriophage M13/genetics , Enzyme-Linked Immunosorbent Assay , Myocardium , Peptide Library , Peroxidase/immunology , Rabbits , Single-Chain Antibodies/immunology , Troponin I/immunology , Troponin I/metabolism
14.
Appl Microbiol Biotechnol ; 101(22): 8165-8179, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29034433

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease caused by sensitization of the immune system to self-antigens. We have previously shown that targeting MG-susceptible alleles can significantly inhibit proliferation of disease-specific T cells. In this work, we humanized a murine monoclonal antibody (mAb) LG11, capable of blocking MG-associated DQ beta 1 (DQB1) allele and reformatted it into single-chain fragment variable (scFv). A fully functional humanized scFv was obtained by optimizing variable domain orientations and linker lengths, along with the optimization of expression conditions and codons to suit Escherichia coli expression machinery. Characterization of humanized scFv (FL8) revealed that the reformatted scFv, despite recognizing the same epitope as the parent murine LG11 mAb, exhibited superior binding affinity (0.97 nM) compared to the LG11 mAb, towards the immunizing antigen (DQB1*0601/70-90) and was able to block the proliferation of T cells cultured from PBLs of MG-patients typed DQB1*0601. The scFv was also capable of binding a variant MG-associated allele (DQB1*0502/70-90) with moderate affinity (18.7 nM), a feature that was absent in the LG11. To our knowledge, this is the first report of humanizing a MG-associated human leukocyte antigen (HLA) scFv for preclinical studies.


Subject(s)
Alleles , Antibodies, Monoclonal, Humanized/immunology , HLA Antigens/genetics , Myasthenia Gravis/immunology , Single-Chain Antibodies/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/genetics , Antibodies, Monoclonal, Humanized/isolation & purification , Antibodies, Monoclonal, Humanized/therapeutic use , Escherichia coli/genetics , HLA Antigens/immunology , Humans , Mice , Myasthenia Gravis/drug therapy , Myasthenia Gravis/genetics , Single-Chain Antibodies/genetics , Single-Chain Antibodies/therapeutic use , Young Adult
15.
Indian J Nucl Med ; 32(2): 133-136, 2017.
Article in English | MEDLINE | ID: mdl-28533644

ABSTRACT

Brown tumors seen in hyperparathyroidism are rare, non-neoplastic lesions because of abnormal bone metabolism, and they can mimic benign bone tumors or malignancy. Although biopsy is considered as the gold standard for diagnosis, it can be inconclusive. As the diagnosis of brown tumors is often challenging, a high index of suspicion is essential for diagnosis. We present a case of 21-year-old woman who presented with multiple painful bony lesions, which were initially misdiagnosed as fibrous dysplasia. Due to persistent bone pain and deterioration in her physical mobility, she was referred to tertiary care centre. After thorough clinical workup, she underwent Tc-99m methylene diphosphonate bone scintigraphy that raised strong clinical suspicion of hyperparathyroidism and brown tumors. Subsequently, Tc-99m-methoxy isobutyl isonitrile (MIBI) parathyroid scintigraphy revealed a solitary MIBI avid focal lesion, suggestive of left inferior parathyroid adenoma. Later parathyroidectomy was performed and histopathological examination confirmed it as atypical parathyroid adenoma.

16.
Methods ; 116: 51-62, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28163103

ABSTRACT

Antibodies are extensively utilized entities in biomedical research, and in the development of diagnostics and therapeutics. Many of these applications require high amounts of antibodies. However, meeting this ever-increasing demand of antibodies in the global market is one of the outstanding challenges. The need to maintain a balance between demand and supply of antibodies has led the researchers to discover better means and methods for optimizing their expression. These strategies aim to increase the volumetric productivity of the antibodies along with the reduction of associated manufacturing costs. Recent years have witnessed major advances in recombinant protein technology, owing to the introduction of novel cloning strategies, gene manipulation techniques, and an array of cell and vector engineering techniques, together with the progress in fermentation technologies. These innovations were also highly beneficial for antibody expression. Antibody expression depends upon the complex interplay of multiple factors that may require fine tuning at diverse levels to achieve maximum yields. However, each antibody is unique and requires individual consideration and customization for optimizing the associated expression parameters. This review provides a comprehensive overview of several state-of-the-art approaches, such as host selection, strain engineering, codon optimization, gene optimization, vector modification and process optimization that are deemed suitable for enhancing antibody expression.


Subject(s)
Antibodies/genetics , Escherichia coli/genetics , Genetic Engineering/methods , Genetic Vectors/chemistry , Pichia/genetics , Animals , Antibodies/isolation & purification , CHO Cells , Codon/chemistry , Codon/metabolism , Cricetulus , Gene Expression , Genetic Vectors/metabolism , HEK293 Cells , Humans , Plants, Genetically Modified/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Spodoptera
17.
Methods ; 116: 84-94, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28012937

ABSTRACT

Antibodies continue to be extremely utilized entities in myriad applications including basic research, imaging, targeted delivery, chromatography, diagnostics, and therapeutics. At production stage, antibodies are generally present in complex matrices and most of their intended applications necessitate purification. Antibody purification has always been a major bottleneck in downstream processing of antibodies, due to the need of high quality products and associated high costs. Over the years, extensive research has focused on finding better purification methodologies to overcome this holdup. Among a plethora of different techniques, affinity chromatography is one of the most selective, rapid and easy method for antibody purification. This review aims to provide a detailed overview on affinity chromatography and the components involved in purification. An array of support matrices along with various classes of affinity ligands detailing their underlying working principles, together with the advantages and limitations of each system in purifying different types of antibodies, accompanying recent developments and important practical methodological considerations to optimize purification procedure are discussed.


Subject(s)
Antibodies/isolation & purification , Chromatography, Affinity/methods , Animals , Antibodies/chemistry , Chromatography, Affinity/instrumentation , Humans , Hydrophobic and Hydrophilic Interactions , Ligands , Protein Binding , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Static Electricity
18.
Trends Pharmacol Sci ; 37(12): 1009-1028, 2016 12.
Article in English | MEDLINE | ID: mdl-27745709

ABSTRACT

Antibody-based therapies have garnered considerable success in recent years. This is due to the availability of strategies to successfully engineer antibodies into humanized forms, better understanding of the biological processes involved in cancer development, the availability of novel recombinant antibody formats, better antibody selection platforms, and improved antibody conjugation methodologies. Such achievements have led to an explosion in the generation of antibodies and antibody-associated constructs for the treatment of cancer and other diseases. In this review, we critically assess recent trends in the development and applications of bispecific antibodies (bsAbs), antibody-drug conjugates (ADCs), and immune checkpoint inhibitors (ICIs) as cancer therapeutics. We also highlight recent US FDA approvals and clinical trials of antibody-based cancer therapies.


Subject(s)
Antibodies/administration & dosage , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Animals , Antibodies/immunology , Antibodies, Bispecific/administration & dosage , Antibodies, Bispecific/immunology , Antineoplastic Agents/immunology , Drug Approval , Drug Design , Humans , Immunoconjugates/administration & dosage , Immunoconjugates/immunology , Neoplasms/immunology , Neoplasms/pathology
19.
Biochim Biophys Acta ; 1864(12): 1678-1685, 2016 12.
Article in English | MEDLINE | ID: mdl-27596061

ABSTRACT

Binding behaviors of the HN and the HC domains of BoNT/A were investigated individually to identify if there exist any differences in their interaction with the cell membrane. Recombinant fragments corresponding to both BoNT/A HN and HC regions were prepared (HN519-845 and HC967-1296) and their binding to synaptic proteins was verified. The binding behaviors of these heavy-chain domains were analyzed by treating the Neuro 2a, a murine neuroblastoma cell line, with compounds known to alter membrane properties. Cholesterol depletion and lipid raft inhibition increased the binding of HN519-845 to Neuro 2a cells without affecting HC967-1296-cell interaction. Sphingolipid depletion decreased the binding of cells to both HC967-1296 and HN519-845 whereas, loading exogenous GD1a, on to the Neuro 2a cells, increased the binding of both the peptides to cells. Microtubule disruption of the Neuro 2a cells by nocodazole decreased the binding of both HC967-1296 and HN519-845 to the treated cells. Inhibition of the clathrin-mediated endocytosis using dynasore, chlorpromazine or potassium (K+) depletion buffer lowered the binding of both HC967-1296 and HN519-845 to the cells, but seemed to exert a more pronounced effect on the binding of HC967-1296 than on the binding of HN519-845. Results indicate that while both the HN and HC domains are involved in the binding of the toxin to neuronal cells there are differences in their behavior which probably stem from their respective amino acid composition and structural location in the toxin three-dimensional structure along with their intended role in translocation and internalization into the cells.


Subject(s)
Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/metabolism , Animals , Botulinum Toxins, Type A/genetics , Caveolae/metabolism , Cell Line , Cell Membrane/metabolism , Clathrin/metabolism , Endocytosis , Membrane Microdomains/metabolism , Mice , Microtubules/metabolism , Models, Molecular , Neurons/metabolism , Neurotoxins/chemistry , Neurotoxins/genetics , Neurotoxins/metabolism , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Protein Domains , Synaptic Vesicles/metabolism
20.
Neurol India ; 64(4): 677-83, 2016.
Article in English | MEDLINE | ID: mdl-27381113

ABSTRACT

BACKGROUND: Lumbar spinal stenosis is a frequent indication for spinal surgery. The clinical symptoms may not be accurately reflected on radiological studies. Treatment is aimed at not only obtaining immediate pain relief but also in preventing its long-term disabling sequelae. The walking ability needs to be correlated with functional outcome measures in assessing patients with symptomatic lumbar canal stenosis (LCS). AIMS AND OBJECTIVES: To study and evaluate the outcome of surgery for degenerative LCS on a clinical, radiological, and functional basis, and to establish an association between various factors that may be influencing the development of LCS. MATERIALS AND METHODS: A prospective study of 48 patients was carried out at the Department of Neurosurgery, Sri Venkateswara Institute of Medical Sciences, Tirupathi, in whom the LCS was confirmed and measured on magnetic resonance imaging images by thecal sac cross-sectional area (CSA). Their functional assessment was done by motorized treadmill test for assessment of the time of appearance of first symptom and the maximum walking distance. The visual analog scale (VAS), Oswestry disability index (ODI), Japanese Orthopedic Association (JOA) score, and Short Form-36 (SF-36) score were also evaluated. CONCLUSIONS: The post-operative VAS, ODI, JOA, SF-36 scores showed a significant improvement from the preoperative ones. Postoperative thecal sac measurements showed a significant neural decompression substantiating the results of improvement in the functional assessment scores after surgery. The treadmill test is a quantifiable means of dynamic function. It may be considered as a useful tool for the assessment of functional impairment and for the correlation of thecal sac CSA with lumbar canal stenosis. A combination of functional scores, treadmill test, and thecal sac CSA may be included in the preoperative assessment of outcome in patients with LCS.


Subject(s)
Decompression, Surgical/methods , Spinal Stenosis/surgery , Humans , Lumbar Vertebrae/surgery , Pain Measurement , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL
...