All available projects have been listed. Do not submit your application until you have selected a project list.

Project 1

Prof. Paul Pfluger

Institute for Diabetes and Obesity

Research Unit NeuroBiology of Diabetes (NBD)

 

Project Description

Spatial and functional characterization of hypothalamic leptin target genes

Obesity and its comorbid sequelae are major health burdens across European nations. Many citizens would greatly benefit from permanent weight loss, but only a few succeed. They rather suffer from weight regain after dieting, often referred to as Yoyo effect. Delineating the largely unexplored, CNS-driven molecular events that impede sustainable weight loss and drive the Yoyo effect is a prerequisite for future therapies, and a major goal of this internship.

In the past, my lab demonstrated unprecedented weight loss in diet-induced obese mice treated with the plant-derived leptin sensitizer celastrol. Our data suggested breakthrough potential for therapeutic anti-obesity strategies built upon hypothalamic leptin re-sensitization, and pointed towards a key role for orexigenic circuitry residing in the hypothalamus. As key objective for this summer internship, we will delineate the functional role of newly identified genes on leptin resistance and leptin re-sensitization in key subpopulations of hypothalamic neurons. First, using a hypothalamic cellular model, the student will manipulate selected genes using Crispr-Cas9-mediated gene editing and conduct proteome as well as transcriptome analyses to study leptin signalling. Second, using hypothalamic cryo-slices of reporter mice subjected to dietary and pharmacological weight loss interventions and techniques such as RNAscope and confocal microscopy, the student will assess the cellular distribution and the relative expression of the same genes in hypothalamic neurons. Within the 2-months research stay, the student will be implemented into a team of international scientists, with direct supervision and training by PhD students and senior scientists.

 

Project 2

Dr. Gerhard Przemeck

Institute for Experimental Genetics

Functional Genetics Group

 

Project Description 

Analysis of the preweaning disease progression in the AKITA mouse 

Mutant INS-gene-induced diabetes of youth (MIDY) is a severe form of monogenic diabetes mellitus characterized by high levels of ER stress due to misfolding mutant proinsulin. This leads to a substantial decrease in insulin availability resulting in early-onset diabetes (<6 months of age). Commonly, it is believed that reduced plasma insulin and, thus, increasing blood glucose levels coincide with a loss of β-cell mass. Our study identified two novel Ins2 mouse models (Ins2C109G and Ins2V26D) on a C3HeB/FeJ genetic background, both displaying overt diabetes from four weeks of age. Surprisingly, these models showed either no or only a slight decrease in β-cell mass up to 22 weeks, contrasting with existing literature on known Ins2 mutants. To investigate this discrepancy, we closely monitored the differentiation and maturation of β-cells throughout the disease progression to assess the impact of mutant insulin on those processes. Confocal
microscopy revealed a noticeable decrease in cells positive for differentiation markers and a near absence of markers for functional maturity even before weaning and no improvement in older animals. To confirm these findings, quantitative immunohistological analysis have been performed on FFPE-pancreas samples collected at various developmental stages: P7, P15, P21, P28, and from aged animals. We now plan to extend the same analysis carried out for our mouse models to the gold standard MIDY model, the AKITA mouse. We are in the process of breeding mouse cohorts for in-depth phenotyping experiments, spanning from ages 4 to 22 weeks. After, we plan to breed cohorts for organ withdrawal between P7 - P28.


As an intern, you will have the opportunity to process the collected FFPE-pancreas and blood samples. Your main tasks will involve cutting and staining sections of FFPE-pancreas using immunohistochemistry techniques. These sections will serve various purposes, including confocal microscopy to generate high-quality images and whole slide scanning for subsequent statistical analysis of α-, β-, and δ-cell mass, as well as the expression of different β-cell maturation and identity markers. For the analysis of those scans, we will utilize the state-of-the-art AI-based image analysis software Visiopharm. All of this will be done in collaboration with our core facility Pathology and Tissue Analytics. Additionally, available plasma samples will be used for determining insulin levels via ELISA. If time permits, further experiments could involve isolating pancreatic islets for in vitro glucose-stimulated insulin secretion assays or finalizing organ collection for mice in the preweaning age.


Our research interest stems from the lack of knowledge regarding the early progression of MIDY disease before animals become overtly diabetic. Existing literature predominantly focuses on the post-weaning age when β-cells are already heavily compromised. However, insulin expression initiates much earlier in development, with the first immunodetection reported as early as E9.5, and strong signals appearing by E12.5. This precedes the expression of critical drivers of β-cell maturation, such as the transcription factor MAFA. Given that mutant insulin itself is the main driver of MIDY disease development, we believe that the misfolding mutant proinsulin significantly influences differentiation and maturation of β-cells processes early on. In our opinion, it is too late to investigate MIDY disease progression only after weaning. Therefore, we aim to gain insights by shifting the starting point of our research to younger mice.


Join us in exploring this crucial phase of MIDY development and contribute to advancing our understanding of this complex disease.     

Project 3

Dr. Mauricio Berriel Diaz

Institute of Diabetes and Cancer, IDC 

Division Metabolism and Cancer 

 

Project Description

NF-κB signaling in muscle wasting and inflammation during cancer cachexia 

Cancer cachexia is a multifactorial syndrome characterized by involuntary weight loss, diminished muscle strength, and atrophy caused by metabolic dysregulation and anorexia, which are induced by cancer. This systemic disorder affects multiple organs and significantly impacts the quality of life and prognosis of cancer patients. Despite extensive research, the precise mechanisms driving cancer cachexia remain elusive, and effective treatment strategies are lacking. 

Nuclear factor kappa B (NF-κB) signaling plays a crucial role in the loss of muscle mass observed during cancer cachexia. Aberrant activation of this pathway promotes muscle atrophy and impairs muscle regeneration. Pro-inflammatory cytokines and tumor-derived factors contribute to NF-κB activation in muscle, which leads to the expression of genes involved in muscle proteolysis and inflammation, as well as inhibition of muscle protein synthesis. 

In this project, the research intern will utilize a comprehensive approach including molecular biology, biochemical, and pharmacological techniques to further elucidate the molecular mechanisms of NF-κB activation and its downstream effects in cancer cachexia-associated muscle wasting. We will generate reporter cell lines, perform western blot analyses to investigate posttranslational modifications, and employ confocal microscopy to study target protein localization. Additionally, we plan to use specific and potent inhibitors to block NF-κB activation and assess their efficacy in preventing muscle atrophy in vitro using murine and human-derived muscle cells. 

Exploring the complex relationship between NF-κB signaling and muscle metabolism in the context of cancer cachexia offers potential for developing targeted treatment apporaches to mitigate muscle wasting, thereby alleviating the devastating impact of cancer cachexia and improving the clinical outcomes in cancer patients. 

 

Project 4

Prof. Timo Müller

Institute for Diabetes and Obesity

Molecular Pharmacology

 

Project Description

The main goal of our lab is to develop and preclinically evaluate novel drugs designed to treat obesity, diabetes and their associated co-morbidities, such as atherosclerosis and fatty liver disease. Together with our collaboration partner Novo Nordisk, we have over the last decade developed a series of novel therapeutics that that improved body weight and glucose control in diet-induced mice. Within this project, we will teach the candidate how to assess the mode-of-action of newly developed therapeutics on systemic energy and glucose metabolism in vitro and in vivo. New drugs will be tested to engage their target receptors using BRET-based technology. Such assays are designed to demonstrate GPCR activation, internalization, degradation and recycling. Apart from these in vitro studies, the students will learn to assess transcriptional and proteomic changes after in vivo drug testing in selected organs mediating food intake and energy metabolism, including the hypothalamus, hind brain and the brown adipose tissue.

Project 5

Dr. Raffaele Teperino

Institute for Experimental Genetics

Environmental Epigenetics Group

 

Project Description

Small non-coding RNAs in metabolic health, disease, and epigenetic inheritance


Small non-coding RNAs (sncRNAs) are abundant RNAs with mostly regulatory functions of cellular and organismal physiology. More recently, they have been shown relevant for the intergenerational effects. For example, mature spermatozoa contain a plethora of sncRNAs, which is sensitive to environmental challenges and contributes to the paternal control of offspring development and adult phenotypic trajectories, especially metabolic and neurobehavioral. In the Environmental Epigenetics group at the Helmholtz Munich, we study the ontogeny and function of these RNAs. We have developed a method to track them from transcriptional source to site of action and identified a sub-population, which is transcribed in mature spermatozoa, inherited at fertilization, and contributes to paternal control of offspring metabolism. 
Goal of this project will be to join a running effort on the characterization of sncRNAs from mouse spermatozoa and placenta. In particular, the selected intern will help extracting RNA, building sncRNA sequencing libraries and learn the computational analysis of sncRNA-sequencing data from quality control to data representation, integration and interpretation.

Intern involvement 
The intern will be fully involved in the wet and dry lab activities of our group, with a focus on a running project in the lab on the characterization of sncRNAs in gametes and during mouse embryonic development. She/he will therefore gain molecular biology and computational skills necessary for the analysis of sncRNAs-sequencing datasets.

Tasks 
The intern will be trained on established protocols for building sncRNA-sequencing libraries. Also, she/he will be trained on computational pipelines established in the group for the analysis, representation, and interpretation of sncRNA-sequencing experiments. Her/his task will be the analysis of a small dataset.
 

Project 6

Dr. Anastasia Georgiadi

Institute for Diabetes and Cancer

Endocrine Pharmacology

 

Project Description:

Using adipose tissue organoids to study adhesion GPCRs thermogenic signaling

Adipose tissue has a high endogenous capacity for remodeling. Under cold exposure adipose tissue undergoes extreme remodeling, to protect body temperature. Heat producing adipocytes are energy consuming adipocytes and they can appear in different fat depots in mice and humans, upon exposure to cold temperature or in response to cold mimicking hormones. Heat producing adipocytes have been found to provide protection against cardiometabolic diseases and to contribute to lower fat mass maintenance, while ageing. In our lab we focus on the discovery of new populations of these energy consuming adipocytes and hormonal signals that can drive their appearance and activation in the white adipose tissue. Particularly we focus on a so far unexplored family of G-protein couple receptors, the adhesion GPCRs in relation to their ability to drive white adipose tissue remodeling to support thermogenesis. In the project proposed for this summer internship, the intern will investigate the role of a novel adhesion GPCR in white adipose tissue remodeling via its actions on adipocytes, adipocytes precursors and endothelial cells. The intern will work with adipose tissue organoids and will perform assays such as lipolysis, Seahorse oxygen consumption, Western blot to assess GPCR signaling and confocal microscopy.

 

 

Project 7

Dr. Alberto Cebrian Serrano

Institute for Diabetes and Obesity

Genetics Unit

 

Project Description:

Developing a novel inducible CRISPR-mediated activation system for the treatment of metabolic 
disease

Project description
CRISPR/Cas9-associated technologies have revolutionized genome editing. Beyond gene editing, the CRISPR/Cas9 technology offers a versatile sequence-specific gene regulation toolset. A nucleasedeficient Cas9 (dCas9), which does not cleave DNA, has been developed. Transcription activators can be tethered to this dCas9 creating a system known as CRISPR activation (CRISPRa). Under the direction of guide RNAs, CRISPRa can increase the expression of endogenous genes of interest. CRISPRa can also upregulate multiple genes at once through the co-delivery of multiple gRNAs. With the aim of temporally controlling gene expression, we have developed a novel inducible CRISPRa system that becomes active in the presence of the plant hormone abscisic acid (ABA). Unlike most of the inducible drugs that act as cell killers or stressors, ABA exhibits insignificant toxic side effects on animal cells. Moreover, ABA’s solubility allows passage through the blood brain barrier, making it an inducible substance with meaningful clinical applications. We have successfully produced endogenous gene expression in different in vitro cell models and in locus with metabolic relevance. Based on these exciting results we herein want to optimise and refine our ABA inducible CRISPRa system. The implement of this mutation-independent strategy to treat metabolic diseases, significantly lowers the barrier for potential clinical use.

Intern Involvement
Structural and biochemical data suggest that there are several ABA analogs might activate our inducible CRISPRa system more efficiently. As a key objective of this summer internship, we will screen for novel ABA agonist components using our inducible CRISPRa system. As an intern, you will have the opportunity to learn about the CRISPR system (dry lab) and molecular biologic techniques (wet lab). Your main tasks will involve in vitro gene transfection and evaluation of gene activation via luciferase assay, qPCR and Western blot/ELISAs analysis. Finally, the results obtained will be analyzed and interpreted. The intern will be trained by PhD students and senior scientist to achieve these goals.