Courses > Semester 8 - Elective courses

Elective courses of Semester 6 plus the following courses:

Intercellular Communication

Theory

Courses combine lectures (26 hours) and analysis of recent original articles (13 hours).

Training seminars

Students are invited to present two 30min seminars on specific topics.

Syllabus

  1. Introduction to Intercellular Communication
    a. Mechanisms and various types of intercellular communication
    b. Types of receptors
    c. Basics of signal transduction
  2. Organization of signaling
    a. Introduction to main signaling pathways
    b. Signaling complexes
    c. Crosstalk between signaling pathways
    d. Signaling networks
    e. Positive and negative feedback in signaling
  3. Physiological role of mediators
    a. Hormones
    b. Cytokines
    c. Growth factors
    d. Neurotransmitters
  4. 2nd messengers, activation pathways, mechanisms of activation, role in signal processing.
  5. Small G proteins, mitogen-activated protein kinases pathways (MAPK)
  6. Signaling by G-protein-coupled receptors (GPCR), heterotrimeric G-proteins
  7. Signaling by receptor tyrosine kinases (RTKs)
  8. Receptors with associated tyrosine kinase activity
    a. The Jak-STAT pathway
    b. Protein tyrosine phosphatases as negative regulators
    c. SOCS (Suppressors of cytokine signaling)
  9. Receptors with intrinsic Ser/Thr kinase activity
    a. TGF-β receptors
    b. SMADS
  10. Signaling by nuclear receptors
    a. Regulation of transcription by nuclear receptors
    b. Nongenomic functions of nuclear receptors
  11. Receptor endocytosis
  12. Regulation of gene expression by transcription factors

Textbooks

  • Biochemistry of signal transduction and regulation, G. Krauss, Wiley-VCH, 2008.
  • Structure and function in cell signaling. J. Nelson, John Willey & Sons, 2007

Genetic Engineering

Structure and use of molecular cloning vectors: lamda-based vectors, other vectors of viral origin (M13), plasmid vectors, cosmids, phagemids, Yeast Artificial Chromosomes, Bacterial Artificial Chromosomes, Phage P1-based vectors.
Methods in Genetic Engineering: In vitro site-directed mutagenesis, in vivo cloning (phage Mu), Gene replacement, Phage display. Gene fusions, Reporter genes.
Characteristics and applications of cloning vectors in Genetic Engineering and Molecular Genetics.
Examples of applications of Genetic Engineering in Biotechnology and Public Health:
In Bacteria: Heterologous gene overexpression. (Vectors, methods). Other biotechnological applications: production of insulin and somatostatin.
In plants: Methods for constructing transgenic plants (Ballistic, Agrobacterium tumefaciens-mediated transformation). Pathogen-derived resistant plants. Transgenic plants resistant to insects (cry-trangenes) and to herbicides.
Transgenic animals.
Health related applications: Construction of recombinant vaccines. DNA vaccines. Gene therapy.

Research Methods in Genetic Engineering

1. A New Toolbox for Recombinant DNA

Methods for rapid and accurate assessment of gene regulation. Useful sequences can be appended to DNA molecules and specific mutations can be generated by synthetic oligonucleotides. Recombination technologies allow rapid exchange of DNA fragments. Advanced systems for the conditional induction of gene expression. Precise genetic alterations using specific recombinases and mechanisms of homologous recombination. Transfer of embryonic stem cell lines into embryos to produce chimeric mice with germline transmission.

2. Genetic Interference by the Use of Appropriate Genetic Elements and the Employment of Basic Mechanisms of Gene Regulation

Te use of transposons as genetic tools for mutagenesis and transgenesis in organisms representing established genetic models. A transposon is resurrected for mutation experiments in mammalian cells. Employment of the RNAi machinery for the knock-down of gene expression in many different organisms. Selective modulation of gene function by miRNAs.

3. From Genome Sequence to Gene Functions

mRNA profiling with microarrays reveals new relationships beween cellular pathways. Chromatin immunoprecipitation and other genome-wide methods can be used to assay modifications in the structure of chromatin in living cells. Determine the locations of proteins in cells and tissues. Arrayed antibodies are used to measure protein levels in cells.

4. The Contribution of Genetic Engineering in Understanding the Genetic Basis of Diseases

Recombinant DNA techniques allow the identification of genes that are responsible for human diseases. The contribution of genetic engineering for targeting growth factor receptors in cancer cells. Microarrays and new technologies offer large scale sequence-based diagnosis. Comparative analysis of mouse models and genomic analysis lead to the discovery of new oncogenes.

5. DNA Fingerprinting

Hypervariable or variable tandem repeat loci can be used to identify genetically associated individuals. Short tandem repeats become the standard for forensic applications. Mitochondrial DNA profiling. Multiplex PCR amplification and fluorescent tags are used to analyze the profile of tandem repeats.

Cancer Biology

The purpose of the course is to present the current knowledge regarding the physiology, the genetics and the biochemistry of cancer cells and the underlying mechanisms leading to carcinogenesis. In addition the course focuses upon:

  • Current methodological approaches applied in cancer studies
  • The translation of the knowledge attained from cancer research into diagnostic methods and therapeutic regimes.

Lecture contents

  • Introduction: The nature of cancer cell
  • The maintenance of genomic integrity and carcinogenesis
  • Cell proliferation and tumorigenesis
  • Apoptosis
  • Cytoplasmic signaling circuits
  • Oncogenes
  • Tumor suppressor genes
  • The biology of angiogenesis
  • Metastasis
  • Current therapeutic approaches

The course consists of three hours lecture 1 hour presentation and discussion of current bibliography by the students, weekly. Attendance is compulsory

Text books

  • The Biology of Cancer Kitraki E Trougkos K (authors) Paschalidis Medical Publications 2006ISBN: 960-399-404-9
  • The Biology of Cancer Robert A Weinberg (author) ISBN-10: 0815340788 Garland Science 09/06/2006

Molecular Neurobiology

1. Induction and Patterning of the Nervous System

Inductive signals control neural cell differentiation. Neural induction involves inhibition of BMP signals. Distinct morphogenetic proteins are shaping the neural plate along its dorsoventral axis. The rostrocaudal axis of the neural tube is patterned in several stages. The actions of homeotic proteins.

2. Generation and Survival of Nerve Cells

The molecular basis of neurogenesis. The role of pro-neural genes. Secreted factors direct the differentiation of neural crest cells into neurons and glia. Neuronal fate in the mammalian cortex is influenced by the timing of cell differentiation. The phenotype of a neuron is controlled by signals emanating from the neuronal target. Control of neuronal survival by neurotrophic factors. The multifaceted role of neurotrophins. Signal transduction by the neurotophic factor receptors.

3. Guidance of Axons to their Targets

Specific molecular cues guide the axons to their targets. The extracellular milieu provides a complex set of commands to the developing axon. Growth cones, integrins, netrins, ephrins, semapahorins. Molecules of distinct protein families interact to guide axons to their destinations.

4. Formation of Synapses and the Fine-tuning of Synaptic Connections

Dynamic interactions of neuronal cells with their targets. The role of neurotrophic factors. Synaptic regression. The recognition of synaptic targets is highly specific. Development of neural circuits and postnatal neuronal activity. Sychronous presynaptic activity enhances the release of neurotrophic factors from their target neurons. Neuronal competition and refinement of synaptic connections.

5. Regeneration of the Nervous System

Regenerative capacity of the nervous system. New neural connections can reform following nerve injury. Axonal regeneration and functional restoration. Biology of the neural stem cell. Functional replacement of neuronal cells. Molecular mechanisms of aging. Alzheimer’s Dementia.

6. Cellular and Molecular Mechanisms of Learning and Memory:

Implicit and explicit memory. Elementary forms of learning: habituation, sensitization, and classical conditioning. Genetic analyses of long-term memory storage in Drosophila. The cAMP-PKA-CREB pathway. Long-term storage of explicit memory in mammals. Genetic interference with long-term potentiation is reflected in the properties of place cells in the hippocampus. Learning and changes in the somatotopic map of the brain.

Bioprocess engineering

Field Ecology

Environmental Sciences