Category Archives: Cell Biology

Synthetic Biology Startup Acquires AI Platform To Disrupt The Drug Industry – Forbes

Sean McClain, Co-Founder and CEO of AbSci.

There has been a lot of recent attention on the challenges of delivering COVID-19 vaccines. But there are also challenges in making them. For some of the newer options like those from Johnson & Johnson and Oxford-AstraZeneca, the modified cells used in vaccine production are struggling under the scale of demand. But synthetic biology company AbScis recent acquisition of the artificial intelligence platform, Denovium, could help mitigate this type of challenge in the future.

Unlike mRNA vaccines, the Johnson & Johnson/Oxford-AstraZeneca class of vaccines rely on a type of virus called adenovirus which is known to cause colds in chimpanzees. To address COVID-19, the adenovirus is genetically altered to express the SARS-CoV-2 spike protein which is what ultimately triggers the bodys immune response. Like mRNA vaccines, adenovirus-based vaccines train the body to recognize and fight COVID-19, foregoing the need to inject a person with a weakened version of SARS-CoV-2.

But producing enough adenovirus cells has been a challenge. To make vaccine doses, large volumes of altered adenovirus are produced by replicating cells in bioreactors. But, the scale of production can also cause the cells to weaken. This can result in a reduced output of adenovirus copies. So while these new vaccines may represent a breakthrough in adenovirus-based therapeutics, the process also highlights some critical roadblocks.

One major issue is that drug discovery and drug manufacturing are often disconnected from one another. Drug discovery typically starts with screeningthe process of finding a set of compounds out of 100,000 combinations that can best neutralize a targeted weak point of a disease. But when a promising protein is identified, it often turns out to be difficult to scale effectively.

Once a therapeutic compound is identified, researchers must then determine if it works well with a group of similar cells called a cell-line. By inserting the compound into the cellswhich then divide and multiply in a bioreactorthe cells act like factories to produce greater volumes of the compound of choice. But, as in the case with adenovirus-producing cells, not all cells can maintain their functions at large volumes. If the protein compound doesnt work well in a scalable cell-line, researchers often have to go back to the drawing board to find a new compound and start again.

Many in the biopharma space are aware of this inefficient process. The synthetic biology company AbSci has spent years developing a platform solution that streamlines the workflow. [Our platform] is simultaneously a drug discovery and manufacturing platform that allows you to discover your drug and the cell line that can manufacture [it], says AbSci CEO, Sean McClain. Were finally uniting drug discovery and manufacturing the first time.

AbSci refers to their core process as their Protein Printing platform, not because it uses ink and paper to make proteins but as an analogy for ease and speed. The first technology [in our platform] is our SoluPro E. coli strain. It has been highly engineered to be more mammalian-like to be able to produce mammalian-like proteins that E. coli wasn't previously capable of doing, says McClain. AbSci also uses what the company calls a folding solution to precisely tailor how proteins fold and therefore function.

Imad Ajjawi, Co-Founder and CBO of Denovium

To find the most effective protein, AbSci alters its folding solutions to create as many protein varieties as possible, often to the order of 10s of millions. The more protein types available, which AbSci refers to as libraries, the higher the likelihood of success. But this also creates a challenge: so many options, but which to choose?

To address this, AbSci recently acquired artificial intelligence company, Denovium. By integrating Denoviums AI platform, AbSci can improve its data analysis via AI models. From there, the company can take the best candidates and find the most effective cell-line to produce the chosen compounds at scale. McClain explains that traditional drug discovery and manufacturing typically takes years. But AbScis platform can take that timeline down to weeks. Were actually able to manufacture [therapeutics] because the dirty secret in pharma is that so many drugs get shelved because [pharma companies] can't actually manufacture them, says McClain.

For McClain, acquiring Denovium is a big step forward for AbScis discovery process. Its going to change the paradigm. Its really a perfect marriage of both data and AI technology. If you don't have good data feeding into your AI model, it's worthless. But if you don't have an AI technology, you can't mine [the data] and get all the benefits, says McClain.

Denoviums co-founder and CBO, Imad Ajjawi, also sees the new collaboration as a significant opportunity. It's really exciting to be a part of AbSci because they have all the data, billions of points that the deep learning engine can now analyze, says Ajjawi. AbScis acquisition also comes on the heels of the companys $65 million Series E in late 2020.

Upgrading the union of biology and AI is important for advancing synthetic biology innovation. But the true potential beneficiaries of this advanced discovery platform are those in need of novel drug options.

AbScis main goal as a company is to bring therapeutics to market more quickly. This technology's impact on healthcare is profound because more drugs and biologics can now enter patients' hands faster, says McClain.

McClain believes that AbScis technology will help speed the process of clinically testing new medications. Faster clinical trial turnarounds could increase the number of drugs approved to address a range of diseases. This could be most impactful for patients with rare or difficult to treat conditions as drug discovery is often prioritized based on how long it takes to find a scalable cell-line.

But though AbSci is working to accelerate drug discovery, the process still takes time. Right now, we have six drugs that are in preclinical or clinical trials. And one of them is actually in phase three. So we could have an improved product here in the next couple of years, says McClain.

As Absci and Denovium finalize their technology integrations, McClain is also looking ahead to build as many partnerships as possible. The more partnerships we do, the more patients were able to affect that at the end of the day, says McClain.

In line with that goal, AbSci today announced a continuation of its partnership with Astellas and Xyphos. AbSci will take on screening and identifying an optimal cell-line for a leading variant of Xyphos MicAbody, a bispecific antibody-like adaptor molecule used in the company's immuno-oncology program.

McClain expects more partnership announcements will follow in the first quarter of 2021. We have some really exciting partnerships that are going to be coming out over this next quarter that I think speak to the [range] of the types of disease states we're working on and the breadth of how the technology can be used within biopharma, says McClain.

Im the founder of SynBioBeta, and some of the companies that I write about are sponsors of the SynBioBeta conference and weekly digest, including AbSci. Thank you to Fiona Mischel and Vinit Parekh for additional research and reporting in this article.

The rest is here:
Synthetic Biology Startup Acquires AI Platform To Disrupt The Drug Industry - Forbes

Severe COVID-19 Outcomes May be Improved With Bevacizumab – Contagionlive.com

Findings included a reduction in fever, an increase in white blood cells and a sharp decrease of c-reactive protein (CRP) levels.

A recent study taking place in Italy and China, led my investigators from the Karolinska Institutet, has found that the anti-cancer therapy bevacizumab may hasten recovery and reduce the rate of mortality in people with the coronavirus disease 2019 (COVID-19). The research was published in the journal Nature Communications.

Bevacizumab has been used to treat different types of cancer since 2004, and works by reducing the formation of blood vessels by inhibiting the growth factor signaling protein vascular endothelial growth factor (VEGF).

It has been seen that patients with a severe infection of COVID-19 have related symptoms to VEGF like excess fluid and disorganized blood vessels in the lungs, as well as elevated levels of the growth factor.

The investigators behind the study recruited 26 patients from two hospitals in Italy and China who had a confirmed case of COVID-19 and displayed symptoms of difficulty breathing, pneumonia and low blood oxygen levels. These patients were then retrospectively compared to 26 other patients with similar symptoms who received the current standard of care.

The recruited participants received standard, care as well as a single, low dose of 7.5 mg/kg bevacizumab. Findings showed that within 24 hours of receiving their therapy, the patients had a significant improvement in their blood oxygen levels compared to the control group. After a 28-day follow-up, 92% of the treated arm no longer needed an equal amount of oxygen support as when they began the trial.

Additionally, none of the participants receiving the therapy died, and 65% improved to such a degree that they were able to be discharged from the hospital in comparison to only 46% of the control arm being discharged. The duration of oxygen-support was also shortened to a median of 9 days for the bevacizumab arm.

"To reduce COVID-19 mortality, we aim to develop an effective therapeutic paradigm for treating patients with severe COVID-19," Yihai Cao, a professor of vascular biology at the Department of Microbiology, Tumor and Cell Biology at Karolinska Institutet and a co-author on the study said. "Our findings suggest that bevacizumab plus standard care is highly beneficial for patients with severe COVID-19 and should be considered as a potential first-line therapeutic regimen for this group."

Link:
Severe COVID-19 Outcomes May be Improved With Bevacizumab - Contagionlive.com

Researchers unlock mysteries of protein that plays key regulatory role in cancer therapies – News-Medical.Net

Researchers from the University of Virginia, Lehigh University, and the Massachusetts Institute of Technology are pooling their respective labs' expertise to unlock the mysteries of a protein that plays a critical regulatory role in human health and disease. Knowing how the protein works could lead to improved therapies for cancers and other diseases.

UVA associate professor of chemical engineering Matthew Lazzara and Lehigh University associate professor of chemistry Damien Thvenin is the project principal investigators. Forest White, a professor of biological engineering at MIT, is also a collaborating investigator.

The project, "Promoting Receptor Protein Tyrosine Phosphatase Activity by Targeting Transmembrane Domain Interactions," is funded by a $1.6 million Project Research Grant (R01) from the National Institute of General Medical Sciences of the National Institutes of Health.

The protein at the center of the project is known as protein tyrosine phosphatase receptor type J (PTPRJ), also sometimes referred to as density-enhanced phosphatase-1 (DEP-1). PTPRJ is a member of the family of receptor-like protein tyrosine phosphatases (RPTP), which target and dephosphorylate, or deactivate, proteins involved in cell proliferation and survival.

The team anticipates that their work on the PTPRJ protein could yield insights that are relevant across the receptor-like protein tyrosine phosphatase family.

"The importance of RPTPs in normal cell function is clear, but we don't yet know much about the structure-function relationships that underpin the regulation of their activity," Lazzara said. "If we knew more, we might be able to design ways to augment their activity in settings, such as cancer, where RPTP substrates need to be turned off."

One goal of the project is to understand how to promote the activity of PTPRJ -- and eventually other RPTPs -- by interfering with the ability of the phosphatase to bind to itself, a process called homodimerization in which two identical proteins form a structure.

"Our collaborators at Lehigh have designed small peptide binders that disrupt PTPRJ homodimerization as a way to promote phosphatase activity," Lazzara said. "Because the phosphatase acts on, and effectively turns off, certain receptors that can promote tumor growth, we think this could eventually lead to a new method to interfere with signaling in cancer cells in a way that would not be circumvented by the common forms of drug resistance we see over and over again in oncology."

"Our approach has all kinds of exciting consequences on cell behavior and therapeutic applications," Thvenin said.

"Indeed, one of the main substrates of RPTPs are receptor tyrosine kinases, which are over-activated, or phosphorylated, in many cancers," Thvenin said. "Existing methods to target tumor-promoting kinases are limited to pharmacological inhibitors and antibodies. While some drug treatments can be highly effective, at least initially, resistance to these inhibitors virtually always arises through mutations or bypass signaling via alternative receptor tyrosine kinases. Promoting the activity of RPTPs could be an effective alternative approach to overcoming common acquired resistance mechanisms, as it should be immune to the effects of gatekeeper mutations."

A second project goal is to identify the circumstances under which interfering with PTPRJ dimerization might be most effective for changing how cells function.

"In cell biology, everything is about context," said Lazzara, who holds a courtesy appointment in biomedical engineering and is a member of the UVA Cancer Center. "The function of a protein in one cellular setting may be different than in another. That can happen for lots of reasons, including differences in the expression of interacting proteins. My lab's main role in the project is to execute a set of experiments designed to capture that complexity and then to use systems biology computational modeling approaches to interpret the data."

White, a former postdoctoral researcher at UVA, will contribute by using mass spectrometry to quantify protein phosphorylation events that change in response to modulating PTPRJ function in the lab. The use of mass spectrometry to quantify signaling protein phosphorylation is an area of expertise for which White is well known, Lazzara said.

Forest's approach can measure hundreds to thousands of unique phosphorylation events at a time in cells, which is substantially greater bandwidth than you can do with many other techniques. There are some other techniques that can measure hundreds of sites, but they are much less quantitative than his approach. Forest has used this method to study many different signaling processes in cancer."

Matthew Lazzara, Associate Professor of Chemical Engineering, University of Virginia

Lazzara noted for prolific and often collaborative research in cell signaling and cellular decision-making has received numerous grants from the National Science Foundation, National Cancer Institute, National Institute of General Medical Sciences, and the American Cancer Society.

His work contributes significantly to UVA chemical engineering's research programs, also providing graduate researchers with opportunities to engage in fundamental and potentially groundbreaking science.

"This project is a great example of how biological researchers are increasingly working collaboratively and integrating multiple areas of expertise to make advances," Lazzara said. "I expect we will continue to see that in cancer research especially."

The rest is here:
Researchers unlock mysteries of protein that plays key regulatory role in cancer therapies - News-Medical.Net

Leveraging systems biology for predicting modulators of inflammation in patients with COVID-19 – Science Advances

Abstract

Dysregulations in the inflammatory response of the body to pathogens could progress toward a hyperinflammatory condition amplified by positive feedback loops and associated with increased severity and mortality. Hence, there is a need for identifying therapeutic targets to modulate this pathological immune response. Here, we propose a single cellbased computational methodology for predicting proteins to modulate the dysregulated inflammatory response based on the reconstruction and analysis of functional cell-cell communication networks of physiological and pathological conditions. We validated the proposed method in 12 human disease datasets and performed an in-depth study of patients with mild and severe symptomatology of the coronavirus disease 2019 for predicting novel therapeutic targets. As a result, we identified the extracellular matrix protein versican and Toll-like receptor 2 as potential targets for modulating the inflammatory response. In summary, the proposed method can be of great utility in systematically identifying therapeutic targets for modulating pathological immune responses.

Inflammation is a key defense mechanism to pathogenic factors, such as infection, chemical substances, or tissue injury, which is mediated by tissue resident and circulating cells recruited from the blood through the establishment of chemokine gradients. To modulate the level of inflammation, these cells release cytokines to communicate with each other and activate cell typespecific functions necessary to clear the pathogenic factor. For instance, phagocytic activity removes cellular debris and pathogens, thereby suppressing inflammation through the release of anti-inflammatory cytokines, such as interleukin-10 (IL-10) and transforming growth factor (1). Typically, the immune response is tightly controlled to minimize tissue injury and restore homeostasis. However, in case the pathogenic factor cannot be cleared, an acute inflammatory response progresses toward a hyperinflammatory condition, commonly referred to as cytokine storm, due to an excessive release of cytokines and an accumulation of immune cells in the tissue.

Recently, a novel, highly contagious coronavirus [severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)] has been identified as the causative pathogen of an ongoing outbreak of viral pneumonia [coronavirus disease 2019 (COVID-19)]. Accumulating evidence suggests that patients with severe symptomatology of COVID-19 develop such a hyperinflammatory immune response, similar to other respiratory infectious diseases such as influenza infection (2). Although initial studies have begun to arise in which the immune status of patients is assessed (3, 4), the molecular mechanisms underlying the hyperinflammatory response of patients with more severe symptoms are not readily known. Hence, the present challenge is how to mitigate this cytokine storm while not impairing viral clearance.

Cytokine storms are most often characterized by high levels of tumor necrosis factor (TNF), interferons, and IL-6, as well as the accumulation of immune cells in the tissue (57). However, the composition of proinflammatory ligands involved in cytokine storms depends on the viral pathogen. For instance, in patients with SARS, hyperinflammation is characterized by high levels of interferon- (IFNG), IL-18, and lymphotoxin- (LT-), whereas patients with severe influenza infections present high levels of TNF, IL-6, and IL-1 (6, 7). Moreover, cytokine storms are mediated by interactions between immune and nonimmune cells, which form positive feedback loops responsible for amplifying and maintaining the immune response. These loops are composed of cell-cell interactions for which incoming signals induce outgoing signals in each participating cell population. For example, in case of influenza infection, a positive feedback loop involves the production of IL-1 and TNF by macrophages in response to granulocyte-macrophage colony-stimulating factor released by nonhematopoietic cells (8). In the case of Epstein-Barr virus infection, the hyperinflammatory response is amplified by multiple, interconnected positive feedback loops between dendritic cells, CD8+ T cells, natural killer (NK) cells, and macrophages that involve several proinflammatory cytokines, including IL-1, IL-6 and IL-18, IFNG, and TNF (9). However, in general, positive feedback loops responsible for the amplification of proinflammatory signals are largely unknown. Furthermore, they differ in the participating cell populations, as well as in the involved inflammatory molecules depending on the tissue type and the viral infection. Consequently, it is critical to characterize the positive feedback loops amplifying and maintaining the hyperinflammatory immune response to develop therapeutic strategies for selectively disrupting cell-cell interactions underlying these conditions.

The development of strategies for modulating the immune response typically relies on the identification of biomarkers as therapeutic targets or the large-scale in vitro screening of compounds. While these approaches have led to the identification of immunomodulatory compounds for a diverse array of underlying diseases (10), they are laborious and resource intensive or, as in the case of biomarkers, mostly not efficacious, which impedes the design of novel therapeutic strategies for modulating inflammation. The upsurge of single-cell sequencing technologies has enabled the analysis of multiple cell populations in a tissue at an unprecedented resolution and permits the development of computational methods that could identify immunomodulatory therapeutic targets and compounds inhibiting them. To date, great efforts have been devoted to the discovery of biomarkers and drug-target interactions (1113). However, to our knowledge, no computational method for predicting immunomodulatory target proteins that could alleviate pathologic dysregulations of the physiological immune response exists.

To address this issue, we present a single cellbased computational method for the systematic prediction of protein targets to modulate the inflammatory response. In particular, given two tissue-specific single-cell profiles of a dysregulated immune response and a physiological control, our method infers the functional cell-cell communication networks of both conditions by integrating a collection of 1756 extracellular receptor-ligand interactions from more than 6000 protein-protein interactions (PPIs) with intracellular signaling and gene regulatory networks. Once the functional cell-cell communication network has been inferred, to construct positive feedback loops, the method searches for those interactions causing the expression of ligands secreted by this cell population. As a plausible therapeutic strategy to modulate the pathological immune response, we propose to target these positive feedback loops. In this regard, our method simulates the effect of perturbing receptor-ligand interactions, prioritizes genes specifically disrupting pathological while preserving physiological feedback loops, and links them to compounds targeting them. To validate our method for predicting immunomodulatory target proteins and to demonstrate its general applicability, we applied it to 12 disease pathologies characterized by hyperinflammatory or chronic inflammation and were able to validate 90% of the predicted target proteins. Similar to a hyperinflammatory immune response, the released cytokines under chronic inflammatory conditions are highly promiscuous, depend on the causative pathogenic factor and the affected tissue, and form positive feedback loops to amplify and maintain an elevated immune response (57, 14).

Last, we applied our method to a recently published dataset of bronchoalveolar lavage fluid from patients with mild and severe symptomatology of COVID-19 (15). Our method revealed that the hyperinflammatory response in patients with severe symptomatology is maintained by interconnected feedback loops involving multiple proinflammatory cytokines and extracellular matrix proteins as well as the reprogramming of the anti-inflammatory immune response by IL-10 into a proinflammatory phenotype. In addition, we demonstrate that T cell recruitment is impaired because of the disruption of a feedback mechanism between T cells, secretory epithelial cells, and macrophages, which explains the defective viral clearance in patients with severe symptomatology. Last, computational perturbation of genes involved in causal feedback loops identified versican (VCAN), an extracellular matrix glycoprotein, and Toll-like receptor 2 (TLR2) as novel target proteins for alleviating the hyperinflammatory response in patients with COVID-19 with severe symptomatology. Thus, in summary, we believe that this method can be of great utility in the systematic identification of therapeutic targets for modulating pathological immune responses.

Because of the well-established effect of cellular feedback in the control of the inflammation, we hypothesized that the positive feedback loops established between immune and nonimmune cells are responsible for amplifying and maintaining an elevated immune response. To detect these feedback loops, we first set out to reconstruct the ligand-receptormediated cell-cell communication network within a tissue (Fig. 1A). In this regard, we manually curated more than 6000 PPIs between receptors and ligands and identified 1756 experimentally validated, extracellular interactions (table S1). Next, we integrated this set of high-confidence interactions with intracellular signaling and gene regulatory networks to infer the cell-cell communication network and positive feedback loops (Fig. 1A). In particular, we first selected transcription factors (TFs) whose expression is preserved across cells in each population and, second, identified the active receptors regulating them using a Markov chain model that assess signal transduction probabilities. Last, cognate ligands were identified for each active receptor, which are expressed in more than 5% of the secreting cell population. Only statistically significant ligand-receptor interactions remained in the final cell-cell communication network of each condition, assessed by the strength of each interaction compared to all potential interactions. Once the functional cell-cell communication network has been inferred, the method searches for those interactions causing the expression of ligands secreted by the receiving population by warranting the existence of a sustained regulatory path from the receptor to the ligand (see Materials and Methods for details). Positive feedback loops are lastly established by combining extracellular and intracellular ligand-receptor interactions. A detailed description of the methodology can be found in Materials and Methods.

(A) The method workflow consists of the following steps: (1) single-cell RNA sequencing (RNA-seq) data with cell type (CT) annotations as an input; (2) each population is examined for the persistent signaling flow from receptors via signaling molecules (pathways) to the phenotype-determining TFs; (3) the phenotype-maintaining receptors form the final interactome along with their cognate ligands from other populations; (4) the interactome is further examined for presence of the positive feedback loops, extracellular (from ligands to receptors) and intracellular (from receptors via signaling pathways to ligands) paths that maintain each other by forming closed loops; (5) disease-specific ligands and receptors that are responsible for the formation of the feedback loops and are ranked on the basis of their ability to disrupt the positive feedback loops unique to pathological immune responses; (6) high-scoring genes are mapped to DrugBank for identifying inhibitory compounds. (B) Percentage of predicted target proteins with literature evidence per example. Except in peripheral age-related macular degeneration (15%) and Crohns disease (60%), all predicted target genes are validated. AERD, aspirin-exacerbated respiratory disease.

To predict immunomodulatory target proteins, we propose to score genes by their ability to disrupt positive feedback loops when perturbed. Namely, given the positive feedback loops underlying pathological and physiological immune responses, each gene is synthetically removed from the feedback loops unique to the pathological condition. The fraction of ligands and receptors that are removed by this synthetic perturbation serves as a score for the gene. We score only those feedback loops and genes that differ between the pathological and corresponding physiological conditions. Last, the method extracts information from DrugBank (16) to select drug candidates targeting the highly scored genes (see Materials and Methods for details).

We set out to validate our approach for identifying immunomodulatory proteins and to demonstrate its general applicability by applying it to a vast array of diseases that are characterized by a pathological immune response. In particular, the considered pathologies include autoimmune diseases, such as lupus nephritis, chronic diseases, such as Crohns disease and type II diabetes, as well as allergic conditions, such as aspirin-exacerbated respiratory disease (table S2). As a result, we identified a median of two top-ranking target proteins having the highest scores, with two notable exceptions. In particular, in case of liver cirrhosis and age-related macular degeneration samples from the peripheral eye region, our method identified 15 and 20 target proteins, respectively. Validation with previous literature revealed evidence for, on average, 90% of the predicted immunomodulatory proteins (Fig. 1B and table S3). Nevertheless, only 60 and 15% of proteins predicted for Crohns disease and peripheral age-related macular degeneration samples could be validated, respectively.

We sought to perform an in-depth case study and analyzed recently published single-cell RNA sequencing (RNA-seq) data of nine Chinese patients with COVID-19 with mild and severe symptomatology and three healthy individuals (15). For each group, we aggregated data of different patients presenting with mild and severe symptoms and healthy individuals into a single representative sample of each condition and clustered cells to identify cell types using known sets of markers (15). We identified 15 common cell types for both groups: B cells, CCR7+ T cells, CD8+ T cells, proliferating T cells, innate T cells, regulatory T cells, four subpopulations of macrophages (Mac1 to Mac4), ciliated cells, secretory cells, plasma cells, myeloid dendritic cells (mDCs), and NK cells. In addition, neutrophils and mast cells were uniquely identified in the severe cases. (Fig. 2A and fig. S1A).

(A) UMAP (uniform manifold approximation and projection) representation of integrated samples from patients with mild (left) and severe (right) symptoms. Treg, regulatory T cells. (B) Expression of selected pro- and anti-inflammatory cytokine and chemokine receptors per identified cell type in patients with mild (top) and severe (bottom) symptoms. (C) Expression of selected pro- and anti-inflammatory cytokines and chemokines per identified cell type in patients with mild (top) and severe (bottom) symptoms.

Before reconstructing the cell-cell communication networks underlying both conditions, we performed differential expression analysis of each cell type under both conditions. As a result, we observed that the mild and severe groups significantly differ in the levels of expression of inflammatory molecules (Fig. 2, B and C). For instance, chemokines, including CCL2 (CC chemokine ligand 2), CCL3, CCL4, and CCL8, are significantly up-regulated in nearly all cell types of patients with severe symptoms. In contrast, chemokines, such as CXCL2 (C-X-C motif chemokine ligand 2), CXCL3, and CXCL9, together with their receptor CXCR4 (C-X-C motif chemokine receptor 4), are up-regulated in specific cell types such as in macrophages. In addition, certain pro- and anti-inflammatory cytokines and their receptors have elevated levels under the severe condition, such as IL-6 and its receptor in macrophages, IL-18 in ciliated cells, IL-4R (IL-4 receptor) and IL-7R. On the contrary, among the up-regulated cytokines under the mild condition are IL-18 in macrophages, CCL20 in innate and regulatory T cells, CXCR6 in NK cells, and VASP (vasodilator-stimulated phosphoprotein) protein in certain macrophage populations and T cells. Known anti-inflammatory cytokines do not show consistent differential expression. Thus, for example, the expression of IL-10 is marginally elevated in Mac3 and regulatory T cell populations of the severe cases, whereas the expression level of IL-1R2 (IL-1 decoy receptor) and IL-1 receptor antagonist (IL-1RN) is elevated in all macrophage populations and mDCs. In contrast to patients with COVID-19, samples obtained from healthy individuals do largely not display expression of cytokines and chemokines (fig. S1, B and C). Notably, CXCL16, the cognate ligand of CXCR6, is expressed in most cells and cell types, which suggests that with increasing severity, CXCL16 expression is substantially decreased (fig. S1D).

To elucidate the intercellular interactions maintaining the differences in the expression of ligands and receptors, we use our method to reconstruct the cell-cell communication networks underlying mild and severe disease courses and observed vast differences, which is signified by a 33% increase in interactions in severe cases (Fig. 3A). Despite the observed increase in cell-cell interactions, the number of unique molecules per condition that are involved in extracellular signaling remains low. The severe condition is characterized by interactions involving the proinflammatory cytokines IL-1B, IL-1A, TNF superfamily member 4 (TNFSF4), and thymic stromal lymphopoietin. In contrast, the mild condition is distinguished by ligands promoting lymphocyte migration, such as CXCL13, CCL24, CCL7 and CCL20 as well as cell-adhesion mediators including tenascin C (TNC). The unique interactions involving these ligands cannot be fully explained by differential expression analysis. For instance, IL-21 nor its receptor IL-21R is differentially expressed in any cell type. However, a significant difference in the signal transduction probabilities of IL-21R can be observed in proliferating T cells, which underscores the advantages of our approach (Fig. 3, B and C).

(A) Number of interactions detected in each cell-cell communication network. (B) Differential signaling activity of ligands and receptors involved in immune response based on SigHotSpotter. (C) Differential expression of ligands and receptors involved in immune response. Dots represent significant differences [adjusted P < 0.05, obtained by MAST analysis, Seurat implementation (34, 35)]. (D and E) Expression of receptors characteristic for neutrophils (D) and mast cells (E) under both conditions. JAK1, Janus kinase 1; TYK2, tyrosine kinase 2.

Next, we investigated whether unique interactions can be explained by differences in the composition of cell populations between the two conditions. While the relative proportion of most cell populations in the cell-cell communication networks differs by at most 1%, neutrophils and mast cell interactions are specific to severe disease courses. Consistent with their role in the induction of inflammation (17, 18), these cells release proinflammatory cytokines, such as TNF, IL-1B, IL-18, and oncostatin M, responsible for the positive regulation of nuclear factor B of activated B cells. Further, neutrophils and mast cells express receptors of proinflammatory cytokines including TNF receptor 1, interferon- receptor 2, IL-18R, and the mast cell growth factor receptor KIT, which suggest their involvement in proinflammatory feedback mechanisms (Fig. 3, D and E).

Although a plethora of clinical trials has been performed or is currently ongoing that probed the efficacy of drugs in the treatment of patients presenting with a hyperinflammatory immune response, no effective treatment has been found. Therefore, we sought to apply our methodology to identify proteins that could be targeted for modulating the immune response in patients with severe symptomatology of the COVID-19. In this regard, we first identified the positive feedback loops underlying mild and severe cases, respectively, and detected three groups (fig. S2). The first group signifies the immune response common to patients with severe and mild symptomatology and is characterized by feedback between pro- and anti-inflammatory cytokines predominantly released by macrophages and dendritic cells (table S4). In particular, proinflammatory cytokine signaling by TNF and IFNG is fine-tuned by anti-inflammatory ligands, such as IL-10 and the TNF receptor antagonist progranulin (GRN). In contrast, the second and third groups contain interactions unique to patients with mild and severe pathologies, respectively. In contrast to patients with COVID-19, healthy individuals display only a single positive feedback loop between fibronectin (FN1) and plasminogen activator, urokinase receptor (PLAUR) not involving any cytokines or chemokines, as expected from the transcriptional analysis. To gain insights into the individual stabilizing mechanisms of the immune response in patients with mild and severe symptomatology, we set out to characterize the feedback mechanisms underlying both groups.

Analysis of the cell-cell communication landscape of mild cases reveals two distinct clusters of positive feedback loops unique to this condition. The first cluster consists of B cells secreting macrophage inhibitory factor (MIF), which is sensed in an autocrine and paracrine manner by human leukocyte antigen class II histocompatibility antigen gamma chain (CD74). This interaction induces B cell survival and proliferation through activation of the PI 3-kinase/Act pathway, thus, rescuing them from apoptosis (19). The absence of autocrine and paracrine MIF stimulation of B cells in patients with severe symptomatology suggests an impaired B cell response to SARS-CoV-2 due to increased apoptosis.

In contrast to the first cluster, the second group of interconnected feedback loops involves a chemotactic interaction between CXCL16 and CXCR6. In particular, CXCL16 and CXCR6 belong to a causal feedback loop between T cells, secretory epithelial cells, and macrophages (Fig. 4A). According to our model, CXCR6 activates IFNG release in innate T cells, which is received by IFNGR1 (IFNG receptor 1) in secretory cells. In turn, IFNG induces the expression of TNC, an extracellular glycoprotein up-regulated in infected tissues, which binds to TLR4 and results in secretion of CXCL16. Mechanistically, the interaction between CXCL16 and CXCR6 is necessary for the recruitment of T lymphocytes to infected tissues. The absence of this interaction together with a 56% decline in innate T cells expressing CXCR6 demonstrates the impaired innate T cell recruitment in patients with severe symptomatology (Fig. 4B). In summary, the feedback loops unique to mild disease courses contribute to survival and recruitment of B and T lymphocytes required for viral clearance.

(A) A feedback loop between T cells, secretory epithelial cells, and macrophages maintain T cell recruitment in patients with mild symptoms. (B) Expression of CXCR6 across cell populations in mild and severe cases. (C) Predicted scores for each potential target protein expressed as the percentage of inflammatory feedback loops inhibited after computational perturbation. (D) Uninterrupted feedback loops after computational inhibition of VCAN in patients with severe symptoms.

Hyperinflammation is maintained in patients with severe symptomatology. In addition to mild disease cases, we analyzed the causal feedback loops identified in patients with severe symptomatology to characterize distinct patterns in the immune response. Severe COVID-19 cases are known to develop a hyperinflammatory response to SARS-CoV-2, which is characterized by an excess of proinflammatory cytokine secretion (20). To elucidate the mechanism that maintains the excessive release of cytokines in severe cases, we first performed a topological analysis of the interconnected feedback loops and assessed differences in network stability between mild and severe cases. In contrast to mild cases, we observed a threefold increase in the interconnectivity of the feedback loops (density, 0.219 and 0.072), which can be attributed to the establishment of redundancy in receptor activation. While, in mild cases, each receptor is activated by a median of one ligand, a median of three ligands activates each receptor in severe cases according to our model (one-sided Wilcoxon rank sum test, P = 0.009). In addition, the number of induced ligands per receptor significantly decreases (one-sided Wilcoxon rank sum test, P = 0.002), which implies that more cellular populations participate in the formation of causal feedback loops in severe cases compared to mild disease courses. In summary, patients with severe symptomatology show a significant redundancy in the activation of receptors and intracellular signaling cascades. Because of the involvement of an increased number of cellular populations in the activation of these receptors, a stable feedback regulation is established that is robust to fluctuations in ligand secretion.

Next, we sought to determine whether this stable feedback regulation maintains the hyperinflammatory immune response by secretion of proinflammatory cytokines. In contrast to patients with mild symptoms, we observed causal feedback loops involving an array of proinflammatory cytokines, namely, TNF, IFNG, LT-, IL-27, IL-6, IL-15, and IL-18. Moreover, these loops contain the extracellular matrix proteins FN1, VCAN, and PLAU, which suggests the induction of proinflammatory ligand secretion by cell adhesion. In agreement with previous reports, our model predicts that these genes induce the expression of the proinflammatory cytokines IL-15 and IFNG (21, 22). Therefore, the hyperinflammatory condition in patients with severe COVID-19 symptomatology is maintained by the redundant secretion of proinflammatory cytokines and signaling through extracellular matrix proteins that reinforces their induction.

IL-10 induces the expression of proinflammatory cytokines in macrophages. In addition to the maintenance of proinflammatory cytokine release through interconnected feedback loops in patients with severe symptomatology, our model predicts that IL-10 induces proinflammatory molecules, including IL-18, FN1, PLAU, and VCAN, in a subpopulation of macrophages, despite its known role as an anti-inflammatory cytokine (23). IL-10 signaling has been shown to suppress proinflammatory cytokine expression through the activation of the signal transducer and activator of transcription 3 (STAT3). In addition, it also can activate STAT1 and STAT5 (24). Because of the induced expression of proinflammatory cytokines in our model, we investigated which downstream TFs are activated by IL-10 signaling in our model and observed that only STAT1, but not STAT3, is activated in macrophages. This finding is consistent with a previous study demonstrating that IL-10 is reprogrammed toward STAT1 induction in the presence of IFNG, which establishes a proinflammatory gene expression profile (25). To further support this finding, we investigated whether known proinflammatory target genes of IL-10 signaling are elevated in this subpopulation compared to patients with mild symptoms where IL-10 signaling activates both STAT1 and STAT3. We observed a statistically significant increase in the expression of IL-6, CXCL8, vascular endothelial growth factor, and IL-17R (P < 6.58 107), the major targets of IL-10 (25, 26). Together, this indicates that IL-10 signaling contributes to the establishment of a proinflammatory gene expression profile in a subpopulation of macrophages by activating STAT1, but not STAT3.

After characterizing the molecular differences and commonalities in patients with mild and severe COVID-19 pathologies, we set out to identify potential target genes for modulating the immune response. Therefore, we sought to identify genes that could be targeted for modulating the immune response in patients with severe symptomatology. On the basis of the identified positive feedback loops in patients with mild and severe symptomatology, we simulated the effect of inhibiting ligands and receptors, by manually removing each gene individually from the positive feedback loops. Then, we ranked the perturbed genes by their ability to disrupt feedback loops unique to severe disease cases (see Materials and Methods for details). As a result, our simulation identifies VCAN and TLR2 as the top-ranking target genes whose inhibition disrupts 76 and 75% of the interconnected feedback loops unique to severe cases, respectively, while not interfering with immune response mechanisms common to both cases (Fig. 4C). In particular, the maintenance of IL-6, IL-18, IL-15, and IL-27 and the induction of proinflammatory cytokines by IL-10 are fully disrupted according to our model, while the anti-inflammatory, autocrine feedback of IL-21 in regulatory T cells remains intact (Fig. 4D). However, to date, no approved compound inhibiting VCAN exists. In addition, adapalene has been identified as an inhibitor of TLR2. However, its current application form (topical administration) is not readily suitable for patients with COVID-19.

Last, we aimed at validating the predicted molecules using two independent bronchoalveolar lavage fluid samples from German patients with severe symptomatology (27). Using our methodology, we reconstructed the positive feedback loops and compared it against the previously detected positive feedback loops of patients with mild symptomatology. As a result, TLR2 and VCAN were able to disrupt the fifth and sixth most feedback loops, only exceeded by CCR6, its cognate ligand CCL20, IL-2RG, and syndecan 1. In contrast to the cell-cell interactome of the German patients where the interaction between CCL20 and CCR6 is present, it could not be detected in the cell-cell interactomes of Chinese patients, since the receptor was not significantly associated with the expression of downstream TFs, which warrants the comparison to samples from patients with mild symptoms having the same genetic background.

In this study, we proposed a computational model for predicting immunomodulatory compounds and target proteins to treat severe symptomatology in patients with COVID-19. The model integrates both intra- and extracellular signaling interactions with gene regulatory networks. Unlike current strategies for identifying potential immunomodulatory proteins and compounds, our method detects and exploits the amplifying feedback loops governing the dysregulated inflammatory response, thereby providing a holistic view of the extracellular cell-cell communication network underlying the disease pathology. In addition, not only the extracellular signaling is modeled by evidence-based cognation of ligand and receptors, but it is also further scrutinized in light of the compatibility with downstream intracellular signaling cascades. In contrast to current strategies for predicting immunomodulatory proteins and compounds, to our knowledge, this is the first method incorporating molecular information about inflammatory processes.

The proposed methodology relies on positive feedback loops, which play a key role in the amplification of the immune response to pathogenic factors (8, 9). Although positive feedback loops can be observed in physiological immune reactions, a runaway inflammation is prevented through the establishment of negative feedback loops (28). In contrast, pathological inflammation is characterized by the presence of positive feedback loops whose amplification is insufficiently restricted by negative feedback loops. Therefore, it is expected to find positive feedback loops under both conditions. In this regard, the proposed strategy of targeting positive feedback loops unique to pathological immune responses constitutes a rational approach for modulating inflammation, since it disrupts loops that are newly established and amplified because of the absence of sufficient negative feedback.

The validity of our method was further corroborated by predicting immunomodulatory proteins and compounds targeting them in the context of 12 diseases characterized by a pathological immune response. In particular, we were able to validate 93% of the top-ranking proteins with previous studies highlighting the efficacy of targeting these predicted genes. In the context of COVID-19, using our method, we were able to identify VCAN and TLR2 as potential targets for immunomodulatory attempts, which was further confirmed by analyzing two independent patients with severe symptomatology. VCAN is an extracellular matrix glycoprotein, which creates a strongly adhesive environment for monocytes and T cells (29, 30). In response to an acute inflammation, VCAN accumulates in the extracellular matrix of the inflamed tissue leading to accumulation of leukocytes (31). Previous studies showed that interference with VCAN significantly dampens virus-induced inflammation and CCL2-induced monocyte migration (32). We hypothesize that the overexpression of VCAN in severe disease cases results in the excessive accumulation of proinflammatory monocytes in the lung, which is further supported by an increased number of monocytes. Thus, VCAN constitutes a plausible, novel target gene for modulating the hyperinflammatory response in patients with COVID-19 with severe symptomatology.

Despite the demonstrated ability of our method to predict immunomodulatory proteins and compounds, it has limitations. Namely, it requires single-cell RNA-seq data of tissues displaying pathological and physiological immune responses, which is currently not widely available. However, the steadily increasing availability of single-cell technologies and publicly available single-cell RNAseq datasets is expected to alleviate this issue in the future. In addition, our method focuses on the effect of ligand-receptormediated cell-cell communication. Nevertheless, other signaling mechanisms exist that are important for establishing a proper inflammatory response, such as through the exchange of exosomes (33), which could extend the current scope of this method. Last, accurate predictions of our method require the correct clustering and annotation of the input data. Although the inherent heterogeneity in single-cell data is an advantage in the detection of strong cell-cell interactions, artificial heterogeneity as a result of inaccurate cell type identification is a notable impediment to the detection of feedback loops. In particular, the association of ligands to causally dependent receptors inducing their expression requires the presence of a sustained regulatory path, which is crucially dependent on the cell type each cell is associated with.

In summary, the presented method provides the first strategy to systematically identify immunomodulatory proteins and the compounds targeting them. Thus, we believe that it will be of great utility in the characterization of pathological immune responses and in the design of novel therapeutic interventions for a wide range of diseases associated with exuberant or persistent inflammation, including COVID-19.

Single-cell RNA-seq datasets were obtained from publicly available databases (table S2). Whenever possible, datasets were obtained in processed form. In case this was not possible, each dataset was processed according to the guidelines in the original studies reporting them. Cell type identification for COVID-19 samples of German patients was performed by transferring the labels of COVID-19 samples of Chinese patients using Seurats TransferData function (34, 35).

A cell-cell communication scaffold was generated by manual curation of PPIs contained in iRefIndex (version 16, 09.10.2019) (36) and in a previously published dataset (37). PPIs from iRefIndex were selected that showed taxon ID 9606, interaction type MI:0407 (Direct Interaction) and contained an HGNC (HUGO Gene Nomenclature Committee) symbol information for both interactors. Furthermore, the interaction had to involve one ligand and receptor, respectively, based on the definition provided in the Cell-Cell Interaction Database (38, 39). An interaction was included in the dataset if the binding occurred extracellularly.

The intracellular signaling network is composed of pathway interactions included in OmniPath (40), Reactome (41), or MetaCore from Thomson Reuters. In particular, all pathways from MetaCore were obtained including all signal transduction interactions while discarding transcriptional gene regulatory interactions.

Gene regulatory interactions were obtained from MetaCore from Thomson Reuters, a manually curated resource of gene-gene interactions, on 01.04.2019 for human genes. Only transcriptional regulatory interactions with known effects, i.e., activation or inhibition, were selected by filtering for direct interactions with reported effects activation or inhibition.

The main algorithm consists of four steps. First, preserved TFs of each cell type are detected. For that, the method discretizes the expression matrix, such that (non)zero counts become 1 (0), respectively, and selects TFs that are expressed in (i) at least 5% of cells of a given cell type and (ii) the 95 percentile of cells.

Second, preserved TFs are connected to the receptors inducing their expression using a Markov chain model of intracellular signaling, called SigHotSpotter (42). SigHotSpotter relies on single-cell RNA-seq data of a single cell type and an intracellular signaling network to simulate the traversal of an extracellular signal through the network as a discrete time Markov chain. Genes with the highest steady-state probability are selected and defined as being active or inactive depending on their compatibility with downstream, preserved TFs. Compatibility is determined by assessing the sign of all shortest paths between an intermediate molecule and its downstream TF targets. A path is considered activating if it consists an even number of inhibitions and inhibiting otherwise, such that both the intermediate and downstream TFs have to be expressed in case of an activating path and not expressed in case of an inhibiting path. An intermediate molecule is considered compatible if a significant number of its target is compatible and assessed with a hypergeometric test (P cutoff = 0.05). Following the same rationale, receptors are identified that target intermediate molecules, which establishes a connection between receptors and preserved TFs.

Third, ligands expressed in at least 5% of cells of each cell type are selected. Last, ligand-receptor interactions are established between two cell populations if (i) the receptor was selected in the first step for the first population, (ii) the ligand was selected in the second step for the second population, and (iii) the receptor-ligand interaction is contained in cell-cell communication scaffold. Every interaction is augmented with an interaction strength defined as the product of the average receptor expression and average ligand expression in all cells of a population expressing the receptor or ligand, respectively. Significance of each interaction is determined on the basis of the scores of all cell-cell interactions in the cell-cell communication scaffold between the two interacting populations. Scores in the top quantile are considered significant.

Causal feedback loops were inferred following three steps. Initially, we linked each receptor in the cell-cell communication network to its coexpressed ligands in each cell population having a conserved regulatory path from the receptor. For that, we first transformed the expression data into binary values as described for the selection of preserved TFs. Then, significantly coexpressed ligands and receptors are detected by comparing the fraction of cells agreeing in binary receptor and ligand expression to a random distribution composed of coexpression values of randomly paired ligands and receptors in the same population. A P value below 0.05 is deemed significant. Eventually, conserved shortest paths between the receptor and its coexpressed ligands are calculated on the TF-gene interactomes (see above) using igraph (43). In particular, starting from TFs being targeted by the receptors in the cell-cell interactomes, the fraction of cells in a subpopulation sharing a shortest path to a coexpressed ligand is computed when the receptor is active and inactive, respectively. A user-defined threshold for the minimum (maximum) fraction of cells having a conserved path when the receptor is active (inactive) links receptors to ligands intracellularly. Here, minimum and maximum thresholds of 0.4 and 0.25 were used, respectively.

To identify causal feedback loops, we combined cell-cell interactions and intracellular links between receptors and the coexpressed ligands in a global network representation using igraph (43).

Using the network of identified positive feedback loops under two conditions, the method computes potential target proteins defined as being unique to the feedback loops of the pathologic condition. Each potential protein is scored as follows: First, the number of ligands participating in pathological feedback loops is computed on the basis of a manually assembled list. Subsequently, the strongly connected components of the pathological feedback loop graphs are computed after removing the potential target gene. Last, the fraction of removed ligands within strongly connected components is computed, which serves as the score for the protein. Compounds inhibiting the top-scoring proteins are obtained from a list of drugs and their inhibited target proteins originally retrieved from DrugBank (16).

Acknowledgments: Funding: This work was supported by the Fonds National de la Recherche (FNR) Luxembourg (references: SysBioCOVID19 & 11662681/InTRinSIC). Author contributions: S.J. wrote the software, performed the analysis, created figures, and wrote the manuscript. I.P. performed the analysis, created figures, and wrote the manuscript. S.C. performed the analysis and created figures. A.d.S. supervised the work, wrote the manuscript, and conceived the idea. Competing interests: The authors declare that they have no competing interests. Data and materials availability: All data supporting the findings of this study are available within the paper and/or the Supplementary Materials. Additional data related to this paper may be requested from the authors. Correspondence and material requests should be directed to A.d.S.

See original here:
Leveraging systems biology for predicting modulators of inflammation in patients with COVID-19 - Science Advances

REGENHU announces its participation in the FLAMIN-GO project aimed at developing "personalized" treatments against rheumatoid arthritis -…

REGENHU today announced its participation in the FLAMIN-GO research project aimed at developing an organ-on-chip technology for clinical trials on Rheumatoid Arthritis (RA).

FLAMIN-GO is an EU-funded project featuring a strong, well-balanced composition of hospital, academia, and industry partners which brings together the know-how of experts in the fields of rheumatology, material science, tissue engineering, nanotechnology, cell biology, and 3D modeling, in a cohesive, transdisciplinary, multi-sectorial approach. The objective is to develop a personalized next-generation synovia-on-chip, that, by effectively mimicking the complexity of an RA joint, will enable personalized clinical trials-on-chip, and ultimately open new avenues toward personalized care in RA.

FLAMIN-GO was designed precisely with the intention of opening up a new pathway towards personalized medicine in the treatment of Rheumatoid Arthritis. The goal is to provide an organ-on-chip solution (OoC) whereby the best drug on the market for treating each patient can be selected, which will also support the development of new drugs. This solution will be based on the design and production of a multi-compartment microfluidic platform for 3D cell culture and perfusion of all joint tissues relevant to the disease, mentioned Annalisa Chiocchetti, project coordinator and Professor of Immunology at the University of Eastern Piedmont in Novara.

REGENHUs participation in the consortium will be three-fold: the company will be providing (a) a customized 3D bioprinter and associated design software capable of printing cell layers into the organ chips in a highly reproducible and accurate manner with high cell viability. (b) training in the use of bioprinter and software, and (c) technical support to the consortium members in resolving issues with printability of biomaterials.

For REGENHU, joining FLAMIN-GO represents a great step forward in maintaining our leadership in 3D bioprinting innovation, said Simon MacKenzie, Chief Executive Officer, REGENHU. We believe that the knowledge generated in the consortium will lead to the discovery of new approaches to help deliver meaningful, novel, alternative solutions for the millions of patients living with RA across Europe.

Rheumatoid arthritis is an autoimmune inflammatory disorder, characterized by synovial joint inflammation, affecting 1% of the overall population. Around 2,9 million patients in the EU are affected by this condition.

Over time, RA can also lead to permanent disability. Currently, there is no cure for RA, but remission of symptoms is more likely when treatment begins early, with disease-modifying antirheumatic drugs (DMARDs).

FLAMIN-GO is a research project designed to develop tailored treatments for each patient suffering from Rheumatoid Arthritis. Under the leadership of the University of Eastern Piedmont, the project originated from a collaboration among different public and private organizations, including the Institute of Nanotechnology of the National Research Council (Cnr Nanotec) at Lecce, Queen Mary University of London, the Max Planck Institute, and the Swiss AO Research Institute Davos, ARI.

REGENHU is a research-driven, Swiss MedTech bioprinter pioneer committed to assisting the research and scientific communities by creating and developing state-of-the-art bioprinting technologies to revolutionize medicine.

Founded in 2007, the dynamic and rapidly expanding company is based in Villaz-St-Pierre, Switzerland, with offices in the United States, and distributors in Asia and Oceania.

See the original post here:
REGENHU announces its participation in the FLAMIN-GO project aimed at developing "personalized" treatments against rheumatoid arthritis -...

Global Cell Counting Market || keyplayers Thermo Fisher Scientific Inc., Becton, Dickinson and Company Merck KGaA, and Bio-Rad Laboratories. KSU |…

The reportGlobal Cell Counting Market, ByProduct (Instruments and Consumables & Accessories), By End-user (Research & Academic Institutes, Hospitals & Diagnostic Labs, Biotechnology & Pharmaceutical Companies, and Others),and By Region (North America, Europe, Asia Pacific, Latin America, and the Middle East & Africa) Trends, Analysis and Forecast till 2029.

Key Highlights:

Analyst View:

Increasing developments in cell counting instruments

Global cell counting market primarily attributed to the major drivers in this market such as the increased need for non-invasive diagnosis, advancements in single cell sequencing technique, and increase in adoption of personalized medicine. These individualized care regimes are improving quality of life of the patients and reducing economic, societal, and clinical burden, projecting a future of prosperity.

The applications of single cell multi-omics primarily include oncology, cell biology, neurology, stem cell and immunology, among others. Aside from the discovery of effective biomarkers for the development of efficient targeted drug therapy, single cell approach also facilitates gene expression and protein expression analyses in an individual cell. Research and academic organizations, biotechnology and biopharmaceutical companies, and diagnostic centers, among others, are prominent end users of single cell multi-omics solutions.

Growing cell counting industry

The applications of single cell multi-omics primarily include oncology, cell biology, neurology, stem cell, and immunology, among others. Aside from the discovery of effective biomarkers for the development of efficient targeted drug therapy, single cell approach also facilitates gene expression and protein expression analyses in an individual cell. Research and academic organizations, biotechnology and biopharmaceutical companies, and diagnostic centers, among others, are prominent end users of single cell multi-omics solutions.

Browse 60 market data tables* and 35figures* through 140 slides and in-depth TOC on GlobalCell CountingMarket, ByProduct (Instruments, and Consumables & Accessories); By Application (Research & Academic Institutes, Hospitals & Diagnostic Labs, Biotechnology & Pharmaceutical Companies, and Others),and By Region (North America, Europe, Asia Pacific, Latin America, and the Middle East & Africa) Trends, Analysis and Forecast till 2029

Key Market Insights from the report:

The global cell counting market accounted for US$ 11.1 billion in 2020 and is estimated to be US$ 19.7 billion by 2029 and is anticipated to register a CAGR of 6.6%. The market report has been segmented on the basis ofproduct, application, and region.

To know the upcoming trends and insights prevalent in this market, click the link below:

https://www.prophecymarketinsights.com/market_insight/Global-Cell-Counting-Market-4522

Competitive Landscape:

The prominent player operating in the global pharmaceutical membrane filtration technologies market includesDanaher Corporation, Thermo Fisher Scientific Inc., Becton, Dickinson and Company Merck KGaA, and Bio-Rad Laboratories.

The market provides detailed information regarding the industrial base, productivity, strengths, manufacturers, and recent trends which will help companies enlarge the businesses and promote financial growth. Furthermore, the report exhibits dynamic factors including segments, sub-segments, regional marketplaces, competition, dominant key players, and market forecasts. In addition, the market includes recent collaborations, mergers, acquisitions, and partnerships along with regulatory frameworks across different regions impacting the market trajectory. Recent technological advances and innovations influencing the global market are included in the report.

About Prophecy Market Insights

Prophecy Market Insights is specialized market research, analytics, marketing/business strategy, and solutions that offers strategic and tactical support to clients for making well-informed business decisions and to identify and achieve high-value opportunities in the target business area. We also help our clients to address business challenges and provide the best possible solutions to overcome them and transform their business.

Some Important Points Answered in this Market Report Are Given Below:

Key Topics Covered

To know more

Contact Us:

Sales

Prophecy Market Insights

Email-sales@prophecymarketinsights.com

Read more:
Global Cell Counting Market || keyplayers Thermo Fisher Scientific Inc., Becton, Dickinson and Company Merck KGaA, and Bio-Rad Laboratories. KSU |...

GenSight Biologics Announces the Publication in Communications Biology of the Proof-of-Concept for GS030-Drug Product in NonHuman Primates – BioSpace

Feb. 4, 2021 06:30 UTC

PARIS--(BUSINESS WIRE)-- Regulatory News:

GenSight Biologics (Paris:SIGHT) (Euronext: SIGHT, ISIN: FR0013183985, PEA-PME eligible), a biopharma company focused on developing and commercializing innovative gene therapies for retinal neurodegenerative diseases and central nervous system disorders, today announced that the journal Communications Biology has published results from the study of GS030- Drug Product (GS030-DP) in non-human primates (NHP).

The paper*, published in the January issue under the title Optogenetic therapy: high spatiotemporal resolution and pattern discrimination compatible with vision restoration in non-human primates, is the first peer-reviewed article constituting a proof-of-concept for retinal ganglion cell (RGC) activation following optogenetic gene therapy with GS030-DP (rAAV2.7m8-ChrimsonR-tdT) in non-human primates. Specifically, the spatiotemporal activation of RGCs allowed for pattern discrimination leading to an estimated Snellen visual acuity of 20/249, superior to the level of legal blindness.

We are proud to have these results, which have been used to support the IND approval of our Phase I/II clinical trial PIONEER with GS030, published in Communications Biology, commented Bernard Gilly, Co-founder and Chief Executive Officer of GenSight. This Phase I/II clinical trial is currently recruiting retinitis pigmentosa patients with bare light perception and its objective is to demonstrate that NHP observations translate into useful visual restoration in these patients.

GS030-DP (rAAV2.7m8-ChrimsonR-tdT) is an optimized viral vector expressing the light-sensitive opsin ChrimsonR. When activated by amber light, ChrimsonR renders its host cell photosensitive, a function lost in retinal diseases causing the degeneration of photoreceptors. Optogenetics combine the cellular expression of light-sensitive opsins with fine-tuned light stimulation generated by a wearable optronic visual stimulation device (GS030-MD).

Preclinical studies generated key findings that supported the initiation of the first-in-human Phase I/II clinical trial PIONEER evaluating the safety and tolerability of the GS030 combined therapy (GS030-DP + GS030-MD) in patients with late-stage retinitis pigmentosa.

This preclinical study represents an important milestone towards the clinical validation of this approach to restore some vision in blinding retinal conditions. This journey that started more than a decade ago with the collaboration between my team at Institut de la Vision in Parisa and Pr. Botond Roska, has also benefited from scientific synergies with the team of Ed Boyden at the MIT, said Jos-Alain Sahel, MD, co-founder of GenSight and of the Institut de la Vision, Director of the IHU FOReSIGHT and Chairman of the Department of Ophthalmology at University of Pittsburgh School of Medicine. We expect that the results of the clinical trial PIONEER will indeed confirm the potency of the approach in the interest of patients.

Expression of ChrimsonR-tdT in the retina of non-human primates was safe and well tolerated

The intravitreal injection of rAAV2.7m8-ChrimsonR-tdT and the expression of ChrimsonR-tdT in the retina did not induce any significant immune reaction or intraocular inflammation. Under ambient lighting, no photophobia or visionrelated changes in behavior was noted in any of the animals injected with rAAV2.7m8-ChrimsonR-tdT. Of note, the wavelength of amber light needed to activate ChrimsonR is much safer than that of highly phototoxic blue-shifted lights.2

The AAV2.7m8 vector showed high transduction efficiency in retinal ganglion cells (RGCs)

The modified viral vector AAV2.7m8 was generated using in vivodirected evolution and selected for its ability to efficiently transduce retina cells when injected in the vitreous.1 The article authored by Gauvain et al. showed that, in macaques injected intravitreally, AAV2.7m8 transduced RGCs more efficiently than the wild-type AAV2 vector. A strong cellular expression of ChrimsonR-tdT was observed in the perifovea, where RGCs are most concentrated. Interestingly, the fluorescent marker protein td-Tomato fused to ChrimsonR seemed to increase the expression of functional opsin.

The therapeutic dose of rAAV2.7m8-ChrimsonR-tdT was defined as 5 1011 vg/eye, which allowed for greater light sensitivity and higher cellular expression in a wider area of the retina.

ChrimsonR-tdT generated a photocurrent with high temporal and spatial resolution

In functional assays (256-mutlielectrode arrays), the RGCs expressing ChrimsonR-tdT were only activated by amber light at a minimal intensity of 1015 photons cm2 s1 and did not show any response to ambient light.

The ex vivo retinal stimulation assays also showed that the electrophysiologic response of RGCs expressing ChrimsonR precisely followed the duration and frequency of the light pulses used to activate the opsin. Moreover, localized stimulation of RGCs induced a response coherent with the size and position of the light pulses.

Optogenetic stimulation of RGCs expressing ChrimsonR-tdT can support restoration of visual acuity

The electrophysiological activity of RGCs expressing ChrimsonR-tdT was consistent with the direction and speed of a moving stimulus. Furthermore, the spatiotemporal activation of treated retinas was specific to the shape of the moving symbols presented (square, circle, cross of different sizes), indicating the ability to discriminate between patterns. This level of pattern discrimination corresponded to a Snellen visual acuity of 20/249 (1.1 LogMAR), a level above the threshold of blindness (20/400) defined by the World Health Organization.3 The authors concluded that These results lay the groundwork for the ongoing clinical trial, PIONEER, with the AAV2.7m8-ChrimsonR-tdT vector for vision restoration in patients with retinitis pigmentosa.

The paper is available at https://www.nature.com/articles/s42003-020-01594-w.

GenSight Biologics expect to release early findings in the first patients of the PIONEER trial later in the first half of 2021.

*About the paper:

Optogenetic therapy: high spatiotemporal resolution and pattern discrimination compatible with vision restoration in non-human primates

Authors:

Gregory Gauvain1, Himanshu Akolkar1,2, Antoine Chaffiol1, Fabrice Arcizet1, Mina A. Khoei1, Mlissa Desrosiers1, Cline Jaillard1, Romain Caplette1, Olivier Marre1, Stphane Bertin3, Claire-Maelle Fovet4, Joanna Demilly4, Valrie Forster1, Elena Brazhnikova1, Philippe Hantraye4, Pierre Pouget5, Anne Douar6, Didier Pruneau6, Jol Chavas6, Jos-Alain Sahel1,2,3, Deniz Dalkara1, Jens Duebel1, Ryad Benosman1,2, Serge Picaud1.

Affiliations:

1 Sorbonne Universit, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France.2 Department of Ophthalmology, University Pittsburgh Medical Center, Pittsburgh, PA, USA.3 CHNO des Quinze-Vingts, INSERM-DGOS CIC 1423, 28 rue de Charenton, F-75012 Paris, France.4 Dpartement des Sciences du Vivant (DSV), MIRcen, Institut dimagerie Biomdicale (I2BM), Commissariat lEnergie Atomique et aux Energies Alternatives (CEA), 92260 Fontenay-auxRoses, France.5 ICM, UMRS 1127 UPMC U 1127 INSERM UMR 7225 CNRS, Paris, France.6 Gensight Biologics, 74 rue du faubourg Saint Antoine, F-75012 Paris, France.

References:

About GenSight Biologics

GenSight Biologics S.A. is a clinical-stage biopharma company focused on developing and commercializing innovative gene therapies for retinal neurodegenerative diseases and central nervous system disorders. GenSight Biologics pipeline leverages two core technology platforms, the Mitochondrial Targeting Sequence (MTS) and optogenetics, to help preserve or restore vision in patients suffering from blinding retinal diseases. GenSight Biologics lead product candidate, LUMEVOQ (GS010; lenadogene nolparvovec), has been submitted for marketing approval in Europe for the treatment of Leber Hereditary Optic Neuropathy (LHON), a rare mitochondrial disease affecting primarily teens and young adults that leads to irreversible blindness. Using its gene therapy-based approach, GenSight Biologics product candidates are designed to be administered in a single treatment to each eye by intravitreal injection to offer patients a sustainable functional visual recovery.

About GS030

GS030 leverages GenSights optogenetics technology platform, a novel approach to restore vision in blind patients using a combination of ocular gene therapy and tailored light-activation of treated retinal cells. In diseases causing degeneration of photoreceptors, a therapeutic gene encoding a light-sensitive protein (ChrimsonR-tdT) is introduced into retinal ganglion cells (RGC) to turn them into photosensitive cells, and thereby restore the ability of the retina to respond to light. Chrimson-tdT is a light-sensitive channelrhodopsin that is activated by high intensities of amber light. An external wearable medical device is therefore needed to stimulate the treated retina. The lightstimulating goggles (GS030-MD) encode the visual scene in real-time and project a light beam at a specific wavelength and intensity onto the treated retina. Treatment with GS030 requires that patients wear the external wearable device to enable restoration of visual function. With the support of the Institut de la Vision in Paris and the team of Dr. Botond Roska at the Friedrich Miescher Institute in Basel, GenSight is developing GS030 combined optogenetic therapy to restore vision in patients suffering from retinitis pigmentosa (RP). Of note, GenSights optogenetics approach is independent from the specific genetic mutations causing blindness. This technology could be applied to other diseases of the retina in which photoreceptors degenerate, like dry agerelated macular degeneration (dry-AMD).

About Optogenetics

Optogenetics is a biological technique that involves the transfer of a gene encoding for a light sensitive protein to cause neuronal cells to respond to light stimulation. As a result, it is a neuromodulation method that can be used to modify or control the activities of individual neurons in living tissue and even in-vivo, with a very high spatial and temporal resolution. Optogenetics combines the use of gene therapy methods to transfer a gene into target neurons with the use of optics and electronics (optronics) to deliver the light to the transduced cells. Optogenetics is widely used by research laboratories throughout the world and holds clinical promise in the field of vision impairment or degenerative neurological disorders.

About Retinitis Pigmentosa (RP)

Retinitis Pigmentosa (RP) is a family of orphan genetic diseases caused by multiple mutations in numerous genes involved in the visual cycle. Over 100 genetic defects have been implicated. RP patients generally begin experiencing vision loss in their young adult years, with progression to blindness by age 40. RP is the most widespread hereditary cause of blindness in developed nations, with a prevalence of about 1.5 million people throughout the world. In Europe and the United States, about 350,000 to 400,000 patients suffer from RP, and every year between 15,000 and 20,000 new patients with RP lose sight. There is currently no existing curative treatment for RP.

a Serge Picaud, Jens Duebel, Deniz Dalkara and Gregory Gauvain

View source version on businesswire.com: https://www.businesswire.com/news/home/20210203005985/en/

Read more:
GenSight Biologics Announces the Publication in Communications Biology of the Proof-of-Concept for GS030-Drug Product in NonHuman Primates - BioSpace

AstraZeneca and UCL to collaborate on two immuno-oncology projects – PharmaTimes

British drugmaker and researchers from the UCL Division of Infection & Immunity will collaborate on two projects which will aim to contribute to the development of new cancer treatments.

The new research collaborations will investigate immune checkpoints key biochemical pathways that regulate the bodys immune responses.

Although immune checkpoints help to keep the bodys immune response at normal levels by not harming healthy cells, they can also block specialist immune cells from attacking and destroying cancer cells.

Over the last decade, the emergence of checkpoint inhibitor drugs have revolutionised cancer treatment and demonstrated benefit in clinical results for patients with solid tumours.

Meanwhile, the two AstraZeneca/UCL projects will focus on increasing understanding of immune checkpoint mechanisms and how to manipulate them.

The ultimate aim of the projects will to be to aid the development of new immunotherapy approaches.

AstraZeneca will provide a number of compounds for the projects, while UCL will use unique preclinical models and an array of molecular and cell biology techniques to study these pathways.

These collaborations with AstraZeneca will bring together some of the very best minds in immuno-oncology, said Dr Kathryn Walsh, executive director, office of the Vice-Provost (Enterprise), UCL.

Working together, experts from both institutions will push the boundaries of our understanding of the role of the bodys immune system. In the future, these insights will play a valuable role in how we will be able to develop new treatments to help patients with solid tumours, she added.

Read the original:
AstraZeneca and UCL to collaborate on two immuno-oncology projects - PharmaTimes

Innate Pharma to Host Key Opinion Leader Discussion on the Potential Role of Lacutamab Across T-Cell LymphomasVirtual event to take place at 2:00 pm…

MARSEILLE, France, Feb. 03, 2021 (GLOBE NEWSWIRE) -- Innate Pharma SA (Euronext Paris: IPH ISIN: FR0010331421; Nasdaq: IPHA) (Innate or the Company) today announced that it will host a virtual key opinion leader (KOL) discussion focused on the potential role of its lead investigational drug candidate, lacutamab, an anti-KIR3DL2 cytotoxicity-inducing antibody in development for T-cell lymphomas. The event will be held virtually on Tuesday, February 9, 2021 at 2:00 p.m. CET / 8:00 a.m. ET.

The event will feature presentations from the Companys executive leadership team, as well as the following KOLs:

Presenters will provide an overview of the treatment landscape and prevalence of the lacutamab target, KIR3DL2, across subtypes of T-cell lymphoma. They will also highlight the potential impact of lacutamab in current and upcoming clinical programs in cutaneous and peripheral T-cell lymphomas.

Details for the Virtual Event

The live webcast of the event will be available at the following link:https://edge.media-server.com/mmc/p/yztsebvs

A telephone number will also be made available. Participants may register in advance of the event at http://emea.directeventreg.com/registration/2284358. Upon registration, participants will be provided with dial-in numbers, a direct event passcode and a unique registrant ID that they may use 10 minutes prior to the event start to access the call. Call reminders will also be sent to registered participants via e-mail the day prior to the event.

A replay of the webcast will be archived on Innates website for 90 days following the event.

This information can also be found in the Investors section of the Innate website, http://www.innate-pharma.com.

About Lacutamab:Lacutamab (IPH4102) is a first-in-class anti-KIR3DL2 humanized cytotoxicity-inducing antibody, which is currently in clinical trials for treatment of cutaneous T-cell lymphoma (CTCL), an orphan disease. This group of rare cutaneous lymphomas of T lymphocytes has a poor prognosis with few efficacious and safe therapeutic options at advanced stages.

KIR3DL2 is an inhibitory receptor of the KIR family, expressed by approximately 65% of patients across all CTCL subtypes and expressed by up 90% of patients with certain aggressive CTCL subtypes, in particular, Szary syndrome. It is expressed by up to 50% of patients with peripheral t-cell lymphoma (PTCL). It has a restricted expression on normal tissues.

About Innate Pharma:

Innate Pharma S.A. is a global, clinical-stage oncology-focused biotech company dedicated to improving treatment and clinical outcomes for patients through therapeutic antibodies that harness the immune system to fight cancer.

Innate Pharmas broad pipeline of antibodies includes several potentially first-in-class clinical and preclinical candidates in cancers with high unmet medical need.

Innate has been a pioneer in the understanding of natural killer cell biology and has expanded its expertise in the tumor microenvironment and tumor-antigens, as well as antibody engineering. This innovative approach has resulted in a diversified proprietary portfolio and major alliances with leaders in the biopharmaceutical industry including Bristol-Myers Squibb, Novo Nordisk A/S, Sanofi, and a multi-products collaboration with AstraZeneca.

Based in Marseille, France, Innate Pharma is listed on Euronext Paris and Nasdaq in the US.

Learn more about Innate Pharma at http://www.innate-pharma.com

Information about Innate Pharma shares:

Disclaimer on forward-looking information and risk factors:

This press release contains certain forward-looking statements, including those within the meaning of the Private Securities Litigation Reform Act of 1995.The use of certain words, including believe, potential, expect and will and similar expressions, is intended to identify forward-looking statements. Although the company believes its expectations are based on reasonable assumptions, these forward-looking statements are subject to numerous risks and uncertainties, which could cause actual results to differ materially from those anticipated. These risks and uncertainties include, among other things, the uncertainties inherent in research and development, including related to safety, progression of and results from its ongoing and planned clinical trials and preclinical studies, review and approvals by regulatory authorities of its product candidates, the Companys commercialization efforts, the Companys continued ability to raise capital to fund its development and the overall impact of the COVID-19 outbreak on the global healthcare system as well as the Companys business, financial condition and results of operations. For an additional discussion of risks and uncertainties which could cause the company's actual results, financial condition, performance or achievements to differ from those contained in the forward-looking statements, please refer to the Risk Factors (Facteurs de Risque") section of the Universal Registration Document filed with the French Financial Markets Authority (AMF), which is available on the AMF website http://www.amf-france.org or on Innate Pharmas website, and public filings and reports filed with the U.S. Securities and Exchange Commission (SEC), including the Companys Annual Report on Form 20-F for the year ended December 31, 2019, and subsequent filings and reports filed with the AMF or SEC, or otherwise made public, by the Company.

This press release and the information contained herein do not constitute an offer to sell or a solicitation of an offer to buy or subscribe to shares in Innate Pharma in any country.

For additional information, please contact:

The rest is here:
Innate Pharma to Host Key Opinion Leader Discussion on the Potential Role of Lacutamab Across T-Cell LymphomasVirtual event to take place at 2:00 pm...

Postdoctoral Researcher, Varjosalo laboratory of Molecular Systems Biology/Pathology job with UNIVERSITY OF HELSINKI | 244729 – Times Higher Education…

The HiLIFE - Institute of Biotechnology (BI) is a research institute that operates at the highest international level. It includes 40 research groups and 250 researchers. The Institute is situated in the Viikki science park. For more information on the Institute, please visit https://www.helsinki.fi/en/hilife-institute-of-biotechnology.

The HiLIFE - Institute of Biotechnology, Varjosalo laboratory of Molecular Systems Biology / Pathology, invites applications for a

POSTDOCTORAL RESEARCHER

The Varjosalo lab has a longstanding interest in the global and comprehensive understanding of how key cellular signaling molecules; protein kinases, protein phosphatases and transcription factors mediate their signaling potential and how the signaling is changed in diseases in the immunology and oncology axis, and more recently Covid-19. For this purpose, we have developed experimental and computational methods to identify the protein-interactions formed by these signaling molecules. Additionally, we employ quantitative mass spectrometry -based proteomics to assess the protein abundance changes, as well as phosphorylation status of the tissues/cells. Furthermore, combining the data from the different sets of omics data (proteomics, HTS and HiSeq), allows us to generate global and systems level understanding on how disease-causing mutations mediate their effects, also enabling the development of therapeutic or diagnostic approaches.

We are seeking a post doc that has extensive practical experience in standard biochemistry/molecular biology/cell biological methods, ideally encompassing proteomics and microscopic techniques and/or bioinformatics. The ideal candidate is an adaptable team-player with excellent communication skills in English , both oral and written, and good organizational skills.

This position will provide multiple opportunities for collaborations with potential lab visits abroad and cross-disciplinary scientific exchange, when applicable. An attitude to drive own research and work in an intra and inter team fashion is highly appreciated. In return, the candidate is offered access to a wide variety of methodologies and advanced techniques, appropriate supervision and help in career development.

More information about the lab, including the latest research and news, can be found here:https://www.helsinki.fi/en/hilife-institute-of-biotechnology/research/st...

The position is initially limited to 2 years with a possibility of extension starting from March 2021. A trial period of 6 months will be applied, and the salary will be based on the Universities salary scheme for teaching and research personnel composed of both task specific and personal performance components.

The deadline for the applications is 15th of March, but the position will be filled immediately once a suitable candidate has been identified.

Application should include the following documents as a single pdf file: CV, list of publications, motivation letter including a description of your research interests, and the names and telephone numbers of at least two referees.

Please submit your application, together with the required attachments, through the University of Helsinki electronic recruitment system by clicking on the Apply for job button. Internal applicants (i.e., current employees of the University of Helsinki) submit their applications through the SAP HR portal.

For further information please contact Research Director Markku Varjosalo, PhDmarkku.varjosalo(at)helsinki.fi.

For technical support regarding the recruitment system, please contact rekrytointi@helsinki.fi.

Finland is a member of the EU, has high quality free schooling (also in English), generous family benefits and healthcare, and was recently ranked as the best country in the world for expat families and in the worlds top ten most livable cities. Finland and the Helsinki region possess top expertise in sciences in terms of a vibrant talent pool, leading research, strong support services and functioning collaboration networks. For more information about working at the University of Helsinki and living in Finland, please see helsinki.fi/en/university/working-at-the-university.

Helsinki Institute of Life Science

Helsinki Institute of Life Science (HiLIFE) is a new institute established in 2017 that supports high quality life science research across the University campuses and faculties. HiLIFE builds on existing strengths and new recruits and partnerships to create an attractive international environment for researchers to solve grand challenges in health, food, and environment. HiLIFE coordinates research infrastructures in life sciences and provides research-based interdisciplinary training.

The University of Helsinki, founded in 1640, is one of the worlds leading universities for multidisciplinary research. The university has an international academic community of 40,000 students and staff members. The University of Helsinki offers comprehensive services to its employees, including occupational health care and health insurance, sports facilities, and professional development opportunities. The International Staff Services office assists employees from abroad with their transition to work and life in Finland.

Due date

15.03.2021 23:59 EET

More:
Postdoctoral Researcher, Varjosalo laboratory of Molecular Systems Biology/Pathology job with UNIVERSITY OF HELSINKI | 244729 - Times Higher Education...