145 results on '"Varki N"'
Search Results
2. The Ashwell-Morell receptor mitigates the lethal coagulopathy of sepsis: 630
- Author
-
Grewal, P. K., Uchiyama, S., Ditto, D., Varki, N., Le, D. T., Nizet, V., and Marth, J. D.
- Published
- 2009
3. Broad Antitumor and Antiangiogenic Activities of AG3340, a Potent and Selective MMP Inhibitor Undergoing Advanced Oncology Clinical Trials
- Author
-
SHALINSKY, D. R., BREKKEN, J., ZOU, H., MCDERMOTT, C. D., FORSYTH, P., EDWARDS, D., MARGOSIAK, S., BENDER, S., TRUITT, G., WOOD, A., VARKI, N. M., and APPELT, K.
- Published
- 1999
4. 12PLECTIN GALACTOSIDE-BINDING SOLUBLE 3 BINDING PROTEIN (LGALS3BP) IS A CANCER-ASSOCIATED LIGAND FOR INHIBITORY SIGLECS
- Author
-
Läubli, H., Stanczak, M., Alisson-Silva, F., Varki, N., and Varki, A.
- Subjects
respiratory system - Abstract
Tumor cells subvert the control of the immune system by downregulation of their antigenicity and production of an immunosuppressive microenvironment including the upregulation and engagement of inhibitory receptors on immune cells. Therapeutic strategies have demonstrated that the immune system can be reactivated and control established cancers by blocking inhibitory receptors on immune cells such CTLA-4 and PD1. While such activation of the immune system is successful in some patients, many patients still show cancer progression after some time. Thus, the definition of new targetable immunomodulatory pathways is needed to improve the outcome in those patients. Recent evidence suggests that sialic acid dependent ligands on tumor cells can engage inhibitory sialic acid binding immunoglobulin-like lectins (Siglecs) on NK cells and cells of the myelomonocytic lineage and thereby facilitate evasion of immune-mediated killing. Moreover, the presence of a natural variant of Siglec-9 with reduced binding capacity to sialic acid dependent ligands in patients with non-small cell lung cancer improved the two year survival in a retrospective multivariate analysis. Here we identify a novel cancer-associated ligand for immuno-inhibitory Siglecs by affinity chromatography and subsequent proteomic analysis. LectinGalactoside-Binding Soluble 3 Binding Protein (LGALS3BP) bound to various inhibitory Siglecs including Siglec-5, Siglec-9 and Siglec-10 with high affinity. LGALS3BP was previously found to be upregulated in various carcinomas such as breast, colorectal, prostate and lung cancer and linked to advanced stage and poor prognosis. The exact function during cancer progression, however, was not yet defined. Our findings provide a novel insight into how LGALS3BP could promote immune evasion by inhibiting immune cell activation through engagement of Siglecs and defines LGALS3BP-Siglec interactions as potential novel target to interfere with cancer progression and reactivate the immune system against carcinomas. Disclosure: All authors have declared no conflicts of interest
- Published
- 2017
5. Program and abstracts for the 2011 Meeting of the Society for Glycobiology
- Author
-
Hollingsworth, MT, Hart, GW, Paulson, JC, Stansell, E, Canis, K, Huang, IC, Panico, M, Morris, H, Haslam, S, Farzan, M, Dell, A, Desrosiers, R, von Itzstein, M, Matroscovich, M, Luther, KB, Hülsmeier, AJ, Schegg, B, Hennet, T, Nycholat, C, McBride, R, Ekiert, D, Xu, R, Peng, W, Razi, N, Gilbert, M, Wakarchuk, W, Wilson, IA, Gahlay, G, Geisler, C, Aumiller, JJ, Moremen, K, Steel, J, Labaer, J, Jarvis, DL, Drickamer, K, Taylor, M, Nizet, V, Rabinovich, G, Lewis, C, Cobb, B, Kawasaki, N, Rademacher, C, Chen, W, Vela, J, Maricic, I, Crocker, P, Kumar, V, Kronenberg, M, Paulson, J, Glenn, K, Mallinger, A, Wen, H, Srivastava, L, Tundup, S, Harn, D, Menon, AK, Yamaguchi, Y, Mkhikian, H, Grigorian, A, Li, C, Chen, HL, Newton, B, Zhou, RW, Beeton, C, Torossian, S, Tatarian, GG, Lee, SU, Lau, K, Walker, E, Siminovitch, KA, Chandy, KG, Yu, Z, Dennis, JW, Demetriou, M, Pandey, MS, Baggenstoss, BA, Washburn, JL, Weigel, PH, Chen, CI, Keusch, JJ, Klein, D, Hofsteenge, J, Gut, H, Szymanski, C, Feldman, M, Schaffer, C, Gao, Y, Strum, S, Liu, B, Schutzbach, JS, Druzhinina, TN, Utkina, NS, Torgov, VI, Szarek, WA, Wang, L, Brockhausen, I, Hitchen, P, Peyfoon, E, Meyer, B, Albers, SV, Chen, C, Newburg, DS, Jin, C, Dinglasan, RD, Beverley, SM, Guo, H, Novozhilova, N, Hickerson, S, Elnaiem, DE, Sacks, D, Turco, SJ, McKay, D, Castro, E, Takahashi, H, Straus, AH, Stalnaker, SH, Live, D, Boons, GJ, Wells, L, Stuart, R, Aoki, K, Boccuto, L, Zhang, Q, Wang, H, Bartel, F, Fan, X, Saul, R, Chaubey, A, Yang, X, Steet, R, Schwartz, C, Tiemeyer, M, Pierce, M, Kraushaar, DC, Condac, E, Nakato, H, Nishihara, S, Sasaki, N, Hirano, K, Nasirikenari, M, Collins, CC, Lau, JT, Devarapu, SK, Jeyaweerasinkam, S, Albiez, RS, Kiessling, L, Gu, J, Clark, GF, Gagneux, P, Ulm, C, Mahavadi, P, Müller, S, Rinné, S, Geyer, H, Gerardy-Schahn, R, Mühlenhoff, M, Günther, A, Geyer, R, Galuska, SP, Shibata, T, Sugihara, K, Nakayama, J, Fukuda, M, Fukuda, MN, Ishikawa, A, Terao, M, Kimura, A, Kato, A, Katayama, I, Taniguchi, N, Miyoshi, E, Aderem, A, Yoneyama, T, Angata, K, Bao, X, Chanda, S, Lowe, J, Sonon, R, Ishihara, M, Talabnin, K, Wang, Z, Black, I, Naran, R, Heiss, C, Azadi, P, Hurum, D, Rohrer, J, Balland, A, Valliere-Douglass, J, Kodama, P, Mujacic, M, Eakin, C, Brady, L, Wang, WC, Wallace, A, Treuheit, M, Reddy, P, Schuman, B, Fisher, S, Borisova, S, Coates, L, Langan, P, Evans, S, Yang, SJ, Zhang, H, Hizal, DB, Tian, Y, Sarkaria, V, Betenbaugh, M, Lütteke, T, Agravat, S, Cholleti, S, Morris, T, Saltz, J, Song, X, Cummings, R, Smith, D, Hofhine, T, Nishida, C, Mialy, R, Sophie, D, Sebastien, F, Patricia, C, Eric, S, Stephane, H, Mokros, D, Joosten, RP, Dominik, A, Vriend, G, Nguyen, LD, Martinez, J, Hinderlich, S, Reissig, HU, Reutter, W, Fan, H, Saenger, W, Moniot, S, Asada, H, Nakahara, T, Miura, Y, Stevenson, T, Yamazaki, T, De Castro, C, Burr, T, Lanzetta, R, Molinaro, A, Parrilli, M, Sule, S, Gerken, TA, Revpredo, L, Thome, J, Cardenas, G, Almeida, I, Leung, MY, Yan, S, Paschinger, K, Bleuler-Martinez, S, Jantsch, V, Wilson, I, Yoshimura, Y, Adlercreutz, D, Mannerstedt, K, Wakarchuk, WW, Dovichi, NJ, Hindsgaul, O, Palcic, MM, Chandrasekaran, A, Bharadwaj, R, Deng, K, Adams, P, Singh, A, Datta, A, Konasani, V, Imamura, A, Lowry, T, Scaman, C, Zhao, Y, Zhou, YD, Yang, K, Zhang, XL, Leymarie, N, Hartshorn, K, White, M, Cafarella, T, Seaton, B, Rynkiewicz, M, Zaia, J, Acosta-Blanco, I, Ortega-Francisco, S, Dionisio-Vicuña, M, Hernandez-Flores, M, Fuentes-Romero, L, Newburg, D, Soto-Ramirez, LE, Ruiz-Palacios, G, Viveros-Rogel, M, Tong, C, Li, W, Kong, L, Qu, M, Jin, Q, Lukyanov, P, Zhang, W, Chicalovets, I, Molchanova, V, Wu, AM, Liu, JH, Yang, WH, Nussbaum, C, Grewal, PK, Sperandio, M, Marth, JD, Yu, R, Usuki, S, Wu, HC, O'Brien, D, Piskarev, V, Ramadugu, SK, Kashyap, HK, Ghirlanda, G, Margulis, C, Brewer, C, Gomery, K, Müller-Loennies, S, Brooks, CL, Brade, L, Kosma, P, Di Padova, F, Brade, H, Evans, SV, Asakawa, K, Kawakami, K, Kushi, Y, Suzuki, Y, Nozaki, H, Itonori, S, Malik, S, Lebeer, S, Petrova, M, Balzarini, J, Vanderleyden, J, Naito-Matsui, Y, Takematsu, H, Murata, K, Kozutsumi, Y, Subedi, GP, Satoh, T, Hanashima, S, Ikeda, A, Nakada, H, Sato, R, Mizuno, M, Yuasa, N, Fujita-Yamaguchi, Y, Vlahakis, J, Nair, DG, Wang, Y, Allingham, J, Anastassiades, T, Strachan, H, Johnson, D, Orlando, R, Harenberg, J, Haji-Ghassemi, O, Mackenzie, R, Lacerda, T, Toledo, M, Straus, A, Takahashi, HK, Woodrum, B, Ruben, M, O'Keefe, B, Samli, KN, Yang, L, Woods, RJ, Jones, MB, Maxwell, J, Song, EH, Manganiello, M, Chow, YH, Convertine, AJ, Schnapp, LM, Stayton, PS, Ratner, DM, Yegorova, S, Rodriguez, MC, Minond, D, Jiménez-Barbero, J, Calle, L, Ardá, A, Gabius, HJ, André, S, Martinez-Mayorga, K, Yongye, AB, Cudic, M, Ali, MF, Chachadi, VB, Cheng, PW, Kiwamoto, T, Na, HJ, Brummet, M, Finn, MG, Hong, V, Polonskaya, Z, Bovin, NV, Hudson, S, Bochner, B, Gallogly, S, Krüger, A, Hanley, S, Gerlach, J, Hogan, M, Ward, C, Joshi, L, Griffin, M, Demarco, C, Deveny, R, Aggeler, R, Hart, C, Nyberg, T, Agnew, B, Akçay, G, Ramphal, J, Calabretta, P, Nguyen, AD, Kumar, K, Eggers, D, Terrill, R, d'Alarcao, M, Ito, Y, Vela, JL, Matsumura, F, Hoshino, H, Lee, H, Kobayashi, M, Borén, T, Jin, R, Seeberger, PH, Pitteloud, JP, Cudic, P, Von Muhlinen, N, Thurston, T, von Muhlinen, N, Wandel, M, Akutsu, M, Foeglein, AÁ, Komander, D, Randow, F, Maupin, K, Liden, D, Haab, B, Dam, TK, Brown, RK, Wiltzius, M, Jokinen, M, Andre, S, Kaltner, H, Bullen, J, Balsbaugh, J, Neumann, D, Hardie, G, Shabanowitz, J, Hunt, D, Hart, G, Mi, R, Ding, X, Van Die, I, Chapman, AB, Cummings, RD, Ju, T, Aryal, R, Ashley, J, Feng, X, Hanover, JA, Wang, P, Keembiyehetty, C, Ghosh, S, Bond, M, Krause, M, Love, D, Radhakrishnan, P, Grandgenet, PM, Mohr, AM, Bunt, SK, Yu, F, Hollingsworth, MA, Ethen, C, Machacek, M, Prather, B, Wu, Z, Kotu, V, Zhao, P, Zhang, D, van der Wel, H, Johnson, JM, West, CM, Abdulkhalek, S, Amith, SR, Jayanth, P, Guo, M, Szewczuk, M, Ohtsubo, K, Chen, M, Olefsky, J, Marth, J, Zapater, J, Foley, D, Colley, K, Kawashima, N, Fujitani, N, Tsuji, D, Itoh, K, Shinohara, Y, Nakayama, K, Zhang, L, Ten Hagen, K, Koren, S, Yehezkel, G, Cohen, L, Kliger, A, Khalaila, I, Finkelstein, E, Parker, R, Kohler, J, Sacoman, J, Badish, L, Hollingsworth, R, Tian, E, Hoffman, M, Hou, X, Tashima, Y, Stanley, P, Kizuka, Y, Kitazume, S, Yoshida, M, Kunze, A, Nasir, W, Bally, M, Hook, F, Larson, G, Mahan, A, Alter, G, Zeidan, Q, Copeland, R, Pokrovskaya, I, Willett, R, Smith, R, Morelle, W, Kudlyk, T, Lupashin, V, Vasudevan, D, Takeuchi, H, Majerus, E, Haltiwanger, RS, Boufala, S, Lee, YA, Min, D, Kim, SH, Shin, MH, Gesteira, T, Pol-Fachin, L, Coulson-Thomas, VJ, Verli, H, Nader, H, Liu, X, Yang, P, Thoden, J, Holden, H, Tytgat, H, Sánchez-Rodríguez, A, Schoofs, G, Verhoeven, T, De Keersmaecker, S, Marchal, K, Ventura, V, Sarah, N, Joann, P, Ding, Y, Jarrell, K, Cook, MC, Gibeault, S, Filippenko, V, Ye, Q, Wang, J, Kunkel, JP, Arteaga-Cabello, FJ, Arciniega-Fuentes, MT, McCoy, J, Ruiz-Palacios, GM, Francoleon, D, Loo, RO, Loo, J, Ytterberg, AJ, Kim, U, Gunsalus, R, Costello, C, Soares, R, Assis, R, Ibraim, I, Noronha, F, De Godoy, AP, Bale, MS, Xu, Y, Brown, K, Blader, I, West, C, Chen, S, Ye, X, Xue, C, Li, G, Yu, G, Yin, L, Chai, W, Gutierrez-Magdaleno, G, Tan, C, Wu, D, Li, Q, Hu, H, Ye, M, Liu, D, Mink, W, Kaese, P, Fujiwara, M, Uchimura, K, Sakai, Y, Nakada, T, Mabashi-Asazuma, H, Toth, AM, Scott, DW, Chacko, BK, Patel, RP, Batista, F, Mercer, N, Ramakrishnan, B, Pasek, M, Boeggeman, E, Verdi, L, Qasba, PK, Tran, D, Lim, JM, Liu, M, Mo, KF, Kirby, P, Yu, X, Lin, C, Costello, CE, Akama, TO, Nakamura, T, Huang, Y, Shi, X, Han, L, Yu, SH, Zhang, Z, Knappe, S, Till, S, Nadia, I, Catarello, J, Quinn, C, Julia, N, Ray, J, Tran, T, Scheiflinger, F, Szabo, C, Dockal, M, Niimi, S, Hosono, T, Michikawa, M, Kannagi, R, Takashima, S, Amano, J, Nakamura, N, Kaneda, E, Nakayama, Y, Kurosaka, A, Takada, W, Matsushita, T, Hinou, H, Nishimura, S, Igarashi, K, Abe, H, Mothere, M, Leonhard-Melief, C, Johnson, H, Nagy, T, Nairn, A, Rosa, MD, Porterfield, M, Kulik, M, Dalton, S, Pierce, JM, Hansen, SF, McAndrew, R, Degiovanni, A, McInerney, P, Pereira, JH, Hadi, M, Scheller, HV, Barb, A, Prestegard, J, Zhang, S, Jiang, J, Tharmalingam, T, Pluta, K, McGettigan, P, Gough, R, Struwe, W, Fitzpatrick, E, Gallagher, ME, Rudd, PM, Karlsson, NG, Carrington, SD, Katoh, T, Panin, V, Gelfenbeyn, K, Freire-de-Lima, L, Handa, K, Hakomori, SI, Bielik, AM, McLeod, E, Landry, D, Mendoza, V, Guthrie, EP, Mao, Y, Wang, X, Moremen, KW, Meng, L, Ramiah, AP, Gao, Z, Johnson, R, Xiang, Y, Rosa, MDEL, Wu, SC, Gilbert, HJ, Karaveg, K, Chen, L, Wang, BC, Mast, S, Sun, B, Fulton, S, Kimzey, M, Pourkaveh, S, Minalla, A, Haxo, T, Wegstein, J, Murray, AK, Nichols, RL, Giannini, S, Grozovsky, R, Begonja, AJ, Hoffmeister, KM, Suzuki-Anekoji, M, Suzuki, A, Yu, SY, Khoo, KH, van Alphen, L, Fodor, C, Wenzel, C, Ashmus, R, Miller, W, Stahl, M, Stintzi, A, Lowary, T, Wiederschain, G, Saba, J, Zumwalt, A, Meitei, NS, Apte, A, Viner, R, Gandy, M, Debowski, A, Stubbs, K, Witzenman, H, Pandey, D, Repnikova, E, Nakamura, M, Islam, R, Kc, N, Caster, C, Chaubard, JL, Krishnamurthy, C, Hsieh-Wilson, L, Pranskevich, J, Rangarajan, J, Guttman, A, Szabo, Z, Karger, B, Chapman, J, Chavaroche, A, Bionda, N, Fields, G, Jacob, F, Tse, BW, Guertler, R, Nixdorf, S, Hacker, NF, Heinzelmann-Schwarz, V, Yang, F, Kohler, JJ, Losfeld, ME, Ng, B, Freeze, HH, He, P, Wondimu, A, Liu, Y, Zhang, Y, Su, Y, Ladisch, S, Grewal, P, Mann, C, Ditto, D, Lardone, R, Le, D, Varki, N, Kulinich, A, Kostjuk, O, Maslak, G, Pismenetskaya, I, Shevtsova, A, Takeishi, S, Okudo, K, Moriwaki, K, Terao, N, Kamada, Y, Kuroda, S, Li, Y, Peiris, D, Markiv, A, Dwek, M, Adamczyk, B, Thanabalasingham, G, Huffman, J, Kattla, J, Novokmet, M, Rudan, I, Gloyn, A, Hayward, C, Reynolds, R, Hansen, T, Klimes, I, Njolstad, P, Wilson, J, Hastie, N, Campbell, H, McCarthy, M, Rudd, P, Owen, K, Lauc, G, Wright, A, Goletz, S, Stahn, R, Danielczyk, A, Baumeister, H, Hillemann, A, Löffler, A, Stöckl, L, Jahn, D, Bahrke, S, Flechner, A, Schlangstedt, M, Karsten, U, Goletz, C, Mikolajczyk, S, Ulsemer, P, Gao, N, Cline, A, Flanagan-Steet, H, Sadler, KC, Lehrman, MA, Coulson-Thomas, YM, Gesteira, TF, Mader, AM, Waisberg, J, Pinhal, MA, Friedl, A, Toma, L, Nader, HB, Mbua, EN, Johnson, S, Wolfert, M, Dimitrievska, S, Huizing, M, Niklason, L, Perdivara, I, Petrovich, R, Tokar, EJ, Waalkes, M, Fraser, P, Tomer, K, Chu, J, Rosa, S, Mir, A, Lehrman, M, Sadler, K, Lauer, M, Hascall, V, Calabro, A, Cheng, G, Swaidani, S, Abaddi, A, Aronica, M, Yuzwa, S, Shan, X, Macauley, M, Clark, T, Skorobogatko, Y, Vosseller, K, Vocadlo, D, Banerjee, A, Baksi, K, Banerjee, D, Melcher, R, Kraus, I, Moeller, D, Demmig, S, Rogoll, D, Kudlich, T, Scheppach, W, Scheurlen, M, Hasilik, A, Steirer, L, Lee, J, Moe, G, Troy, FA, Wang, F, Xia, B, Wang, B, Yi, S, Yu, H, Suzuki, M, Kobayashi, T, Sato, Y, Zhou, H, Briscoe, A, Lee, R, Wolfert, MA, Matsumoto, Y, Hamamura, K, Yoshida, T, Akita, K, Okajima, T, Furukawa, K, Urano, T, Ruhaak, LR, Miyamoto, S, and Lebrilla, CB
- Subjects
Embryogenesis ,Cancer screening ,Cancer research ,medicine ,Cell migration ,Neural cell adhesion molecule ,Biology ,medicine.disease ,Biochemistry ,Metastasis - Abstract
Cell surface mucins configure the cell surface by presenting extended protein backbones that are heavily O-glycosylated. The glycopeptide structures establish physicochemical properties at the cell surface that enable and block the formation of biologically important molecular complexes. Some mucins, such as MUC1, associate with receptor tyrosine kinases and other cell surface receptors, and engage in signal transduction in order to communicate information regarding conditions at the cell surface to the nucleus. In that context, the MUC1 cytoplasmic tail (MUC1CT) receives phosphorylation signals from receptor tyrosine kinases and serine/threonine kinases, which enables its association with different signaling complexes that conduct these signals to the nucleus and perhaps other subcellular organelles. We have detected the MUC1CT at promoters of over 500 genes, in association with several different transcription factors, and have shown that promoter occupancy can vary under different growth factor conditions. However, the full biochemical nature of the nuclear forms of MUC1 and its function at these promoter regions remain undefined. I will present evidence that nuclear forms of the MUC1CT include extracellular and cytoplasmic tail domains. In addition, I will discuss evidence for a hypothesis that the MUC1CT possesses a novel catalytic function that enables remodeling of the transcription factor occupancy of promoters, and thereby engages in regulation of gene expression.
- Published
- 2016
6. IL-1 beta and TNF-alpha upregulate angiotensin II type 1 (AT(1)) receptors on cardiac fibroblasts and are associated with increased AT(1) density in the post-MI heart
- Author
-
Gurantz, D, Cowling, R T, Varki, N, Frikovsky, E, Moore, C D, and Greenberg, Barry H
- Subjects
post-myocardial infarction remodeling ,interleukin 1 beta (IL-1 beta) ,receptor ,cardiac fibroblast ,AT(1) ,tumor necrosis factor-alpha TNF-alpha - Abstract
Angiotensin (Ang) II plays an important role in post-myocardial infarction (MI) cardiac remodeling. The Ang II type I (AT(1)) receptor which mediates most Ang II effects is upregulated on non-myocytes in the post-MI heart. We have shown that pro-inflammatory cytokines increase AT(1) receptor density on cardiac fibroblasts through a mechanism involving NF-kappa B activation. This study examines the in vitro kinetics of tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 beta (IL-1 beta) induced AT(1) receptor upregulation in neonatal rat cardiac fibroblasts and assesses temporal and spatial associations between the appearance of these agents and increased AT(1) receptor density post-MI. The results show that IL-1 beta more rapidly induces AT(1) receptor upregulation than does TNF-alpha, an effect that can be mimicked by a NF-kappa B-dependent luciferase reporter gene. Moreover, the effects of these pro-inflammatory cytokines are additive. Using immunohistochemistry in the post-MI rat heart we found strong temporal and spatial correlations between TNF-alpha, IL-1 beta and AT(1) receptor proteins in the peri-infarction (PI) zone in fibroblasts and macrophages. Labeling intensity for the cytokines and the AT(1) receptor increased from 1 to 7 days post-MI in the PI zone in conjunction with replacement scar formation. This labeling persisted in non-myocytes bordering the scar for up to 83 days post-MI. These findings suggest that IL-1 beta and TNF-alpha act coordinately to increase AT(1) receptor density on non-myocytes in the post-MI heart and that this effect may contribute to extracellular matrix remodeling and fibrosis. (c) 2005 Published by Elsevier Ltd.
- Published
- 2005
7. 12P - Lectin Galactoside-Binding Soluble 3 Binding Protein (LGALS3BP) is a Cancer-Associated Ligand for Inhibitory Siglecs
- Author
-
Läubli, H., Stanczak, M., Alisson-Silva, F., Varki, N., and Varki, A.
- Published
- 2014
- Full Text
- View/download PDF
8. Effects of ACE2 inhibition in the post-myocardial infarction heart.
- Author
-
Kim MA, Yang D, Kida K, Molotkova N, Yeo SJ, Varki N, Iwata M, Dalton ND, Peterson KL, Siems WE, Walther T, Cowling RT, Kjekshus J, Greenberg B, Kim, Myung-A, Yang, Dongheon, Kida, Keisuke, Molotkova, Natalia, Yeo, Seon Ju, and Varki, Nissi
- Abstract
Background: There is evidence that angiotensin-converting enzyme 2 (ACE2) is cardioprotective. To assess this in the post-myocardial infarction (MI) heart, we treated adult male Sprague-Dawley rats with either placebo (PL) or C16, a selective ACE2 inhibitor, after permanent coronary artery ligation or sham operation.Methods and Results: Coronary artery ligation resulting in MI between 25% to 50% of the left ventricular (LV) circumference caused substantial cardiac remodeling. Daily C16 administration from postoperative days 2 to 28 at a dose that inhibited myocardial ACE2 activity was associated with a significant increase in MI size and reduction in LV % fractional shortening. Treatment with C16 did not significantly affect post-MI increases in LV end-diastolic dimension but did inhibit increases in wall thickness and fibrosis in non-infarcted LV. On postoperative day 7, C16 had no significant effect on the increased level of apoptosis in the infarct and border zones nor did it significantly affect capillary density surrounding the MI. It did, however, significantly reduce the number of c-kit(+) cells in the border region.Conclusions: These findings support the notion that ACE2 exerts cardioprotective effects by preserving jeopardized cardiomyocytes in the border zone. The reduction in hypertrophy and fibrosis with C16, however, suggests that ACE2 activity has diverse effects on post-MI remodeling. [ABSTRACT FROM AUTHOR]- Published
- 2010
- Full Text
- View/download PDF
9. Immunohistochemical identification of tissue factor in solid tumors.
- Author
-
Callander, Natalie S., Varki, Nissi, Vijaya Rao, L. Mohan, Callander, N S, Varki, N, and Rao, L V
- Published
- 1992
- Full Text
- View/download PDF
10. Two human tumors with high basement-membrane-producing potential.
- Author
-
Barsky, Sanford H., Layfield, Lester, Varki, Nissi, Bhuta, Sunita, Barsky, S H, Layfield, L, Varki, N, and Bhuta, S
- Published
- 1988
- Full Text
- View/download PDF
11. Lymphocyte-mediated alopecia in C57BL/6 mice following successful immunotherapy for melanoma
- Author
-
Becker, J.C., Varki, N., Brocker, E.B., and Reisfeld, R.A.
- Subjects
Immunotherapy -- Physiological aspects ,Baldness -- Physiological aspects -- Complications and side effects -- Care and treatment ,Alopecia -- Physiological aspects -- Complications and side effects -- Care and treatment ,Melanoma -- Care and treatment -- Complications and side effects ,Health ,Care and treatment ,Physiological aspects ,Complications and side effects - Abstract
Lymphocyte-Mediated Alopecia in C57BL/6 Mice Following Successful Immunotherapy for Melanoma.' Journal of Investigative Dermatology, October 1996;107(4):627-632. According to the authors' abstract of an article published in Journal of Investigative Dermatology, [...]
- Published
- 1996
12. 223Embryonic stem cells survive and proliferate after intra-peritoneal in utero transplantation and produce teratocarcinomas
- Author
-
Nedelcu, E., Srivastava, A., Varki, N., Assatourians, G., and Carrier, E.
- Published
- 2003
- Full Text
- View/download PDF
13. 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
14. Chromogranin a epitopes: Clues from synthetic peptides and peptide mapping
- Author
-
Gill, B.M., Barbosa, J.A., Hogue-Angeletti, R., Varki, N., and O'Connor, D.T.
- Published
- 1992
- Full Text
- View/download PDF
15. Human-specific elimination of epithelial Siglec-XII suppresses the risk of inflammation-driven colorectal cancers.
- Author
-
Cuello HA, Sinha S, Verhagen AL, Varki N, Varki A, and Ghosh P
- Subjects
- Animals, Humans, Mice, Lectins genetics, Lectins metabolism, Mice, Knockout, Sialic Acid Binding Immunoglobulin-like Lectins genetics, Sialic Acid Binding Immunoglobulin-like Lectins metabolism, Membrane Proteins genetics, Membrane Proteins metabolism, Colorectal Neoplasms genetics, Colorectal Neoplasms pathology, Colorectal Neoplasms metabolism, Inflammation genetics, Inflammation metabolism
- Abstract
Carcinomas are common in humans but rare among closely related "great apes." Plausible explanations, including human-specific genomic alterations affecting the biology of sialic acids, are proposed, but causality remains unproven. Here, an integrated evolutionary genetics-phenome-transcriptome approach studied the role of SIGLEC12 gene (encoding Siglec-XII) in epithelial transformation and cancer. Exogenous expression of the protein in cell lines and genetically engineered mice recapitulated approximately 30% of the human population in whom the protein is expressed in a form that cannot bind ligand because of a fixed, homozygous, human-universal missense mutation. Siglec-XII-null cells/mice recapitulated the remaining approximately 70% of the human population in whom an additional polymorphic frameshift mutation eliminates the entire protein. Siglec-XII expression drove several pro-oncogenic phenotypes in cell lines and increased tumor burden in mice challenged with chemical carcinogen and inflammation. Transcriptomic studies yielded a 29-gene signature of Siglec-XII-positive disease and when used as a computational tool for navigating human data sets, pinpointed with surprising precision that SIGLEC12 expression (model) recapitulates a very specific type of colorectal carcinomas (disease) that is associated with mismatch-repair defects and inflammation, disproportionately affects European Americans, and carries a favorable prognosis. They revealed a hitherto-unknown evolutionary genetic mechanism for an ethnic/environmental predisposition of carcinogenesis.
- Published
- 2024
- Full Text
- View/download PDF
16. Comparative physiological anthropogeny: exploring molecular underpinnings of distinctly human phenotypes.
- Author
-
Vaill M, Kawanishi K, Varki N, Gagneux P, and Varki A
- Subjects
- Animals, Humans, Genome, Phenotype, Biological Evolution, Hominidae genetics
- Abstract
Anthropogeny is a classic term encompassing transdisciplinary investigations of the origins of the human species. Comparative anthropogeny is a systematic comparison of humans and other living nonhuman hominids (so-called "great apes"), aiming to identify distinctly human features in health and disease, with the overall goal of explaining human origins. We begin with a historical perspective, briefly describing how the field progressed from the earliest evolutionary insights to the current emphasis on in-depth molecular and genomic investigations of "human-specific" biology and an increased appreciation for cultural impacts on human biology. While many such genetic differences between humans and other hominids have been revealed over the last two decades, this information remains insufficient to explain the most distinctive phenotypic traits distinguishing humans from other living hominids. Here we undertake a complementary approach of "comparative physiological anthropogeny," along the lines of the preclinical medical curriculum, i.e., beginning with anatomy and considering each physiological system and in each case considering genetic and molecular components that are relevant. What is ultimately needed is a systematic comparative approach at all levels from molecular to physiological to sociocultural, building networks of related information, drawing inferences, and generating testable hypotheses. The concluding section will touch on distinctive considerations in the study of human evolution, including the importance of gene-culture interactions.
- Published
- 2023
- Full Text
- View/download PDF
17. Long-term exposure to house dust mites accelerates lung cancer development in mice.
- Author
-
Wang D, Li W, Albasha N, Griffin L, Chang H, Amaya L, Ganguly S, Zeng L, Keum B, González-Navajas JM, Levin M, AkhavanAghdam Z, Snyder H, Schwartz D, Tao A, Boosherhri LM, Hoffman HM, Rose M, Estrada MV, Varki N, Herdman S, Corr M, Webster NJG, Raz E, and Bertin S
- Subjects
- Humans, Animals, Mice, NLR Family, Pyrin Domain-Containing 3 Protein metabolism, Pyroglyphidae, Lung pathology, Disease Models, Animal, Tumor Microenvironment, Asthma metabolism, Asthma pathology, Lung Neoplasms chemically induced, Lung Neoplasms metabolism
- 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)., (© 2023. The Author(s).)- Published
- 2023
- Full Text
- View/download PDF
18. Development and applications of sialoglycan-recognizing probes (SGRPs) with defined specificities: exploring the dynamic mammalian sialoglycome.
- Author
-
Srivastava S, Verhagen A, Sasmal A, Wasik BR, Diaz S, Yu H, Bensing BA, Khan N, Khedri Z, Secrest P, Sullam P, Varki N, Chen X, Parrish CR, and Varki A
- Subjects
- Humans, Mice, Animals, Mice, Inbred C57BL, Mammals metabolism, Polysaccharides, Sialic Acids chemistry, Hemagglutinins, Viral
- Abstract
Glycans that are abundantly displayed on vertebrate cell surface and secreted molecules are often capped with terminal sialic acids (Sias). These diverse 9-carbon-backbone monosaccharides are involved in numerous intrinsic biological processes. They also interact with commensals and pathogens, while undergoing dynamic changes in time and space, often influenced by environmental conditions. However, most of this sialoglycan complexity and variation remains poorly characterized by conventional techniques, which often tend to destroy or overlook crucial aspects of Sia diversity and/or fail to elucidate native structures in biological systems, i.e. in the intact sialome. To date, in situ detection and analysis of sialoglycans has largely relied on the use of plant lectins, sialidases, or antibodies, whose preferences (with certain exceptions) are limited and/or uncertain. We took advantage of naturally evolved microbial molecules (bacterial adhesins, toxin subunits, and viral hemagglutinin-esterases) that recognize sialoglycans with defined specificity to delineate 9 classes of sialoglycan recognizing probes (SGRPs: SGRP1-SGRP9) that can be used to explore mammalian sialome changes in a simple and systematic manner, using techniques common in most laboratories. SGRP candidates with specificity defined by sialoglycan microarray studies were engineered as tagged probes, each with a corresponding nonbinding mutant probe as a simple and reliable negative control. The optimized panel of SGRPs can be used in methods commonly available in most bioscience labs, such as ELISA, western blot, flow cytometry, and histochemistry. To demonstrate the utility of this approach, we provide examples of sialoglycome differences in tissues from C57BL/6 wild-type mice and human-like Cmah-/- mice., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
19. 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 A, Khan N, Khedri Z, Kellman BP, Srivastava S, Verhagen A, Yu H, Bruntse AB, Diaz S, Varki N, Beddoe T, Paton AW, Paton JC, Chen X, Lewis NE, and Varki A
- Subjects
- Salmonella typhi chemistry, Sialic Acids, Polysaccharides, Cholera Toxin, Escherichia coli, Bacterial Toxins chemistry
- 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)., (© The Author(s) 2022. Published by Oxford University Press. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
20. Lp(a), oxidized phospholipids and oxidation-specific epitopes are increased in subjects with keloid formation.
- Author
-
Ruder S, Mansfield B, Immelman AR, Varki N, Miu P, Raal F, and Tsimikas S
- Subjects
- Humans, Epitopes, Case-Control Studies, Lipoprotein(a), Apolipoproteins B, Apolipoprotein B-100, Oxidation-Reduction, Malondialdehyde, Immunoglobulin M, Immunoglobulin G, Antigen-Antibody Complex, Phospholipids
- Abstract
Background: Keloid 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., Methods: This 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., Results: Cases 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., Conclusion: Darkly 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., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. Sialoglycan-binding patterns of bacterial AB 5 toxin B subunits correlate with host range and toxicity, indicating evolution independent of A subunits.
- Author
-
Khan N, Sasmal A, Khedri Z, Secrest P, Verhagen A, Srivastava S, Varki N, Chen X, Yu H, Beddoe T, Paton AW, Paton JC, and Varki A
- Subjects
- Animals, Evolution, Molecular, Mammals metabolism, N-Acetylneuraminic Acid metabolism, Phylogeny, Plague microbiology, Protein Binding, Protein Subunits metabolism, Yersinia pestis metabolism, Bacteria classification, Bacteria metabolism, Bacterial Toxins metabolism, Bacterial Toxins toxicity, Host Specificity, Polysaccharides metabolism
- Abstract
Many pathogenic bacteria secrete AB
5 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., Competing Interests: Conflict of interest The authors declare that they have no conflicts of interest with the contents of this article., (Copyright © 2022 The Authors. Published by Elsevier Inc. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
22. Targeting glycan sulfation in a CD11c+ myeloid population inhibits early KRAS-mutant lung neoplasia.
- Author
-
Kim SY, Johns SC, Gupta P, Varki N, and Fuster MM
- Subjects
- Adenoma etiology, Adenoma metabolism, Animals, CD8-Positive T-Lymphocytes, Heparitin Sulfate chemistry, Humans, Lung Neoplasms etiology, Lung Neoplasms metabolism, Lymphocytes, Tumor-Infiltrating immunology, Mice, Mice, Inbred C57BL, Mice, Transgenic, Myeloid Cells metabolism, Myeloid Cells pathology, Sulfates metabolism, Sulfotransferases physiology, T-Lymphocytes, Regulatory immunology, Adenoma pathology, CD11c Antigen metabolism, Lung Neoplasms pathology, Mutation, Myeloid Cells immunology, Polysaccharides chemistry, Proto-Oncogene Proteins p21(ras) genetics
- Abstract
Early lung carcinoma development may be modulated by innate host cellular mechanisms that promote tumor growth and invasion. We recently identified how a loss-of-function mutation in the glycan sulfating enzyme N-deacetylase/N-sulfotransferase-1 (Ndst1; involved in heparan sulfate biosynthesis) targeted to antigen presenting cells (APCs) may augment acquired anti-tumor T cell immune mechanisms. Crossing this mutation (Ndst1f/f CD11cCre+) onto a model of inducible spontaneous Kras mutant lung cancer [CCSP-rtTA; (tetO7) CMV-Kras-G12D] allowed us to examine how the APC mutation affects the formation and growth of early lung carcinoma. We examined early bronchocentric adenoma formation in the model, and the frequency of such events was significantly reduced on the mutant background. This was associated with significant reductions in tumor associated FOXP3+ cellular infiltration and CD163+ M2-type macrophage infiltration. The findings evolved prior to effector CD8+ T cell infiltration into tumors. The impact of this unique glycan under-sulfating mutation on inhibiting early Kras G12D mutant bronchocentric adenoma formation along with a cellular phenotype of inhibited tumor infiltration by cells involved in suppressive T-regulatory cell signaling (FOXP3+ cells) or tumor-permissive M2 macrophage functions (CD163+ cells) provides insight on how glycan targeting may modulate innate cellular mechanisms during early lung tumor development. The findings may also impact the future design of host-centered immunologic anti-tumor therapeutic strategies., (Copyright © 2021. Published by Elsevier Inc.)
- Published
- 2021
- Full Text
- View/download PDF
23. Dietary Neu5Ac Intervention Protects Against Atherosclerosis Associated With Human-Like Neu5Gc Loss-Brief Report.
- Author
-
Kawanishi K, Coker JK, Grunddal KV, Dhar C, Hsiao J, Zengler K, Varki N, Varki A, and Gordts PLSM
- Subjects
- Animal Feed, Animals, Antibodies metabolism, Aorta pathology, Aortic Diseases genetics, Aortic Diseases metabolism, Aortic Diseases pathology, Atherosclerosis genetics, Atherosclerosis metabolism, Atherosclerosis pathology, Diet, High-Fat, Disease Models, Animal, Foam Cells metabolism, Foam Cells pathology, Humans, Male, Mice, Inbred C57BL, Mice, Knockout, Mixed Function Oxygenases genetics, Mixed Function Oxygenases metabolism, N-Acetylneuraminic Acid metabolism, Neuraminic Acids immunology, Neuraminic Acids metabolism, Pan troglodytes, Receptors, LDL genetics, Receptors, LDL metabolism, Sialadenitis metabolism, Sialadenitis pathology, THP-1 Cells, Mice, Aorta metabolism, Aortic Diseases prevention & control, Atherosclerosis prevention & control, Dietary Supplements, N-Acetylneuraminic Acid administration & dosage, Neuraminic Acids administration & dosage, Plaque, Atherosclerotic
- Abstract
Objective: Species-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 Results: We 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., Conclusions: Interventional 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
- Full Text
- View/download PDF
24. Endothelial Heparan Sulfate Mediates Hepatic Neutrophil Trafficking and Injury during Staphylococcus aureus Sepsis.
- Author
-
Golden GJ, Toledo AG, Marki A, Sorrentino JT, Morris C, Riley RJ, Spliid C, Chen Q, Cornax I, Lewis NE, Varki N, Le D, Malmström J, Karlsson C, Ley K, Nizet V, and Esko JD
- Subjects
- Animals, Disease Models, Animal, Endothelial Cells immunology, Female, Glycocalyx metabolism, Glycocalyx pathology, Liver immunology, Liver microbiology, Liver pathology, Lung immunology, Lung microbiology, Lung pathology, Male, Mice, Mice, Inbred C57BL, Staphylococcus aureus pathogenicity, Endothelial Cells metabolism, Heparitin Sulfate genetics, Heparitin Sulfate metabolism, Neutrophil Activation, Neutrophils pathology, Sepsis microbiology, Staphylococcus aureus immunology
- 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
- Full Text
- View/download PDF
25. Human-specific polymorphic pseudogenization of SIGLEC12 protects against advanced cancer progression.
- Author
-
Siddiqui SS, Vaill M, Do R, Khan N, Verhagen AL, Zhang W, Lenz HJ, Johnson-Pais TL, Leach RJ, Fraser G, Wang C, Feng GS, Varki N, and Varki A
- 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., Competing Interests: The authors declare no competing interest with the content of this manuscript., (© 2020 The Authors. FASEB BioAdvances published by the Federation of American Societies for Experimental Biology.)
- Published
- 2020
- Full Text
- View/download PDF
26. Evaluation of IL-17D in Host Immunity to Group A Streptococcus Infection.
- Author
-
Washington A Jr, Varki N, Valderrama JA, Nizet V, and Bui JD
- Subjects
- Animals, Chemokine CCL2 immunology, Immunity, Innate immunology, Killer Cells, Natural immunology, Mice, Mice, Inbred C57BL, Neutrophils immunology, Pathogen-Associated Molecular Pattern Molecules immunology, Th17 Cells immunology, Interleukin-27 immunology, Streptococcal Infections immunology, Streptococcus 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., (Copyright © 2020 by The American Association of Immunologists, Inc.)- Published
- 2020
- Full Text
- View/download PDF
27. DDX5 promotes oncogene C3 and FABP1 expressions and drives intestinal inflammation and tumorigenesis.
- Author
-
Abbasi N, Long T, Li Y, Yee BA, Cho BS, Hernandez JE, Ma E, Patel PR, Sahoo D, Sayed IM, Varki N, Das S, Ghosh P, Yeo GW, and Huang WJM
- Subjects
- Animals, Carcinogenesis metabolism, Colitis chemically induced, Complement C3 genetics, DEAD-box RNA Helicases physiology, Dextran Sulfate adverse effects, Epithelial Cells metabolism, Fatty Acid-Binding Proteins genetics, Female, Gene Expression genetics, Gene Expression Regulation, Neoplastic genetics, Inflammation, Intestinal Mucosa metabolism, Intestine, Small metabolism, Intestines pathology, Male, Mice, Mice, Inbred C57BL, Oncogenes genetics, Signal Transduction, Complement C3 metabolism, DEAD-box RNA Helicases metabolism, Fatty Acid-Binding Proteins metabolism
- 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., (© 2020 Abbasi et al.)
- Published
- 2020
- Full Text
- View/download PDF
28. Multiple Genomic Events Altering Hominin SIGLEC Biology and Innate Immunity Predated the Common Ancestor of Humans and Archaic Hominins.
- Author
-
Khan N, de Manuel M, Peyregne S, Do R, Prufer K, Marques-Bonet T, Varki N, Gagneux P, and Varki A
- Subjects
- Animals, Gene Expression, Genome, Hominidae immunology, Humans, Immunity, Innate genetics, Multigene Family, Mutation, Polymorphism, Genetic, Selection, Genetic, Evolution, Molecular, Hominidae genetics, Sialic Acid Binding Ig-like Lectin 3 genetics
- 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., (© The Author(s) 2020. Published by Oxford University Press on behalf of the Society for Molecular Biology and Evolution.)
- Published
- 2020
- Full Text
- View/download PDF
29. Proteomic atlas of organ vasculopathies triggered by Staphylococcus aureus sepsis.
- Author
-
Toledo AG, Golden G, Campos AR, Cuello H, Sorrentino J, Lewis N, Varki N, Nizet V, Smith JW, and Esko JD
- Subjects
- Animals, Hyaluronic Acid metabolism, Liver pathology, Mice, Mice, Inbred C57BL, Multiple Organ Failure microbiology, Proteomics, Staphylococcal Infections immunology, Vascular Diseases metabolism, Vascular Remodeling, Methicillin-Resistant Staphylococcus aureus immunology, Proteome, Sepsis metabolism, Staphylococcal Infections complications, Vascular Diseases microbiology
- 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
- Full Text
- View/download PDF
30. Human species-specific loss of CMP- N -acetylneuraminic acid hydroxylase enhances atherosclerosis via intrinsic and extrinsic mechanisms.
- Author
-
Kawanishi K, Dhar C, Do R, Varki N, Gordts PLSM, and Varki A
- Subjects
- Animals, Cattle, Cytokines metabolism, Diet, High-Fat, Female, Humans, Hyperglycemia pathology, Inflammation pathology, Macrophages metabolism, Macrophages pathology, Male, Mice, Inbred C57BL, Mixed Function Oxygenases metabolism, Models, Biological, Phenotype, Receptors, LDL deficiency, Receptors, LDL metabolism, Sialic Acids metabolism, Species Specificity, Atherosclerosis enzymology, Mixed Function Oxygenases deficiency
- 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., Competing Interests: The authors declare no conflict of interest.- Published
- 2019
- Full Text
- View/download PDF
31. MST1R kinase accelerates pancreatic cancer progression via effects on both epithelial cells and macrophages.
- Author
-
Babicky ML, Harper MM, Chakedis J, Cazes A, Mose ES, Jaquish DV, French RP, Childers B, Alakus H, Schmid MC, Foubert P, Miyamoto J, Holman PJ, Walterscheid ZJ, Tang CM, Varki N, Sicklick JK, Messer K, Varner JA, Waltz SE, and Lowy AM
- Subjects
- Animals, Carcinoma, Pancreatic Ductal enzymology, Disease Progression, Female, Gene Knockdown Techniques, Humans, Intracellular Signaling Peptides and Proteins, Male, Mice, Mice, Inbred C57BL, Mice, Transgenic, Pancreatic Neoplasms enzymology, Proof of Concept Study, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Receptor Protein-Tyrosine Kinases genetics, Signal Transduction, Tumor Microenvironment, Carcinoma, Pancreatic Ductal pathology, Epithelial Cells pathology, Macrophages pathology, Pancreatic Neoplasms pathology, Receptor Protein-Tyrosine Kinases metabolism
- 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
- Full Text
- View/download PDF
32. Evolution of the exclusively human pathogen Neisseria gonorrhoeae : Human-specific engagement of immunoregulatory Siglecs.
- Author
-
Landig CS, Hazel A, Kellman BP, Fong JJ, Schwarz F, Agarwal S, Varki N, Massari P, Lewis NE, Ram S, and Varki A
- 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
- Full Text
- View/download PDF
33. Human evolutionary loss of epithelial Neu5Gc expression and species-specific susceptibility to cholera.
- Author
-
Alisson-Silva F, Liu JZ, Diaz SL, Deng L, Gareau MG, Marchelletta R, Chen X, Nizet V, Varki N, Barrett KE, and Varki A
- Subjects
- Animals, Cholera metabolism, Cholera pathology, Epithelial Cells pathology, Female, Humans, Intestine, Small metabolism, Intestine, Small microbiology, Intestine, Small pathology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Species Specificity, Vibrio cholerae pathogenicity, Biological Evolution, Cholera microbiology, Disease Susceptibility, Epithelial Cells metabolism, Mixed Function Oxygenases physiology, Neuraminic Acids metabolism, Vibrio cholerae classification
- 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., Competing Interests: The authors have declared that no competing interests exists.
- Published
- 2018
- Full Text
- View/download PDF
34. The plasminogen receptor , Plg-R KT , is essential for mammary lobuloalveolar development and lactation.
- Author
-
Miles LA, Baik N, Bai H, Makarenkova HP, Kiosses WB, Krajewski S, Castellino FJ, Valenzuela A, Varki NM, Mueller BM, and Parmer RJ
- Subjects
- Animals, Apoptosis, Cell Proliferation, Extracellular Matrix metabolism, Extracellular Matrix pathology, Female, Fibrinogen genetics, Fibrinogen metabolism, Fibrosis, Genotype, Macrophages metabolism, Macrophages pathology, Mammary Glands, Animal growth & development, Mammary Glands, Animal pathology, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Phenotype, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Fibrin metabolism, Lactation, Mammary Glands, Animal metabolism, Morphogenesis, Receptors, Cell Surface deficiency
- Abstract
Essentials Plg-R
KT -/- female mice give birth, but no offspring of Plg-RKT -/- female mice survive to weaning. Causal mechanisms of potential lactational failure in Plg-RKT -/- mice are unknown. Plg-RKT regulates extracellular matrix remodeling, cell proliferation, apoptosis, fibrin surveillance. Plg-RKT is essential for lactogenesis and mammary lobuloalveolar development., Summary: Background Lactational competence requires plasminogen, the zymogen of the serine protease, plasmin. Plg-RKT is a unique transmembrane plasminogen receptor that promotes plasminogen activation to plasmin on cell surfaces. Plg-RKT -/- mice are viable, but no offspring of Plg-RKT -/- female mice survive to weaning. Objectives We investigated potential lactational failure in Plg-RKT -/- mice and addressed causal mechanisms. Methods Fibrin accumulation, macrophage infiltration, processing of extracellular matrix components, effects of genetic deletion of fibrinogen, expression of fibrosis genes, and proliferation and apoptosis of epithelial cells were examined in lactating mammary glands of Plg-RKT -/- and Plg-RKT +/+ mice. Results Milk was not present in the stomachs of offspring of Plg-RKT -/- female mice and the pups were rescued by foster mothers. Although the mammary ductal tree developed normally in Plg-RKT -/- glands, lobuloalveolar development was blocked by a hypertrophic fibrotic stroma and infiltrating macrophages were present. A massive accumulation of fibrin was also present in Plg-RKT -/- alveoli and ducts. Although this accumulation was decreased when Plg-RKT -/- mice were made genetically heterozygous for fibrinogen, defects in lobuloalveolar development were not rescued by fibrinogen heterozygosity. Transcriptional profiling revealed that EGF was downregulated 12-fold in Plg-RKT -/- glands. Furthermore, proliferation of epithelial cells was not detectable. In addition, the pro-survival protein, Mcl-1, was markedly downregulated and apoptosis was observed in Plg-RKT -/- but not Plg-RKT +/+ glands. Conclusions Plg-RKT is essential for lactogenesis and functions to maintain the appropriate stromal extracellular matrix environment, regulate epithelial cell proliferation and apoptosis, and, by regulating fibrinolysis, preserve alveolar and ductal patency., (© 2018 International Society on Thrombosis and Haemostasis.)- Published
- 2018
- Full Text
- View/download PDF
35. Paired Siglec receptors generate opposite inflammatory responses to a human-specific pathogen.
- Author
-
Schwarz F, Landig CS, Siddiqui S, Secundino I, Olson J, Varki N, Nizet V, and Varki A
- Subjects
- Animals, Cytokines metabolism, Escherichia coli immunology, Escherichia coli pathogenicity, Escherichia coli Infections immunology, Escherichia coli Infections pathology, Humans, Immune Evasion, Macrophages immunology, Macrophages microbiology, Mice, Mice, Transgenic, Microbial Viability, Inflammation pathology, Lectins metabolism, Membrane Proteins metabolism, Sialic Acid Binding Immunoglobulin-like Lectins metabolism, Sialic Acids metabolism
- 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., (© 2017 The Authors.)
- Published
- 2017
- Full Text
- View/download PDF
36. Studies on the Detection, Expression, Glycosylation, Dimerization, and Ligand Binding Properties of Mouse Siglec-E.
- Author
-
Siddiqui S, Schwarz F, Springer S, Khedri Z, Yu H, Deng L, Verhagen A, Naito-Matsui Y, Jiang W, Kim D, Zhou J, Ding B, Chen X, Varki N, and Varki A
- Subjects
- Amino Acid Substitution, Animals, Antibodies chemistry, Antigens, CD chemistry, Antigens, CD genetics, Antigens, Differentiation, B-Lymphocyte chemistry, Antigens, Differentiation, B-Lymphocyte genetics, Dendritic Cells cytology, Dendritic Cells metabolism, Glycosylation, Humans, Macrophages cytology, Macrophages metabolism, Mice, Mice, Knockout, Monocytes cytology, Monocytes metabolism, Mutagenesis, Mutation, Missense, Neutrophils cytology, Neutrophils metabolism, Protein Tyrosine Phosphatase, Non-Receptor Type 6, Rats, Rats, Inbred Lew, Sialic Acid Binding Immunoglobulin-like Lectins chemistry, Sialic Acid Binding Immunoglobulin-like Lectins genetics, Sialic Acid Binding Immunoglobulin-like Lectins metabolism, Antigens, CD metabolism, Antigens, Differentiation, B-Lymphocyte metabolism, Gene Expression Regulation physiology, Protein Multimerization physiology
- 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., (© 2017 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
37. Deficiency of plasminogen receptor, Plg-R KT , causes defects in plasminogen binding and inflammatory macrophage recruitment in vivo.
- Author
-
Miles LA, Baik N, Lighvani S, Khaldoyanidi S, Varki NM, Bai H, Mueller BM, and Parmer RJ
- Subjects
- Animals, Blood Cell Count, Cell Membrane metabolism, Female, Fibrinolysin chemistry, Homeostasis, Humans, Inflammation, Male, Mice, Mice, Transgenic, Protein Binding, Protein Domains, Thrombolytic Therapy, Macrophages cytology, Plasminogen chemistry, Receptors, Cell Surface chemistry
- Abstract
Essentials Plg-R
KT is a novel integral membrane plasminogen receptor. The functions of Plg-RKT in vivo are not known. Plg-RKT is a key player in macrophage recruitment in the inflammatory response in vivo. Plg-RKT deficiency is not compatible with survival of the species., Summary: Background Plg-RKT is a novel integral membrane plasminogen receptor that binds plasminogen via a C-terminal lysine exposed on the cell surface and promotes plasminogen activation on the cell surface by both tissue plasminogen activator and urokinase plasminogen activator. Objectives To evaluate the role of Plg-RKT in vivo we generated Plg-RKT -/- mice using a homologous recombination technique. Methods We characterized the effect of Plg-RKT deletion on reproduction, viability, health and spontaneous thrombosis and inflammation. Results Plg-RKT -/- mice were viable and fertile. Survival of Plg-RKT -/- mice and Plg-RKT +/+ littermates was not significantly different. However, quite strikingly, all pups of Plg-RKT -/- females died within 2 days of birth, consistent with a lactation defect in Plg-RKT -/- mothers. Additionally, there was a significant effect of Plg-RKT deficiency on the growth rates of female, but not male, mice. In experimental peritonitis studies, Plg-RKT -/- mice exhibited a marked defect in macrophage recruitment. As a contributing mechanism, the capacity of Plg-RKT -/- macrophages for plasminogen binding was markedly decreased. Conclusions These studies demonstrate that Plg-RKT is required for plasminogen binding and macrophage migration in vivo. In addition, Plg-RKT deficiency is not compatible with survival of the species, due to the death of all offspring of Plg-RKT -/- females. This new mouse model will be important for future studies aimed at delineating the role of cell surface plasminogen activation in challenge and disease models in vivo., Competing Interests: Authors state no conflicts of interests., (© 2016 International Society on Thrombosis and Haemostasis.)- Published
- 2017
- Full Text
- View/download PDF
38. Esophageal Submucosal Injection of Capsaicin but Not Acid Induces Symptoms in Normal Subjects.
- Author
-
Lee RH, Korsapati H, Bhalla V, Varki N, and Mittal RK
- Abstract
Background/aims: Transient receptor potential vanilloid-1 (TRPV1) is a candidate for mediating acid-induced symptoms in the esophagus. We conducted studies to determine if the presence of acid in the mucosa/submucosa and direct activation of TRPV1 by capsaicin elicited symptoms in normal healthy subjects. We also studied the presence of TRPV1 receptors in the esophagus., Methods: Unsedated endoscopy was performed on healthy subjects with no symptoms. Using a sclerotherapy needle, normal saline (pH 2.0-7.5) was injected into the mucosa/submucosa, 5 cm above the Z line. In a separate group of healthy subjects, injection of capsaicin and vehicle was also studied. Quality of symptoms was reported using the McGill Pain Questionnaire, and symptom intensity using the visual analogue scale (VAS). Immunohistochemistry was performed on 8 surgical esophagus specimens using TRPV1 antibody., Results: Acid injection either did not elicit or elicited mild symptoms in subjects at all pH solutions. Capsaicin but not the vehicle elicited severe heartburn/chest pain in all subjects. Mean VAS for capsaicin was 91 ± 3 and symptoms lasted for 25 ± 1 minutes. Immunohistochemistry revealed a linear TRPV1 staining pattern between the epithelial layer and the submucosa that extended into the papillae. Eighty-five percent of papillae stained positive for TRPV1 with a mean 1.1 positive papillae per high-powered field., Conclusions: The mechanism of acid-induced heartburn and chest pain is not the simple interaction of hydrogen ions with afferents located in the esophageal mucosa and submucosa. TRPV1 receptors are present in the lamina propria and their activation induces heartburn and chest pain.
- Published
- 2016
- Full Text
- View/download PDF
39. Siglec receptors impact mammalian lifespan by modulating oxidative stress.
- Author
-
Schwarz F, Pearce OM, Wang X, Samraj AN, Läubli H, Garcia JO, Lin H, Fu X, Garcia-Bingman A, Secrest P, Romanoski CE, Heyser C, Glass CK, Hazen SL, Varki N, Varki A, and Gagneux P
- Subjects
- Animals, Body Weight, Homeostasis, Immunomodulation, Male, Mammals genetics, Mice, Inbred C57BL, Multigene Family, Phenotype, Phylogeny, Reactive Oxygen Species metabolism, Sialic Acid Binding Immunoglobulin-like Lectins deficiency, Sialic Acid Binding Immunoglobulin-like Lectins metabolism, Longevity, Mammals physiology, Oxidative Stress, Receptors, Cell Surface metabolism
- Abstract
Aging is a multifactorial process that includes the lifelong accumulation of molecular damage, leading to age-related frailty, disability and disease, and eventually death. In this study, we report evidence of a significant correlation between the number of genes encoding the immunomodulatory CD33-related sialic acid-binding immunoglobulin-like receptors (CD33rSiglecs) and maximum lifespan in mammals. In keeping with this, we show that mice lacking Siglec-E, the main member of the CD33rSiglec family, exhibit reduced survival. Removal of Siglec-E causes the development of exaggerated signs of aging at the molecular, structural, and cognitive level. We found that accelerated aging was related both to an unbalanced ROS metabolism, and to a secondary impairment in detoxification of reactive molecules, ultimately leading to increased damage to cellular DNA, proteins, and lipids. Taken together, our data suggest that CD33rSiglecs co-evolved in mammals to achieve a better management of oxidative stress during inflammation, which in turn reduces molecular damage and extends lifespan.
- Published
- 2015
- Full Text
- View/download PDF
40. The E-Id protein axis modulates the activities of the PI3K-AKT-mTORC1-Hif1a and c-myc/p19Arf pathways to suppress innate variant TFH cell development, thymocyte expansion, and lymphomagenesis.
- Author
-
Miyazaki M, Miyazaki K, Chen S, Chandra V, Wagatsuma K, Agata Y, Rodewald HR, Saito R, Chang AN, Varki N, Kawamoto H, and Murre C
- Subjects
- Animals, Cell Proliferation, Cyclin-Dependent Kinase Inhibitor p16 metabolism, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Hypoxia-Inducible Factor 1, alpha Subunit metabolism, Inhibitor of Differentiation Proteins genetics, Lymphoid Tissue cytology, Mechanistic Target of Rapamycin Complex 1, Mice, Multiprotein Complexes metabolism, Oncogene Protein v-akt metabolism, Phosphatidylinositol 3-Kinases metabolism, Proto-Oncogene Proteins c-myc metabolism, STAT1 Transcription Factor, TOR Serine-Threonine Kinases metabolism, Basic Helix-Loop-Helix Leucine Zipper Transcription Factors metabolism, Cell Differentiation, Inhibitor of Differentiation Proteins metabolism, Lymphoma physiopathology, T-Lymphocytes, Helper-Inducer cytology, Thymocytes cytology
- Abstract
It is now well established that the E and Id protein axis regulates multiple steps in lymphocyte development. However, it remains unknown how E and Id proteins mechanistically enforce and maintain the naïve T-cell fate. Here we show that Id2 and Id3 suppressed the development and expansion of innate variant follicular helper T (TFH) cells. Innate variant TFH cells required major histocompatibility complex (MHC) class I-like signaling and were associated with germinal center B cells. We found that Id2 and Id3 induced Foxo1 and Foxp1 expression to antagonize the activation of a TFH transcription signature. We show that Id2 and Id3 acted upstream of the Hif1a/Foxo/AKT/mTORC1 pathway as well as the c-myc/p19Arf module to control cellular expansion. We found that mice depleted for Id2 and Id3 expression developed colitis and αβ T-cell lymphomas. Lymphomas depleted for Id2 and Id3 expression displayed elevated levels of c-myc, whereas p19Arf abundance declined. Transcription signatures of Id2- and Id3-depleted lymphomas revealed similarities to genetic deficiencies associated with Burkitt lymphoma. We propose that, in response to antigen receptor and/or cytokine signaling, the E-Id protein axis modulates the activities of the PI3K-AKT-mTORC1-Hif1a and c-myc/p19Arf pathways to control cellular expansion and homeostatic proliferation., (© 2015 Miyazaki et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2015
- Full Text
- View/download PDF
41. Advanced Technologies in Sialic Acid and Sialoglycoconjugate Analysis.
- Author
-
Kitajima K, Varki N, and Sato C
- Subjects
- Aging metabolism, Carcinogenesis chemistry, Carcinogenesis pathology, Chromatography, High Pressure Liquid instrumentation, Fluorometry instrumentation, Glycoconjugates chemistry, Glycolipids analysis, Glycolipids chemistry, Glycoproteins analysis, Glycoproteins chemistry, Humans, Immunity, Innate, Immunohistochemistry instrumentation, N-Acetylneuraminic Acid chemistry, Neuraminic Acids chemistry, Neurogenesis physiology, Oligosaccharides analysis, Oligosaccharides chemistry, Polysaccharides analysis, Polysaccharides chemistry, Sialic Acids analysis, Sialic Acids chemistry, Chromatography, High Pressure Liquid methods, Fluorometry methods, Glycoconjugates analysis, Immunohistochemistry methods, N-Acetylneuraminic Acid analysis, Neuraminic Acids analysis
- Abstract
Although the structural diversity of sialic acid (Sia) is rapidly expanding, understanding of its biological significance has lagged behind. Advanced technologies to detect and probe diverse structures of Sia are absolutely necessary not only to understand further biological significance but also to pursue medicinal and industrial applications. Here we describe analytical methods for detection of Sia that have recently been developed or improved, with a special focus on 9-O-acetylated N-acetylneuraminic acid (Neu5,9Ac), N-glycolylneuraminic acid (Neu5Gc), deaminoneuraminic acid (Kdn), O-sulfated Sia (SiaS), and di-, oligo-, and polysialic acid (diSia/oligoSia/polySia) in glycoproteins and glycolipids. Much more attention has been paid to these Sia and sialoglycoconjugates during the last decade, in terms of regulation of the immune system, neural development and function, tumorigenesis, and aging.
- Published
- 2015
- Full Text
- View/download PDF
42. Host adaptation of a bacterial toxin from the human pathogen Salmonella Typhi.
- Author
-
Deng L, Song J, Gao X, Wang J, Yu H, Chen X, Varki N, Naito-Matsui Y, Galán JE, and Varki A
- Subjects
- Animals, Bacterial Toxins genetics, Cell Line, Cells, Cultured, Crystallography, X-Ray, Host Specificity, Humans, Jurkat Cells, Mice, Mice, Inbred C57BL, Models, Molecular, Neuraminic Acids metabolism, Pan troglodytes, Salmonella typhi pathogenicity, Typhoid Fever microbiology, Bacterial Toxins chemistry, Bacterial Toxins metabolism, Membrane Glycoproteins chemistry, N-Acetylneuraminic Acid chemistry, N-Acetylneuraminic Acid metabolism, Salmonella typhi chemistry
- Abstract
Salmonella Typhi is an exclusive human pathogen that causes typhoid fever. Typhoid toxin is a S. Typhi virulence factor that can reproduce most of the typhoid fever symptoms in experimental animals. Toxicity depends on toxin binding to terminally sialylated glycans on surface glycoproteins. Human glycans are unusual because of the lack of CMAH, which in other mammals converts N-acetylneuraminic acid (Neu5Ac) to N-glycolylneuraminic acid (Neu5Gc). Here, we report that typhoid toxin binds to and is toxic toward cells expressing glycans terminated in Neu5Ac (expressed by humans) over glycans terminated in Neu5Gc (expressed by other mammals). Mice constitutively expressing CMAH thus displaying Neu5Gc in all tissues are resistant to typhoid toxin. The atomic structure of typhoid toxin bound to Neu5Ac reveals the structural bases for its binding specificity. These findings provide insight into the molecular bases for Salmonella Typhi's host specificity and may help the development of therapies for typhoid fever., (Copyright © 2014 Elsevier Inc. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
43. Lectin galactoside-binding soluble 3 binding protein (LGALS3BP) is a tumor-associated immunomodulatory ligand for CD33-related Siglecs.
- Author
-
Läubli H, Alisson-Silva F, Stanczak MA, Siddiqui SS, Deng L, Verhagen A, Varki N, and Varki A
- Subjects
- Animals, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, Biomarkers, Tumor genetics, Biomarkers, Tumor immunology, Carrier Proteins genetics, Carrier Proteins immunology, Colorectal Neoplasms genetics, Colorectal Neoplasms immunology, Colorectal Neoplasms pathology, Female, Gene Expression Regulation, Neoplastic immunology, Glycoproteins genetics, Glycoproteins immunology, HEK293 Cells, Humans, Immunologic Factors genetics, Immunologic Factors immunology, Male, Mice, Neoplasm Proteins genetics, Neoplasm Proteins immunology, Prostatic Neoplasms genetics, Prostatic Neoplasms immunology, Prostatic Neoplasms pathology, Sialic Acid Binding Ig-like Lectin 3 genetics, Sialic Acid Binding Ig-like Lectin 3 immunology, Tumor Escape, Up-Regulation immunology, Antigens, Neoplasm biosynthesis, Biomarkers, Tumor biosynthesis, Carrier Proteins biosynthesis, Colorectal Neoplasms metabolism, Glycoproteins biosynthesis, Immunologic Factors biosynthesis, Neoplasm Proteins metabolism, Prostatic Neoplasms metabolism, Sialic Acid Binding Ig-like Lectin 3 metabolism
- Abstract
Lectin galactoside-binding soluble 3 binding protein (LGALS3BP, also called Mac-2 binding protein) is a heavily glycosylated secreted molecule that has been shown previously to be up-regulated in many cancers and has been implicated in tumor metastatic processes, as well as in other cell adhesion and immune functions. The CD33-related subset of sialic acid-binding immunoglobulin-like lectins (Siglecs) consists of immunomodulatory molecules that have recently been associated with the modulation of immune responses to cancer. Because up-regulation of Siglec ligands in cancer tissue has been observed, the characterization of these cancer-associated ligands that bind to inhibitory CD33-related Siglecs could provide novel targets for cancer immunomodulatory therapy. Here we used affinity chromatography of tumor cell extracts to identify LGALS3BP as a novel sialic acid-dependent ligand for human Siglec-9 and for other immunomodulatory Siglecs, such as Siglec-5 and Siglec-10. In contrast, the mouse homolog Siglec-E binds to murine LGALS3BP with lower affinity. LGALS3BP has been observed to be up-regulated in human colorectal and prostate cancer specimens, particularly in the extracellular matrix. Finally, LGALS3BP was able to inhibit neutrophil activation in a sialic acid- and Siglec-dependent manner. These findings suggest a novel immunoinhibitory function for LGALS3BP that might be important for immune evasion of tumor cells during cancer progression., (© 2014 by The American Society for Biochemistry and Molecular Biology, Inc.)
- Published
- 2014
- Full Text
- View/download PDF
44. Id2 and Id3 maintain the regulatory T cell pool to suppress inflammatory disease.
- Author
-
Miyazaki M, Miyazaki K, Chen S, Itoi M, Miller M, Lu LF, Varki N, Chang AN, Broide DH, and Murre C
- Subjects
- Animals, Base Sequence, CD8-Positive T-Lymphocytes immunology, Cell Differentiation immunology, Cell Movement immunology, Cell Proliferation, Female, Forkhead Transcription Factors biosynthesis, Gene Expression Regulation immunology, Green Fluorescent Proteins genetics, Inflammation genetics, Inhibitor of Differentiation Protein 2 biosynthesis, Inhibitor of Differentiation Protein 2 genetics, Inhibitor of Differentiation Proteins biosynthesis, Inhibitor of Differentiation Proteins genetics, Interleukin-10 biosynthesis, Mice, Mice, Inbred C57BL, Mice, Knockout, Receptors, Antigen, T-Cell immunology, Receptors, CXCR5 biosynthesis, Sequence Analysis, RNA, Inflammation immunology, Inhibitor of Differentiation Protein 2 immunology, Inhibitor of Differentiation Proteins immunology, T-Lymphocytes, Regulatory immunology
- Abstract
Regulatory T (Treg) cells suppress the development of inflammatory disease, but our knowledge of transcriptional regulators that control this function remains incomplete. Here we show that expression of Id2 and Id3 in Treg cells was required to suppress development of fatal inflammatory disease. We found that T cell antigen receptor (TCR)-driven signaling initially decreased the abundance of Id3, which led to the activation of a follicular regulatory T (TFR) cell-specific transcription signature. However, sustained lower abundance of Id2 and Id3 interfered with proper development of TFR cells. Depletion of Id2 and Id3 expression in Treg cells resulted in compromised maintenance and localization of the Treg cell population. Thus, Id2 and Id3 enforce TFR cell checkpoints and control the maintenance and homing of Treg cells.
- Published
- 2014
- Full Text
- View/download PDF
45. Siglec-5 and Siglec-14 are polymorphic paired receptors that modulate neutrophil and amnion signaling responses to group B Streptococcus.
- Author
-
Ali SR, Fong JJ, Carlin AF, Busch TD, Linden R, Angata T, Areschoug T, Parast M, Varki N, Murray J, Nizet V, and Varki A
- Subjects
- Adult, Amnion metabolism, Amnion microbiology, Antigens, CD genetics, Antigens, CD metabolism, Antigens, Differentiation, Myelomonocytic genetics, Antigens, Differentiation, Myelomonocytic metabolism, Blotting, Western, Cell Line, Tumor, Cells, Cultured, DNA-Binding Proteins genetics, DNA-Binding Proteins immunology, DNA-Binding Proteins metabolism, Female, Gene Expression drug effects, Gene Expression immunology, Genotype, Host-Pathogen Interactions immunology, Humans, Infant, Newborn, Lectins genetics, Lectins metabolism, Lipopolysaccharides immunology, Lipopolysaccharides pharmacology, Monocytes immunology, Monocytes metabolism, Monocytes microbiology, Neutrophils metabolism, Neutrophils microbiology, Phosphorylation, Polymorphism, Genetic, Pregnancy, Proto-Oncogene Proteins c-akt metabolism, Receptors, Cell Surface genetics, Receptors, Cell Surface metabolism, Reverse Transcriptase Polymerase Chain Reaction, Signal Transduction immunology, Streptococcal Infections microbiology, Streptococcus agalactiae genetics, Streptococcus agalactiae physiology, Amnion immunology, Antigens, CD immunology, Antigens, Differentiation, Myelomonocytic immunology, Lectins immunology, Neutrophils immunology, Receptors, Cell Surface immunology, Streptococcal Infections immunology, Streptococcus agalactiae immunology
- Abstract
Group B Streptococcus (GBS) causes invasive infections in human newborns. We recently showed that the GBS β-protein attenuates innate immune responses by binding to sialic acid-binding immunoglobulin-like lectin 5 (Siglec-5), an inhibitory receptor on phagocytes. Interestingly, neutrophils and monocytes also express Siglec-14, which has a ligand-binding domain almost identical to Siglec-5 but signals via an activating motif, raising the possibility that these are paired Siglec receptors that balance immune responses to pathogens. Here we show that β-protein-expressing GBS binds to both Siglec-5 and Siglec-14 on neutrophils and that the latter engagement counteracts pathogen-induced host immune suppression by activating p38 mitogen-activated protein kinase (MAPK) and AKT signaling pathways. Siglec-14 is absent from some humans because of a SIGLEC14-null polymorphism, and homozygous SIGLEC14-null neutrophils are more susceptible to GBS immune subversion. Finally, we report an unexpected human-specific expression of Siglec-5 and Siglec-14 on amniotic epithelium, the site of initial contact of invading GBS with the fetus. GBS amnion immune activation was likewise influenced by the SIGLEC14-null polymorphism. We provide initial evidence that the polymorphism could influence the risk of prematurity among human fetuses of mothers colonized with GBS. This first functionally proven example of a paired receptor system in the Siglec family has multiple implications for regulation of host immunity., (© 2014 Ali et al.)
- Published
- 2014
- Full Text
- View/download PDF
46. Corrigendum: involvement of a non-human sialic Acid in human cancer.
- Author
-
Samraj AN, Läubli H, Varki N, and Varki A
- Published
- 2014
- Full Text
- View/download PDF
47. Rapid evolution of binding specificities and expression patterns of inhibitory CD33-related Siglecs in primates.
- Author
-
Padler-Karavani V, Hurtado-Ziola N, Chang YC, Sonnenburg JL, Ronaghy A, Yu H, Verhagen A, Nizet V, Chen X, Varki N, Varki A, and Angata T
- Subjects
- Animals, Antibody Specificity, Base Sequence, DNA Primers, Immunohistochemistry, Primates immunology, Sialic Acid Binding Ig-like Lectin 3 immunology, Sialic Acid Binding Immunoglobulin-like Lectins immunology
- Abstract
Siglecs are sialic acid-binding Ig-like lectins that recognize sialoglycans via amino-terminal V-set domains. CD33-related Siglecs (CD33rSiglecs) on innate immune cells recognize endogenous sialoglycans as "self-associated molecular patterns" (SAMPs), dampening immune responses via cytosolic immunoreceptor tyrosine-based inhibition motifs that recruit tyrosine phosphatases. However, sialic acid-expressing pathogens subvert this mechanism through molecular mimicry. Meanwhile, endogenous host SAMPs must continually evolve to evade other pathogens that exploit sialic acids as invasion targets. We hypothesized that these opposing selection forces have accelerated CD33rSiglec evolution. We address this by comparative analysis of major CD33rSiglec (Siglec-3, Siglec-5, and Siglec-9) orthologs in humans, chimpanzees, and baboons. Recombinant soluble molecules displaying ligand-binding domains show marked quantitative and qualitative interspecies differences in interactions with strains of the sialylated pathogen, group B Streptococcus, and with sialoglycans presented as gangliosides or in the form of sialoglycan microarrays, including variations such as N-glycolyl and O-acetyl groups. Primate Siglecs also show quantitative and qualitative intra- and interspecies variations in expression patterns on leukocytes, both in circulation and in tissues. Taken together our data explain why the CD33rSiglec-encoding gene cluster is undergoing rapid evolution via multiple mechanisms, driven by the need to maintain self-recognition by innate immune cells, while escaping 2 distinct mechanisms of pathogen subversion.
- Published
- 2014
- Full Text
- View/download PDF
48. Involvement of a non-human sialic Acid in human cancer.
- Author
-
Samraj AN, Läubli H, Varki N, and Varki A
- Abstract
Sialic acids are common monosaccharides that are widely expressed as outer terminal units on all vertebrate cell surfaces, and play fundamental roles in cell-cell and cell-microenvironment interactions. The predominant sialic acids on most mammalian cells are N-glycolylneuraminic acid (Neu5Gc) and N-acetylneuraminic acid (Neu5Ac). Neu5Gc is notable for its deficiency in humans due to a species-specific and species-universal inactivating deletion in the CMAH gene encoding the hydroxylase that converts CMP-Neu5Ac to CMP-Neu5Gc. However, Neu5Gc is metabolically incorporated into human tissues from dietary sources (particularly red meat), and detected at even higher levels in some human cancers. Early life exposure to Neu5Gc-containing foods in the presence of certain commensal bacteria that incorporate dietary Neu5Gc into lipooligosaccharides can lead to generation of antibodies that are also cross-reactive against Neu5Gc-containing glycans in human tissues ("xeno-autoantigens"). Such anti-Neu5Gc "xeno-autoantibodies" are found in all humans, although ranging widely in levels among individuals, and displaying diverse and variable specificities for the underlying glycan. Experimental evidence in a human-like Neu5Gc-deficient Cmah(-) (/) (-) mouse model shows that inflammation due to "xenosialitis" caused by this antigen-antibody interaction can promote tumor progression, suggesting a likely mechanism for the well-known epidemiological link between red meat consumption and carcinoma risk. In this review, we discuss the history of this field, mechanisms of Neu5Gc incorporation into tissues, the origin and specificities of human anti-Neu5Gc antibodies, their use as possible cancer biomarkers, implications of xenosialitis in cancer initiation and progression, and current and future approaches toward immunotherapy that could take advantage of this unusual human-specific phenomenon.
- Published
- 2014
- Full Text
- View/download PDF
49. Tumor infiltrating B-cells are increased in prostate cancer tissue.
- Author
-
Woo JR, Liss MA, Muldong MT, Palazzi K, Strasner A, Ammirante M, Varki N, Shabaik A, Howell S, Kane CJ, Karin M, and Jamieson CA
- Subjects
- Animals, Antigens, CD20 metabolism, Cell Count, Demography, Humans, Image Processing, Computer-Assisted, Immunohistochemistry, Male, Mice, Middle Aged, Prostatectomy, Prostatic Neoplasms surgery, Treatment Outcome, B-Lymphocytes immunology, B-Lymphocytes pathology, Lymphocytes, Tumor-Infiltrating immunology, Lymphocytes, Tumor-Infiltrating pathology, Prostatic Neoplasms immunology, Prostatic Neoplasms pathology
- Abstract
Background: The presence of increased B-cell tumor infiltrating lymphocytes (TILs) was seen in mouse prostate cancer (PCa) but has not been fully documented in human PCa. We, therefore, investigated the density of infiltrating B cells within human PCa utilizing a quantitative computational method., Methods: Archived radical prostatectomy specimens from 53 patients with known clinical outcome and D'Amico risk category were obtained and immunohistochemically (IHC) stained for the B cell marker, CD20. Slides were reviewed by a genitourinary pathologist who manually delineated the tumoral regions of PCa. Slides were digitally scanned and a computer algorithm quantified the area of CD20 stained B-cells as a measure of B cell density within the outlined regions of prostate cancer (intra-tumoral region), versus extra-tumoral prostate tissue. Correlations were analyzed between B-cell density and demographic and clinical variables, including D'Amico risk groups and disease recurrence., Results: For the entire cohort, the mean intra-tumoral B cell density was higher (3.22 SE = 0.29) than in the extra-tumoral region of each prostatectomy section (2.24, SE = 0.19) (paired t test; P < 0.001). When analyzed according to D'Amico risk group, the intra-tumoral B cell infiltration in low risk (0.0377 vs. 0.0246; p = 0.151) and intermediate risk (0.0260 vs. 0.0214; p = 0.579) patient prostatectomy specimens did not show significantly more B-cells within the PCa tumor. However, patient specimens from the high-risk group (0.0301 vs. 0.0197; p < 0.001) and from those who eventually had PCa recurrence or progression (0.0343 vs. 0.0246; p = 0.019) did show significantly more intra-tumoral CD20+ B-cell staining. Extent of B-cell infiltration in the prostatectomy specimens did not correlate with any other clinical parameters., Conclusions: Our study shows that higher B-cell infiltration was present within the intra-tumoral PCa regions compared to the extra-tumoral benign prostate tissue regions in prostatectomy sections. For this study we developed a new method to measure B-cells using computer-assisted digitized image analysis. Accurate, consistent quantitation of B-cells in prostatectomy specimens is essential for future clinical trials evaluating the effect of B cell ablating antibodies. The interaction of B-cells and PCa may serve as the basis for new therapeutic targets.
- Published
- 2014
- Full Text
- View/download PDF
50. Free fatty acids induce Lhb mRNA but suppress Fshb mRNA in pituitary LβT2 gonadotropes and diet-induced obesity reduces FSH levels in male mice and disrupts the proestrous LH/FSH surge in female mice.
- Author
-
Sharma S, Morinaga H, Hwang V, Fan W, Fernandez MO, Varki N, Olefsky JM, and Webster NJ
- Subjects
- Animals, Diet, High-Fat adverse effects, Dose-Response Relationship, Drug, Female, Follicle Stimulating Hormone, beta Subunit blood, Follicle Stimulating Hormone, beta Subunit genetics, Follicle Stimulating Hormone, beta Subunit metabolism, Gene Expression drug effects, Gonadotrophs cytology, Gonadotrophs metabolism, Immunoblotting, Luteinizing Hormone, beta Subunit blood, Luteinizing Hormone, beta Subunit genetics, Luteinizing Hormone, beta Subunit metabolism, Male, Mice, Mice, Inbred C57BL, Mitogen-Activated Protein Kinases metabolism, Obesity etiology, Obesity genetics, Ovary drug effects, Ovary metabolism, Pituitary Gland cytology, Proestrus genetics, Proestrus metabolism, RNA Interference, RNA, Messenger genetics, Reverse Transcriptase Polymerase Chain Reaction, Toll-Like Receptor 2 genetics, Toll-Like Receptor 2 metabolism, Toll-Like Receptor 4 genetics, Toll-Like Receptor 4 metabolism, Fatty Acids, Nonesterified pharmacology, Gonadotrophs drug effects, Obesity metabolism, Proestrus drug effects, RNA, Messenger metabolism
- Abstract
Female obesity is associated with insulin resistance, hyperandrogenemia, and reproductive dysfunction. We hypothesized that elevated free fatty acids (FFAs) might directly modulate pituitary gonadotropin production. FFAs caused a time- and dose-dependent increase in phosphorylation of the MAPKs p38MAPK, c-Jun N-terminal kinase (JNK)-1/2, and ERK1/2 in LβT2 gonadotrope cells. Furthermore, FFAs up-regulated Lhb mRNA expression acutely, an effect that was blocked by JNK inhibition, but suppressed Fshb mRNA expression, an effect that was independent of MAPK signaling. FFAs enhanced the activation of the MAPKs in the presence of GnRH, although the cotreatment did not alter Lhb induction but did eliminate the GnRH induction of Fshb. FFAs also suppressed activin-induced Fshb expression. Knockdown experiments showed that the FFA effect on the inflammatory kinases p38MAPK and JNK and on Lhb, but not Fshb, mRNA expression is mediated via toll-like receptor-2 and toll-like receptor-4 and was mimicked by lipopolysaccharide stimulation. In vivo, male C57BL/6 mice on a high-fat diet showed reduced FSH levels consistent with the suppression of Fshb seen in vitro. Histological analysis of the testes showed an increased number of abnormal seminiferous tubules. Female mice on a high-fat diet lacked the expected proestrus LH and FSH surge and exhibited an increase in the number of days at estrus and a reduced number of days at proestrus, and ovaries had significantly fewer corpora lutea. Taken together, our findings suggest that lipid excess can lead to reproductive defects in both male and female mice.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.