3,320 results on '"Varki A"'
Search Results
2. Chemoenzymatic synthesis of N-acetyl analogues of 9-O-acetylated b-series gangliosides
- Author
-
Yu, Hai, Zheng, Zimin, Zhang, Libo, Yang, Xiaohong, Varki, Ajit, and Chen, Xi
- Subjects
Medicinal and Biomolecular Chemistry ,Organic Chemistry ,Chemical Sciences ,b-Series ganglioside ,Chemoenzymatic synthesis ,Glycosphingolipid ,9-N-acetyl sialic acid ,O-acetyl sialic acid ,One-pot multienzyme ,b-series ganglioside ,chemoenzymatic synthesis ,glycosphingolipid ,one-pot multienzyme ,Biochemistry and cell biology ,Medicinal and biomolecular chemistry ,Organic chemistry - Abstract
The stable N-acetyl analogues of biologically important 9-O-acetylated b-series gangliosides including 9NAc-GD3, 9NAc-GD2, 9NAc-GD1b, and 9NAc-GT1b were chemoenzymatically synthesized from a GM3 sphingosine. Two chemoenzymatic methods using either 6-azido-6-deoxy-N-acetylmannosamine (ManNAc6N3) as a chemoenzymatic synthon or 6-acetamido-6-deoxy-N-acetylmannosamine (ManNAc6NAc) as an enzymatic precursor for 9-acetamido-9-deoxy-N-acetylneuraminic acid (Neu5Ac9NAc) were developed and compared for the synthesis of 9NAc-GD3. The latter method was found to be more efficient and was used to produce the desired 9-N-acetylated glycosylsphingosines. Furthermore, glycosylsphingosine acylation reaction conditions were improved to obtain target 9-N-acetylated gangliosides in a faster reaction with an easier purification process compared to the previous acylation conditions.
- Published
- 2023
3. Re²Pair: Increasing the Scalability of RePair by Decreasing Memory Usage.
- Author
-
Justin Kim, Rahul Varki, Marco Oliva, and Christina Boucher 0001
- Published
- 2024
- Full Text
- View/download PDF
4. Cataloging natural sialic acids and other nonulosonic acids (NulOs), and their representation using the Symbol Nomenclature for Glycans.
- Author
-
Frank, Martin, Lütteke, Thomas, Knirel, Yuriy, Schoenhofen, Ian, Varki, Ajit, Vinogradov, Evgeny, Woods, Robert, Zachara, Natasha, Zhang, Jian, Kamerling, Johannis, Neelamegham, Sriram, Toukach, Philip, Bolton, Evan, Chen, Xi, and Lewis, Amanda
- Subjects
NulO ,glycans ,nonulosonic acid ,sialic acid ,symbol nomenclature ,Animals ,Sialic Acids ,N-Acetylneuraminic Acid ,Polysaccharides ,Monosaccharides ,Cataloging - Abstract
Nonulosonic acids or non-2-ulosonic acids (NulOs) are an ancient family of 2-ketoaldonic acids (α-ketoaldonic acids) with a 9-carbon backbone. In nature, these monosaccharides occur either in a 3-deoxy form (referred to as sialic acids) or in a 3,9-dideoxy sialic-acid-like form. The former sialic acids are most common in the deuterostome lineage, including vertebrates, and mimicked by some of their pathogens. The latter sialic-acid-like molecules are found in bacteria and archaea. NulOs are often prominently positioned at the outermost tips of cell surface glycans, and have many key roles in evolution, biology and disease. The diversity of stereochemistry and structural modifications among the NulOs contributes to more than 90 sialic acid forms and 50 sialic-acid-like variants described thus far in nature. This paper reports the curation of these diverse naturally occurring NulOs at the NCBI sialic acid page (https://www.ncbi.nlm.nih.gov/glycans/sialic.html) as part of the NCBI-Glycans initiative. This includes external links to relevant Carbohydrate Structure Databases. As the amino and hydroxyl groups of these monosaccharides are extensively derivatized by various substituents in nature, the Symbol Nomenclature For Glycans (SNFG) rules have been expanded to represent this natural diversity. These developments help illustrate the natural diversity of sialic acids and related NulOs, and enable their systematic representation in publications and online resources.
- Published
- 2023
5. Simple and practical sialoglycan encoding system reveals vast diversity in nature and identifies a universal sialoglycan-recognizing probe derived from AB5 toxin B subunits.
- Author
-
Sasmal, Aniruddha, Khan, Naazneen, Khedri, Zahra, Kellman, Benjamin, Srivastava, Saurabh, Verhagen, Andrea, Bruntse, Anders, Diaz, Sandra, Varki, Nissi, Beddoe, Travis, Paton, Adrienne, Paton, James, Lewis, Nathan, Varki, Ajit, Chen, Xi, and Yu, Hai
- Subjects
Yersinia enterocolitica ,bacterial toxin ,glycan coding ,glycan microarray ,toxin B subunit ,Escherichia coli ,Salmonella typhi ,Sialic Acids ,Bacterial Toxins ,Polysaccharides ,Cholera Toxin - Abstract
Vertebrate sialic acids (Sias) display much diversity in modifications, linkages, and underlying glycans. Slide microarrays allow high-throughput explorations of sialoglycan-protein interactions. A microarray presenting ~150 structurally defined sialyltrisaccharides with various Sias linkages and modifications still poses challenges in planning, data sorting, visualization, and analysis. To address these issues, we devised a simple 9-digit code for sialyltrisaccharides with terminal Sias and underlying two monosaccharides assigned from the nonreducing end, with 3 digits assigning a monosaccharide, its modifications, and linkage. Calculations based on the encoding system reveal >113,000 likely linear sialyltrisaccharides in nature. Notably, a biantennary N-glycan with 2 terminal sialyltrisaccharides could thus have >1010 potential combinations and a triantennary N-glycan with 3 terminal sequences, >1015 potential combinations. While all possibilities likely do not exist in nature, sialoglycans encode enormous diversity. While glycomic approaches are used to probe such diverse sialomes, naturally occurring bacterial AB5 toxin B subunits are simpler tools to track the dynamic sialome in biological systems. Sialoglycan microarray was utilized to compare sialoglycan-recognizing bacterial toxin B subunits. Unlike the poor correlation between B subunits and species phylogeny, there is stronger correlation with Sia-epitope preferences. Further supporting this pattern, we report a B subunit (YenB) from Yersinia enterocolitica (broad host range) recognizing almost all sialoglycans in the microarray, including 4-O-acetylated-Sias not recognized by a Yersinia pestis orthologue (YpeB). Differential Sia-binding patterns were also observed with phylogenetically related B subunits from Escherichia coli (SubB), Salmonella Typhi (PltB), Salmonella Typhimurium (ArtB), extra-intestinal E.coli (EcPltB), Vibrio cholera (CtxB), and cholera family homologue of E. coli (EcxB).
- Published
- 2022
6. P-selectin, carcinoma metastasis and heparin: novel mechanistic connections with therapeutic implications
- Author
-
Varki A. and Varki N.M.
- Subjects
heparin ,carcinomas ,P-selectin ,mucin ,metastasis ,Medicine (General) ,R5-920 ,Biology (General) ,QH301-705.5 - Abstract
Metastasis is a multistep cascade initiated when malignant cells penetrate the tissue surrounding the primary tumor and enter the bloodstream. Classic studies indicated that blood platelets form complexes around tumor cells in the circulation and facilitate metastases. In other work, the anticoagulant drug heparin diminished metastasis in murine models, as well is in preliminary human studies. However, attempts to follow up the latter observation using vitamin K antagonists failed, indicating that the primary mechanism of heparin action was unrelated to its anticoagulant properties. Other studies showed that the overexpression of sialylated fucosylated glycans in human carcinomas is associated with a poor prognosis. We have now brought all these observations together into one mechanistic explanation, which has therapeutic implications. Carcinoma cells expressing sialylated fucosylated mucins can interact with platelets, leukocytes and endothelium via the selectin family of cell adhesion molecules. The initial organ colonization of intravenously injected carcinoma cells is attenuated in P-selectin-deficient mice, in mice receiving tumor cells pretreated with O-sialoglycoprotease (to selectively remove mucins from cell surfaces), or in mice receiving a single dose of heparin prior to tumor cell injection. In each case, we found that formation of a platelet coating on cancer cells was impeded, allowing increased access of leukocytes to the tumor cells. Several weeks later, all animals showed a decrease in the extent of established metastasis, indicating a long-lasting effect of the short-term intervention. The absence of obvious synergism amongst the three treatments suggests that they all act via a common pathway. Thus, a major mechanism of heparin action in cancer may be inhibition of P-selectin-mediated platelet coating of tumor cells during the initial phase of the metastatic process. We therefore suggest that heparin use in cancer be re-explored, specifically during the time interval between initial visualization of a primary tumor until just after definitive surgical removal.
- Published
- 2001
7. Long-term exposure to house dust mites accelerates lung cancer development in mice
- Author
-
Wang, Dongjie, Li, Wen, Albasha, Natalie, Griffin, Lindsey, Chang, Han, Amaya, Lauren, Ganguly, Sneha, Zeng, Liping, Keum, Bora, González-Navajas, José M, Levin, Matt, AkhavanAghdam, Zohreh, Snyder, Helen, Schwartz, David, Tao, Ailin, Boosherhri, Laela M, Hoffman, Hal M, Rose, Michael, Estrada, Monica Valeria, Varki, Nissi, Herdman, Scott, Corr, Maripat, Webster, Nicholas JG, Raz, Eyal, and Bertin, Samuel
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Cancer ,Lung ,Health Effects of Indoor Air Pollution ,Lung Cancer ,Aetiology ,2.1 Biological and endogenous factors ,Respiratory ,Good Health and Well Being ,Humans ,Animals ,Mice ,NLR Family ,Pyrin Domain-Containing 3 Protein ,Pyroglyphidae ,Asthma ,Lung Neoplasms ,Disease Models ,Animal ,Tumor Microenvironment ,Lung cancer ,Kras ,Urethane ,House dust mites ,Chronic inflammation ,NLRP3 ,IL-1 beta ,CCL2 ,Macrophages ,Tumor microenvironment ,IL-1β ,Oncology and carcinogenesis - Abstract
BackgroundIndividuals with certain chronic inflammatory lung diseases have a higher risk of developing lung cancer (LC). However, the underlying mechanisms remain largely unknown. Here, we hypothesized that chronic exposure to house dust mites (HDM), a common indoor aeroallergen associated with the development of asthma, accelerates LC development through the induction of chronic lung inflammation (CLI). METHODS: The effects of HDM and heat-inactivated HDM (HI-HDM) extracts were evaluated in two preclinical mouse models of LC (a chemically-induced model using the carcinogen urethane and a genetically-driven model with oncogenic KrasG12D activation in lung epithelial cells) and on murine macrophages in vitro. Pharmacological blockade or genetic deletion of the Nod-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome, caspase-1, interleukin-1β (IL-1β), and C-C motif chemokine ligand 2 (CCL2) or treatment with an inhaled corticosteroid (ICS) was used to uncover the pro-tumorigenic effect of HDM. RESULTS: Chronic intranasal (i.n) instillation of HDM accelerated LC development in the two mouse models. Mechanistically, HDM caused a particular subtype of CLI, in which the NLRP3/IL-1β signaling pathway is chronically activated in macrophages, and made the lung microenvironment conducive to tumor development. The tumor-promoting effect of HDM was significantly decreased by heat treatment of the HDM extract and was inhibited by NLRP3, IL-1β, and CCL2 neutralization, or ICS treatment.ConclusionsCollectively, these data indicate that long-term exposure to HDM can accelerate lung tumorigenesis in susceptible hosts (e.g., mice and potentially humans exposed to lung carcinogens or genetically predisposed to develop LC).
- Published
- 2023
8. Sialoglycan-binding patterns of bacterial AB5 toxin B subunits correlate with host range and toxicity, indicating evolution independent of A subunits.
- Author
-
Khan, Naazneen, Sasmal, Aniruddha, Khedri, Zahra, Secrest, Patrick, Verhagen, Andrea, Srivastava, Saurabh, Varki, Nissi, Chen, Xi, Yu, Hai, Beddoe, Travis, Paton, Adrienne W, Paton, James C, and Varki, Ajit
- Subjects
Animals ,Mammals ,Bacteria ,Yersinia pestis ,Plague ,N-Acetylneuraminic Acid ,Polysaccharides ,Protein Subunits ,Bacterial Toxins ,Evolution ,Molecular ,Phylogeny ,Protein Binding ,Host Specificity ,bacterial ,cytotoxicity ,evolution ,host range ,pathogenesis ,phylogenetic ,sialoglycan microarray ,toxin ,Vector-Borne Diseases ,Biodefense ,Prevention ,Emerging Infectious Diseases ,Infectious Diseases ,Vaccine Related ,2.2 Factors relating to the physical environment ,Aetiology ,Infection ,Chemical Sciences ,Biological Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology - Abstract
Many pathogenic bacteria secrete AB5 toxins that can be virulence factors. Cytotoxic A subunits are delivered to the cytosol following B subunit binding to specific host cell surface glycans. Some B subunits are not associated with A subunits, for example, YpeB of Yersinia pestis, the etiologic agent of plague. Plague cannot be eradicated because of Y. pestis' adaptability to numerous hosts. We previously showed selective binding of other B5 pentamers to a sialoglycan microarray, with sialic acid (Sia) preferences corresponding to those prominently expressed by various hosts, for example, N-acetylneuraminic acid (Neu5Ac; prominent in humans) or N-glycolylneuraminic acid (Neu5Gc; prominent in ruminant mammals and rodents). Here, we report that A subunit phylogeny evolved independently of B subunits and suggest a future B subunit nomenclature based on bacterial species names. We also found via phylogenetic analysis of B subunits, which bind Sias, that homologous molecules show poor correlation with species phylogeny. These data indicate ongoing lateral gene transfers between species, including mixing of A and B subunits. Consistent with much broader host range of Y. pestis, we show that YpeB recognizes all mammalian Sia types, except for 4-O-acetylated ones. Notably, YpeB alone causes dose-dependent cytotoxicity, which is abolished by a mutation (Y77F) eliminating Sia recognition, suggesting that cell proliferation and death are promoted via lectin-like crosslinking of cell surface sialoglycoconjugates. These findings help explain the host range of Y. pestis and could be important for pathogenesis. Overall, our data indicate ongoing rapid evolution of both host Sias and pathogen toxin-binding properties.
- Published
- 2022
9. O‑Acetyl Migration within the Sialic Acid Side Chain: A Mechanistic Study Using the Ab Initio Nanoreactor
- Author
-
Oh, Lisa, Ji, Yang, Li, Wanqing, Varki, Ajit, Chen, Xi, and Wang, Lee-Ping
- Subjects
Biological Sciences ,Organic Chemistry ,Chemical Sciences ,Acetylation ,Esters ,Glycerol ,N-Acetylneuraminic Acid ,Nanotechnology ,Sialic Acids ,Medicinal and Biomolecular Chemistry ,Biochemistry and Cell Biology ,Medical Biochemistry and Metabolomics ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical biochemistry and metabolomics ,Medicinal and biomolecular chemistry - Abstract
Many disease-causing viruses target sialic acids on the surface of host cells. Some viruses bind preferentially to sialic acids with O-acetyl modification at the hydroxyl group of C7, C8, or C9 on the glycerol-like side chain. Studies of proteins binding to sialosides containing O-acetylated sialic acids are crucial in understanding the related diseases but experimentally difficult due to the lability of the ester group. We recently showed that O-acetyl migration among hydroxyl groups of C7, C8, and C9 in sialic acids occurs in all directions in a pH-dependent manner. In the current study, we elucidate a full mechanistic pathway for the migration of O-acetyl among C7, C8, and C9. We used an ab initio nanoreactor to explore potential reaction pathways and density functional theory, pKa calculations, and umbrella sampling to investigate elementary steps of interest. We found that when a base is present, migration is easy in any direction and involves three key steps: deprotonation of the hydroxyl group, cyclization between the two carbons, and the migration of the O-acetyl group. This dynamic equilibrium may play a defensive role against pathogens that evolve to gain entry to the cell by binding selectively to one acetylation state.
- Published
- 2022
10. Evolution of Human-Specific Alleles Protecting Cognitive Function of Grandmothers
- Author
-
Saha, Sudeshna, Khan, Naazneen, Comi, Troy, Verhagen, Andrea, Sasmal, Aniruddha, Diaz, Sandra, Yu, Hai, Chen, Xi, Akey, Joshua M, Frank, Martin, Gagneux, Pascal, and Varki, Ajit
- Subjects
Genetics ,Human Genome ,Clinical Research ,2.1 Biological and endogenous factors ,Aetiology ,Alleles ,Amino Acids ,Animals ,Cognition ,Grandparents ,Hominidae ,Humans ,CD33 ,pathogen ,sialic acids ,archaic genome ,molecular dynamics simulation ,phylogenetic analysis ,Biochemistry and Cell Biology ,Evolutionary Biology - Abstract
The myelomonocytic receptor CD33 (Siglec-3) inhibits innate immune reactivity by extracellular V-set domain recognition of sialic acid (Sia)-containing "self-associated molecular patterns" (SAMPs). We earlier showed that V-set domain-deficient CD33-variant allele, protective against late-onset Alzheimer's Disease (LOAD), is derived and specific to the hominin lineage. We now report multiple hominin-specific CD33 V-set domain mutations. Due to hominin-specific, fixed loss-of-function mutation in the CMAH gene, humans lack N-glycolylneuraminic acid (Neu5Gc), the preferred Sia-ligand of ancestral CD33. Mutational analysis and molecular dynamics (MD)-simulations indicate that fixed change in amino acid 21 of hominin V-set domain and conformational changes related to His45 corrected for Neu5Gc-loss by switching to N-acetylneuraminic acid (Neu5Ac)-recognition. We show that human-specific pathogens Neisseria gonorrhoeae and Group B Streptococcus selectively bind human CD33 (huCD33) as part of immune-evasive molecular mimicry of host SAMPs and that this binding is significantly impacted by amino acid 21 modification. In addition to LOAD-protective CD33 alleles, humans harbor derived, population-universal, cognition-protective variants at several other loci. Interestingly, 11 of 13 SNPs in these human genes (including CD33) are not shared by genomes of archaic hominins: Neanderthals and Denisovans. We present a plausible evolutionary scenario to compile, correlate, and comprehend existing knowledge about huCD33-evolution and suggest that grandmothering emerged in humans.
- Published
- 2022
11. Dietary Neu5Ac Intervention Protects Against Atherosclerosis Associated With Human-Like Neu5Gc Loss—Brief Report
- Author
-
Kawanishi, Kunio, Coker, Joanna K, Grunddal, Kaare V, Dhar, Chirag, Hsiao, Jason, Zengler, Karsten, Varki, Nissi, Varki, Ajit, and Gordts, Philip LSM
- Subjects
Clinical Research ,Prevention ,Cardiovascular ,Atherosclerosis ,Aging ,Nutrition ,Animal Feed ,Animals ,Antibodies ,Aorta ,Aortic Diseases ,Diet ,High-Fat ,Dietary Supplements ,Disease Models ,Animal ,Foam Cells ,Humans ,Male ,Mice ,Inbred C57BL ,Mice ,Knockout ,Mixed Function Oxygenases ,N-Acetylneuraminic Acid ,Neuraminic Acids ,Pan troglodytes ,Plaque ,Atherosclerotic ,Receptors ,LDL ,Sialadenitis ,THP-1 Cells ,atherosclerosis ,inflammation ,mice ,mucins ,N-acetylneuraminic acid ,Cardiorespiratory Medicine and Haematology ,Clinical Sciences ,Cardiovascular System & Hematology - Abstract
ObjectiveSpecies-specific pseudogenization of the CMAH gene during human evolution eliminated common mammalian sialic acid N-glycolylneuraminic acid (Neu5Gc) biosynthesis from its precursor N-acetylneuraminic acid (Neu5Ac). With metabolic nonhuman Neu5Gc incorporation into endothelia from red meat, the major dietary source, anti-Neu5Gc antibodies appeared. Human-like Ldlr-/-Cmah-/- mice on a high-fat diet supplemented with a Neu5Gc-enriched mucin, to mimic human red meat consumption, suffered increased atherosclerosis if human-like anti-Neu5Gc antibodies were elicited.Approach and resultsWe now ask whether interventional Neu5Ac feeding attenuates metabolically incorporated Neu5Gc-mediated inflammatory acceleration of atherogenesis in this Cmah-/-Ldlr-/- model system. Switching to a Neu5Gc-free high-fat diet or adding a 5-fold excess of Collocalia mucoid-derived Neu5Ac in high-fat diet protects against accelerated atherosclerosis. Switching completely from a Neu5Gc-rich to a Neu5Ac-rich diet further reduces severity. Remarkably, feeding Neu5Ac-enriched high-fat diet alone has a substantial intrinsic protective effect against atherosclerosis in Ldlr-/- mice even in the absence of dietary Neu5Gc but only in the human-like Cmah-null background.ConclusionsInterventional Neu5Ac feeding can mitigate or prevent the red meat/Neu5Gc-mediated increased risk for atherosclerosis, and has an intrinsic protective effect, even in the absence of Neu5Gc feeding. These findings suggest that similar interventions should be tried in humans and that Neu5Ac-enriched diets alone should also be investigated further.
- Published
- 2021
12. Lp(a), oxidized phospholipids and oxidation-specific epitopes are increased in subjects with keloid formation
- Author
-
Ruder, Sundeep, Mansfield, Brett, Immelman, Andrew Ronald, Varki, Nissi, Miu, Phuong, Raal, Frederick, and Tsimikas, Sotirios
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Cardiovascular ,Clinical Research ,Atherosclerosis ,Humans ,Antigen-Antibody Complex ,Phospholipids ,Epitopes ,Case-Control Studies ,Lipoprotein(a) ,Apolipoproteins B ,Apolipoprotein B-100 ,Oxidation-Reduction ,Malondialdehyde ,Immunoglobulin M ,Immunoglobulin G ,Other Information and Computing Sciences ,Medical Biochemistry and Metabolomics ,Nutrition and Dietetics ,Nutrition & Dietetics ,Medical biochemistry and metabolomics ,Nutrition and dietetics - Abstract
BackgroundKeloid formation following trauma or surgery is common among darkly pigmented individuals. Since lipoprotein(a) [Lp(a)] has been postulated to have a putative role in wound healing, and also mediates atherosclerotic cardiovascular disease, it was assessed whether Lp(a), its associated oxidized phospholipids and other oxidation-specific biomarkers were associated with keloid formation.MethodsThis case-control study included darkly pigmented individuals of African ancestry, 100 with keloid scarring and 100 non-keloid controls. The lipid panel, hsCRP, Lp(a), oxidized phospholipids on apolipoprotein B-100 (OxPL-apoB), IgG and IgM apoB-immune complexes and IgG and IgM autoantibodies to a malondialdehyde mimotope (MDA-mimotope) were measured. Immunohistochemistry of keloid specimens was performed for both Lp(a) and OxPL staining.ResultsCases and controls were well matched for age, sex and lipid profile. Mean Lp(a) (57.8 vs. 44.2 mg/dL; P = 0.01, OxPL-apoB 17.4 vs. 15.7 nmol/L; P = 0.009) and IgG and IgM apoB-immune complexes and IgG and IgM MDA-mimotope levels were significantly higher in keloid cases. Keloid tissue stained strongly for OxPL.ConclusionDarkly pigmented individuals of African ancestry with keloids have higher plasma levels of Lp(a), OxPL-apoB and oxidation-specific epitopes. The commonality of excessive wound healing in keloids and chronic complications from coronary revascularization suggests avenues of investigation to define a common mechanism driven by Lp(a) and the innate response to oxidized lipids.
- Published
- 2022
13. SARS-CoV-2 and MERS-CoV Spike Protein Binding Studies Support Stable Mimic of Bound 9-O-Acetylated Sialic Acids
- Author
-
Oh, Lisa, Varki, Ajit, Chen, Xi, and Wang, Lee-Ping
- Subjects
Medicinal and Biomolecular Chemistry ,Organic Chemistry ,Chemical Sciences ,Coronaviruses ,Rare Diseases ,Lung ,Emerging Infectious Diseases ,Infectious Diseases ,Binding Sites ,COVID-19 ,Humans ,Middle East Respiratory Syndrome Coronavirus ,Pandemics ,Protein Binding ,Receptors ,Virus ,SARS-CoV-2 ,Sialic Acids ,Spike Glycoprotein ,Coronavirus ,MERS-CoV ,CoV S protein ,sialic acid ,MM-PBSA ,SOMD ,binding free energy simulations ,molecular dynamics ,Theoretical and Computational Chemistry ,Medicinal and biomolecular chemistry ,Organic chemistry - Abstract
Many disease-causing viruses target sialic acids (Sias), a class of nine-carbon sugars known to coat the surface of many cells, including those in the lungs. Human beta coronaviridae, known for causing respiratory tract diseases, often bind Sias, and some preferentially bind to those with 9-O-Ac-modification. Currently, co-binding of SARS-CoV-2, a beta coronavirus responsible for the COVID-19 pandemic, to human Sias has been reported and its preference towards α2-3-linked Neu5Ac has been shown. Nevertheless, O-acetylated Sias-protein binding studies are difficult to perform, due to the ester lability. We studied the binding free energy differences between Neu5,9Ac2α2-3GalβpNP and its more stable 9-NAc mimic binding to SARS-CoV-2 spike protein using molecular dynamics and alchemical free energy simulations. We identified multiple Sia-binding pockets, including two novel sites, with similar binding affinities to those of MERS-CoV, a known co-binder of sialic acid. In our binding poses, 9-NAc and 9-OAc Sias bind similarly, suggesting an experimentally reasonable mimic to probe viral mechanisms.
- Published
- 2022
14. Human‐specific polymorphic pseudogenization of SIGLEC12 protects against advanced cancer progression
- Author
-
Siddiqui, Shoib S, Vaill, Michael, Do, Raymond, Khan, Naazneen, Verhagen, Andrea L, Zhang, Wu, Lenz, Heinz‐Josef, Johnson‐Pais, Teresa L, Leach, Robin J, Fraser, Gary, Wang, Charles, Feng, Gen‐Sheng, Varki, Nissi, and Varki, Ajit
- Subjects
Biological Sciences ,Genetics ,Biotechnology ,Human Genome ,Urologic Diseases ,Cancer ,2.1 Biological and endogenous factors ,advanced carcinoma ,dot blot ,immunohistochemistry ,pseudogenization ,SIGLEC12 ,Biological sciences - Abstract
Compared with our closest living evolutionary cousins, humans appear unusually prone to develop carcinomas (cancers arising from epithelia). The SIGLEC12 gene, which encodes the Siglec-XII protein expressed on epithelial cells, has several uniquely human features: a fixed homozygous missense mutation inactivating its natural ligand recognition property; a polymorphic frameshift mutation eliminating full-length protein expression in ~60%-70% of worldwide human populations; and, genomic features suggesting a negative selective sweep favoring the pseudogene state. Despite the loss of canonical sialic acid binding, Siglec-XII still recruits Shp2 and accelerates tumor growth in a mouse model. We hypothesized that dysfunctional Siglec-XII facilitates human carcinoma progression, correlating with known tumorigenic signatures of Shp2-dependent cancers. Immunohistochemistry was used to detect Siglec-XII expression on tissue microarrays. PC-3 prostate cancer cells were transfected with Siglec-XII and transcription of genes enriched with Siglec-XII was determined. Genomic SIGLEC12 status was determined for four different cancer cohorts. Finally, a dot blot analysis of human urinary epithelial cells was established to determine the Siglec-XII expressors versus non-expressors. Forced expression in a SIGLEC12 null carcinoma cell line enriched transcription of genes associated with cancer progression. While Siglec-XII was detected as expected in ~30%-40% of normal epithelia, ~80% of advanced carcinomas showed strong expression. Notably, >80% of late-stage colorectal cancers had a functional SIGLEC12 allele, correlating with overall increased mortality. Thus, advanced carcinomas are much more likely to occur in individuals whose genomes have an intact SIGLEC12 gene, likely because the encoded Siglec-XII protein recruits Shp2-related oncogenic pathways. The finding has prognostic, diagnostic, and therapeutic implications.
- Published
- 2021
15. Chemoenzymatic Synthesis of Sialosides Containing 7‑N- or 7,9-Di‑N‑acetyl Sialic Acid as Stable O‑Acetyl Analogues for Probing Sialic Acid-Binding Proteins
- Author
-
Kooner, Anoopjit Singh, Diaz, Sandra, Yu, Hai, Santra, Abhishek, Varki, Ajit, and Chen, Xi
- Subjects
Medicinal and Biomolecular Chemistry ,Organic Chemistry ,Chemical Sciences ,Infectious Diseases ,Carrier Proteins ,Mannose ,N-Acetylneuraminic Acid ,Sialic Acids ,Sialyltransferases ,Medicinal and biomolecular chemistry ,Organic chemistry - Abstract
A novel chemoenzymatic synthon strategy has been developed to construct a comprehensive library of α2-3- and α2-6-linked sialosides containing 7-N- or 7,9-di-N-acetyl sialic acid, the stable analogue of naturally occurring 7-O-acetyl- or 7,9-di-O-acetyl-sialic acid. Diazido and triazido-mannose derivatives that were readily synthesized chemically from inexpensive galactose were shown to be effective chemoenzymatic synthons. Together with bacterial sialoside biosynthetic enzymes with remarkable substrate promiscuity, they were successfully used in one-pot multienzyme (OPME) sialylation systems for highly efficient synthesis of sialosides containing multiple azido groups. Conversion of the azido groups to N-acetyl groups generated the desired sialosides. The hydrophobic and UV-detectable benzyloxycarbonyl (Cbz) group introduced in the synthetic acceptors of sialyltransferases was used as a removable protecting group for the propylamine aglycon of the target sialosides. The resulting N-acetyl sialosides were novel stable probes for sialic acid-binding proteins such as plant lectin MAL II, which bond strongly to sialyl T antigens with or without an N-acetyl at C7 or at both C7 and C9 in the sialic acid.
- Published
- 2021
16. Endothelial Heparan Sulfate Mediates Hepatic Neutrophil Trafficking and Injury during Staphylococcus aureus Sepsis
- Author
-
Golden, Gregory J, Toledo, Alejandro Gómez, Marki, Alex, Sorrentino, James T, Morris, Claire, Riley, Raquel J, Spliid, Charlotte, Chen, Qiongyu, Cornax, Ingrid, Lewis, Nathan E, Varki, Nissi, Le, Dzung, Malmström, Johan, Karlsson, Christofer, Ley, Klaus, Nizet, Victor, and Esko, Jeffrey D
- Subjects
Liver Disease ,Sepsis ,Infectious Diseases ,Hematology ,Digestive Diseases ,Emerging Infectious Diseases ,Clinical Research ,Chronic Liver Disease and Cirrhosis ,2.1 Biological and endogenous factors ,Aetiology ,Cardiovascular ,Inflammatory and immune system ,Animals ,Disease Models ,Animal ,Endothelial Cells ,Female ,Glycocalyx ,Heparitin Sulfate ,Liver ,Lung ,Male ,Mice ,Mice ,Inbred C57BL ,Neutrophil Activation ,Neutrophils ,Staphylococcus aureus ,heparan sulfate ,intravital microscopy ,liver ,neutrophils ,proteomics ,sepsis ,thrombosis ,Microbiology - Abstract
Hepatic failure is an important risk factor for poor outcome in septic patients. Using a chemical tagging workflow and high-resolution mass spectrometry, we demonstrate that rapid proteome remodeling of the vascular surfaces precedes hepatic damage in a murine model of Staphylococcus aureus sepsis. These early changes include vascular deposition of neutrophil-derived proteins, shedding of vascular receptors, and altered levels of heparin/heparan sulfate-binding factors. Modification of endothelial heparan sulfate, a major component of the vascular glycocalyx, diminishes neutrophil trafficking to the liver and reduces hepatic coagulopathy and organ damage during the systemic inflammatory response to infection. Modifying endothelial heparan sulfate likewise reduces neutrophil trafficking in sterile hepatic injury, reflecting a more general role of heparan sulfate contribution to the modulation of leukocyte behavior during inflammation. IMPORTANCE Vascular glycocalyx remodeling is critical to sepsis pathology, but the glycocalyx components that contribute to this process remain poorly characterized. This article shows that during Staphylococcus aureus sepsis, the liver vascular glycocalyx undergoes dramatic changes in protein composition associated with neutrophilic activity and heparin/heparan sulfate binding, all before organ damage is detectable by standard circulating liver damage markers or histology. Targeted manipulation of endothelial heparan sulfate modulates S. aureus sepsis-induced hepatotoxicity by controlling the magnitude of neutrophilic infiltration into the liver in both nonsterile and sterile injury. These data identify an important vascular glycocalyx component that impacts hepatic failure during nonsterile and sterile injury.
- Published
- 2021
17. Reversible O‑Acetyl Migration within the Sialic Acid Side Chain and Its Influence on Protein Recognition
- Author
-
Ji, Yang, Sasmal, Aniruddha, Li, Wanqing, Oh, Lisa, Srivastava, Saurabh, Hargett, Audra A, Wasik, Brian R, Yu, Hai, Diaz, Sandra, Choudhury, Biswa, Parrish, Colin R, Freedberg, Darón I, Wang, Lee-Ping, Varki, Ajit, and Chen, Xi
- Subjects
Biological Sciences ,Organic Chemistry ,Chemical Sciences ,Infectious Diseases ,Biotechnology ,Emerging Infectious Diseases ,Acetylation ,Animals ,Cattle ,Chromatography ,High Pressure Liquid ,Hemagglutinins ,Viral ,Molecular Structure ,Oxidation-Reduction ,Periodic Acid ,Phenylenediamines ,Polysaccharides ,Protein Binding ,Sialic Acids ,Torovirus ,Viral Fusion Proteins ,Biological sciences ,Chemical sciences - Abstract
O-Acetylation is a common naturally occurring modification of carbohydrates and is especially widespread in sialic acids, a family of nine-carbon acidic monosaccharides. O-Acetyl migration within the exocyclic glycerol-like side chain of mono-O-acetylated sialic acid reported previously was from the C7- to C9-hydroxyl group with or without an 8-O-acetyl intermediate, which resulted in an equilibrium that favors the formation of the 9-O-acetyl sialic acid. Herein, we provide direct experimental evidence demonstrating that O-acetyl migration is bidirectional, and the rate of equilibration is influenced predominantly by the pH of the sample. While the O-acetyl group on sialic acids and sialoglycans is stable under mildly acidic conditions (pH < 5, the rate of O-acetyl migration is extremely low), reversible O-acetyl migration is observed readily at neutral pH and becomes more significant when the pH increases to slightly basic. Sialoglycan microarray studies showed that esterase-inactivated porcine torovirus hemagglutinin-esterase bound strongly to sialoglycans containing a more stable 9-N-acetylated sialic acid analog, but these compounds were less resistant to periodate oxidation treatment compared to their 9-O-acetyl counterparts. Together with prior studies, the results support the possible influence of sialic acid O-acetylation and O-acetyl migration to host-microbe interactions and potential application of the more stable synthetic N-acetyl mimics.
- Published
- 2021
18. Multiple Genomic Events Altering Hominin SIGLEC Biology and Innate Immunity Predated the Common Ancestor of Humans and Archaic Hominins.
- Author
-
Khan, Naazneen, de Manuel, Marc, Peyregne, Stephane, Do, Raymond, Prufer, Kay, Marques-Bonet, Tomas, Varki, Nissi, Gagneux, Pascal, and Varki, Ajit
- Subjects
CD33rSiglecs ,Neanderthal/Denisovan ,archaic hominin ,common ancestor ,evolution ,great apes ,hominin ,sialic acid ,Biochemistry and Cell Biology ,Evolutionary Biology ,Genetics ,Developmental Biology - Abstract
Human-specific pseudogenization of the CMAH gene eliminated the mammalian sialic acid (Sia) Neu5Gc (generating an excess of its precursor Neu5Ac), thus changing ubiquitous cell surface "self-associated molecular patterns" that modulate innate immunity via engagement of CD33-related-Siglec receptors. The Alu-fusion-mediated loss-of-function of CMAH fixed ∼2-3 Ma, possibly contributing to the origins of the genus Homo. The mutation likely altered human self-associated molecular patterns, triggering multiple events, including emergence of human-adapted pathogens with strong preference for Neu5Ac recognition and/or presenting Neu5Ac-containing molecular mimics of human glycans, which can suppress immune responses via CD33-related-Siglec engagement. Human-specific alterations reported in some gene-encoding Sia-sensing proteins suggested a "hotspot" in hominin evolution. The availability of more hominid genomes including those of two extinct hominins now allows full reanalysis and evolutionary timing. Functional changes occur in 8/13 members of the human genomic cluster encoding CD33-related Siglecs, all predating the human common ancestor. Comparisons with great ape genomes indicate that these changes are unique to hominins. We found no evidence for strong selection after the Human-Neanderthal/Denisovan common ancestor, and these extinct hominin genomes include almost all major changes found in humans, indicating that these changes in hominin sialobiology predate the Neanderthal-human divergence ∼0.6 Ma. Multiple changes in this genomic cluster may also explain human-specific expression of CD33rSiglecs in unexpected locations such as amnion, placental trophoblast, pancreatic islets, ovarian fibroblasts, microglia, Natural Killer(NK) cells, and epithelia. Taken together, our data suggest that innate immune interactions with pathogens markedly altered hominin Siglec biology between 0.6 and 2 Ma, potentially affecting human evolution.
- Published
- 2020
19. Serum Antibodies to N-Glycolylneuraminic Acid Are Elevated in Duchenne Muscular Dystrophy and Correlate with Increased Disease Pathology in Cmah-/-mdx Mice.
- Author
-
Martin, Paul T, Kawanishi, Kunio, Ashbrook, Anna, Golden, Bethannie, Samraj, Annie, Crowe, Kelly E, Zygmunt, Deborah A, Okerblom, Jonathan, Yu, Hai, Maki, Agatha, Diaz, Sandra, Chen, Xi, Janssen, Paul ML, and Varki, Ajit
- Subjects
Animals ,Mice ,Inbred mdx ,Mice ,Knockout ,Humans ,Mice ,Muscular Dystrophy ,Duchenne ,Disease Models ,Animal ,Neuraminic Acids ,Autoantibodies ,Autoantigens ,Child ,Male ,Cardiovascular ,Duchenne/ Becker Muscular Dystrophy ,Nutrition ,Rare Diseases ,Brain Disorders ,Muscular Dystrophy ,Pediatric ,Clinical Research ,Intellectual and Developmental Disabilities (IDD) ,2.1 Biological and endogenous factors ,Aetiology ,Musculoskeletal ,Medical and Health Sciences ,Pathology - Abstract
Humans cannot synthesize the common mammalian sialic acid N-glycolylneuraminic acid (Neu5Gc) because of an inactivating deletion in the cytidine-5'-monophospho-(CMP)-N-acetylneuraminic acid hydroxylase (CMAH) gene responsible for its synthesis. Human Neu5Gc deficiency can lead to development of anti-Neu5Gc serum antibodies, the levels of which can be affected by Neu5Gc-containing diets and by disease. Metabolic incorporation of dietary Neu5Gc into human tissues in the face of circulating antibodies against Neu5Gc-bearing glycans is thought to exacerbate inflammation-driven diseases like cancer and atherosclerosis. Probing of sera with sialoglycan arrays indicated that patients with Duchenne muscular dystrophy (DMD) had a threefold increase in overall anti-Neu5Gc antibody titer compared with age-matched controls. These antibodies recognized a broad spectrum of Neu5Gc-containing glycans. Human-like inactivation of the Cmah gene in mice is known to modulate severity in a variety of mouse models of human disease, including the X chromosome-linked muscular dystrophy (mdx) model for DMD. Cmah-/-mdx mice can be induced to develop anti-Neu5Gc-glycan antibodies as humans do. The presence of anti-Neu5Gc antibodies, in concert with induced Neu5Gc expression, correlated with increased severity of disease pathology in Cmah-/-mdx mice, including increased muscle fibrosis, expression of inflammatory markers in the heart, and decreased survival. These studies suggest that patients with DMD who harbor anti-Neu5Gc serum antibodies might exacerbate disease severity when they ingest Neu5Gc-rich foods, like red meats.
- Published
- 2021
20. Sialoglycan recognition is a common connection linking acidosis, zinc, and HMGB1 in sepsis
- Author
-
Siddiqui, Shoib S, Dhar, Chirag, Sundaramurthy, Venkatasubramaniam, Sasmal, Aniruddha, Yu, Hai, Bandala-Sanchez, Esther, Li, Miaomiao, Zhang, Xiaoxiao, Chen, Xi, Harrison, Leonard C, Xu, Ding, and Varki, Ajit
- Subjects
Biochemistry and Cell Biology ,Biomedical and Clinical Sciences ,Biological Sciences ,Sepsis ,Infectious Diseases ,Hematology ,Inflammatory and immune system ,Acidosis ,Carrier Proteins ,HMGB1 Protein ,Humans ,Hydrogen-Ion Concentration ,Immunity ,Innate ,Lipopolysaccharides ,Polysaccharides ,Sialic Acids ,Sialoglycoproteins ,Zinc ,sialic acid ,Neu5Ac ,COVID-19 ,cytokine storm ,HMGB1 - Abstract
Blood pH is tightly maintained between 7.35 and 7.45, and acidosis (pH
- Published
- 2021
21. Evolutionary conservation of human ketodeoxynonulosonic acid production is independent of sialoglycan biosynthesis
- Author
-
Kawanishi, Kunio, Saha, Sudeshna, Diaz, Sandra, Vaill, Michael, Sasmal, Aniruddha, Siddiqui, Shoib S, Choudhury, Biswa P, Sharma, Kumar, Chen, Xi, Schoenhofen, Ian C, Sato, Chihiro, Kitajima, Ken, Freeze, Hudson H, Münster-Kühnel, Anja, and Varki, Ajit
- Subjects
2.1 Biological and endogenous factors ,Aetiology ,Atherosclerosis ,HEK293 Cells ,Human Umbilical Vein Endothelial Cells ,Humans ,Kidney Failure ,Chronic ,N-Acetylneuraminic Acid ,Polysaccharides ,Glycobiology ,Metabolism ,Nephrology ,Medical and Health Sciences ,Immunology - Abstract
Human metabolic incorporation of nonhuman sialic acid (Sia) N-glycolylneuraminic acid into endogenous glycans generates inflammation via preexisting antibodies, which likely contributes to red meat-induced atherosclerosis acceleration. Exploring whether this mechanism affects atherosclerosis in end-stage renal disease (ESRD), we instead found serum accumulation of 2-keto-3-deoxy-d-glycero-d-galacto-2-nonulosonic acid (Kdn), a Sia prominently expressed in cold-blooded vertebrates. In patients with ESRD, levels of the Kdn precursor mannose also increased, but within a normal range. Mannose ingestion by healthy volunteers raised the levels of urinary mannose and Kdn. Kdn production pathways remained conserved in mammals but were diminished by an M42T substitution in a key biosynthetic enzyme, N-acetylneuraminate synthase. Remarkably, reversion to the ancestral methionine then occurred independently in 2 lineages, including humans. However, mammalian glycan databases contain no Kdn-glycans. We hypothesize that the potential toxicity of excess mannose in mammals is partly buffered by conversion to free Kdn. Thus, mammals probably conserve Kdn biosynthesis and modulate it in a lineage-specific manner, not for glycosylation, but to control physiological mannose intermediates and metabolites. However, human cells can be forced to express Kdn-glycans via genetic mutations enhancing Kdn utilization, or by transfection with fish enzymes producing cytidine monophosphate-Kdn (CMP-Kdn). Antibodies against Kdn-glycans occur in pooled human immunoglobulins. Pathological conditions that elevate Kdn levels could therefore result in antibody-mediated inflammatory pathologies.
- Published
- 2021
22. Chronic OVA allergen challenged Siglec-F deficient mice have increased mucus, remodeling, and epithelial Siglec-F ligands which are up-regulated by IL-4 and IL-13
- Author
-
Doherty Taylor A, Dayan Shanna, Miller Marina, Rosenthal Peter, Pham Alexa, Song Dae Jae, Cho Jae Youn, Varki Ajit, and Broide David H
- Subjects
Diseases of the respiratory system ,RC705-779 - Abstract
Abstract Background In this study we examined the role of Siglec-F, a receptor highly expressed on eosinophils, in contributing to mucus expression, airway remodeling, and Siglec-F ligand expression utilizing Siglec-F deficient mice exposed to chronic allergen challenge. Methods Wild type (WT) and Siglec-F deficient mice were sensitized and challenged chronically with OVA for one month. Levels of airway inflammation (eosinophils), Siglec-F ligand expresion and remodeling (mucus, fibrosis, smooth muscle thickness, extracellular matrix protein deposition) were assessed in lung sections by image analysis and immunohistology. Airway hyperreactivity to methacholine was assessed in intubated and ventilated mice. Results Siglec-F deficient mice challenged with OVA for one month had significantly increased numbers of BAL and peribronchial eosinophils compared to WT mice which was associated with a significant increase in mucus expression as assessed by the number of periodic acid Schiff positive airway epithelial cells. In addition, OVA challenged Siglec-F deficient mice had significantly increased levels of peribronchial fibrosis (total lung collagen, area of peribronchial trichrome staining), as well as increased numbers of peribronchial TGF-β1+ cells, and increased levels of expression of the extracellular matrix protein fibronectin compared to OVA challenged WT mice. Lung sections immunostained with a Siglec-Fc to detect Siglec-F ligand expression demonstrated higher levels of expression of the Siglec-F ligand in the peribronchial region in OVA challenged Siglec-F deficient mice compared to WT mice. WT and Siglec-F deficient mice challenged intranasally with IL-4 or IL-13 had significantly increased levels of airway epithelial Siglec-F ligand expression, whereas this was not observed in WT or Siglec-F deficient mice challenged with TNF-α. There was a significant increase in the thickness of the peribronchial smooth muscle layer in OVA challenged Siglec-F deficient mice, but this was not associated with significant increased airway hyperreactivity compared to WT mice. Conclusions Overall, this study demonstrates an important role for Siglec-F in modulating levels of chronic eosinophilic airway inflammation, peribronchial fibrosis, thickness of the smooth muscle layer, mucus expression, fibronectin, and levels of peribronchial Siglec-F ligands suggesting that Siglec-F may normally function to limit levels of chronic eosinophilic inflammation and remodeling. In addition, IL-4 and IL-13 are important regulators of Siglec-F ligand expression by airway epithelium.
- Published
- 2010
- Full Text
- View/download PDF
23. Exploring the Impact of Ketodeoxynonulosonic Acid in Host-Pathogen Interactions Using Uptake and Surface Display by Nontypeable Haemophilus influenzae.
- Author
-
Saha, Sudeshna, Coady, Alison, Sasmal, Aniruddha, Kawanishi, Kunio, Choudhury, Biswa, Yu, Hai, Sorensen, Ricardo U, Inostroza, Jaime, Schoenhofen, Ian C, Chen, Xi, Münster-Kühnel, Anja, Sato, Chihiro, Kitajima, Ken, Ram, Sanjay, Nizet, Victor, and Varki, Ajit
- Subjects
Animals ,Mice ,Inbred C57BL ,Humans ,Mice ,Haemophilus influenzae ,Haemophilus Infections ,Sugar Acids ,Sialic Acids ,N-Acetylneuraminic Acid ,Glycoconjugates ,Immunoglobulin M ,Antigens ,CD ,Antibodies ,Molecular Mimicry ,Protein Binding ,Biological Transport ,Complement C3 ,Female ,Host-Pathogen Interactions ,Sialic Acid Binding Immunoglobulin-like Lectins ,CMAH ,Kdn ,Neu5Ac ,antibody ,bacterial pathogenesis ,glycobiology ,molecular mimicry ,nontypeable Haemophilus influenzae ,sialic acid ,Infectious Diseases ,Biotechnology ,Clinical Research ,2.2 Factors relating to the physical environment ,Infection ,Microbiology - Abstract
Surface expression of the common vertebrate sialic acid (Sia) N-acetylneuraminic acid (Neu5Ac) by commensal and pathogenic microbes appears structurally to represent "molecular mimicry" of host sialoglycans, facilitating multiple mechanisms of host immune evasion. In contrast, ketodeoxynonulosonic acid (Kdn) is a more ancestral Sia also present in prokaryotic glycoconjugates that are structurally quite distinct from vertebrate sialoglycans. We detected human antibodies against Kdn-terminated glycans, and sialoglycan microarray studies found these anti-Kdn antibodies to be directed against Kdn-sialoglycans structurally similar to those on human cell surface Neu5Ac-sialoglycans. Anti-Kdn-glycan antibodies appear during infancy in a pattern similar to those generated following incorporation of the nonhuman Sia N-glycolylneuraminic acid (Neu5Gc) onto the surface of nontypeable Haemophilus influenzae (NTHi), a human commensal and opportunistic pathogen. NTHi grown in the presence of free Kdn took up and incorporated the Sia into its lipooligosaccharide (LOS). Surface display of the Kdn within NTHi LOS blunted several virulence attributes of the pathogen, including Neu5Ac-mediated resistance to complement and whole blood killing, complement C3 deposition, IgM binding, and engagement of Siglec-9. Upper airway administration of Kdn reduced NTHi infection in human-like Cmah null (Neu5Gc-deficient) mice that express a Neu5Ac-rich sialome. We propose a mechanism for the induction of anti-Kdn antibodies in humans, suggesting that Kdn could be a natural and/or therapeutic "Trojan horse" that impairs colonization and virulence phenotypes of free Neu5Ac-assimilating human pathogens.IMPORTANCE All cells in vertebrates are coated with a dense array of glycans often capped with sugars called sialic acids. Sialic acids have many functions, including serving as a signal for recognition of "self" cells by the immune system, thereby guiding an appropriate immune response against foreign "nonself" and/or damaged cells. Several pathogenic bacteria have evolved mechanisms to cloak themselves with sialic acids and evade immune responses. Here we explore a type of sialic acid called "Kdn" (ketodeoxynonulosonic acid) that has not received much attention in the past and compare and contrast how it interacts with the immune system. Our results show potential for the use of Kdn as a natural intervention against pathogenic bacteria that take up and coat themselves with external sialic acid from the environment.
- Published
- 2021
24. Display of the human mucinome with defined O-glycans by gene engineered cells
- Author
-
Nason, Rebecca, Büll, Christian, Konstantinidi, Andriana, Sun, Lingbo, Ye, Zilu, Halim, Adnan, Du, Wenjuan, Sørensen, Daniel M, Durbesson, Fabien, Furukawa, Sanae, Mandel, Ulla, Joshi, Hiren J, Dworkin, Leo Alexander, Hansen, Lars, David, Leonor, Iverson, Tina M, Bensing, Barbara A, Sullam, Paul M, Varki, Ajit, Vries, Erik de, de Haan, Cornelis AM, Vincentelli, Renaud, Henrissat, Bernard, Vakhrushev, Sergey Y, Clausen, Henrik, and Narimatsu, Yoshiki
- Subjects
Rare Diseases ,Genetic Engineering ,Glycosylation ,HEK293 Cells ,Humans ,Microbiota ,Mucin-1 ,Mucins ,Mucous Membrane ,Polysaccharides - Abstract
Mucins are a large family of heavily O-glycosylated proteins that cover all mucosal surfaces and constitute the major macromolecules in most body fluids. Mucins are primarily defined by their variable tandem repeat (TR) domains that are densely decorated with different O-glycan structures in distinct patterns, and these arguably convey much of the informational content of mucins. Here, we develop a cell-based platform for the display and production of human TR O-glycodomains (~200 amino acids) with tunable structures and patterns of O-glycans using membrane-bound and secreted reporters expressed in glycoengineered HEK293 cells. Availability of defined mucin TR O-glycodomains advances experimental studies into the versatile role of mucins at the interface with pathogenic microorganisms and the microbiome, and sparks new strategies for molecular dissection of specific roles of adhesins, glycoside hydrolases, glycopeptidases, viruses and other interactions with mucin TRs as highlighted by examples.
- Published
- 2021
25. Evaluation of IL-17D in Host Immunity to Group A Streptococcus Infection
- Author
-
Washington, Allen, Varki, Nissi, Valderrama, J Andrés, Nizet, Victor, and Bui, Jack D
- Subjects
Biodefense ,Infectious Diseases ,Prevention ,Vaccine Related ,Emerging Infectious Diseases ,Aetiology ,2.1 Biological and endogenous factors ,Inflammatory and immune system ,Infection ,Animals ,Chemokine CCL2 ,Immunity ,Innate ,Interleukin-27 ,Killer Cells ,Natural ,Mice ,Mice ,Inbred C57BL ,Neutrophils ,Pathogen-Associated Molecular Pattern Molecules ,Streptococcal Infections ,Streptococcus ,Th17 Cells ,Immunology - Abstract
IL-17D is a cytokine that belongs to the IL-17 family and is conserved in vertebrates and invertebrates. In contrast to IL-17A and IL-17F, which are expressed in Th17 cells, IL-17D is expressed broadly in nonimmune cells. IL-17D can promote immune responses to cancer and viruses in part by inducing chemokines and recruiting innate immune cells such as NK cells. Although bacterial infection can induce IL-17D in fish and invertebrates, the role of mammalian IL-17D in antibacterial immunity has not been established. To determine whether IL-17D has a role in mediating host defense against bacterial infections, we studied i.p. infection by group A Streptococcus (GAS) in wild-type (WT) and Il17d -/- mice. Compared with WT animals, mice deficient in IL-17D experienced decreased survival, had greater weight loss, and showed increased bacterial burden in the kidney and peritoneal cavity following GAS challenge. In WT animals, IL-17D transcript was induced by GAS infection and correlated to increased levels of chemokine CCL2 and greater neutrophil recruitment. Of note, GAS-mediated IL-17D induction in nonimmune cells required live bacteria, suggesting that processes beyond recognition of pathogen-associated molecular patterns were required for IL-17D induction. Based on our results, we propose a model in which nonimmune cells can discriminate between nonviable and viable GAS cells, responding only to the latter by inducing IL-17D.
- Published
- 2020
26. Human species-specific loss of CMP-N-acetylneuraminic acid hydroxylase enhances atherosclerosis via intrinsic and extrinsic mechanisms.
- Author
-
Kawanishi, Kunio, Dhar, Chirag, Do, Raymond, Varki, Nissi, Gordts, Philip LSM, and Varki, Ajit
- Subjects
Macrophages ,Animals ,Mice ,Inbred C57BL ,Cattle ,Humans ,Hyperglycemia ,Inflammation ,Sialic Acids ,Mixed Function Oxygenases ,Receptors ,LDL ,Cytokines ,Species Specificity ,Phenotype ,Models ,Biological ,Female ,Male ,Atherosclerosis ,Diet ,High-Fat ,CMAH ,N-glycolylneuraminic acid ,atherosclerosis ,cytidine-5′-monophosphate (CMP)-N-acetylneuraminic acid (Neu5Ac) hydroxylase ,human evolution ,Cardiovascular ,Nutrition ,Heart Disease ,Prevention ,cytidine-5 '-monophosphate (CMP)-N-acetylneuraminic acid (Neu5Ac) hydroxylase - Abstract
Cardiovascular disease (CVD) events due to atherosclerosis cause one-third of worldwide deaths and risk factors include physical inactivity, age, dyslipidemia, hypertension, diabetes, obesity, smoking, and red meat consumption. However, ∼15% of first-time events occur without such factors. In contrast, coronary events are extremely rare even in closely related chimpanzees in captivity, despite human-like CVD-risk-prone blood lipid profiles, hypertension, and mild atherosclerosis. Similarly, red meat-associated enhancement of CVD event risk does not seem to occur in other carnivorous mammals. Thus, heightened CVD risk may be intrinsic to humans, and genetic changes during our evolution need consideration. Humans exhibit a species-specific deficiency of the sialic acid N-glycolylneuraminic acid (Neu5Gc), due to pseudogenization of cytidine monophosphate-N-acetylneuraminic acid (Neu5Ac) hydroxylase (CMAH), which occurred in hominin ancestors ∼2 to 3 Mya. Ldlr -/- mice with human-like Cmah deficiency fed a sialic acids (Sias)-free high-fat diet (HFD) showed ∼1.9-fold increased atherogenesis over Cmah wild-type Ldlr -/- mice, associated with elevated macrophage cytokine expression and enhanced hyperglycemia. Human consumption of Neu5Gc (from red meat) acts as a "xeno-autoantigen" via metabolic incorporation into endogenous glycoconjugates, as interactions with circulating anti-Neu5Gc "xeno-autoantibodies" potentiate chronic inflammation ("xenosialitis"). Cmah -/- Ldlr -/- mice immunized with Neu5Gc-bearing antigens to generate human-like anti-Neu5Gc antibodies suffered a ∼2.4-fold increased atherosclerosis on a Neu5Gc-rich HFD, compared with Neu5Ac-rich or Sias-free HFD. Lesions in Neu5Gc-immunized and Neu5Gc-rich HFD-fed Cmah -/- Ldlr -/- mice were more advanced but unexplained by lipoprotein or glucose changes. Human evolutionary loss of CMAH likely contributes to atherosclerosis predisposition via multiple intrinsic and extrinsic mechanisms, and future studies could consider this more human-like model.
- Published
- 2019
27. Evolution of the exclusively human pathogen Neisseria gonorrhoeae: Human‐specific engagement of immunoregulatory Siglecs
- Author
-
Landig, Corinna S, Hazel, Ashley, Kellman, Benjamin P, Fong, Jerry J, Schwarz, Flavio, Agarwal, Sarika, Varki, Nissi, Massari, Paola, Lewis, Nathan E, Ram, Sanjay, and Varki, Ajit
- Subjects
Biological Sciences ,Ecology ,Evolutionary Biology ,Genetics ,Urologic Diseases ,Clinical Research ,Sexually Transmitted Infections ,HIV/AIDS ,Infectious Diseases ,Inflammatory and immune system ,Good Health and Well Being ,disease biology ,evolutionary medicine ,gonorrhea ,microbial biology ,polymorphism ,population genetics ,sialic acid ,Siglecs ,Medicinal and Biomolecular Chemistry ,Evolutionary biology - Abstract
Neisseria gonorrhoeae causes the sexually transmitted disease gonorrhea exclusively in humans and uses multiple strategies to infect, including acquisition of host sialic acids that cap and mask lipooligosaccharide termini, while restricting complement activation. We hypothesized that gonococci selectively target human anti-inflammatory sialic acid-recognizing Siglec receptors on innate immune cells to blunt host responses and that pro-inflammatory Siglecs and SIGLEC pseudogene polymorphisms represent host evolutionary adaptations to counteract this interaction. N. gonorrhoeae can indeed engage multiple human but not chimpanzee CD33rSiglecs expressed on innate immune cells and in the genitourinary tract--including Siglec-11 (inhibitory) and Siglec-16 (activating), which we detected for the first time on human cervical epithelium. Surprisingly, in addition to LOS sialic acid, we found that gonococcal porin (PorB) mediated binding to multiple Siglecs. PorB also bound preferentially to human Siglecs and not chimpanzee orthologs, modulating host immune reactions in a human-specific manner. Lastly, we studied the distribution of null SIGLEC polymorphisms in a Namibian cohort with a high prevalence of gonorrhea and found that uninfected women preferentially harbor functional SIGLEC16 alleles encoding an activating immune receptor. These results contribute to the understanding of the human specificity of N. gonorrhoeae and how it evolved to evade the human immune defense.
- Published
- 2019
28. DDX5 promotes oncogene C3 and FABP1 expressions and drives intestinal inflammation and tumorigenesis
- Author
-
Abbasi, Nazia, Long, Tianyun, Li, Yuxin, Yee, Brian A, Cho, Benjamin S, Hernandez, Juan E, Ma, Evelyn, Patel, Parth R, Sahoo, Debashis, Sayed, Ibrahim M, Varki, Nissi, Das, Soumita, Ghosh, Pradipta, Yeo, Gene W, and Huang, Wendy Jia Men
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Cancer ,Autoimmune Disease ,Colo-Rectal Cancer ,Genetics ,Inflammatory Bowel Disease ,Digestive Diseases ,2.1 Biological and endogenous factors ,Aetiology ,Animals ,Carcinogenesis ,Colitis ,Complement C3 ,DEAD-box RNA Helicases ,Dextran Sulfate ,Epithelial Cells ,Fatty Acid-Binding Proteins ,Female ,Gene Expression ,Gene Expression Regulation ,Neoplastic ,Inflammation ,Intestinal Mucosa ,Intestine ,Small ,Intestines ,Male ,Mice ,Mice ,Inbred C57BL ,Oncogenes ,Signal Transduction ,Biological sciences ,Biomedical and clinical sciences - Abstract
Tumorigenesis in different segments of the intestinal tract involves tissue-specific oncogenic drivers. In the colon, complement component 3 (C3) activation is a major contributor to inflammation and malignancies. By contrast, tumorigenesis in the small intestine involves fatty acid-binding protein 1 (FABP1). However, little is known of the upstream mechanisms driving their expressions in different segments of the intestinal tract. Here, we report that the RNA-binding protein DDX5 binds to the mRNA transcripts of C3 and Fabp1 to augment their expressions posttranscriptionally. Knocking out DDX5 in epithelial cells protected mice from intestinal tumorigenesis and dextran sodium sulfate (DSS)-induced colitis. Identification of DDX5 as a common upstream regulator of tissue-specific oncogenic molecules provides an excellent therapeutic target for intestinal diseases.
- Published
- 2020
29. Modified Sialic Acids on Mucus and Erythrocytes Inhibit Influenza A Virus Hemagglutinin and Neuraminidase Functions
- Author
-
Barnard, Karen N, Alford-Lawrence, Brynn K, Buchholz, David W, Wasik, Brian R, LaClair, Justin R, Yu, Hai, Honce, Rebekah, Ruhl, Stefan, Pajic, Petar, Daugherity, Erin K, Chen, Xi, Schultz-Cherry, Stacey L, Aguilar, Hector C, Varki, Ajit, and Parrish, Colin R
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Clinical Sciences ,Biological Sciences ,Biodefense ,Infectious Diseases ,Pneumonia & Influenza ,Emerging Infectious Diseases ,Influenza ,2.1 Biological and endogenous factors ,A549 Cells ,Animals ,Dogs ,Erythrocytes ,Female ,Hemagglutinin Glycoproteins ,Influenza Virus ,Hemagglutinins ,Humans ,Influenza A virus ,Influenza ,Human ,Madin Darby Canine Kidney Cells ,Male ,Mice ,Mixed Function Oxygenases ,Mucus ,N-Acetylneuraminic Acid ,Neuraminidase ,Orthomyxoviridae ,Receptors ,Virus ,Saliva ,influenza ,mucus ,sialic acids ,Agricultural and Veterinary Sciences ,Medical and Health Sciences ,Virology ,Agricultural ,veterinary and food sciences ,Biological sciences ,Biomedical and clinical sciences - Abstract
Sialic acids (Sia) are the primary receptors for influenza viruses and are widely displayed on cell surfaces and in secreted mucus. Sia may be present in variant forms that include O-acetyl modifications at C-4, C-7, C-8, and C-9 positions and N-acetyl or N-glycolyl at C-5. They can also vary in their linkages, including α2-3 or α2-6 linkages. Here, we analyze the distribution of modified Sia in cells and tissues of wild-type mice or in mice lacking CMP-N-acetylneuraminic acid hydroxylase (CMAH) enzyme, which synthesizes N-glycolyl (Neu5Gc) modifications. We also examined the variation of Sia forms on erythrocytes and in saliva from different animals. To determine the effect of Sia modifications on influenza A virus (IAV) infection, we tested for effects on hemagglutinin (HA) binding and neuraminidase (NA) cleavage. We confirmed that 9-O-acetyl, 7,9-O-acetyl, 4-O-acetyl, and Neu5Gc modifications are widely but variably expressed in mouse tissues, with the highest levels detected in the respiratory and gastrointestinal (GI) tracts. Secreted mucins in saliva and surface proteins of erythrocytes showed a high degree of variability in display of modified Sia between different species. IAV HAs from different virus strains showed consistently reduced binding to both Neu5Gc- and O-acetyl-modified Sia; however, while IAV NAs were inhibited by Neu5Gc and O-acetyl modifications, there was significant variability between NA types. The modifications of Sia in mucus may therefore have potent effects on the functions of IAV and may affect both pathogens and the normal flora of different mucosal sites.IMPORTANCE Sialic acids (Sia) are involved in numerous different cellular functions and are receptors for many pathogens. Sia come in chemically modified forms, but we lack a clear understanding of how they alter interactions with microbes. Here, we examine the expression of modified Sia in mouse tissues, on secreted mucus in saliva, and on erythrocytes, including those from IAV host species and animals used in IAV research. These Sia forms varied considerably among different animals, and their inhibitory effects on IAV NA and HA activities and on bacterial sialidases (neuraminidases) suggest a host-variable protective role in secreted mucus.
- Published
- 2020
30. Maximum reproductive lifespan correlates with CD33rSIGLEC gene number: Implications for NADPH oxidase‐derived reactive oxygen species in aging
- Author
-
Khan, Naazneen, Kim, Stuart K, Gagneux, Pascal, Dugan, Laura L, and Varki, Ajit
- Subjects
Biomedical and Clinical Sciences ,Immunology ,Aging ,1.1 Normal biological development and functioning ,Underpinning research ,Good Health and Well Being ,Animals ,Gene Dosage ,Humans ,Longevity ,NADPH Oxidases ,Neutrophils ,Reactive Oxygen Species ,Sialic Acid Binding Ig-like Lectin 3 ,Whale ,Killer ,CD33rSIGLEC ,NADPH-oxidase ,prolonged post-reproductive lifespan ,reactive oxygen species ,CD33rSIGLEC ,Biochemistry and Cell Biology ,Physiology ,Medical Physiology ,Biochemistry & Molecular Biology ,Biochemistry and cell biology ,Medical physiology - Abstract
Humans and orcas are among the very rare species that have a prolonged post-reproductive lifespan (PRLS), during which the aging process continues. Reactive oxygen species (ROS) derived from mitochondria and from the NADPH oxidase (NOX) enzymes of innate immune cells are known to contribute to aging, with the former thought to be dominant. CD33-related-Siglecs are immune receptors that recognize self-associated-molecular-patterns and modulate NOX-derived-ROS. We herewith demonstrate a strong correlation of lifespan with CD33rSIGLEC gene number in 26 species, independent of body weight or phylogeny. The correlation is stronger when considering total CD33rSIGLEC gene number rather than those encoding inhibitory and activating subsets, suggesting that lifetime balancing of ROS is important. Combining independent lines of evidence including the short half-life and spontaneous activation of neutrophils, we calculate that even without inter-current inflammation, a major source of lifetime ROS exposure may actually be neutrophil NOX-derived. However, genomes of human supercentenarians (>110 years) do not harbor a significantly higher number of functional CD33rSIGLEC genes. Instead, lifespan correlation with CD33rSIGLEC gene number was markedly strengthened by excluding the post-reproductive lifespan of humans and orcas (R2 = 0.83; P
- Published
- 2020
31. Siglec-14 Enhances NLRP3-Inflammasome Activation in Macrophages
- Author
-
Tsai, Chih-Ming, Riestra, Angelica M, Ali, Syed Raza, Fong, Jerry J, Liu, Janet Z, Hughes, Gillian, Varki, Ajit, and Nizet, Victor
- Subjects
Biodefense ,Vaccine Related ,Emerging Infectious Diseases ,Prevention ,Clinical Research ,2.1 Biological and endogenous factors ,Aetiology ,Inflammatory and immune system ,Infection ,Humans ,Inflammasomes ,Lectins ,Macrophages ,NLR Family ,Pyrin Domain-Containing 3 Protein ,Receptors ,Cell Surface ,Streptococcal Infections ,Streptococcus agalactiae ,THP-1 Cells ,Siglec ,Interleukin-1 beta ,Caspase-1 ,Inflammasome ,Innate immunity ,Vimentin ,Interleukin-1β ,Medical and Health Sciences - Abstract
Pathogenic microorganisms are sensed by the inflammasome, resulting in the release of the pro-immune and proinflammatory cytokine interleukin-1β (IL-1β). In humans, the paired sialic acid-binding Ig-like lectin receptors Siglec-5 (inhibitory) and Siglec-14 (activating) have been shown to have reciprocal roles in regulating macrophage immune responses, but their interaction with IL-1β signaling and the inflammasome has not been characterized. Here we show that in response to known inflammasome activators (ATP, nigericin) or the sialic acid-expressing human bacterial pathogen group B Streptococcus (GBS), the presence of Siglec-14 enhances, whereas Siglec-5 reduces, inflammasome activation and macrophage IL-1β release. Human THP-1 macrophages stably transfected with Siglec-14 exhibited increased caspase-1 activation, IL-1β release and pyroptosis after GBS infection, in a manner blocked by a specific inhibitor of nucleotide-binding domain leucine-rich repeat protein 3 (NLRP3), a protein involved in inflammasome assembly. Another leading pathogen, Streptococcus pneumoniae, lacks sialic acid but rather prominently expresses a sialidase, which cleaves sialic acid from macrophages, eliminating cis- interactions with the lectin receptor, thus attenuating Siglec-14 induced IL-1β secretion. Vimentin, a cytoskeletal protein released during macrophage inflammatory activation is known to induce the inflammasome. We found that vimentin has increased interaction with Siglec-14 compared to Siglec-5, and this interaction heightened IL-1β production by Siglec-14-expressing cells. Siglec-14 is absent from some humans because of a SIGLEC5/14 fusion polymorphism, and we found increased IL-1β expression in primary macrophages from SIGLEC14+/+ individuals compared to those with the SIGLEC14-/+ and SIGLEC14-/- genotypes. Collectively, our results identify a new immunoregulatory role of Siglec-14 as a positive regulator of NLRP3 inflammasome activation.
- Published
- 2020
32. Long-term exposure to house dust mites accelerates lung cancer development in mice
- Author
-
Dongjie Wang, Wen Li, Natalie Albasha, Lindsey Griffin, Han Chang, Lauren Amaya, Sneha Ganguly, Liping Zeng, Bora Keum, José M. González-Navajas, Matt Levin, Zohreh AkhavanAghdam, Helen Snyder, David Schwartz, Ailin Tao, Laela M. Boosherhri, Hal M. Hoffman, Michael Rose, Monica Valeria Estrada, Nissi Varki, Scott Herdman, Maripat Corr, Nicholas J. G. Webster, Eyal Raz, and Samuel Bertin
- Subjects
Lung cancer ,Kras ,Urethane ,House dust mites ,Chronic inflammation ,NLRP3 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Individuals with certain chronic inflammatory lung diseases have a higher risk of developing lung cancer (LC). However, the underlying mechanisms remain largely unknown. Here, we hypothesized that chronic exposure to house dust mites (HDM), a common indoor aeroallergen associated with the development of asthma, accelerates LC development through the induction of chronic lung inflammation (CLI). Methods The effects of HDM and heat-inactivated HDM (HI-HDM) extracts were evaluated in two preclinical mouse models of LC (a chemically-induced model using the carcinogen urethane and a genetically-driven model with oncogenic Kras G12D activation in lung epithelial cells) and on murine macrophages in vitro. Pharmacological blockade or genetic deletion of the Nod-like receptor family pyrin domain-containing protein 3 (NLRP3) inflammasome, caspase-1, interleukin-1β (IL-1β), and C–C motif chemokine ligand 2 (CCL2) or treatment with an inhaled corticosteroid (ICS) was used to uncover the pro-tumorigenic effect of HDM. Results Chronic intranasal (i.n) instillation of HDM accelerated LC development in the two mouse models. Mechanistically, HDM caused a particular subtype of CLI, in which the NLRP3/IL-1β signaling pathway is chronically activated in macrophages, and made the lung microenvironment conducive to tumor development. The tumor-promoting effect of HDM was significantly decreased by heat treatment of the HDM extract and was inhibited by NLRP3, IL-1β, and CCL2 neutralization, or ICS treatment. Conclusions Collectively, these data indicate that long-term exposure to HDM can accelerate lung tumorigenesis in susceptible hosts (e.g., mice and potentially humans exposed to lung carcinogens or genetically predisposed to develop LC).
- Published
- 2023
- Full Text
- View/download PDF
33. Gut bacteria responding to dietary change encode sialidases that exhibit preference for red meat-associated carbohydrates
- Author
-
Zaramela, Livia S, Martino, Cameron, Alisson-Silva, Frederico, Rees, Steven D, Diaz, Sandra L, Chuzel, Léa, Ganatra, Mehul B, Taron, Christopher H, Secrest, Patrick, Zuñiga, Cristal, Huang, Jianbo, Siegel, Dionicio, Chang, Geoffrey, Varki, Ajit, and Zengler, Karsten
- Subjects
Nutrition ,Digestive Diseases ,Metabolic and endocrine ,Oral and gastrointestinal ,Animals ,Bacteria ,Bacteroides ,Clostridiales ,Crystallography ,X-Ray ,Diet ,Feces ,Female ,Gastrointestinal Microbiome ,Humans ,Male ,Metagenomics ,Mice ,Mice ,Inbred C57BL ,Neuraminic Acids ,Neuraminidase ,Polysaccharides ,Red Meat ,Microbiology ,Medical Microbiology - Abstract
Dietary habits have been associated with alterations of the human gut resident microorganisms contributing to obesity, diabetes and cancer1. In Western diets, red meat is a frequently eaten food2, but long-term consumption has been associated with increased risk of disease3,4. Red meat is enriched in N-glycolylneuraminic acid (Neu5Gc) that cannot be synthesized by humans5. However, consumption can cause Neu5Gc incorporation into cell surface glycans6, especially in carcinomas4,7. As a consequence, an inflammatory response is triggered when Neu5Gc-containing glycans encounter circulating anti-Neu5Gc antibodies8,9. Although bacteria can use free sialic acids as a nutrient source10-12, it is currently unknown if gut microorganisms contribute to releasing Neu5Gc from food. We found that a Neu5Gc-rich diet induces changes in the gut microbiota, with Bacteroidales and Clostridiales responding the most. Genome assembling of mouse and human shotgun metagenomic sequencing identified bacterial sialidases with previously unobserved substrate preference for Neu5Gc-containing glycans. X-ray crystallography revealed key amino acids potentially contributing to substrate preference. Additionally, we verified that mouse and human sialidases were able to release Neu5Gc from red meat. The release of Neu5Gc from red meat using bacterial sialidases could reduce the risk of inflammatory diseases associated with red meat consumption, including colorectal cancer4 and atherosclerosis13.
- Published
- 2019
34. Lp(a), oxidized phospholipids and oxidation-specific epitopes are increased in subjects with keloid formation
- Author
-
Sundeep Ruder, Brett Mansfield, Andrew Ronald Immelman, Nissi Varki, Phuong Miu, Frederick Raal, and Sotirios Tsimikas
- Subjects
Nutritional diseases. Deficiency diseases ,RC620-627 - Abstract
Highlights Keloid formation is common among individuals of African ancestry. Lp(a) and OxPL are associated with acute and chronic CVD events. Lp(a) and OxPL-apoB levels are elevated in patients with keloids. Keloids express OxPL staining but not Lp(a) staining. OxPL on Lp(a) may be mechanistically involved in keloid formation.
- Published
- 2022
- Full Text
- View/download PDF
35. Human evolutionary loss of epithelial Neu5Gc expression and species-specific susceptibility to cholera.
- Author
-
Alisson-Silva, Frederico, Liu, Janet Z, Diaz, Sandra L, Deng, Lingquan, Gareau, Mélanie G, Marchelletta, Ronald, Chen, Xi, Nizet, Victor, Varki, Nissi, Barrett, Kim E, and Varki, Ajit
- Subjects
Intestine ,Small ,Epithelial Cells ,Animals ,Mice ,Inbred C57BL ,Mice ,Knockout ,Humans ,Mice ,Vibrio cholerae ,Cholera ,Disease Susceptibility ,Neuraminic Acids ,Mixed Function Oxygenases ,Species Specificity ,Female ,Male ,Biological Evolution ,Intestine ,Small ,Inbred C57BL ,Knockout ,Virology ,Microbiology ,Immunology ,Medical Microbiology - Abstract
While infectious agents have typical host preferences, the noninvasive enteric bacterium Vibrio cholerae is remarkable for its ability to survive in many environments, yet cause diarrheal disease (cholera) only in humans. One key V. cholerae virulence factor is its neuraminidase (VcN), which releases host intestinal epithelial sialic acids as a nutrition source and simultaneously remodels intestinal polysialylated gangliosides into monosialoganglioside GM1. GM1 is the optimal binding target for the B subunit of a second virulence factor, the AB5 cholera toxin (Ctx). This coordinated process delivers the CtxA subunit into host epithelia, triggering fluid loss via cAMP-mediated activation of anion secretion and inhibition of electroneutral NaCl absorption. We hypothesized that human-specific and human-universal evolutionary loss of the sialic acid N-glycolylneuraminic acid (Neu5Gc) and the consequent excess of N-acetylneuraminic acid (Neu5Ac) contributes to specificity at one or more steps in pathogenesis. Indeed, VcN was less efficient in releasing Neu5Gc than Neu5Ac. We show enhanced binding of Ctx to sections of small intestine and isolated polysialogangliosides from human-like Neu5Gc-deficient Cmah-/- mice compared to wild-type, suggesting that Neu5Gc impeded generation of the GM1 target. Human epithelial cells artificially expressing Neu5Gc were also less susceptible to Ctx binding and CtxA intoxication following VcN treatment. Finally, we found increased fluid secretion into loops of Cmah-/- mouse small intestine injected with Ctx, indicating an additional direct effect on ion transport. Thus, V. cholerae evolved into a human-specific pathogen partly by adapting to the human evolutionary loss of Neu5Gc, optimizing multiple steps in cholera pathogenesis.
- Published
- 2018
36. Proteomic atlas of organ vasculopathies triggered by Staphylococcus aureus sepsis.
- Author
-
Toledo, Alejandro Gómez, Golden, Gregory, Campos, Alexandre Rosa, Cuello, Hector, Sorrentino, James, Lewis, Nathan, Varki, Nissi, Nizet, Victor, Smith, Jeffrey W, and Esko, Jeffrey D
- Subjects
Liver ,Animals ,Mice ,Inbred C57BL ,Mice ,Staphylococcal Infections ,Sepsis ,Vascular Diseases ,Multiple Organ Failure ,Hyaluronic Acid ,Proteome ,Proteomics ,Methicillin-Resistant Staphylococcus aureus ,Vascular Remodeling ,Inbred C57BL - Abstract
Sepsis is a life-threatening condition triggered by a dysregulated host response to microbial infection resulting in vascular dysfunction, organ failure and death. Here we provide a semi-quantitative atlas of the murine vascular cell-surface proteome at the organ level, and how it changes during sepsis. Using in vivo chemical labeling and high-resolution mass spectrometry, we demonstrate the presence of a vascular proteome that is perfusable and shared across multiple organs. This proteome is enriched in membrane-anchored proteins, including multiple regulators of endothelial barrier functions and innate immunity. Further, we automated our workflows and applied them to a murine model of methicillin-resistant Staphylococcus aureus (MRSA) sepsis to unravel changes during systemic inflammatory responses. We provide an organ-specific atlas of both systemic and local changes of the vascular proteome triggered by sepsis. Collectively, the data indicates that MRSA-sepsis triggers extensive proteome remodeling of the vascular cell surfaces, in a tissue-specific manner.
- Published
- 2019
37. An Atlas of Human Glycosylation Pathways Enables Display of the Human Glycome by Gene Engineered Cells
- Author
-
Narimatsu, Yoshiki, Joshi, Hiren J, Nason, Rebecca, Van Coillie, Julie, Karlsson, Richard, Sun, Lingbo, Ye, Zilu, Chen, Yen-Hsi, Schjoldager, Katrine T, Steentoft, Catharina, Furukawa, Sanae, Bensing, Barbara A, Sullam, Paul M, Thompson, Andrew J, Paulson, James C, Büll, Christian, Adema, Gosse J, Mandel, Ulla, Hansen, Lars, Bennett, Eric Paul, Varki, Ajit, Vakhrushev, Sergey Y, Yang, Zhang, and Clausen, Henrik
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,Biotechnology ,Genetics ,Human Genome ,Emerging Infectious Diseases ,Epitopes ,Genetic Engineering ,Glycosylation ,Glycosyltransferases ,HEK293 Cells ,Humans ,Metabolic Networks and Pathways ,Oligosaccharides ,Polysaccharides ,Proteins ,adhesin ,carbohydrate ,galectin ,glycan array ,glycoengineering ,glycosylation ,glycosyltransferase ,lectin ,microarray ,siglec ,Medical and Health Sciences ,Developmental Biology ,Biological sciences ,Biomedical and clinical sciences ,Health sciences - Abstract
The structural diversity of glycans on cells-the glycome-is vast and complex to decipher. Glycan arrays display oligosaccharides and are used to report glycan hapten binding epitopes. Glycan arrays are limited resources and present saccharides without the context of other glycans and glycoconjugates. We used maps of glycosylation pathways to generate a library of isogenic HEK293 cells with combinatorially engineered glycosylation capacities designed to display and dissect the genetic, biosynthetic, and structural basis for glycan binding in a natural context. The cell-based glycan array is self-renewable and reports glycosyltransferase genes required (or blocking) for interactions through logical sequential biosynthetic steps, which is predictive of structural glycan features involved and provides instructions for synthesis, recombinant production, and genetic dissection strategies. Broad utility of the cell-based glycan array is demonstrated, and we uncover higher order binding of microbial adhesins to clustered patches of O-glycans organized by their presentation on proteins.
- Published
- 2019
38. MST1R kinase accelerates pancreatic cancer progression via effects on both epithelial cells and macrophages
- Author
-
Babicky, Michele L, Harper, Megan M, Chakedis, Jeffery, Cazes, Alex, Mose, Evangeline S, Jaquish, Dawn V, French, Randall P, Childers, Betzaira, Alakus, Hakan, Schmid, Michael C, Foubert, Phillippe, Miyamoto, Jaclyn, Holman, Patrick J, Walterscheid, Zakkary J, Tang, Chih-Min, Varki, Nissi, Sicklick, Jason K, Messer, Karen, Varner, Judith A, Waltz, Susan E, and Lowy, Andrew M
- Subjects
Biomedical and Clinical Sciences ,Oncology and Carcinogenesis ,Immunology ,Pancreatic Cancer ,Rare Diseases ,Cancer ,Digestive Diseases ,Aetiology ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,2.1 Biological and endogenous factors ,Animals ,Carcinoma ,Pancreatic Ductal ,Disease Progression ,Epithelial Cells ,Female ,Gene Knockdown Techniques ,Humans ,Intracellular Signaling Peptides and Proteins ,Macrophages ,Male ,Mice ,Mice ,Inbred C57BL ,Mice ,Transgenic ,Pancreatic Neoplasms ,Proof of Concept Study ,Protein Serine-Threonine Kinases ,Receptor Protein-Tyrosine Kinases ,Signal Transduction ,Tumor Microenvironment ,Clinical Sciences ,Oncology & Carcinogenesis ,Biochemistry and cell biology ,Oncology and carcinogenesis - Abstract
The MST1R (RON) kinase is overexpressed in >80% of human pancreatic cancers, but its role in pancreatic carcinogenesis is unknown. In this study, we examined the relevance of Mst1r kinase to Kras driven pancreatic carcinogenesis using genetically engineered mouse models. In the setting of mutant Kras, Mst1r overexpression increased acinar-ductal metaplasia (ADM), accelerated the progression of pancreatic intraepithelial neoplasia (PanIN), and resulted in the accumulation of (mannose receptor C type 1) MRC1+, (arginase 1) Arg+ macrophages in the tumor microenvironment. Conversely, absence of a functional Mst1r kinase slowed PanIN initiation, resulted in smaller tumors, prolonged survival and a reduced tumor-associated macrophage content. Mst1r expression was associated with increased production of its ligand Mst1, and in orthotopic models, suppression of Mst1 expression resulted in reduced tumor size, changes in macrophage polarization and enhanced T cell infiltration. This study demonstrates the functional significance of Mst1r during pancreatic cancer initiation and progression. Further, it provides proof of concept that targeting Mst1r can modulate pancreatic cancer growth and the microenvironment. This study provides further rationale for targeting Mst1r as a therapeutic strategy.
- Published
- 2019
39. Dual actions of group B Streptococcus capsular sialic acid provide resistance to platelet-mediated antimicrobial killing
- Author
-
Uchiyama, Satoshi, Sun, Josh, Fukahori, Kyoko, Ando, Nao, Wu, Mengyou, Schwarz, Flavio, Siddiqui, Shoib S, Varki, Ajit, Marth, Jamey D, and Nizet, Victor
- Subjects
Medical Microbiology ,Biomedical and Clinical Sciences ,Biological Sciences ,Prevention ,Infectious Diseases ,Emerging Infectious Diseases ,Clinical Research ,Hematology ,Development of treatments and therapeutic interventions ,5.1 Pharmaceuticals ,Infection ,Adult ,Animals ,Antigens ,CD ,Antigens ,Differentiation ,B-Lymphocyte ,Bacterial Capsules ,Blood Bactericidal Activity ,Blood Platelets ,Female ,Glycocalyx ,Humans ,Male ,Mice ,Mice ,Knockout ,N-Acetylneuraminic Acid ,Platelet Activation ,Sialic Acid Binding Immunoglobulin-like Lectins ,Streptococcal Infections ,Streptococcus agalactiae ,Virulence Factors ,platelets ,group B Streptococcus ,innate immunity ,sialic acid ,Siglec - Abstract
Circulating platelets have important functions in thrombosis and in modulating immune and inflammatory responses. However, the role of platelets in innate immunity to bacterial infection is largely unexplored. While human platelets rapidly kill Staphylococcus aureus, we found the neonatal pathogen group B Streptococcus (GBS) to be remarkably resistant to platelet killing. GBS possesses a capsule polysaccharide (CPS) with terminal α2,3-linked sialic acid (Sia) residues that mimic a common epitope present on the human cell surface glycocalyx. A GBS mutant deficient in CPS Sia was more efficiently killed by human platelets, thrombin-activated platelet releasate, and synthetic platelet-associated antimicrobial peptides. GBS Sia is known to bind inhibitory Sia-recognizing Ig superfamily lectins (Siglecs) to block neutrophil and macrophage activation. We show that human platelets also express high levels of inhibitory Siglec-9 on their surface, and that GBS can engage this receptor in a Sia-dependent manner to suppress platelet activation. In a mouse i.v. infection model, antibody-mediated platelet depletion increased susceptibility to platelet-sensitive S. aureus but did not alter susceptibility to platelet-resistant GBS. Elimination of murine inhibitory Siglec-E partially reversed platelet suppression in response to GBS infection. We conclude that GBS Sia has dual roles in counteracting platelet antimicrobial immunity: conferring intrinsic resistance to platelet-derived antimicrobial components and inhibiting platelet activation through engagement of inhibitory Siglecs. We report a bacterial virulence factor for evasion of platelet-mediated innate immunity.
- Published
- 2019
40. Inhibition of Nuclear PTEN Tyrosine Phosphorylation Enhances Glioma Radiation Sensitivity through Attenuated DNA Repair
- Author
-
Ma, Jianhui, Benitez, Jorge A, Li, Jie, Miki, Shunichiro, de Albuquerque, Claudio Ponte, Galatro, Thais, Orellana, Laura, Zanca, Ciro, Reed, Rachel, Boyer, Antonia, Koga, Tomoyuki, Varki, Nissi M, Fenton, Tim R, Marie, Suely Kazue Nagahashi, Lindahl, Erik, Gahman, Timothy C, Shiau, Andrew K, Zhou, Huilin, DeGroot, John, Sulman, Erik P, Cavenee, Webster K, Kolodner, Richard D, Chen, Clark C, and Furnari, Frank B
- Subjects
Genetics ,Rare Diseases ,Brain Disorders ,Cancer ,Neurosciences ,Brain Cancer ,6.1 Pharmaceuticals ,Evaluation of treatments and therapeutic interventions ,Animals ,Brain Neoplasms ,Cell Nucleus ,DNA Repair ,Female ,Glioma ,Humans ,Male ,Mice ,PTEN Phosphohydrolase ,Phosphorylation ,Pyrimidines ,Rad51 Recombinase ,Radiation Tolerance ,Receptor ,Fibroblast Growth Factor ,Type 2 ,Tyrosine ,Xenograft Model Antitumor Assays ,DNA damage ,FGFR2 ,GBM ,PTEN ,ionizing radiation ,tyrosine phosphorylation ,Oncology and Carcinogenesis ,Oncology & Carcinogenesis - Abstract
Ionizing radiation (IR) and chemotherapy are standard-of-care treatments for glioblastoma (GBM) patients and both result in DNA damage, however, the clinical efficacy is limited due to therapeutic resistance. We identified a mechanism of such resistance mediated by phosphorylation of PTEN on tyrosine 240 (pY240-PTEN) by FGFR2. pY240-PTEN is rapidly elevated and bound to chromatin through interaction with Ki-67 in response to IR treatment and facilitates the recruitment of RAD51 to promote DNA repair. Blocking Y240 phosphorylation confers radiation sensitivity to tumors and extends survival in GBM preclinical models. Y240F-Pten knockin mice showed radiation sensitivity. These results suggest that FGFR-mediated pY240-PTEN is a key mechanism of radiation resistance and is an actionable target for improving radiotherapy efficacy.
- Published
- 2019
41. User-friendly bioorthogonal reactions click to explore glycan functions in complex biological systems
- Author
-
Xi Chen and Ajit Varki
- Subjects
Medicine - Published
- 2023
- Full Text
- View/download PDF
42. Paired Siglec receptors generate opposite inflammatory responses to a human‐specific pathogen
- Author
-
Schwarz, Flavio, Landig, Corinna S, Siddiqui, Shoib, Secundino, Ismael, Olson, Joshua, Varki, Nissi, Nizet, Victor, and Varki, Ajit
- Subjects
Genetics ,Nanotechnology ,Bioengineering ,Infectious Diseases ,2.1 Biological and endogenous factors ,Aetiology ,Infection ,Inflammatory and immune system ,Animals ,Cytokines ,Escherichia coli ,Escherichia coli Infections ,Humans ,Immune Evasion ,Inflammation ,Lectins ,Macrophages ,Membrane Proteins ,Mice ,Mice ,Transgenic ,Microbial Viability ,Sialic Acid Binding Immunoglobulin-like Lectins ,Sialic Acids ,Escherichia coli K1 ,Siglec ,molecular mimicry ,paired receptors ,polysialic acid ,Biological Sciences ,Information and Computing Sciences ,Medical and Health Sciences ,Developmental Biology - Abstract
Paired immune receptors display near-identical extracellular ligand-binding regions but have intracellular sequences with opposing signaling functions. While inhibitory receptors dampen cellular activation by recognizing self-associated molecules, the functions of activating counterparts are less clear. Here, we studied the inhibitory receptor Siglec-11 that shows uniquely human expression in brain microglia and engages endogenous polysialic acid to suppress inflammation. We demonstrated that the human-specific pathogen Escherichia coli K1 uses its polysialic acid capsule as a molecular mimic to engage Siglec-11 and escape killing. In contrast, engagement of the activating counterpart Siglec-16 increases elimination of bacteria. Since mice do not have paired Siglec receptors, we generated a model by replacing the inhibitory domain of mouse Siglec-E with the activating module of Siglec-16. Siglec-E16 enhanced proinflammatory cytokine expression and bacterial killing in macrophages and boosted protection against intravenous bacterial challenge. These data elucidate uniquely human interactions of a pathogen with Siglecs and support the long-standing hypothesis that activating counterparts of paired immune receptors evolved as a response to pathogen molecular mimicry of host ligands for inhibitory receptors.
- Published
- 2017
43. Studies on the Detection, Expression, Glycosylation, Dimerization, and Ligand Binding Properties of Mouse Siglec-E*
- Author
-
Siddiqui, Shoib, Schwarz, Flavio, Springer, Stevan, Khedri, Zahra, Yu, Hai, Deng, Lingquan, Verhagen, Andrea, Naito-Matsui, Yuko, Jiang, Weiping, Kim, Daniel, Zhou, Jie, Ding, Beibei, Chen, Xi, Varki, Nissi, and Varki, Ajit
- Subjects
Biochemistry and Cell Biology ,Biological Sciences ,1.1 Normal biological development and functioning ,Underpinning research ,Inflammatory and immune system ,Amino Acid Substitution ,Animals ,Antibodies ,Antigens ,CD ,Antigens ,Differentiation ,B-Lymphocyte ,Dendritic Cells ,Gene Expression Regulation ,Glycosylation ,Humans ,Macrophages ,Mice ,Mice ,Knockout ,Monocytes ,Mutagenesis ,Mutation ,Missense ,Neutrophils ,Protein Multimerization ,Protein Tyrosine Phosphatase ,Non-Receptor Type 6 ,Rats ,Rats ,Inbred Lew ,Sialic Acid Binding Immunoglobulin-like Lectins ,Siglec-9 ,Siglec-E ,cell signaling ,dimerization ,flow cytometry ,monoclonal antibody ,sialic acid ,Chemical Sciences ,Medical and Health Sciences ,Biochemistry & Molecular Biology ,Biological sciences ,Biomedical and clinical sciences ,Chemical sciences - Abstract
CD33-related Siglecs are a family of proteins widely expressed on innate immune cells. Binding of sialylated glycans or other ligands triggers signals that inhibit or activate inflammation. Immunomodulation by Siglecs has been extensively studied, but relationships between structure and functions are poorly explored. Here we present new data relating to the structure and function of Siglec-E, the major CD33-related Siglec expressed on mouse neutrophils, monocytes, macrophages, and dendritic cells. We generated nine new rat monoclonal antibodies specific to mouse Siglec-E, with no cross-reactivity to Siglec-F. Although all antibodies detected Siglec-E on transfected human HEK-293T cells, only two reacted with mouse bone marrow neutrophils by flow cytometry and on spleen sections by immunohistochemistry. Moreover, whereas all antibodies recognized Siglec-E-Fc on immunoblots, binding was dependent on intact disulfide bonds and N-glycans, and only two antibodies recognized native Siglec-E within spleen lysates. Thus, we further investigated the impact of Siglec-E homodimerization. Homology-based structural modeling predicted a cysteine residue (Cys-298) in position to form a disulfide bridge between two Siglec-E polypeptides. Mutagenesis of Cys-298 confirmed its role in dimerization. In keeping with the high level of 9-O-acetylation found in mice, sialoglycan array studies indicate that this modification has complex effects on recognition by Siglec-E, in relationship to the underlying structures. However, we found no differences in phosphorylation or SHP-1 recruitment between dimeric and monomeric Siglec-E expressed on HEK293A cells. Phylogenomic analyses predicted that only some human and mouse Siglecs form disulfide-linked dimers. Notably, Siglec-9, the functionally equivalent human paralog of Siglec-E, occurs as a monomer.
- Published
- 2017
44. Siglec-7 engagement by GBS β-protein suppresses pyroptotic cell death of natural killer cells
- Author
-
Fong, Jerry J, Tsai, Chih-Ming, Saha, Sudeshna, Nizet, Victor, Varki, Ajit, and Bui, Jack D
- Subjects
Biodefense ,Vaccine Related ,Prevention ,Emerging Infectious Diseases ,Infectious Diseases ,2.1 Biological and endogenous factors ,Underpinning research ,Aetiology ,1.1 Normal biological development and functioning ,Infection ,Inflammatory and immune system ,Good Health and Well Being ,Antigens ,Differentiation ,Myelomonocytic ,Cells ,Cultured ,DNA-Binding Proteins ,Humans ,Immunity ,Innate ,Inflammation Mediators ,Killer Cells ,Natural ,Lectins ,Pyroptosis ,natural killer cells ,Siglec ,group B Streptococcus ,pyroptosis ,inflammasome - Abstract
Natural killer (NK) cells are innate immune lymphocytes that recognize and destroy abnormal host cells, such as tumor cells or those infected by viral pathogens. To safely accomplish these functions, NK cells display activating receptors that detect stress molecules or viral ligands displayed at the cell surface, balanced by inhibitory receptors that bind to self-molecules. To date, such activating and inhibitory receptors on NK cells are not known to recognize bacterial determinants. Moreover, NK cell responses to direct interactions with extracellular bacteria are poorly explored. In this study, we observed the human neonatal pathogen group B Streptococcus (GBS) can directly engage human NK cells. The interaction was mediated through the B6N segment of streptococcal β-protein, binding to the inhibitory receptor Siglec-7 via its amino-terminal V-set domain. Unlike classical Siglec binding, the interaction is also independent of its sialic acid recognition property. In contrast to WT GBS, mutants lacking β-protein induced efficient pyroptosis of NK cells through the NLRP3 inflammasome, with production and secretion of the proinflammatory cytokine IL-1β and dissemination of the cytotoxic molecule granzyme B. We postulate that GBS evolved β-protein engagement of inhibitory human Siglec-7 to suppress the pyroptotic response of NK cells and thereby block recruitment of a broader innate immune response, i.e., by "silencing the sentinel."
- Published
- 2018
45. Human-like Cmah inactivation in mice increases running endurance and decreases muscle fatigability: implications for human evolution
- Author
-
Okerblom, Jonathan, Fletes, William, Patel, Hemal H, Schenk, Simon, Varki, Ajit, and Breen, Ellen C
- Subjects
Genetics ,Rare Diseases ,Musculoskeletal ,Animals ,Male ,Mice ,Mice ,Knockout ,Mixed Function Oxygenases ,Physical Conditioning ,Animal ,Running ,human ,evolution ,running ,hunting ,Biological Sciences ,Agricultural and Veterinary Sciences ,Medical and Health Sciences - Abstract
Compared to other primates, humans are exceptional long-distance runners, a feature that emerged in genus Homo approximately 2 Ma and is classically attributed to anatomical and physiological adaptations such as an enlarged gluteus maximus and improved heat dissipation. However, no underlying genetic changes have currently been defined. Two to three million years ago, an exon deletion in the CMP-Neu5Ac hydroxylase (CMAH) gene also became fixed in our ancestral lineage. Cmah loss in mice exacerbates disease severity in multiple mouse models for muscular dystrophy, a finding only partially attributed to differences in immune reactivity. We evaluated the exercise capacity of Cmah-/- mice and observed an increased performance during forced treadmill testing and after 15 days of voluntary wheel running. Cmah-/- hindlimb muscle exhibited more capillaries and a greater fatigue resistance in situ Maximal coupled respiration was also higher in Cmah null mice ex vivo and relevant differences in metabolic pathways were also noted. Taken together, these data suggest that CMAH loss contributes to an improved skeletal muscle capacity for oxygen use. If translatable to humans, CMAH loss could have provided a selective advantage for ancestral Homo during the transition from forest dwelling to increased resource exploration and hunter/gatherer behaviour in the open savannah.
- Published
- 2018
46. Polyclonal human antibodies against glycans bearing red meat-derived non-human sialic acid N-glycolylneuraminic acid are stable, reproducible, complex and vary between individuals: Total antibody levels are associated with colorectal cancer risk
- Author
-
Samraj, Annie N, Bertrand, Kimberly A, Luben, Robert, Khedri, Zahra, Yu, Hai, Nguyen, Dzung, Gregg, Christopher J, Diaz, Sandra L, Sawyer, Sherilyn, Chen, Xi, Eliassen, Heather, Padler-Karavani, Vered, Wu, Kana, Khaw, Kay-Tee, Willett, Walter, and Varki, Ajit
- Subjects
Biological Sciences ,Biomedical and Clinical Sciences ,Immunology ,Prevention ,Digestive Diseases ,Nutrition ,Cancer ,Colo-Rectal Cancer ,Clinical Research ,Adult ,Aged ,Antibodies ,Atherosclerosis ,Autoantigens ,Colorectal Neoplasms ,Diabetes Mellitus ,Type 2 ,Epitopes ,Female ,Humans ,Middle Aged ,N-Acetylneuraminic Acid ,Neuraminic Acids ,Polysaccharides ,Red Meat ,Risk Factors ,General Science & Technology - Abstract
BackgroundN-glycolylneuraminic acid (Neu5Gc) is a non-human red-meat-derived sialic acid immunogenic to humans. Neu5Gc can be metabolically incorporated into glycan chains on human endothelial and epithelial surfaces. This represents the first example of a "xeno-autoantigen", against which circulating human "xeno-autoantibodies" can react. The resulting inflammation ("xenosialitis") has been demonstrated in human-like Neu5Gc-deficient mice and contributed to carcinoma progression via antibody-mediated inflammation. Anti-Neu5Gc antibodies have potential as biomarkers for diseases associated with red meat consumption such as carcinomas, atherosclerosis, and type 2 diabetes.MethodsELISA assays measured antibodies against Neu5Gc or Neu5Gc-glycans in plasma or serum samples from the Nurses' Health Studies, the Health Professionals Follow-up Study, and the European Prospective Investigation into Cancer and Nutrition, including inter-assay reproducibility, stability with delayed sample processing, and within-person reproducibility over 1-3 years in archived samples. We also assessed associations between antibody levels and coronary artery disease risk (CAD) or red meat intake. A glycan microarray was used to detected antibodies against multiple Neu5Gc-glycan epitopes. A nested case-control study design assessed the association between total anti-Neu5Gc antibodies detected in the glycan array assay and the risk of colorectal cancer (CRC).ResultsELISA assays showed a wide range of anti-Neu5Gc responses and good inter-assay reproducibility, stability with delayed sample processing, and within-person reproducibility over time, but these antibody levels did not correlate with CAD risk or red meat intake. Antibodies against Neu5Gc alone or against individual Neu5Gc-bearing epitopes were also not associated with colorectal cancer (CRC) risk. However, a sialoglycan microarray study demonstrated positive association with CRC risk when the total antibody responses against all Neu5Gc-glycans were combined. Individuals in the top quartile of total anti-Neu5Gc IgG antibody concentrations had nearly three times the risk compared to those in the bottom quartile (Multivariate Odds Ratio comparing top to bottom quartile: 2.98, 95% CI: 0.80, 11.1; P for trend = 0.02).ConclusionsFurther work harnessing the utility of these anti-Neu5Gc antibodies as biomarkers in red meat-associated diseases must consider diversity in individual antibody profiles against different Neu5Gc-bearing glycans. Traditional ELISA assays for antibodies directed against Neu5Gc alone, or against specific Neu5Gc-glycans may not be adequate to define risk associations. Our finding of a positive association of total anti-Neu5Gc antibodies with CRC risk also warrants confirmation in larger prospective studies.
- Published
- 2018
47. Unravelling the specificity and mechanism of sialic acid recognition by the gut symbiont Ruminococcus gnavus.
- Author
-
Owen, C David, Tailford, Louise E, Monaco, Serena, Šuligoj, Tanja, Vaux, Laura, Lallement, Romane, Khedri, Zahra, Yu, Hai, Lecointe, Karine, Walshaw, John, Tribolo, Sandra, Horrex, Marc, Bell, Andrew, Chen, Xi, Taylor, Gary L, Varki, Ajit, Angulo, Jesus, and Juge, Nathalie
- Subjects
Goblet Cells ,Colon ,Cell Line ,Animals ,Mice ,Inbred C57BL ,Humans ,Ruminococcus ,N-Acetylneuraminic Acid ,Neuraminidase ,Glycoproteins ,Lactose ,Adhesins ,Bacterial ,Mucins ,Crystallography ,X-Ray ,Mutagenesis ,Site-Directed ,Computational Biology ,Symbiosis ,Catalytic Domain ,Protein Binding ,Substrate Specificity - Abstract
Ruminococcus gnavus is a human gut symbiont wherein the ability to degrade mucins is mediated by an intramolecular trans-sialidase (RgNanH). RgNanH comprises a GH33 catalytic domain and a sialic acid-binding carbohydrate-binding module (CBM40). Here we used glycan arrays, STD NMR, X-ray crystallography, mutagenesis and binding assays to determine the structure and function of RgNanH_CBM40 (RgCBM40). RgCBM40 displays the canonical CBM40 β-sandwich fold and broad specificity towards sialoglycans with millimolar binding affinity towards α2,3- or α2,6-sialyllactose. RgCBM40 binds to mucus produced by goblet cells and to purified mucins, providing direct evidence for a CBM40 as a novel bacterial mucus adhesin. Bioinformatics data show that RgCBM40 canonical type domains are widespread among Firmicutes. Furthermore, binding of R. gnavus ATCC 29149 to intestinal mucus is sialic acid mediated. Together, this study reveals novel features of CBMs which may contribute to the biogeography of symbiotic bacteria in the gut.
- Published
- 2017
48. Evolution of host adaptation in the Salmonella typhoid toxin.
- Author
-
Gao, Xiang, Deng, Lingquan, Stack, Gabrielle, Yu, Hai, Chen, Xi, Naito-Matsui, Yuko, Varki, Ajit, and Galán, Jorge E
- Subjects
Cell Line ,Animals ,Humans ,Mice ,Salmonella typhi ,Typhoid Fever ,Neuraminic Acids ,N-Acetylneuraminic Acid ,Polysaccharides ,Bacterial Proteins ,Transcription Factors ,Bacterial Toxins ,Endotoxins ,Virulence Factors ,Crystallography ,X-Ray ,Adaptation ,Physiological ,Binding Sites ,Amino Acid Sequence ,Models ,Molecular ,Male ,Glycomics ,HEK293 Cells ,Host Specificity ,Crystallography ,X-Ray ,Adaptation ,Physiological ,Models ,Molecular ,Microbiology ,Medical Microbiology - Abstract
The evolution of virulence traits is central for the emergence or re-emergence of microbial pathogens and for their adaptation to a specific host 1-5 . Typhoid toxin is an essential virulence factor of the human-adapted bacterial pathogen Salmonella Typhi 6,7 , the cause of typhoid fever in humans 8-12 . Typhoid toxin has a unique A2B5 architecture with two covalently linked enzymatic 'A' subunits, PltA and CdtB, associated with a homopentameric 'B' subunit made up of PltB, which has binding specificity for the N-acetylneuraminic acid (Neu5Ac) sialoglycans 6,13 prominently present in humans 14 . Here, we examine the functional and structural relationship between typhoid toxin and ArtAB, an evolutionarily related AB5 toxin encoded by the broad-host Salmonella Typhimurium 15 . We find that ArtA and ArtB, homologues of PltA and PltB, can form a functional complex with the typhoid toxin CdtB subunit after substitution of a single amino acid in ArtA, while ArtB can form a functional complex with wild-type PltA and CdtB. We also found that, after addition of a single-terminal Cys residue, a CdtB homologue from cytolethal distending toxin can form a functional complex with ArtA and ArtB. In line with the broad host specificity of S. Typhimurium, we found that ArtB binds human glycans, terminated in N-acetylneuraminic acid, as well as glycans terminated in N-glycolylneuraminic acid (Neu5Gc), which are expressed in most other mammals 14 . The atomic structure of ArtB bound to its receptor shows the presence of an additional glycan-binding site, which broadens its binding specificity. Despite equivalent toxicity in vitro, we found that the ArtB/PltA/CdtB chimaeric toxin exhibits reduced lethality in an animal model, indicating that the host specialization of typhoid toxin has optimized its targeting mechanisms to the human host. This is a remarkable example of a toxin evolving to broaden its enzymatic activities and adapt to a specific host.
- Published
- 2017
49. Human‐specific polymorphic pseudogenization of SIGLEC12 protects against advanced cancer progression
- Author
-
Shoib S Siddiqui, Michael Vaill, Raymond Do, Naazneen Khan, Andrea L Verhagen, Wu Zhang, Heinz‐Josef Lenz, Teresa L Johnson‐Pais, Robin J Leach, Gary Fraser, Charles Wang, Gen‐Sheng Feng, Nissi Varki, and Ajit Varki
- Subjects
advanced carcinoma ,dot blot ,immunohistochemistry ,pseudogenization ,SIGLEC12 ,Biology (General) ,QH301-705.5 - Abstract
Abstract Compared with our closest living evolutionary cousins, humans appear unusually prone to develop carcinomas (cancers arising from epithelia). The SIGLEC12 gene, which encodes the Siglec‐XII protein expressed on epithelial cells, has several uniquely human features: a fixed homozygous missense mutation inactivating its natural ligand recognition property; a polymorphic frameshift mutation eliminating full‐length protein expression in ~60%–70% of worldwide human populations; and, genomic features suggesting a negative selective sweep favoring the pseudogene state. Despite the loss of canonical sialic acid binding, Siglec‐XII still recruits Shp2 and accelerates tumor growth in a mouse model. We hypothesized that dysfunctional Siglec‐XII facilitates human carcinoma progression, correlating with known tumorigenic signatures of Shp2‐dependent cancers. Immunohistochemistry was used to detect Siglec‐XII expression on tissue microarrays. PC‐3 prostate cancer cells were transfected with Siglec‐XII and transcription of genes enriched with Siglec‐XII was determined. Genomic SIGLEC12 status was determined for four different cancer cohorts. Finally, a dot blot analysis of human urinary epithelial cells was established to determine the Siglec‐XII expressors versus non‐expressors. Forced expression in a SIGLEC12 null carcinoma cell line enriched transcription of genes associated with cancer progression. While Siglec‐XII was detected as expected in ~30%–40% of normal epithelia, ~80% of advanced carcinomas showed strong expression. Notably, >80% of late‐stage colorectal cancers had a functional SIGLEC12 allele, correlating with overall increased mortality. Thus, advanced carcinomas are much more likely to occur in individuals whose genomes have an intact SIGLEC12 gene, likely because the encoded Siglec‐XII protein recruits Shp2‐related oncogenic pathways. The finding has prognostic, diagnostic, and therapeutic implications.
- Published
- 2021
- Full Text
- View/download PDF
50. The spillover effect of product recalls on competitors’ market value: The role of corporate product reliability
- Author
-
Liu, Dong and Varki, Sajeev
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.