Endocytic recycling is central to circadian collagen fibrillogenesis and disrupted in fibrosis

Curation statements for this article:
  • Curated by eLife

    eLife logo

    eLife assessment

    This important work substantially advances our understanding of how collagen fibrils are built and maintained in a manner regulated by circadian rhythms in intracellular secretory trafficking pathways. The evidence supporting the data are solid, although further data regarding the molecular mechanisms regulating endocytic recycling of collagen would have strengthened the study. The work will be of considerable interest to those who study extracellular matrix assembly or collagen homeostasis.

This article has been Reviewed by the following groups

Read the full article See related articles

Abstract

Collagen-I fibrillogenesis is crucial to health and development, where dysregulation is a hallmark of fibroproliferative diseases. Here, we show that collagen-I fibril assembly required a functional endocytic system that recycles collagen-I to assemble new fibrils. Endogenous collagen production was not required for fibrillogenesis if exogenous collagen was available, but the circadian-regulated vacuolar protein sorting (VPS) 33b and collagen-binding integrin-α11 subunit were crucial to fibrillogenesis. Cells lacking VPS33b secrete soluble collagen-I protomers but were deficient in fibril formation, thus secretion and assembly are separately controlled. Overexpression of VPS33b led to loss of fibril rhythmicity and over-abundance of fibrils, which was mediated through integrin α11β1. Endocytic recycling of collagen-I was enhanced in human fibroblasts isolated from idiopathic pulmonary fibrosis, where VPS33b and integrin-α11 subunit were overexpressed at the fibrogenic front; this correlation between VPS33b, integrin-α11 subunit, and abnormal collagen deposition was also observed in samples from patients with chronic skin wounds. In conclusion, our study showed that circadian-regulated endocytic recycling is central to homeostatic assembly of collagen fibrils and is disrupted in diseases.

Article activity feed

  1. eLife assessment

    This important work substantially advances our understanding of how collagen fibrils are built and maintained in a manner regulated by circadian rhythms in intracellular secretory trafficking pathways. The evidence supporting the data are solid, although further data regarding the molecular mechanisms regulating endocytic recycling of collagen would have strengthened the study. The work will be of considerable interest to those who study extracellular matrix assembly or collagen homeostasis.

  2. Reviewer #1 (Public Review):

    Summary:

    The authors describe that the endocytic pathway is crucial for ColI fibrillogenesis. ColI is endocytosed by fibroblasts, prior to exocytosis and formation of fibrils, which can include a mixture of endogenous/nascent ColI chains and exogenous ColI. ColI uptake and fibrillogenesis are regulated by circadian rhythm as described by the authors in 2020, thanks to the dependence of this pathway on circadian-clock-regulated protein VPS33B. Cells are capable of forming fibrils with recently endocytosed ColI when nascent chains are not available. Previously identified VPS33B is demonstrated not to have a role in endocytosis of ColI, but to play a role in fibril formation, which the authors demonstrate by showing the loss of fibril formation in VPS33B KO, and an excess of insoluble fibrils - along-side a decrease in soluble ColI secretion - in VPS33B overexpression conditions. A VPS33B binding protein VIPAS39 is also shown to be required for fibrillogenesis and to colocalise with ColI. The authors thus conclude that ColI is internalised into endosomal structures within the cell, and that ColI, VPS33B, and VIPA39 are co-trafficked to the site of fibrillogenesis, where along with ITGA11, which by mass spectrometric analysis is shown to be regulated by VPS33B levels, ColI fibrils are formed. Interestingly, in involved human skin sections from idiopathic pulmonary fibrosis (IPF) patients, ITGA11 and VPS33B expression is increased compared to healthy tissue, while in patient-derived fibroblasts, uptake of fluorescently-labelled ColI is also increased. This suggests that there may be a significant contribution of endocytosis-dependent fibrillogenesis in the formation of fibrotic and chronic wound-healing diseases in humans.

    Strengths:

    This is an interesting paper that contributes an exciting novel understanding of the formation of fibrotic disease, which despite its high occurrence, still has no robust therapeutic options. The precise mechanisms of fibrillogenesis are also not well understood, so a study devoted to this complex and key mechanism is well appreciated. The dependence of fibrillogenesis on VPS33B and VIPA39 is convincing and robust, while the distinction between soluble ColI secretion and insoluble fibrillar ColI is interesting and informative.

    Weaknesses:

    There are a number of limitations to this study in its current state. Inhibition of ColI uptake is performed using Dyngo4a, which although proposed as an inhibitor of Clathrin-dependent endocytosis is known to be quite un-specific. This may not be a problem however, as the endocytic mechanism for ColI also does not seem to be well defined in the literature, in fact, the principle mechanism described in the papers referred to by the authors is that of phagocytosis. It would be interesting to explore this important part of the mechanism further, especially in relation to the intracellular destination of ColI. The circadian regulation does not appear as robust as the authors' last paper, however, there could be a larger lag between endocytosis of ColI and realisation of fibrils. The authors state that the endocytic pathway is the mechanism of trafficking and that they show ColI, VPS33B, and VIPA39 are co-trafficked. However, the only link that is put forward to the endosomes is rather tenuously through VPS33B/VIPA39. There is no direct demonstration of ColI localisation to endosomes (ie. immunofluorescence), and this is overstated throughout the text. Demonstrating the intracellular trafficking and localisation of ColI, and its actual relationship to VPS33B and VIPA39, followed by ITGA11, would broaden the relevance of this paper significantly to incorporate the field of protein trafficking. Finally, the "self-formation" of ColI fibrils is discussed in relation to the literature and the concentration of fluorescently-tagged ColI, however as the key message of the paper is the fibrillogenesis from exocytosed colI, I do not feel like it is demonstrated to leave no doubt. Specific inhibition of intracellular trafficking steps, or following the progressive formation of ColI fibrils over time by immunofluorescence would demonstrate without any further doubt that ColI must be endocytosed first, to form fibrils as a secondary step, rather than externally-added ColI being incorporated directly to fibrils, independent of cellular uptake.

  3. Reviewer #2 (Public Review):

    Summary:

    In this manuscript, the authors describe a mechanism, by which fluorescently-labelled Collagen type I is taken up by cells via endocytosis and then incorporated into newly synthesized fibers via an ITGA11 and VPS33B-dependent mechanism. The authors claim the existence of this collagen recycling mechanism and link it to fibrotic diseases such as IPF and chronic wounds.

    Strengths:

    The manuscript is well-written, and experimentally contains a broad variation of assays to support their conclusions. Also, the authors added data of IPF patient-derived fibroblasts, patient-derived lung samples, and patient-derived samples of chronic wounds that highlight a potential in vivo disease correlation of their findings.

    The authors were also analyzing the membrane topology of VPS33B and could unravel a likely 'hairpin' like conformation in the ER membrane.

    Weaknesses:

    Experimental evidence is missing that supports the non-degradative endocytosis of the labeled collagen.

    The authors show and mention in the text that the endocytosis inhibitor Dyngo®4a shows an effect on collagen secretion. It is not clear to me how specific this readout is if the inhibitor affects more than endocytosis. This issue was unfortunately not further discussed. The authors use commercial rat tail collagen, it is unclear to me which state the collagen is in when it's endocytosed. Is it fully assembled as collagen fiber or are those single heterotrimers or homotrimers?

    The Cy-labeled collagen is clearly incorporated into new fibers, but I'm not sure whether the collagen is needed to be endocytosed to be incorporated into the fibers or if that is happening in the extracellular space mediated by the cells.

    In general for the collagen blots, due to the lack of molecular weight markers, what chain/form of collagen type I are you showing here?

    Besides the VPS33B siRNA transfected cells the authors also use CRISPR/Cas9-generated KO. The KO cells do not seem to be a clean system, as there is still a lot of mRNA produced. Were the clones sequenced to verify the KO on a genomic level? For the siRNA transfection, a control blot for efficiency would be great to estimate the effect size. To me it is not clear where the endocytosed collagen and VPS33B eventually meet in the cells and whether they interact. Or is ITGA11 required to mediate this process, in case VPS33B is not reaching the lumen?

    The authors show an upregulation of ITGA11 and VPS33B in IPF patients-derived fibroblasts, which can be correlated to an increased level of ColI uptake, however, it is not clear whether this increased uptake in those cells is due to the elevated levels of VPS33B and/or ITGA11.

  4. Reviewer #3 (Public Review):

    Summary:

    Chang et al. investigated the mechanisms governing collagen fibrillogenesis, firstly demonstrating that cells within tail tendons are able to uptake exogenous collagen and use this to synthesize new collagen-1 fibrils. Using an endocytic inhibitor, the authors next showed that endocytosis was required for collagen fibrillogenesis and that this process occurs in a circadian rhythmic manner. Using knockdown and overexpression assays, it was then demonstrated that collagen fibril formation is controlled by vacuolar protein sorting 33b (VPS33b), and this VPS33b-dependent fibrillogenesis is mediated via Integrin alpha-11 (ITGA11). Finally, the authors demonstrated increased expression of VPS33b and ITGA11 at the gene level in fibroblasts from patients with idiopathic pulmonary fibrosis (IPF), and greater expression of these proteins in both lung samples from IPF patients and in chronic skin wounds, indicating that endocytic recycling is disrupted in fibrotic diseases.

    Strengths:

    The authors have performed a comprehensive functional analysis of the regulators of endocytic recycling of collagen, providing compelling evidence that VPS33b and ITGA11 are crucial regulators of this process.

    Weaknesses:

    Throughout the study, several different cell types have been used (immortalised tail tendon fibroblasts, NIHT3T cells, and HEK293T cells). In general, it is not clear which cells have been used for a particular experiment, and the rationale for using these different cell types is not explained. In addition, some experimental details are missing from the methods.

    There is also a lack of functional studies in patient-derived IPF fibroblasts which means the link between endocytic recycling of collagen and the role of VPS33b and ITGA11 cannot be fully established.