Druggability of neurological disease associated cell signalling
Our lab develops and adapts molecular tools we then use to identify druggable mechanisms of synaptic dysfunction implicated in neurological disease. Our current focus is on Ras-signalling related rare neurodevelopmental disorders and Alzheimer’s Disease. Together with corresponding data analytical pipelines, some tools may also be used in screening assays, either for the generation of chemical biology reagents supporting target validation, or to screen drug or diversity libraries for repurposing or drug discovery.
As cell-based models of disease become more realistic and complex, mechanistic investigations and druggability studies demand precise and selective analytical tools and approaches. Longitudinal follow-up in a live-cell imaging setting is increasingly required. Our lab applies parallelised longitudinal imaging and optical actuation of complex cell systems, paired with the use of proteome-wide and proximity analysis at bulk and single synapse levels. These approaches are supported by advanced instrumentation at the core facilities situated in our department, especially the high-throughput microscopy instrumentation and other automation platforms at the Turku Screening Unit as well as the Turku Proteomics Core
Key words:
Neurobiology, Alzheimer’s disease, Disease mechanisms, Optical Pathway Reporters, Optogenetics, Protein engineering, Image processing, Data analysis, Druggability, Drug discovery
Selected Earlier Results:
Our lab previously identified druggable sites on proteins forming a novel disease associated pathway involving nNOS and NOS1AP. We developed molecules targeting the pathway, enabling us to demonstrate in vivo POC in several disease models (1-5). To gain further insight in disease mechanisms, we developed a simple optogenetic tool design strategy that allowed temporally precise encoding of pathway inhibition (6). We also identified methods to ease the application of optogenetics, for example by reducing blue-light burden of long-term use and developing a method for real time direct monitoring of switching and relaxation to support the optimisation of actuation protocols (7).
Current Research Activities :
We design new optical reporters, optogenetic actuators, labelling strategies and data analysis pipelines to investigate synaptopathies. One particular aim is to reveal the functional deficits of missense variants of SynGAP1 known to cause the rare disease SynGAP non-syndromic intellectual disability (8-9). Assays developed using these tools may identify neomorphic properties and, in future studies, support drug repurposing screens and help clarify the impact of variants of uncertain significance (VUS). Another project aims to define phenotypic response signatures in human neuron models of a panel of rare diseases to carry out repurposing drug screens. A more basic research project examines the turnover, protein composition and function of the neuronal synapse (10) and how it is regulated by adaptor proteins like NOS1AP, and another explores the roles of temporal encoding of neuronal signalling pathways and imbalances in the synaptic proteome on regulation of connectivity and disease-associated synaptic dysfunction.
Collaborations and network:
Local collaborations include those with Assoc. Prof. Pekka Postila and Prof. Olli Pentikäinen (Institute of Biomedicine) on the structural biology of SynGAP1, with Prof. Pekka Ruusuvuori on the applications of machine learning to data analysis pipelines, and Assoc. Prof. Otto Kauko on development of single synapse proteomics methods. International collaborations include those with Dr. Florian Freudenberg (University of Frankfurt) on NOS1AP function in psychiatric disorder endophenotypes, and the EURAS consortium (www.euras-project.eu/) on neurodevelopmental rasopathies. In addition, our team is responsible for the Lab Automation site of the Turku Screening Unit, which represents Turku at the National Drug Discovery and Chemical Biology Platform of Biocenter Finland and is a partner site of the European infrastructure project EU-OPENSCREEN.
Selected publications:
- Li LL, Ginet V, Liu X, Vergun O, Tuittila M, Mathieu M, Bonny C, Puyal J, Truttmann AC, Courtney MJ. The nNOS-p38MAPK pathway is mediated by NOS1AP during neuronal death. J Neurosci. 2013 May 8;33(19):8185-201. doi: 10.1523/JNEUROSCI.4578-12.2013. PMID: 23658158; PMCID: PMC6619617.
- Li LL, Melero-Fernandez de Mera RM, Chen J, Ba W, Kasri NN, Zhang M, Courtney MJ. Unexpected Heterodivalent Recruitment of NOS1AP to nNOS Reveals Multiple Sites for Pharmacological Intervention in Neuronal Disease Models. J Neurosci. 2015 May 13;35(19):7349-64. doi: 10.1523/JNEUROSCI.0037-15.2015. PMID: 25972165; PMCID: PMC6705433.
- Lee WH, Carey LM, Li LL, Xu Z, Lai YY, Courtney MJ, Hohmann AG. ZLc002, a putative small-molecule inhibitor of nNOS interaction with NOS1AP, suppresses inflammatory nociception and chemotherapy-induced neuropathic pain and synergizes with paclitaxel to reduce tumor cell viability. Mol Pain. 2018 Jan-Dec;14:1744806918801224. doi: 10.1177/1744806918801224. Epub 2018 Aug 29. PMID: 30157705; PMCID: PMC6144507.
- Lee WH, Carey LM, Li LL, Xu Z, Lai YY, Courtney MJ, Hohmann AG. ZLc002, a putative small-molecule inhibitor of nNOS interaction with NOS1AP, suppresses inflammatory nociception and chemotherapy-induced neuropathic pain and synergizes with paclitaxel to reduce tumor cell viability. Mol Pain. 2018 Jan-Dec;14:1744806918801224. doi: 10.1177/1744806918801224. Epub 2018 Aug 29. PMID: 30157705; PMCID: PMC6144507.
- Esen-Sehir D, Li L, Reif A, Courtney MJ, Freudenberg F (2020) P.504 Targeting nitric oxide synthase 1 adaptor protein interactions as a possible treatment for schizophrenia: an initial proof-of-concept study in mice, European Neuropsychopharmacology, Volume 40, Supplement 1, 2020, Pages S283, ISSN 0924-977X, https://doi.org/10.1016/j.euroneuro.2020.09.368 (https://www.sciencedirect.com/science/article/pii/S0924977X20306416)
- Melero-Fernandez de Mera RM, Li LL, Popinigis A, Cisek K, Tuittila M, Yadav L, Serva A, Courtney MJ. A simple optogenetic MAPK inhibitor design reveals resonance between transcription-regulating circuitry and temporally-encoded inputs. Nat Commun. 2017 May 12;8:15017. doi: 10.1038/ncomms15017. PMID: 28497795; PMCID: PMC5437309.
- Li LL, Klein FM, Li Greci L, Popinigis A, Freudenberg F, Courtney MJ. Resonance energy transfer sensitises and monitors in situ switching of LOV2-based optogenetic actuators. Nat Commun. 2020 Oct 9;11(1):5107. doi: 10.1038/s41467-020-18816-8. PMID: 33037199; PMCID: PMC7547724.
- Courtney MJ et al. 2021, Webinar “Investigating the functional single cell biology of SynGAP1 pathways”, SynGAP research fund, USA (virtual) webinar https://www.syngapresearchfund.org/webinars/investigating-the-functional-single-cell-biology-of-syngap1-pathways
- Yotova AY, Li LL, O'Leary A, Tegeder I, Reif A, Courtney MJ, Slattery DA, Freudenberg F. Embryonic and adult synaptic proteome perturbations after maternal immune activation: Identification of persistent changes relevant for early intervention. Res Sq [Preprint]. 2023 Jul 3:rs.3.rs-3100753. doi: 10.21203/rs.3.rs-3100753/v1. PMID: 37461513; PMCID: PMC10350178.
Michael Courtney
Group Leader
Computational tools for cellular communication and multi-omics integration in metabolic diseases
This project develops advanced computational methods for analyzing cellular communication and its disruption in metabolic diseases, with a specific focus on obesity. By integrating single-cell and multi-omics data (e.g. transcriptomics, epigenomics, proteomics), we aim to reveal molecular pathways that drive disease progression in obesity and to identify key regulatory factors involved. A particular focus is on understanding how cells in different tissues communicate and how these interactions are disrupted in metabolic disorders.
We will develop new computational methods and models to investigate how cellular behavior and communication change over time and their role in disease progression. Advanced machine learning techniques and graph-based approaches will be utilized to infer key regulatory networks and to identify key cellular components and pathways that drive metabolic disorders. Integration of multi-omics and spatial datasets will enable a comprehensive view of these processes. The ultimate goal is to identify new biomarkers or therapeutic targets that would facilitate new strategies for prevention or treatment of obesity and related metabolic diseases.
Although the primary focus is on obesity and metabolic diseases, the computational tools developed will be widely applicable to other diseases and complex biological systems. The tools will be made available as open-source software to benefit the broader research community.
This project is well-suited for candidates with expertise in computational biology, bioinformatics, machine learning, or related fields.
Selected publications:
- Smolander J, Junttila S, Elo LL. Cell-connectivity-guided trajectory inference from single-cell data. Bioinformatics 39(9):btad515, 2023.
- Biehl A, Venäläinen MS, Suojanen LU, Kupila S, Ahola AJ, Pietiläinen KH, Elo LL. Development and validation of a weight-loss predictor to assist weight loss management. Sci Rep 13(1):20661, 2023.
- Suomi T*, Starskaia I*, Kalim UU*, Rasool O, Jaakkola MK, Grönroos T, Välikangas T, Brorson C, Mazzoni G, Bruggraber S, Overbergh L, Dunger D, Peakman M, Chmura P, Brunak S, Schulte AM, Mathieu C, Knip M, Lahesmaa R*, Elo LL*. Gene expression signature predicts rate of type 1 diabetes progression. eBioMedicine 92:104625, 2023
- Välikangas T, Suomi T, Chandler CE, Scott AJ, Tran BQ, Ernst RK, Goodlett DR, Elo LL. Benchmarking tools for detecting longitudinal differential expression in proteomics data allows establishing a robust reproducibility optimization regression approach. Nat Commun 13(1):7877, 2022.
Laura Elo
Research Director, Turku Bioscience Centre
Professor, Turku Bioscience Centre
InFLAMES Flagship
Towards a better understanding of enterovirus infections – innovative strategies for improved detection, prevention and treatment
Viruses are the most abundant biological entity on Earth infecting all forms of cellular life and a major cause of human infectious disease. Our example virus family of Picornaviridae consist of more than 300 known human pathogenic viruses with a diverse range of transmission routes including through the gastrointestinal and respiratory tract, skin lesions, placenta and organ transplantations. The symptoms and disease caused by these viruses range from asymptomatic infections or mild cough, skin lesions and nausea to very severe acute presentations such myocarditis, meningitis, encephalitis and acute flaccid paralysis (AFP). There is increasing and convincing recent evidence for associations of specific enterovirus types with chronic diseases, such as asthma, cardiomyopathy and diabetes.
Human pathogenic enteroviruses can be divided into a number of species and serotypes of enteroviruses, rhinoviruses and parechoviruses. Interestingly, enteroviruses (more specifically echoviruses, such as E9 and E30, and parechovirus type 3A) account for over 90% of pediatric meningitis cases where diagnosis has been achieved. AFP, another severely disabling neurological outcome from infection linked to enteroviruses, usually follows from a respiratory tract infection and can lead to permanent disability in those affected. Types associated with AFP include polioviruses, EV-D68 and EV-A71; although traditionally associated with poliovirus, recent analyses find paralytic symptoms to be around 10-times more common following EV-D68 infection (1% of diagnosed cases) than after poliovirus infection (0.1% of those infected). Substantial morbidity is also observed in hand, foot and mouth disease (HFMD), a common childhood disease mostly associated with EV-A71, coxsackieviruses A16 (CVA16) and A6 (CVA6). Although most children present with self-limited fever with skin eruption on hands and feet and vesicles or ulcerations in the mouth, severe and sometimes fatal complications, such as neurological disorders or cardiorespiratory failure, have been reported in EV-A71 associated HFMD cases in Asia and in Europe.
These small RNA viruses rapidly evolve though either mutation or recombination events, influencing their replication capability, transmissibility and potential escape from host immune responses (Pons-Salort & Grassly, 2018). In addition to known and potential virus factors influencing pathogenicity and infection outcomes, several studies have provided evidence for the existence of a range of host factors predictive of more severe systemic infection, particularly those linked to innate immunity. For example, EV-A71 encephalitis with cardio-respiratory compromise has been linked to elevated interleukin 1beta, interleukin 1 receptor antagonist and granulocyte colony stimulating factor levels (Griffiths et al., 2012). Further work has identified potential inborn errors in TLR3- or MDA5-dependent type I IFN immunity in children with severe rhombencephalitis linked to E30 and EV-A71 infections (Chen et al., 2021).
Although traditionally regarded as causing acute resolving infections, recent studies have provided convincing evidence for enterovirus persistence and involvement in longer term disease processes. Coxsackievirus B3 infection, for example, can remain asymptomatic or mild, or the virus can initiate acute infection of cardiac tissue and, in some cases, establish a long-term persistent infection that can lead to serious disease sequelae, including dilated cardiomyopathy. Interestingly, the presence of 5′ terminally deleted forms of enterovirus RNAs has been found in heart tissues derived from patients with acute myocarditis (Chapman et al, 2008) and dilated cardiomyopathy (Bouin et al., 2019. These deleted RNAs are found in association with very low levels of full-length enterovirus genomic RNAs, an interaction that may facilitate continued persistence while limiting virus particle production. Whether these outcomes represent further examples of the impact of host susceptibility factors on virus clearance remain to be explored further.
Our aim is to obtain a better understanding of enterovirus infection: why some individuals encounter a severe form while other remains completely asymptomatic and whether these different outcomes arise from virus pathogenicity determinants or host genetic background, and their further potential role in determining why enterovirus infections in some individual lead to a persistently and/or chronicity but not in all those infected. The tools applied can include comparative analysis of transcriptomics, proteomics and metabolomics of plasma, cerebrospinal fluids or respiratory samples obtained from individuals with varying severity of infection, and analysis of tissue or blood derived viral genomes. The findings will also contribute towards a better diagnosis of enterovirus infections, and potentially open new innovative avenues for prevention and treatment.
These investigations will be supported by a strong European Non-Polio Enterovirus Network (ENPEN) as a source of virus isolates, sequences and hospital-based surveillance data. ENPEN was established in 2017 and now composes scientists from over 70 institutions in Europe. With ENPEN, we are currently collaborating also with the European Centre for Disease Control (ECDC) and WHO European office.
Key words:
Virology, Microbiology, Infectious Diseases, Neurovirulence, Acute versus Chronic Infection, Metagenomics, Transcriptome, Omics, Diagnosis, Better Health, Public Health
Selected publications:
- Harvala H, Broberg E, Benschop K, Berginc N, Ladhani S, Susi P, Christiansen C, McKenna J, Allen D, Makiello P, McAllister G, Carmen M, Zakikhany K, Dyrdak R, Nielsen X, Madsen T, Paul J, Moore C, von Eije K, Piralla A, Carlier M, Vanoverschelde L, Poelman R, Anton A, López-Labrador FX, Pellegrinelli L, Keeren K, Maier M, Cassidy H, Derdas S, Savolainen-Kopra C, Diedrich S, Nordbø S, Buesa J, Bailly JL, Baldanti F, MacAdam A, Mirand A, Dudman S, Schuffenecker I, Kadambari S, Neyts J, Griffiths MJ, Richter J, Margaretto C, Govind S, Morley U, Adams O, Krokstad S, Dean J, Pons-Salort M, Prochazka B, Cabrerizo M, Majumdar M, Nebbia G, Wiewel M, Cottrell S, Coyle P, Martin J, Moore C, Midgley S, Horby P, Wolthers K, Simmonds P, Niesters H, Fischer TK. 2018. Recommendations for enterovirus diagnostics and characterisation within and beyond Europe. Journal of Clinical Virology 101:11-17.
- Bubba L, Broberg EK, Jasir A, Harvala H for Enterovirus study collaborators. 2020. Circulation of non-polio enteroviruses in 24 EU/EEA countries between 2015 and 2017: clinical relevance and considerations for systematic surveillance. Lancet Infectious Diseases 20(3):350-361.
- Harvala H, Robertson I, Chieochansin T, McWilliam Leitch EC, Templeton K, Simmonds P. 2009. Association of human parechovirus type 3 with sepsis and fever in young infants identified by direct typing of cerebrospinal fluid samples. Journal of Infectious Diseases 199:1753-60.
- Kamau E, Nguyen D, Celma C, Blomqvist S, Horby P, Simmonds P, Harvala H. 2021. Seroprevalence and Virologic Surveillance of Enterovirus 71 and Coxsackievirus A6, United Kingdom, 2006-2017. Emerging Infectious Diseases 27(9):2261-2268.
- Pons-Salort M, Lambert B, Kamau E, Pebody R, Harvala H, Simmonds P, Grassly NC. 2023. Changes in transmission of Enterovirus D68 (EV-D68) in England inferred from seroprevalence data. Elife 9;12:e76609.
- Fischer TK, Simmonds P, Harvala H. 2022. The importance of enterovirus surveillance in a post-polio world. Lancet Infect Diseases Jan;22(1):e35-e40.
Heli Harvala
Professor of Virology
Exploring the role of SHANK3 in the vasculature and the development of vascular anomalies
Our earlier discoveries in neurons, epithelial and cancer cells place SHANK3, a scaffolding protein previously linked to autism, at the nexus of complex cell signalling pathways. More recently, our preliminary data, supported by publicly available datasets, demonstrates high SHANK3 expression in endothelial cells (blood and lymphatic vasculature) where its function is unknown. In addition, SHANK3 is highly expressed in brain arteriovenous malformations (bAVMs), vascular anomalies that can trigger haemorrhagic stroke, permanent disability and death in children and young adults.
We aim to investigate the link between SHANK3 and bAVM development by first unravelling the role of SHANK3 within the normal endothelium. We will then apply the unprecedented knowledge gained from this investigation to bAVMs in mouse models and patient samples.
Key words:
SHANK3, Scaffold protein, Actin cytoskeleton, Cytoskeletal reorganisation, Cell adhesion, Integrin signalling, KRAS, Vascular anomaly, bAVM, Stroke, Brain health
Main points of the research topic:
- SHANK3 is highly expressed in normal human vasculature and in bAVM, where its role is unknown
- SHANK3 is at the nexus of several key signalling pathways, including KRAS signalling, Rap1-mediated integrin activation and actin cytoskeletal reorganisation – these pathways regulate cell adhesion, morphology and migration
- In cancer models, targeting SHANK3 is effective against KRAS-driven cancers in in vitro and in vivo models.
- In more than 50% of bAVM cases, a causative mutation in the oncogenic RAS-RAF-MEK-ERK pathway has been observed.
- We will combine this expertise to understand the role of SHANK3 in the endothelium and as a potential target in bAVM.
Methodology:
- endothelial cell culture
- microfluidics
- KRAS signalling readouts
- protein-protein interaction
- proteomic analyses
- Immunofluorescence and immunohistochemistry
- analysis of patient samples
Suitable background experience :
- PhD degree and proven expertise (A publication within the research topic) related to the research theme (e.g. vascular biology, cell signalling, SHANK3 biology, KRAS signalling, animal models of endothelial barriers)
- Excellent communication skills in both spoken and written English.
Selected publications:
- J. Lilja, J. Kaivola, J. R. W. Conway, J. Vuorio, H. Parkkola, P. Roivas, T. Varila, G. Jacquemet, E. Peuhu, E. Wang, U. Pentikäinen, I. M. D. Posada, H. Hamidi, A. K. Najumudeen, O. J. Sansom, I. L. Barsukov, D. Abankwa, I. Vattulainen, M. Salmi, J. Ivaska, Targeting a broad spectrum of KRAS-mutant cancers by hyperactivation-induced cell death. BioRxiv (2022), p. 2022.09.21.508660, doi:10.1101/2022.09.21.508660.
- S. I. Salomaa, M. Miihkinen, E. Kremneva, I. Paatero, J. Lilja, G. Jacquemet, J. Vuorio, L. Antenucci, K. Kogan, F. Hassani Nia, P. Hollos, A. Isomursu, I. Vattulainen, E. T. Coffey, H.-J. Kreienkamp, P. Lappalainen, J. Ivaska, SHANK3 conformation regulates direct actin binding and crosstalk with Rap1 signaling. Curr Biol. 31, 4956-4970.e9 (2021).
- J. Lilja, J. Ivaska, Integrin activity in neuronal connectivity. J Cell Sci. 131, jcs212803 (2018).
- J. Lilja, T. Zacharchenko, M. Georgiadou, G. Jacquemet, N. De Franceschi, E. Peuhu, H. Hamidi, J. Pouwels, V. Martens, F. H. Nia, M. Beifuss, T. Boeckers, H.-J. Kreienkamp, I. L. Barsukov, J. Ivaska, SHANK proteins limit integrin activation by directly interacting with Rap1 and R-Ras. Nat Cell Biol. 19, 292–305 (2017).
Professor Johanna Ivaska
Principal Investigator
Dr Hellyeh Hamidi
Research Support Coordinator
Single-cell analyses of immune responses in cardiovascular disease
T cells and other inflammatory cells are crucial in the growth and instability of atherosclerotic plaques. Recent single-cell analyses have shown that the T cells accumulating in the atherosclerotic aorta are phenotypically distinct from those in circulation, underscoring the significance of regulatory events within the aortic tissue microenvironment.
Our laboratory is dedicated to understanding the interaction between immune and stromal cells in atherosclerosis and autoimmune disorders, utilizing single-cell transcriptomics, immune receptor sequencing, and spatial omics. We are particularly focused on obesity-associated remodeling of perivascular adipose tissue and its impact on immune cell migration and plaque formation. Our research integrates data from both mouse models and patient samples, employing multidisciplinary wet lab and bioinformatics approaches. Our ultimate aim is to identify novel peripheral biomarkers and potential therapeutic targets.
We collaborate closely with the single-cell omics core facility at Turku Bioscience Centre, ensuring access to cutting-edge precision genomics and immunoprofiling techniques. Additionally, we are part of the InFLAMES research flagship, a prestigious immunological R&D cluster. Our team is international and interdisciplinary, with a robust network of collaborators in immunology, genomics, bioinformatics, and cardiovascular health. We are committed to fostering equality, diversity, and a healthy work-life balance.
We are seeking a highly qualified PhD-level scientist with a degree in life sciences, bioinformatics, or a related field. Experience in immunology, cardiovascular diseases, genomics, or proficiency with command-line tools is an asset. Proficiency in English, both oral and written, is required as it is our working language. We also value strong interpersonal skills and the ability to collaborate with individuals from diverse backgrounds.
Key words:
Atherosclerosis, Adipose, Obesity, Immunity, Single-cell, Omics, Transcriptomics, Bioinformatics
Selected Earlier Results:
We have previously used single-cell transcriptomics and epigenomics to characterise the cellular landscape in atherosclerosis in both human (1, 2) and in hyperlipidaemic mouse model (3). These studies have identified disease-associated cell states and explored their relationships with known genetic risk loci. In a complementary line of studies, we have used functional assays and single-cell sequencing to measure the immunoregulatory functions of adipose tissue-derived stem cells in lean and obese individuals, and how these functions are modified by metabolic syndrome (4).
Collaborations and network:
We collaborate locally with the teams of Prof. Pekka Ruusuvuori (imaging-based bioinformatics), Prof. Anne Roivainen (PET), and Prof. Laura Elo (single-cell bioinformatics). We have extensive long-term collaboration with teams of Prof. Minna Kaikkonen-Määttä and Merja Heinäniemi at the University of Eastern Finland, who are both experts in cardiovascular diseases and genomics methods. In addition, we have an ongoing collaboration with group of Prof. Anders Woetmann Andersen at the University of Copenhagen, Denmark. We are also part of the InFLAMES research flagship, funded by the Research Council of Finland, which provides excellent collaborative network including connections to local pharma and biotech companies.
Selected publications:
- Depuydt MAC, Prange KHM, Slenders L, Örd T, Elbersen D, Boltjes A, de Jager SCA, Asselbergs FW, de Borst GJ, Aavik E, Lönnberg T, Lutgens E, Glass CK, Kaikkonen MU, Bot I, Slütter B, van der Laan SW, Yla-Herttuala S, Mokry M, Kuiper J, de Winther MPJ & Pasterkamp G: Microanatomy of the human atherosclerotic plaque by single-cell transcriptomics. Circulation Research 2020 127(11):1437-1455. PMID: 32981416
- Örd T, Õunap K, Stolze LK, Aherrahrou R, Nurminen V, Toropainen A, Selvarajan I, Lönnberg T, Aavik E, Ylä-Herttuala S, Civelek M, Romanoski CE, Kaikkonen MU: Single-Cell Epigenomics and Functional Fine-Mapping of Atherosclerosis GWAS Loci. Circulation Research 2021 129(2):240-258. PMID: 34024118
- Örd T, Lönnberg T, Nurminen V, Ravindran A, Niskanen H, Kiema M, Öunap K, Maria M, Moreau PR, Mishra PP, Palani S, Virta J, Liljenbäck H, Aavik E, Roivainen A, Ylä-Herttuala S, Laakkonen JP, Lehtimäki T & Kaikkonen MU: Dissecting the polygenic basis of atherosclerosis via disease-associated cell state signatures. The American Journal of Human Genetics 2023 110:1-19. PMID: 37060905
- Oliva-Olivera W, Castellano-Castillo D, von Meyenn F, Cardona F, Lönnberg T & Tinahones FJ: Human adipose tissue-derived stem cell paracrine networks vary according metabolic risk and after TNFα-induced death: an analysis at the single-cell level. Metabolism 2021; 116:154466. PMID: 33333081
Tapio Lönnberg
Principal Investigator, Docent
Guardians of Balance: Macrophages Bridging the Neuroendocrine and Immune Systems
The nervous, endocrine, and immune systems are intricately connected, both anatomically and functionally, with their continuous communication playing a crucial role in maintaining our body’s physiological balance. At the heart of this complex network are the hypothalamus—a key brain region—and the pituitary gland, located directly beneath it, which act as central regulatory hubs. These structures integrate signals from the central nervous system with the peripheral endocrine system, coordinating the body’s responses to internal and external changes. Any disruption in the function of the hypothalamus or pituitary, either individually or together, can profoundly impact essential processes like hormonal regulation, stress responses, growth, reproduction, and overall homeostasis. Notably, both organs house macrophages strategically positioned to interact directly with neurons and endocrine cells. We hypothesize that these macrophages play an active role in shaping how neurons and endocrine cells function and adapt, influencing the body’s ability to respond to changing physiological conditions.
Macrophages, traditionally recognized for their role as immune cells, are now understood to have diverse functions that extend far beyond pathogen defense. Tissue-resident macrophages can adapt and actively participate in the development and maintenance of homeostatic processes within the tissues in which they reside by intercellular communication between macrophages and neighboring cells. This adaptability allows macrophages to influence the function of neighboring cells, including those in the hypothalamus and pituitary gland, making them crucial players in neuroendocrine regulation. Our research aims to uncover the precise mechanisms by which macrophages interact with neurons and endocrine cells across different physiological conditions. We are particularly interested in exploring whether these interactions differ between genders, as this could provide insights into gender-specific responses in various disease conditions.
How Do We Do It?
We employ a range of cutting-edge techniques, including the analysis of human tissue samples, single-cell RNA sequencing (scRNA-seq), advanced flow cytometry, and state-of-the-art in vivo animal models. By integrating these approaches, we aim to gain a detailed understanding of the complex roles macrophages play within the hypothalamic-pituitary axis and their broader implications for human health and disease.
Who Are We Looking For?
We seek passionate and innovative researchers with expertise in immunology, neurobiology, or endocrinology to join our dynamic team. If you are driven by curiosity and a desire to contribute to groundbreaking research that bridges the immune and neuroendocrine systems, we want to hear from you!
Selected publications:
- Henna Lehtonen, Heli Jokela, Julian Hofmann, Arfa Mehmood, Lauriina Tola, Florent Ginhoux, Burkhard Becher, Melanie Greter, Marko Salmi, Heide Gerke, Pia Rantakari. Early precursor-derived pituitary gland tissue-resident macrophages play a pivotal role in modulating hormonal balance. bioRxiv 2024. Preprint doi: 10.1101/2024.05.07.592305 and in re-submission
- Dolfi B, Gallerand A, Firulyova M, Xu Y, Merlin J, Dumont A, Castiglione A, Vaillant N, Quemener S, Gerke H, Stunault M, Schrank P, Kim S, Zhu A, Ding J, Gilleron J, Magnone V, Barbry P, Dombrowicz D, Duranton C, Wakkach A, Blin-Wakkach C, Becher B, Pagnotta S, Argüello R, Rantakari P, Chakarov S, Ginhoux F, Zaitsev K, Kim K, Yvan-Charvet L, Guinamard R, Williams J, Ivanov S. Unravelling the sex-specific diversity and functions of adrenal gland macrophages. Cell Rep. 2022. PMID: 35705045 doi: 10.1038/s41467-020-18206-0
- Lokka E, Lintukorpi L, Cisneros-Montalvo S, Mäkelä J-A, Tyystjärvi S, Ojasalo V, Gerke H, Toppari J, Rantakari P#, Salmi M# Generation, localization and functions of macrophages during the maturation of testis. Nature Communications 2020. PMID: 32873797 doi: 10.1038/s41467-020-18206-0 #Denotes equal contribution
- Jokela H, Lokka E, Kiviranta M, Tyystjärvi S, Gerke H, Elima K, Salmi M#, Rantakari P#. Fetal –derived macrophages persist and sequentially maturate in ovaries after birth. Eur J Immunol. 2020. PMID: 32459864 doi: 10.1002/eji.202048531 #Denotes equal contribution
Pia Rantakari
Group leader
InFLAMES Research Flagship Center
Turku Bioscience Centre
Infection and Immunity Research Unit Institute of Biomedicine
University of Turku, and Åbo Academi University
Systems-biology based target discovery for new disease-modifying heart failure treatments
We are looking for a motivated postdoctoral fellow to help us discover new ways to treat heart diseases. Our research aims to elucidate molecular mechanisms mediating cardiac regeneration and remodelling, to identify and validate new therapeutic targets, and eventually to develop new pharmacological treatments that stop or reverse the progression of heart failure.
The most common causes of heart failure are myocardial infarction (MI) caused by coronary artery disease and hypertension. In adult mammals, cell loss during and after an MI results in formation of a fibrotic scar and remodelling of the surrounding cardiac muscle, and often leads to heart failure. Remarkably, some lower vertebrates and neonatal mammals can fully regenerate their hearts after an injury. This capacity is however lost soon after birth due to cardiomyocyte cell cycle exit. The exact molecular mechanisms regulating cardiac regeneration and the postnatal loss or regenerative capacity are however not known, which hinders the development of regenerative therapies. Furthermore, hypertension and MI increase cardiac workload and thereby cause structural and functional changes in the myocardium, including cardiomyocyte hypertrophy and fibrosis, which eventually lead to contractile dysfunction and heart failure. Current pharmacological treatments cannot effectively stop or reverse the progression of pathological remodelling and heart failure, and thus there is an urgent need for the development of therapies that would promote cardiac regeneration or stop and reverse cardiac remodelling.
Key words:
Cardiac remodelling, cardiac regeneration, hypertrophy, fibrosis, human pluripotent stem cell, cardiomyocyte, drug target, cell biology, systems biology, high content analysis
Methods:
Our research is mainly based on hPSC-based cardiac in vitro models, including co-cultures and 3D microtissues consisting of two to three cardiac cell types: cardiomyocytes, endothelial cells, and cardiac fibroblasts. In addition to standard lab techniques, we utilise omics technologies (transcriptomics, proteomics, metabolomics) and phenotypic screening techniques such as high content imaging and cell painting.
Requirements:
Successful candidates should have a PhD degree and proven expertise related to the research theme (e.g. hPSC-based cardiac in vitro models, bioinformatics, cell signalling, molecular biology), excellent communication skills in both spoken and written English, and ability to work as a team in a dynamic multicultural research environment.
Selected publications:
- Majid QA, Ghimire BR, Merkely B, Randi AM, Harding SE, Talman V*, Földes G*: Generation and characterisation of scalable and stable human pluripotent stem-cell derived microvascular-like endothelial cells for cardiac applications. Angiogenesis 2024; 27: 561-582. https://doi.org/10.1007/s10456-024-09929-5 *Co-senior authors
- Pohjolainen L, Kinnunen SM, Auno S, Kiriazis A, Pohjavaara S, Kari-Koskinen J, Zore M, Jumppanen M, Yli-Kauhaluoma J, Talman V, Ruskoaho H, Välimäki M: Switching hypertrophic signalling towards enhanced cardiomyocyte maturity by GATA4-targeted compound. Stem Cell Res Ther 2024; 15:5. https://doi.org/10.1186/s13287-023-03623-x
- Chaudhari U, Pohjolainen L, Ruskoaho H, Talman V: Genome-wide profiling of miRNA-gene regulatory networks in mouse postnatal heart development - implications for cardiac regeneration. Front Cardiovasc Med 2023; 10:1148618. https://doi.org/10.3389/fcvm.2023.1148618
- Pohjolainen L, Ruskoaho H, Talman V: Transcriptomics reveal stretched human pluripotent stem cell-derived cardiomyocytes as an advantageous hypertrophy model. J Mol Cell Cardiol Plus 2022, 2:100020. https://doi.org/10.1016/j.jmccpl.2022.100020
- Karhu ST, Ruskoaho H, Talman V: Distinct regulation of cardiac fibroblast proliferation and transdifferentiation by classical and novel protein kinase C isoforms: possible implications for new antifibrotic therapies. Mol Pharmacol 2021; 99:104-113. https://doi.org/10.1124/molpharm.120.000094
- Talman V, Teppo J, Pöhö P, Movahedi P, Vaikkinen A, Karhu ST, Trošt K, Suvitaival T, Heikkonen J, Pahikkala T, Kotiaho T, Kostiainen R, Varjosalo M, Ruskoaho H: Molecular atlas of postnatal heart development. J Am Heart Assoc 2018; 7:e010378. https://doi.org/10.1161/JAHA.118.010378