Abstract
The extracellular matrix is an active participant, modulator and mediator of the cell, tissue, organ and organismal response to injury. Recent research has highlighted the role of hyaluronan, an abundant glycosaminoglycan constituent of the extracellular matrix, in many fundamental biological processes underpinning homeostasis and disease development. From this basis, emerging studies have demonstrated the therapeutic potential of strategies which target hyaluronan synthesis, biology and signaling, with significant promise as therapeutics for a variety of inflammatory and immune diseases. This review summarizes the state of the art in this field and discusses challenges and opportunities in what could emerge as a new class of therapeutic agents, that we term “matrix biologics”.
Keywords: hyaluronan, hyaladherins, signaling, treatment
1. Introduction
The extracellular matrix (ECM) is not simply an inert filler of intercellular space; on the contrary, the ECM is an active participant, modulator, and mediator of cell fate, signaling and injury response. In a way, the ECM and the cells constitute a microscopic “host-environment” unit which ultimately dictates the macroscopically observed host-environment interactions that can be observed after injury and in disease processes. ECM biology is complex, derived in part from a dynamic mix of ECM components. One such prominent ECM component is hyaluronan (HA), a ubiquitously expressed glycosaminoglycan sugar. HA structure is deceptively simple, consisting of repeating disaccharide chains of N-acetyl-glucosamine and glucuronic acid, however recent research has shed light on its fascinatingly complex biology and has strongly suggested that HA plays a central role in fundamental biological processes that ultimately dictate homeostasis or disease development. Such processes include cell and organ development, tissue injury response, inflammation (including autoimmunity), cell migration, cancer formation and cancer resistance. Recent research has shed considerable light into the mechanistic effects of HA biology, which seems to be regulated by four major mechanisms: 1) physical properties, size and molecular weight of HA; 2) HA binding partners in a matrix and the formation of a macromolecular structure; 3) HA metabolism (synthesis and degradation); and 4) HA receptor signaling (1). These insights, in turn, have enabled the translation of HA biology into the human health arena, including the ability to intelligently design or modify HA molecules to specific therapeutic ends.
There are obvious benefits to using HA in medicine: it is naturally occurring, easily biodegradable, non-antigenic and highly hydrophilic. Consequently, HA has been in wide use in cosmetics (dermal filler), orthopedics (intra-articular injections in degenerative disease) and ophthalmology (as a filler of the corpus vitreum in eye surgery) (2–8). However, most of these applications are utilizing the biocompatibility and physical properties of HA. In this review, we will advance the argument that HA-related applications can be vastly expanded, taking advantage of HA biological (as opposed to, or in addition to its physical) properties. Furthermore, approaches that modify HA biology (e.g. synthesis, interaction with other proteins) can also be viewed as belonging to the same realm of pharmaceuticals. We will give examples of recent experimental evidence of the clinical utility of such approaches and highlight opportunities for further development in the future.
2. Mechanistic background
The following chapters will address the basic mechanistic steps of HA biology which can be targeted in a pharmacological setting: HA synthesis and degradation; HA association with extracellular proteins to form a HA matrix; and HA signaling.
2.1. Hyaluronan synthesis and metabolism
HA is synthesized at the plasma membrane (9) by one of three isozymes, Hyaluronan synthase (HAS) 1, 2, and 3. HAS are evolutionarily conserved, highly homologous (55–70% protein identity) (10, 11), and catalyze the addition of UDP-D-glucuronic acid (GlcA) and UDP-N-acetyl-D-glucosamine (GlcNAC) monomers in an alternating assembly to form HA (12, 13). HAS isoforms are different in their half-life, rate of HA synthesis, affinity for HA substrates (14) and molecular weight of synthesized HA: HAS1 and HAS2 synthesize larger polymers (2×105 to 2×106 Da, HAS1 product size being somewhat lower than HAS2), while HAS3 synthesized the shortest HA polymer sizes (1×105 to 1×106 Da) (14). It is still not clear how mammalian HAS regulate the length of their HA product, in contrast to bacterial HAS, where the length of the final product depends on the availability and stoichiometry of UDP-sugar precursors (15). In any case, HAS enzymes are the first modulator of HA biological activity, by way of producing HA of different sizes, which may have different biological functions, as we will see below.
At the tissue level, HAS gene expression and subsequent HA synthesis is regulated by a wide range of cytokines and growth factors (16, 17). HAS genes may respond similarly or differentially to a particular signal; for example, TGF-β1 induces downregulation of HAS3 but upregulation of HAS1 expression (18). Furthermore, cell- or tissue-specific HAS expression may lead to opposing effects. For example, HAS2 expression in airway epithelia leads to resistance against injury and decrease in experimental fibrosis (19, 20), while HAS2 expression in fibroblasts promotes fibrosis (21, 22). HA synthesis is also under tight metabolic control. For example, the half-life time of HAS2 is only 17 min; however, after the post-translational ligation of a single O-linked-β-N-acetylglucosamine (O-GlcNAc) on the hydroxyl group of its serine 221 residue, HAS2 half-life time is extended substantially to >5 hours (23, 24), thus linking HA synthesis to metabolic processes in the cell and the organism, like hyperglycemia (23). Conversely, HAS2 can be inhibited by phosphorylation by the AMP-activated protein kinase, a master metabolic regulator activated by low ATP/AMP ratios, and epigenetically by sirtuin 1, (SIRT1) another important energetic sensor. The SIRT1 action is very interesting and intricate, in that it suppreses expression of an antisense nucleotide (HAS2-Antisense1), which is known to stabilize HAS2 mRNA and induce HAS upregulation. Therefore, decreased HAS2-AS1 leads to HAS2 downregulation (25). In aggregate, mounting evidence points towards an intricate and complex regulatory network that can influence HA synthesis, underscoring the importance of this molecule for biological processes.
Experimental evidence suggests that HAS genes play important roles in disease and injury. In the naked mole rat, expression of a particularly high molecular weight HA by HAS2 was shown to be the reason behind the longevity and cancer resistance of this rodent (26). In cancer, overexpression of HAS influences tumor growth, metastatic potential, and progression in prostate, colon, breast, and endometrial tumors (27). New evidence suggests that during cancer-induced tissue remodeling, an abundant HA matrix is produced, based on a dramatic metabolic reprogramming of cancel cells, with overabundance of HA substrate saccharides and overexpression of HAS, particularly HAS2 (reviewed in (28). Ectopic expression of HAS genes may also functionally alter the biological responses of cells to injury in vivo (19, 21). Thus, HAS activity may have opposing effects dependent on context and tissue. HAS activity can be chemically inhibited (29, 30) and this can be utilized for therapeutic purposes, as we will see below.
HA turnover is achieved either non-enzymatically or enzymatically. Reactive oxygen species such as superoxide, hydrogen peroxide, nitric oxide, peroxynitrite, and hypohalous acids, either generated during inflammation or introduced via environmental exposures, degrade HA (31, 32). Enzymatically, hyaluronidases specifically induce HA digestion. Several HA degrading enzymes exist in mammals: hyaluronidases 1–3 (HYAL1–3), PH20 (33, 34), the newly discovered CEMIP, also known as KIAA1199 (which may not be directly degrading HA but facilitate HA uptake leading to degradation (35)) and CEMIP2, also known as TMEM2 (36–39). HYALs are highly homologous endoglycosidases, which hydrolyze the β−1,4 linkage of the HA molecule, which is a linear polysaccharide composed of repeating β−1,4-linked D-glucuronic acid (GlcA) and β−1,3-linked N-acetyl-D-glucosamine (GlcNAc) disaccharide units (34, 40, 41). HYAL activity is regulated by environmental cues, such as acidity: for example, HYAL1–3 are primarily active at an acidic pH supporting their activity in lysosomes, while PH20 optimum activity is at a neutral pH (33, 42) and TMEM2 activity is at pH5–8 (39). TMEM2 is found in all organs, HYAL2 is found in heart, skeletal muscle, colon, spleen, kidney, liver, placenta, and lung, while HYAL1 is more limited in scope but is high in liver (which is a primary location of HA degradation), while also found in plasma and urine. PH20 is specific to the acrosome, an organelle of sperm cells and has a role in fertilization (33). HA degradation is accomplished by TMEM2, HYAL1 and HYAL2 acting in concert. TMEM2 and HYAL2 appear to be active on the cell membrane, possibly in an organ- and cell-specific fashion. The acidic environment necessary for HYAL2 activity is provided by NHE2 (43). Thus generated HA fragments are internalized into endolysosomes, where HYAL1 further degrades the HA into tetrasaccharides (44).
Perturbations in HA catabolism have significant biological effects. Hyaluronidase deficiency in humans leads to elevated HA levels in plasma (45) and a lysosomal storage disorder called mucopolysaccharidosis IX characterized by accumulation of HA, short stature, and multiple soft tissue masses in the joints (46). In mice, HYAL2 deficiency also leads to craniofacial and hematological defects (47, 48). CEMIP deficiency enhanced the inflammatory response to cutaneous infection (49). On the other end of the spectrum, local overexpression of HYAL1 in mouse skin induced dendritic cell (DC) migration and maturation, which in turn muted the response to contact hypersensitization (CHS) via activation of the innate immune receptor Tlr4 by HA oligosaccharides (50), while overexpression of TMEM2 resulted in increased lifespan, ER stress resistance and infection resistance in the C. elegans model (51). Further, increased hyaluronidase levels have been found in several cancers, and tend to correlate with more invasive and metastatic phenotypes (42).
2.2. Hyaluronan association with extracellular molecules
HA does not exist in the ECM as an isolated molecule. Instead, it has been known for decades that ECM HA is found in covalent binding with other proteins or glycoproteins which have been grouped under the portmanteau term “hyaladherins”. Importantly, many signaling properties of HA only come to be when HA is bound to hyaladherins, but not with free, unbound HA (52, 53). We will discuss 4 major hyaladherins, Inter-α-inhibitor (IαI), Tumor necrosis factor stimulated gene 6 (TSG-6), Pentraxin-3 (PTX3), and Versican, because their interactions with HA have been well described and clear therapeutic targets identified.
IαI is a family of composite proteins comprised of a common light chain and several homologous heavy chains (HCs). The light chain consists of a chondroitin 4-sulfate domain and a core protease inhibitory moiety, bikunin, which lends IαI its name. One bikunin molecule is usually linked to 1 or 2 HCs via a unique ester bond, producing pre-α-trypsin inhibitor (Pαl, HC3-bikunin in humans) and lαI (HC1·bikunin-HC2 in humans) (54–56). IαI are produced in the liver and coupled with bikunin before release into the circulation, but also can be produced in other solid organs including lung, kidney and brain (57, 58). IαI HCs can be bound to HA in a transesterification reaction catalyzed by tumor necrosis factor-stimulated gene 6 (TSG-6), a 35-kDa secreted ECM hyaladherin. TSG-6 is induced by TNFα, and secreted by immune and structural cells (59, 60). Thus, during inflammation, induction of TSG-6, and extravasation (as well as local de novo production) of IαI promote the deposition of a “pathological” HA-HC matrix (61–63).
TSG-6 and IαI demonstrate very interesting paradoxical effects in the pathogenesis of inflammation. Both proteins are increased in diseases such as asthma, COPD, cystic fibrosis, lung fibrosis, atherosclerosis, kidney disease, and many others (58, 61–69). On the one hand, both proteins are necessary for the generation of a pathological HA matrix and the development of airway inflammation and hyperresponsiveness in allergic and TLR4-mediated lung injury (62, 63, 68, 70). On the other hand, both have prominent anti-inflammatory activities in other disease models such as lung inflammation after endotoxin (71) and bleomycin exposure (72), pancreatitis, arthritis and many others (58, 69). TSG-6 is thought to mediate the beneficial effects of stem cells (73–75) in regenerative models. IαI binds to coagulation factor IX (76) and is a strong factor XI inhibitor (77). IαI inhibits plasmin (78) and significantly ameliorates disseminated intravascular coagulation and lung injury in LPS-induced sepsis (79). Furthermore, IαI inhibits complement activation (80–82) and prevents complement-induced lung injury (80). Finally, IαI may directly bind pathogen-associated molecular patterns such as the dengue virus (83), adenovirus and HIV (84, 85), or danger-associated molecular patterns like histones (86, 87) and HMGB1 (88), thus sequestering infectious and danger-associated molecular patterns and inhibiting inflammation.
IαI HC in the HA matrix are crosslinked through PTX3 (89). IαI, TSG-6 and PTX3 are indispensable for the normal development of a functional HA matrix in the cumulus oophorus complex in the ovulating ovary, and their absence results in severe female infertility (89–92). During inflammation, PTX3 can be induced via TLR activation (93) and has a role in injury repair (94), antimicrobial defense (95–102), and allograft survival (103). PTX3 also ameliorates allergic lung inflammation (104), LPS-induced lung injury (like TSG-6 and IαI) (105), and promotes injury repair, fibrin deposition and fibrosis in a lung injury model of acid aspiration (94). In further parallels with IαI, PTX3 can bind viral antigens, such as the influenza hemagglutinin (106) and interacts with the complement, but with conflicting effects on activation (107, 108).
These complex and partly conflicting reports on the role of TSG-6, IαI and PTX3 in inflammation may be best explained if we take into consideration the inflammatory mechanism underlying a given disease or disease model: when pathological HA matrix formation is necessary for the development of disease, for example airway hyperresponsiveness after lung injury or allograft dysfunction (63–65, 70, 109–112), then PTX3, IαI and TSG-6 promote disease by mediating the formation of pathological HA matrix. However, in non-HA-mediated processes (71, 73), PTX3-, TSG-6- and IαI-supported HA matrices may promote an anti-inflammatory role, by ameliorating complement- or coagulation factor-dependent pathways. Generally, acute processes appear to benefit from the accumulation of LMW-HA matrix which promotes inflammation and clearance of the offending agents, while chronic or degenerative processes, which lack homeostasis, may be adversely affected by the accumulation of HA matrix.
HA is also decorated by the proteoglycan Versican, and this serves to further modulate the inflammatory response in a feedback loop (113, 114). Versican is induced by TLR activation, infection or aseptic injury (115–118) and regulates the activity of chemokines (113), the retention of immune cells, and chemotaxis (115, 119). As we have seen with HAS2 above, Versican also has opposing effects depending on cell type: anti-inflammatory (macrophage expression) (117) or pro-inflammatory (mesenchymal cell expression (115, 119–121). Taken together, available research suggests that a HA matrix, including cross-linked TSG-6, PTX3 and IαI HC, and Versican, modulates the immune response to injury in complex and cell-specific ways. Indeed, HA biology cannot be appreciated without including HA interactions with hyaladherins. The hyaluronan matrix is effectively a canvas, which is dynamically decorated by hyaladherins, and it is this interaction that may dictate hyaluronan-specific effects and roles after injury (114).
2.3. Hyaluronan signaling
There are several cell surface receptors for HA such as CD44, RHAMM, HARE, LYVE1, layilin, and the innate immune receptors TLR2, TLR4 and TLR5 (19, 122–132). Importantly, HA size is a crucial determinant for receptor signaling, perhaps because signal transduction is dependent on receptor clustering on the membrane (19, 133). Although there is no generally accepted nomenclature describing HA sizes, for the purposes of this review we will be referring to high molecular weight HA (HMW-HA, 1 million Da or higher, the naturally occurring form), low-molecular-weight hyaluronan (LMW-HA, between 100–500 kDa, occurs during inflammation and tissue injury) and HA oligosaccharides (oHA, 10 HA disaccharides or less, the minimum size able to engage receptors but cannot induce receptor clustering (134). This is probably because longer chains of HA possess multivalent sites for CD44 binding while oHA have only 1 or 2 binding sites (135–137). It is important to stress that HA biological effects depend on HA size. HMW-HA is anti-inflammatory and anti-angiogenic (17), while LMW-HA is pro-inflammatory (138) and pro-angiogenic, promoting tumor progression (133, 139). In general, LMW-HA seems to be pro-inflammatory and rather deleterious while HMW-HA appears to be, by and large, beneficial. Interestingly, oHA of can either stimulate or inhibit inflammation depending on cell type and disease model. oHA can increase angiogenesis during wound healing (140) and promote inflammation in synovial fibroblasts (141), but have been also shown to ameliorate TLR3-mediated inflammation (142), inhibit HA/CD44-mediated activation of intracellular kinases (143, 144), retard tumor growth and sensitize resistant cancer cells to chemotherapeutics (133).
An important aspect of HA signaling is its ability to signal through toll-like receptors (TLRs). LMW-HA engages a receptor complex of CD44 and TLRs and induces cytokine release (145) and airway hyperresponsiveness (AHR) (70, 109) in macrophages and naïve mice, respectively. In bleomycin-induced acute lung injury, LMW-HA and HMW-HA signal through TLR2 and TLR4, but not CD44 (19). Again, HA size appears to be a major determinant of TLR-mediated signaling pathways. In general, LMW-HA induces TLR-mediated inflammation (19, 109, 146), while HMW-HA is inert (or antagonizes LMW-HA effects) (68, 70). However, HMW-HA can also signal through TLR in some contexts, although the effect is not pro-inflammatory. For example, overexpression of HAS2, which produces HMW-HA (14, 147), promotes epithelial resiliency to injury and alveolar progenitor cell renewal through TLR4 and TLR2 activation (19, 20). Thus, both HMW-HA and LMW-HA activate TLR signaling, but their effects differ (inflammation vs. epithelial resilience). The reasons and mechanisms underlying these differences are unclear but may involve different receptor clusters in each cell type. For example, LMW-HA induces activation of dendritic cells via TLR4, independently of TLR2 (123), while it activates macrophages via TLR2 pathway independently of TLR4 (148); in another example oHA activate TLR4 and CD44 in chondrocytes (149–151), but TLR2 and TLR4, and not CD44, in synovial fibroblasts (141). oHA can signal through CD44 or TLRs (152) but not both at the same time (123, 149, 153, 154). A recent study suggested that the N-acetyl groups on HA are important for TLR4 signaling, and that partial de-acetylation or selective butyrylation of HA led to inert or anti-inflammatory molecules along the TLR4 axis (155). However, much remains unknown about HA signaling through TLR, including an understanding of how the signaling occurs, since no one has been able to show direct binding of HA to these receptors. Indeed, recent studies suggest that some of the original findings of TLR-mediated LMW-HA signaling may be due to contamination by the TLR ligands, notably lipopolysaccharide (endotoxin), which activates TLR4. Some reports suggest that endotoxin-free LMW-HA does not have TLR4-activating properties (156); other work has shown that although LMW-HA causes inflammation, TLR4 actually ameliorates this inflammatory response (157). These contradictory findings could be due to differences in cells studied, cell provenance (e.g. which organism), their activation states, and the endpoints that were assayed (152). It may also be that LMW-HA does indeed activated TLR4 under some conditions, but not others, and that some of the action of HA is not direct, but due to either coordinate action by different-sized fragments (158, 159). Finally, it could be that the effects of some HA fragments are indirect: it is possible that differently-sized HA fragments compete for receptor engagement, and it is well understood that clustering of receptors is dependent on HA fragment size (134). Overall, it is probable that different mechanisms apply in different contexts and conditions.
HA signaling is normally initiated at the cell membrane. However, HA has also been detected intracellularly. It may be there as part of a physiologic pathway; for example, HA associates with the mitotic spindle, microtubules, and the receptor RHAMM during mitosis (160, 161). However, there is also the possibility that HA is produced aberrantly during pathological processes; for example, in the setting of hyperglycemia hyaluronan is produced in the Golgi and initiates an endoplasmic reticulum stress and autophagy response (162–166).
3. Therapeutic applications of hyaluronan signaling
In aggregate, the available literature may suggest the following (necessarily simplified) general principles regarding HA signaling: HMW-HA is the prevailing molecule during health and homeostasis, and it promotes cellular resilience and longevity, aided by its hyaladherin matrix. During acute inflammation, HMW-HA is degraded into LMW-HA fragments, which together with the associated hyaladherins promote the inflammatory response with the biological goal of removing the offending agents and restoring homeostasis. The process can be derailed in two major ways: first, chronic inflammation (e.g. in disease) or aberrant inflammation (e.g. through exposure to pollutants) may transform the LMW-HA pathway into a maladaptive response which supports pathology instead of homeostasis, because it fails to resolve; secondly, aberrant cells (e.g. in cancer) may co-opt the HA signaling pathway to aid local survival at the expense of the organism. In such cases, disrupting the LMW-HA signaling pathway, or restoring the HMW-HA milieu would promote disease resolution. This, then, is the guiding principle for therapeutic applications of HA that will be explored in this chapter.
Here, we will first summarize the known associations of HA with human disease and then introduce principles of therapeutic applications for HA signaling. As the preceding chapters suggest, HA biology is far-reaching, and the spectrum of potential therapeutic targets is vast. Therefore, rather than enumerating every possible disease that can be addressed with HA signaling axis, we will focus on mechanistic considerations, based on the above principles, that can guide intelligent design of HA-targeting drugs and careful selection of disease processes, with the ultimate goal to maximize therapeutic benefit and minimize side effect profiles. Clearly, more than one pathway can be invoked for given diseases. For example, inhibiting the formation of HA complexes or HA synthesis will also impair HA signaling downstream of these processes. Nevertheless, we believe that the mechanistic approach is more conducive to a thoughtful drug design process.
3.1. Associations and roles for hyaluronan matrix in human disease
Dysregulation of HA metabolism is a hallmark of many inflammatory and degenerative diseases. Increased levels of circulating or tissue HA have been found in many lung disorders including asthma, COPD, interstitial lung disease, and pulmonary hypertension (167, 168), as well as inflammatory, fibrotic and degenerative diseases like rheumatoid arthritis (169), hepatitis (170, 171), cirrhosis (172, 173), chronic kidney disease (174), atherosclerosis (175), cardiac remodeling in failure (176), diabetic pancreas (177), allograft rejection (110, 178, 179), virtually every type of solid cancer (133, 180–182), etc. In most, or all these diseases, elevated serum levels of HA have been associated with adverse outcomes and worse prognosis. Whenever HA size has been evaluated, it was most commonly LMW-HA. Furthermore, abnormal HAS expression is often found in malignancies and tissue injury and is also associated with clinical outcomes. Hematologic malignancies (monoclonal gammopathy of undetermined significance, multiple myeloma and Waldenström’s macroglobulinemia) and solid cancers (bladder cancer) contain cells with aberrant HAS1 splice variants, which are associated with poor prognosis (183). Increased HAS2 expression also negatively correlates with outcomes in breast, oral, endometrial and brain cancers (184). The prevailing theory is that increased HA expression in the tumor stroma promotes cancer progression and metastasis by enhancing pro-proliferation signaling, promoting evasion of apoptosis, inducing angiogenesis and lymphangiogenesis, promoting invasivity and metastatic potential, reprogramming energy metabolism and aiding immune evasion (184).
Abnormal HA matrix deposition (HA associated with hyaladherins) is observed in many pathological processes associated with tissue remodeling like pulmonary hypertension (185), inflammatory bowel disease (186) and asthma (61). Furthermore, in conditions of endoplasmic reticulum stress cells produce so-called “HA cables” (HA associated with IαI HC, and sometimes TSG-6, that appears in cable-like structures) that originate in the perinuclear area and protrude into the extracellular space, where they create an adhesive matrix entrapping inflammatory cells (187). Versican deposition is increased in airways of patients with idiopathic pulmonary fibrosis (188), severe ARDS (189) and asthma (190, 191). In human lung transplant patients, increased PTX3 levels are associated with graft dysfunction (i.e. ischemia reperfusion injury) (192) and genetic variability in PTX3 that leads to increased serum PTX3 levels, predisposes to allograft dysfunction (103). On the opposite side of the spectrum, genetic variability in PTX3 that leads to decreased PTX3 function, is associated with increased susceptibility to invasive aspergillosis in stem cell transplant patients (95). TSG-6 levels in synovial fluid correlate with disease progression in patients with osteoarthritis (193). Finally, there is a wealth of literature suggesting that circulating levels of IαI are associated with survival in human infections/inflammatory diseases like severe sepsis in adults, necrotizing enterocolitis in newborns and severe dengue in children (194–198).
3.2. Established applications of HA therapeutics in medicine
In this chapter we will briefly summarize established applications of HA in clinical medicine. As mentioned above, many or most of these applications are based on viscoelastic and physical properties of HA, and thus will not be a focus of this review, which is targeting biological translation of HA signaling. Nevertheless, this list highlights and supports the translational potential, safety profile and applicability of HA pharmaceuticals in clinical practice. It should also be noted that the physical and signaling effects of HA are likely to be linked, in that specific HA sizes demonstrate distinct biological and physical behavior that go in parallel. Thus, many future applications of HA will integrate its biology with its physics (199–202), as in, for example, the production of biomaterials that promote stem cell growth (HA biology) along with tissue integration and compatibility (HA physics).
Dermatology:
Perhaps the most common application of HA preparations is in cosmetic dermatology, both used superficially (included in many skin cosmetic creams) and injected as filler for wrinkle or scar treatment. HA-containing gels are used in skin wounds (including burn wounds (203)) and ulcer care (2).
ENT:
Along similar lines, injected HA is used for augmentation or repair of vocal cords. HA sprays are also used successfully as adjunct (hydrating) agents after sinus surgery to promote healing, reduce crusting and expedite post-operative recovery (204, 205).
Ophthalmology:
HA fillers are used in eye surgeries, in particular as replacement of corpus vitreum lost during surgical manipulation in cataract surgery, lens implantation and corneal transplantation. HA is also a very common lubricant in “artificial tear” eye drops.
Rheumatology/Orthopedics:
Rheumatoid and degenerative (osteo-) arthritis are both characterized by loss and degradation of HMW-HA in the synovial fluid and cartilage destruction. Intra-articular injections with HMW-HA have been shown to improve symptoms.
Gynecology:
HA preparations are used in two main applications: either as vaginal suppositories or creams to alleviate symptoms of vaginal atrophy or injury after menopause, surgery, irradiation etc. (206–209), or as surgically applied gels to prevent adhesions after hysteroscopic lysis (210).
Tissue Engineering:
HA biodegradability, biocompatibility and immune tolerance also make it an ideal candidate as a tissue engineering scaffold material. Skin, bone and soft tissue grafts have been engineered with the use of HA, and the range of applications is ever increasing (2, 211, 212).
3.3. Novel applications of HA Therapeutics: Modulation of HA signaling
3.3.1. HA oligosaccharides
Hyaluronan oligosaccharides (oHA) bind monovalently to HA receptors (135) and participate in irreversible binding with ECM molecules like the IαI HC (213), thus disrupting HA signaling. oHA inhibit HA activation of the PI3/Akt pathway and the complex formation between HA receptor CD44 and tyrosine kinases as well as CD44 and Emmprin, and induce PTEN (143, 214–217), thus inhibiting cancer cell growth. The inhibition of CD44/Emmprin association further blocks plasma membrane localization of monocarboxylate transporters MCT1 and MCT4, which results in the inhibition of lactate efflux and induces metabolic reprogramming of the cancer cell (218, 219). Finally, oHA inhibit the association of CD44 with drug transporters ABCB1 and ABCG2, thus decreasing the chemoresistance of tumor cells to chemotherapy drugs (215, 218, 220). These affects raised interest in the use of oHA in cancer. Indeed, studies in preclinical models have demonstrated that oHA can arrest or delay the growth of grafted tumors in vivo (214, 215, 217, 221, 222). Modification of oHA, such as sulfation of their side chains, may add to their activity by inducing inhibition of HA synthesis (223). oHA are attractive because they are small molecules (in general less than 10 disaccharide units) that can be easily formulated and delivered, and, like all other HA molecules, are non-immunogenic.
3.3.2. LMW-HA
Although most literature suggests that LMW-HA fragments are pro-inflammatory, there is some evidence that they can be utilized as therapeutics, primarily from the lung disease literature. LMW-HA promotes motile cilia beating through activation of its receptor RHAMM (224) and induces the expression of mucins (225, 226), thus potentially improving mucociliary clearance. There is a substantial body of literature suggesting that LMW-HA may protect from the development of emphysema, possibly by ameliorating elastic fiber degradation as was shown in animal models of emphysema (227–232). Inhaled LMW-HA was safe in patients with COPD and preliminary data suggested that it was effective in reducing breakdown of elastin fragments even after only 2 weeks of treatment (232, 233). In the intestine, LMW-HA induces the expression of antibacterial defensins and promotes epithelial integrity ((234) more about this below). These results suggest that there is still much to be found about mechanisms of HA signaling depending on organs, and that HA effects in disease (e.g. in COPD) or in conditions of heavy exposure burden (e.g. in the microbiome-filled intestine) may be very different from these seen in isolated cell models or exposures in naïve animals and sterile conditions.
3.3.3. HMW-HA
Preclinical and clinical studies support a role for HMW-HA as a treatment in airway disease (19, 229, 235). In several preclinical models, HMW-HA ameliorates epithelial injury, inflammation and airway hyperresponsiveness (19, 53, 68, 70, 109, 236–242). In allergic asthma, HMW-HA promotes regulatory T-cell activity and suppression of adaptive immunity (238, 239), but in all models HMW-HA can prevent repolarization and calcium flux-mediated contraction of myocytes (243), suggesting utility in preventing airway constriction across diseases. In humans, HMW-HA protects against exercise-induced airway hyperresponsiveness in patients with bronchial asthma (235). In COPD patients, HMW-HA ameliorates severe acute disease exacerbations, reduced the duration of non-invasive ventilation treatment and shortened the hospital length of stay (244).
It is worth noting that inhaled HMW-HA has been used for several years in Europe, especially in patients with cystic fibrosis, with a remarkably good safety profile (245–248) and evidence suggests that it does not undergo fragmentation or promote inflammation (249). Based on these data, HMW-HA can be considered a mature treatment candidate for inflammatory airway disease.
Further applications of HMW-HA in tissue injury and fibrosis are possible, based on available evidence. HMW-HA (induced by the anti-inflammatory cytokine IL-10) improved renal fibrosis after injury (250) and skin injury (251) in a mouse model, while the HA-production inhibitor 4MU was deleterious in these conditions. In another mouse model HMW-HA improved allograft survival by competitively inhibiting the effects of LMW-HA (110). Taken together, these results all point towards HMW-HA as being a powerful candidate agent protecting against epithelial injury, fibrosis and chronic inflammation.
3.4. Novel applications of HA Therapeutics: Inhibition of synthesis (4-Methylumbelliferone)
Inhibition of HA synthesis may be an attractive modality when exuberant HA production is part of the disease pathogenesis. In this context we may benefit from the existence of a well-known drug, 4-methylumbelliferone (4-MU), which is a coumarin derivative that inhibits HA synthesis in two ways. First, by acting in competition with uridine diphosphate (UDP) as a substrate for UDP-glucuronyltransferase (an enzyme involved in HA synthesis), it leads to a reduction in the availability of the UDP-glucuronyltransferase product UDP-glucuronic acid (UDP-GlcUA) in the cytosol. Since the abundance of UDP-GlcUA is one of the determinants of HAS synthetic activity, this in turn leads to a reduced HA abundance. Another, more recently discovered and less well understood effect of 4-MU isthe downregulatiion of HAS expression (252, 253). 4-MU has been in clinical use for over 30 years in the treatment of bile duct disorders (254–256) and is very well tolerated. This may be somewhat counterintuitive, since we just described multiple beneficial effects of HA, which 4-MU inhibits. It should be noted however, that 4-MU is not a complete inhibitor of HA production. Thus, when given in conditions of high LMW-HA production, like inflammation, it is possible that 4-MU preferentially inhibits pro-inflammatory HA expression, while still permitting the expression of native, HMW-HA which may be metabolized at a lower rate.
Aberrant HA signaling has been implicated in several autoimmune diseases, including Type 1 Diabetes mellitus. HMW-HA, in complex with TSG-6 and IαI, is abundant in the extracellular matrix if healthy pancreatic islets, where it may support normal cell function (257, 258). However, HMW-HA and its matrix are degraded to LMW-HA molecules during the development of autoimmune insulitis in humans and animal models of the disease (177, 259), and inhibition of HA synthesis with 4-MU has been effective in preventing autoimmune destruction of pancreatic islets, and diabetes progression (260).
Primary Sclerosing Cholangitis is an autoimmune disease of the biliary ducts resulting in fibrosis and ultimately cirrhosis, without known curative treatment short of transplantation. Serum HA levels are associated with PSC progression (261) and 4-MU was shown to be beneficial in preclinical models of PSC and liver fibrosis (22, 262). Because 4-MU has been known to ameliorate bile duct spasm for many years (263), it is now investigated as a potential therapeutic in PSC. A dose-finding study of 4-MU (NCT02780752) has just been completed and results are eagerly anticipated, as this could be the first step towards the study of 4-MU in PSC. Other autoimmune diseases, such as multiple sclerosis, thyroiditis and rheumatoid arthritis have been associated with elevated HA levels in serum and ECM, and may similarly benefit from inhibition of HA synthesis (264).
Given the roles of HA in cancer, described above, inhibition of HA synthesis is an attractive treatment option in cancer (265). 4-MU induces apoptosis and inhibits cancer growth in vitro (266, 267) while promoting the immune response in cancer tissue, inhibiting angiogenesis and improving survival in vivo (268–271).
3.5. Novel applications of HA Therapeutics: Hyaluronidase use
Hyaluronidase has been used for several years as an adjunct to local injection treatment (for example with local anesthetics or subcutaneously administered systemic medications), since the degradation of skin HA aids in drug absorption thus improving effectiveness (272–274). A similar use of hyaluronidase as adjunct in clinical care explored the use of hyaluronidase-assisted subcutaneous resuscitation in severely dehydrated children (aged 2 months or older) who could not get intravenous fluids (275).
Based on preclinical data (276, 277), hyaluronidase treatment has been mostly considered in cancer. In a study using recombinant human hyaluronidase in association with chemotherapy in pancreatic cancer, a significant improvement in survival was observed with hyaluronidase (278). Unfortunately these results were not replicated in a follow up, larger study, where there was no improvement in survival, although there was an improvement in objective response rate (decrease of cancer lesion size) (279). However, questions about patient selection and adequacy of dosing were raised (280), suggesting that hyaluronidase treatment may have utility in carefully selected cancer patients.
Non-cancer disease processes may also be candidates for hyaluronidase treatment. For example, hyaluronan deposition occurs after intraventricular hemorrhage in premature infants, resulting in inflammation, arrested oligodendrocyte maturation, and reduced white matter myelination. In a rabbit model of intraventricular hemorrhage, hyaluronidase treatment reduced CD44 and TLR4 expression and inflammation, and promoted oligodendrocyte maturation and myelination (281).
3.6. Novel applications of HA Therapeutics: Inhibition of HA-hyaladherin matrix formation
As mentioned in Chapter 2, a pathological HA matrix is deposited during chronic inflammation and mediates the development of disease. Disruption of this pathological matrix may thus ameliorate the disease. Most approaches have taken advantage of the fact that this pathological HA matrix seems to be reversible and amenable to resolution by the addition of exogenous HA molecules which remove hyaladherins from the deposited LMW-HA. In a mouse model of allergic asthma, instillation of HMW-HA into the airways led to reduction of pathological HA-HC complexes and amelioration of cellular and humoral inflammation (53). In the rabbit intraventricular hemorrhage model discussed above, HA-HC complexes contribute to inflammation and intraventricular injection of oHA reduced inflammation and enhanced myelination by depleting HC-HA levels (281).
3.7. Novel applications of HA Therapeutics: Therapeutic applications of hyaladherins (IαI, TSG-6)
In the Introduction to Chapter 3 we suggested that deposition of HA matrix is a physiological response to acute inflammation and has protective effects in this context. Along those lines, hyaladherins TSG-6, IαI and PTX-3 have been extensively found to be protective in many models of acute disease. TSG-6 may be one of the main mediators of the beneficial effects of mesenchymal stem cells whenever they are applied therapeutically, and was successful in ameliorating disease in many preclinical models e.g. myocardial infarction, skin and eye wound healing, colitis, traumatic brain injury and acute lung injury (69). IαI is perhaps the closest to pharmacological application. Based on promising results in preclinical models (87, 282–285), a pharmaceutical company is currently producing IαI for applications in severe pneumonia, adult and neonatal severe sepsis, and adult and neonatal ischemic brain injury (286). Overall, there is a promising future in the utilization of hyaladherins, either alone or in a HA matrix composition for medical purposes. Hyaladherins may affect the conformation of HA, thus its ability to cluster and engage receptors, they may also vastly broaden the spectrum of receptors and other ECM molecules that the HA matrix can interact with (114), and lastly they can also affect the metabolism and degradation of HA matrices.
3.8. HA in host-environment interfaces (lung, skin, intestine) think interactions with mucins and rheology, infections
HA is abundant in skin (287), in the luminal surface of the conducting airways (288), and other interfaces like the urinary bladder (289). HA can also be ingested under natural conditions: for example, HA of different molecular weights (ranging from oligosaccharides to MHW-HA) can be found in human breast milk (290), and human milk HA can promote epithelial antimicrobial defenses in vitro and in vivo via activation of CD44, ERK1/2 and the expression of β-defensins (158). Specifically-sized HA molecules have shown comparable effects. A 750 kDa HMW-HA molecule (given intraperitoneally) protects against intestinal epithelial injury in vivo via activation of TLR4 and COX2 (291, 292). A much smaller 35 kDa LMW-HA molecule, given orally, was able to promote β-defensin expression and antibacterial response in a TLR4-dependent but CD44 independent fashion (293) while also promoting epithelial integrity via activation of the HA receptor layilin and protecting against infectious and necrotizing colitis in vivo (234, 294–296). Oral administration of this HA fragment was well tolerated by healthy adult humans (297), supporting the potential utility of HA as a dietary supplement.
In the skin, emerging evidence suggests that HA fulfills a complex role linking innate and adaptive immunity (298). Degradation of HA into oHA fragments may serve to fight cutaneous infections, but may promote or ameliorate cutaneous sensitization to allergens depending on the timing of HA exposure in relation to allergen exposure: oHA exacerbate allergy when present at the time of sensitization, but may prevent it if present prior to sensitization (298). While these data provide tantalizing clues for a utility of HA as a treatment in skin infection and inflammatory diseases (like atopic dermatitis), it is still too early to fully appreciate the mechanisms and potential applications.
A much more mature arena is the role of HA at the airway interface. HA interacts with the cystic fibrosis transmembrane conductance regulator (299) which has emerged as a pathogenic factor not only in cystic fibrosis but also in COPD (300–304), and forms complexes with IαI, which blocks epithelial sodium channel activation, thus promoting mucus fluidity (305). As discussed above, LMW-HA promotes ciliary beating (224) and induces mucin expression (225, 226), while in COPD cells, HMWHA promotes mucociliary clearance without affecting ciliary beating frequency or airway surface layer hydration (244). In children with recurrent upper respiratory tract infections, aerosolized HMW-HA improved symptoms and ciliary motility while reducing adenoid hypertrophy, neutrophilic inflammation and microbial growth (306). In patients with nasal polyposis undergoing sinus surgery, intranasal HMW-HA improved mucociliary clearance and nasal obstruction (307). In aggregate, these data support a potentially fundamental role of HA in airway clearance. Furthermore, HMW-HA has bacteriostatic (308, 309), antibacterial (310) and antiviral (311) properties, and can sterically inhibit the diffusion of particles (such as micro-organisms) through mucin (312). HMW-HA is thus a well-studied modality in airway disease and is ripe for investigation in novel applications and disease settings (313, 314).
HMW-HA can also impact infections by interfering with the development of bacterial biofilms, i.e. bacteria attached to surfaces (either organic or medical devices) and held together by self-produced polymer matrices. Approximately 65–80% of microbial infections in the body, including almost all chronic infections, involve bacterial biofilms (315). Important biofilm-producing bacteria are well known pathogens like Streptococcus pneumoniae, S. pyogenes, Haemophilus influenzae, Moraxella catarrhalis and Pseudomonas aeruginosa (316–318). In studies of children with chronic or recurrent otitis and sinusitis media, mucosal biofilm was found in 54–95% of cases (319, 320). Biofilms insulate bacteria against antibiotics, thus fostering antibiotic resistance (315, 321, 322). Thus, bacterial biofilm inhibition contributes to antibacterial properties. HMW-HA inhibits bacterial adhesion to cellular substrates and has notable antiadhesive properties and high antibiofilm activity (323). In patients with recurrent or refractory cystitis, bladder instillation of HA significantly reduced the prevalence of recurrent infections and symptoms or irritation in affected patients. (324–326). Together, available data support the consideration of topical HA as a treatment agent in diseases with inflammatory and infectious etiology.
3.9. HA as delivery system
Hyaluronan has gained significant utility as a delivery system for cancer therapeutics but also other drugs, and there are several reviews on this functionality (2, 327–329). The bioavailability, degradability, non-immunogenicity of HA, and ability to target tissues with an abundance of HA receptors (e.g., cancer tissue which overexpresses the HA receptor CD44), make HA an attractive delivery system. Several applications are either in development or in clinical testing and have been reviewed elsewhere (327, 329). For reasons of expedience, we will only mention one example: intravesical application of a conjugate of paclitaxel and hyaluronic acid has shown promise in patients with carcinoma in situ (CIS) of the bladder that is not responsive to traditional treatment with bacillus Calmette-Guérin (BCG) (330). Undoubtedly, HA-linked delivery systems are an intense area of development in pharmaceutics (329).
4. Conclusions and Future Directions
The sum of available information suggests that HA is an attractive target for pharmaceutical development. HA (and its hyaladherins) are natural, cheap, biodegradable, biocompatible molecules. There is a long history of use with an outstanding safety profile. Furthermore, novel findings strongly suggest that there are exciting new horizons in HA applied biology. The HA matrix, in its many forms, is shown to participate in the maintenance of homeostasis and the development of disease in many areas of clinical medicine. There are several challenges to be overcome in the process. Size and viscosity of HMW-HA preparations can complicate the formulation of easily dispensable preparations. Also, because effects of the HA matrix can differ based on cell type, disease process and pathogenesis, the candidate diseases for a clinical translation must be carefully selected. HA is easily degradable in its native form, and formulations need to be explored which prolong half-life time without compromising efficacy. Finally, large-scale, cost-efficient production of high-purity HA, without bacterial contamination, will have to be implemented. These challenges, however, are not insurmountable. Thus, the time seems ripe to explore HA as a pharmaceutical not simply based on its physical characteristics, but by exploiting its unique and versatile biology. HA can be a prominent member of a new drug family, that may be called Matrix Biologics, which will unleash the potential of the extracellular matrix in shaping health and disease.
Figure 1.

HA metabolism and signaling pathways that may be amenable to therapeutic targeting. HA synthases produce HA which is then bound to hyaladherins (TSG-6, PTX-3, IαI, Versican) in the extracellular matrix, and indirectly to other extracellular molecules like complement, fibronectin, collagen etc. In disease, enzymatic (hyaluronidases) and non-enzymatic degradation of HA generates a LMW-HA matrix which can mediate disease progression or development through interactions with surface HA receptors such as CD44, RHAMM and toll-like receptors, which mediate its biological effects via kinase activation and receptor signaling. There are several pharmacological targets and interventions in this cycle (highlighted in bold letters), including: inhibition of HA synthases with 4-methylumbelliferone (4-MU); targeting hyaluronidases; and using HMW-HA or oHA to disturb LMW-HA matrix formation and signaling. Furthermore, in select diseases use of hyaladherins like TSG-6 or IαI may be of therapeutic benefit.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Garantziotis S, and Savani RC. Hyaluronan biology: A complex balancing act of structure, function, location and context. Matrix biology : journal of the International Society for Matrix Biology. 2019;78–79:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Valachova K, and Soltes L. Hyaluronan as a Prominent Biomolecule with Numerous Applications in Medicine. International journal of molecular sciences. 2021;22(13). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Balazs EA, and Denlinger JL. Clinical uses of hyaluronan. Ciba Found Symp. 1989;143:265–75; discussion 75–80, 81–5. [DOI] [PubMed] [Google Scholar]
- 4.Balazs EA. Viscosupplementation for treatment of osteoarthritis: from initial discovery to current status and results. Surg Technol Int. 2004;12:278–89. [PubMed] [Google Scholar]
- 5.Price RD, Myers S, Leigh IM, and Navsaria HA. The role of hyaluronic acid in wound healing: assessment of clinical evidence. Am J Clin Dermatol. 2005;6(6):393–402. [DOI] [PubMed] [Google Scholar]
- 6.Volpi N, Schiller J, Stern R, and Soltes L. Role, metabolism, chemical modifications and applications of hyaluronan. Curr Med Chem. 2009;16(14):1718–45. [DOI] [PubMed] [Google Scholar]
- 7.Gaffney J, Matou-Nasri S, Grau-Olivares M, and Slevin M. Therapeutic applications of hyaluronan. Mol Biosyst. 2010;6(3):437–43. [DOI] [PubMed] [Google Scholar]
- 8.Prestwich GD. Hyaluronic acid-based clinical biomaterials derived for cell and molecule delivery in regenerative medicine. J Control Release. 2011;155(2):193–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Laurent TC, and Fraser JR. Hyaluronan. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 1992;6(7):2397–404. [PubMed] [Google Scholar]
- 10.Dicker KT, Gurski LA, Pradhan-Bhatt S, Witt RL, Farach-Carson MC, and Jia X. Hyaluronan: a simple polysaccharide with diverse biological functions. Acta biomaterialia. 2014;10(4):1558–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Spicer AP, and McDonald JA. Characterization and molecular evolution of a vertebrate hyaluronan synthase gene family. The Journal of biological chemistry. 1998;273(4):1923–32. [DOI] [PubMed] [Google Scholar]
- 12.Prehm P. Hyaluronate is synthesized at plasma membranes. Biochem J. 1984;220(2):597–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Prehm P. Identification and regulation of the eukaryotic hyaluronate synthase. Ciba Foundation symposium. 1989;143:21–30; discussion −40, 281–5. [DOI] [PubMed] [Google Scholar]
- 14.Itano N, Sawai T, Yoshida M, Lenas P, Yamada Y, Imagawa M, et al. Three isoforms of mammalian hyaluronan synthases have distinct enzymatic properties. The Journal of biological chemistry. 1999;274(35):25085–92. [DOI] [PubMed] [Google Scholar]
- 15.Jing W, and DeAngelis PL. Synchronized chemoenzymatic synthesis of monodisperse hyaluronan polymers. The Journal of biological chemistry. 2004;279(40):42345–9. [DOI] [PubMed] [Google Scholar]
- 16.Jiang D, Liang J, and Noble PW. Hyaluronan in tissue injury and repair. Annual review of cell and developmental biology. 2007;23:435–61. [DOI] [PubMed] [Google Scholar]
- 17.Jiang D, Liang J, and Noble PW. Hyaluronan as an immune regulator in human diseases. Physiological reviews. 2011;91(1):221–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Stuhlmeier KM, and Pollaschek C. Differential effect of transforming growth factor beta (TGF-beta) on the genes encoding hyaluronan synthases and utilization of the p38 MAPK pathway in TGF-beta-induced hyaluronan synthase 1 activation. The Journal of biological chemistry. 2004;279(10):8753–60. [DOI] [PubMed] [Google Scholar]
- 19.Jiang D, Liang J, Fan J, Yu S, Chen S, Luo Y, et al. Regulation of lung injury and repair by Toll-like receptors and hyaluronan. Nature medicine. 2005;11(11):1173–9. [DOI] [PubMed] [Google Scholar]
- 20.Liang J, Zhang Y, Xie T, Liu N, Chen H, Geng Y, et al. Hyaluronan and TLR4 promote surfactant-protein-C-positive alveolar progenitor cell renewal and prevent severe pulmonary fibrosis in mice. Nature medicine. 2016;22(11):1285–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Li Y, Jiang D, Liang J, Meltzer EB, Gray A, Miura R, et al. Severe lung fibrosis requires an invasive fibroblast phenotype regulated by hyaluronan and CD44. The Journal of experimental medicine. 2011;208(7):1459–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Yang YM, Noureddin M, Liu C, Ohashi K, Kim SY, Ramnath D, et al. Hyaluronan synthase 2-mediated hyaluronan production mediates Notch1 activation and liver fibrosis. Science translational medicine. 2019;11(496). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Vigetti D, Viola M, Karousou E, De Luca G, and Passi A. Metabolic control of hyaluronan synthases. Matrix biology : journal of the International Society for Matrix Biology. 2014;35:8–13. [DOI] [PubMed] [Google Scholar]
- 24.Vigetti D, Deleonibus S, Moretto P, Karousou E, Viola M, Bartolini B, et al. Role of UDP-N-acetylglucosamine (GlcNAc) and O-GlcNAcylation of hyaluronan synthase 2 in the control of chondroitin sulfate and hyaluronan synthesis. The Journal of biological chemistry. 2012;287(42):35544–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Caon I, Parnigoni A, Viola M, Karousou E, Passi A, and Vigetti D. Cell Energy Metabolism and Hyaluronan Synthesis. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2021;69(1):35–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tian X, Azpurua J, Hine C, Vaidya A, Myakishev-Rempel M, Ablaeva J, et al. High-molecular-mass hyaluronan mediates the cancer resistance of the naked mole rat. Nature. 2013;499(7458):346–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Adamia S, Pilarski PM, Belch AR, and Pilarski LM. Aberrant splicing, hyaluronan synthases and intracellular hyaluronan as drivers of oncogenesis and potential drug targets. Current cancer drug targets. 2013;13(4):347–61. [DOI] [PubMed] [Google Scholar]
- 28.Passi A, Vigetti D, Buraschi S, and Iozzo RV. Dissecting the role of hyaluronan synthases in the tumor microenvironment. The FEBS journal. 2019;286(15):2937–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Nagy N, Gurevich I, Kuipers HF, Ruppert SM, Marshall PL, Xie BJ, et al. 4-Methylumbelliferyl glucuronide contributes to hyaluronan synthesis inhibition. The Journal of biological chemistry. 2019;294(19):7864–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Kuipers HF, Nagy N, Ruppert SM, Sunkari VG, Marshall PL, Gebe JA, et al. The pharmacokinetics and dosing of oral 4-methylumbelliferone for inhibition of hyaluronan synthesis in mice. Clinical and experimental immunology. 2016;185(3):372–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Soltes L, Mendichi R, Kogan G, Schiller J, Stankovska M, and Arnhold J. Degradative action of reactive oxygen species on hyaluronan. Biomacromolecules. 2006;7(3):659–68. [DOI] [PubMed] [Google Scholar]
- 32.Monzon ME, Fregien N, Schmid N, Falcon NS, Campos M, Casalino-Matsuda SM, et al. Reactive oxygen species and hyaluronidase 2 regulate airway epithelial hyaluronan fragmentation. The Journal of biological chemistry. 2010;285(34):26126–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Csoka AB, Frost GI, and Stern R. The six hyaluronidase-like genes in the human and mouse genomes. Matrix biology : journal of the International Society for Matrix Biology. 2001;20(8):499–508. [DOI] [PubMed] [Google Scholar]
- 34.Stern R, and Jedrzejas MJ. Hyaluronidases: their genomics, structures, and mechanisms of action. Chemical reviews. 2006;106(3):818–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Yoshida H, Nagaoka A, Kusaka-Kikushima A, Tobiishi M, Kawabata K, Sayo T, et al. KIAA1199, a deafness gene of unknown function, is a new hyaluronan binding protein involved in hyaluronan depolymerization. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(14):5612–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Totong R, Schell T, Lescroart F, Ryckebusch L, Lin YF, Zygmunt T, et al. The novel transmembrane protein Tmem2 is essential for coordination of myocardial and endocardial morphogenesis. Development. 2011;138(19):4199–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.De Angelis JE, Lagendijk AK, Chen H, Tromp A, Bower NI, Tunny KA, et al. Tmem2 Regulates Embryonic Vegf Signaling by Controlling Hyaluronic Acid Turnover. Dev Cell. 2017;40(2):123–36. [DOI] [PubMed] [Google Scholar]
- 38.Yamamoto H, Tobisawa Y, Inubushi T, Irie F, Ohyama C, and Yamaguchi Y. A mammalian homolog of the zebrafish transmembrane protein 2 (TMEM2) is the long-sought-after cell-surface hyaluronidase. The Journal of biological chemistry. 2017;292(18):7304–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Yamaguchi Y, Yamamoto H, Tobisawa Y, and Irie F. TMEM2: A missing link in hyaluronan catabolism identified? Matrix biology : journal of the International Society for Matrix Biology. 2019;78–79:139–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Meyer K Chemical structure of hyaluronic acid. Federation proceedings. 1958;17(4):1075–7. [PubMed] [Google Scholar]
- 41.Stern R Hyaluronan catabolism: a new metabolic pathway. European journal of cell biology. 2004;83(7):317–25. [DOI] [PubMed] [Google Scholar]
- 42.Lokeshwar VB, and Selzer MG. Hyalurondiase: both a tumor promoter and suppressor. Seminars in cancer biology. 2008;18(4):281–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Bourguignon LY, Singleton PA, Diedrich F, Stern R, and Gilad E. CD44 interaction with Na+-H+ exchanger (NHE1) creates acidic microenvironments leading to hyaluronidase-2 and cathepsin B activation and breast tumor cell invasion. The Journal of biological chemistry. 2004;279(26):26991–7007. [DOI] [PubMed] [Google Scholar]
- 44.Erickson M, and Stern R. Chain gangs: new aspects of hyaluronan metabolism. Biochemistry research international. 2012;2012:893947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Natowicz MR, Short MP, Wang Y, Dickersin GR, Gebhardt MC, Rosenthal DI, et al. Clinical and biochemical manifestations of hyaluronidase deficiency. The New England journal of medicine. 1996;335(14):1029–33. [DOI] [PubMed] [Google Scholar]
- 46.Triggs-Raine B, Salo TJ, Zhang H, Wicklow BA, and Natowicz MR. Mutations in HYAL1, a member of a tandemly distributed multigene family encoding disparate hyaluronidase activities, cause a newly described lysosomal disorder, mucopolysaccharidosis IX. Proceedings of the National Academy of Sciences of the United States of America. 1999;96(11):6296–300. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Jadin L, Wu X, Ding H, Frost GI, Onclinx C, Triggs-Raine B, et al. Skeletal and hematological anomalies in HYAL2-deficient mice: a second type of mucopolysaccharidosis IX? FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2008;22(12):4316–26. [DOI] [PubMed] [Google Scholar]
- 48.Chowdhury B, Hemming R, Hombach-Klonisch S, Flamion B, and Triggs-Raine B. Murine hyaluronidase 2 deficiency results in extracellular hyaluronan accumulation and severe cardiopulmonary dysfunction. The Journal of biological chemistry. 2013;288(1):520–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Dokoshi T, Zhang LJ, Li F, Nakatsuji T, Butcher A, Yoshida H, et al. Hyaluronan Degradation by Cemip Regulates Host Defense against Staphylococcus aureus Skin Infection. Cell Rep. 2020;30(1):61–8 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Muto J, Morioka Y, Yamasaki K, Kim M, Garcia A, Carlin AF, et al. Hyaluronan digestion controls DC migration from the skin. The Journal of clinical investigation. 2014;124(3):1309–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Schinzel RT, Higuchi-Sanabria R, Shalem O, Moehle EA, Webster BM, Joe L, et al. The Hyaluronidase, TMEM2, Promotes ER Homeostasis and Longevity Independent of the UPR(ER). Cell. 2019;179(6):1306–18 e18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Stober VP, Johnson CG, Majors A, Lauer ME, Cali V, Midura RJ, et al. TNF-stimulated gene 6 promotes formation of hyaluronan-inter-alpha-inhibitor heavy chain complexes necessary for ozone-induced airway hyperresponsiveness. The Journal of biological chemistry. 2017;292(51):20845–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Johnson CG, Stober VP, Cyphert-Daly JM, Trempus CS, Flake GP, Cali V, et al. High molecular weight hyaluronan ameliorates allergic inflammation and airway hyperresponsiveness in the mouse. American journal of physiology Lung cellular and molecular physiology. 2018;315(5):L787–L98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Salier JP, Rouet P, Raguenez G, and Daveau M. The inter-alpha-inhibitor family: from structure to regulation. Biochem J. 1996;315 (Pt 1):1–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Zhuo L, Hascall VC, and Kimata K. Inter-alpha-trypsin inhibitor, a covalent protein-glycosaminoglycan-protein complex. The Journal of biological chemistry. 2004;279(37):38079–82. [DOI] [PubMed] [Google Scholar]
- 56.Zhuo L, and Kimata K. Structure and function of inter-alpha-trypsin inhibitor heavy chains. Connective tissue research. 2008;49(5):311–20. [DOI] [PubMed] [Google Scholar]
- 57.Garantziotis S, Zudaire E, Trempus CS, Hollingsworth JW, Jiang D, Lancaster LH, et al. Serum inter-alpha-trypsin inhibitor and matrix hyaluronan promote angiogenesis in fibrotic lung injury. American journal of respiratory and critical care medicine. 2008;178(9):939–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Lord MS, Melrose J, Day AJ, and Whitelock JM. The Inter-alpha-Trypsin Inhibitor Family: Versatile Molecules in Biology and Pathology. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2020;68(12):907–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Milner CM, and Day AJ. TSG-6: a multifunctional protein associated with inflammation. Journal of cell science. 2003;116(Pt 10):1863–73. [DOI] [PubMed] [Google Scholar]
- 60.Milner CM, Higman VA, and Day AJ. TSG-6: a pluripotent inflammatory mediator? Biochemical Society transactions. 2006;34(Pt 3):446–50. [DOI] [PubMed] [Google Scholar]
- 61.Lauer ME, Majors AK, Comhair S, Ruple LM, Matuska B, Subramanian A, et al. Hyaluronan and Its Heavy Chain Modification in Asthma Severity and Experimental Asthma Exacerbation. The Journal of biological chemistry. 2015;290(38):23124–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Stober VP, Johnson CG, Majors A, Lauer ME, Cali V, Midura RJ, et al. TNF-stimulated gene 6 promotes formation of hyaluronan-inter-alpha-inhibitor heavy chain complexes necessary for ozone-induced airway hyperresponsiveness. The Journal of biological chemistry. 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Swaidani S, Cheng G, Lauer ME, Sharma M, Mikecz K, Hascall VC, et al. TSG-6 protein is crucial for the development of pulmonary hyaluronan deposition, eosinophilia, and airway hyperresponsiveness in a murine model of asthma. The Journal of biological chemistry. 2013;288(1):412–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Cheng G, Swaidani S, Sharma M, Lauer ME, Hascall VC, and Aronica MA. Hyaluronan deposition and correlation with inflammation in a murine ovalbumin model of asthma. Matrix biology : journal of the International Society for Matrix Biology. 2011;30(2):126–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Cheng G, Swaidani S, Sharma M, Lauer ME, Hascall VC, and Aronica MA. Correlation of hyaluronan deposition with infiltration of eosinophils and lymphocytes in a cockroach-induced murine model of asthma. Glycobiology. 2013;23(1):43–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Matuska B, Comhair S, Farver C, Chmiel J, Midura RJ, Bonfield T, et al. Pathological Hyaluronan Matrices in Cystic Fibrosis Airways and Secretions. American journal of respiratory cell and molecular biology. 2016;55(4):576–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Lazrak A, Jurkuvenaite A, Ness EC, Zhang S, Woodworth BA, Muhlebach MS, et al. Inter-alpha-inhibitor blocks epithelial sodium channel activation and decreases nasal potential differences in DeltaF508 mice. American journal of respiratory cell and molecular biology. 2014;50(5):953–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Lazrak A, Creighton J, Yu Z, Komarova S, Doran SF, Aggarwal S, et al. Hyaluronan mediates airway hyperresponsiveness in oxidative lung injury. American journal of physiology Lung cellular and molecular physiology. 2015;308(9):L891–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Day AJ, and Milner CM. TSG-6: A multifunctional protein with anti-inflammatory and tissue-protective properties. Matrix biology : journal of the International Society for Matrix Biology. 2019;78–79:60–83. [DOI] [PubMed] [Google Scholar]
- 70.Garantziotis S, Li Z, Potts EN, Kimata K, Zhuo L, Morgan DL, et al. Hyaluronan mediates ozone-induced airway hyperresponsiveness in mice. The Journal of biological chemistry. 2009;284(17):11309–17. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 71.Danchuk S, Ylostalo JH, Hossain F, Sorge R, Ramsey A, Bonvillain RW, et al. Human multipotent stromal cells attenuate lipopolysaccharide-induced acute lung injury in mice via secretion of tumor necrosis factor-alpha-induced protein 6. Stem cell research & therapy. 2011;2(3):27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Foskett AM, Bazhanov N, Ti X, Tiblow A, Bartosh TJ, and Prockop DJ. Phase-directed therapy: TSG-6 targeted to early inflammation improves bleomycin-injured lungs. American journal of physiology Lung cellular and molecular physiology. 2014;306(2):L120–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Lee RH, Pulin AA, Seo MJ, Kota DJ, Ylostalo J, Larson BL, et al. Intravenous hMSCs improve myocardial infarction in mice because cells embolized in lung are activated to secrete the anti-inflammatory protein TSG-6. Cell stem cell. 2009;5(1):54–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Choi H, Lee RH, Bazhanov N, Oh JY, and Prockop DJ. Anti-inflammatory protein TSG-6 secreted by activated MSCs attenuates zymosan-induced mouse peritonitis by decreasing TLR2/NF-kappaB signaling in resident macrophages. Blood. 2011;118(2):330–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Xie J, Broxmeyer HE, Feng D, Schweitzer KS, Yi R, Cook TG, et al. Human adipose-derived stem cells ameliorate cigarette smoke-induced murine myelosuppression via secretion of TSG-6. Stem Cells. 2015;33(2):468–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Winkler CJ, Jorba N, Shitanishi KT, and Herring SW. Protein sieving characteristics of sub-20-nm pore size filters at varying ionic strength during nanofiltration of Coagulation Factor IX. Biologicals : journal of the International Association of Biological Standardization. 2013;41(3):176–83. [DOI] [PubMed] [Google Scholar]
- 77.Delaria KA, Muller DK, Marlor CW, Brown JE, Das RC, Roczniak SO, et al. Characterization of placental bikunin, a novel human serine protease inhibitor. The Journal of biological chemistry. 1997;272(18):12209–14. [DOI] [PubMed] [Google Scholar]
- 78.Kobayashi H, Shinohara H, Takeuchi K, Itoh M, Fujie M, Saitoh M, et al. Inhibition of the soluble and the tumor cell receptor-bound plasmin by urinary trypsin inhibitor and subsequent effects on tumor cell invasion and metastasis. Cancer research. 1994;54(3):844–9. [PubMed] [Google Scholar]
- 79.Jourdain M, Carrette O, Tournoys A, Fourrier F, Mizon C, Mangalaboyi J, et al. Effects of inter-alpha-inhibitor in experimental endotoxic shock and disseminated intravascular coagulation. American journal of respiratory and critical care medicine. 1997;156(6):1825–33. [DOI] [PubMed] [Google Scholar]
- 80.Garantziotis S, Hollingsworth JW, Ghanayem RB, Timberlake S, Zhuo L, Kimata K, et al. Inter-alpha-trypsin inhibitor attenuates complement activation and complement-induced lung injury. J Immunol. 2007;179(6):4187–92. [DOI] [PubMed] [Google Scholar]
- 81.Garantziotis S Modulation of plasma complement by the initial dose of epirubicin/docetaxel therapy in breast cancer and its predictive value. British journal of cancer. 2011;104(3):542; author reply 3–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Okroj M, Holmquist E, Sjolander J, Corrales L, Saxne T, Wisniewski HG, et al. Heavy chains of inter alpha inhibitor (IalphaI) inhibit the human complement system at early stages of the cascade. The Journal of biological chemistry. 2012;287(24):20100–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Huerta V, Ramos Y, Yero A, Pupo D, Martin D, Toledo P, et al. Novel interactions of domain III from the envelope glycoprotein of dengue 2 virus with human plasma proteins. Journal of proteomics. 2016;131:205–13. [DOI] [PubMed] [Google Scholar]
- 84.Parker AL, Waddington SN, Nicol CG, Shayakhmetov DM, Buckley SM, Denby L, et al. Multiple vitamin K-dependent coagulation zymogens promote adenovirus-mediated gene delivery to hepatocytes. Blood. 2006;108(8):2554–61. [DOI] [PubMed] [Google Scholar]
- 85.Munch J, Rucker E, Standker L, Adermann K, Goffinet C, Schindler M, et al. Semen-derived amyloid fibrils drastically enhance HIV infection. Cell. 2007;131(6):1059–71. [DOI] [PubMed] [Google Scholar]
- 86.Pemberton AD, Brown JK, and Inglis NF. Proteomic identification of interactions between histones and plasma proteins: implications for cytoprotection. Proteomics. 2010;10(7):1484–93. [DOI] [PubMed] [Google Scholar]
- 87.Chaaban H, Keshari RS, Silasi-Mansat R, Popescu NI, Mehta-D’Souza P, Lim YP, et al. Inter-alpha inhibitor protein and its associated glycosaminoglycans protect against histone-induced injury. Blood. 2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Hatayama K, Chen RH, Hanson J, Teshigawara K, Qiu J, Santoso A, et al. High-mobility group box-1 and inter-alpha inhibitor proteins: In vitro binding and co-localization in cerebral cortex after hypoxic-ischemic injury. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2021;35(3):e21399. [DOI] [PubMed] [Google Scholar]
- 89.Salustri A, Garlanda C, Hirsch E, De Acetis M, Maccagno A, Bottazzi B, et al. PTX3 plays a key role in the organization of the cumulus oophorus extracellular matrix and in in vivo fertilization. Development. 2004;131(7):1577–86. [DOI] [PubMed] [Google Scholar]
- 90.Scarchilli L, Camaioni A, Bottazzi B, Negri V, Doni A, Deban L, et al. PTX3 interacts with inter-alpha-trypsin inhibitor: implications for hyaluronan organization and cumulus oophorus expansion. The Journal of biological chemistry. 2007;282(41):30161–70. [DOI] [PubMed] [Google Scholar]
- 91.Zhuo L, and Kimata K. Cumulus oophorus extracellular matrix: its construction and regulation. ell structure and function. 2001;26(4):189–96. [DOI] [PubMed] [Google Scholar]
- 92.Zhuo L, Yoneda M, Zhao M, Yingsung W, Yoshida N, Kitagawa Y, et al. Defect in SHAP-hyaluronan complex causes severe female infertility. A study by inactivation of the bikunin gene in mice. The Journal of biological chemistry. 2001;276(11):7693–6. [DOI] [PubMed] [Google Scholar]
- 93.Balhara J, Koussih L, Zhang J, and Gounni AS. Pentraxin 3: an immuno-regulator in the lungs. Front Immunol. 2013;4:127. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Doni A, Musso T, Morone D, Bastone A, Zambelli V, Sironi M, et al. An acidic microenvironment sets the humoral pattern recognition molecule PTX3 in a tissue repair mode. The Journal of experimental medicine. 2015;212(6):905–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Cunha C, Aversa F, Lacerda JF, Busca A, Kurzai O, Grube M, et al. Genetic PTX3 deficiency and aspergillosis in stem-cell transplantation. The New England journal of medicine. 2014;370(5):421–32. [DOI] [PubMed] [Google Scholar]
- 96.Jaillon S, Mancuso G, Hamon Y, Beauvillain C, Cotici V, Midiri A, et al. Prototypic long pentraxin PTX3 is present in breast milk, spreads in tissues, and protects neonate mice from Pseudomonas aeruginosa lung infection. J Immunol. 2013;191(4):1873–82. [DOI] [PubMed] [Google Scholar]
- 97.Mauri T, Coppadoro A, Bombino M, Bellani G, Zambelli V, Fornari C, et al. Alveolar pentraxin 3 as an early marker of microbiologically confirmed pneumonia: a threshold-finding prospective observational study. Crit Care. 2014;18(5):562. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Chiarini M, Sabelli C, Melotti P, Garlanda C, Savoldi G, Mazza C, et al. PTX3 genetic variations affect the risk of Pseudomonas aeruginosa airway colonization in cystic fibrosis patients. Genes Immun. 2010;11(8):665–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Moalli F, Paroni M, Veliz Rodriguez T, Riva F, Polentarutti N, Bottazzi B, et al. The therapeutic potential of the humoral pattern recognition molecule PTX3 in chronic lung infection caused by Pseudomonas aeruginosa. J Immunol. 2011;186(9):5425–34. [DOI] [PubMed] [Google Scholar]
- 100.Olesen R, Wejse C, Velez DR, Bisseye C, Sodemann M, Aaby P, et al. DC-SIGN (CD209), pentraxin 3 and vitamin D receptor gene variants associate with pulmonary tuberculosis risk in West Africans. Genes Immun. 2007;8(6):456–67. [DOI] [PubMed] [Google Scholar]
- 101.Soares AC, Souza DG, Pinho V, Vieira AT, Nicoli JR, Cunha FQ, et al. Dual function of the long pentraxin PTX3 in resistance against pulmonary infection with Klebsiella pneumoniae in transgenic mice. Microbes and infection / Institut Pasteur. 2006;8(5):1321–9. [DOI] [PubMed] [Google Scholar]
- 102.Kabbani D, Bhaskaran A, Singer LG, Bhimji A, Rotstein C, Keshavjee S, et al. Pentraxin 3 levels in bronchoalveolar lavage fluid of lung transplant recipients with invasive aspergillosis. The Journal of heart and lung transplantation : the official publication of the International Society for Heart Transplantation. 2017;36(9):973–9. [DOI] [PubMed] [Google Scholar]
- 103.Diamond JM, Meyer NJ, Feng R, Rushefski M, Lederer DJ, Kawut SM, et al. Variation in PTX3 is associated with primary graft dysfunction after lung transplantation. American journal of respiratory and critical care medicine. 2012;186(6):546–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Balhara J, Shan L, Zhang J, Muhuri A, Halayko AJ, Almiski MS, et al. Pentraxin 3 deletion aggravates allergic inflammation through a TH17-dominant phenotype and enhanced CD4 T-cell survival. The Journal of allergy and clinical immunology. 2017;139(3):950–63 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Han B, Haitsma JJ, Zhang Y, Bai X, Rubacha M, Keshavjee S, et al. Long pentraxin PTX3 deficiency worsens LPS-induced acute lung injury. Intensive care medicine. 2011;37(2):334–42. [DOI] [PubMed] [Google Scholar]
- 106.Reading PC, Bozza S, Gilbertson B, Tate M, Moretti S, Job ER, et al. Antiviral activity of the long chain pentraxin PTX3 against influenza viruses. J Immunol. 2008;180(5):3391–8. [DOI] [PubMed] [Google Scholar]
- 107.Ma YJ, Doni A, Skjoedt MO, Honore C, Arendrup M, Mantovani A, et al. Heterocomplexes of mannose-binding lectin and the pentraxins PTX3 or serum amyloid P component trigger cross-activation of the complement system. The Journal of biological chemistry. 2011;286(5):3405–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Bonavita E, Gentile S, Rubino M, Maina V, Papait R, Kunderfranco P, et al. PTX3 is an extrinsic oncosuppressor regulating complement-dependent inflammation in cancer. Cell. 2015;160(4):700–14. [DOI] [PubMed] [Google Scholar]
- 109.Garantziotis S, Li Z, Potts EN, Lindsey JY, Stober VP, Polosukhin VV, et al. TLR4 is necessary for hyaluronan-mediated airway hyperresponsiveness after ozone inhalation. American journal of respiratory and critical care medicine. 2010;181(7):666–75. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 110.Todd JL, Wang X, Sugimoto S, Kennedy VE, Zhang HL, Pavlisko EN, et al. Hyaluronan contributes to bronchiolitis obliterans syndrome and stimulates lung allograft rejection through activation of innate immunity. American journal of respiratory and critical care medicine. 2014;189(5):556–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Stober VP, Szczesniak C, Childress Q, Heise RL, Bortner C, Hollingsworth JW, et al. Bronchial epithelial injury in the context of alloimmunity promotes lymphocytic bronchiolitis through hyaluronan expression. American journal of physiology Lung cellular and molecular physiology. 2014;306(11):L1045–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Cui Y, Liu K, Monzon-Medina ME, Padera RF, Wang H, George G, et al. Therapeutic lymphangiogenesis ameliorates established acute lung allograft rejection. The Journal of clinical investigation. 2015;125(11):4255–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Wight TN, Frevert CW, Debley JS, Reeves SR, Parks WC, and Ziegler SF. Interplay of extracellular matrix and leukocytes in lung inflammation. Cellular immunology. 2017;312:1–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Tighe RM, and Garantziotis S. Hyaluronan interactions with innate immunity in lung biology. Matrix biology : journal of the International Society for Matrix Biology. 2019;78–79:84–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Evanko SP, Potter-Perigo S, Bollyky PL, Nepom GT, and Wight TN. Hyaluronan and versican in the control of human T-lymphocyte adhesion and migration. Matrix biology : journal of the International Society for Matrix Biology. 2012;31(2):90–100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Chang MY, Tanino Y, Vidova V, Kinsella MG, Chan CK, Johnson PY, et al. A rapid increase in macrophage-derived versican and hyaluronan in infectious lung disease. Matrix biology : journal of the International Society for Matrix Biology. 2014;34:1–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Chang MY, Kang I, Gale M Jr., Manicone AM, Kinsella MG, Braun KR, et al. Versican is produced by Trif- and type I interferon-dependent signaling in macrophages and contributes to fine control of innate immunity in lungs. American journal of physiology Lung cellular and molecular physiology. 2017;313(6):L1069–L86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Asplund A, Stillemark-Billton P, Larsson E, Rydberg EK, Moses J, Hulten LM, et al. Hypoxic regulation of secreted proteoglycans in macrophages. Glycobiology. 2010;20(1):33–40. [DOI] [PubMed] [Google Scholar]
- 119.Kang I, Harten IA, Chang MY, Braun KR, Sheih A, Nivison MP, et al. Versican Deficiency Significantly Reduces Lung Inflammatory Response Induced by Polyinosine-Polycytidylic Acid Stimulation. The Journal of biological chemistry. 2017;292(1):51–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Hope C, Ollar SJ, Heninger E, Hebron E, Jensen JL, Kim J, et al. TPL2 kinase regulates the inflammatory milieu of the myeloma niche. Blood. 2014;123(21):3305–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Kim S, Takahashi H, Lin WW, Descargues P, Grivennikov S, Kim Y, et al. Carcinoma-produced factors activate myeloid cells through TLR2 to stimulate metastasis. Nature. 2009;457(7225):102–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Hardwick C, Hoare K, Owens R, Hohn HP, Hook M, Moore D, et al. Molecular cloning of a novel hyaluronan receptor that mediates tumor cell motility. The Journal of cell biology. 1992;117(6):1343–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Termeer C, Benedix F, Sleeman J, Fieber C, Voith U, Ahrens T, et al. Oligosaccharides of Hyaluronan activate dendritic cells via toll-like receptor 4. The Journal of experimental medicine. 2002;195(1):99–111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Forteza RM, Casalino-Matsuda SM, Falcon NS, Valencia Gattas M, and Monzon ME. Hyaluronan and layilin mediate loss of airway epithelial barrier function induced by cigarette smoke by decreasing E-cadherin. The Journal of biological chemistry. 2012;287(50):42288–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Bono P, Rubin K, Higgins JM, and Hynes RO. Layilin, a novel integral membrane protein, is a hyaluronan receptor. Molecular biology of the cell. 2001;12(4):891–900. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Mouta Carreira C, Nasser SM, di Tomaso E, Padera TP, Boucher Y, Tomarev SI, et al. LYVE-1 is not restricted to the lymph vessels: expression in normal liver blood sinusoids and downregulation in human liver cancer and cirrhosis. Cancer research. 2001;61(22):8079–84. [PubMed] [Google Scholar]
- 127.Prevo R, Banerji S, Ferguson DJ, Clasper S, and Jackson DG. Mouse LYVE-1 is an endocytic receptor for hyaluronan in lymphatic endothelium. The Journal of biological chemistry. 2001;276(22):19420–30. [DOI] [PubMed] [Google Scholar]
- 128.Banerji S, Ni J, Wang SX, Clasper S, Su J, Tammi R, et al. LYVE-1, a new homologue of the CD44 glycoprotein, is a lymph-specific receptor for hyaluronan. The Journal of cell biology. 1999;144(4):789–801. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Weigel JA, Raymond RC, McGary C, Singh A, and Weigel PH. A blocking antibody to the hyaluronan receptor for endocytosis (HARE) inhibits hyaluronan clearance by perfused liver. The Journal of biological chemistry. 2003;278(11):9808–12. [DOI] [PubMed] [Google Scholar]
- 130.Weigel JA, Raymond RC, and Weigel PH. The hyaluronan receptor for endocytosis (HARE) is not CD44 or CD54 (ICAM-1). Biochemical and biophysical research communications. 2002;294(4):918–22. [DOI] [PubMed] [Google Scholar]
- 131.Zhou B, Weigel JA, Fauss L, and Weigel PH. Identification of the hyaluronan receptor for endocytosis (HARE). The Journal of biological chemistry. 2000;275(48):37733–41. [DOI] [PubMed] [Google Scholar]
- 132.Hussain S, Johnson CG, Sciurba J, Meng X, Stober VP, Liu C, et al. TLR5 participates in the TLR4 receptor complex and promotes MyD88-dependent signaling in environmental lung injury. Elife. 2020;9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Toole BP, Ghatak S, and Misra S. Hyaluronan oligosaccharides as a potential anticancer therapeutic. Current pharmaceutical biotechnology. 2008;9(4):249–52. [DOI] [PubMed] [Google Scholar]
- 134.Yang C, Cao M, Liu H, He Y, Xu J, Du Y, et al. The high and low molecular weight forms of hyaluronan have distinct effects on CD44 clustering. The Journal of biological chemistry. 2012;287(51):43094–107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Lesley J, Hascall VC, Tammi M, and Hyman R. Hyaluronan binding by cell surface CD44. The Journal of biological chemistry. 2000;275(35):26967–75. [DOI] [PubMed] [Google Scholar]
- 136.Wolny PM, Banerji S, Gounou C, Brisson AR, Day AJ, Jackson DG, et al. Analysis of CD44-hyaluronan interactions in an artificial membrane system: insights into the distinct binding properties of high and low molecular weight hyaluronan. The Journal of biological chemistry. 2010;285(39):30170–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Schmits R, Filmus J, Gerwin N, Senaldi G, Kiefer F, Kundig T, et al. CD44 regulates hematopoietic progenitor distribution, granuloma formation, and tumorigenicity. Blood. 1997;90(6):2217–33. [PubMed] [Google Scholar]
- 138.Horton MR, McKee CM, Bao C, Liao F, Farber JM, Hodge-DuFour J, et al. Hyaluronan fragments synergize with interferon-gamma to induce the C-X-C chemokines mig and interferon-inducible protein-10 in mouse macrophages. The Journal of biological chemistry. 1998;273(52):35088–94. [DOI] [PubMed] [Google Scholar]
- 139.Toole BP, Zoltan-Jones A, Misra S, and Ghatak S. Hyaluronan: a critical component of epithelial-mesenchymal and epithelial-carcinoma transitions. Cells, tissues, organs. 2005;179(1–2):66–72. [DOI] [PubMed] [Google Scholar]
- 140.Gao F, Yang CX, Mo W, Liu YW, and He YQ. Hyaluronan oligosaccharides are potential stimulators to angiogenesis via RHAMM mediated signal pathway in wound healing. Clinical and investigative medicine Medecine clinique et experimentale. 2008;31(3):E106–16. [DOI] [PubMed] [Google Scholar]
- 141.Campo GM, Avenoso A, D’Ascola A, Prestipino V, Scuruchi M, Nastasi G, et al. 4-mer hyaluronan oligosaccharides stimulate inflammation response in synovial fibroblasts in part via TAK-1 and in part via p38-MAPK. Current medicinal chemistry. 2013;20(9):1162–72. [DOI] [PubMed] [Google Scholar]
- 142.Kim MY, Muto J, and Gallo RL. Hyaluronic acid oligosaccharides suppress TLR3-dependent cytokine expression in a TLR4-dependent manner. PloS one. 2013;8(8):e72421. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Ghatak S, Misra S, and Toole BP. Hyaluronan constitutively regulates ErbB2 phosphorylation and signaling complex formation in carcinoma cells. The Journal of biological chemistry. 2005;280(10):8875–83. [DOI] [PubMed] [Google Scholar]
- 144.Misra S, Toole BP, and Ghatak S. Hyaluronan constitutively regulates activation of multiple receptor tyrosine kinases in epithelial and carcinoma cells. The Journal of biological chemistry. 2006;281(46):34936–41. [DOI] [PubMed] [Google Scholar]
- 145.Taylor KR, Yamasaki K, Radek KA, Di Nardo A, Goodarzi H, Golenbock D, et al. Recognition of hyaluronan released in sterile injury involves a unique receptor complex dependent on Toll-like receptor 4, CD44, and MD-2. The Journal of biological chemistry. 2007;282(25):18265–75. [DOI] [PubMed] [Google Scholar]
- 146.Voelcker V, Gebhardt C, Averbeck M, Saalbach A, Wolf V, Weih F, et al. Hyaluronan fragments induce cytokine and metalloprotease upregulation in human melanoma cells in part by signalling via TLR4. Exp Dermatol. 2008;17(2):100–7. [DOI] [PubMed] [Google Scholar]
- 147.Koprunner M, Mullegger J, and Lepperdinger G. Synthesis of hyaluronan of distinctly different chain length is regulated by differential expression of Xhas1 and 2 during early development of Xenopus laevis. Mech Dev. 2000;90(2):275–8. [DOI] [PubMed] [Google Scholar]
- 148.Scheibner KA, Lutz MA, Boodoo S, Fenton MJ, Powell JD, and Horton MR. Hyaluronan fragments act as an endogenous danger signal by engaging TLR2. J Immunol. 2006;177(2):1272–81. [DOI] [PubMed] [Google Scholar]
- 149.Campo GM, Avenoso A, Campo S, D’Ascola A, Nastasi G, and Calatroni A. Small hyaluronan oligosaccharides induce inflammation by engaging both toll-like-4 and CD44 receptors in human chondrocytes. Biochemical pharmacology. 2010;80(4):480–90. [DOI] [PubMed] [Google Scholar]
- 150.Campo GM, Avenoso A, Campo S, D’Ascola A, Traina P, and Calatroni A. Differential effect of molecular size HA in mouse chondrocytes stimulated with PMA. Biochimica et biophysica acta. 2009;1790(10):1353–67. [DOI] [PubMed] [Google Scholar]
- 151.Campo GM, Avenoso A, D’Ascola A, Prestipino V, Scuruchi M, Nastasi G, et al. Hyaluronan differently modulates TLR-4 and the inflammatory response in mouse chondrocytes. Biofactors. 2012;38(1):69–76. [DOI] [PubMed] [Google Scholar]
- 152.Powell JD, and Horton MR. Threat matrix: low-molecular-weight hyaluronan (HA) as a danger signal. Immunologic research. 2005;31(3):207–18. [DOI] [PubMed] [Google Scholar]
- 153.Taylor KR, Trowbridge JM, Rudisill JA, Termeer CC, Simon JC, and Gallo RL. Hyaluronan fragments stimulate endothelial recognition of injury through TLR4. The Journal of biological chemistry. 2004;279(17):17079–84. [DOI] [PubMed] [Google Scholar]
- 154.Termeer CC, Hennies J, Voith U, Ahrens T, Weiss JM, Prehm P, et al. Oligosaccharides of hyaluronan are potent activators of dendritic cells. J Immunol. 2000;165(4):1863–70. [DOI] [PubMed] [Google Scholar]
- 155.Babasola O, Rees-Milton KJ, Bebe S, Wang J, and Anastassiades TP. Chemically modified N-acylated hyaluronan fragments modulate proinflammatory cytokine production by stimulated human macrophages. The Journal of biological chemistry. 2014;289(36):24779–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Dong Y, Arif A, Olsson M, Cali V, Hardman B, Dosanjh M, et al. Endotoxin free hyaluronan and hyaluronan fragments do not stimulate TNF-alpha, interleukin-12 or upregulate co-stimulatory molecules in dendritic cells or macrophages. Scientific reports. 2016;6:36928. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Zhao H, Leu SW, Shi L, Dedaj R, Zhao G, Garg HG, et al. TLR4 is a negative regulator in noninfectious lung inflammation. J Immunol. 2010;184(9):5308–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Hill DR, Rho HK, Kessler SP, Amin R, Homer CR, McDonald C, et al. Human milk hyaluronan enhances innate defense of the intestinal epithelium. The Journal of biological chemistry. 2013;288(40):29090–104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159.de la Motte CA, and Kessler SP. The role of hyaluronan in innate defense responses of the intestine. International journal of cell biology. 2015;2015:481301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Evanko SP, Parks WT, and Wight TN. Intracellular hyaluronan in arterial smooth muscle cells: association with microtubules, RHAMM, and the mitotic spindle. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2004;52(12):1525–35. [DOI] [PubMed] [Google Scholar]
- 161.Evanko SP, and Wight TN. Intracellular localization of hyaluronan in proliferating cells. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 1999;47(10):1331–42. [DOI] [PubMed] [Google Scholar]
- 162.Wang AJ, Ren J, Abbadi A, Wang A, and Hascall VC. Heparin affects cytosolic glucose responses of hyperglycemic dividing mesangial cells. The Journal of biological chemistry. 2019;294(16):6591–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163.Wang CP, Hascall VC, Zhang F, Linhardt RJ, Abbadi A, and Wang A. The Responses of Hyperglycemic Dividing Mesangial Cells to Heparin Are Mediated by the Non-reducing Terminal Trisaccharide. The Journal of biological chemistry. 2015;290(48):29045–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Wang A, Sankaranarayanan NV, Yanagishita M, Templeton DM, Desai UR, Sugahara K, et al. Heparin interaction with a receptor on hyperglycemic dividing cells prevents intracellular hyaluronan synthesis and autophagy responses in models of type 1 diabetes. Matrix biology : journal of the International Society for Matrix Biology. 2015;48:36–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Wang A, Ren J, Wang CP, and Hascall VC. Heparin prevents intracellular hyaluronan synthesis and autophagy responses in hyperglycemic dividing mesangial cells and activates synthesis of an extensive extracellular monocyte-adhesive hyaluronan matrix after completing cell division. The Journal of biological chemistry. 2014;289(13):9418–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Wang A, and Hascall VC. Hyperglycemia, intracellular hyaluronan synthesis, cyclin D3 and autophagy. Autophagy. 2009;5(6):864–5. [DOI] [PubMed] [Google Scholar]
- 167.Lennon FE, and Singleton PA. Role of hyaluronan and hyaluronan-binding proteins in lung pathobiology. American journal of physiology Lung cellular and molecular physiology. 2011;301(2):L137–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Lauer ME, Dweik RA, Garantziotis S, and Aronica MA. The Rise and Fall of Hyaluronan in Respiratory Diseases. International journal of cell biology. 2015;2015:712507. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Poole AR, Witter J, Roberts N, Piccolo F, Brandt R, Paquin J, et al. Inflammation and cartilage metabolism in rheumatoid arthritis. Studies of the blood markers hyaluronic acid, orosomucoid, and keratan sulfate. Arthritis and rheumatism. 1990;33(6):790–9. [DOI] [PubMed] [Google Scholar]
- 170.Guechot J, Loria A, Serfaty L, Giral P, Giboudeau J, and Poupon R. Serum hyaluronan as a marker of liver fibrosis in chronic viral hepatitis C: effect of alpha-interferon therapy. J Hepatol. 1995;22(1):22–6. [DOI] [PubMed] [Google Scholar]
- 171.Peters L, Mocroft A, Soriano V, Rockstroh J, Rauch A, Karlsson A, et al. Hyaluronic acid levels predict risk of hepatic encephalopathy and liver-related death in HIV/viral hepatitis coinfected patients. PloS one. 2013;8(5):e64283. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Plevris JN, Haydon GH, Simpson KJ, Dawkes R, Ludlum CA, Harrison DJ, et al. Serum hyaluronan--a non-invasive test for diagnosing liver cirrhosis. Eur J Gastroenterol Hepatol. 2000;12(10):1121–7. [DOI] [PubMed] [Google Scholar]
- 173.Gudowska M, Gruszewska E, Panasiuk A, Cylwik B, Flisiak R, Swiderska M, et al. Hyaluronic acid concentration in liver diseases. Clinical and experimental medicine. 2016;16(4):523–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Stridh S, Palm F, and Hansell P. Renal interstitial hyaluronan: functional aspects during normal and pathological conditions. American journal of physiology Regulatory, integrative and comparative physiology. 2012;302(11):R1235–49. [DOI] [PubMed] [Google Scholar]
- 175.Fischer JW. Role of hyaluronan in atherosclerosis: Current knowledge and open questions. Matrix biology : journal of the International Society for Matrix Biology. 2019;78–79:324–36. [DOI] [PubMed] [Google Scholar]
- 176.Hackert K, Homann S, Mir S, Beran A, Gorressen S, Funk F, et al. 4-Methylumbelliferone Attenuates Macrophage Invasion and Myocardial Remodeling in Pressure-Overloaded Mouse Hearts. Hypertension. 2021;77(6):1918–27. [DOI] [PubMed] [Google Scholar]
- 177.Bogdani M, Johnson PY, Potter-Perigo S, Nagy N, Day AJ, Bollyky PL, et al. Hyaluronan and hyaluronan-binding proteins accumulate in both human type 1 diabetic islets and lymphoid tissues and associate with inflammatory cells in insulitis. Diabetes. 2014;63(8):2727–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Wells A, Larsson E, Hanas E, Laurent T, Hallgren R, and Tufveson G. Increased hyaluronan in acutely rejecting human kidney grafts. Transplantation. 1993;55(6):1346–9. [DOI] [PubMed] [Google Scholar]
- 179.Cui Y, Liu K, Monzon-Medina ME, Padera RF, Wang H, George G, et al. Therapeutic lymphangiogenesis ameliorates established acute lung allograft rejection. Journal of Clinical Investigation. 2015;125(11):4255–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Toole BP. Hyaluronan: from extracellular glue to pericellular cue. Nature reviews. 2004;4(7):528–39. [DOI] [PubMed] [Google Scholar]
- 181.Toole BP. The CD147-HYALURONAN Axis in Cancer. Anat Rec (Hoboken). 2019. [DOI] [PubMed] [Google Scholar]
- 182.Tammi RH, Kultti A, Kosma VM, Pirinen R, Auvinen P, and Tammi MI. Hyaluronan in human tumors: pathobiological and prognostic messages from cell-associated and stromal hyaluronan. Seminars in cancer biology. 2008;18(4):288–95. [DOI] [PubMed] [Google Scholar]
- 183.Adamia S, Kriangkum J, Belch AR, and Pilarski LM. Aberrant posttranscriptional processing of hyaluronan synthase 1 in malignant transformation and tumor progression. Advances in cancer research. 2014;123:67–94. [DOI] [PubMed] [Google Scholar]
- 184.Caon I, Bartolini B, Parnigoni A, Carava E, Moretto P, Viola M, et al. Revisiting the hallmarks of cancer: The role of hyaluronan. Seminars in cancer biology. 2020;62:9–19. [DOI] [PubMed] [Google Scholar]
- 185.Aytekin M, Comhair SA, de la Motte C, Bandyopadhyay SK, Farver CF, Hascall VC, et al. High levels of hyaluronan in idiopathic pulmonary arterial hypertension. American journal of physiology Lung cellular and molecular physiology. 2008;295(5):L789–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186.Majors AK, Austin RC, de la Motte CA, Pyeritz RE, Hascall VC, Kessler SP, et al. Endoplasmic reticulum stress induces hyaluronan deposition and leukocyte adhesion. The Journal of biological chemistry. 2003;278(47):47223–31. [DOI] [PubMed] [Google Scholar]
- 187.Hascall VC, Majors AK, De La Motte CA, Evanko SP, Wang A, Drazba JA, et al. Intracellular hyaluronan: a new frontier for inflammation? Biochimica et biophysica acta. 2004;1673(1–2):3–12. [DOI] [PubMed] [Google Scholar]
- 188.Bensadoun ES, Burke AK, Hogg JC, and Roberts CR. Proteoglycan deposition in pulmonary fibrosis. American journal of respiratory and critical care medicine. 1996;154(6 Pt 1):1819–28. [DOI] [PubMed] [Google Scholar]
- 189.Morales MM, Pires-Neto RC, Inforsato N, Lancas T, da Silva LF, Saldiva PH, et al. Small airway remodeling in acute respiratory distress syndrome: a study in autopsy lung tissue. Crit Care. 2011;15(1):R4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 190.Lv N, Xiao L, Camargo CA Jr., Wilson SR, Buist AS, Strub P, et al. Abdominal and general adiposity and level of asthma control in adults with uncontrolled asthma. Annals of the American Thoracic Society. 2014;11(8):1218–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 191.de Medeiros Matsushita M, da Silva LF, dos Santos MA, Fernezlian S, Schrumpf JA, Roughley P, et al. Airway proteoglycans are differentially altered in fatal asthma. The Journal of pathology. 2005;207(1):102–10. [DOI] [PubMed] [Google Scholar]
- 192.Diamond JM, Lederer DJ, Kawut SM, Lee J, Ahya VN, Bellamy S, et al. Elevated plasma long pentraxin-3 levels and primary graft dysfunction after lung transplantation for idiopathic pulmonary fibrosis. American journal of transplantation : official journal of the American Society of Transplantation and the American Society of Transplant Surgeons. 2011;11(11):2517–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Wisniewski HG, Colon E, Liublinska V, Karia RJ, Stabler TV, Attur M, et al. TSG-6 activity as a novel biomarker of progression in knee osteoarthritis. Osteoarthritis and cartilage / OARS, Osteoarthritis Research Society. 2014;22(2):235–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Baek YW, Brokat S, Padbury JF, Pinar H, Hixson DC, and Lim YP. Inter-alpha inhibitor proteins in infants and decreased levels in neonatal sepsis. The Journal of pediatrics. 2003;143(1):11–5. [DOI] [PubMed] [Google Scholar]
- 195.Lim YP, Bendelja K, Opal SM, Siryaporn E, Hixson DC, and Palardy JE. Correlation between mortality and the levels of inter-alpha inhibitors in the plasma of patients with severe sepsis. J Infect Dis. 2003;188(6):919–26. [DOI] [PubMed] [Google Scholar]
- 196.Opal SM, Lim YP, Siryaporn E, Moldawer LL, Pribble JP, Palardy JE, et al. Longitudinal studies of inter-alpha inhibitor proteins in severely septic patients: a potential clinical marker and mediator of severe sepsis. Critical care medicine. 2007;35(2):387–92. [DOI] [PubMed] [Google Scholar]
- 197.Chaaban H, Singh K, Huang J, Siryaporn E, Lim YP, and Padbury JF. The role of inter-alpha inhibitor proteins in the diagnosis of neonatal sepsis. The Journal of pediatrics. 2009;154(4):620–2 e1. [DOI] [PubMed] [Google Scholar]
- 198.Koraka P, Lim YP, Shin MD, Setiati TE, Mairuhu AT, van Gorp EC, et al. Plasma levels of inter-alpha inhibitor proteins in children with acute Dengue virus infection. PloS one. 2010;5(4):e9967. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Cowman MK, Schmidt TA, Raghavan P, and Stecco A. Viscoelastic Properties of Hyaluronan in Physiological Conditions. F1000Res. 2015;4:622. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 200.Vasvani S, Kulkarni P, and Rawtani D. Hyaluronic acid: A review on its biology, aspects of drug delivery, route of administrations and a special emphasis on its approved marketed products and recent clinical studies. Int J Biol Macromol. 2020;151:1012–29. [DOI] [PubMed] [Google Scholar]
- 201.Robert L. Hyaluronan, a truly “youthful” polysaccharide. Its medical applications. Pathologiebiologie. 2015;63(1):32–4. [DOI] [PubMed] [Google Scholar]
- 202.Burdick JA, and Prestwich GD. Hyaluronic acid hydrogels for biomedical applications. Adv Mater. 2011;23(12):H41–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Longinotti C The use of hyaluronic acid based dressings to treat burns: A review. Burns Trauma. 2014;2(4):162–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204.Casale M, Ciglia G, Frari V, Incammisa A, Mazzola F, Baptista P, et al. The potential role of hyaluronic acid in postoperative radiofrequency surgery for chronic inferior turbinate hypertrophy. Am J Rhinol Allergy. 2013;27(3):234–6. [DOI] [PubMed] [Google Scholar]
- 205.Casale M, Sabatino L, Frari V, Mazzola F, Dell’Aquila R, Baptista P, et al. The potential role of hyaluronan in minimizing symptoms and preventing exacerbations of chronic rhinosinusitis. Am J Rhinol Allergy. 2014;28(4):345–8. [DOI] [PubMed] [Google Scholar]
- 206.Carter J, Goldfarb S, Baser RE, Goldfrank DJ, Seidel B, Milli L, et al. A single-arm clinical trial investigating the effectiveness of a non-hormonal, hyaluronic acid-based vaginal moisturizer in endometrial cancer survivors. Gynecol Oncol. 2020;158(2):366–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207.De Seta F, Caruso S, Di Lorenzo G, Romano F, Mirandola M, and Nappi RE. Efficacy and safety of a new vaginal gel for the treatment of symptoms associated with vulvovaginal atrophy in postmenopausal women: A double-blind randomized placebo-controlled study. Maturitas. 2021;147:34–40. [DOI] [PubMed] [Google Scholar]
- 208.Jokar A, Davari T, Asadi N, Ahmadi F, and Foruhari S. Comparison of the Hyaluronic Acid Vaginal Cream and Conjugated Estrogen Used in Treatment of Vaginal Atrophy of Menopause Women: A Randomized Controlled Clinical Trial. Int J Community Based Nurs Midwifery. 2016;4(1):69–78. [PMC free article] [PubMed] [Google Scholar]
- 209.Chen J, Geng L, Song X, Li H, Giordan N, and Liao Q. Evaluation of the efficacy and safety of hyaluronic acid vaginal gel to ease vaginal dryness: a multicenter, randomized, controlled, open-label, parallel-group, clinical trial. J Sex Med. 2013;10(6):1575–84. [DOI] [PubMed] [Google Scholar]
- 210.Fei Z, Bin Z, Xin X, Fei H, and Yuechong C. Meta-analysis on the use of hyaluronic acid gel to prevent recurrence of intrauterine adhesion after hysteroscopic adhesiolysis. Taiwan J Obstet Gynecol. 2019;58(6):731–6. [DOI] [PubMed] [Google Scholar]
- 211.Sodhi H, and Panitch A. Glycosaminoglycans in Tissue Engineering: A Review. Biomolecules. 2020;11(1). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Jensen G, Holloway JL, and Stabenfeldt SE. Hyaluronic Acid Biomaterials for Central Nervous System Regenerative Medicine. Cells. 2020;9(9). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 213.Lauer ME, Glant TT, Mikecz K, DeAngelis PL, Haller FM, Husni ME, et al. Irreversible heavy chain transfer to hyaluronan oligosaccharides by tumor necrosis factor-stimulated gene-6. The Journal of biological chemistry. 2013;288(1):205–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Ghatak S, Misra S, and Toole BP. Hyaluronan oligosaccharides inhibit anchorage-independent growth of tumor cells by suppressing the phosphoinositide 3-kinase/Akt cell survival pathway. The Journal of biological chemistry. 2002;277(41):38013–20. [DOI] [PubMed] [Google Scholar]
- 215.Slomiany MG, Dai L, Bomar PA, Knackstedt TJ, Kranc DA, Tolliver L, et al. Abrogating drug resistance in malignant peripheral nerve sheath tumors by disrupting hyaluronan-CD44 interactions with small hyaluronan oligosaccharides. Cancer research. 2009;69(12):4992–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 216.Slomiany MG, Dai L, Tolliver LB, Grass GD, Zeng Y, and Toole BP. Inhibition of Functional Hyaluronan-CD44 Interactions in CD133-positive Primary Human Ovarian Carcinoma Cells by Small Hyaluronan Oligosaccharides. Clinical cancer research : an official journal of the American Association for Cancer Research. 2009;15(24):7593–601. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Urakawa H, Nishida Y, Knudson W, Knudson CB, Arai E, Kozawa E, et al. Therapeutic potential of hyaluronan oligosaccharides for bone metastasis of breast cancer. J Orthop Res. 2012;30(4):662–72. [DOI] [PubMed] [Google Scholar]
- 218.Slomiany MG, Grass GD, Robertson AD, Yang XY, Maria BL, Beeson C, et al. Hyaluronan, CD44, and emmprin regulate lactate efflux and membrane localization of monocarboxylate transporters in human breast carcinoma cells. Cancer research. 2009;69(4):1293–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Toole BP, and Slomiany MG. Hyaluronan, CD44 and Emmprin: partners in cancer cell chemoresistance. Drug Resist Updat. 2008;11(3):110–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Cordo Russo RI, Garcia MG, Alaniz L, Blanco G, Alvarez E, and Hajos SE. Hyaluronan oligosaccharides sensitize lymphoma resistant cell lines to vincristine by modulating P-glycoprotein activity and PI3K/Akt pathway. International journal of cancer Journal international du cancer. 2008;122(5):1012–8. [DOI] [PubMed] [Google Scholar]
- 221.Zeng C, Toole BP, Kinney SD, Kuo JW, and Stamenkovic I. Inhibition of tumor growth in vivo by hyaluronan oligomers. International journal of cancer Journal international du cancer. 1998;77(3):396–401. [DOI] [PubMed] [Google Scholar]
- 222.Hosono K, Nishida Y, Knudson W, Knudson CB, Naruse T, Suzuki Y, et al. Hyaluronan oligosaccharides inhibit tumorigenicity of osteosarcoma cell lines MG-63 and LM-8 in vitro and in vivo via perturbation of hyaluronan-rich pericellular matrix of the cells. The American journal of pathology. 2007;171(1):274–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Benitez A, Yates TJ, Lopez LE, Cerwinka WH, Bakkar A, and Lokeshwar VB. Targeting hyaluronidase for cancer therapy: antitumor activity of sulfated hyaluronic acid in prostate cancer cells. Cancer research. 2011;71(12):4085–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Forteza R, Lieb T, Aoki T, Savani RC, Conner GE, and Salathe M. Hyaluronan serves a novel role in airway mucosal host defense. FASEB Journal. 2001;15(12):2179–86. [DOI] [PubMed] [Google Scholar]
- 225.Casalino-Matsuda SM, Monzon ME, Day AJ, and Forteza RM. Hyaluronan fragments/CD44 mediate oxidative stress-induced MUC5B up-regulation in airway epithelium. American journal of respiratory cell and molecular biology. 2009;40(3):277–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Monzon ME, Forteza RM, and Casalino-Matsuda SM. MCP-1/CCR2B-dependent loop upregulates MUC5AC and MUC5B in human airway epithelium. American journal of physiology Lung cellular and molecular physiology. 2011;300(2):L204–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Cantor JO, Cerreta JM, Armand G, and Turino GM. Aerosolized hyaluronic acid decreases alveolar injury induced by human neutrophil elastase. Proc Soc Exp Biol Med. 1998;217(4):471–5. [DOI] [PubMed] [Google Scholar]
- 228.Cantor JO, Cerreta JM, Ochoa M, Ma S, Chow T, Grunig G, et al. Aerosolized hyaluronan limits airspace enlargement in a mouse model of cigarette smoke-induced pulmonary emphysema. Experimental lung research. 2005;31(4):417–30. [DOI] [PubMed] [Google Scholar]
- 229.Cantor JO, Cerreta JM, Ochoa M, Ma S, Liu M, and Turino GM. Therapeutic effects of hyaluronan on smoke-induced elastic fiber injury: does delayed treatment affect efficacy? Lung. 2011;189(1):51–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Cantor JO, Shteyngart B, Cerreta JM, Liu M, Armand G, and Turino GM. The effect of hyaluronan on elastic fiber injury in vitro and elastase-induced airspace enlargement in vivo. Proc Soc Exp Biol Med. 2000;225(1):65–71. [DOI] [PubMed] [Google Scholar]
- 231.Turino GM, and Cantor JO. Hyaluronan in respiratory injury and repair. American journal of respiratory and critical care medicine. 2003;167(9):1169–75. [DOI] [PubMed] [Google Scholar]
- 232.Turino GM, Ma S, Lin YY, and Cantor JO. The Therapeutic Potential of Hyaluronan in COPD. Chest. 2018;153(4):792–8. [DOI] [PubMed] [Google Scholar]
- 233.Cantor J, Ma S, and Turino G. A pilot clinical trial to determine the safety and efficacy of aerosolized hyaluronan as a treatment for COPD. International journal of chronic obstructive pulmonary disease. 2017;12:2747–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Kim Y, Kessler SP, Obery DR, Homer CR, McDonald C, and de la Motte CA. Hyaluronan 35kDa treatment protects mice from Citrobacter rodentium infection and induces epithelial tight junction protein ZO-1 in vivo. Matrix biology : journal of the International Society for Matrix Biology. 2017;62:28–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235.Petrigni G, and Allegra L. Aerosolised hyaluronic acid prevents exercise-induced bronchoconstriction, suggesting novel hypotheses on the correction of matrix defects in asthma. Pulmonary pharmacology & therapeutics. 2006;19(3):166–71. [DOI] [PubMed] [Google Scholar]
- 236.Hussain S, Ji Z, Taylor AJ, DeGraff LM, George M, Tucker CJ, et al. Multiwalled Carbon Nanotube Functionalization with High Molecular Weight Hyaluronan Significantly Reduces Pulmonary Injury. ACS nano. 2016;10(8):7675–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237.Zhou T, Yu Z, Jian MY, Ahmad I, Trempus C, Wagener BM, et al. Instillation of hyaluronan reverses acid instillation injury to the mammalian blood gas barrier. American journal of physiology Lung cellular and molecular physiology. 2018;314(5):L808–L21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Bollyky PL, Falk BA, Wu RP, Buckner JH, Wight TN, and Nepom GT. Intact extracellular matrix and the maintenance of immune tolerance: high molecular weight hyaluronan promotes persistence of induced CD4+CD25+ regulatory T cells. Journal of leukocyte biology. 2009;86(3):567–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Bollyky PL, Lord JD, Masewicz SA, Evanko SP, Buckner JH, Wight TN, et al. Cutting edge: high molecular weight hyaluronan promotes the suppressive effects of CD4+CD25+ regulatory T cells. Journal of Immunology. 2007;179(2):744–7. [DOI] [PubMed] [Google Scholar]
- 240.Liu YY, Lee CH, Dedaj R, Zhao H, Mrabat H, Sheidlin A, et al. High-molecular-weight hyaluronan--a possible new treatment for sepsis-induced lung injury: a preclinical study in mechanically ventilated rats. Crit Care. 2008;12(4):R102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Li Z, Potts-Kant EN, Garantziotis S, Foster WM, and Hollingsworth JW. Hyaluronan signaling during ozone-induced lung injury requires TLR4, MyD88, and TIRAP. PloS one. 2011;6(11):e27137. [DOI] [PMC free article] [PubMed] [Google Scholar] [Research Misconduct Found]
- 242.Feng F, Li Z, Potts-Kant EN, Wu Y, Foster WM, Williams KL, et al. Hyaluronan activation of the Nlrp3 inflammasome contributes to the development of airway hyperresponsiveness. Environmental health perspectives. 2012;120(12):1692–8. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 243.Lazrak A, Creighton JR, Yu Z, Komarova S, Doran SF, Aggarwal S, et al. Hyaluronan mediates airway hyper-responsiveness in oxidative lung injury. Am J Physiol Lung Cell Mol Physiol. 2015:ajplung 00377 2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Galdi F, Pedone C, McGee CA, George M, Rice AB, Hussain SS, et al. Inhaled high molecular weight hyaluronan ameliorates respiratory failure in acute COPD exacerbation: a pilot study. Respiratory research. 2021;22(1):30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Varricchio A, Capasso M, Avvisati F, Varricchio AM, De Lucia A, Brunese FP, et al. Inhaled hyaluronic acid as ancillary treatment in children with bacterial acute rhinopharyngitis. Journal of biological regulators and homeostatic agents. 2014;28(3):537–43. [PubMed] [Google Scholar]
- 246.Ros M, Casciaro R, Lucca F, Troiani P, Salonini E, Favilli F, et al. Hyaluronic acid improves the tolerability of hypertonic saline in the chronic treatment of cystic fibrosis patients: a multicenter, randomized, controlled clinical trial. Journal of aerosol medicine and pulmonary drug delivery. 2014;27(2):133–7. [DOI] [PubMed] [Google Scholar]
- 247.Cresta F, Naselli A, Favilli F, and Casciaro R. Inhaled hypertonic saline+hyaluronic acid in cystic fibrosis with asthma-like symptoms: a new therapeutic chance. BMJ case reports. 2013;2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 248.Furnari ML, Termini L, Traverso G, Barrale S, Bonaccorso MR, Damiani G, et al. Nebulized hypertonic saline containing hyaluronic acid improves tolerability in patients with cystic fibrosis and lung disease compared with nebulized hypertonic saline alone: a prospective, randomized, double-blind, controlled study. Therapeutic advances in respiratory disease. 2012;6(6):315–22. [DOI] [PubMed] [Google Scholar]
- 249.Lamas A, Marshburn J, Stober VP, Donaldson SH, and Garantziotis S. Effects of inhaled high-molecular weight hyaluronan in inflammatory airway disease. Respiratory research. 2016;17(1):123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.Wang X, Balaji S, Steen EH, Blum AJ, Li H, Chan CK, et al. High-molecular weight hyaluronan attenuates tubulointerstitial scarring in kidney injury. JCI Insight. 2020;5(12). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 251.Balaji S, Wang X, King A, Le LD, Bhattacharya SS, Moles CM, et al. Interleukin-10-mediated regenerative postnatal tissue repair is dependent on regulation of hyaluronan metabolism via fibroblast-specific STAT3 signaling. FASEB journal : official publication of the Federation of American Societies for Experimental Biology. 2017;31(3):868–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252.Kakizaki I, Kojima K, Takagaki K, Endo M, Kannagi R, Ito M, et al. A novel mechanism for the inhibition of hyaluronan biosynthesis by 4-methylumbelliferone. The Journal of biological chemistry. 2004;279(32):33281–9. [DOI] [PubMed] [Google Scholar]
- 253.Kultti A, Pasonen-Seppanen S, Jauhiainen M, Rilla KJ, Karna R, Pyoria E, et al. 4-Methylumbelliferone inhibits hyaluronan synthesis by depletion of cellular UDP-glucuronic acid and downregulation of hyaluronan synthase 2 and 3. Experimental cell research. 2009;315(11):1914–23. [DOI] [PubMed] [Google Scholar]
- 254.Abate A, Dimartino V, Spina P, Costa PL, Lombardo C, Santini A, et al. Hymecromone in the treatment of motor disorders of the bile ducts: a multicenter, double-blind, placebo-controlled clinical study. Drugs under experimental and clinical research. 2001;27(5–6):223–31. [PubMed] [Google Scholar]
- 255.Quaranta S, Rossetti S, and Camarri E. [Double-blind clinical study on hymecromone and placebo in motor disorders of the bile ducts after cholecystectomy]. Clin Ter. 1984;108(6):513–7. [PubMed] [Google Scholar]
- 256.Coccia G, Michetti P, Rossetti S, and Dodero M. [Double-blind cross-over study of imecromone and placebo in the treatment of dyspeptic syndrome]. Minerva Dietol Gastroenterol. 1985;31(2):293–8. [PubMed] [Google Scholar]
- 257.Hull RL, Johnson PY, Braun KR, Day AJ, and Wight TN. Hyaluronan and hyaluronan binding proteins are normal components of mouse pancreatic islets and are differentially expressed by islet endocrine cell types. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2012;60(10):749–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Bollyky PL, Bogdani M, Bollyky JB, Hull RL, and Wight TN. The role of hyaluronan and the extracellular matrix in islet inflammation and immune regulation. Curr Diab Rep. 2012;12(5):471–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Bogdani M, Speake C, Dufort MJ, Johnson PY, Larmore MJ, Day AJ, et al. Hyaluronan deposition in islets may precede and direct the location of islet immune-cell infiltrates. Diabetologia. 2020;63(3):549–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Nagy N, Kaber G, Johnson PY, Gebe JA, Preisinger A, Falk BA, et al. Inhibition of hyaluronan synthesis restores immune tolerance during autoimmune insulitis. The Journal of clinical investigation. 2015;125(10):3928–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Vesterhus M, Hov JR, Holm A, Schrumpf E, Nygard S, Godang K, et al. Enhanced liver fibrosis score predicts transplant-free survival in primary sclerosing cholangitis. Hepatology. 2015;62(1):188–97. [DOI] [PubMed] [Google Scholar]
- 262.Sawada N, Mikami K, Igarashi G, Endo T, Sato K, Kakizaki I, et al. Beneficial effect of 4-methylumbelliferone against bile duct ligation-induced hepatic fibrosis in rats. Hirosaki Medical Journal. 2016;66(2–4):143–51. [Google Scholar]
- 263.McFarland RJ, Corbett CR, Taylor P, and Nash AG. The relaxant action of hymecromone and lignocaine on induced spasm of the bile duct sphincter. British journal of clinical pharmacology. 1984;17(6):766–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Nagy N, Kuipers HF, Marshall PL, Wang E, Kaber G, and Bollyky PL. Hyaluronan in immune dysregulation and autoimmune diseases. Matrix biology : journal of the International Society for Matrix Biology. 2019;78–79:292–313. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Nagy N, Kuipers HF, Frymoyer AR, Ishak HD, Bollyky JB, Wight TN, et al. 4-methylumbelliferone treatment and hyaluronan inhibition as a therapeutic strategy in inflammation, autoimmunity, and cancer. Front Immunol. 2015;6:123. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Saito T, Tamura D, Nakamura T, Makita Y, Ariyama H, Komiyama K, et al. 4-methylumbelliferone leads to growth arrest and apoptosis in canine mammary tumor cells. Oncology reports. 2013;29(1):335–42. [DOI] [PubMed] [Google Scholar]
- 267.Lokeshwar VB, Lopez LE, Munoz D, Chi A, Shirodkar SP, Lokeshwar SD, et al. Antitumor activity of hyaluronic acid synthesis inhibitor 4-methylumbelliferone in prostate cancer cells. Cancer research. 2010;70(7):2613–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 268.Malvicini M, Fiore E, Ghiaccio V, Piccioni F, Rizzo M, Olmedo Bonadeo L, et al. Tumor Microenvironment Remodeling by 4-Methylumbelliferone Boosts the Antitumor Effect of Combined Immunotherapy in Murine Colorectal Carcinoma. Molecular therapy : the journal of the American Society of Gene Therapy. 2015;23(9):1444–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Piccioni F, Fiore E, Bayo J, Atorrasagasti C, Peixoto E, Rizzo M, et al. 4-methylumbelliferone inhibits hepatocellular carcinoma growth by decreasing IL-6 production and angiogenesis. Glycobiology. 2015;25(8):825–35. [DOI] [PubMed] [Google Scholar]
- 270.Piccioni F, Malvicini M, Garcia MG, Rodriguez A, Atorrasagasti C, Kippes N, et al. Antitumor effects of hyaluronic acid inhibitor 4-methylumbelliferone in an orthotopic hepatocellular carcinoma model in mice. Glycobiology. 2012;22(3):400–10. [DOI] [PubMed] [Google Scholar]
- 271.Rodriguez MM, Onorato A, Cantero MJ, Dominguez L, Bayo J, Fiore E, et al. 4-methylumbelliferone-mediated polarization of M1 macrophages correlate with decreased hepatocellular carcinoma aggressiveness in mice. Scientific reports. 2021;11(1):6310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Weber GC, Buhren BA, Schrumpf H, Wohlrab J, and Gerber PA. Clinical Applications of Hyaluronidase. Advances in experimental medicine and biology. 2019;1148:255–77. [DOI] [PubMed] [Google Scholar]
- 273.Sanchez L, Richter J, Cho HJ, Jagannath S, Madduri D, Parekh S, et al. Subcutaneous daratumumab and hyaluronidase-fihj in newly diagnosed or relapsed/refractory multiple myeloma. Ther Adv Hematol. 2021;12:2040620720987075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Duco MR, Murdock JL, and Reeves DJ. Trastuzumab/Hyaluronidase-oysk: A New Option for Patients With HER2-Positive Breast Cancer. The Annals of pharmacotherapy. 2020;54(3):254–61. [DOI] [PubMed] [Google Scholar]
- 275.Zubairi H, Nelson BD, Tulshian P, Fredricks K, Altawil Z, Mireles S, et al. Hyaluronidase-Assisted Resuscitation in Kenya for Severely Dehydrated Children. Pediatr Emerg Care. 2019;35(10):692–5. [DOI] [PubMed] [Google Scholar]
- 276.Thompson CB, Shepard HM, O’Connor PM, Kadhim S, Jiang P, Osgood RJ, et al. Enzymatic depletion of tumor hyaluronan induces antitumor responses in preclinical animal models. Mol Cancer Ther. 2010;9(11):3052–64. [DOI] [PubMed] [Google Scholar]
- 277.Provenzano PP, Cuevas C, Chang AE, Goel VK, Von Hoff DD, and Hingorani SR. Enzymatic targeting of the stroma ablates physical barriers to treatment of pancreatic ductal adenocarcinoma. Cancer cell. 2012;21(3):418–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Hingorani SR, Zheng L, Bullock AJ, Seery TE, Harris WP, Sigal DS, et al. HALO 202: Randomized Phase II Study of PEGPH20 Plus Nab-Paclitaxel/Gemcitabine Versus Nab-Paclitaxel/Gemcitabine in Patients With Untreated, Metastatic Pancreatic Ductal Adenocarcinoma. J Clin Oncol. 2018;36(4):359–66. [DOI] [PubMed] [Google Scholar]
- 279.Van Cutsem E, Tempero MA, Sigal D, Oh DY, Fazio N, Macarulla T, et al. Randomized Phase III Trial of Pegvorhyaluronidase Alfa With Nab-Paclitaxel Plus Gemcitabine for Patients With Hyaluronan-High Metastatic Pancreatic Adenocarcinoma. J Clin Oncol. 2020;38(27):3185–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280.Hakim N, Patel R, Devoe C, and Saif MW. Why HALO 301 Failed and Implications for Treatment of Pancreatic Cancer. Pancreas (Fairfax). 2019;3(1):e1–e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Vinukonda G, Dohare P, Arshad A, Zia MT, Panda S, Korumilli R, et al. Hyaluronidase and Hyaluronan Oligosaccharides Promote Neurological Recovery after Intraventricular Hemorrhage. The Journal of neuroscience : the official journal of the Society for Neuroscience. 2016;36(3):872–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 282.Singh K, Zhang LX, Bendelja K, Heath R, Murphy S, Sharma S, et al. Inter-alpha inhibitor protein administration improves survival from neonatal sepsis in mice. Pediatric research. 2010;68(3):242–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Chen X, Nakada S, Donahue JE, Chen RH, Tucker R, Qiu J, et al. Neuroprotective effects of inter-alpha inhibitor proteins after hypoxic-ischemic brain injury in neonatal rats. Exp Neurol. 2019;317:244–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 284.Stober VP, Lim YP, Opal S, Zhuo L, Kimata K, and Garantziotis S. Inter-alpha-inhibitor Ameliorates Endothelial Inflammation in Sepsis. Lung. 2019;197(3):361–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Schuffels S, Nakada S, Wu Y, Lim YP, Chen X, and Stonestreet BS. Effects of inter-alpha inhibitor proteins on brain injury after exposure of neonatal rats to severe hypoxia-ischemia. Exp Neurol. 2020;334:113442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286.Lim YP. ProThera Biologics, Inc.: a novel immunomodulator and biomarker for life-threatening diseases. R I Med J (2013). 2013;96(2):16–8. [PubMed] [Google Scholar]
- 287.Papakonstantinou E, Roth M, and Karakiulakis G. Hyaluronic acid: A key molecule in skin aging. Dermatoendocrinol. 2012;4(3):253–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Abbadi A, Lauer M, Swaidani S, Wang A, and Hascall V. Hyaluronan Rafts on Airway Epithelial Cells. The Journal of biological chemistry. 2016;291(3):1448–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 289.Poggi MM, Johnstone PA, and Conner RJ. Glycosaminoglycan content of human bladders. a method of analysis using cold-cup biopsies. Urologic oncology. 2000;5(5):234–7. [DOI] [PubMed] [Google Scholar]
- 290.Yuan H, Amin R, Ye X, de la Motte CA, and Cowman MK. Determination of hyaluronan molecular mass distribution in human breast milk. Anal Biochem. 2015;474:78–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 291.Riehl TE, Ee X, and Stenson WF. Hyaluronic acid regulates normal intestinal and colonic growth in mice. American Journal of Physiology: Gastrointestinal and Liver Physiology. 2012;303(3):G377–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Riehl TE, Foster L, and Stenson WF. Hyaluronic acid is radioprotective in the intestine through a TLR4 and COX-2-mediated mechanism. American Journal of Physiology: Gastrointestinal and Liver Physiology. 2012;302(3):G309–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293.Hill DR, Kessler SP, Rho HK, Cowman MK, and de la Motte CA. Specific-sized hyaluronan fragments promote expression of human beta-defensin 2 in intestinal epithelium. The Journal of biological chemistry. 2012;287(36):30610–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Kim Y, West GA, Ray G, Kessler SP, Petrey AC, Fiocchi C, et al. Layilin is critical for mediating hyaluronan 35kDa-induced intestinal epithelial tight junction protein ZO-1 in vitro and in vivo. Matrix biology : journal of the International Society for Matrix Biology. 2018;66:93–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Kessler SP, Obery DR, Nickerson KP, Petrey AC, McDonald C, and de la Motte CA. Multifunctional Role of 35 Kilodalton Hyaluronan in Promoting Defense of the Intestinal Epithelium. The journal of histochemistry and cytochemistry : official journal of the Histochemistry Society. 2018;66(4):273–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 296.Gunasekaran A, Eckert J, Burge K, Zheng W, Yu Z, Kessler S, et al. Hyaluronan 35 kDa enhances epithelial barrier function and protects against the development of murine necrotizing enterocolitis. Pediatric research. 2020;87(7):1177–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Bellar A, Kessler SP, Obery DR, Sangwan N, Welch N, Nagy LE, et al. Safety of Hyaluronan 35 in Healthy Human Subjects: A Pilot Study. Nutrients. 2019;11(5). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Muto J, Sayama K, Gallo RL, and Kimata K. Emerging evidence for the essential role of hyaluronan in cutaneous biology. Journal of dermatological science. 2019;94(1):190–5. [DOI] [PubMed] [Google Scholar]
- 299.Schulz T, Schumacher U, Prante C, Sextro W, and Prehm P. Cystic fibrosis transmembrane conductance regulator can export hyaluronan. Pathobiology. 2010;77(4):200–9. [DOI] [PubMed] [Google Scholar]
- 300.Rowe SM, Jones I, Dransfield MT, Haque N, Gleason S, Hayes KA, et al. Efficacy and Safety of the CFTR Potentiator Icenticaftor (QBW251) in COPD: Results from a Phase 2 Randomized Trial. International journal of chronic obstructive pulmonary disease. 2020;15:2399–409. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 301.Solomon GM, Fu L, Rowe SM, and Collawn JF. The therapeutic potential of CFTR modulators for COPD and other airway diseases. Curr Opin Pharmacol. 2017;34:132–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 302.Raju SV, Tate JH, Peacock SK, Fang P, Oster RA, Dransfield MT, et al. Impact of heterozygote CFTR mutations in COPD patients with chronic bronchitis. Respiratory research. 2014;15:18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Rab A, Rowe SM, Raju SV, Bebok Z, Matalon S, and Collawn JF. Cigarette smoke and CFTR: implications in the pathogenesis of COPD. American journal of physiology Lung cellular and molecular physiology. 2013;305(8):L530–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Dransfield MT, Wilhelm AM, Flanagan B, Courville C, Tidwell SL, Raju SV, et al. Acquired cystic fibrosis transmembrane conductance regulator dysfunction in the lower airways in COPD. Chest. 2013;144(2):498–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Lazrak A, Jurkuvenaite A, Ness EC, Zhang S, Woodworth BA, Muhlebach MS, et al. Inter-alpha-inhibitor blocks epithelial sodium channel activation and decreases nasal potential differences in DeltaF508 mice. Am J Respir Cell Mol Biol. 2014;50(5):953–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Macchi A, Castelnuovo P, Terranova P, and Digilio E. Effects of sodium hyaluronate in children with recurrent upper respiratory tract infections: results of a randomised controlled study. Int J Immunopathol Pharmacol. 2013;26(1):127–35. [DOI] [PubMed] [Google Scholar]
- 307.Gelardi M, Guglielmi AV, De Candia N, Maffezzoni E, Berardi P, and Quaranta N. Effect of sodium hyaluronate on mucociliary clearance after functional endoscopic sinus surgery. Eur Ann Allergy Clin Immunol. 2013;45(3):103–8. [PubMed] [Google Scholar]
- 308.Carlson GA, Dragoo JL, Samimi B, Bruckner DA, Bernard GW, Hedrick M, et al. Bacteriostatic properties of biomatrices against common orthopaedic pathogens. Biochem Biophys Res Commun. 2004;321(2):472–8. [DOI] [PubMed] [Google Scholar]
- 309.Pirnazar P, Wolinsky L, Nachnani S, Haake S, Pilloni A, and Bernard GW. Bacteriostatic effects of hyaluronic acid. J Periodontol. 1999;70(4):370–4. [DOI] [PubMed] [Google Scholar]
- 310.Liu A, Park JH, Zhang X, Sugita S, Naito Y, Lee JH, et al. Therapeutic Effects of Hyaluronic Acid in Bacterial Pneumonia in Ex Vivo Perfused Human Lungs. American journal of respiratory and critical care medicine. 2019;200(10):1234–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Cermelli C, Cuoghi A, Scuri M, Bettua C, Neglia RG, Ardizzoni A, et al. In vitro evaluation of antiviral and virucidal activity of a high molecular weight hyaluronic acid. Virol J. 2011;8:141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Hansen IM, Ebbesen MF, Kaspersen L, Thomsen T, Bienk K, Cai Y, et al. Hyaluronic Acid Molecular Weight-Dependent Modulation of Mucin Nanostructure for Potential Mucosal Therapeutic Applications. Mol Pharm. 2017;14(7):2359–67. [DOI] [PubMed] [Google Scholar]
- 313.Maiz Carro L, and Martinez-Garcia MA. Use of Hyaluronic Acid (HA) in Chronic Airway Diseases. Cells. 2020;9(10). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 314.Garantziotis S, Brezina M, Castelnuovo P, and Drago L. The role of hyaluronan in the pathobiology and treatment of respiratory disease. American journal of physiology Lung cellular and molecular physiology. 2016;310(9):L785–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315.Davies D Understanding biofilm resistance to antibacterial agents. Nat Rev Drug Discov. 2003;2(2):114–22. [DOI] [PubMed] [Google Scholar]
- 316.Hall-Stoodley L, Hu FZ, Gieseke A, Nistico L, Nguyen D, Hayes J, et al. Direct detection of bacterial biofilms on the middle-ear mucosa of children with chronic otitis media. Jama. 2006;296(2):202–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Torretta S, Drago L, Marchisio P, Gaffuri M, Clemente IA, and Pignataro L. Topographic distribution of biofilm-producing bacteria in adenoid subsites of children with chronic or recurrent middle ear infections. Ann Otol Rhinol Laryngol. 2013;122(2):109–13. [DOI] [PubMed] [Google Scholar]
- 318.Vergison A Microbiology of otitis media: a moving target. Vaccine. 2008;26 Suppl 7:G5–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Kania RE, Lamers GE, Vonk MJ, Dorpmans E, Struik J, Tran Ba Huy P, et al. Characterization of mucosal biofilms on human adenoid tissues. Laryngoscope. 2008;118(1):128–34. [DOI] [PubMed] [Google Scholar]
- 320.Coticchia J, Zuliani G, Coleman C, Carron M, Gurrola J 2nd, Haupert M, et al. Biofilm surface area in the pediatric nasopharynx: Chronic rhinosinusitis vs obstructive sleep apnea. Arch Otolaryngol Head Neck Surg. 2007;133(2):110–4. [DOI] [PubMed] [Google Scholar]
- 321.Romano CL, Toscano M, Romano D, and Drago L. Antibiofilm agents and implant-related infections in orthopaedics: where are we? J Chemother. 2013;25(2):67–80. [DOI] [PubMed] [Google Scholar]
- 322.Watnick P, and Kolter R. Biofilm, city of microbes. J Bacteriol. 2000;182(10):2675–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323.Drago L, Cappelletti L, De Vecchi E, Pignataro L, Torretta S, and Mattina R. Antiadhesive and antibiofilm activity of hyaluronic acid against bacteria responsible for respiratory tract infections. APMIS : acta pathologica, microbiologica, et immunologica Scandinavica. 2014;122(10):1013–9. [DOI] [PubMed] [Google Scholar]
- 324.Kim A, Lim B, Song M, and Choo MS. Pretreatment features to influence effectiveness of intravesical hyaluronic Acid instillation in refractory interstitial cystitis/painful bladder syndrome. Int Neurourol J. 2014;18(3):163–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 325.De Vita D, Antell H, and Giordano S. Effectiveness of intravesical hyaluronic acid with or without chondroitin sulfate for recurrent bacterial cystitis in adult women: a meta-analysis. Int Urogynecol J. 2013;24(4):545–52. [DOI] [PubMed] [Google Scholar]
- 326.De Vita D, and Giordano S. Effectiveness of intravesical hyaluronic acid/chondroitin sulfate in recurrent bacterial cystitis: a randomized study. Int Urogynecol J. 2012;23(12):1707–13. [DOI] [PubMed] [Google Scholar]
- 327.Kotla NG, Bonam SR, Rasala S, Wankar J, Bohara RA, Bayry J, et al. Recent advances and prospects of hyaluronan as a multifunctional therapeutic system. J Control Release. 2021. [DOI] [PubMed] [Google Scholar]
- 328.Grosso R, and de-Paz MV. Thiolated-Polymer-Based Nanoparticles as an Avant-Garde Approach for Anticancer Therapies-Reviewing Thiomers from Chitosan and Hyaluronic Acid. Pharmaceutics. 2021;13(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329.Harrer D, Sanchez Armengol E, Friedl JD, Jalil A, Jelkmann M, Leichner C, et al. Is hyaluronic acid the perfect excipient for the pharmaceutical need? Int J Pharm. 2021;601:120589. [DOI] [PubMed] [Google Scholar]
- 330.Bassi PF, Volpe A, D’Agostino D, Palermo G, Renier D, Franchini S, et al. Paclitaxel-hyaluronic acid for intravesical therapy of bacillus Calmette-Guerin refractory carcinoma in situ of the bladder: results of a phase I study. The Journal of urology. 2011;185(2):445–9. [DOI] [PubMed] [Google Scholar]
