Autophagic flux regulation by the cholesterol/H+ antiporter PTCH1

Lead Research Organisation: University of Leeds
Department Name: Inst of Molecular & Cellular Biology

Abstract

Development of a fertilized egg into a properly formed embryo requires a number of signals that perfectly orchestrate the formation of tissues and organs. One of those important signalling events is the so-called "Hedgehog" pathway that when absent provokes very serious congenital defects incompatible with life. Conversely, excessive activity of the Hedgehog pathway after birth is a common cause of childhood brain tumours and adult brain, skin, lung, prostate, and gut cancers. My group has contributed to the understanding of how the Hedgehog signals are perceived by cells and transmitted into different outcomes, depending on the tissue type and the context. Here, we propose to investigate a novel function of the Hedgehog pathway in regulating autophagy, the "self-eating" behaviour of cells. We found that the Hedgehog receptor PTCH1, a tumour suppressor, inhibits autophagy. Cells normally feed themselves from available nutrients; however, in conditions of starvation like those encountered by tumour cells that grow faster that the vasculature that nourishes them, cells degrade part of their contents to survive. Autophagy is essential for cancer cells survival. We will investigate how the Hedgehog pathway regulates autophagy at the molecular level. We will build onto our recent publication that shows that interaction between the C-terminal domain of PTCH1 and ATG101 is necessary for inhibition of autophagy, characterised by reduced number of acidic vesicles where degradation occurs. We propose that PTCH1 acts as a transporter, dissipating the proton gradient of those vesicles (autolysosomes) by a mechanism coupled to cholesterol transport. We will test this hypothesis and will investigate if this function of PTCH1 is lost by mutations of the C-terminal domain frequently found in cancer. We hope that our findings will increase the knowledge on new targets for cancer therapy and will reveal new ways in which cells adapt and survive to different stressors.

Technical Summary

The Hedgehog signalling pathway is a master regulator of growth and differentiation. Binding of any of three Hh isoforms to their receptor Patched1 (PTCH1) activates both canonical and non-canonical signals. PTCH1 is a 12-transmembrane protein with homology to bacterial RND permeates and to some cholesterol transporters. Recent the cryo-EM structures of PTCH1 revealed that it contains a tunnel with aligned cholesterol-like densities, which together with functional studies support the notion that PTCH1 is a cholesterol transporter. We have recently reported that PTCH1 interaction with ATG101 through its C-terminal domain (CTD) reduces autophagic flux and acidification of autolysosomes. This is a novel non-canonical function of the tumour suppressor PTCH1, since autophagy is essential for detoxification of protein aggregates and damaged organelles and for survival of cancer cells under conditions of scarce nutrient availability. Here, we aim to define the mechanism by which PTCH1 inhibits autophagic flux. We propose that interaction with ATG101 is required to localise PTCH1 to the autophagome/autolysosome membrane, where it causes alkalisation of the autolysosomal lumen by means of its cholesterol/H+ antiporter activity. We will determine if PTCH1 impairs autolysosome acidification by its RND domain and if cholesterol transport is necessary for inhibition of the autophagic flux by PTCH1. We also investigate if the Niemann-Pick C1 (NPC1)/NPC2 cholesterol handling system is necessary to stimulate PTCH1's translocation of cholesterol inside autolysosomes and dissipation of the proton gradient. We have also identified three frequent mutations in the CTD of PTCH1 in some epithelial cancers and will test if these mutations abolish the interaction of PTCH1 with ATG101 and, therefore, cannot inhibit autophagy, giving a growth advantage to cancer cells.

Planned Impact

This research project will generate fundamental knowledge about the tumour suppressor PTCH1 that will be useful to both increase its anti-autophagic activity (in cancer) and decrease it (in regenerative medicine, tissue engineering). Because of the tumour suppressor role of PTCH1, applications that mimic its function and increase its stability will be useful in the oncology field. Conversely, strategies that reduce PTCH1 activity will be of value for applications in stem cell-based therapies for neurodegenerative diseases, regenerative medicine (spinal cord injury), and skin replacement tissue engineering, which will benefit of transient controlled activation of canonical Hedgehog signalling.
The outcomes of our research will directly benefit the commercial private sector, since it will offer a platform for analysis, validation, and development of small inhibitors and tools for research of commercial value. We are interested in working with Avacta, a mid-size biotechnology company based in the UK (Wheterby). We will pursue a partnership, a formal collaboration to test lead inhibitors in canonical and non-canonical Hedgehog signalling at the University of Leeds with a small budget for pilot experiments and exchange of personnel for short periods of 2-3 months. At the end of the grant life, we will pursue an application for a CASE studentship with the industrial partner.
Avacta has strong links with the University of Leeds and the Astbury Biostructure Laboratory to screen and refine affimers (adhirons) that target proteins with affinities equivalent to antibodies, but which are cheap to generate in large quantities in bacteria. We will seek to identify adhirons that compete for binding of some regulators of PTCH1 that will serve for many purposes: 1) as research tools to investigate the effect of blocking single protein-protein interactions in the academic setting (academic beneficiaries), 2) as potential diagnostic tools to investigate the presence of common truncation in PTCH1 in cancer (leading toward personalised medicine), and 3) as the basis for modifications to increase their cell permeability, including specific targeting (therapeutics). This avenue of impact will increase research tools and therapeutic development, enrich the already fruitful collaboration between Avacta and many investigators at the University of Leeds, increase PDRAs training in a highly competitive technology and increase PDRAs employment attractiveness.
Dr. Riobo-Del Galdo will further reach to public beneficiaries through ideation and delivery of lectures targeted to high school student audience (KS4). She has established connections with a group of Science teachers at Allerton High School in Leeds, Mr. Jonathan Allcock and Dr. Kirsty Bryant, and has already been a guest lecturer at the school. Dr. Riobo-Del Galdo will undertake a training session of People Like Me, which disarms stereotypes in STEM careers for girls aged 11-14 to deliver an activity following the lectures at the school to inspire girls to opt in STEM-related A-level subjects. She will also continue her participation in University Open Days to educate prospective Sixth Form students and their parents in the development of impact that starts from basic cellular and structural biology research.
 
Description While this award is 3 year and 3 months in, our timeline was severely impacted by the COVID pandemic, resulting in a temporary change of focus from mechanistic investigation of how the tumour suppressor PTCH1 suppresses autophagy to investigating what happens when PTCH1 cannot exert this function. For this, we pushed forward a new collaboration with the University of Rome, from which we had generated cells carrying a mutation in PTCH1 described in colon cancer, which we predicted to abolish its function on autophagy. We found that the PTCH1 mutations prevented its interaction with the ATG101 protein, impaired suppression of autophagy and increased cell division and survival (2 publications, and another article, describing the oncogenic effects of the loss of this regulatory domain of PTCH1, independent of canonical HH signalling is also in preparation). Comparison of the genes expressed in those mutant cells with normal cells was done by bioinformatic analysis of RNA-seq- which we could carry out remotely. We found a profound change in many signalling pathways, in addition to those regulating autophagy: metabolic pathways in cancer, MAPK signalling, and ribosomal proteins, which will be further investigated in future projects for which we are submitting applications.
After the labs re-opened, we returned to the proposed project plan. We successfully generated PTCH1, NPC1 and NPC2-deficient HEK293 and HeLa cells using CRISPR/Cas9, which have been now fully characterised by western blot and fillipin staining. We now confirmed the hypothesis that the capacity of PTCH1 to block autophagy completion requires an intact NPC1/NPC2 cholesterol shuffling system. Furthermore, we have discovered that PTCH1 overexpression induces cholesterol accumulation on lysosomes, suggesting that it acts as a cholesterol pump driving cholesterol from the cytosolic side of the lysosomal membrane to the lumen, opposite of the direction of transport of NPC1. This remarkable finding also supports our hypothesis that cyclic shuttling of cholesterol between PTCH1 and NPC1 drives the increase in luminal pH and the impairment of autophagic degradation. We are currently performing experiments of lysosomal pH changes and assessing a PTHC1 mutant with an inactive cation gate.
Another collaboration studying cholesterol transport in PTCH-like proteins resulted in a recent publication.
Exploitation Route The main findings of this project (in progress):
1- we identified and confirmed the existence of somatic mutations in the terminal domain of PTCH1 in ~ 20% of right side colon cancer
2- PTCH1 CTD mutations do not perturb canonical signalling but increase autophagic flux, MAPK signalling and induce metabolic adaptations that confer a proliferative advantage of the cells carrying the mutation.
3- Indeed, we confirmed that around 5% of cancers of the colon (exclusively right-sided) and 8% of endometrial cancers carry these mutations, making autophagy an attractive process to target in a group of patients.
4- We have generated isogenic normal and transformed epithelial cell lines with specific knockout of NPC1, NPC2 and PTCH1. These cells have been instrumental in confirming our hypothesis that PTCH1 regulates cholesterol transport in the lysosomes and that this can mediate its inhibitory effect on autophagic cargo degradation.
5- We have generated isogenic colon cancer cell lines with a truncation of the PTCH1 C-tail at S1223. Those cells are more aggressive and have a proliferative advantage. We are using that model to investigate the potential sensitivities of PTCH1-mutant colon cancers to current treatments and using them as a model for antibody validation for an industry interested party. These studies are beyond the scope of this grant and have taken off using other sources of funding, but were possible by some key findings of this award.
Sectors Healthcare,Pharmaceuticals and Medical Biotechnology

 
Description Lattice Light Sheet Microscopy for the Biosciences
Amount £651,012 (GBP)
Funding ID BB/V01904X/1 
Organisation Biotechnology and Biological Sciences Research Council (BBSRC) 
Sector Public
Country United Kingdom
Start 05/2021 
End 05/2022
 
Title HCT116-PTCH1 CTD mut 
Description HCT116 colon cancer cells engineered to harbour a truncation in the CTD of PTCH1 at S1223 by CRISPR/Cas9. 
Type Of Material Cell line 
Year Produced 2020 
Provided To Others? No  
Impact Phenotypic characterisation of these cells will be published in one of the manuscripts in preparation. They exhibit enhanced autophagic flux, but no growth advantage , which we attributed to constitutive AKT signalling due to an PI3KCA activating mutation. 
 
Title HEK293-NPC1KO 
Description HEK293 cells deficient in NPC1 expression by introduction of a frameshift mutation by CRISPR/Cas9. Cell phenotype is validated by sequencing, western blot and accumulation of cholesterol in lysosomes (filipin staining). 
Type Of Material Cell line 
Year Produced 2021 
Provided To Others? No  
Impact The cell lines will be shared to any investigator upon publication of our findings. 
 
Title HEK293-NPC2KO 
Description HEK293 cells deficient in NPC2 expression by introduction of a frameshift mutation by CRISPR/Cas9. Cell phenotype is validated by sequencing, western blot and accumulation of cholesterol in lysosomes (filipin staining). 
Type Of Material Cell line 
Year Produced 2021 
Provided To Others? No  
Impact Cells will be available to share upon publication. 
 
Title HeLa-NPC1KO 
Description HeLa cells deficient in NPC1 expression by introduction of a frameshift mutation by CRISPR/Cas9. Cell phenotype is validated by sequencing, western blot and accumulation of cholesterol in lysosomes (filipin staining). 
Type Of Material Cell line 
Year Produced 2021 
Provided To Others? No  
Impact Cells will be available upon publication. 
 
Title HeLa-NPC2KO 
Description HeLa cells deficient in NPC2 expression by introduction of a frameshift mutation by CRISPR/Cas9. Cell phenotype is validated by sequencing, western blot and accumulation of cholesterol in lysosomes (filipin staining). 
Type Of Material Cell line 
Year Produced 2021 
Provided To Others? No  
Impact Cells will be shared upon publication. 
 
Title SW620-PTCH1 CTD mut 
Description SW620 cells (colon cancer cells) engineered to carry a truncation at S1223 of PTCH1 CTD. 
Type Of Material Cell line 
Year Produced 2022 
Provided To Others? Yes  
Impact The mutant cancer cells exhibit a very aggressive phenotype, including increased autophagic flux, as predicted in the current grant proposal. The mutations results in increased proliferation, migration, ERK and AKT signalling, autophagy and tumour formation in animals. The cells are central to 2 publicaitons in preparation, and have been used in a collaboration (output listed). 
 
Description BBSRC -IPA 
Organisation Ruder Boskovic Institute
Country Croatia 
Sector Public 
PI Contribution Starting collaboration through an International Partnering Award - officially beginning in April 2022
Collaborator Contribution Partners will contribute to development of a Hedgehog signalling website resource and provide genetically modified human-derived cancer cell lines, advanced training in mass spec of GLI proteins PTMs, and transgenic mice.
Impact First Hh signalling workshop organised in January 2023 at the University of Leeds. 20 delegates attended and presented talks in a 2-day networking event.
Start Year 2022
 
Description BBSRC -IPA 
Organisation Sapienza University of Rome
Country Italy 
Sector Academic/University 
PI Contribution Starting collaboration through an International Partnering Award - officially beginning in April 2022
Collaborator Contribution Partners will contribute to development of a Hedgehog signalling website resource and provide genetically modified human-derived cancer cell lines, advanced training in mass spec of GLI proteins PTMs, and transgenic mice.
Impact First Hh signalling workshop organised in January 2023 at the University of Leeds. 20 delegates attended and presented talks in a 2-day networking event.
Start Year 2022
 
Description BBSRC -IPA 
Organisation University of Warsaw
Country Poland 
Sector Academic/University 
PI Contribution Starting collaboration through an International Partnering Award - officially beginning in April 2022
Collaborator Contribution Partners will contribute to development of a Hedgehog signalling website resource and provide genetically modified human-derived cancer cell lines, advanced training in mass spec of GLI proteins PTMs, and transgenic mice.
Impact First Hh signalling workshop organised in January 2023 at the University of Leeds. 20 delegates attended and presented talks in a 2-day networking event.
Start Year 2022
 
Description Collaboration with Prof. Gianluca Canettieri 
Organisation Sapienza University of Rome
Country Italy 
Sector Academic/University 
PI Contribution Our study of PTCH1 in regulation of autophagy generated an interest of studying its role in cancer development. This was formalised by joint supervision of a PGR student Ms Begoña Caballero Ruiz, funded by the University of Rome. The PGR has already generated a large amount of data from colon cancer cells engineered to carry a truncation in the C-terminal domain of PTCH1, which is necessary for its regulatory role on autophagy. We corroborated that the mutant cells show enhanced basal autophagy and generated RNA-seq data with very interesting leads (2 papers published). The latter findings are in preparation for a 3rd publication. The PGR has successfully graduated and hold a postdoctoral position in another group at the University of Leeds. Gianluca and I are co-sponsoring a second PGR to continue Begoña's work across UK-Italy (proposed start November 2023).
Collaborator Contribution The partner contributes to the full stipend and tuition support of the PRG, as well as to consumables related to about 50% of the PhD duration. His institution will sponsor a second PGR under the same conditions.
Impact Published: 1- Induction of Pro-Fibrotic CLIC4 in Dermal Fibroblasts by TGF-ß/Wnt3a Is Mediated by GLI2 Upregulation. Wasson CW, Caballero-Ruiz B, Gillespie J, Derrett-Smith E, Mankouri J, Denton CP, Canettieri G, Riobo-Del Galdo NA, Del Galdo F. Cells. 2022 Feb 3;11(3):530. 2- Partial Truncation of the C-Terminal Domain of PTCH1 in Cancer Enhances Autophagy and Metabolic Adaptability. Caballero-Ruiz B, Gkotsi DS, Ollerton H, Morales-Alcala CC, Bordone R, Jenkins GML, Di Magno L, Canettieri G, Riobo-Del Galdo NA. Cancers (Basel). 2023 Jan 6;15(2):369. Articles in preparation: 1- Mutations in the C-terminal domain of PTCH1 promote colorectal cancer cell growth. M. Begoña Caballero Ruiz, Danai-Stella Gkotsi, Rosa Bordone, Gianluca Canettieri and Natalia Riobo-Del Galdo Abstracts for conferences (EACR Congress 2022): 1- PTCH1 C-terminal domain mutations enhance colon cancer tumorigenesis B. Caballero-Ruiz, S. Coni, H. Wood, R. Bordone, G. Canettieri, N.A. Riobo-Del Galdo
Start Year 2019
 
Description Investigation of PTCH-domain containing family 
Organisation University of Helsinki
Department Helsinki Biobank
Country Finland 
Sector Academic/University 
PI Contribution We have contributed our expertise in Hedgehog signalling, specifically: recombinant Shh production, Gli-luciferase assays and confocal imaging.
Collaborator Contribution The partner has contributed technician time and protein expression and purificaiton of PTCHD1, as well as in silico structural analysis and docking.
Impact 1 publication (doi: 10.3390/ijms24032682) co-applicant in 2 Finish grant applications (1 unsuccessful, 1 pending)
Start Year 2019