Chemical probes for lysosomal biology

09 - 10 September 2024 09:00 - 17:00 Hilton York Free
Request an invitation
Fluorescent cells

Theo Murphy meeting organised by Dr Simon Wheeler and Professor Elizabeth New.

Lysosomal function underpins both cellular health and disease. Despite this our understanding of how these organelles work, and how they contribute to important pathologies, is far from complete. The use of chemical sensors to elucidate these mechanisms provides an exciting but largely unexploited opportunity to facilitate fundamental research and ultimately to develop therapies.

The schedule of talks and speaker biographies are available below. Speaker abstracts will be available closer to the meeting date.

Poster session

There will be a poster session on Monday 09 September. If you would like to present a poster, please submit your proposed title, abstract (up to 200 words), author list, and the name of the proposed presenter and institution to the Scientific Programmes team no later than Friday 26 July 2024.

Attending this event

This event is intended for researchers in relevant fields, and is a residential meeting taking place at Hilton York, 1 Tower Street, York, YO1 9WD.

  • Free to attend
  • Advance registration essential (more information about registration will be available soon)
  • This is an in person meeting
  • Catering options are available to purchase during registration. Participants are responsible for their own accommodation booking.

Enquiries: contact the Scientific Programmes team

Organisers

  • Simon Wheeler

    Dr Simon Wheeler, De Montfort University, UK

    Simon Wheeler studied chemistry at the University of Bristol before working in the pharma industry for 14 years as a synthetic chemist. Redundancy prompted him to retrain as a pharmacist at De Montfort University but he quickly found that this profession offered insufficient opportunity for his curiosity. He returned to De Montfort to study for a PhD in cell biology (under the supervision of Dr Dan Sillence) where he examined the cellular mechanisms of lysosomal storage disorders especially Niemann-Pick type C. After a period of post-doctoral work on fluorescent europium-based probes with Dr Steve Butler at Loughborough University he returned to De Montfort University in August 2022 as a lecturer in pharmaceutical chemistry. His research interests are in fragment based drug discovery and crown ether-based fluorescent probes for Ca2+.

  • Elizabeth New

    Professor Elizabeth New, University of Sydney, Australia

    Dr Elizabeth New undertook her undergraduate and Masters studies at the University of Sydney. She completed her PhD studies in 2010 at the University of Durham with Professor David Parker. Liz was then a Royal Commission for the Exhibition of 1851 Research Fellow at the University of California, Berkeley, working with Professor Chris Chang. In 2012, she returned to the University of Sydney, holding a Discovery Early Career Research Fellowship from the Australian Research Council from 2012-2014, and a Westpac Research Fellowship from 2016. Liz’s research interests lie in the development of small molecule sensors for the study of oxidative stress and metal ions in biology.

Schedule

Chair

Frances Platt

Professor Frances Platt FMedSci FRS, University of Oxford, UK

09:00-09:05 Welcome and introduction
Dr Simon Wheeler, De Montfort University, UK

Dr Simon Wheeler, De Montfort University, UK

09:05-09:45 Controlling ion flux across the lysosome through two-pore channels

Much evidence points to lysosomes as non-canonical stores of Ca2+. This extends the role of lysosomes to signalling processes. A number of Ca2+-permeable lysosomal ion channels have been identified and shown to regulate numerous functions including many aspects of membrane trafficking. Malfunction of these channels is linked to a growing number of diseases. In this talk, I will cover recent advances in understanding how two-pore channels (TPCs) control flux of Ca2+ and other ions across the lysosomal membrane. The picture that emerges is an unusual one in which TPCs are able to switch their ion selectivity in an agonist dependent manner to differentially control lysosomal function. I will discuss molecular mechanisms underpinning their activation by natural and newly identified synthetic mimics. And how the local Ca2+ release events that TPCs generate are converted into global Ca2+ signals through cross-talk with other Ca2+ channels on neighbouring canonical Ca2+ stores.

Professor Sandip Patel, UCL, UK

Professor Sandip Patel, UCL, UK

09:45-10:30 Lysosomes at the nexus of cellular homeostasis and cancer

Transmembrane proteins represent ~20-30% of the human proteome and are subjected to turnover by endocytic uptake and delivery to the lysosome, where their degradation poses a unique challenge due to their hydrophobic, phospholipid-embedded a-helical domains that are inaccessible to endopeptidases. Here, we answer the long-standing question of how lysosomes degrade transmembrane proteins: a key missing piece in the life cycle of this important class of proteins that mediate signal transduction, nutrient import, adhesion, and migration. Combining lysosomal proteomics with functional genomic data mining, untargeted metabolomics, and biochemical reconstitution, we find that a previously mischaracterised enzyme that we rename Lysosomal Leucine Aminopeptidase (LyLAP) is most tightly associated with elevated endocytic activity and enables transmembrane protein degradation. LyLAP performs a unique function not found among known lysosomal hydrolases, namely, processive disassembly of hydrophobic a-helices triggered by their N-terminal hydrophobic (often Leucine) residues. Importantly, LyLAP is upregulated in pancreatic ductal adenocarcinoma (PDA), an aggressive cancer that relies on macropinocytosis for nutrient uptake. Strikingly, loss of LyLAP activity has catastrophic consequences for lysosomes, ultimately leading to PDA cell death. Thus, LyLAP enables lysosomal degradation of membrane proteins, and may represent a targetable vulnerability in highly endocytic cancer cells.

Dr Aakriti Jain, University of California, Berkeley, USA

Dr Aakriti Jain, University of California, Berkeley, USA

10:30-11:00 Break
11:00-11:45 Lysosomes as targets for cancer therapy

Being originally discovered as cellular recycling bins, lysosomes are today recognised as versatile signalling organelles that control a wide range of cellular functions that are essential not only for the well-being of normal cells but also for malignant transformation and cancer progression. In addition to their core functions in waste disposal and recycling of macromolecules and energy, lysosomes serve as an indispensable support system for malignant phenotype by promoting cell growth, cytoprotective autophagy, drug resistance, pH homeostasis, invasion, metastasis and genomic integrity. On the other hand, malignant transformation reduces the stability of lysosomal membranes rendering cancer cells sensitive to lysosome-dependent cell death. Notably, many clinically approved cationic amphiphilic drugs widely used for the treatment of other diseases accumulate in lysosomes, interfere with their cancer-promoting and cancer-supporting functions, and destabilise their membranes thereby opening intriguing possibilities for cancer therapy. Here, I will discuss the emerging evidence that supports the supplementation of current cancer therapies with lysosome-targeting cationic amphiphilic drugs and the molecular mechanisms by which they disturb lysosomal function and permeabilise lysosomal membranes.

Funding: Danish Cancer Society, Danish National Research Foundation, European Research Council, and Novo Nordisk Foundation.

Dr Marja Jäättelä, Danish Cancer Institute, Denmark

Dr Marja Jäättelä, Danish Cancer Institute, Denmark

11:45-12:30 GBA1 and parkinsonism: new tools for new times

To explore the pathogenesis of GBA1-associated synucleinopathies, we genetically engineered GBA1 in an iPSC line derived from a patient with both GD (GBA1: N370S/N370S) and PD using CRISPR/Cas9 and piggyBac systems and generated a panel of isogenic lines (GBA1: KO/KO, and WT/WT) to model the impact of enzyme and lipid substrate levels.

DANs differentiated from these isogenic lines demonstrated descending levels of GCase protein: the WT/WT line exhibited the highest, followed by the N370S/N370S, whereas the KO/KO line showed only a trace. Glycosylation analysis demonstrated endoplasmic reticulum (ER) retention of a modest fraction of GCase N370S. Immunofluorescence with the sensitive GCase antibody hGCase-1/23 revealed targeting of GCase N370S to lysosomes. Both in vitro GCase activity assay using the fluorogenic substrate 4MU and live-cell GCase assay with LysoFQ-GBA demonstrated reduced GCase activity in the N370S/N370S and no activity in the KO/KO line. Surprisingly, lipid analysis using supercritical fluid chromatography-mass spectrometry revealed substantial glucosylceramide and glucosylsphingosine accumulation only in the KO/KO line, despite reduced GCase activity in the N370S/N370S.

Proteomic profiling of lysosomes purified via the LysoIP approach revealed that the KO/KO lysosomes had substantial lipid accumulation and reduced levels of Cathepsin F. Ongoing experiments aim to further elucidate the mechanistic implications of these findings.

Yu Chen, National Human Genome Research Institute, USA

Yu Chen, National Human Genome Research Institute, USA

Chair

Elizabeth New

Professor Elizabeth New, University of Sydney, Australia

13:30-14:15 Photo-activatable and clickable lipid analogs as tools to investigate the transport of lysosomal sphingosine

Sphingolipids are major eukaryotic lipids defined by a common sphingoid backbone. They are particularly enriched at the plasma membrane and they undergo constitutive turnover along the endocytic pathway. The bulk of sphingolipid recycling takes place in late endosomes and lysosomes. Here, catabolism of complex sphingolipids produces sphingosine and long chain sphingoid bases, which are then exported to the ER for synthesis of new sphingolipids. This process, called sphingosine recycling, is critical for cellular lipid homeostasis, yet its mechanisms are largely unknown. To address this, we utilised a recently developed functionalised sphingosine probe. The probe pre-localises into the lysosome and is activated by light. Its other functionalities allow the detection of interaction partners as well as following its subcellular distribution and metabolic conversions. Using this tool, we investigated the role of the sterol transporter STARD3 in sphingosine recycling. Overexpression of STARD3 in cells increased the probe’s metabolic conversion into higher sphingolipids, whereas STARD3 depletion led to delayed export from the lysosome. In vitro approaches showed that the lipid binding domain of STARD3 can bind sphingosine, supporting the hypothesis that STARD3 is a sphingosine transporter operating at the lysosome – ER contact sites. This work paves the way for further research on protein mediated sphingosine transport and on mechanisms that regulate the recycling of cellular sphingolipids. 

Dr Denisa Jamecna, University of Osnabrück, Germany

Dr Denisa Jamecna, University of Osnabrück, Germany

14:15-15:00 Visualisation of endosome/ lysosomal labile iron by ferrous ion-selective fluorescent probes

Iron is the most abundant transition metal in the human body and plays numerous essential roles. The homeostasis of iron is tightly regulated, as both deficiency and overload can lead to various dysfunctions. However, much of intracellular iron homeostasis and trafficking remains poorly understood, largely due to the lack of effective fluorescent probes for detecting the labile iron pool. Lysosomes and endosomes are key organelles in intracellular iron trafficking, as they are involved in endocytic iron uptake, storage, and subcellular iron recycling processes. We have developed a series of ferrous ion-selective fluorescent probes using N-oxide chemistry, some of which have been applied to study iron status in endosomes and lysosomes under both physiological and pathological conditions. Among these, a lysosome-specific fluorescent probe, HMRhoNox, was designed and employed to monitor transferrin-mediated iron uptake and the lysosomal accumulation of ferrous ions during ferroptosis. Additionally, we developed a plasma membrane-anchoring fluorescent probe to enable real-time imaging of iron uptake. This presentation will detail the development of these probes, their imaging capabilities, and recent findings from their application.

Dr Takusu Hirayama, Gifu Pharmaceutical University, Japan

Dr Takusu Hirayama, Gifu Pharmaceutical University, Japan

15:00-15:30 Break
15:30-16:15 Organelle-targeted sensors and multimodal probes for cellular imaging

Understanding the biochemical composition of cells and organelles is crucial for comprehending physiological and pathological processes. This requires tools that can identify and monitor the chemistry of organelles within cells. We are interested in developing fluorogenic labels, and responsive fluorescent sensors for cellular redox state and redox-active metal ions. To this end, we are developing lysosome-targeted redox sensors and investigating more robust lysosomal-targeting groups.

Multimodal imaging, which integrates the benefits of two or more imaging techniques, is becoming increasingly popular. Conventional probes are typically limited to a single analyte; by combining multiple imaging modalities, more comprehensive information can be obtained from a single sample. Developing multimodal probes compatible with several imaging techniques is therefore highly desirable. We have prepared a set of lysosomally-targeted multimodal imaging agents that can be used for fluorescence microscopy and additional modalities, including phosphorescence, Raman, and X-ray fluorescence microscopies.

Dr Liam Adair, The University of Sydney, Australia

Dr Liam Adair, The University of Sydney, Australia

16:15-17:00 Towards fluorescent sensors for lysosomal Ca 2+ and K+
Dr Simon Wheeler, De Montfort University, UK

Dr Simon Wheeler, De Montfort University, UK

Chair

Simon Wheeler

Dr Simon Wheeler, De Montfort University, UK

09:00-09:45 A genetically encoded lysosomal pH sensor for in vivo applications in aging and neurodegenerative disease

A highly acidic pH is required for the optimal functioning of lysosomal enzymes. Loss of lysosomal intraluminal acidity can disrupt cellular protein and lipid homeostasis and is linked to age-related diseases such as neurodegeneration. To understand how lysosomal pH changes with age and disease, we have developed a robust new lysosomal pH biosensor that we call FIRE-pHLy for Fluorescent Indicator REporting on pH of the Lysosome. Using FIRE-pHLy, we have performed cell-based high-throughput fluorescence screening assays for modifiers of lysosomal pH in undifferentiated and differentiated SH-SY5Y neuroblastoma cells. We have also performed a whole genome screen for genetic modifiers of lysosomal pH in iPSC-derived neurons (induced neurons or iNeurons.) Finally, we have expressed FIRE-pHLy in C. elegans and M. musculus. In C. elegans, lysosomal pH changes during development and aging, and manipulation of lysosomal pH can directly impact lifespan. Thus, lysosomal pH serves as both a biomarker and therapeutic target in aging and neurodegeneration.

Dr Aimee Kao, University of California San Francisco, USA

Dr Aimee Kao, University of California San Francisco, USA

09:45-10:30 Interrogation of lysosome biology by pH sensitive nanoparticles

Engineered and tailor-made nanomaterials (NM) are of increasing relevance for current and future developments in the life and material sciences for applications, eg as drug carriers, fluorescent sensors, and multimodal labels in bioanalytical assays, and reporters for imaging applications. For instance, NM-based reporters and sensors, that are labelled or stained with a multitude of conventional or sensor dyes, have several advantages as compared to molecular probes like enhanced brightness, ie amplified signals, ease of designing ratiometric systems by combining analyte-sensitive and inert reference dyes, and increased photostability. Moreover, this can enable the use of hydrophobic dyes in aqueous environments. For rational NM design, choosing and tailoring the intrinsic physicochemical properties, such as particle size, size distribution, morphology, and surface chemistry of the NM application-specific considerations like biocompatibility, ease and low cost of preparation, and colloidal stability and performance in the targeted environment must be considered. In this lecture, different design concepts of inorganic, organic, and hybrid NM and microparticles with hydrophilic surface chemistries and different functionalities are presented that can be used for the targeting of lysosomes; and to monitor functional parameters of endo-lysosomal compartments, like pH or enable oxygen sensing.

Dr Isabella Tavernaro, Federal Institute for Materials Research and Testing, Germany

Dr Isabella Tavernaro, Federal Institute for Materials Research and Testing, Germany

10:30-11:00 Break
11:00-11:45 Progress and challenges for protein-based probes of metal ions

An ever-growing selection of high performance fluorescent protein-based biosensors is revolutionizing our ability to visualize metal ion concentrations in cells. In this seminar I will describe our most recent efforts to use protein engineering to make a new generation of genetically encoded biosensors, and chemigenetic biosensors augmented with synthetic molecules, for calcium ion, potassium ion, and sodium ion. Key to this effort is our reliance on the use of directed protein evolution to iteratively and reliably improve the properties of biosensors. By creating biosensors with different colours, specificities, and affinities, we are opening up new opportunities for researchers to use multiplexed imaging to investigate dynamics changes in metal ion concentrations in organelles, cells, and whole tissues in model organisms.

Professor Robert E Campbell, The University of Tokyo, Japan

Professor Robert E Campbell, The University of Tokyo, Japan

11:45-12:30 Understanding the role of solute carriers in lysosomal homeostasis and signalling

Solute carriers (SLCs) play key roles in lysosomal homeostasis and intracellular signalling as essential regulators of metabolite and ion exchange with the cytoplasm. Recent advances in structural biology have revealed an unexpected evolutionary links between SLCs and intracellular receptors, opening a new and exciting time in transporter biology. In this talk I will discuss our recent results in understanding the concepts and mechanisms through which SLC proteins can both transport and signal across membranes and the unique role of lysosomal lipids in regulating SLC function.

Professor Simon Newstead, University of Oxford, UK

Professor Simon Newstead, University of Oxford, UK

Chair

Aimee Kao

Dr Aimee Kao, University of California San Francisco, USA

13:30-14:15 Super-resolution imaging of sub-cellular dynamics of endolysosomes

As a process of cellular uptake, endocytosis, with gradient acidity in different endocytic vesicles, is vital for the homeostasis of intracellular nutrients and other functions. To study the dynamics of endolysosomes, we developed pH-sensitive molecular probes imaging the sub-cellular dynamics of endocytic vesicles using a super-resolution fluorescence technique, structured illumination microscopy (SIM). Firstly, a membrane-anchored pH probe, ECGreen was synthesised to visualise endocytic vesicles under SIM. Being sensitive to acidity with increasing fluorescence at low pH, ECGreen, can differentiate early and late endosomes as well as endolysosomes. Meanwhile, membrane-anchoring not only improves the durability of ECGreen, but also provides an excellent anti-photobleaching property for long-time imaging with SIM, which makes ECGreen a good probe for elucidating the interaction between mitochondria and endocytic vesicles in autophagy. Based on ECGreen, a multi-dimensional model containing spatial, temporal, and pH information was successfully developed to analyse endocytic dynamics for its unclear roles in cellular functions. Secondly, by taking advantages of intramolecular charge transfer and a pH-sensitive pyridine unit, we developed a cationic quinolinium-based fluorescent probe, PyQPMe, which exhibits excellent ratiometric fluorescent response to endolysosomes at different stage of interest. By applying PyQPMe as a small molecular probe, we were able to reveal a constant conversion rate from early endosome to late endosome during autophagy using the super-resolution imaging.  

Dr Jiajie Diao, University of Cincinnati College of Medicine, USA

Dr Jiajie Diao, University of Cincinnati College of Medicine, USA

14:15-15:00 Morpho-functional characterisation of lysosomes by correlative light and electron microscopy

Cells reprogram and adapt their endo-lysosomal systems to their metabolic requirements in physiology and pathology. Yet, molecular and ultrastructural adaptations of endo-lysosomal organelles to metabolic stimuli at nanoscale, and how this is exploited particularly by cancer cells remain notoriously obscure. Addressing this, we have developed endo-lysosomal biology tailored correlative microscopy approaches to combine molecular specificity and live-cell capabilities of fluorescence microscopy (FM) with high-resolution ultrastructure of electron microscopy (EM). We label and visualise endo-lysosomal organelles in (live) cells with fluorescent biosensors after which they are processed for EM and imaged using a (volume) EM technique. We correlate hundreds of fluorescently labelled organelles enabling quantitative analysis of the molecular-functional-ultrastructural data within a single dataset.

We have employed our methods to map the ultrastructural distribution of endo-lysosomal proteins; to link single organelle dynamics to their 3D ultrastructure; to image transient contact sites between the endoplasmic reticulum and lysosomes; and to monitor functional parameters of endo-lysosomal compartments, ie enzyme activities, pH, and calcium content. The presented CLEM workflows are compatible with a large repertoire of probes and can integrate information on protein localisation, functional status, dynamics and 3D ultrastructure, all in a single sample and at the level of single organelles.

Dr Nalan Liv, University Medical Center Utrecht, The Netherlands

Dr Nalan Liv, University Medical Center Utrecht, The Netherlands

15:00-15:30 Break
15:30-16:15 New sub-cellular chemical imaging techniques with potential to probe lysosomal biology

Vibrational spectroscopy has found many applications in the biological sciences. Its capabilities include direct, label-free and in situ characterisation of protein secondary structures (including protein aggregates), lipid profiling, and investigation of metabolism (eg lactate, glucose, glycogen, organic phosphates). Unfortunately, a common form of vibrational spectroscopy, infrared spectroscopy, suffers from relatively poor spatial resolution due to the longer wavelengths of infrared light (compared to bright field or fluorescence microscopy). Other modalities of vibrational spectroscopy, such as Raman spectroscopy offer improved diffraction-limited spatial resolution, but can suffer substantial limitations from cell and tissue autofluorescence. Substantial efforts have been undertaken for both infrared and Raman spectroscopy to develop improved instrumentation, light sources, and imaging protocols to enable direct sub-cellular chemical imaging with these two techniques. In my talk I will present the recent developments in the field of vibrational spectroscopy, along with approaches to integrate these techniques in multi-modal or correlative workflows, which could open new possibilities to probe the biology of lysosomes and other organelles. 

Dr Mark Hackett, Curtin University, Australia

Dr Mark Hackett, Curtin University, Australia

16:15-16:55 Rational design of fluorescent nano probes for long term tracking and super-resolution imaging of lysosomal dynamics

Lysosomes are membrane-enclosed small spherical cytoplasmic organelles. Malfunctioning and abnormalities in lysosomes can cause a plethora of neurodegenerative diseases. Consequently, understanding the structural information on lysosomes down to a subnanometer level is essential. Recently, super-resolution imaging enables us to visualise dynamical processes in living cells down to subnanometer accuracy by breaking the diffraction limit. Herein, the advancement of various fluorescent nanoprobes that have been utilised in our lab recently for super-resolution imaging of lysosomal dynamics are presented. Dr Nandi will discuss the detail single molecule level photophysical of coinage metal nanoclusters for their potential use in the super-resolution imaging of lysosomes. He will also highlight the rational design of fluorescent superparamagnetic iron oxide nanoparticles (SPIONs) to enhance their photon budget as well as the magnetic resonance imaging contrast. The SPIONs were highly specific in staining and imaging the lysosomes of HeLa cells with high contrast.

Dr Chayan Nandi, Indian Institute of Technology Mandi, India

Dr Chayan Nandi, Indian Institute of Technology Mandi, India

16:55-17:00 Closing remarks