Role of the myelomonocytic lineage in Noonan syndrome-associated clinical traits

PhD proposal

Supervisor: A. Yart

Team P. Valet – I2MC

The general aim of the thesis project is to better understand the pathophysiology of Noonan syndrome (NS), a relatively frequent (1/2000 live births) genetic disorder associating cranio-facial features, cardiopathies, short stature and learning disability. Beside these congenital involvements, patients with NS also display a wide range of clinical traits that are reminiscent of early ageing, the pathophysiology of which is misunderstood: bone defects (e.g. decreased bone mass), muscle weakness, predisposition to myeloproliferative disorders (e.g. juvenile myelomonocytic leukaemia, JMML) and metabolic imbalance (e.g. insulin resistance). Interestingly, we and other obtained striking evidences that many of NS impairments are related to the dysfunction of cells from the myelomonocytic lineage : myeloid precursors in JMML, osteoclast in bone defects, monocytes/macrophages in insulin resistance. As a result, our project now aims

  1. to understand the cellular and molecular mechanisms that confer on these cells their pathophysiological potential with a particular focus on the pro-senescent potential of RAS/MAPK hyperactivation,
  2. to assess the relative contribution of myelomonocytic cells to the development of the different traits of the disease, and
  3. to translate the obtained observations to the clinics.

For this, we will favour a translational, comprehensive approach taking advantage of our expertise on the pathophysiology of NS, a wide range of approaches to specifically alter myeloid cells in pre-clinical models (knock in mouse and zebrafish) and evaluate the consequences on major postnatal/evolutive symptoms of the disease and a large and well-documented cohort of patients associated to a full biological collection and results from ongoing clinical trials.

 We expect to demonstrate that a dysfunction of cells from the myeloid lineage, in line with increased senescence, participates to multiple traits of NS, thereby highlighting a founding pathophysiological mechanism which could pave the way for the development of “many birds/one stone” therapeutic strategies. Beyond rare diseases, these results could help better understanding monocyte/macrophage biology and provide new insights into frequent pathologies involving myelomonocytic cells.

Key words: genetic diseases, macrophage, senescence, inflammation

*

Modeling TNF-induced ceramide metabolism changes in T-cell immuno-senescence and resistance of melanoma patients to immune checkpoint inhibitors

PhD proposal

Supervisor: B. Segui

Team T. Levade – CRCT

Cutaneous melanoma is a very aggressive and immunogenic skin cancer that can be treated by immunotherapy based on the administration of antibodies neutralizing CTLA-4 and PD-1, two immune checkpoints expressed on the surface of activated T lymphocytes, acting as a real brake on lymphocyte activation. To date, the best clinical responses in patients with advanced melanoma have been obtained with the combination of ipilimumab (anti-CTLA-4) and nivolumab (anti-PD-1). However, almost half of patients with metastatic melanoma do not respond to this treatment and there are no biomarkers predictive of resistance. In this context, cellular senescence, a process of genetic, epigenetic and metabolic reprogramming as well as morphological modifications of the cell during aging, seems to play a dual role in resistance to immunotherapy.

While the senescence of cancer cells and regulatory T cells appears to facilitate the response to immunotherapies in mice and humans, the senescence of effector T cells may contribute to resistance mechanisms. Among the mechanisms of cellular senescence, a distinction is made between replicative senescence, linked to the shortening of telomeres as cell divisions progress, premature senescence induced by stressors such as pro-inflammatory cytokines (IFNg and TNF) and senescence induced by oncogenes such as BRAFV600E, which is expressed in 50% of melanomas.

Our laboratory has demonstrated two mechanisms of resistance to immunotherapies in murine melanoma models, one based on the production of TNF in the tumor microenvironment, the other dependent on the expression in melanoma cells of sphingosine kinase 1 (SK1), a lipid metabolism enzyme from the ceramide family. In addition, we have shown that the expression of SK1 in human tumor biopsies is a predictive biomarker of resistance to anti-PD-1. Our work constitutes the scientific rationale of two clinical trials in patients with advanced melanoma, TICIMEL and IMMUSPHINX, which aim to assess the impact of anti-TNF agents in melanoma immunotherapy and to identify in the biological samples of patients (blood and tumor biopsies) of biomarkers of resistance / response to immunotherapies. Biomarkers are sought among cytokines and metabolites of ceramide in plasma (measured by mesoscale and mass spectrometry, respectively), profile, gene expression and methylation of peripheral blood mononuclear cells (evaluated by flow cytometry, RNA Seq / CITE Seq single Seq RNA Seq, methyloma), leukocyte infiltrate of tumors (evaluated by immunohistochemistry) and the expression of enzymes of the metabolism of ceramide in tumors (evaluated by in situ hybridization).

The thesis work will consist of developing computer modeling approaches of all the big data collected in the two clinical trials TICIMEL and IMMUSPHINX in order to identify signatures of senescence in regulatory T lymphocytes and effectors predictive of response and resistance to immunotherapies, respectively. In addition, we will study whether these signatures are associated with alterations in the metabolism of ceramide dependent on TNF signaling.

Key words: Cancer, immunotherapy, Clinical trials, Biomarkers, big data, bioinformatics

*

Blood platelets, Thrombosis and Aging: deciphering novel molecular mechanisms for therapy

PhD proposal

Supervisor: S. SEVERIN

Team B. Payrastre – I2MC

The thrombotic risk, mainly due to blood platelet dysfunctions, has a considerable impact in the cardiovascular complications in elderly. However, the mechanisms involved in the increased thrombotic risk and platelet dysfunctions associated to aging are still poorly characterized and therapeutic strategies are very limited. In this context, it seems urgent (i) to better understand the mechanisms involved in the dysfunction of platelet production and activation in an aging context and (ii) to find new pharmacological platelet targets or biomarkers in order to prevent and limit thrombotic complications. The research PhD project aims to finely study the processes of production and activation of blood platelets, from molecular mechanisms to pathophysiology, in an aging context. This project will use transgenic mouse models and a panel of in vivo experimental procedures in mice as well as in vitro and ex vivo imaging and biochemical techniques.

Key words: Blood platelets, Megakaryocytes, Thrombosis, Aging

*

Multiomics analysis and modeling to predict metabolic response in the resistance to leukemia therapies

PhD proposal

Supervisor: J.E. SARRY

Team J.E. Sarry – CRCT

Despite a high rate of complete remission after treatment with genotoxic agents, the prognosis is poor in human acute myeloid leukemia (AML). Front-line chemotherapy is highly effective in ablating leukemic cells, but relapses caused by tumor regrowth initiated by resistant leukemic clones (RLCs) are observed in the majority of patients. The biology of therapeutic resistance currently represents an active area of research. However, the molecular mechanisms underlying drug resistance are still poorly understood in AML, especially in the in vivo context. To address this issue and to characterize chemoresistance and minimal residual disease (MRD), we established a robust patient-derived xenograft (PDX)-based preclinical model that predicts response to chemothepeutics in AML patients. Taking advantage of these in vivo models, we demonstrated that in vivo drug tolerant/resistant AML cells present an enhanced mitochondrial oxidative metabolism. Furthermore, our studies shown that the catabolic flexibility, the inflammatory response, and the metabolic cooperation between stromal and leukemic compartments are key players of mitochondrial activities of chemoresistant AML cells13,14. Therefore our previous results provide not only new targets but also a strong scientific rationale for ongoing clinical trials that assessed emerging combinatory therapies with different mitochondrial inhibitors in AML. Altogether, our work suggests that the mitochondrial function, the metabolic cooperation and symbiosis between the stromal and leukemic compartments inside the bone marrow niche, and inflammatory/stress responses, play a crucial role in drug resistance of AML.

 In light of our current results and evidences, our working hypothesis is that the microenvironment-tumor interplay regulates mitochondrial metabolism of AML cells through the activation of key transcriptional factors and regulons, and of stress responses, leading to drug resistance in vivo. To address this hypothesis and to further decipher the heterogeneity of these drug resistance pathways at both single cell and tissue levels, our methodological strategy is based on functional approaches using well-characterized AML cell lines and primary patient samples in vitro and in our xenograft NSG model coupled to bulk and single cell omics and computational approaches. Finally, our preliminary single cell transcriptomic analysis of cells collected from PDX and patients at diagnosis and relapse uncovered a subpopulation endowed with a specific transcriptional profile implicated into (and predictive of) the relapse in AML.

Key Words: AML, drug resistance, metabolism, in silico modeling, computational prediction

*

Adipose tissue microbiota and ageing

PhD proposal

Supervisor: J. Galitzky

Team A. Bouloumié – I2MC

The development of adipose tissue is dependent on its interaction with the environment and particularly the nutritional one. These last years, the intestinal microbiota has emerged as an important vector in the link between nutrition and fat mass growth. But other components including gender, age and location of the fat depots have also to be considered. Our hypothesis is that there is a molecular relationship between the microbiota which is influenced by age and nutrition and the immune and progenitor cells of the fat depots which govern the structure, diversity and metabolic plasticity of the tissues. We have recently shown that bacteria are present in the tissue and could be essential factors in the interface between the environment and the physiology of the tissue. The research project proposes to study the molecular interaction between bacteria, particularly tissue bacteria, and the microenvironment of fat depots including progenitor cells and immune cells with a particular focus on cell senescence.

Using metagenomic approaches carried out on subcutaneous and visceral adipose depots from human cohorts (FLORINASH cohort), we will analyze the composition and diversity of the tissue microbiota of human adipose depots and the impact of obesity, gender, age and associated pathologies. In parallel, we will dissect the molecular aspects of the interaction between bacterial populations and cells of the adipose microenvironment. We will study the impact of bacteria of interest and their products (identified by our previous work and by the approaches mentioned above) on cell senescence in the microenvironment of fat depots and inversely the impact of cell senescence on bacteria sensing, growth and metabolism. The intracellular pathways will be analyzed with an interest on pathways common to bacterial sensing and senescence including TLRs, NLRs, inflammasome and STING pathways. Finally, the causality of bacterial populations and / or their products of interest in controlling the diversity and metabolic plasticity of fat depots will be studied in vivo in the unique model developed in the team of humanized axenic mice.

Key words: Progenitor cells, senescence, Bacteria, Inflammation, Metabolism, Plasticity

*