Network


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

Hotspot


Dive into the research topics where Johanna Vendelin is active.

Publication


Featured researches published by Johanna Vendelin.


Journal of Medical Genetics | 2007

The human GIMAP5 gene has a common polyadenylation polymorphism increasing risk to systemic lupus erythematosus

Anna Hellquist; Marco Zucchelli; Katja Kivinen; Ulpu Saarialho-Kere; Sari Koskenmies; Elisabeth Widen; Heikki Julkunen; Andrew Wong; Marja-Liisa Karjalainen-Lindsberg; Tiina Skoog; Johanna Vendelin; Deborah S. Cunninghame-Graham; Timothy J. Vyse; Juha Kere; Cecilia M. Lindgren

Background: Several members of the GIMAP gene family have been suggested as being involved in different aspects of the immune system in different species. Recently, a mutation in the GIMAP5 gene was shown to cause lymphopenia in a rat model of autoimmune insulin-dependent diabetes. Thus it was hypothesised that genetic variation in GIMAP5 may be involved in susceptibility to other autoimmune disorders where lymphopenia is a key feature, such as systemic lupus erythematosus (SLE). Material and methods: To investigate this, seven single nucleotide polymorphisms in GIMAP5 were analysed in five independent sets of family-based SLE collections, containing more than 2000 samples. Result: A significant increase in SLE risk associated with the most common GIMAP5 haplotype was found (OR 1.26, 95% CI 1.02 to 1.54, p = 0.0033). In families with probands diagnosed with trombocytopenia, the risk was increased (OR 2.11, 95% CI 1.09 to 4.09, p = 0.0153). The risk haplotype bears a polymorphic polyadenylation signal which alters the 3′ part of GIMAP5 mRNA by producing an inefficient polyadenylation signal. This results in higher proportion of non-terminated mRNA for homozygous individuals (p<0.005), a mechanism shown to be causal in thalassaemias. To further assess the functional effect of the polymorphic polyadenylation signal in the risk haplotype, monocytes were treated with several cytokines affecting apoptosis. All the apoptotic cytokines induced GIMAP5 expression in two monocyte cell lines (1.5–6 times, p<0.0001 for all tests). Conclusion: Taken together, the data suggest the role of GIMAP5 in the pathogenesis of SLE.


Neurogastroenterology and Motility | 2009

Neuropeptide S receptor 1 expression in the intestine and skin – putative role in peptide hormone secretion

L. Sundman; Ulpu Saarialho-Kere; Johanna Vendelin; Katri Lindfors; G. Assadi; Katri Kaukinen; M. Westerholm‐ormio; Erkki Savilahti; Markku Mäki; Harri Alenius; M. D’amato; Ville Pulkkinen; Juha Kere; Päivi Saavalainen

Abstract  Neuropeptide S receptor 1 (NPSR1) was recently found to be genetically associated with inflammatory bowel disease in addition to asthma and related traits. Epithelia of several organs express NPSR1 isoforms A and B, including the intestine and the skin, and NPSR1 appears to be upregulated in inflammation. In this study, we used cell lines and tissue samples to characterize the expression of NPSR1 and its ligand neuropeptide S (NPS) in inflammation. We used polyclonal and monoclonal antibodies to investigate the expression of NPS and NPSR1 in intestinal diseases, such as celiac disease and food allergy, and in cutaneous inflammatory disorders. We found that NPSR1‐A was expressed by the enteroendocrine cells of the gut. Overall, the expression pattern of NPS was similar to its receptor suggesting an autocrine mechanism. In an NPSR1‐A overexpressing cell model, stimulation with NPS resulted in a dose‐dependent upregulation of glycoprotein hormone, alpha polypeptide (CGA), tachykinin 1 (TAC1), neurotensin (NTS) and galanin (GAL) encoding peptide hormones secreted by enteroendocrine cells. Because NPSR1 was also expressed in macrophages, neutrophils, and intraepithelial lymphocytes, we demonstrated that stimulation with the pro‐inflammatory cytokines tumour necrosis factor alpha and interferon gamma increased NPSR1 expression in the THP‐1 monocytic cells. In conclusion, similar to other neuropeptides and their receptors, NPSR1 signalling might play a dual role along the gut–brain axis. The NPS/NPSR1 pathway may participate in the regulation of the peptide hormone production in enteroendocrine cells of the small intestine.


Human Molecular Genetics | 2008

Biological and genetic interaction between Tenascin C and Neuropeptide S receptor 1 in allergic diseases

Christina Orsmark-Pietras; Erik Melén; Johanna Vendelin; Sara Bruce; Annika Laitinen; Lauri A. Laitinen; Roger Lauener; Josef Riedler; Erika von Mutius; Gert Doekes; Magnus Wickman; Marianne van Hage; Göran Pershagen; Annika Scheynius; Fredrik Nyberg; Juha Kere

Neuropeptide S receptor 1 (NPSR1, GPRA 154, GPRA) has been verified as a susceptibility gene for asthma and related phenotypes. The ligand for NPSR1, Neuropeptide S (NPS), activates signalling through NPSR1 and microarray analysis has identified Tenascin C (TNC) as a target gene of NPS-NPSR1 signalling. TNC has previously been implicated as a risk gene for asthma. We aimed therefore to study the genetic association of TNC in asthma- and allergy-related disorders as well as the biological and genetic interactions between NPSR1 and TNC. Regulation of TNC was investigated using NPS stimulated NPSR1 transfected cells. We genotyped 12 TNC SNPs in the cross-sectional PARSIFAL study (3113 children) and performed single SNP association, haplotype association and TNC and NPSR1 gene-gene interaction analyses. Our experimental results show NPS-dependent upregulation of TNC-mRNA. The genotyping results indicate single SNP and haplotype associations for several SNPs in TNC with the most significant association to rhinoconjunctivitis for a haplotype, with a frequency of 29% in cases (P = 0.0005). In asthma and atopic sensitization significant gene-gene interactions were found between TNC and NPSR1 SNPs, indicating that depending on the NPSR1 genotype, TNC can be associated with either an increased or a decreased risk of disease. We conclude that variations in TNC modifies, not only risk for asthma, but also for rhinoconjunctivitis. Furthermore, we show epistasis based on both a direct suggested regulatory effect and a genetic interaction between NPSR1 and TNC. These results suggest merging of previously independent pathways of importance in the development of asthma- and allergy-related traits.


PLOS ONE | 2013

Interaction between Retinoid Acid Receptor-Related Orphan Receptor Alpha (RORA) and Neuropeptide S Receptor 1 (NPSR1) in Asthma

Nathalie Acevedo; Annika Sääf; Cilla Söderhäll; Erik Melén; Jami Mandelin; Christina Orsmark Pietras; Sini Ezer; Johanna Vendelin; Gustav Boije af Gennäs; Jari Yli-Kauhaluoma; Harri Alenius; Erika von Mutius; Gert Doekes; Charlotte Braun-Fahrländer; Josef Riedler; Marianne van Hage; Mauro D’Amato; Annika Scheynius; Göran Pershagen; Juha Kere; Ville Pulkkinen

Retinoid acid receptor-related Orphan Receptor Alpha (RORA) was recently identified as a susceptibility gene for asthma in a genome-wide association study. To investigate the impact of RORA on asthma susceptibility, we performed a genetic association study between RORA single nucleotide polymorphisms (SNPs) in the vicinity of the asthma-associated SNP (rs11071559) and asthma-related traits. Because the regulatory region of a previously implicated asthma susceptibility gene, Neuropeptide S receptor 1 (NPSR1), has predicted elements for RORA binding, we hypothesized that RORA may interact biologically and genetically with NPSR1. 37 RORA SNPs and eight NPSR1 SNPs were genotyped in the Swedish birth cohort BAMSE (2033 children) and the European cross-sectional PARSIFAL study (1120 children). Seven RORA SNPs confined into a 49 kb region were significantly associated with physician-diagnosed childhood asthma. The most significant association with rs7164773 (T/C) was driven by the CC genotype in asthma cases (OR = 2.0, 95%CI 1.36–2.93, p = 0.0003 in BAMSE; and 1.61, 1.18–2.19, p = 0.002 in the combined BAMSE-PARSIFAL datasets, respectively), and strikingly, the risk effect was dependent on the Gln344Arg mutation in NPSR1. In cell models, stimulation of NPSR1 activated a pathway including RORA and other circadian clock genes. Over-expression of RORA decreased NPSR1 promoter activity further suggesting a regulatory loop between these genes. In addition, Rora mRNA expression was lower in the lung tissue of Npsr1 deficient mice compared to wildtype littermates during the early hours of the light period. We conclude that RORA SNPs are associated with childhood asthma and show epistasis with NPSR1, and the interaction between RORA and NPSR1 may be of biological relevance. Combinations of common susceptibility alleles and less common functional polymorphisms may modify the joint risk effects on asthma susceptibility.


BMC Pulmonary Medicine | 2011

The asthma candidate gene NPSR1 mediates isoform specific downstream signalling

Christina Orsmark Pietras; Johanna Vendelin; Francesca Anedda; Sara Bruce; Mikael Adner; Lilli Sundman; Ville Pulkkinen; Harri Alenius; Mauro D'Amato; Cilla Söderhäll; Juha Kere

BackgroundNeuropeptide S Receptor 1 (NPSR1, GPRA, GPR154) was first identified as an asthma candidate gene through positional cloning and has since been replicated as an asthma and allergy susceptibility gene in several independent association studies. In humans, NPSR1 encodes two G protein-coupled receptor variants, NPSR1-A and NPSR1-B, with unique intracellular C-termini. Both isoforms show distinct expression pattern in asthmatic airways. Although NPSR1-A has been extensively studied, functional differences and properties of NPSR1-B have not yet been clearly examined. Our objective was to investigate downstream signalling properties of NPSR1-B and functional differences between NPSR1-A and NPSR1-B.MethodsHEK-293 cells transiently overexpressing NPSR1-A or NPSR1-B were stimulated with the ligand neuropeptide S (NPS) and downstream signalling effects were monitored by genome-scale affymetrix expression-arrays. The results were verified by NPS concentration-response and time series analysis using qRT-PCR, cAMP and Ca2+ assays, and cAMP/PKA, MAPK/JNK and MAPK/ERK pathway specific reporter assays.ResultsNPSR1-B signalled through the same pathways and regulated the same genes as NPSR1-A, but NPSR1-B yielded lower induction on effector genes than NPSR1-A, with one notable exception, CD69, a marker of regulatory T cells.ConclusionsWe conclude that NPSR1-B is regulating essentially identical set of genes as NPSR1-A, with few, but possibly important exceptions, and that NPSR1-A induces stronger signalling effects than NPSR1-B. Our findings suggest an isoform-specific link to pathogenetic processes in asthma and allergy.


Science | 2004

Characterization of a Common Susceptibility Locus for Asthma-Related Traits

Tarja Laitinen; Anne Polvi; Pia Rydman; Johanna Vendelin; Ville Pulkkinen; Paula Salmikangas; Siru Mäkelä; Marko Rehn; Asta Pirskanen; Anna Rautanen; Marco Zucchelli; Harriet Gullstén; Marina Leino; Harri Alenius; Tuula Petäys; Tari Haahtela; Annika Laitinen; Catherine Laprise; Thomas J. Hudson; Lauri A. Laitinen; Juha Kere


American Journal of Respiratory Cell and Molecular Biology | 2005

Characterization of GPRA, a Novel G Protein–Coupled Receptor Related to Asthma

Johanna Vendelin; Ville Pulkkinen; Marko Rehn; Asta Pirskanen; Anne Räisänen-Sokolowski; Annika Laitinen; Lauri A. Laitinen; Juha Kere; Tarja Laitinen


Human Molecular Genetics | 2006

Neuropeptide S and G protein-coupled receptor 154 modulate macrophage immune responses

Ville Pulkkinen; Marja-Leena Majuri; Guoying Wang; P. Holopainen; Yasushi Obase; Johanna Vendelin; Henrik Wolff; Paula Rytilä; Lauri A. Laitinen; Tari Haahtela; Tarja Laitinen; Harri Alenius; Juha Kere; Marko Rehn


Human Molecular Genetics | 2006

Downstream target genes of the neuropeptide S–NPSR1 pathway

Johanna Vendelin; Sara Bruce; P. Holopainen; Ville Pulkkinen; Paula Rytilä; Asta Pirskanen; Marko Rehn; Tarja Laitinen; Lauri A. Laitinen; Tari Haahtela; Ulpu Saarialho-Kere; Annika Laitinen; Juha Kere


Archive | 2008

Asthma Susceptibility Locus

Tarja Laitinen; Juha Kere; Lauri A. Laitinen; Anne Polvi; Siru Mäkelä; Johanna Vendelin; Ville Pulkkinen; Paula Salminkangas

Collaboration


Dive into the Johanna Vendelin's collaboration.

Top Co-Authors

Avatar

Juha Kere

Karolinska Institutet

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lauri A. Laitinen

Helsinki University Central Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge