Subscribe to RSS

DOI: 10.1055/a-2599-4925
HSP47 at the Crossroads of Thrombosis and Collagen Dynamics: Unlocking Therapeutic Horizons and Debates
Abstract
Heat shock protein 47 (HSP47), a collagen-specific molecular chaperone encoded by the SERPINH1 gene, has emerged as a groundbreaking focus in thrombosis research. Recent findings published in “Science” have revolutionized our understanding of thrombosis, identifying HSP47 as a critical mediator in a new thrombosis target for treatment. This discovery not only unveils a novel pathway in thrombosis but also opens new avenues for therapeutic intervention. HSP47's significance extends beyond thrombosis, influencing pathological processes such as fibrosis and cancer. In fibrosis, its upregulation promotes collagen deposition, while its dysregulation in osteogenesis imperfecta (OI) Type X underscores the protein's indispensable role in collagen biosynthesis. The therapeutic challenge lies in balancing HSP47 inhibition to reduce fibrotic burden without impairing its essential physiological functions. In cancer, HSP47 plays dual roles. It supports tumor progression through collagen stabilization and metastasis facilitation while contributing to tissue repair under hyperthermia treatment combined with radiotherapy or chemotherapy. However, its overexpression can exacerbate tumor aggressiveness via mechanisms such as angiogenesis and epithelial–mesenchymal transition.
This review emphasizes the pivotal discovery of HSP47's thrombogenic role and its broader implications in disease biology. These findings mark a paradigm shift in thrombosis research and underscore the potential of HSP47 as a target in diverse pathological contexts, from platelet-driven diseases to fibrotic and oncological disorders.
#
Keywords
heat shock protein 47 - collagen - thrombosis - angiogenesis - cancer therapy - hyperthermiaIntroduction
Type I collagen serves as the primary structural component of the extracellular matrix (ECM) and plays a critical role in fibrosis and platelet adhesion.[1] Structurally, type I collagen comprises two α1 chains and a single α2 chain, which assemble into a triple-helical configuration within the endoplasmic reticulum (ER) before being transported to the Golgi apparatus.[2] This collagen is inherently thermally unstable at physiological temperatures, necessitating the assistance of specialized chaperone proteins to ensure proper folding and stability.[3] One such indispensable chaperone is heat shock protein (HSP) 47 (HSP47), a collagen-specific binding protein encoded by the SERPINH1 gene and residing in the ER.[4] HSP47 is structurally related to the serpin family but does not function as a serine protease inhibitor.[5] [6] Unlike other chaperones such as HSP60, HSP70, and HSP90, which interact with a wide range of substrates,[7] [8] HSP47 exhibits unique specificity, binding exclusively to procollagen. HSP47 is crucial for the proper folding of procollagen within this compartment. Indeed, this ER-resident molecular chaperone binds to the Gly-Xaa-Arg motif in procollagen, stabilizing the triple helix.[5] [6] By preventing local unfolding and aggregation, HSP47 enables the successful formation of the collagen's helical structure ([Fig. 1]).[5] [6] The acidification of organelles, particularly the ER and Golgi apparatus, plays a crucial role in the function of HSPs. HSP47 relies on the pH gradient within these organelles to regulate collagen maturation and secretion. However, its dissociation from procollagen is pH-dependent, occurring in the acidic environment of the cis-Golgi. In the ER, which maintains a neutral pH of approximately 7.2 to 7.4, HSP47 binds newly synthesized procollagen and prevents premature aggregation. This binding is critical for the correct folding and stabilization of procollagen. As procollagen is transported to the Golgi, the pH progressively decreases, reaching approximately 6.3 to 6.7 in the cis-Golgi. This acidification triggers the release of HSP47 from the collagen molecule.[5] [9] The dissociation of HSP47 in this environment is essential for the proper trafficking of procollagen to the ECM. If the acidification process is disrupted, such as through the inhibition of vacuolar H+-ATPase by compounds like bafilomycin A1, HSP47 remains bound to procollagen, leading to its retention and impaired secretion. In the trans-Golgi, where the pH is further reduced to around 6.0, HSP47 is recycled back to the ER via coat protein complex I-mediated retrograde transport. This recycling process ensures that HSP47 is available to assist in the maturation of newly synthesized procollagen molecules. Acidification within organelles is essential for the function of various HSPs involved in protein maturation, trafficking, and degradation. Other HSP families, such as HSP70 and HSP90, rely on pH-sensitive mechanisms to regulate protein folding and release in the ER, protein degradation in lysosomes, and the activation of stress responses in acidic microenvironments.


HSP47 involvement in both physiological and pathological processes is now well established.[10] Its involvement in collagen synthesis and maturation has many implications for diseases where collagen integrity is compromised, such as OI, fibrosis, and certain cancers.[11] Recent studies have expanded our understanding of HSP47's functions, suggesting that it also plays significant roles in thrombosis or angiogenesis. In thrombosis, HSP47 is found on activated platelets, where it supports platelet aggregation by enhancing collagen interactions and thrombus formation.[12] [13] This association with thrombosis, particularly in conditions involving immobilization, has spurred interest in HSP47 as a target for thromboprotective therapies, as demonstrated in studies involving hibernating animals,[14] which exhibit natural resistance to clot formation. However, targeting HSP47 for thrombosis prevention or other therapeutic applications, like idiopathic pulmonary fibrosis (IPF), must take into account its essential role in collagen stability, as deficiency or inhibition of HSP47 can lead to severe skeletal defects, as observed in OI Type X.[15]
This review aims to consolidate current knowledge on HSP47's diverse roles, exploring its implications in collagen biology, thrombosis, angiogenesis, but also questions about the relevance of HSP47 target in fibrosis and cancer. By addressing both the therapeutic promise and the challenges of targeting HSP47, we hope to elucidate its potential as a multifaceted biomarker and therapeutic target across a spectrum of diseases, emphasizing the importance of context in understanding HSP47's complex biology.
HSP47 as a Target in Pulmonary Fibrosis and Cancer
Many diseases are associated with either collagen accumulation or malfunction.[16] Fibrosis is one such condition, particularly when abnormal collagen accumulates in organs such as the liver,[17] [18] lungs,[19] and kidneys.[20] [21] We will focus in the next paragraph on pulmonary fibrosis. Indeed, HSP47 expression is upregulated by key profibrotic factors in pulmonary fibrosis models.[22] TGF-β1 enhances HSP47 and collagen type I expression in normal human fibroblasts and alveolar epithelial cells.[23] Similarly, IL-1β promote HSP47 synthesis.[24] In bleomycin-induced pulmonary fibrosis, HSP47 expression is elevated in myofibroblasts, alveolar epithelial cells, and macrophages, correlating with increased collagen deposition.[25] [26] Aging amplifies this response, with older mice showing greater HSP47 and collagen levels.[27] Exposure to TGF-β1, a key extracellular regulator of fibrosis, induced an upregulation in both HSP47 and collagen type I at the mRNA and protein levels in normal human lung fibroblasts and alveolar epithelial cells.[28] [29] ER stress and unfolded protein response pathways further regulate HSP47, as demonstrated by increased expression in fibrotic lungs.[30] Conversely, HSP47 levels decrease in conditions like acute respiratory distress syndrome associated with diffuse alveolar damage.[31] Finally, HSP47 expression is significantly higher in fibrotic lungs of patients with IPF and correlates with disease severity.[11] [25] [32] [33] It is localized to myofibroblasts and type II alveolar epithelial cells in areas of active fibrosis.[34] Serum HSP47 levels are elevated in acute IPF exacerbations and acute interstitial pneumonia, making it a potential biomarker.[11] Additionally, anti-HSP47 autoantibodies are prevalent in autoimmune and fibrotic diseases, likely triggered by tissue damage and inflammation.[35] Given HSP47's central role in fibrosis, therapeutic strategies aimed at modulating HSP47 expression and downstream effects may help mitigate fibrotic diseases. Nintedanib, an already approved treatment for IPF and other progressive fibrosing interstitial lung diseases, has been shown to suppress lung fibroblast proliferation, motility, and myofibroblast differentiation.[36] [37] [38] [39] [40] [41] [42] It also reduces TGF-β1-induced collagen production but has minimal effect on HSP47 protein levels, despite lowering its mRNA through SERPINH1 suppression. The other validated treatment for IPF, pirfenidone, is an orally administered synthetic molecule that slows disease progression and reduces fibrosis in organs such as the kidney, liver, and heart.[43] It achieves this by inhibiting profibrotic mediators such as TGF-β1, TNF-α, and IL-1β, while also exhibiting anti-inflammatory and antioxidant properties. Notably, pirfenidone decreases HSP47 and collagen expression in lung cells and reduces the presence of HSP47-positive fibroblasts in fibrotic tissues.[28] [29] [41] A variety of drugs have demonstrated the ability to influence HSP47, shedding light on its potential as a therapeutic target in fibrosis. Diallyl sulfide, a sulfur compound derived from garlic, has shown significant antifibrotic effects by suppressing both HSP47 and α-smooth muscle actin expression in models of lung fibrosis.[44] Aminoguanidine, known for its ability to inhibit advanced glycation end-products linked to inflammation and fibrosis, markedly reduces HSP47 and collagen expression in fibrotic tissues.[45] [46] Corticosteroids like budesonide and fluticasone are also effective, downregulating SERPINH1 mRNA and collagen production in lung fibroblasts, making them relevant in treating fibrotic conditions.[47] Moreover, prieurianin and its analogs, such as dregeanin—naturally occurring limonoids from plants like Trichilia prieuriana and Aphanamixis polystachya—exhibit diverse biological properties, including antifibrotic activity, by targeting HSP47 at the protein–collagen interface.[48] Specific compounds designed to target the pH-dependent dissociation of Hsp47 from procollagen in the Golgi are still under active investigation. However, several general approaches to modulating Hsp47 function and its interaction with procollagen have been explored. Glycine analogs, such as Gly-Pro-Arg, mimic collagen sequences and have shown potential to interfere with Hsp47 binding, which could indirectly affect its dissociation from procollagen. Agents that alter the pH gradient in the Golgi, including ammonium chloride and bafilomycin A1 (a vacuolar H+-ATPase inhibitor), can disrupt the acidic conditions required for Hsp47 release, thus influencing its activity.[49] [50] Adding to these strategies, SERPINH1 siRNA has emerged as a promising approach, effectively silencing HSP47 expression in animal models, reducing fibrosis markers, and improving lung function.[51] Collectively, these therapeutic interventions emphasize the critical role of HSP47 in fibrosis and its value as a focal point for developing treatments to enhance patient outcomes ([Table 1])[5] [41] [44] [45] [47] [48] [49] [51] [52] [53] and across multiple fibrotic diseases (e.g., pulmonary, liver, kidney, or skin fibrosis; [Table 2]).[54] [55] [56] [57] [58] [59] [60] [61] [62] [63] [64] [65]
Drug/Compound |
Target disease/condition |
Effect on HSP47 |
References |
---|---|---|---|
Nintedanib |
Idiopathic pulmonary fibrosis |
Reduces SERPINH1 mRNA (HSP47 precursor) but minimal effect on HSP47 protein levels |
Knüppel et al. (2017)[41] |
Pirfenidone |
IPF, fibrotic diseases |
Decreases HSP47 and collagen expression in lung cells, reduces HSP47-positive fibroblasts—reduces SERPINH1 mRNA (HSP47 precursor) but minimal effect on HSP47 protein levels |
Knüppel et al. (2017)[41] |
Diallylsulfide |
Lung fibrosis |
Suppresses HSP47 and α-SMA expression |
Kalayarasan et al. (2013)[114] |
Aminoguanidine |
Fibrosis, inflammation |
Reduces HSP47 and collagen expression |
Chen et al. (2009)[45] |
Corticosteroids (budesonide, fluticasone) |
Fibrosis, lung diseases |
Downregulate SERPINH1 mRNA, decrease collagen production |
Goulet et al. (2007)[47] |
Prieurianin and Dregeanin |
Fibrosis |
Targets HSP47 at protein–collagen interface |
Vergoten et al. (2024)[48] |
Bafilomycin A1 |
Various diseases |
Disrupts acidic conditions required for HSP47 release from procollagen |
Satoh et al. (1996)[49] |
SERPINH1 siRNA |
Fibrosis (lung, liver, kidney) |
Suppresses HSP47 expression, reduces fibrosis markers |
Li et al. (2021)[115] |
Epigallocatechin gallate |
? |
? |
Okuno et al. (2021)[52] |
Col003 |
Ischemic stroke and fibrosis |
Blocking interactions between collagen and HSP47 |
|
Toolkit peptides |
? |
Peptides blocking interactions between collagen and HSP47 |
Cai et al. (2021)[84] |
Abbreviations: HSP47, heat shock protein 47; IPF, idiopathic pulmonary fibrosis.
This table presents an overview of key compounds and drug candidates investigated for their effects on HSP47 in the context of fibrosis, inflammation, and thromboinflammatory disorders. The agents listed—ranging from U.S. Food and Drug Administration-approved drugs (e.g., nintedanib, pirfenidone) to experimental peptides—target various stages of HSP47 regulation, including transcription (SERPINH1 mRNA), translation, and protein–collagen interaction. Mechanisms include suppression of collagen synthesis, disruption of HSP47–collagen binding, and inhibition of fibroblast activation. The table also highlights the variability in therapeutic impact, including instances of incomplete protein-level inhibition despite reduced mRNA levels.
Disease |
Model |
Organ |
Therapeutic outcome |
References |
---|---|---|---|---|
Scleroderma |
Subcutaneous implantation of keloid in mice |
Skin |
Reduced collagen—reduced keloid volume |
Chen et al. (2011)[54] |
Pulmonary fibrosis |
Oral administration of paraquat (20 mg/kg/day) in rats |
Lung |
Reduction in pulmonary fibrosis with reduced HSP47 and collagen |
Hagiwara et al. (2007)[55] |
Pulmonary fibrosis |
Intratracheal administration of bleomycin in rats |
Lung |
Reduced HSP47 and collagen expression—improved survival |
Hagiwara et al. (2007)[56] |
Liver fibrosis |
Percutaneous instillation of japonicum cercariae |
Liver |
Improved survival, reduced liver markers and fibrosis |
Huang et al. (2014)[57] |
Scleroderma |
Intradermal administration of Bleomycin in mice |
Skin |
Reduced NOX4, α-SMA, collagen type I—reduced skin thickness |
Morry et al. (2015)[58] |
Kidney fibrosis |
Unilateral ureteral obstruction in mice |
Kidney |
Reduced collagens I, II, IV—decreased macrophage infiltration |
Xia et al. (2008)[59] |
Peritoneal fibrosis |
Intraperitoneal injection of chlorhexidine gluconate |
Peritoneum |
Suppressed collagen and macrophage infiltration |
Obata et al. (2012)[60] |
Liver cirrhosis |
Intraperitoneal injection of dimethylnitrosamine in rats |
Liver |
Prolonged survival—reduced collagen and fibrosis |
Sato et al. (2008)[61] |
Pancreatic fibrosis |
Jugular injection of dibutyltin dichloride |
Pancreas |
Reduced collagen and fibrosis |
Ishiwatari et al. (2013)[62] |
Pulmonary fibrosis |
Intratracheal administration of bleomycin |
Lung |
Reduced collagen and cytokines—improved lung structure |
Otsuka et al. (2017)[63] |
Dry eye |
Mouse model of GVHD |
Eye |
Reduced HSP47—restored tear secretion—reduced fibrosis |
Ohigashi et al. (2019)[64] |
Skin fibrosis |
Cutaneous chronic GVHD in mice |
Skin |
Targeting HSP47+ myofibroblasts without inducing immunosuppression |
Yamakawa et al. (2018)[65] |
Abbreviation: HSP47, heat shock protein 47; GVHD, graft-versus-host disease.
This table summarizes preclinical studies evaluating the outcomes of targeting HSP47 in various fibrotic diseases, including pulmonary, liver, kidney, pancreatic, and skin fibrosis. HSP47 inhibition was achieved through diverse models involving bleomycin, paraquat, apioncin, or chemical inducers. These studies support the therapeutic relevance of HSP47 modulation but also underscore the need for controlled targeting to prevent excessive tissue remodeling or impairment of physiological repair responses.
Fibrosis and cancer share common features, particularly in their reliance on collagen remodeling and cellular proliferation to drive disease progression. In both conditions, excessive collagen deposition alters the structure and function of the ECM, leading to tissue stiffness and disruption of normal cellular environments. HSP47 is overexpressed in various cancers,[66] [67] [68] where its upregulation enhances tumor aggressiveness, though the underlying mechanisms remain unclear. The SERPINH1 gene, which encodes the heat-inducible protein HSP47, is situated on chromosome 11q13.5, a genomic hotspot commonly amplified in various human cancers.[69] Moreover, data from the University of ALabama at Birmingham CANcer (UALCAN) cancer database reveal that HSP47 mRNA levels are significantly elevated in multiple tumor types, including bladder, breast, colon, liver, lung, and stomach cancers, compared with normal tissues. Kaplan–Meier survival analysis further shows that higher HSP47 expression is strongly associated with poorer overall survival in patients with several cancers, such as bladder, liver, and lung adenocarcinomas.[70] Understanding these links is critical for advancing cancer treatment strategies. HSP47 plays a multifaceted role in cancer progression, influencing tumor growth, metastasis, and angiogenesis.[70] Overexpression of HSP47 promotes tumor cell proliferation by interacting with ER chaperones and stabilizing collagen production, essential for the ECM.[66] [71] It also enhances epithelial–mesenchymal transition, a process critical for metastasis, by regulating key proteins in the Wnt/β-catenin and TGF-β pathways.[72] Additionally, HSP47 stabilizes Discoidin domain receptors (a class of collagen-binding receptor tyrosine kinases) DDR2, facilitating cancer cell migration and invasion.[73] In metastatic breast cancer, HSP47 strengthens actin filament contractility, enhancing the spread of cancer cells.[74] It also promotes cancer cell–platelet interactions, which protect circulating tumor cells and support their colonization in distant organs through the HSP47–collagen axis.[71] Furthermore, HSP47 contributes to angiogenesis[75] by activating extracellular signal-regulated kinase signaling and upregulating VEGF-A via hypoxia-inducible factor-1α (HIF-1α). These interconnected pathways highlight HSP47 as a central player in cancer development and a promising target for therapeutic intervention. HSP47 might actively promote angiogenesis in glioma through the HIF1α–VEGFR2 pathway.[75] In bladder cancer, the ERK signaling pathway, in conjunction with C–C motif chemokine ligand 2 (CCL2), fosters HSP47-mediated angiogenesis.[76] Studies indicate that HSP47 inhibitors can counteract VEGF-driven fibrovascular changes in retinal diseases, reducing the fibrovascular formation that contributes to conditions like retinal fibrosis.[77] This effect suggests a broader therapeutic potential for HSP47 inhibitors in controlling pathological angiogenesis beyond oncology.
#
HSP47 Inhibition as a New Treatment for Thrombosis
Thrombosis contributes to approximately 25% of global mortality, underscoring its critical status as a leading public health threat. It is central to the pathophysiology of numerous cardiovascular complications, including myocardial infarction, stroke, and venous thromboembolism (VTE)—a condition encompassing deep vein thrombosis (DVT) and pulmonary embolism.[78] Risk factors for VTE range from classic contributors like cancer and surgery to immobilization alone, which independently elevates thrombotic risk even in otherwise healthy individuals.[79] The distinction between arterial and venous thrombosis is key: While platelets dominate arterial clotting through their adhesion to exposed collagen and subsequent aggregation, venous thrombosis primarily involves fibrin deposition, driven by coagulation factors and inflammatory mediators rather than platelets. Amidst the ongoing exploration of thrombogenic mechanisms, HSP47, traditionally known for its intracellular chaperoning of collagen, has emerged as a novel player in platelet biology. Proteomic analyses identified HSP47 on the surface of GPVI-activated platelets, challenging conventional views of its localization.[12] Functional studies revealed that HSP47 directly binds to collagen fibrils and facilitates platelet aggregation through collagen–GPVI interactions. Inhibiting HSP47 significantly diminished these interactions, highlighting its modulatory role in platelet–collagen adhesion.[13] [80] Knockout mice lacking platelet-specific HSP47 or treated with HSP47 inhibitors displayed impaired thrombus formation under both in vivo and flow-based in vitro conditions, despite maintaining normal platelet counts. These mice also exhibited prolonged bleeding times, revealing HSP47's integral role in thrombus stability and hemostasis.[13]
Interestingly, HSP47 functions not as a classical transmembrane receptor but more likely as a co-receptor or surface scaffold.[12] [13] It enhances GPVI signaling pathways that lead to platelet adhesion and spreading, rather than initiating full activation independently. Studies demonstrate that collagen motifs bound by HSP47 support morphological changes like shape change, without inducing granule release or integrin αIIbβ3 activation. Moreover, HSP47 binds to the GFOGER motif—a recognized integrin α2β1 collagen-binding sequence—but does not directly modulate this interaction,[80] further supporting its role as an accessory regulator rather than a primary receptor.
Mechanistically, HSP47 is stored in the dense tubular system of resting platelets and translocates to the membrane upon activation via actin dynamics, similar to protein disulfide isomerase.[80] [81] It enhances cytoskeletal remodeling, a trait observed in other cell types like fibroblasts and epithelial cells,[32] [82] and is essential for promoting platelet spreading. It also stabilizes GPVI receptor dimerization and downstream phosphorylation of key signaling proteins such as spleen tyrosine kinase (Syk), linker for activation of T-cells (LAT), and phospholipase C gamma 2 (PLCγ2)—integral elements of the GPVI signaling axis.[80]
Recent compelling data have further connected HSP47 to thrombin-mediated activation. Thienel et al.[14] demonstrated that HSP47 facilitates thrombin binding to platelets and is essential for downstream GPVI signaling, even though thrombin is traditionally associated with protease-activated receptor receptor activation.[14] Inhibition of HSP47 abrogated thrombin-triggered platelet aggregation, granule release, and neutrophil extracellular trap (NET) formation—events central to thromboinflammation. These findings position HSP47 as a nexus linking thrombin engagement to GPVI signal amplification.[14]
In cancer-associated thrombosis (CT), HSP47 has also been implicated in platelet–cancer cell crosstalk through collagen secretion pathways,[71] reinforcing the protein's versatility beyond classical clot formation. Furthermore, studies in hibernating mammals such as brown bears reveal that their natural resistance to VTE during prolonged immobilization is associated with suppressed HSP47 levels, suggesting that downregulation of HSP47 may be an evolutionary thromboprotective adaptation.[14] Experimental models corroborate these observations. In PF4-Cre (+)/HSP47−/− knockout mice subjected to inferior vena cava thrombosis, both thrombus size and frequency were significantly reduced. These mice also showed diminished NET formation and reduced circulating D-dimer and citH3/DNA complexes, further emphasizing HSP47's pivotal role in thromboinflammatory pathways.[14]
Moreover, recent findings from whole exome sequencing of SERPINH1 have identified 99 variants, including missense and loss-of-function mutations. These variants were analyzed for differences in allele frequency between individuals with and without VTE.[83] The Malmö Diet and Cancer cohort, comprising 28,794 participants, was explored with a follow-up period extending until December 31, 2018. Among this population, 2,584 individuals experienced VTE events. Variants were assessed for their association with VTE using Cox regression models adjusted for age, sex, and additional risk factors such as body mass index (BMI), smoking, and Factor V Leiden. Kaplan–Meier survival analysis for thrombosis-free survival showed no significant differences in outcomes between carriers and non-carriers of SERPINH1 variants. Furthermore, genetic databases such as Genebass and UK Biobank exomes did not reveal significant links between SERPINH1 and thrombotic risk. Thus, the study concludes that SERPINH1 genetic variation is not associated with an increased risk of VTE in the general population, including both common and rare variants. While HSP47 has a critical biological role in platelet function and collagen interaction, its genetic variability does not appear to be a significant determinant of VTE risk in the studied population.[83]
From a translational standpoint, this creates a compelling case for therapeutic targeting. Unlike conventional antiplatelet agents that often carry bleeding risks, HSP47 inhibition appears to offer antithrombotic benefits without significantly impairing primary hemostasis. Thienel et al. showed that small-molecule HSP47 inhibitors (SMI I and II) reduced thrombus formation in vivo without affecting bleeding time[14]—potentially overcoming one of the greatest limitations of current therapies like P2Y12 or GPIIb/IIIa inhibitors. Furthermore, recent studies indicate that HSP47 inhibitors reduce cerebral vascular damage in ischemic stroke models.[53] Structural work by Cai et al.[84] delineated the collagen-binding domains of HSP47 and identified potent inhibitory compounds capable of blocking its interaction with native collagens. These agents provide a rational basis for the development of selective, structure-guided inhibitors that target pathologic thrombosis without impairing physiological clotting mechanisms. Moreover, venous thrombosis is a common and severe complication in cancer patients, often driven by tumor-derived prothrombogenic extracellular vesicles (EVs).[85] [86] [87] Recent evidence, including data from Osorio et al.,[88] highlights the presence of HSP47 on the surface of small EVs isolated from human blood. This is particularly relevant given HSP47's established roles in collagen stabilization and thrombosis via platelet–collagen interactions. Although HSP47 was traditionally viewed as an intracellular chaperone for collagen, its extracellular role—especially when bound to vesicular membranes—suggests a broader functional spectrum. The detection of HSP47 in small extracellular vesicles secreted in heart failure[88] supports the hypothesis that this chaperone may be externalized in pathological microenvironments such as those found in tumors. Since tumor hypoxia is a hallmark of aggressive malignancy and a known trigger for thromboinflammatory signaling, the presence of HSP47 in tumor-derived EVs may facilitate thrombin generation and platelet activation indirectly through interactions with collagen or other matrix components exposed at sites of vascular damage or inflammation. This aligns with prior findings where platelet-surface HSP47 supported GPVI signaling and promoted thrombus formation. Hence, in cancer-related thrombosis, tumor-derived EVs carrying HSP47 could mimic or enhance platelet–collagen interactions, thereby contributing to a procoagulant state. From a translational standpoint, this suggests that targeting vesicular HSP47—either directly through inhibitory antibodies or indirectly via blockade of EV uptake—may represent a novel antithrombotic strategy in oncology. Given the dual role of HSP47 in both fibrosis and thrombosis, its detection in EVs also opens avenues for biomarker development, helping to stratify cancer patients at elevated thrombotic risk. Finally, in regenerative medicine contexts, HSP47 appears to exert indirect influence on platelets through matrix remodeling. For instance, platelet-rich plasma and platelet-rich fibrin therapies have been shown to stimulate HSP47 expression in surrounding osteoblasts and tendon cells, promoting collagen synthesis and healing.[89] [90] These findings suggest that activated platelets may influence extracellular HSP47 signaling in wound healing, even when HSP47 is not acting directly within the platelet itself.
Altogether, these multilayered insights into HSP47's role—spanning collagen binding, GPVI modulation, thrombin signaling, and matrix biology—support its candidacy as a highly promising therapeutic target in thrombotic diseases. Future efforts should aim to bridge preclinical discoveries with human applications, refine delivery mechanisms for selective inhibition, and delineate the contexts in which HSP47 modulation offers the greatest clinical value.
#
HSP47 Inhibition: Is It Beneficial or Harmful?
The prospect of inhibiting HSP47 as a therapeutic strategy presents both opportunities and challenges, necessitating a nuanced approach to its application in medicine. HSP47, a collagen-specific molecular chaperone, plays a crucial role in ensuring proper collagen folding and stability. Its significance extends across various physiological processes, from maintaining structural integrity in bones to supporting ECM remodeling. Several compelling examples highlight the complexity of inhibiting HSP47 and emphasize the need for caution: The impact of HSP47 on bone integrity in OI, its potential necessity in hyperthermia (HT)-based cancer therapies, but also the potential impact on bleeding or autoimmune disorders.
First, OI is a genetic disorder characterized by fragile bones due to mutations affecting collagen production or structure, primarily type I collagen. OI Type X, a rare form of the disease, is caused by mutations in the SERPINH1 gene, which encodes HSP47.[15] In this condition, defective HSP47 disrupts the proper folding and assembly of collagen, resulting in brittle bones, frequent fractures, and compromised skeletal integrity. Recent data have explored the role of the SERPINH1 gene in another collagen disorder: hypermobile Ehlers–Danlos Syndrome (hEDS). The study investigates potential genetic contributions by sequencing the SERPINH1 gene in 100 Polish patients diagnosed with hEDS.[91] Four heterozygous missense variants in the SERPINH1 gene were identified in the study group. However, these variants were classified as benign. The study found no pathogenic or likely pathogenic mutations in SERPINH1, suggesting that the gene does not play a direct role in the etiology of hEDS. This result emphasizes the pivotal role of HSP47 in collagen biosynthesis and underscores the complexity of its potential involvement in various connective tissue diseases. The findings provide a foundation for further exploration into molecular mechanisms underlying hEDS and related disorders. Given its critical function, the inhibition of HSP47 could inadvertently mimic or exacerbate the pathological features of OI. Beyond bones, such inhibition may impair connective tissues throughout the body, leading to weakened tendons, poor wound healing, and vascular instability due to compromised collagen quality. Collagen is also vital for ECM remodeling, and disrupting this process could impair normal tissue repair and regeneration. To minimize these risks, therapeutic strategies targeting HSP47 must be carefully tailored, prioritizing selective inhibition in specific tissues or partial suppression to balance therapeutic benefits with essential physiological functions.
Second, HT, or thermal therapy, involves raising tissue temperatures to approximately 39 °C to 42 °C for about an hour.[92] [93] [94] [95] This approach is widely studied in oncology, where it is often combined with chemotherapy, radiotherapy, or immunotherapy to enhance treatment efficacy. HT induces a range of beneficial cellular responses, including the upregulation of HSPs, such as HSP47.[96] [97] This upregulation has been observed across species, including fish[98] [99] and humans, where HSP47 is significantly elevated during thermal stress, aiding in collagen remodeling and tissue repair. In humans, several studies have demonstrated that HT induces an elevation in HSP47 mRNA transcription (Northern blot analysis) and protein translation (Western blot analysis) shortly after treatment.[100] [101] [102] [103] [104] [105] This transcriptional and translational up-regulation is evident by the 6th hour, reaching its peak at the 9th hour.[106] Research in human dermal fibroblasts has also demonstrated that pulsed heat shocks at 60 °C result in greater collagen synthesis and reduced collagen degradation, leading to a higher overall net collagen content compared with heat shocks at 45 °C.[103] This stresses the importance of both temperature and pulse duration, as optimal pulses lasting approximately 2 seconds per rise significantly influence collagen remodeling and tissue repair. Additionally, these short-pulsed heat shocks at 45 °C and 60 °C led to an upregulation of both procollagen type I and procollagen type III. Studies examining HSP47 mRNA have also found a decrease in HSP47 transcription with aging. This phenomenon was mostly evident in cells from older individuals treated with HT, suggesting that the reduction in HSP47's heat response may contribute to the deteriorated quality and quantity of collagen observed with aging.[107] These examples highlight the dual nature of HSP47's role in medicine. Last but not least, HT shows promise in preventing thrombosis.[76] Indeed, in this recent study, the synergistic benefits of combining HT with intermittent pneumatic compression to enhance blood flow velocity and reduce the risk of DVT has been demonstrated. Temperatures between 42 °C and 45 °C were particularly effective, demonstrating superior hemodynamic outcomes while preserving vascular integrity. These findings suggest HT as a valuable intervention in thrombosis management following orthopedic procedures.[108]
Thus, on one hand, HSP47 inhibition may offer therapeutic benefits in conditions like fibrosis or cancer. On the other hand, its essential role in maintaining collagen stability and tissue integrity, as evidenced by its inhibition in OI and its beneficial induction in HT-based therapies, highlights the potential risks associated with modulating HSP47. Moreover, the relationship between HT, thrombosis, and HSP47 regulation remains complex and somewhat controversial. The recent findings by Tong et al. underscore the dual nature of HT's effects.[108] While it effectively boosts blood flow and promotes venous return, it may also upregulate HSP47 could potentially contribute to thrombosis by promoting a procoagulant environment as described in [Fig. 2]. These contrasting effects emphasize the need for cautious and context-specific therapeutic strategies. Modulating HSP47 requires a balanced approach to harness its benefits while mitigating potential risks.


Moreover, while our review has primarily centered on HSP47's role in thrombosis and fibrosis, its potential involvement in bleeding complications is worth further exploration. A study published in the “Journal of Thrombosis and Haemostasis” by Sasikumar highlights HSP47 as a platelet collagen-binding protein that contributes to thrombosis and hemostasis.[13] The study notes an increase in bleeding time associated with HSP47 activity, although there are currently no direct data available regarding its role in human cancer-related bleeding.
Finally, while targeting HSP47 has demonstrated promising therapeutic potential in fibrotic and thrombotic conditions, concerns arise regarding its inhibition in autoimmune diseases such as rheumatoid arthritis, where collagen integrity is already compromised.[109] [110] Elevated HSP47 expression and the presence of anti-HSP47 autoantibodies are associated with autoimmune pathologies such as rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis.[35] [111] [112] [113] These findings suggest that inappropriate or non-selective inhibition of HSP47 may risk exacerbating inflammation or compromising tissue structural integrity in already vulnerable patients. Importantly, HSP47 is not only implicated in collagen maturation but also plays broader roles in angiogenesis, cellular stress responses, and immune modulation. It can be expressed aberrantly on the surface of activated cells and may act as a proinflammatory mediator in damaged tissues. This dual functionality underlines the necessity for strategies that achieve therapeutic inhibition of HSP47 while minimizing collateral effects on healthy tissue or immune tolerance. To address these risks, several innovative approaches are being developed. Tissue-specific delivery systems, such as antibody-guided or nanoparticle-based formulations, could allow targeted inhibition of HSP47 in fibrotic or thrombotic lesions while sparing tissues involved in joint integrity or immune regulation. Intermittent or low-dose administration may provide therapeutic effects during disease flares without compromising baseline collagen maintenance. Additionally, targeting HSP47 within the vasculature—particularly in endothelial cells or platelets implicated in thrombosis—may allow selective modulation of thromboinflammatory processes with a lower risk of systemic immune disruption. Furthermore, monitoring biomarkers such as circulating anti-HSP47 antibodies may help stratify patients according to risk and tailor dosing regimens appropriately. Such approaches, informed by the context-specific role of HSP47, would support the safe and effective development of HSP47-targeted therapies, especially in patients with coexisting autoimmune and fibrotic diseases.
#
#
Conclusion
All in all, the discovery of HSP47's surface localization on platelets and its critical role in thrombosis represents one of the most extraordinary advancements in recent years within the thrombosis field. This revelation fundamentally alters our understanding of thrombus formation after immobilization, identifying HSP47 as a previously unrecognized but essential player in thrombosis. Its targeting presents an unparalleled opportunity for developing innovative antithrombotic therapies. Beyond thrombosis, HSP47's roles in fibrosis and cancer highlight its versatility and complexity. While it promotes collagen stability and tissue repair, its dysregulation can drive pathological processes such as fibrotic scarring and tumor progression. However, the relationship between HSP47 and HT introduces a nuanced perspective, particularly in oncology. This paradox suggests that inhibiting HSP47 in cancer may inadvertently undermine therapeutic strategies that rely on its beneficial effects during HT treatments. These dual roles demand careful modulation of HSP47 activity. While inhibition could alleviate conditions like fibrosis or thrombosis, it risks impairing essential collagen biosynthesis, as seen in OI Type X, or contradicting therapeutic gains in cancer HT protocols. This extraordinary discovery of HSP47's thrombogenic function underscores its centrality in disease biology. Harnessing this insight will not only advance therapeutic strategies for thrombosis but also provide a framework for addressing its roles in thrombosis, fibrosis, and cancer. Future research must prioritize tissue-specific and context-dependent modulation of HSP47 to unlock its full therapeutic potential while safeguarding its critical physiological functions. Overall, refining delivery strategies and therapeutic windows will be essential for translating HSP47 inhibition into a viable clinical intervention across diverse pathologies.
#
#
Conflict of Interest
M.M.A. is the founder of BTT Medical Institute.
Acknowledgments
We thank Dr Mohammad Hosseine-Farid for helpful discussions.
Authors' Contributions
D.M.S. and M.M.A. supervised the work and wrote the paper. A.F.C. participated in writing and editing of the paper.
-
References
- 1 Ricard-Blum S. The collagen family. Cold Spring Harb Perspect Biol 2011; 3 (01) a004978
- 2 Shoulders MD, Raines RT. Collagen structure and stability. Annu Rev Biochem 2009; 78: 929-958
- 3 Fujii KK, Taga Y, Sakai T. et al. Lowering the culture temperature corrects collagen abnormalities caused by HSP47 gene knockout. Sci Rep 2019; 9 (01) 17433
- 4 Nagai N, Hosokawa M, Itohara S. et al. Embryonic lethality of molecular chaperone hsp47 knockout mice is associated with defects in collagen biosynthesis. J Cell Biol 2000; 150 (06) 1499-1506
- 5 Ito S, Nagata K. Roles of the endoplasmic reticulum-resident, collagen-specific molecular chaperone Hsp47 in vertebrate cells and human disease. J Biol Chem 2019; 294 (06) 2133-2141
- 6 Ito S, Nagata K. Biology of Hsp47 (Serpin H1), a collagen-specific molecular chaperone. Semin Cell Dev Biol 2017; 62: 142-151
- 7 Singh MK, Shin Y, Ju S. et al. Heat shock response and heat shock proteins: Current understanding and future opportunities in human diseases. Int J Mol Sci 2024; 25 (08) 4209
- 8 Niwa T, Kanamori T, Ueda T, Taguchi H. Global analysis of chaperone effects using a reconstituted cell-free translation system. Proc Natl Acad Sci U S A 2012; 109 (23) 8937-8942
- 9 Oecal S, Socher E, Uthoff M. et al. The pH-dependent client release from the collagen-specific chaperone HSP47 is triggered by a tandem histidine pair. J Biol Chem 2016; 291 (24) 12612-12626
- 10 Taguchi T, Razzaque MS. The collagen-specific molecular chaperone HSP47: is there a role in fibrosis?. Trends Mol Med 2007; 13 (02) 45-53
- 11 Bellaye P-S, Burgy O, Bonniaud P, Kolb M. HSP47: a potential target for fibrotic diseases and implications for therapy. Expert Opin Ther Targets 2021; 25 (01) 49-62
- 12 Kaiser WJ, Holbrook L-M, Tucker KL, Stanley RG, Gibbins JM. A functional proteomic method for the enrichment of peripheral membrane proteins reveals the collagen binding protein Hsp47 is exposed on the surface of activated human platelets. J Proteome Res 2009; 8 (06) 2903-2914
- 13 Sasikumar P, AlOuda KS, Kaiser WJ. et al. The chaperone protein HSP47: a platelet collagen binding protein that contributes to thrombosis and hemostasis. J Thromb Haemost 2018; 16 (05) 946-959
- 14 Thienel M, Müller-Reif JB, Zhang Z. et al. Immobility-associated thromboprotection is conserved across mammalian species from bear to human. Science 2023; 380 (6641): 178-187
- 15 Herbert A. Osteogenesis imperfecta type 10 and the cellular scaffolds underlying common immunological diseases. Genes Immun 2024; 25 (04) 265-276
- 16 Myllyharju J, Kivirikko KI. Collagens and collagen-related diseases. Ann Med 2001; 33 (01) 7-21
- 17 Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021; 18 (03) 151-166
- 18 Karsdal MA, Daniels SJ, Holm Nielsen S. et al. Collagen biology and non-invasive biomarkers of liver fibrosis. Liver Int 2020; 40 (04) 736-750
- 19 Staab-Weijnitz CA. Fighting the fiber: Targeting collagen in lung fibrosis. Am J Respir Cell Mol Biol 2022; 66 (04) 363-381
- 20 Bülow RD, Boor P. Extracellular matrix in kidney fibrosis: More than just a scaffold. J Histochem Cytochem 2019; 67 (09) 643-661
- 21 Eddy AA. Overview of the cellular and molecular basis of kidney fibrosis. Kidney Int Suppl (2011) 2014; 4 (01) 2-8
- 22 Zhang X, Zhang X, Huang W, Ge X. The role of heat shock proteins in the regulation of fibrotic diseases. Biomed Pharmacother 2021; 135: 111067
- 23 Miyamura T, Sakamoto N, Kakugawa T. et al. Small molecule inhibitor of HSP47 prevents pro-fibrotic mechanisms of fibroblasts in vitro. Biochem Biophys Res Commun 2020; 530 (03) 561-565
- 24 Bellaye P-S, Burgy O, Causse S, Garrido C, Bonniaud P. Heat shock proteins in fibrosis and wound healing: good or evil?. Pharmacol Ther 2014; 143 (02) 119-132
- 25 Ishii H, Mukae H, Kakugawa T. et al. Increased expression of collagen-binding heat shock protein 47 in murine bleomycin-induced pneumopathy. Am J Physiol Lung Cell Mol Physiol 2003; 285 (04) L957-L963
- 26 Kakugawa T, Mukae H, Hishikawa Y. et al. Localization of HSP47 mRNA in murine bleomycin-induced pulmonary fibrosis. Virchows Arch 2010; 456 (03) 309-315
- 27 Ham SY, Pyo MJ, Kang M. et al. HSP47 increases the expression of type I collagen in fibroblasts through IRE1α activation, XBP1 splicing, and nuclear translocation of β-catenin. Cells 2024; 13 (06) 527
- 28 Nakayama S, Mukae H, Sakamoto N. et al. Pirfenidone inhibits the expression of HSP47 in TGF-beta1-stimulated human lung fibroblasts. Life Sci 2008; 82 (3-4): 210-217
- 29 Hisatomi K, Mukae H, Sakamoto N. et al. Pirfenidone inhibits TGF-β1-induced over-expression of collagen type I and heat shock protein 47 in A549 cells. BMC Pulm Med 2012; 12: 24
- 30 Kakugawa T, Mukae H, Hayashi T. et al. Expression of HSP47 in usual interstitial pneumonia and nonspecific interstitial pneumonia. Respir Res 2005; 6 (01) 57
- 31 Kim DJ, Park SH, Sheen MR. et al. Comparison of experimental lung injury from acute renal failure with injury due to sepsis. Respiration 2006; 73 (06) 815-824
- 32 Razzaque MS, Nazneen A, Taguchi T. Immunolocalization of collagen and collagen-binding heat shock protein 47 in fibrotic lung diseases. Mod Pathol 1998; 11 (12) 1183-1188
- 33 Iwashita T, Kadota J, Naito S. et al. Involvement of collagen-binding heat shock protein 47 and procollagen type I synthesis in idiopathic pulmonary fibrosis: contribution of type II pneumocytes to fibrosis. Hum Pathol 2000; 31 (12) 1498-1505
- 34 Sakamoto N, Okuno D, Tokito T. et al. HSP47: A therapeutic target in pulmonary fibrosis. Biomedicines 2023; 11 (09) 2387
- 35 Yokota S, Kubota H, Matsuoka Y. et al. Prevalence of HSP47 antigen and autoantibodies to HSP47 in the sera of patients with mixed connective tissue disease. Biochem Biophys Res Commun 2003; 303 (02) 413-418
- 36 Hilberg F, Roth GJ, Krssak M. et al. BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 2008; 68 (12) 4774-4782
- 37 Hostettler KE, Zhong J, Papakonstantinou E. et al. Anti-fibrotic effects of nintedanib in lung fibroblasts derived from patients with idiopathic pulmonary fibrosis. Respir Res 2014; 15 (01) 157
- 38 Wollin L, Wex E, Pautsch A. et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45 (05) 1434-1445
- 39 Wollin L, Distler JHW, Redente EF. et al. Potential of nintedanib in treatment of progressive fibrosing interstitial lung diseases. Eur Respir J 2019; 54 (03) 1900161
- 40 Wollin L, Maillet I, Quesniaux V, Holweg A, Ryffel B. Antifibrotic and anti-inflammatory activity of the tyrosine kinase inhibitor nintedanib in experimental models of lung fibrosis. J Pharmacol Exp Ther 2014; 349 (02) 209-220
- 41 Knüppel L, Ishikawa Y, Aichler M. et al. A novel antifibrotic mechanism of nintedanib and pirfenidone. Inhibition of collagen fibril assembly. Am J Respir Cell Mol Biol 2017; 57 (01) 77-90
- 42 Lehmann M, Buhl L, Alsafadi HN. et al. Differential effects of Nintedanib and Pirfenidone on lung alveolar epithelial cell function in ex vivo murine and human lung tissue cultures of pulmonary fibrosis. Respir Res 2018; 19 (01) 175
- 43 Wu W, Qiu L, Wu J, Liu X, Zhang G. Efficacy and safety of pirfenidone in the treatment of idiopathic pulmonary fibrosis patients: a systematic review and meta-analysis of randomised controlled trials. BMJ Open 2021; 11 (12) e050004
- 44 Kalayarasan S, Sriram N, Sudhandiran G. Diallyl sulfide attenuates bleomycin-induced pulmonary fibrosis: critical role of iNOS, NF-kappaB, TNF-alpha and IL-1beta. Life Sci 2008; 82 (23-24): 1142-1153
- 45 Chen L, Wang T, Wang X. et al. Blockade of advanced glycation end product formation attenuates bleomycin-induced pulmonary fibrosis in rats. Respir Res 2009; 10 (01) 55
- 46 Liu J, Jin Z, Wang X, Jakoš T, Zhu J, Yuan Y. RAGE pathways play an important role in regulation of organ fibrosis. Life Sci 2023; 323: 121713
- 47 Goulet S, Bihl MP, Gambazzi F, Tamm M, Roth M. Opposite effect of corticosteroids and long-acting beta(2)-agonists on serum- and TGF-beta(1)-induced extracellular matrix deposition by primary human lung fibroblasts. J Cell Physiol 2007; 210 (01) 167-176
- 48 Vergoten G, Bailly C. Insights into the mechanism of action of the degraded limonoid prieurianin. Int J Mol Sci 2024; 25 (07) 3597
- 49 Satoh M, Hirayoshi K, Yokota S, Hosokawa N, Nagata K. Intracellular interaction of collagen-specific stress protein HSP47 with newly synthesized procollagen. J Cell Biol 1996; 133 (02) 469-483
- 50 Ko MK, Kay EP. Hsp47-dependent and -independent intracellular trafficking of type I collagen in corneal endothelial cells. Mol Vis 1999; 5: 17
- 51 Liu Y, Liu J, Quimbo A. et al. Anti-HSP47 siRNA lipid nanoparticle ND-L02-s0201 reverses interstitial pulmonary fibrosis in preclinical rat models. ERJ Open Res 2021; 7 (02) 00733-02020
- 52 Okuno D, Sakamoto N, Tagod MSO. et al. Screening of inhibitors targeting heat shock protein 47 involved in the development of idiopathic pulmonary fibrosis. ChemMedChem 2021; 16 (16) 2515-2523
- 53 Wu S, Liang C, Xie X. et al. Hsp47 inhibitor Col003 attenuates collagen-induced platelet activation and cerebral ischemic-reperfusion injury in rats. Front Pharmacol 2022; 12: 792263
- 54 Chen J-J, Jin P-S, Zhao S. et al. Effect of heat shock protein 47 on collagen synthesis of keloid in vivo. ANZ J Surg 2011; 81 (06) 425-430
- 55 Hagiwara S, Iwasaka H, Matsumoto S, Noguchi T. An antisense oligonucleotide to HSP47 inhibits paraquat-induced pulmonary fibrosis in rats. Toxicology 2007; 236 (03) 199-207
- 56 Hagiwara S, Iwasaka H, Matsumoto S, Noguchi T. Antisense oligonucleotide inhibition of heat shock protein (HSP) 47 improves bleomycin-induced pulmonary fibrosis in rats. Respir Res 2007; 8 (01) 37
- 57 Huang J-Q, Tao R, Li L. et al. Involvement of heat shock protein 47 in Schistosoma japonicum-induced hepatic fibrosis in mice. Int J Parasitol 2014; 44 (01) 23-35
- 58 Morry J, Ngamcherdtrakul W, Gu S. et al. Dermal delivery of HSP47 siRNA with NOX4-modulating mesoporous silica-based nanoparticles for treating fibrosis. Biomaterials 2015; 66: 41-52
- 59 Xia Z, Abe K, Furusu A. et al. Suppression of renal tubulointerstitial fibrosis by small interfering RNA targeting heat shock protein 47. Am J Nephrol 2008; 28 (01) 34-46
- 60 Obata Y, Nishino T, Kushibiki T. et al. HSP47 siRNA conjugated with cationized gelatin microspheres suppresses peritoneal fibrosis in mice. Acta Biomater 2012; 8 (07) 2688-2696
- 61 Sato Y, Murase K, Kato J. et al. Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat Biotechnol 2008; 26 (04) 431-442
- 62 Ishiwatari H, Sato Y, Murase K. et al. Treatment of pancreatic fibrosis with siRNA against a collagen-specific chaperone in vitamin A-coupled liposomes. Gut 2013; 62 (09) 1328-1339
- 63 Otsuka M, Shiratori M, Chiba H. et al. Treatment of pulmonary fibrosis with siRNA against a collagen-specific chaperone HSP47 in vitamin A-coupled liposomes. Exp Lung Res 2017; 43 (6-7): 271-282
- 64 Ohigashi H, Hashimoto D, Hayase E. et al. Ocular instillation of vitamin A-coupled liposomes containing HSP47 siRNA ameliorates dry eye syndrome in chronic GVHD. Blood Adv 2019; 3 (07) 1003-1010
- 65 Yamakawa T, Ohigashi H, Hashimoto D. et al. Vitamin A-coupled liposomes containing siRNA against HSP47 ameliorate skin fibrosis in chronic graft-versus-host disease. Blood 2018; 131 (13) 1476-1485
- 66 Zhu J, Xiong G, Fu H, Evers BM, Zhou BP, Xu R. Chaperone Hsp47 drives malignant growth and invasion by modulating an ECM gene network. Cancer Res 2015; 75 (08) 1580-1591
- 67 Mori K, Toiyama Y, Okugawa Y. et al. Preoperative heat shock protein 47 levels identify colorectal cancer patients with lymph node metastasis and poor prognosis. Oncol Lett 2020; 20 (06) 333
- 68 Mori K, Toiyama Y, Otake K. et al. Proteomics analysis of differential protein expression identifies heat shock protein 47 as a predictive marker for lymph node metastasis in patients with colorectal cancer. Int J Cancer 2017; 140 (06) 1425-1435
- 69 Huang X, Gollin SM, Raja S, Godfrey TE. High-resolution mapping of the 11q13 amplicon and identification of a gene, TAOS1, that is amplified and overexpressed in oral cancer cells. Proc Natl Acad Sci U S A 2002; 99 (17) 11369-11374
- 70 Shi R, Yu R, Lian F. et al. Targeting HSP47 for cancer treatment. Anticancer Drugs 2024; 35 (07) 623-637
- 71 Xiong G, Chen J, Zhang G. et al. Hsp47 promotes cancer metastasis by enhancing collagen-dependent cancer cell-platelet interaction. Proc Natl Acad Sci U S A 2020; 117 (07) 3748-3758
- 72 Tian S, Peng P, Li J. et al. SERPINH1 regulates EMT and gastric cancer metastasis via the Wnt/β-catenin signaling pathway. Aging (Albany NY) 2020; 12 (04) 3574-3593
- 73 Chen J, Wang S, Zhang Z, Richards CI, Xu R. Heat shock protein 47 (HSP47) binds to discoidin domain-containing receptor 2 (DDR2) and regulates its protein stability. J Biol Chem 2019; 294 (45) 16846-16854
- 74 Yoneda A, Minomi K, Tamura Y. HSP47 promotes metastasis of breast cancer by interacting with myosin IIA via the unfolded protein response transducer IRE1α. Oncogene 2020; 39 (23) 4519-4537
- 75 Wu ZB, Cai L, Lin SJ. et al. Heat shock protein 47 promotes glioma angiogenesis. Brain Pathol 2016; 26 (01) 31-42
- 76 Ma W, Ou T, Cui X. et al. HSP47 contributes to angiogenesis by induction of CCL2 in bladder cancer. Cell Signal 2021; 85: 110044
- 77 Recchia FM, Xu L. Differential expression of the collagen-binding protein Hsp47 in experimental retinal neovascularization. Invest Ophthalmol Vis Sci 2005; 46: 3162
- 78 Wendelboe A, Weitz JI. Global health burden of venous thromboembolism. Arterioscler Thromb Vasc Biol 2024; 44 (05) 1007-1011
- 79 Khan F, Tritschler T, Kahn SR, Rodger MA. Venous thromboembolism. Lancet 2021; 398 (10294): 64-77
- 80 AlOuda SK, Sasikumar P, AlThunayan T. et al. Role of heat shock protein 47 in platelet glycoprotein VI dimerization and signaling. Res Pract Thromb Haemost 2023; 7 (06) 102177
- 81 Crescente M, Pluthero FG, Li L. et al. Intracellular trafficking, localization, and mobilization of platelet-borne Thiol isomerases. Arterioscler Thromb Vasc Biol 2016; 36 (06) 1164-1173
- 82 Liu D, Razzaque MS, Cheng M, Taguchi T. The renal expression of heat shock protein 47 and collagens in acute and chronic experimental diabetes in rats. Histochem J 2001; 33 (11-12): 621-628
- 83 Manderstedt E, Lind-Halldén C, Halldén C. et al.; Regeneron Genetics Center. SERPINH1 variants and thrombotic risk among middle-aged and older adults: a population-based cohort study. J Thromb Haemost 2024; 22 (03) 869-873
- 84 Cai H, Sasikumar P, Little G. et al. Identification of HSP47 binding site on native collagen and its implications for the development of HSP47 inhibitors. Biomolecules 2021; 11 (07) 983
- 85 Charles S, Fatrara T, Bouriche T. et al. Tissue factor-bearing extracellular vesicles, procoagulant phospholipids and D-dimer as potential biomarkers for venous thromboembolism in patients with newly diagnosed multiple myeloma: A comprehensive analysis. Thromb Res 2025; 247: 109256
- 86 Lucotti S, Ogitani Y, Kenific CM. et al. Extracellular vesicles from the lung pro-thrombotic niche drive cancer-associated thrombosis and metastasis via integrin beta 2. Cell 2025; 188 (06) 1642-1661.e24
- 87 Shibata C, Otsuka M, Ishigaki K, Seimiya T, Kishikawa T, Fujishiro M. CA19-9-positive extracellular vesicle is a risk factor for cancer-associated thrombosis in pancreatic cancer. Gastro Hep Adv 2024; 3 (04) 551-561
- 88 Osorio LA, Lozano M, Soto P. et al. Levels of small extracellular vesicles containing hERG-1 and Hsp47 as potential biomarkers for cardiovascular diseases. Int J Mol Sci 2024; 25 (09) 4913
- 89 Kajikawa Y, Morihara T, Sakamoto H. et al. Platelet-rich plasma enhances the initial mobilization of circulation-derived cells for tendon healing. J Cell Physiol 2008; 215 (03) 837-845
- 90 Wu C-L, Lee S-S, Tsai C-H, Lu KH, Zhao JH, Chang YC. Platelet-rich fibrin increases cell attachment, proliferation and collagen-related protein expression of human osteoblasts. Aust Dent J 2012; 57 (02) 207-212
- 91 Junkiert-Czarnecka A, Pilarska-Deltow M, Bąk A, Heise M, Haus O. The role of gene encoding collagen secretion protein (SERPINH1) in the pathogenesis of a hypermobile type of Ehlers-Danlos syndrome. Postepy Dermatol Alergol 2023; 40 (01) 102-106
- 92 Smadja DM. Hyperthermia for targeting cancer and cancer stem cells: Insights from novel cellular and clinical approaches. Stem Cell Rev Rep 2024; 20 (06) 1532-1539
- 93 Mallory M, Gogineni E, Jones GC, Greer L, Simone II CB. Therapeutic hyperthermia: The old, the new, and the upcoming. Crit Rev Oncol Hematol 2016; 97: 56-64
- 94 Smadja DM, Abreu MM. Hyperthermia and targeting heat shock proteins: innovative approaches for neurodegenerative disorders and Long COVID. Front Neurosci 2025; 19: 1475376
- 95 Abreu MM, Chocron AF, Smadja DM. From cold to hot: mechanisms of hyperthermia in modulating tumor immunology for enhanced immunotherapy. Front Immunol 2025; 16: 1487296
- 96 Lukácsi S, Munkácsy G, Győrffy B. Harnessing hyperthermia: Molecular, cellular, and immunological insights for enhanced anticancer therapies. Integr Cancer Ther 2024; 23: 15 347354241242094
- 97 Sharma HS, Hoopes PJ. Hyperthermia induced pathophysiology of the central nervous system. Int J Hyperthermia 2003; 19 (03) 325-354
- 98 Beemelmanns A, Zanuzzo FS, Xue X, Sandrelli RM, Rise ML, Gamperl AK. The transcriptomic responses of Atlantic salmon (Salmo salar) to high temperature stress alone, and in combination with moderate hypoxia. BMC Genomics 2021; 22 (01) 261
- 99 Akbarzadeh A, Günther OP, Houde AL. et al. Developing specific molecular biomarkers for thermal stress in salmonids. BMC Genomics 2018; 19 (01) 749
- 100 Nagata K, Saga S, Yamada KM. A major collagen-binding protein of chick embryo fibroblasts is a novel heat shock protein. J Cell Biol 1986; 103 (01) 223-229
- 101 Verrico AK, Haylett AK, Moore JV. In vivo expression of the collagen-related heat shock protein HSP47, following hyperthermia or photodynamic therapy. Lasers Med Sci 2001; 16 (03) 192-198
- 102 Abdelnasir A, Sun JR, Cheng YF. et al. Evaluation of Hsp47 expression in heat-stressed rat myocardial cells in vitro and in vivo. Genet Mol Res 2014; 13 (04) 10787-10802
- 103 Dams SD, De Liefde-van Beest M, Nuijs AM. et al. Pulsed heat shocks enhance procollagen type I and procollagen type III expression in human dermal fibroblasts. Skin Res Technol 2010; 16: 354-364 . Accessed October 1, 2024 at: https://onlinelibrary.wiley.com/doi/10.1111/j.1600-0846.2010.00441.x
- 104 Haylett AK, Higley K, Chiu M, Shackley DC, Moore JV. Collagen secretion after photodynamic therapy versus scar-inducing anti-cancer modalities: an in vitro study. Photochem Photobiol Sci 2002; 1 (09) 673-677
- 105 Shackley DC, Haylett A, Whitehurst C. et al. Comparison of the cellular molecular stress responses after treatments used in bladder cancer. BJU Int 2002; 90 (09) 924-932
- 106 Verrico AK, Moore JV. Expression of the collagen-related heat shock protein HSP47 in fibroblasts treated with hyperthermia or photodynamic therapy. Br J Cancer 1997; 76 (06) 719-724
- 107 Miyaishi O, Ito Y, Kozaki K. et al. Age-related attenuation of HSP47 heat response in fibroblasts. Mech Ageing Dev 1995; 77 (03) 213-226
- 108 Tong M, Zhang S, Ma P. et al. Efficacy analysis of intermittent pneumatic compression combined with hyperthermia at different temperatures for prevention of deep vein thrombosis after simulated orthopaedic surgery in male rabbits. Am J Transl Res 2024; 16 (10) 5337-5346
- 109 Khan ES, Däinghaus T. HSP47 in human diseases: Navigating pathophysiology, diagnosis and therapy. Clin Transl Med 2024; 14 (08) e1755
- 110 Morito D, Nagata K. ER stress proteins in autoimmune and inflammatory diseases. Front Immunol 2012; 3: 48
- 111 Fujimoto M, Hamaguchi Y, Yazawa N, Komura K, Takehara K, Sato S. Autoantibodies to a collagen-specific molecular chaperone, heat-shock protein 47, in systemic sclerosis. Clin Exp Immunol 2004; 138 (03) 534-539
- 112 Kakugawa T, Yokota S, Mukae H. et al. High serum concentrations of autoantibodies to HSP47 in nonspecific interstitial pneumonia compared with idiopathic pulmonary fibrosis. BMC Pulm Med 2008; 8: 23
- 113 Izquierdo E, Cañete JD, Celis R. et al. Synovial fibroblast hyperplasia in rheumatoid arthritis: clinicopathologic correlations and partial reversal by anti-tumor necrosis factor therapy. Arthritis Rheum 2011; 63 (09) 2575-2583
- 114 Kalayarasan S, Sriram N, Sudhandiran G. Life Sci 2008; 82 (23-24): 1142-1153
- 115 Liu Y, Liu J, Quimbo A. et al Anti-HSP47 siRNA lipid nanoparticle ND-L02-s0201 reverses interstitial pulmonary fibrosis in preclinical rat models. ERJ Open Res 2021; 7 (02) 0733-2020
Address for correspondence
Publication History
Received: 05 February 2025
Accepted: 22 April 2025
Article published online:
05 June 2025
© 2025. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution License, permitting unrestricted use, distribution, and reproduction so long as the original work is properly cited. (https://creativecommons.org/licenses/by/4.0/)
Georg Thieme Verlag KG
Oswald-Hesse-Straße 50, 70469 Stuttgart, Germany
David M. Smadja, Alberto F. Chocron, M. Marc Abreu. HSP47 at the Crossroads of Thrombosis and Collagen Dynamics: Unlocking Therapeutic Horizons and Debates. TH Open 2025; 09: a25994925.
DOI: 10.1055/a-2599-4925
-
References
- 1 Ricard-Blum S. The collagen family. Cold Spring Harb Perspect Biol 2011; 3 (01) a004978
- 2 Shoulders MD, Raines RT. Collagen structure and stability. Annu Rev Biochem 2009; 78: 929-958
- 3 Fujii KK, Taga Y, Sakai T. et al. Lowering the culture temperature corrects collagen abnormalities caused by HSP47 gene knockout. Sci Rep 2019; 9 (01) 17433
- 4 Nagai N, Hosokawa M, Itohara S. et al. Embryonic lethality of molecular chaperone hsp47 knockout mice is associated with defects in collagen biosynthesis. J Cell Biol 2000; 150 (06) 1499-1506
- 5 Ito S, Nagata K. Roles of the endoplasmic reticulum-resident, collagen-specific molecular chaperone Hsp47 in vertebrate cells and human disease. J Biol Chem 2019; 294 (06) 2133-2141
- 6 Ito S, Nagata K. Biology of Hsp47 (Serpin H1), a collagen-specific molecular chaperone. Semin Cell Dev Biol 2017; 62: 142-151
- 7 Singh MK, Shin Y, Ju S. et al. Heat shock response and heat shock proteins: Current understanding and future opportunities in human diseases. Int J Mol Sci 2024; 25 (08) 4209
- 8 Niwa T, Kanamori T, Ueda T, Taguchi H. Global analysis of chaperone effects using a reconstituted cell-free translation system. Proc Natl Acad Sci U S A 2012; 109 (23) 8937-8942
- 9 Oecal S, Socher E, Uthoff M. et al. The pH-dependent client release from the collagen-specific chaperone HSP47 is triggered by a tandem histidine pair. J Biol Chem 2016; 291 (24) 12612-12626
- 10 Taguchi T, Razzaque MS. The collagen-specific molecular chaperone HSP47: is there a role in fibrosis?. Trends Mol Med 2007; 13 (02) 45-53
- 11 Bellaye P-S, Burgy O, Bonniaud P, Kolb M. HSP47: a potential target for fibrotic diseases and implications for therapy. Expert Opin Ther Targets 2021; 25 (01) 49-62
- 12 Kaiser WJ, Holbrook L-M, Tucker KL, Stanley RG, Gibbins JM. A functional proteomic method for the enrichment of peripheral membrane proteins reveals the collagen binding protein Hsp47 is exposed on the surface of activated human platelets. J Proteome Res 2009; 8 (06) 2903-2914
- 13 Sasikumar P, AlOuda KS, Kaiser WJ. et al. The chaperone protein HSP47: a platelet collagen binding protein that contributes to thrombosis and hemostasis. J Thromb Haemost 2018; 16 (05) 946-959
- 14 Thienel M, Müller-Reif JB, Zhang Z. et al. Immobility-associated thromboprotection is conserved across mammalian species from bear to human. Science 2023; 380 (6641): 178-187
- 15 Herbert A. Osteogenesis imperfecta type 10 and the cellular scaffolds underlying common immunological diseases. Genes Immun 2024; 25 (04) 265-276
- 16 Myllyharju J, Kivirikko KI. Collagens and collagen-related diseases. Ann Med 2001; 33 (01) 7-21
- 17 Kisseleva T, Brenner D. Molecular and cellular mechanisms of liver fibrosis and its regression. Nat Rev Gastroenterol Hepatol 2021; 18 (03) 151-166
- 18 Karsdal MA, Daniels SJ, Holm Nielsen S. et al. Collagen biology and non-invasive biomarkers of liver fibrosis. Liver Int 2020; 40 (04) 736-750
- 19 Staab-Weijnitz CA. Fighting the fiber: Targeting collagen in lung fibrosis. Am J Respir Cell Mol Biol 2022; 66 (04) 363-381
- 20 Bülow RD, Boor P. Extracellular matrix in kidney fibrosis: More than just a scaffold. J Histochem Cytochem 2019; 67 (09) 643-661
- 21 Eddy AA. Overview of the cellular and molecular basis of kidney fibrosis. Kidney Int Suppl (2011) 2014; 4 (01) 2-8
- 22 Zhang X, Zhang X, Huang W, Ge X. The role of heat shock proteins in the regulation of fibrotic diseases. Biomed Pharmacother 2021; 135: 111067
- 23 Miyamura T, Sakamoto N, Kakugawa T. et al. Small molecule inhibitor of HSP47 prevents pro-fibrotic mechanisms of fibroblasts in vitro. Biochem Biophys Res Commun 2020; 530 (03) 561-565
- 24 Bellaye P-S, Burgy O, Causse S, Garrido C, Bonniaud P. Heat shock proteins in fibrosis and wound healing: good or evil?. Pharmacol Ther 2014; 143 (02) 119-132
- 25 Ishii H, Mukae H, Kakugawa T. et al. Increased expression of collagen-binding heat shock protein 47 in murine bleomycin-induced pneumopathy. Am J Physiol Lung Cell Mol Physiol 2003; 285 (04) L957-L963
- 26 Kakugawa T, Mukae H, Hishikawa Y. et al. Localization of HSP47 mRNA in murine bleomycin-induced pulmonary fibrosis. Virchows Arch 2010; 456 (03) 309-315
- 27 Ham SY, Pyo MJ, Kang M. et al. HSP47 increases the expression of type I collagen in fibroblasts through IRE1α activation, XBP1 splicing, and nuclear translocation of β-catenin. Cells 2024; 13 (06) 527
- 28 Nakayama S, Mukae H, Sakamoto N. et al. Pirfenidone inhibits the expression of HSP47 in TGF-beta1-stimulated human lung fibroblasts. Life Sci 2008; 82 (3-4): 210-217
- 29 Hisatomi K, Mukae H, Sakamoto N. et al. Pirfenidone inhibits TGF-β1-induced over-expression of collagen type I and heat shock protein 47 in A549 cells. BMC Pulm Med 2012; 12: 24
- 30 Kakugawa T, Mukae H, Hayashi T. et al. Expression of HSP47 in usual interstitial pneumonia and nonspecific interstitial pneumonia. Respir Res 2005; 6 (01) 57
- 31 Kim DJ, Park SH, Sheen MR. et al. Comparison of experimental lung injury from acute renal failure with injury due to sepsis. Respiration 2006; 73 (06) 815-824
- 32 Razzaque MS, Nazneen A, Taguchi T. Immunolocalization of collagen and collagen-binding heat shock protein 47 in fibrotic lung diseases. Mod Pathol 1998; 11 (12) 1183-1188
- 33 Iwashita T, Kadota J, Naito S. et al. Involvement of collagen-binding heat shock protein 47 and procollagen type I synthesis in idiopathic pulmonary fibrosis: contribution of type II pneumocytes to fibrosis. Hum Pathol 2000; 31 (12) 1498-1505
- 34 Sakamoto N, Okuno D, Tokito T. et al. HSP47: A therapeutic target in pulmonary fibrosis. Biomedicines 2023; 11 (09) 2387
- 35 Yokota S, Kubota H, Matsuoka Y. et al. Prevalence of HSP47 antigen and autoantibodies to HSP47 in the sera of patients with mixed connective tissue disease. Biochem Biophys Res Commun 2003; 303 (02) 413-418
- 36 Hilberg F, Roth GJ, Krssak M. et al. BIBF 1120: triple angiokinase inhibitor with sustained receptor blockade and good antitumor efficacy. Cancer Res 2008; 68 (12) 4774-4782
- 37 Hostettler KE, Zhong J, Papakonstantinou E. et al. Anti-fibrotic effects of nintedanib in lung fibroblasts derived from patients with idiopathic pulmonary fibrosis. Respir Res 2014; 15 (01) 157
- 38 Wollin L, Wex E, Pautsch A. et al. Mode of action of nintedanib in the treatment of idiopathic pulmonary fibrosis. Eur Respir J 2015; 45 (05) 1434-1445
- 39 Wollin L, Distler JHW, Redente EF. et al. Potential of nintedanib in treatment of progressive fibrosing interstitial lung diseases. Eur Respir J 2019; 54 (03) 1900161
- 40 Wollin L, Maillet I, Quesniaux V, Holweg A, Ryffel B. Antifibrotic and anti-inflammatory activity of the tyrosine kinase inhibitor nintedanib in experimental models of lung fibrosis. J Pharmacol Exp Ther 2014; 349 (02) 209-220
- 41 Knüppel L, Ishikawa Y, Aichler M. et al. A novel antifibrotic mechanism of nintedanib and pirfenidone. Inhibition of collagen fibril assembly. Am J Respir Cell Mol Biol 2017; 57 (01) 77-90
- 42 Lehmann M, Buhl L, Alsafadi HN. et al. Differential effects of Nintedanib and Pirfenidone on lung alveolar epithelial cell function in ex vivo murine and human lung tissue cultures of pulmonary fibrosis. Respir Res 2018; 19 (01) 175
- 43 Wu W, Qiu L, Wu J, Liu X, Zhang G. Efficacy and safety of pirfenidone in the treatment of idiopathic pulmonary fibrosis patients: a systematic review and meta-analysis of randomised controlled trials. BMJ Open 2021; 11 (12) e050004
- 44 Kalayarasan S, Sriram N, Sudhandiran G. Diallyl sulfide attenuates bleomycin-induced pulmonary fibrosis: critical role of iNOS, NF-kappaB, TNF-alpha and IL-1beta. Life Sci 2008; 82 (23-24): 1142-1153
- 45 Chen L, Wang T, Wang X. et al. Blockade of advanced glycation end product formation attenuates bleomycin-induced pulmonary fibrosis in rats. Respir Res 2009; 10 (01) 55
- 46 Liu J, Jin Z, Wang X, Jakoš T, Zhu J, Yuan Y. RAGE pathways play an important role in regulation of organ fibrosis. Life Sci 2023; 323: 121713
- 47 Goulet S, Bihl MP, Gambazzi F, Tamm M, Roth M. Opposite effect of corticosteroids and long-acting beta(2)-agonists on serum- and TGF-beta(1)-induced extracellular matrix deposition by primary human lung fibroblasts. J Cell Physiol 2007; 210 (01) 167-176
- 48 Vergoten G, Bailly C. Insights into the mechanism of action of the degraded limonoid prieurianin. Int J Mol Sci 2024; 25 (07) 3597
- 49 Satoh M, Hirayoshi K, Yokota S, Hosokawa N, Nagata K. Intracellular interaction of collagen-specific stress protein HSP47 with newly synthesized procollagen. J Cell Biol 1996; 133 (02) 469-483
- 50 Ko MK, Kay EP. Hsp47-dependent and -independent intracellular trafficking of type I collagen in corneal endothelial cells. Mol Vis 1999; 5: 17
- 51 Liu Y, Liu J, Quimbo A. et al. Anti-HSP47 siRNA lipid nanoparticle ND-L02-s0201 reverses interstitial pulmonary fibrosis in preclinical rat models. ERJ Open Res 2021; 7 (02) 00733-02020
- 52 Okuno D, Sakamoto N, Tagod MSO. et al. Screening of inhibitors targeting heat shock protein 47 involved in the development of idiopathic pulmonary fibrosis. ChemMedChem 2021; 16 (16) 2515-2523
- 53 Wu S, Liang C, Xie X. et al. Hsp47 inhibitor Col003 attenuates collagen-induced platelet activation and cerebral ischemic-reperfusion injury in rats. Front Pharmacol 2022; 12: 792263
- 54 Chen J-J, Jin P-S, Zhao S. et al. Effect of heat shock protein 47 on collagen synthesis of keloid in vivo. ANZ J Surg 2011; 81 (06) 425-430
- 55 Hagiwara S, Iwasaka H, Matsumoto S, Noguchi T. An antisense oligonucleotide to HSP47 inhibits paraquat-induced pulmonary fibrosis in rats. Toxicology 2007; 236 (03) 199-207
- 56 Hagiwara S, Iwasaka H, Matsumoto S, Noguchi T. Antisense oligonucleotide inhibition of heat shock protein (HSP) 47 improves bleomycin-induced pulmonary fibrosis in rats. Respir Res 2007; 8 (01) 37
- 57 Huang J-Q, Tao R, Li L. et al. Involvement of heat shock protein 47 in Schistosoma japonicum-induced hepatic fibrosis in mice. Int J Parasitol 2014; 44 (01) 23-35
- 58 Morry J, Ngamcherdtrakul W, Gu S. et al. Dermal delivery of HSP47 siRNA with NOX4-modulating mesoporous silica-based nanoparticles for treating fibrosis. Biomaterials 2015; 66: 41-52
- 59 Xia Z, Abe K, Furusu A. et al. Suppression of renal tubulointerstitial fibrosis by small interfering RNA targeting heat shock protein 47. Am J Nephrol 2008; 28 (01) 34-46
- 60 Obata Y, Nishino T, Kushibiki T. et al. HSP47 siRNA conjugated with cationized gelatin microspheres suppresses peritoneal fibrosis in mice. Acta Biomater 2012; 8 (07) 2688-2696
- 61 Sato Y, Murase K, Kato J. et al. Resolution of liver cirrhosis using vitamin A-coupled liposomes to deliver siRNA against a collagen-specific chaperone. Nat Biotechnol 2008; 26 (04) 431-442
- 62 Ishiwatari H, Sato Y, Murase K. et al. Treatment of pancreatic fibrosis with siRNA against a collagen-specific chaperone in vitamin A-coupled liposomes. Gut 2013; 62 (09) 1328-1339
- 63 Otsuka M, Shiratori M, Chiba H. et al. Treatment of pulmonary fibrosis with siRNA against a collagen-specific chaperone HSP47 in vitamin A-coupled liposomes. Exp Lung Res 2017; 43 (6-7): 271-282
- 64 Ohigashi H, Hashimoto D, Hayase E. et al. Ocular instillation of vitamin A-coupled liposomes containing HSP47 siRNA ameliorates dry eye syndrome in chronic GVHD. Blood Adv 2019; 3 (07) 1003-1010
- 65 Yamakawa T, Ohigashi H, Hashimoto D. et al. Vitamin A-coupled liposomes containing siRNA against HSP47 ameliorate skin fibrosis in chronic graft-versus-host disease. Blood 2018; 131 (13) 1476-1485
- 66 Zhu J, Xiong G, Fu H, Evers BM, Zhou BP, Xu R. Chaperone Hsp47 drives malignant growth and invasion by modulating an ECM gene network. Cancer Res 2015; 75 (08) 1580-1591
- 67 Mori K, Toiyama Y, Okugawa Y. et al. Preoperative heat shock protein 47 levels identify colorectal cancer patients with lymph node metastasis and poor prognosis. Oncol Lett 2020; 20 (06) 333
- 68 Mori K, Toiyama Y, Otake K. et al. Proteomics analysis of differential protein expression identifies heat shock protein 47 as a predictive marker for lymph node metastasis in patients with colorectal cancer. Int J Cancer 2017; 140 (06) 1425-1435
- 69 Huang X, Gollin SM, Raja S, Godfrey TE. High-resolution mapping of the 11q13 amplicon and identification of a gene, TAOS1, that is amplified and overexpressed in oral cancer cells. Proc Natl Acad Sci U S A 2002; 99 (17) 11369-11374
- 70 Shi R, Yu R, Lian F. et al. Targeting HSP47 for cancer treatment. Anticancer Drugs 2024; 35 (07) 623-637
- 71 Xiong G, Chen J, Zhang G. et al. Hsp47 promotes cancer metastasis by enhancing collagen-dependent cancer cell-platelet interaction. Proc Natl Acad Sci U S A 2020; 117 (07) 3748-3758
- 72 Tian S, Peng P, Li J. et al. SERPINH1 regulates EMT and gastric cancer metastasis via the Wnt/β-catenin signaling pathway. Aging (Albany NY) 2020; 12 (04) 3574-3593
- 73 Chen J, Wang S, Zhang Z, Richards CI, Xu R. Heat shock protein 47 (HSP47) binds to discoidin domain-containing receptor 2 (DDR2) and regulates its protein stability. J Biol Chem 2019; 294 (45) 16846-16854
- 74 Yoneda A, Minomi K, Tamura Y. HSP47 promotes metastasis of breast cancer by interacting with myosin IIA via the unfolded protein response transducer IRE1α. Oncogene 2020; 39 (23) 4519-4537
- 75 Wu ZB, Cai L, Lin SJ. et al. Heat shock protein 47 promotes glioma angiogenesis. Brain Pathol 2016; 26 (01) 31-42
- 76 Ma W, Ou T, Cui X. et al. HSP47 contributes to angiogenesis by induction of CCL2 in bladder cancer. Cell Signal 2021; 85: 110044
- 77 Recchia FM, Xu L. Differential expression of the collagen-binding protein Hsp47 in experimental retinal neovascularization. Invest Ophthalmol Vis Sci 2005; 46: 3162
- 78 Wendelboe A, Weitz JI. Global health burden of venous thromboembolism. Arterioscler Thromb Vasc Biol 2024; 44 (05) 1007-1011
- 79 Khan F, Tritschler T, Kahn SR, Rodger MA. Venous thromboembolism. Lancet 2021; 398 (10294): 64-77
- 80 AlOuda SK, Sasikumar P, AlThunayan T. et al. Role of heat shock protein 47 in platelet glycoprotein VI dimerization and signaling. Res Pract Thromb Haemost 2023; 7 (06) 102177
- 81 Crescente M, Pluthero FG, Li L. et al. Intracellular trafficking, localization, and mobilization of platelet-borne Thiol isomerases. Arterioscler Thromb Vasc Biol 2016; 36 (06) 1164-1173
- 82 Liu D, Razzaque MS, Cheng M, Taguchi T. The renal expression of heat shock protein 47 and collagens in acute and chronic experimental diabetes in rats. Histochem J 2001; 33 (11-12): 621-628
- 83 Manderstedt E, Lind-Halldén C, Halldén C. et al.; Regeneron Genetics Center. SERPINH1 variants and thrombotic risk among middle-aged and older adults: a population-based cohort study. J Thromb Haemost 2024; 22 (03) 869-873
- 84 Cai H, Sasikumar P, Little G. et al. Identification of HSP47 binding site on native collagen and its implications for the development of HSP47 inhibitors. Biomolecules 2021; 11 (07) 983
- 85 Charles S, Fatrara T, Bouriche T. et al. Tissue factor-bearing extracellular vesicles, procoagulant phospholipids and D-dimer as potential biomarkers for venous thromboembolism in patients with newly diagnosed multiple myeloma: A comprehensive analysis. Thromb Res 2025; 247: 109256
- 86 Lucotti S, Ogitani Y, Kenific CM. et al. Extracellular vesicles from the lung pro-thrombotic niche drive cancer-associated thrombosis and metastasis via integrin beta 2. Cell 2025; 188 (06) 1642-1661.e24
- 87 Shibata C, Otsuka M, Ishigaki K, Seimiya T, Kishikawa T, Fujishiro M. CA19-9-positive extracellular vesicle is a risk factor for cancer-associated thrombosis in pancreatic cancer. Gastro Hep Adv 2024; 3 (04) 551-561
- 88 Osorio LA, Lozano M, Soto P. et al. Levels of small extracellular vesicles containing hERG-1 and Hsp47 as potential biomarkers for cardiovascular diseases. Int J Mol Sci 2024; 25 (09) 4913
- 89 Kajikawa Y, Morihara T, Sakamoto H. et al. Platelet-rich plasma enhances the initial mobilization of circulation-derived cells for tendon healing. J Cell Physiol 2008; 215 (03) 837-845
- 90 Wu C-L, Lee S-S, Tsai C-H, Lu KH, Zhao JH, Chang YC. Platelet-rich fibrin increases cell attachment, proliferation and collagen-related protein expression of human osteoblasts. Aust Dent J 2012; 57 (02) 207-212
- 91 Junkiert-Czarnecka A, Pilarska-Deltow M, Bąk A, Heise M, Haus O. The role of gene encoding collagen secretion protein (SERPINH1) in the pathogenesis of a hypermobile type of Ehlers-Danlos syndrome. Postepy Dermatol Alergol 2023; 40 (01) 102-106
- 92 Smadja DM. Hyperthermia for targeting cancer and cancer stem cells: Insights from novel cellular and clinical approaches. Stem Cell Rev Rep 2024; 20 (06) 1532-1539
- 93 Mallory M, Gogineni E, Jones GC, Greer L, Simone II CB. Therapeutic hyperthermia: The old, the new, and the upcoming. Crit Rev Oncol Hematol 2016; 97: 56-64
- 94 Smadja DM, Abreu MM. Hyperthermia and targeting heat shock proteins: innovative approaches for neurodegenerative disorders and Long COVID. Front Neurosci 2025; 19: 1475376
- 95 Abreu MM, Chocron AF, Smadja DM. From cold to hot: mechanisms of hyperthermia in modulating tumor immunology for enhanced immunotherapy. Front Immunol 2025; 16: 1487296
- 96 Lukácsi S, Munkácsy G, Győrffy B. Harnessing hyperthermia: Molecular, cellular, and immunological insights for enhanced anticancer therapies. Integr Cancer Ther 2024; 23: 15 347354241242094
- 97 Sharma HS, Hoopes PJ. Hyperthermia induced pathophysiology of the central nervous system. Int J Hyperthermia 2003; 19 (03) 325-354
- 98 Beemelmanns A, Zanuzzo FS, Xue X, Sandrelli RM, Rise ML, Gamperl AK. The transcriptomic responses of Atlantic salmon (Salmo salar) to high temperature stress alone, and in combination with moderate hypoxia. BMC Genomics 2021; 22 (01) 261
- 99 Akbarzadeh A, Günther OP, Houde AL. et al. Developing specific molecular biomarkers for thermal stress in salmonids. BMC Genomics 2018; 19 (01) 749
- 100 Nagata K, Saga S, Yamada KM. A major collagen-binding protein of chick embryo fibroblasts is a novel heat shock protein. J Cell Biol 1986; 103 (01) 223-229
- 101 Verrico AK, Haylett AK, Moore JV. In vivo expression of the collagen-related heat shock protein HSP47, following hyperthermia or photodynamic therapy. Lasers Med Sci 2001; 16 (03) 192-198
- 102 Abdelnasir A, Sun JR, Cheng YF. et al. Evaluation of Hsp47 expression in heat-stressed rat myocardial cells in vitro and in vivo. Genet Mol Res 2014; 13 (04) 10787-10802
- 103 Dams SD, De Liefde-van Beest M, Nuijs AM. et al. Pulsed heat shocks enhance procollagen type I and procollagen type III expression in human dermal fibroblasts. Skin Res Technol 2010; 16: 354-364 . Accessed October 1, 2024 at: https://onlinelibrary.wiley.com/doi/10.1111/j.1600-0846.2010.00441.x
- 104 Haylett AK, Higley K, Chiu M, Shackley DC, Moore JV. Collagen secretion after photodynamic therapy versus scar-inducing anti-cancer modalities: an in vitro study. Photochem Photobiol Sci 2002; 1 (09) 673-677
- 105 Shackley DC, Haylett A, Whitehurst C. et al. Comparison of the cellular molecular stress responses after treatments used in bladder cancer. BJU Int 2002; 90 (09) 924-932
- 106 Verrico AK, Moore JV. Expression of the collagen-related heat shock protein HSP47 in fibroblasts treated with hyperthermia or photodynamic therapy. Br J Cancer 1997; 76 (06) 719-724
- 107 Miyaishi O, Ito Y, Kozaki K. et al. Age-related attenuation of HSP47 heat response in fibroblasts. Mech Ageing Dev 1995; 77 (03) 213-226
- 108 Tong M, Zhang S, Ma P. et al. Efficacy analysis of intermittent pneumatic compression combined with hyperthermia at different temperatures for prevention of deep vein thrombosis after simulated orthopaedic surgery in male rabbits. Am J Transl Res 2024; 16 (10) 5337-5346
- 109 Khan ES, Däinghaus T. HSP47 in human diseases: Navigating pathophysiology, diagnosis and therapy. Clin Transl Med 2024; 14 (08) e1755
- 110 Morito D, Nagata K. ER stress proteins in autoimmune and inflammatory diseases. Front Immunol 2012; 3: 48
- 111 Fujimoto M, Hamaguchi Y, Yazawa N, Komura K, Takehara K, Sato S. Autoantibodies to a collagen-specific molecular chaperone, heat-shock protein 47, in systemic sclerosis. Clin Exp Immunol 2004; 138 (03) 534-539
- 112 Kakugawa T, Yokota S, Mukae H. et al. High serum concentrations of autoantibodies to HSP47 in nonspecific interstitial pneumonia compared with idiopathic pulmonary fibrosis. BMC Pulm Med 2008; 8: 23
- 113 Izquierdo E, Cañete JD, Celis R. et al. Synovial fibroblast hyperplasia in rheumatoid arthritis: clinicopathologic correlations and partial reversal by anti-tumor necrosis factor therapy. Arthritis Rheum 2011; 63 (09) 2575-2583
- 114 Kalayarasan S, Sriram N, Sudhandiran G. Life Sci 2008; 82 (23-24): 1142-1153
- 115 Liu Y, Liu J, Quimbo A. et al Anti-HSP47 siRNA lipid nanoparticle ND-L02-s0201 reverses interstitial pulmonary fibrosis in preclinical rat models. ERJ Open Res 2021; 7 (02) 0733-2020



