Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Malin Bäckström is active.

Publication


Featured researches published by Malin Bäckström.


Cellular and Molecular Life Sciences | 2011

Composition and functional role of the mucus layers in the intestine

Malin E. V. Johansson; Daniel Ambort; Thaher Pelaseyed; André Schütte; Jenny K. Gustafsson; Anna Ermund; Durai B. Subramani; Jessica Holmén-Larsson; Kristina A. Thomsson; Joakim H. Bergström; Sjoerd van der Post; Ana M. Rodríguez-Piñeiro; Henrik Sjövall; Malin Bäckström; Gunnar C. Hansson

In discussions on intestinal protection, the protective capacity of mucus has not been very much considered. The progress in the last years in understanding the molecular nature of mucins, the main building blocks of mucus, has, however, changed this. The intestinal enterocytes have their apical surfaces covered by transmembrane mucins and the whole intestinal surface is further covered by mucus, built around the gel-forming mucin MUC2. The mucus of the small intestine has only one layer, whereas the large intestine has a two-layered mucus where the inner, attached layer has a protective function for the intestine, as it is impermeable to the luminal bacteria.


Journal of Biological Chemistry | 2006

The ST6GalNAc-I Sialyltransferase Localizes throughout the Golgi and Is Responsible for the Synthesis of the Tumor-associated Sialyl-Tn O-Glycan in Human Breast Cancer

Robert Sewell; Malin Bäckström; Martin Dalziel; Steven Gschmeissner; Hasse Karlsson; Thomas Noll; Jochem Gätgens; Henrik Clausen; Gunnar C. Hansson; Joy Burchell; Joyce Taylor-Papadimitriou

The functional properties of glycoproteins are strongly influenced by their profile of glycosylation, and changes in this profile are seen in malignancy. In mucin-type O-linked glycosylation these changes can result in the production of mucins such as MUC1, carrying shorter sialylated O-glycans, and with different site occupancy. Of the tumor-associated sialylated O-glycans, the disaccharide, sialyl-Tn (sialic acid α2,6GalNAc), is expressed by 30% of breast carcinomas and is the most tumor-specific. The ST6GalNAc-I glycosyltransferase, which can catalyze the transfer of sialic acid to GalNAc, shows a highly restricted pattern of expression in normal adult tissues, being largely limited to the gastrointestinal tract and absent in mammary gland. In breast carcinomas, however, a complete correlation between the expression of RNA-encoding ST6GalNAc-I and the expression of sialyl-Tn is evident, demonstrating that the expression of sialyl-Tn results from switching on expression of hST6GalNAc-I. Endogenous or exogenous expression of hST6GalNAc-I (but not ST6GalNAc-II) always results in the expression of sialyl-Tn. This ability to override core 1/core 2 pathways of O- linked glycosylation is explained by the localization of ST6GalNAc-I, which is found throughout the Golgi stacks. The development of a Chinese hamster ovary (CHO) cell line expressing MUC1 and ST6GalNAc-I allowed the large scale production of MUC1 carrying 83% sialyl-Tn O-glycans. The presence of ST6GalNAc-I in the CHO cells reduced the number of O-glycosylation sites occupied in MUC1, from an average of 4.3 to 3.8 per tandem repeat. The availability of large quantities of this MUC1 glycoform will allow the evaluation of its efficacy as an immunogen for immunotherapy of MUC1/STn-expressing tumors.


Molecular & Cellular Proteomics | 2010

Comparison of Methods for Profiling O-Glycosylation HUMAN PROTEOME ORGANISATION HUMAN DISEASE GLYCOMICS/PROTEOME INITIATIVE MULTI-INSTITUTIONAL STUDY OF IgA1

Yoshinao Wada; Anne Dell; Stuart M. Haslam; Bérangère Tissot; Kevin Canis; Parastoo Azadi; Malin Bäckström; Catherine E. Costello; Gunnar C. Hansson; Yoshiyuki Hiki; Mayumi Ishihara; Hiromi Ito; Kazuaki Kakehi; Niclas G. Karlsson; Catherine E. Hayes; Koichi Kato; Nana Kawasaki; Kay Hooi Khoo; Kunihiko Kobayashi; Daniel Kolarich; Akihiro Kondo; Carlito B. Lebrilla; Miyako Nakano; Hisashi Narimatsu; Jan Novak; Milos V. Novotny; Erina Ohno; Nicolle H. Packer; Elizabeth Palaima; Matthew B. Renfrow

The Human Proteome Organisation Human Disease Glycomics/Proteome Initiative recently coordinated a multi-institutional study that evaluated methodologies that are widely used for defining the N-glycan content in glycoproteins. The study convincingly endorsed mass spectrometry as the technique of choice for glycomic profiling in the discovery phase of diagnostic research. The present study reports the extension of the Human Disease Glycomics/Proteome Initiatives activities to an assessment of the methodologies currently used for O-glycan analysis. Three samples of IgA1 isolated from the serum of patients with multiple myeloma were distributed to 15 laboratories worldwide for O-glycomics analysis. A variety of mass spectrometric and chromatographic procedures representative of current methodologies were used. Similar to the previous N-glycan study, the results convincingly confirmed the pre-eminent performance of MS for O-glycan profiling. Two general strategies were found to give the most reliable data, namely direct MS analysis of mixtures of permethylated reduced glycans in the positive ion mode and analysis of native reduced glycans in the negative ion mode using LC-MS approaches. In addition, mass spectrometric methodologies to analyze O-glycopeptides were also successful.


Molecular & Cellular Proteomics | 2009

Comparison of Methods for Profiling O-glycosylation: HUPO Human Disease Glycomics/Proteome Initiative Multi-Institutional Study of IgA1

Yoshinao Wada; Anne Dell; Stuart M. Haslam; Bérangère Tissot; Kevin Canis; Parastoo Azadi; Malin Bäckström; Catherine E. Costello; Gunnar C. Hansson; Yoshiyuki Hiki; Mayumi Ishihara; Hiromi Ito; Kazuaki Kakehi; Niclas G. Karlsson; Koichi Kato; Nana Kawasaki; Kay-Hooi Khoo; Kunihiko Kobayashi; Daniel Kolarich; Akihiro Kondo; Carlito B. Lebrilla; Miyako Nakano; Hisashi Narimatsu; Jan Novak; Milos V. Novotny; Erina Ohno; Nicolle H. Packer; Matthew B. Renfrow; Michiko Tajiri; Naoyuki Taniguchi

The Human Proteome Organisation Human Disease Glycomics/Proteome Initiative recently coordinated a multi-institutional study that evaluated methodologies that are widely used for defining the N-glycan content in glycoproteins. The study convincingly endorsed mass spectrometry as the technique of choice for glycomic profiling in the discovery phase of diagnostic research. The present study reports the extension of the Human Disease Glycomics/Proteome Initiatives activities to an assessment of the methodologies currently used for O-glycan analysis. Three samples of IgA1 isolated from the serum of patients with multiple myeloma were distributed to 15 laboratories worldwide for O-glycomics analysis. A variety of mass spectrometric and chromatographic procedures representative of current methodologies were used. Similar to the previous N-glycan study, the results convincingly confirmed the pre-eminent performance of MS for O-glycan profiling. Two general strategies were found to give the most reliable data, namely direct MS analysis of mixtures of permethylated reduced glycans in the positive ion mode and analysis of native reduced glycans in the negative ion mode using LC-MS approaches. In addition, mass spectrometric methodologies to analyze O-glycopeptides were also successful.


Biochemical Journal | 2003

Recombinant MUC1 mucin with a breast cancer-like O-glycosylation produced in large amounts in Chinese-hamster ovary cells.

Malin Bäckström; Thomas Link; Fredrik J. Olson; Hasse Karlsson; Rosalind Graham; Gianfranco Picco; Joy Burchell; Joyce Taylor-Papadimitriou; Thomas Noll; Gunnar C. Hansson

We have developed an expression system for the production of large quantities of recombinant MUC1 mucin in CHO-K1 (Chinese-hamster ovary K1) cells. The extracellular part of human MUC1, including 16 MUC1 tandem repeats, was produced as a fusion protein with murine IgG Fc, with an intervening enterokinase cleavage site for the removal of the Fc tail. Stable MUC1-IgG-producing CHO-K1 clones were generated and were found to secrete MUC1-IgG into the culture medium. After adaptation to suspension culture in protein-free medium in a bioreactor, the fusion protein was secreted in large quantities (100 mg/l per day) into the culture supernatant. From there, MUC1 could be purified to homogeneity using a two-step procedure including enterokinase cleavage and ion-exchange chromatography. Capillary liquid chromatography MS of released oligosaccharides from CHO-K1-produced MUC1 identified the main O-glycans as Galbeta1-3GalNAc (core 1) and mono- and di-sialylated core 1. The glycans occupied on average 4.3 of the five potential O-glycosylation sites in the tandem repeats, as determined by nano-liquid chromatography MS of partially deglycosylated Clostripain-digested protein. A very similar O-glycan profile and site occupancy was found in MUC1-IgG produced in the breast carcinoma cell line T47D, which has O-glycosylation typical for breast cancer. In contrast, MUC1-IgG produced in another breast cancer cell line, MCF-7, showed a more complex pattern with both core 1- and core 2-based O-glycans. This is the first reported production of large quantities of recombinant MUC1 with a breast cancer-like O-glycosylation that could be used for the immunotherapy of breast cancer.


Journal of Immunology | 2005

Recombinant tumor-associated MUC1 glycoprotein impairs the differentiation and function of dendritic cells

Aurelia Rughetti; Ilenia Pellicciotta; Mauro Biffoni; Malin Bäckström; Thomas Link; Eric P. Bennet; Henrik Clausen; Thomas Noll; Gunnar C. Hansson; Joy Burchell; Luigi Frati; Joyce Taylor-Papadimitriou; Marianna Nuti

Tumors exploit several strategies to evade immune recognition, including the production of a large number of immunosuppressive factors, which leads to reduced numbers and impaired functions of dendritic cells (DCs) in the vicinity of tumors. We have investigated whether a mucin released by tumor cells could be involved in causing these immunomodulating effects on DCs. We used a recombinant purified form of the MUC1 glycoprotein, an epithelial associated mucin that is overexpressed, aberrantly glycosylated, and shed during cancer transformation. The O-glycosylation profile of the recombinant MUC1 glycoprotein (ST-MUC1) resembled that expressed by epithelial tumors in vivo, consisting of large numbers of sialylated core 1 (sialyl-T, ST) oligosaccharides. When cultured in the presence of ST-MUC1, human monocyte-derived DCs displayed a modified phenotype with decreased expression of costimulatory molecules (CD86, CD40), Ag-presenting molecules (DR and CD1d), and differentiation markers (CD83). In contrast, markers associated with an immature phenotype, CD1a and CD206 (mannose receptor), were increased. This effect was already evident at day 4 of DC culture and was dose dependent. The modified phenotype of DCs corresponded to an altered balance in IL-12/IL-10 cytokine production, with DC expressing an IL-10highIL-12low phenotype after exposure to ST-MUC1. These DCs were defective in their ability to induce immune responses in both allogeneic and autologous settings, as detected in proliferation and ELISPOT assays. The altered DC differentiation and Ag presentation function induced by the soluble sialylated tumor-associated mucin may represent a mechanism by which epithelial tumors can escape immunosurveillance.


Journal of Biological Chemistry | 2013

Site-specific O-Glycosylation on the MUC2 Mucin Protein Inhibits Cleavage by the Porphyromonas gingivalis Secreted Cysteine Protease (RgpB)

Sjoerd van der Post; Durai B. Subramani; Malin Bäckström; Malin E. V. Johansson; Malene Bech Vester-Christensen; Ulla Mandel; Eric P. Bennett; Henrik Clausen; Gunnar Dahlén; Aneta Sroka; Jan Potempa; Gunnar C. Hansson

Background: MUC2 polymers form the mucus layer of colon that separates luminal bacteria from the epithelium. Results: P. gingivalis secrets a protease that cleaves the MUC2 mucin, a cleavage modulated by O-glycosylation. Conclusion: Bacteria can disrupt the MUC2 polymer via proteolytic cleavage. However, O-glycosylation can inhibit this process. Significance: Bacteria can dissolve the protective inner mucus layer, potentially triggering colitis. The colonic epithelial surface is protected by an inner mucus layer that the commensal microflora cannot penetrate. We previously demonstrated that Entamoeba histolytica secretes a protease capable of dissolving this layer that is required for parasite penetration. Here, we asked whether there are bacteria that can secrete similar proteases. We screened bacterial culture supernatants for such activity using recombinant fragments of the MUC2 mucin, the major structural component, and the only gel-forming mucin in the colonic mucus. MUC2 has two central heavily O-glycosylated mucin domains that are protease-resistant and has cysteine-rich N and C termini responsible for polymerization. Culture supernatants of Porphyromonas gingivalis, a bacterium that secretes proteases responsible for periodontitis, cleaved the MUC2 C-terminal region, whereas the N-terminal region was unaffected. The active enzyme was isolated and identified as Arg-gingipain B (RgpB). Two cleavage sites were localized to IR↓TT and NR↓QA. IR↓TT cleavage will disrupt the MUC2 polymers. Because this site has two potential O-glycosylation sites, we tested whether recombinant GalNAc-transferases (GalNAc-Ts) could glycosylate a synthetic peptide covering the IRTT sequence. Only GalNAc-T3 was able to glycosylate the second Thr in IRTT, rendering the sequence resistant to cleavage by RgpB. Furthermore, when GalNAc-T3 was expressed in CHO cells expressing the MUC2 C terminus, the second threonine was glycosylated, and the protein became resistant to RgpB cleavage. These findings suggest that bacteria can produce proteases capable of dissolving the inner protective mucus layer by specific cleavages in the MUC2 mucin and that this cleavage can be modulated by site-specific O-glycosylation.


Vaccine | 2010

Intranasal immunization with a proteoliposome-derived cochleate containing recombinant gD protein confers protective immunity against genital herpes in mice

Judith del Campo; Madelene Lindqvist; Maribel Cuello; Malin Bäckström; Osmir Cabrerra; Josefine Persson; Oliver Pérez; Ali M. Harandi

The purpose of this study was to investigate the potential of intranasal (IN) immunization with Neisseria meningitides B proteoliposome (AFPL1) and AFPL1-derived cochleate (AFCo1), containing glycoprotein D (gD) of herpes simplex virus type 2 (HSV-2) for induction of protective immunity against genital herpes infection in mice. We could show that IN immunization with both AFPL1 and AFCo1 containing gD induced gD-specific IgG antibody and lymphoproliferative responses. However, IFN-gamma response could only be detected in CD4(+) splenic cells and genital lymph node cells of the AFCo1gD immunized mice upon recall antigen stimulation in vitro. Importantly, IN immunization with AFCo1gD could elicit a complete protection against an otherwise lethal vaginal challenge with HSV-2, while the AFPL1gD immunized mice were only partially protected. Further, we could show that the IFN-gamma response and protective immunity observed after IN immunization with AFCo1gD are mediated via the adaptor molecule myeloid differentiation factor 88. These data may have implications for the development of a mucosal vaccine against genital herpes.


Gene | 1994

Insertion of a HIV-1-neutralizing epitope in a surface-exposed internal region of the cholera toxin B-subunit.

Malin Bäckström; Michael Lebens; Florian Schödel; Jan Holmgren

The non-toxic B-subunit of cholera toxin (CTB) is a powerful immunogen and has been investigated as a carrier for foreign peptide epitopes, with peptides genetically fused to either the N- or C terminus of CTB. In the present study, we have constructed a plasmid encoding a novel intrachain CTB fusion protein with a peptide epitope inserted into an internal region of CTB: eight amino acids (aa) in CTB (56-63) were substituted with a 10-aa peptide from the third variable (V3) loop of the HIV-1 envelope protein gp120. The resulting chimeric protein retained important functional characteristics of the native CTB including pentamerization and GM1 ganglioside receptor binding. The internal hybrid protein was also shown to be resistant to proteolytic degradation during production in Vibrio cholerae, whereas a terminal hybrid protein, where the same gp120-epitope was fused to the N terminus of CTB, was rapidly cleaved during culture. The inserted epitope, which is known to give rise to HIV-1 neutralizing Ab, could be detected with a V3 loop-specific monoclonal Ab when the chimeric protein was analyzed in ELISA and immunoblot, indicating that the epitope inserted at this site is presented on the surface of the protein. Consistent with these observations, immunization of mice with the CTB::HIV hybrid protein elicited a high titered serum Ab response to the CTB moiety and also, in some but not all animals, a detectable response to the inserted gp120 epitope.


Vaccine | 2003

A mucosally administered recombinant fusion protein vaccine against schistosomiasis protecting against immunopathology and infection

Michael Lebens; Jia-Bin Sun; Hamid Sadeghi; Malin Bäckström; Ida Olsson; Nathalie Mielcarek; Bin-Ling Li; André Capron; Cecil Czerkinsky; Jan Holmgren

We have constructed and efficiently produced and purified a candidate vaccine against schistosomiasis consisting of a novel hybrid protein in which two dominant T- and B-cell epitopes from Schistosoma mansoni 28 kDa glutathione-S-transferase (Sm28GST) antigen (a.a 24-43 and 191-212) are fused to cholera toxin B subunit (CTB). Intranasal treatment of S. mansoni-infected mice with the hybrid protein, which similar to native CTB was assembled into receptor binding pentamers, significantly reduced total worm burden and liver egg counts due to the induction of Sm28GST-specific antibodies. Immunopathologic granuloma formation in the liver was also significantly suppressed and there was an almost complete suppression of delayed-type hypersensitivity reactions to both Sm28GST and to total soluble egg antigen in infected animals. The results suggest that this type of hybrid protein could be used as a combined anti-immunopathology and anti-infection vaccine against schistosomiasis.

Collaboration


Dive into the Malin Bäckström's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hasse Karlsson

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar

Michael Lebens

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Thomas Link

Forschungszentrum Jülich

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jan Holmgren

University of Gothenburg

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge