Next Article in Journal
Antifibrotic Effects of an α7 Nicotinic Acetylcholine Receptor Agonist in Keloid Fibroblasts and a Rat Scar Model
Next Article in Special Issue
Human Leukocyte Antigen-DR Expression on Monocytes Is a Useful Predictor in a Systemic Inflammation Response-Based Prognostic Model in Advanced Non-Small Cell Lung Cancer
Previous Article in Journal
The Future of Fascia—A Scoping Review on Emerging Research Trends and Recommended Sample Sizes and Effect Sizes in Fascia Studies
Previous Article in Special Issue
The Roles of SHCBP1 in Cancer Hallmarks: Molecular Mechanisms and Therapeutic Implications
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

CD44 as a Central Integrator of Inflammation and Fibrosis: From Molecular Signaling to Environmental Modulation

by
Agnieszka Pedrycz-Wieczorska
1,
Patrycja Chylińska-Wrzos
2,
Anna Grzywacz
3,4,
Ewa Zieliński
5,
Andrzej Bartosiński
6,
Kornelia Kędziora-Kornatowska
7,
Marta Lis-Sochocka
2,
Paulina Mertowska
8,*,
Sebastian Mertowski
8,
Krzysztof Bojarski
9,
Mansur Rahnama-Hezavah
10,
Tomasz Urbanowicz
11 and
Ewelina Grywalska
8
1
Faculty of Medicine and Health Sciences, University of Applied Sciences in Tarnow, 33-100 Tarnów, Poland
2
Department of Histology, Embryology and Cytophysiology, Medical University of Lublin, 20-059 Lublin, Poland
3
Independent Laboratory of Behavioral Genetics and Epigenetics, Pomeranian Medical University in Szczecin, 70-111 Szczecin, Poland
4
Department of Medical Sciences and Public Health, Gdansk University of Physical Education and Sport, Kazimierza Gorskiego 1 St., 80-336 Gdansk, Poland
5
Department of Emergency Medical Services, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University Toruń, 85-067 Bydgoszcz, Poland
6
Faculty of Medicine, Collegium Medicum, Mazovian University in Płock, 09-402 Płock, Poland
7
Department of Geriatrics, Faculty of Health Sciences, L. Rydygier Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Skłodowskiej Curie 9 Street, 85-094 Bydgoszcz, Poland
8
Department of Experimental Immunology, Medical University of Lublin, 20-059 Lublin, Poland
9
General Surgery Department, SP ZOZ in Łęczna, 21-010 Łęczna, Poland
10
Department of Dental Surgery, Medical University of Lublin, 20-059 Lublin, Poland
11
Cardiac Surgery and Transplantology Department, Poznan University of Medical Sciences, 61-701 Poznan, Poland
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2025, 26(18), 8870; https://doi.org/10.3390/ijms26188870
Submission received: 7 August 2025 / Revised: 30 August 2025 / Accepted: 2 September 2025 / Published: 11 September 2025
(This article belongs to the Special Issue Biomarkers of Tumor Progression, Prognosis and Therapy: 2nd Edition)

Abstract

CD44, a multi-isoform adhesion receptor for hyaluronic acid (HA), plays a crucial role in regulating cell interactions with the extracellular matrix, cell migration, differentiation, and survival in both physiological and pathological contexts. Accumulating experimental evidence suggests that CD44 is not merely a passive marker of mesenchymal cell activation but rather an active signaling hub driving fibrosis in many organs, including the lung, skin, heart, and liver. Its involvement in fibroblast differentiation into myofibroblasts, as well as induction of the invasive phenotype of these cells, shows striking analogies to the mechanisms of epithelial-to-mesenchymal transition (EMT) known from cancer progression. In this paper, we discuss both the molecular mechanisms of CD44-dependent signaling (including through EGFR, MAPK/ERK, CaMKII, lipid rafts, and Smad) and the influence of its modulation (knockout, antibodies, blockade of HA synthesis) on the course of fibrosis in in vitro and in vivo models. In addition, we present the influence of environmental pollutants—such as heavy metals, particulate matter, endocrine disruptors, and microplastics—on the activation of the HA-CD44 axis in connective tissue, with particular emphasis on their role in the induction of chronic inflammation, EMT, and extracellular matrix deposition. The collected evidence suggests that CD44 serves as a central integrator of inflammatory and fibrogenic signals, and its pharmacological modulation may represent a novel therapeutic strategy for treating fibrotic diseases and chronic inflammatory conditions.

1. Introduction

Connective tissue is a highly organized biological structure that plays a key role not only in providing support and mechanical integrity of organs, but also in maintaining the homeostasis of the cellular microenvironment. It participates in regenerative processes, the transmission of biochemical and immunological signals, and in creating a protective barrier against pathogenic factors. An essential element regulating these functions is surface receptors, which enable bidirectional communication between cells and the components of the extracellular matrix (ECM) [1,2,3]. A special place in this signaling network is occupied by CD44—a transmembrane glycoprotein belonging to the family of adhesion receptors, constituting the primary receptor for hyaluronic acid (HA) and also participating in interactions with other ECM components, such as osteopontin, collagens, fibronectin, and laminin [4,5,6]. The CD44 protein is characterized by a complex molecular structure and high heterogeneity resulting from alternative splicing, which leads to the formation of many isoforms (so-called CD44v). Individual variants differ in the length of the extracellular domain, the degree of glycosylation, and ligand affinity [7,8,9,10].
In physiological conditions, CD44 regulates several biological processes, including adhesion and migration of fibroblasts, lymphocytes, and progenitor cells, participates in the recruitment of immunocompetent cells, and also participates in mechanotransduction, ECM remodeling, and myofibroblast differentiation [11,12]. In pathological situations, such as chronic inflammation, organ fibrosis, or neoplastic progression, CD44 becomes a key mediator of disease progression, among others, through the activation of the phosphoinositide 3-kinase/protein kinase B (PI3K/AKT), mitogen-activated protein kinase (MAPK), and Ras homolog family member A/Rho-associated protein kinase (RhoA/ROCK) signaling pathways, as well as through interactions with transforming growth factor-beta (TGF-β) receptors, epidermal growth factor receptor (EGFR), and various growth factor binding molecules [13,14,15,16,17,18,19,20]. CD44, as a transmembrane glycoprotein acting as a cell surface receptor, plays a crucial role not only in binding hyaluronic acid but also in modulating signaling pathways, including the PI3K/Akt/mTOR pathway. Both PI3Kβ and mTOR play a crucial role in regulating key cellular processes, including survival, proliferation, metabolism, and the immune response. Under pathological conditions, particularly in inflammatory and neoplastic diseases, activation of these pathways via CD44 can lead to uncontrolled cell growth or dysregulation of the immune response. PI3Kβ is involved in, among other things, neutrophil activation and regulation of phagocytosis, while mTOR controls T-cell metabolism and inhibits autophagy, which is crucial for the course of infection and sepsis. Therefore, interactions of CD44 with extracellular matrix components may initiate signaling leading to pathological activation of PI3Kβ and mTOR, making these pathways potential therapeutic targets [21]. At the same time, recent years have seen a shift in attention to the anti-inflammatory potential of CD44, resulting from its interaction with molecules regulating the immune response, such as interleukin 37 (IL-37). In addition to its classic role as an HA receptor, CD44 can function as a co-receptor in a complex with IL-1R8—essential for IL-37 action. This anti-inflammatory cytokine inhibits the expression of numerous pro-inflammatory mediators (IL-1, TNF, IL-6, IL-17, CCL2), limits mTOR activation, and influences innate and adaptive immune cells, including macrophages, dendritic cells, and T lymphocytes. The dependence of IL-37 action on the presence of IL-1R8 and previous reports of this receptor’s interaction with CD44 indicate that CD44 may also participate in mechanisms of inflammatory response suppression. This suggests a dual function of CD44—as a promoter of pro-inflammatory signals and a potential stabilizer of the IL-37/IL-1R8 complex—which opens up new therapeutic possibilities in the context of diseases associated with immune system deregulation, such as sepsis, viral infections (e.g., COVID-19) or autoimmune diseases [22].
At the same time, increasing environmental pollution—including air, water, and soil—is associated with an increase in the incidence of immunological disorders and chronic tissue-based diseases. Particularly hazardous are particulate matter (PM2.5 and PM10), heavy metals (e.g., cadmium, lead, and arsenic), endocrine-disrupting compounds (EDCs, such as phthalates and bisphenol A), dioxins, and micro- and nanoplastics. Exposure to these substances can lead to oxidative stress, epigenetic modifications, damage to cell membranes, and activation of macrophages. As a result, ECM fragments are released, including degraded HA, which, as a DAMP (damage-associated molecular pattern) molecule, can strongly bind to CD44, activating pro-inflammatory, cytokine, and pro-fibrotic cascades [23,24].
These observations support the hypothesis that CD44 functions not only as an adhesion receptor but also as a sensor of environmental signals, integrating external stimuli with the cellular response and the remodeling of the tissue microenvironment [23]. CD44 expression and activity may undergo significant modifications under the influence of toxic environmental factors, which may lead to excessive activation of fibroblasts and myofibroblasts, disturbances in cell-ECM interactions, increased invasiveness of epithelial and neoplastic cells, and chronic inflammation in tissues exposed to toxins. The role of CD44 as an element of the “environment-microenvironment-cellular response” axis may vary depending on the type of exposure, isoform variant, tissue type, and cell population characteristics.
This paper aims to review the current literature on the expression, structure, and function of the CD44 protein, with a particular emphasis on its role as a key regulator of biological processes in connective tissue. An additional goal is to analyze the impact of environmental pollution factors, including chemicals, dust, heavy metals, and microplastics, on the regulation of the CD44 signaling pathway and its interactions with hyaluronan. Particular attention was paid to molecular mechanisms that may provide a basis for using CD44 as a biomarker of environmental exposure or a therapeutic target in diseases associated with chronic inflammation and connective tissue degradation.

2. Molecular Characteristics of the CD44 Protein: Structure, Isoforms, Biological Functions

The CD44 protein (also known as the CD44 antigen) is a cell surface glycoprotein encoded by the CD44 gene in humans (uniprot.org). It belongs to the family of CD (cluster of differentiation) particles, such as the CD44 antigen. The CD44 gene is located on chromosome 11 (region 11p13) and is sometimes referred to by various synonyms, including LHR, MDU2, MDU3, and MIC4 [25]. The protein itself functions under many alternative names reflecting its diverse properties. The most common ones include: CD44 antigen (official name), CDw44, H-CAM (Homing Cell Adhesion Molecule), Hermes antigen, hyaluronate receptor, Epican, Extracellular matrix receptor III (ECMR-III) and terms indicating its proteoglycan character: heparan sulfate proteoglycan or chondroitin sulfate proteoglycan 8 (CSPG8) [26]. The CD44 protein is a significant component of the Indian blood group system (ISBT 023), which is one of the rare antigen systems of erythrocytes. The presence of specific antigens of this system—Ina and Inb—is conditioned by the substitution of a single amino acid in position 46 of the CD44 polypeptide chain (arginine for Inb, proline for Ina). These antigens are encoded by allelic variants of the CD44 gene, located on chromosome 11 (11p13), and are primarily expressed on hematopoietic cells, including erythrocytes, lymphocytes, and monocytes. The Indian system exhibits autosomal codominant inheritance and may play a clinical role in transfusion medicine, particularly in cases of transfusion reactions or hemolytic disease of the newborn (Hemolytic Disease of the Newborn, HDFN). Although this system is rare in the general population, its presence has been reported to be more frequent in individuals of Indian and Pakistani descent, reflecting its historical name [25,27,28].
In the UniProt database, CD44 is listed under the accession number P16070 as a peer-reviewed entry within the UniProtKB/Swiss-Prot resource, designated with the entry name CD44_HUMAN. The canonical amino acid sequence of this protein includes 742 amino acid residues, while its molecular mass in the precursor form (including the signal sequence) is estimated at approximately 80–82 kDa [29]. CD44 is characterized by a high degree of structural heterogeneity, resulting from both the presence of numerous alternative isoforms (alternative splicing products) and various post-translational modifications, such as glycosylation, sulfation, and phosphorylation. This molecular variability is reflected in the diverse spectrum of biological functions that this protein performs in different cell types and physiological and pathological contexts. Many naturally occurring polymorphic variants have also been identified within the CD44 polypeptide sequence that can modify its three-dimensional structure, ligand binding ability, stability, and immunogenicity. Of particular importance are single amino acid substitutions (SAAS), some of which have been documented in both the UniProt database and the dbSNP set, and others have been associated with specific clinical phenotypes and disease states (Table 1) [12,30,31].

2.1. Biological Functions

CD44 is a transmembrane adhesion glycoprotein belonging to the class I receptor family, playing a crucial role in a wide range of biological processes, including both physiological mechanisms of homeostasis and pathological changes associated with inflammatory and neoplastic diseases [12,32]. The primary function of CD44 is to mediate interactions between cells and with the surrounding extracellular matrix (ECM). The primary ligand of this receptor is hyaluronic acid (HA), a key component of the ECM, responsible for maintaining the structural integrity and elasticity of tissues. In addition to HA, CD44 exhibits affinity for several other ECM components, including osteopontin, fibronectin, laminin, and collagens, as well as for proteolytic enzymes such as matrix metalloproteinases (MMP-2, MMP-9), which makes it an important integrator of signals from the extracellular environment [4,13,33,34,35,36,37,38,39,40,41,42] (Table 2).
In the context of the immune system, CD44 is involved in the adhesion and migration of leukocytes, enables the homing of T lymphocytes to lymphoid organs, and participates in the processes of their activation, survival, and proliferation [43,44,45]. The HCELL variant (Hematopoietic Cell E-/L-selectin Ligand) functions as a ligand for E- and L-selectins, enabling leukocytes to roll on the vascular endothelium, which is the first step in their recruitment to sites of inflammation [46,47,48,49]. CD44 also plays an essential role in hematopoiesis, progenitor cell migration, and bone marrow regeneration. Through its extracellular domain, it can bind cytokines, chemokines, proteases, and growth factors (e.g., Transforming Growth Factor Beta (TGF-β), Epidermal Growth Factor (EGF), and Vascular Endothelial Growth Factor (VEGF)), creating a local microenvironment that promotes the intensification of the cellular response [50,51,52,53]. The cytoplasmic domain of CD44, although short, binds to adaptor proteins (ezrin, radixin, and moesin), tyrosine kinases, and phospholipase C, initiating the activation of numerous signaling pathways, including the PI3K/AKT, MAPK/ERK, RhoA/ROCK, and GTPase pathways (Rac1 and Cdc42) [15,16,17,18,19,30,54,55,56,57,58,59,60,61].
CD44 activation leads to the mobilization of calcium ions, reorganization of the actin cytoskeleton, and alterations in cell adhesion and morphology, which are essential components of processes such as migration, proliferation, differentiation, and inflammatory responses [62,63,64,65,66]. In pathological conditions, particularly in cancers, CD44 plays a crucial role in promoting the invasion, migration, and metastasis of cancer cells. As a marker of cancer stem cells (CSCs), CD44 facilitates the formation of invadopodia, specialized structures that enable ECM degradation and tissue penetration [67,68,69]. Altered CD44 expression, including the presence of specific alternative isoforms (e.g., CD44v6), correlates with increased tumor aggressiveness and a poorer clinical prognosis in numerous types of cancer, including colon, breast, and gastric cancers, as well as leukemias. Disorders in CD44 function or expression lead to a wide range of abnormalities, including impaired leukocyte migration, chronic inflammation, excessive activation of fibroblasts and myofibroblasts, pathological tissue fibrosis, and increased invasiveness of cancer cells. Due to the central position of CD44 as an integrator of environmental signals and a regulator of dynamic cellular interactions, this protein remains the subject of intensive research as a potential diagnostic and prognostic biomarker and therapeutic target in numerous disease entities (Figure 1) [4,70,71,72,73,74,75].

2.2. Subcellular Localization and Expression Profile of CD44 Isoforms

CD44 tends to preferentially localize in specialized domains of the cell membrane, particularly on the surface of microvilli and other membrane protrusions that increase the cell’s surface area of contact with its environment. In dynamic conditions, such as migration or cellular reorganization processes, this receptor concentrates in regions of active cell movement, including within the structures of membrane protrusions—lamellipodia and filopodia [80,81,82]. In vitro models, particularly in the context of wound healing, have demonstrated the colocalization of CD44 with actin filaments in the anterior regions of migrating cells, suggesting its involvement in regulating directional migration and cytoskeletal reorganization [55,83].
From the cytoplasmic side, the intracellular domain of CD44 interacts with the ERM family of anchoring proteins (ezrin, radexin, moesin), which connect it to the cortical layer of the actin cytoskeleton (Table 3). These interactions provide mechanical stability to the receptor complex while simultaneously enabling the transduction of signals from the cell surface to its interior, thereby activating, among others, the Rho- and Rac-dependent GTPase pathways. CD44 is also preferentially located in the so-called lipid rafts—microdomains of the cell membrane rich in cholesterol and sphingolipids, which promote the recruitment of signaling and adhesion proteins and participate in the modulation of signal transmission [52,54,56,84,85].
In terms of tissue expression, CD44 has a wide distribution and is constitutively expressed on the surface of many cell types, including both hematopoietic and non-hematopoietic cells [91,92,93]. The canonical form of CD44 (also known as CD44s, the standard form) is abundantly expressed in immune cells, including T lymphocytes, monocytic cells, and mesenchymal stromal cells. However, the CD44 gene undergoes extensive alternative splicing, resulting in the emergence of numerous variant isoforms (CD44v), the expression of which is often tissue-specific and variable, depending on the physiological or pathological condition. For example, the isoform CD44v10, also known as the epithelial variant (CD44E), is highly expressed in epithelial cells and is described in detail in the oncology literature in the context of epithelial neoplasms (carcinoma). Its presence correlates with progression, invasiveness, and metastatic potential of many types of cancers, including breast, colon, and pancreatic cancers, making it a potential diagnostic and prognostic marker. A distinct CD44v expression profile is observed in neuroectodermal tumors, such as neuroblastoma, where reduced expression levels of specific variant isoforms have been identified, which may reflect tissue-dependent molecular differentiation and phenotypic plasticity of tumor cells [10,25,83,94,95].

2.3. Protein Structure and Isoforms

CD44 is a protein characterized by a differentiated domain architecture, the structure of which determines the receptor’s ability to participate in dynamic adhesion processes, signaling, and spatial organization of the cell (Table 4). The structure of the mature polypeptide, resulting from the cleavage of the signal sequence directing translocation to the endoplasmic reticulum, includes 722 amino acids and consists of three main functional segments: the extracellular domain, the transmembrane segment, and the cytoplasmic domain [96,97].
The most extensive part of the molecule is the extracellular domain (residues 21–649), which is responsible for direct interaction with extracellular matrix ligands. Its N-terminal part contains a conserved LINK-type domain, with a structure homologous to glycosaminoglycan-binding proteins, which ensures high affinity for HA and ligand selectivity. In the further part of the extracellular domain, there are regions encoded by alternative exons, the presence of which is the result of the differential splicing mechanism. These fragments, often rich in hydroxyl residues (serine, threonine) and proline residues, are potential sites of post-translational modifications, including O-glycosylation and the attachment of glycosaminoglycan chains (e.g., chondroitin sulfate), which functionally classifies CD44 as a transmembrane proteoglycan. Additionally, the presence of conserved N-glycosylation sites on asparagine residues affects the protein’s proper folding, conformational stability, and correct localization in the plasma membrane [25,98,99].
The extracellular domain is separated from the cytoplasmic domain by a transmembrane segment (residues 650–670), which takes the form of a hydrophobic α-helix stably anchored in the lipid bilayer. This region is rich in apolar residues, such as leucine, alanine, and valine, which are responsible for the correct placement of the molecule in the membrane and the orientation of the domains. Specifically, the N-terminus is located on the external side, while the C-terminus is positioned in the cytoplasm. The C-terminal cytoplasmic domain (residues 671–742), devoid of a permanent tertiary structure (intrinsically disordered region), shows high functional flexibility and plays a central role in signal transduction. It contains basic motifs (rich in lysine and arginine) and serine-threonine phosphorylation sites, regulating signaling activity and the stability of interactions with molecular partners. Of key importance are the bindings to adaptor proteins of the ERM family (ezrin, radexin, moesin), which enable anchoring of the receptor in membrane domains with high signaling activity (e.g., lipid rafts) and physical coupling with the actin cytoskeleton. Through these interactions, CD44 integrates signals from the cellular environment, influencing the reorganization of cytoskeletal structures, cell polarity, and its motility and invasiveness [25,30,100,101,102].

2.3.1. Alternative Splicing of the CD44 Gene and Characterization of Selected Isoforms

The CD44 gene is subject to complex transcriptomic regulation, including intensive alternative splicing, which is a key mechanism generating its structural and functional diversity. This process involves the variable inclusion of up to 10 of the 19 exons encoding the extracellular domain, as well as the use of internal splicing sites and alternative splicing of the C-terminal region, including the cytoplasmic tail. As a result of such multivariability, numerous isoforms are formed—both membrane, with different lengths and compositions of the extracellular domain, and soluble forms, lacking the transmembrane segment, which can be secreted into the extracellular space. At least 19 different isoform variants of the CD44 protein have been described in the UniProt database, although some of them remain unconfirmed at the protein level in proteomic studies (Table 5, Figure 2) [99,100,101,103].
The reference isoform is considered to be P16070-1, which includes the complete set of possible exons and encodes a 742-residue polypeptide of approximately 82 kDa. Although sometimes referred to as CD44H or the “full” form of CD44, it is not the same as the so-called standard isoform (CD44s), which in cell biology nomenclature means a simplified, shortened version devoid of variant exons. CD44s corresponds to the P16070-2 isoform and is widely expressed in hematopoietic cells, such as T lymphocytes, where it functions as a basal adhesion receptor. In addition to these two basic variants, numerous isoforms with specific tissue expression or characteristic of pathological conditions have been identified. An example is the P16070-10 isoform, known as CD44E, which occurs in epithelial cells and lacks exons 6–11; this variant often correlates with the presence and aggressiveness of epithelial neoplasms. Another isoform, CD44R2 (P16070-11), lacks exons 6–13, whereas CDw44 (isoform 12) includes a form that excludes exons 6–14, which was initially detected in reticulocytes. Another interesting variant is CD44R5 (isoform 14), in which sequences encoded by exons 6–11, 13, and 14 are excluded, which significantly affects the topology of the extracellular domain. In turn, the Hermes isoform (P16070-15), which lacks exons 6–14 and 19, has been described as an antigen involved in lymphocyte homing—the ability of lymphocytes to migrate and colonize lymphoid organs selectively. Of particular note is the P16070-19 isoform, known as CD44RC, which is a soluble form lacking a transmembrane segment that can be released into the extracellular environment; it has an increased ability to bind hyaluronic acid and can act as a so-called decoy receptor, modulating the availability of the ligand for other forms of CD44. The expression of individual CD44 isoforms is precisely regulated in a manner dependent on the stage of development, cell type, and physiological or pathological condition of the organism. In healthy somatic tissues, the standard form of the protein dominates, while isoforms containing additional exons, collectively referred to as CD44v (variant), appear with greater frequency in pathological conditions, especially in the context of cancer. Molecular observations indicate that variants containing exons v6 and v10 correlate with the intensity of invasive and metastatic features of cancer cells, which suggests their significant participation in the clinical progression of the disease [101,102,103,104,105,106,107].
Table 5. Summary of CD44 Isoforms (based on [106,108,109,110,111,112,113]). The table provides an overview of canonical and alternative CD44 isoforms generated by extensive alternative splicing events. Each isoform differs in amino acid length, molecular mass, isoelectric point, and hydropathic profile, reflecting distinct structural and functional characteristics. Specific deletions or amino acid substitutions associated with exon skipping or alternative splice donor/acceptor usage determine ligand-binding capacity, subcellular localization, and signaling potential. Collectively, the diversity of CD44 isoforms underlies their specialized roles in processes such as immune cell homing, epithelial differentiation, tissue remodeling, and cancer progression.
Table 5. Summary of CD44 Isoforms (based on [106,108,109,110,111,112,113]). The table provides an overview of canonical and alternative CD44 isoforms generated by extensive alternative splicing events. Each isoform differs in amino acid length, molecular mass, isoelectric point, and hydropathic profile, reflecting distinct structural and functional characteristics. Specific deletions or amino acid substitutions associated with exon skipping or alternative splice donor/acceptor usage determine ligand-binding capacity, subcellular localization, and signaling potential. Collectively, the diversity of CD44 isoforms underlies their specialized roles in processes such as immune cell homing, epithelial differentiation, tissue remodeling, and cancer progression.
IsoformLength (aa)Mass (Da)pI% Hydrophilic% HydrophobicDescription
P16070-174281,5384.9857.95%42.05%Canonical isoform.
P16070-2 (CD44SP)2933279.3733.33%66.67%Lacks exons 6–14. Differences: 23–29: DLNITCR → GVGRRKS; 30–742: deleted.
P16070-371177,9835.558.23%41.77%Alternative splice donor/acceptor in exon 5. Differences: 192 G→A; 193–223 deleted.
P16070-4 (Epidermal)69976,6124.9468.24%31.76%Lacks exon 6. Differences: 223 T→S; 224–266 deleted.
P16070-573480,7904.9867.71%32.29%Alternative splice donor/acceptor in exon 7. Differences: 266–273 deleted.
P16070-669976,7054.9567.38%32.62%Lacks exon 10. Differences: 385 I→T; 386–428 deleted.
P16070-771378,4465.0560.17%39.83%Lacks exon 13. Differences: 506 Q→R; 507–535 deleted.
P16070-867474,3884.8860.83%39.17%Lacks exon 14. Differences: 536 N→R; 537–604 deleted.
P16070-967574,1964.9161.19%38.81%Lacks exon 19. Differences: 675 R→S; 676–742 deleted.
P16070-10 (CD44E, CD44R1, Epithelial, Keratinocyte)49353,4115.0266.13%33.67%Lacks exons 6–11. Differences: 223 T→N; 224–472 deleted.
P16070-11 (CD44R2)42946,5655.2456.88%43.12%Lacks 223–535 region.
P16070-12 (CDw44, Reticulocyte)36139,4165.0450.97%49.03%Lacks exons 6–14. Differences: 223 T→R; 224–604 deleted.
P16070-13 (CD44R4)42546,2614.8659.06%40.94%Lacks exons 6–11 and 14. Differences: 223 T→N; 224–472 and 537–604 deleted; 536 N→R.
P16070-14 (CD44R5)39643,1694.9959.60%40.40%Lacks exons 6–11, 13 and 14. Differences: 223 T→N; 224–472, 507–535 and 537–604 deleted; 506 Q→R; 536 N→R.
P16070-15 (Hermes)29432,0754.8654.76%45.24%Lacks exons 6–14 and 19. Differences: 223 T→R; 224–604 and 676–742 deleted; 675 R→S.
P16070-1666873,1505.0360.63%39.37%Alternative splice donor/acceptor on exon 5; lacks exon 10. Differences: 192 G→A; 193–223 and 386–428 deleted; 385 I→T.
P16070-1769175,9574.9567.15%32.85%Alternative splice donor/acceptor on exon 7; lacks exon 10. Differences: 266–273 and 386–428 deleted; 385 I→T.
P16070-1834037,2785.1561.47%38.53%Differences: 223 T→R; 224–604 and 605–625 deleted.
P16070-19 (CD44RC)13915,6357.7361.87%38.13%Soluble isoform; enhanced HA binding. Differences: 78–139 replaced with SLHCSQQSKK...QGVVRNSRPVYDS; 140–742 deleted.
Figure 2. Structural organization of CD44 isoforms resulting from alternative splicing. Diagram shows the canonical CD44 isoform (P16070-1) and numerous alternatively spliced variants, showing their exonic composition within the N-terminal domain, the variable region (alternative splicing), and the C-terminal domain. Exons encoding the constant N-terminal region (S1–S5, blue) are present in all major variants, while the central variable region (V2–V10, purple/green) reflects the high splicing diversity. The C-terminal region (S6–S10, blue) is conserved in most isoforms, accounting for the transmembrane and cytoplasmic domains. Green labels indicate exons with alternative splice donor/acceptor sites. Individual isoforms (e.g., CD44v2, CD44v4, CD44v6, CD44R1, Hermes) differ in the characteristic presence or absence of selected exons, which determines their diverse biological functions, such as adhesion, cell migration, regulation of the immune response or cancer progression (based on [106,108,109,110,111,112,113]).
Figure 2. Structural organization of CD44 isoforms resulting from alternative splicing. Diagram shows the canonical CD44 isoform (P16070-1) and numerous alternatively spliced variants, showing their exonic composition within the N-terminal domain, the variable region (alternative splicing), and the C-terminal domain. Exons encoding the constant N-terminal region (S1–S5, blue) are present in all major variants, while the central variable region (V2–V10, purple/green) reflects the high splicing diversity. The C-terminal region (S6–S10, blue) is conserved in most isoforms, accounting for the transmembrane and cytoplasmic domains. Green labels indicate exons with alternative splice donor/acceptor sites. Individual isoforms (e.g., CD44v2, CD44v4, CD44v6, CD44R1, Hermes) differ in the characteristic presence or absence of selected exons, which determines their diverse biological functions, such as adhesion, cell migration, regulation of the immune response or cancer progression (based on [106,108,109,110,111,112,113]).
Ijms 26 08870 g002

2.3.2. Protein Interactions and Complexes

CD44 acts as a dynamic transmembrane receptor that integrates signals from the extracellular and intracellular spaces. Its extracellular domain enables the binding of extracellular matrix components, such as hyaluronic acid, collagens, osteopontin, or fibronectin, which promote cell adhesion, migration, and communication with the environment [107,114]. The cytoplasmic domain of CD44 interacts with cytoskeleton-anchoring proteins (e.g., ERM, merlin), kinases, and GTPase regulatory proteins, enabling the activation of intracellular signaling pathways. CD44 also forms complexes with other membrane receptors (e.g., EGFR, PDPN) and coreceptors (CD74), which affect cell proliferation, survival, and motility in response to environmental signals [108,109,115,116,117,118,119]. Through these complex interactions, CD44 acts as a central signaling platform, integrating the mechanisms of adhesion, migration, cytoskeletal regulation, and cellular response to external stimuli (Figure 3).

2.3.3. Post-Translational Modifications (PTMs)

The CD44 protein is subject to numerous post-translational modifications (PTMs) that are crucial for its localization, stability, molecular interactions, and biological functions. The most important of these include glycosylation, phosphorylation, and proteolysis, as well as rarer modifications such as ubiquitination. Variability in the type, location, and intensity of these modifications contributes to the considerable functional heterogeneity of CD44, especially in the context of tumor pathologies and interactions with the extracellular matrix (Table 6).

3. The Role of CD44 in Pathologies

In addition to its physiological functions, CD44 plays a crucial role in the pathogenesis of neoplastic, autoimmune, and inflammatory diseases, where its expression and function change. Different CD44 isoforms (standard and variant) play various roles depending on the disease context [12,129,141,142,143,144] (Table 7).
In the context of neoplastic diseases, CD44 acts in a multidirectional manner, promoting the malignant phenotype of cells. Overexpression of this protein is common in tumors showing features of invasiveness, resistance to treatment, and high metastatic potential. CD44-positive cells are characterized by the activation of signaling pathways that increase their survival, migration, and resistance to environmental stress, especially in hypoxic conditions and during cytotoxic therapies. CD44 interacts with the tumor microenvironment, influencing its remodeling, and also supports the formation of pre-metastatic niches through adhesion to the extracellular matrix and localization of proteases, such as MMP-9, on the cell surface [145,146].
Variant isoforms, including CD44v6 and CD44v3, play an additional role—they participate in angiogenesis, activation of MET and VEGFR2 receptors, and binding of growth factors using heparan sulfate residues [147,148]. Switching splicing from the standard form (CD44s) to the variant or vice versa is closely correlated with the stage of cancer progression, e.g., in colon or liver cancer. Overexpression of CD44, regardless of its form, is often associated with shorter patient survival, more advanced TNM stages, and a higher risk of metastases, making it not only a prognostic marker but also a potential target for molecularly targeted therapy [149,150,151]. In pathologies with an immune basis, CD44 plays a crucial role in the pathomechanisms of chronic inflammation, primarily by facilitating the migration, activation, and retention of leukocytes in affected tissues. In rheumatoid arthritis, increased expression of CD44—both on immune cells and synovial fibroblasts—promotes the perpetuation of inflammatory infiltrate and cartilage degradation. Antibodies against CD44 reduce the migration capacity of fibroblasts and alleviate symptoms of inflammation in animal models [149,150,151,152,153,154,155,156].
In lupus nephritis (LN), the presence of CD44 on tubular epithelial cells and infiltrating leukocytes is associated with increased inflammation and fibrosis. Inhibition of CD44 activity in a mouse model led to reduced proteinuria, collagen deposition, and lymphocytic infiltration [140,157,158]. Furthermore, serum levels of the soluble form of CD44 (sCD44) are a useful biomarker of disease activity, with its increase preceding clinical exacerbations, suggesting its potential use in monitoring patients. In inflammatory bowel disease (IBD), such as Crohn’s disease and ulcerative colitis, increased expression of CD44 variants—particularly CD44v3, CD44v6, and CD44v7—has been reported in the intestinal mucosa. Functionally, CD44v7 interactions with osteopontin promote IL-6 production by macrophages, which in turn supports inhibition of T regulatory cell differentiation and aggravates inflammation. Blocking this interaction in animal models reduces the severity of colitis and restores immune balance. As a result, the CD44v7–osteopontin pathway represents a promising target for novel targeted therapies in IBD [129,141,142,143,144,145,159].
Table 7. Pathological role of CD44 in selected disease entities (based on [74,135,146,147,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174]). The table highlights the diverse pathological functions of CD44 across malignant and inflammatory diseases. Distinct isoforms, including CD44s, CD44v, and soluble CD44, contribute to tumor initiation, invasion, therapy resistance, and immune dysregulation through interactions with HA, osteopontin, growth factor receptors, and proteases. These mechanisms involve activation of STAT3, PI3K/Akt, and other signaling cascades, supporting processes such as epithelial–mesenchymal transition, fibrosis, or chronic inflammation. Clinically, CD44 expression and isoform profiles serve as prognostic and predictive biomarkers and represent potential therapeutic targets in both oncology and autoimmune disorders.
Table 7. Pathological role of CD44 in selected disease entities (based on [74,135,146,147,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174]). The table highlights the diverse pathological functions of CD44 across malignant and inflammatory diseases. Distinct isoforms, including CD44s, CD44v, and soluble CD44, contribute to tumor initiation, invasion, therapy resistance, and immune dysregulation through interactions with HA, osteopontin, growth factor receptors, and proteases. These mechanisms involve activation of STAT3, PI3K/Akt, and other signaling cascades, supporting processes such as epithelial–mesenchymal transition, fibrosis, or chronic inflammation. Clinically, CD44 expression and isoform profiles serve as prognostic and predictive biomarkers and represent potential therapeutic targets in both oncology and autoimmune disorders.
Disease/Cancer TypeRole of CD44CD44 IsoformsMechanism of ActionClinical Significance
Breast cancerCSC marker, EMT, therapy resistanceCD44s, CD44^highSTAT3 and PI3K/Akt activation; splicing toward CD44sPoor prognosis, tumor recurrence
Pancreatic cancerInvasion, metastasisCD44v6MMP-9 localization, interactions with HAPromotes metastasis, reduces overall survival (OS)
Lung cancerProliferation, chemoresistanceCD44vMET/VEGFR2, PI3K/Akt signalingAccelerated tumor growth
Rheumatoid arthritis (RA)Leukocyte adhesion, fibroblast activationCD44v (various)HA interactions, pannus formationCD44 blockade reduces inflammation
Lupus nephritis (LN)Inflammation, renal fibrosisCD44s, sCD44CD4+, CD19+ recruitment, fibroblast activationDisease activity biomarker, therapeutic target
Crohn’s diseaseIL-6 production, Treg deficiencyCD44v7Interaction with osteopontinCD44v7 blockade protects against colitis

3.1. CD44 as a Regulator of Tissue Fibrosis and Myofibroblast Differentiation

Fibroblast differentiation into myofibroblasts is an essential step in the repair and fibrotic processes that occur in response to tissue damage. Myofibroblasts, through the expression of α-smooth muscle actin (α-SMA) and contractile properties, are capable of remodeling the ECM and restoring tissue integrity (Figure 4) [175,176,177,178]. However, their sustained activation and excessive numbers underlie many fibrotic pathologies, such as idiopathic pulmonary fibrosis, liver cirrhosis, and restrictive cardiomyopathies. The central factor inducing this phenotype is TGF-β1, which activates the canonical Smad2/3 signaling pathway, leading to the expression of myofibroblast-specific genes. However, increasing evidence suggests that the presence of TGF-β1 alone is insufficient to activate the differentiation program fully. Simultaneous signaling from HA and its primary receptor, the glycoprotein CD44, which acts as a coreceptor to amplify and modulate signal transmission, is necessary [179,180,181]. CD44, as an HA receptor, participates in the formation of a dynamic signaling structure on the surface of fibroblasts. The presence of high-molecular-weight HA and its binding to CD44 enables the localization of the receptor complex in lipid rafts of the cell membrane—specialized domains rich in cholesterol and sphingolipids that act as signaling platforms. In these microdomains, CD44 co-localizes with other receptors, including EGFR, which triggers non-canonical signaling pathways independent of Smads. It has been demonstrated that inhibition of HA synthesis (e.g., by knocking down HAS2 synthase) or blocking CD44 function prevents the activation of α-SMA, even in the presence of TGF-β1, confirming the necessity of the HA-CD44 axis in this process. Moreover, HA level affects the presence of CD44 in lipid rafts, thereby conditioning the efficiency of the entire signal transduction process [175,182,183,184].
Despite lacking its kinase activity, CD44 plays a crucial role in initiating signaling cascades by forming dynamic complexes with other membrane receptors and adaptor proteins. One of the most critical mechanisms of CD44 signaling activation is its functional cooperation with the EGFR, which is conditioned by the presence of HA and TGF-β1. In response to these signals, CD44 and EGFR co-localize in lipid rafts—specialized microdomains of the cell membrane rich in cholesterol and sphingolipids, which are platforms for concentrating signaling components. The formation of the CD44–EGFR complex within lipid rafts is necessary for the activation of intracellular pathways leading to the differentiation of fibroblasts into myofibroblasts [185,186]. Disruption of this organization, by destabilizing rafts or disrupting CD44 binding to HA, results in disorganized signaling and inhibition of further transmission. Signal initiation in rafts first leads to the activation of MAPK/ERK kinase, and then to the activation of calmodulin-dependent kinase II (CaMKII). It has been experimentally demonstrated that ERK phosphorylation is a prerequisite for CaMKII activation, and the blockade of either of these pathways effectively inhibits the expression of myofibroblast markers, such as α-SMA, and prevents complete differentiation. The discussed CD44–EGFR–ERK/CaMKII cascade acts in parallel and synergistically with the classical Smad2/3 pathway, activated by TGF-β receptors. Integrated signaling from both pathways enables the maximal activation of pro-fibrotic genes and the full phenotypic transformation of fibroblasts into contractile, ECM-modifying myofibroblasts [167,176,187,188,189].
Notably, the functionality of lipid rafts appears to be critical for effective CD44-dependent signaling. Restriction of HA synthesis—e.g., by knockdown of the HAS2 gene—leads to impaired localization of CD44 in these microdomains and reduced ERK/CaMKII signaling. CD44 can also interact with other signaling proteins, such as ERM family proteins (ezrin, radexin, moesin), which link it to the actin cytoskeleton and enhance cellular stability [30].
Figure 4. The multifunctional role of CD44 in fibrosis and myofibroblast differentiation (based on [175,179,180,181,182,183,184,185,186,190]). The figure illustrates the pleiotropic functions of CD44 during fibrotic processes, emphasizing its dual role as both a promoter of inflammation and an inhibitor of myofibroblast differentiation. CD44 regulates fibroblast and immune cell recruitment, ECM remodeling, and collagen metabolism through interactions with hyaluronic acid, ERM proteins, and metalloproteinases. Its activity is highly context-dependent, influenced by tissue type, molecular environment, and the presence of mediators such as TGF-β or inflammatory factors. Moreover, specific molecular interactions—including hyaluronan binding, pentraxin 3–dependent signaling, and Wnt/β-catenin pathway regulation—shape CD44’s impact on cell migration, survival, proliferation, and myofibroblast differentiation, ultimately determining the balance between tissue repair and pathological fibrosis.
Figure 4. The multifunctional role of CD44 in fibrosis and myofibroblast differentiation (based on [175,179,180,181,182,183,184,185,186,190]). The figure illustrates the pleiotropic functions of CD44 during fibrotic processes, emphasizing its dual role as both a promoter of inflammation and an inhibitor of myofibroblast differentiation. CD44 regulates fibroblast and immune cell recruitment, ECM remodeling, and collagen metabolism through interactions with hyaluronic acid, ERM proteins, and metalloproteinases. Its activity is highly context-dependent, influenced by tissue type, molecular environment, and the presence of mediators such as TGF-β or inflammatory factors. Moreover, specific molecular interactions—including hyaluronan binding, pentraxin 3–dependent signaling, and Wnt/β-catenin pathway regulation—shape CD44’s impact on cell migration, survival, proliferation, and myofibroblast differentiation, ultimately determining the balance between tissue repair and pathological fibrosis.
Ijms 26 08870 g004

3.2. In Vitro and In Vivo Experimental Evidence for the Role of CD44 in Fibrosis (Lung, Skin, Heart, Liver)

In numerous experimental models, activation of the HA–CD44 pathway has been shown to promote the transformation of fibroblasts into myofibroblasts, as well as to maintain their pro-fibrotic phenotype. This relationship has been confirmed both in vitro studies on fibroblasts isolated from human and animal tissues, as well as in vivo studies in animal models of fibrosis of various organs. Notably, interventions targeting CD44, such as its genetic silencing or antibody blockade, effectively limit collagen deposition, reduce fibroblast activation, and slow the progression of fibrosis (Table 8).
Accumulating preclinical data from various in vitro and in vivo models indicate that modulation of CD44 activity, both through genetic interventions and pharmacological approaches, significantly affects the course and intensity of fibrotic processes in numerous organs. These results support the view that CD44 is not only a marker of the profibrotic phenotype but also plays a role as an actively involved mediator of fibrosis pathogenesis, integrating extracellular with intracellular transduction pathways that lead to fibroblast activation, myofibroblast induction, and the overproduction of ECM components.
In several animal models, the genetic deletion of CD44 (Cd44/) has been shown to attenuate the fibrotic response significantly. For example, Cd44 knockout mice are protected against bleomycin-induced lung fibrosis as well as against myocardial fibrotic remodeling induced by chronic angiotensin II exposure. In both cases, there was a decrease in myofibroblast and macrophage activity, limited collagen accumulation, and reduced expression of pro-fibrotic genes, clearly indicating the crucial role of CD44 in amplifying pro-fibrotic signals in the injured tissue environment [198,199,200].
It is worth emphasizing that similar effects were also obtained using the CD44 immunoneutralization strategy. Administration of monoclonal antibodies directed against CD44 (e.g., clone IM7) to mice resulted in a significant attenuation of the progression of liver and lung fibrosis, which was accompanied by a reduction in the expression of activated myofibroblast markers (e.g., α-SMA) and a decrease in the collagen content in the ECM [201]. Equally promising results were achieved by pharmacological intervention at the level of the CD44 ligand–hyaluronic acid. In models of renal, hepatic, and pulmonary fibrosis, the use of 4-methylumbelliferone (4-MU)—an inhibitor of HA synthesis—led to a reduction in its deposition in the tissue and indirect quenching of CD44 signaling. In particular, in a model of lupus nephritis, both 4-MU and anti-CD44 antibody were shown to reduce the inflammatory infiltrate, limit interstitial fibrosis, and preserve the architecture of the glomeruli and tubules, without affecting the degree of cytotoxic hepatocyte damage or the intensity of inflammation [202,203,204,205]. In addition, in models of fibrosis induced by toxic agents (e.g., CCl4) and by hemodynamic factors (e.g., venous congestion in the IVC ligation model), it has been shown that blockade of the HA–CD44 axis translates into limited activation of stellate cells, reduced expression of markers such as S100A4 or TGF-β1, and consequently—limited progression of fibrosis [206]. Therefore, the possibility of therapies targeting the HA–CD44 pathway is on the horizon, whether through the administration of anti-CD44 antibodies, competitive peptides blocking HA binding, inhibition of HA synthesis enzymes, or modulation of alternative CD44 splicing (e.g., variant forms associated with pathology). In diseases such as idiopathic pulmonary fibrosis, where there are currently no effective drugs to reverse the scarring process, intervening in the CD44–HA interaction is a promising new strategy. CD44 has already been proposed as a biomarker of fibrosis activity (e.g., in congestive hepatopathy or scleroderma) and as a target for drugs that inhibit disease progression. High CD44 expression and HA production are observed not only in disease but also in regenerative processes, especially in the skin [207,208]. Therefore, anti-CD44 therapies must be carefully designed to precisely target pathological fibrogenesis niches, thereby limiting long-term CD44 blockade in healthy tissues. Further understanding of the complexity of CD44 signaling (dependent on tissue context and cell type) is needed to develop safe and effective therapeutic approaches. However, the data to date are encouraging—CD44 is emerging as a key regulatory node in the fibrosis signaling network and as a promising target, the modulation of which may benefit patients with progressive fibrotic diseases, for whom treatment options are currently limited.

3.3. CD44 and the Invasive Phenotype of Myofibroblasts—Analogies to EMT in Cancer

Advanced stages of fibrotic processes are characterized by the emergence of a population of myofibroblasts with an invasive phenotype, which acquire the ability to migrate actively, degrade the ECM, and penetrate surrounding tissue structures. This aggressive phenotype not only maintains the chronic profibrotic state but also promotes spatial expansion of fibrosis and its progression towards adjacent, previously unchanged organ areas [192,209,210,211].
What is particularly important from the point of view of pathobiological mechanisms is that the behavior of invasive myofibroblasts during advanced fibrosis exhibits striking similarities to the phenotype of cancer cells undergoing EMT. This fundamental phenomenon facilitates the migration, invasion, and formation of metastases by cancer cells. In both cases, increased cell motility, cytoskeletal reorganization, degradation of the basement membrane, and reprogramming of cell adhesion genes, such as E-cadherin, N-cadherin, and vimentin, are observed [212,213,214].
Fibroblasts isolated from the fibrotic lungs of patients with IPF exhibit a highly invasive phenotype that resembles that of EMT cells in terms of their behavior. They can degrade ECM components, cross the basement membrane barrier, and move toward chemotactic gradients—characteristics of transformed cancer cells. A key molecular factor enabling this phenotype is signaling initiated by the HA–CD44 complex. Blocking CD44 or interrupting its interaction with HA (e.g., by using neutralizing antibodies) has been shown to effectively abolish the ability of myofibroblasts to degrade matrix and migrate, thereby inhibiting the progression of fibrosis [214,215,216,217].
An analogous mechanism is observed in the context of malignant tumor progression. During EMT, induced, among other things, by the action of TGF-β1, epithelial cells acquire the features of mesenchymal cells, which allows them to detach from the primary tumor focus and initiate invasion. In many tumor models (including breast, lung, and pancreatic cancer), TGF-β1 has been shown to induce de novo HA synthesis and CD44 expression, which leads to the formation of the HA–CD44 complex and its interaction with kinase receptors such as EGFR. This signaling axis leads to the activation of the PI3K/AKT and MAPK/ERK pathways, as well as cytoskeletal reorganization, resulting in the loss of epithelial features, increased invasiveness, and the acquisition of the complete EMT phenotype [216]. In cancer cells undergoing EMT, experimental inhibition of HA synthesis (e.g., 4-MU) or CD44 silencing (e.g., by RNAi) leads to the suppression of EGFR activity and downstream signaling pathways (ERK, AKT), resulting in the reversal of the EMT phenotype and attenuation of cell invasiveness [218,219,220].

4. Impact of Environmental Pollutants on CD44 in Connective Tissue

Environmental factors, including heavy metals, particulate matter, endocrine-disrupting compounds (EDCs), and microplastics, can impact the function and expression of CD44, thereby modulating inflammatory and fibrotic processes in connective tissue. The characteristic tissue response to these factors is chronic damage with concomitant inflammation and extracellular matrix remodeling.

4.1. Heavy Metals

Exposure to toxic metals (e.g., lead, cadmium) disrupts the homeostasis of the immune system and connective tissue functions, including the regulation of leukocyte adhesion molecules. It has been observed that chronic exposure to lead leads to a decrease in CD44 expression on the surface of blood cells, for example, in children living in areas contaminated with electronic waste (e-waste). High blood Pb levels have been correlated with lower expression of CD44 and CD58 adhesion molecules on erythrocytes [200,201]. This may impair the ability of blood cells to interact with the endothelium and matrix, weakening proper repair and immune responses. On the other hand, in response to damage caused by heavy metals (such as oxidative stress and cytotoxicity), the body triggers a chronic inflammatory state, characterized by an influx of immune cells. Macrophages and neutrophils accumulating at metal accumulation sites typically exhibit high expression of CD44, which is essential for their adhesion and migration in tissues rich in hyaluronan. It can therefore be assumed that heavy metals indirectly enhance signaling through CD44, e.g., by inducing the release of HA fragments in damaged tissues (HA then acts as a DAMP activating the immune response). The aforementioned population studies in children from e-waste areas have indeed shown an association between increased levels of metals (Pb, Cd) and reduced expression of adhesion molecules (CD44, CD58) on blood cells, which indicates a chronic effect of metals on the hematopoietic and immune systems. In summary, heavy metals can both directly change the level of CD44 on cells (e.g., reduction in CD44 on erythrocytes in lead poisoning) and indirectly, through induced inflammation, increase the demand for CD44 functions in repair processes. Paradoxically, chronic activation of the CD44 pathway can promote uncontrolled fibrosis (e.g., scarring in organs after metal poisoning) [202,203,204].

4.2. Particulate Matter (PM)

Air pollutants, especially fine PM2.5 dust particles and crystalline silica (e.g., mine dust), have a documented ability to cause chronic inflammation in the airways and lung fibrosis. CD44 plays a crucial role in the lung’s response to inhaled particles. For example, during exposure to ozone, a strong oxidant present in smog, recruitment of inflammatory cells to the lungs requires the interaction of CD44 with hyaluronic acid. Studies in mice have shown that individuals lacking CD44 are protected from ozone-induced bronchial hyperresponsiveness despite a similar increase in HA concentration in the lungs [205,206,207,208]. This means that damage to lung tissue by ozone leads to the release of HA fragments, which then signal via CD44 on macrophages and neutrophils, facilitating their influx into the alveoli. In the case of silica dust (causing silicosis), chronic inhalation leads to a gradual, fibrotic remodeling of the lungs. As described earlier, this process is mainly dependent on the HA–CD44 pathway. Studies in a mouse model have shown that blockade of CD44 (with the IM7 antibody) alleviates the effects of silica dust, fibroblast activation, and collagen deposition in the lung, and respiratory function is improved. This suggests that dust interaction with the lung initiates an inflammatory-repair cascade mediated by CD44. First, acute inflammation (characterized by the influx of CD44+ cells) occurs, followed by chronic fibrosis (resulting from CD44-dependent activation of fibroblasts). Interestingly, finer dust particles can penetrate deeper into the respiratory system and induce a more severe inflammatory-fibrotic response. It has been found that the smaller the particles (even on the nanoscale), the greater their toxicity and ability to induce fibrosis—this is due to their larger surface area relative to mass and the potential to generate oxidants. The effect, among others, is a stronger polarization of the immune response towards Th2 and more intensive matrix deposition in the lungs after exposure to fine dust. In addition, suspended dust can induce epigenetic changes in cells. For example, PM2.5 particles can modulate DNA methylation and other epigenetic modifications, thereby affecting the expression of various genes. It is possible that this also includes CD44 variant genes, although this aspect requires further research. In summary, environmental particulate matter acts as a trigger for chronic inflammation and fibrosis (especially in the lungs), and CD44 is one of the key receptors involved in the development and maintenance of these pathological processes [192,208,209,210,211].

4.3. Endocrine-Disrupting Compounds (EDCs)

EDCs include pesticides, bisphenols (e.g., BPA), phthalates—substances capable of interfering with the endocrine system and causing abnormal metabolic and inflammatory responses. Increasing evidence suggests that exposure to EDCs can stimulate processes characteristic of EMT (epithelial–mesenchymal transition) and fibrosis in various tissues. For example, bisphenol A (BPA) and styrene derivatives can induce EMT in epithelial cells of the respiratory and reproductive systems. Under the influence of these compounds, epithelial cells undergo a transition to a mesenchymal phenotype, becoming more migratory and invasive [211,212]. Since the EMT is accompanied by increased CD44 expression, it is logical to assume that EDCs may secondarily increase the activity of the CD44 pathway in the tissues affected by their action. In addition to inducing EMT, EDCs promote chronic low-grade inflammation [213,214,215]. For example, in people with high blood levels of phthalates, elevated inflammatory markers and matrix remodeling in the liver (nonalcoholic fatty liver disease with fibrosis) are observed [213]. Experimental studies confirm this correlation—chronic exposure to low doses of the phthalate DEHP in mice leads to increased liver damage, inflammatory infiltration (leukocyte infiltration), and increased collagen deposition [214]. Similar changes occur in the adipose tissue of obese people, where some EDCs accumulate. There, increased synthesis of hyaluronic acid and osteopontin was observed, along with enhanced expression of CD44 on macrophages infiltrating adipose tissue [215,216]. Moreover, some EDCs can directly modulate CD44 expression. For example, BPA has been shown to activate the estrogen-related receptor (ERRγ) in A549 lung cancer cells, which is associated with the induction of EMT [218,219]. This suggests that substances with estrogenic activity (such as parabens, alkylphenols) may potentially increase CD44 levels in target cells responding to estrogen signals. Although the effect of specific EDCs on the CD44 pathway requires further study, it is already clear that EDCs contribute to chronic inflammation and fibrosis in tissues, where CD44 acts as a mediator, participating in immune infiltration and phenotypic changes (e.g., EMT) of cells exposed to these compounds.

4.4. Microplastics

Microscopic plastic particles (originating from degradation of packaging, textiles, or tire abrasion, among others) are a ubiquitous environmental pollutant. They can enter the body through ingestion or inhalation. It has been proven that the accumulation of microplastics in tissues causes inflammatory reactions and fibrosis and also disrupts the composition of the microbiome [219,220,221]. The immune system recognizes these particles as foreign bodies; at the site of their deposition, macrophage recruitment occurs, and granulomas form (analogous to the inhalation of silica dust or asbestos fibers) [222,223,224,225]. Fibroblasts and myofibroblasts participate in the formation of granulation tissue and the encystation of microplastics, the migration and activation of which depend, among other things, on the interaction of CD44 with the matrix rich in hyaluronan. It can be predicted that in places of microplastic accumulation, there is increased expression of CD44 on macrophages (which form HA-rich granulation tissue) and on fibroblasts that build a fibrous capsule around foreign particles. Long-term presence of microplastics may maintain a chronic inflammation—continuous influx of monocytes and lymphocytes sustains the inflammatory response, which is mediated by signaling through CD44 and its ligands (HA, osteopontin, etc.). Moreover, microplastics often act as carriers for other contaminants (e.g., heavy metals, polychlorinated biphenyls), enhancing the combined toxic effect [226,227,228]. An analogy can be drawn to silicosis—just as silica dust induces lung fibrosis (silicosis) via CD44-dependent mechanisms, microplastics can induce “plasticosis,” characterized by chronic inflammation and scarring of organs (e.g., lungs, gastrointestinal tract) following the accumulation of plastics [229]. This is a newly defined disease term, but case reports (e.g., in seabirds eating plastic) confirm the occurrence of extensive scarring and fibrotic tissue remodeling in the presence of microplastics. This suggests the same molecular pathways as in other forms of chronic inflammation around a foreign body, with a significant contribution from CD44 signaling (Table 9).

5. Limitations and Future Research Directions

Despite the abundant evidence indicating the vital role of CD44 in the pathogenesis of tissue fibrosis and inflammatory responses, the current state of knowledge is primarily based on preclinical models, mainly animal and in vitro studies. There is a lack of extensive translational and clinical studies confirming the efficacy and safety of CD44 targeting in humans. Most interventions (e.g., neutralizing antibodies, gene knockouts, or HA synthesis inhibitors) have been performed under controlled conditions, which limits their direct translation into the complex environment of the human body.
In addition, the heterogeneity of CD44 isoforms (CD44s, CD44v3, v6, v7, etc.) and their dynamic expression, which varies depending on the cell type, inflammation, or microenvironment, makes it challenging to determine which form of the molecule is the key pathogenic factor. Most studies do not distinguish between isoforms in analyses, which may mask subtle but critical functional differences.
Another limitation is the insufficient understanding of the long-term effects of modulating the CD44–HA axis. Although inhibition of this pathway is beneficial in fibrosis models, it is unclear whether it also disrupts physiological healing, regeneration, or immune responses. Furthermore, the influence of environmental pollutants on CD44 expression and function has been described mainly in a correlative context. There is a need for cause-and-effect studies that will unequivocally confirm the participation of this receptor in mediating the harmful effects of factors such as microplastics, EDCs, or heavy metals. In light of the growing interest in targeted therapies in chronic diseases, CD44 appears as a promising molecular target in the context of both antifibrotic and immunomodulatory interventions. Future studies should focus on the development of selective inhibitors of CD44 isoforms that will allow for precise modulation of its activity without interfering with the physiological functions of this receptor in regenerative and immune processes. It will also be crucial to develop and validate biomarkers based on CD44 expression profiles—both its membrane and soluble forms (sCD44)—and hyaluronic acid (HA) levels, which will enable dynamic monitoring of fibrosis progression and response to treatment. A key direction of development is the use of combined therapies, including simultaneous inhibition of the CD44–HA axis and canonical TGF-β/Smad pathways, which may yield a synergistic effect in limiting uncontrolled matrix remodeling. It will also be critical to deepen our understanding of the interactions between environmental factors and the regulation of CD44 expression, taking into account the influence of epigenetic modifications and tissue microbiota disorders. Further studies should also include population and clinical assessments of the efficacy and safety of CD44 inhibitors and HA antagonists (such as 4-methylumbelliferone) in patients with idiopathic pulmonary fibrosis, liver cirrhosis or systemic sclerosis. In parallel, it is worth exploring analogies with oncogenesis, including the mechanisms of EMT and acquisition of an invasive phenotype, which may enable translation of therapeutic strategies used in oncology (e.g., anti-CD44 antibodies) to the treatment of fibrogenic diseases. An integrated approach, combining molecular, toxicological, environmental, and clinical studies, may contribute to a more comprehensive understanding of the role of CD44 in connective tissue homeostasis and pathology and initiate a new generation of therapies targeting this multifunctional receptor.

6. Conclusions

CD44 is an essential pathophysiological factor in the processes of fibrosis and remodeling of connective tissues. Its activity, dependent on interaction with hyaluronic acid and the presence of specific signaling pathways (EGFR, MAPK, Smad), affects fibroblast differentiation, inflammatory cell migration, myofibroblast formation, and intensification of the inflammatory response. Experimental models of lung, skin, heart, and liver fibrosis demonstrate that genetic deletion of CD44, its pharmacological blockade, or inhibition of HA biosynthesis leads to inhibition of the progression of fibrogenic changes. At the same time, the similarity between the phenotypes of invasive myofibroblasts and cancer cells after EMT emphasizes the commonality of mechanisms driving aggressive tissue remodeling. Finally, environmental factors—from heavy metals to microplastics—modulate CD44 activity and may enhance its pathogenic effect by stimulating inflammation, EMT, and matrix overproduction. The collected data identify CD44 as a potential target for therapeutic interventions in fibrotic diseases, as well as a biomarker for assessing exposure to environmental factors.

Author Contributions

Conceptualization, P.C.-W., A.P.-W., A.G., E.Z., A.B., K.B., P.M., S.M. and E.G.; methodology, A.P.-W.; validation, A.G. and P.M.; formal analysis, A.G.; investigation, P.C.-W. and A.B.; resources, E.Z.; data curation, A.P.-W.; writing—original draft preparation, P.C.-W., A.P.-W., A.G., M.L.-S., E.Z., A.B., K.B., P.M., S.M., K.K.-K. and E.G.; writing—review and editing, P.M., S.M., M.R.-H., T.U. and E.G.; visualization, P.M., S.M., T.U. and M.R.-H.; supervision, E.G.; project administration, E.G.; funding acquisition, M.R.-H., T.U. and E.G. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the University of Applied Sciences in Tarnów.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were generated or analyzed in this study. All data cited and discussed are publicly available from the original publications referenced in the manuscript.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
CD44Cluster of Differentiation 44
HAHyaluronic Acid
TGF-βTransforming Growth Factor Beta
EGFEpidermal Growth Factor
VEGFVascular Endothelial Growth Factor
PI3KPhosphoinositide 3-Kinase
AKTAKT Serine/Threonine Kinase
MAPKMitogen-Activated Protein Kinase
RhoARas Homolog Family Member A
ROCKRho-associated Protein Kinase
ERMEzrin–Radixin–Moesin
EMTEpithelial–Mesenchymal Transition
CSCCancer Stem Cell
ECMExtracellular Matrix
MMPMatrix Metalloproteinase
STAT3Signal Transducer and Activator of Transcription 3
NF-κBNuclear Factor Kappa-light-chain-enhancer of Activated B Cells
ALTAlanine Aminotransferase
SScSystemic Sclerosis
IPFIdiopathic Pulmonary Fibrosis
RZSRheumatoid Arthritis (RA)
LNLupus Nephritis
EDCsEndocrine Disrupting Chemicals
BPABisphenol A
DEHPDi(2-ethylhexyl) Phthalate
ERRγEstrogen-Related Receptor Gamma
PM2.5Particulate Matter ≤ 2.5 µm
4-MU4-Methylumbelliferone
OSOverall Survival
KOKnockout
sCD44Soluble CD44
DAMPDamage-Associated Molecular Pattern
PCBPolychlorinated Biphenyls

References

  1. Kamrani, P.; Marston, G.; Arbor, T.C.; Jan, A. Anatomy, Connective Tissue. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2025. [Google Scholar]
  2. Di, X.; Gao, X.; Peng, L.; Ai, J.; Jin, X.; Qi, S.; Li, H.; Wang, K.; Luo, D. Cellular Mechanotransduction in Health and Diseases: From Molecular Mechanism to Therapeutic Targets. Signal Transduct. Target. Ther. 2023, 8, 282. [Google Scholar] [CrossRef]
  3. Boraldi, F.; Lofaro, F.D.; Bonacorsi, S.; Mazzilli, A.; Garcia-Fernandez, M.; Quaglino, D. The Role of Fibroblasts in Skin Homeostasis and Repair. Biomedicines 2024, 12, 1586. [Google Scholar] [CrossRef]
  4. Chaudhry, G.-S.; Akim, A.; Naveed Zafar, M.; Safdar, N.; Sung, Y.Y.; Muhammad, T.S.T. Understanding Hyaluronan Receptor (CD44) Interaction, HA-CD44 Activated Potential Targets in Cancer Therapeutics. Adv. Pharm. Bull. 2021, 11, 426–438. [Google Scholar] [CrossRef] [PubMed]
  5. Thorne, R.F.; Legg, J.W.; Isacke, C.M. The Role of the CD44 Transmembrane and Cytoplasmic Domains in Co-Ordinating Adhesive and Signalling Events. J. Cell Sci. 2004, 117, 373–380. [Google Scholar] [CrossRef] [PubMed]
  6. Chen, C.; Zhao, S.; Karnad, A.; Freeman, J.W. The Biology and Role of CD44 in Cancer Progression: Therapeutic Implications. J. Hematol. Oncol. 2018, 11, 64. [Google Scholar] [CrossRef] [PubMed]
  7. Martegani, M.P.; Del Prete, F.; Gasbarri, A.; Natali, P.G.; Bartolazzi, A. Structural Variability of CD44v Molecules and Reliability of Immunodetection of CD44 Isoforms Using mAbs Specific for CD44 Variant Exon Products. Am. J. Pathol. 1999, 154, 291–300. [Google Scholar] [CrossRef][Green Version]
  8. Maltseva, D.; Tonevitsky, A. RNA-Binding Proteins Regulating the CD44 Alternative Splicing. Front. Mol. Biosci. 2023, 10, 1326148. [Google Scholar] [CrossRef]
  9. Gaiteiro, C.; Soares, J.; Relvas-Santos, M.; Peixoto, A.; Ferreira, D.; Brandão, A.; Fernandes, E.; Azevedo, R.; Paulo, P.; Palmeira, C.; et al. Glycoproteogenomics Characterizes the CD44 Splicing Code Driving Bladder Cancer Invasion. Theranostics 2021, 12, 3150–3177. [Google Scholar] [CrossRef]
  10. Bajorath, J. Molecular Organization, Structural Features, and Ligand Binding Characteristics of CD44, a Highly Variable Cell Surface Glycoprotein with Multiple Functions. Proteins 2000, 39, 103–111. [Google Scholar] [CrossRef]
  11. Tsuneki, M.; Madri, J.A. CD44 Influences Fibroblast Behaviors Via Modulation of Cell-Cell and Cell-Matrix Interactions, Affecting Survivin and Hippo Pathways. J. Cell. Physiol. 2016, 231, 731–743. [Google Scholar] [CrossRef]
  12. Jordan, A.R.; Racine, R.R.; Hennig, M.J.P.; Lokeshwar, V.B. The Role of CD44 in Disease Pathophysiology and Targeted Treatment. Front. Immunol. 2015, 6, 182. [Google Scholar] [CrossRef]
  13. Xu, H.; Niu, M.; Yuan, X.; Wu, K.; Liu, A. CD44 as a Tumor Biomarker and Therapeutic Target. Exp. Hematol. Oncol. 2020, 9, 36. [Google Scholar] [CrossRef]
  14. Herishanu, Y.; Gibellini, F.; Njuguna, N.; Hazan-Halevy, I.; Keyvanfar, K.; Lee, E.; Wilson, W.; Wiestner, A. CD44 Signaling via PI3K/AKT and MAPK/ERK Pathways Protects CLL Cells from Spontaneous and Drug Induced Apoptosis through MCL-1. Leuk. Lymphoma 2011, 52, 1758–1769. [Google Scholar] [CrossRef]
  15. Herishanu, Y.; Gibellini, F.; Njuguna, N.; Keyvanfar, K.; Wiestner, A. CD44 Signaling Via PI3K/AKT and MAPK/ERK Pathways Protects CLL Cells from Spontaneous and Drug Induced Apoptosis. Blood 2008, 112, 541. [Google Scholar] [CrossRef]
  16. Robbins, E.W.; Travanty, E.A.; Yang, K.; Iczkowski, K.A. MAP Kinase Pathways and Calcitonin Influence CD44 Alternate Isoform Expression in Prostate Cancer Cells. BMC Cancer 2008, 8, 260. [Google Scholar] [CrossRef]
  17. Bai, R.-J.; Liu, D.; Li, Y.-S.; Tian, J.; Yu, D.-J.; Li, H.-Z.; Zhang, F.-J. OPN Inhibits Autophagy through CD44, Integrin and the MAPK Pathway in Osteoarthritic Chondrocytes. Front. Endocrinol. 2022, 13, 919366. [Google Scholar] [CrossRef] [PubMed]
  18. Ohata, H.; Ishiguro, T.; Aihara, Y.; Sato, A.; Sakai, H.; Sekine, S.; Taniguchi, H.; Akasu, T.; Fujita, S.; Nakagama, H.; et al. Induction of the Stem-like Cell Regulator CD44 by Rho Kinase Inhibition Contributes to the Maintenance of Colon Cancer-Initiating Cells. Cancer Res. 2012, 72, 5101–5110. [Google Scholar] [CrossRef] [PubMed]
  19. Zhang, Y.; Xia, H.; Ge, X.; Chen, Q.; Yuan, D.; Chen, Q.; Leng, W.; Chen, L.; Tang, Q.; Bi, F. CD44 Acts through RhoA to Regulate YAP Signaling. Cell. Signal. 2014, 26, 2504–2513. [Google Scholar] [CrossRef] [PubMed]
  20. Huo, X.; Dai, Y.; Yang, T.; Zhang, Y.; Li, M.; Xu, X. Decreased Erythrocyte CD44 and CD58 Expression Link E-Waste Pb Toxicity to Changes in Erythrocyte Immunity in Preschool Children. Sci. Total Environ. 2019, 664, 690–697. [Google Scholar] [CrossRef]
  21. Avivar-Valderas, A. Inhibition of PI3Kβ and mTOR influence the immune response and the defense mechanism against pathogens. Int. J. Infect. 2023, 7, 46–49. [Google Scholar]
  22. Toniato, E. IL-37 is an inhibitory cytokine that could be useful for treating infections. Int. J. Infect. 2024, 8, 1–2. [Google Scholar]
  23. Chou, Y.-E.; Hsieh, M.-J.; Hsin, C.-H.; Chiang, W.-L.; Lai, Y.-C.; Lee, Y.-H.; Huang, S.-C.; Yang, S.-F.; Lin, C.-W. CD44 Gene Polymorphisms and Environmental Factors on Oral Cancer Susceptibility in Taiwan. PLoS ONE 2014, 9, e93692. [Google Scholar] [CrossRef]
  24. Cirillo, N. The Hyaluronan/CD44 Axis: A Double-Edged Sword in Cancer. Int. J. Mol. Sci. 2023, 24, 15812. [Google Scholar] [CrossRef]
  25. UniProt. Available online: https://www.uniprot.org/uniprotkb/P16070/entry (accessed on 10 July 2025).
  26. Xu, Q. The Indian Blood Group System. Immunohematology 2011, 27, 89–93. [Google Scholar] [CrossRef]
  27. Joshi, S.R.; Sheladiya, A.; Mendapara-Dobariya, K.V. INRA, a New High-Frequency Antigen in the INDIAN (IN023) Blood Group System. Asian J. Transfus. Sci. 2017, 11, 121–123. [Google Scholar] [CrossRef] [PubMed]
  28. Poole, J.; Tilley, L.; Warke, N.; Spring, F.A.; Overbeeke, M.A.M.; van der Mark-Zoet, J.A.C.M.; Ahrens, N.; Armstrong, D.; Williams, M.; Daniels, G. Two Missense Mutations in the CD44 Gene Encode Two New Antigens of the Indian Blood Group System. Transfusion 2007, 47, 1306–1311. [Google Scholar] [CrossRef] [PubMed]
  29. Telen, M.J.; Udani, M.; Washington, M.K.; Levesque, M.C.; Lloyd, E.; Rao, N. A Blood Group-Related Polymorphism of CD44 Abolishes a Hyaluronan-Binding Consensus Sequence without Preventing Hyaluronan Binding. J. Biol. Chem. 1996, 271, 7147–7153. [Google Scholar] [CrossRef] [PubMed]
  30. Gerhard, D.S.; Wagner, L.; Feingold, E.A.; Shenmen, C.M.; Grouse, L.H.; Schuler, G.; Klein, S.L.; Old, S.; Rasooly, R.; Good, P.; et al. The Status, Quality, and Expansion of the NIH Full-Length cDNA Project: The Mammalian Gene Collection (MGC). Genome Res. 2004, 14, 2121–2127. [Google Scholar] [CrossRef]
  31. Günthert, U. CD44: A Multitude of Isoforms with Diverse Functions. In Adhesion in Leukocyte Homing and Differentiation; Dunon, D., Mackay, C.R., Imhof, B.A., Eds.; Springer: Berlin/Heidelberg, Germany, 1993; pp. 47–63. ISBN 978-3-642-78253-4. [Google Scholar]
  32. Senbanjo, L.T.; Chellaiah, M.A. CD44: A Multifunctional Cell Surface Adhesion Receptor Is a Regulator of Progression and Metastasis of Cancer Cells. Front. Cell Dev. Biol. 2017, 5, 18. [Google Scholar] [CrossRef]
  33. Misra, S.; Hascall, V.C.; Markwald, R.R.; Ghatak, S. Interactions between Hyaluronan and Its Receptors (CD44, RHAMM) Regulate the Activities of Inflammation and Cancer. Front. Immunol. 2015, 6, 201. [Google Scholar] [CrossRef]
  34. Klement, J.D.; Paschall, A.V.; Redd, P.S.; Ibrahim, M.L.; Lu, C.; Yang, D.; Celis, E.; Abrams, S.I.; Ozato, K.; Liu, K. An Osteopontin/CD44 Immune Checkpoint Controls CD8+ T Cell Activation and Tumor Immune Evasion. J. Clin. Investig. 2018, 128, 5549–5560. [Google Scholar] [CrossRef]
  35. Shirasaki, T.; Honda, M.; Yamashita, T.; Nio, K.; Shimakami, T.; Shimizu, R.; Nakasyo, S.; Murai, K.; Shirasaki, N.; Okada, H.; et al. The Osteopontin-CD44 Axis in Hepatic Cancer Stem Cells Regulates IFN Signaling and HCV Replication. Sci. Rep. 2018, 8, 13143. [Google Scholar] [CrossRef]
  36. Ou, J.; Deng, J.; Wei, X.; Xie, G.; Zhou, R.; Yu, L.; Liang, H. Fibronectin Extra Domain A (EDA) Sustains CD133(+)/CD44(+) Subpopulation of Colorectal Cancer Cells. Stem Cell Res. 2013, 11, 820–833. [Google Scholar] [CrossRef]
  37. Ozer, E.; Canda, T.; Kurtodlu, B. The Role of Angiogenesis, Laminin and CD44 Expression in Metastatic Behavior of Early-Stage Low-Grade Invasive Breast Carcinomas. Cancer Lett. 1997, 121, 119–123. [Google Scholar] [CrossRef]
  38. Song, T.; Yang, Y.; Wang, Y.; Ni, Y.; Yang, Y.; Zhang, L. Bulk and Single-Cell RNA Sequencing Reveal the Contribution of Laminin Γ2 -CD44 to the Immune Resistance in Lymphocyte-Infiltrated Squamous Lung Cancer Subtype. Heliyon 2024, 10, e31299. [Google Scholar] [CrossRef] [PubMed]
  39. Rousselle, P.; Beck, K. Laminins and Matrix Metalloproteinases Connection: A Subtle Relationship That Can Go Wrong in a Tumor Context, Particularly If CD44 Gets Involved. In The Extracellular Matrix and the Tumor Microenvironment; Kovalszky, I., Franchi, M., Alaniz, L.D., Eds.; Springer International Publishing: Cham, Switzerland, 2022; pp. 219–246. ISBN 978-3-030-99708-3. [Google Scholar]
  40. Govindaraju, P.; Todd, L.; Shetye, S.; Monslow, J.; Puré, E. CD44-Dependent Inflammation, Fibrogenesis, and Collagenolysis Regulates Extracellular Matrix Remodeling and Tensile Strength during Cutaneous Wound Healing. Matrix Biol. 2019, 75–76, 314–330. [Google Scholar] [CrossRef] [PubMed]
  41. Onyszczuk, M.; Rynkiewicz, M.; Kiczmer, P.; Drozdzowska, B. Prognostic and Clinicopathological Significance of CD44, MMP-2, and MMP-9 Expression in Clear Cell Renal Cell Carcinoma. Med. Res. J. 2025, 10, 9–18. [Google Scholar] [CrossRef]
  42. Chrabańska, M.; Rynkiewicz, M.; Kiczmer, P.; Drozdzowska, B. Immunohistochemical Expression of CD44, MMP-2, MMP-9, and Ki-67 as the Prognostic Markers in Non-Clear Cell Renal Cell Carcinomas-A Prospective Cohort Study. J. Clin. Med. 2022, 11, 5196. [Google Scholar] [CrossRef]
  43. Baaten, B.J.; Li, C.-R.; Bradley, L.M. Multifaceted Regulation of T Cells by CD44. Commun. Integr. Biol. 2010, 3, 508–512. [Google Scholar] [CrossRef]
  44. Gutjahr, J.C.; Greil, R.; Hartmann, T.N. The Role of CD44 in the Pathophysiology of Chronic Lymphocytic Leukemia. Front. Immunol. 2015, 6, 177. [Google Scholar] [CrossRef]
  45. Martin, T.A.; Harrison, G.; Mansel, R.E.; Jiang, W.G. The Role of the CD44/Ezrin Complex in Cancer Metastasis. Crit. Rev. Oncol. Hematol. 2003, 46, 165–186. [Google Scholar] [CrossRef]
  46. AbuSamra, D.B.; Al-Kilani, A.; Hamdan, S.M.; Sakashita, K.; Gadhoum, S.Z.; Merzaban, J.S. Quantitative Characterization of E-Selectin Interaction with Native CD44 and P-Selectin Glycoprotein Ligand-1 (PSGL-1) Using a Real Time Immunoprecipitation-Based Binding Assay. J. Biol. Chem. 2015, 290, 21213–21230. [Google Scholar] [CrossRef] [PubMed]
  47. Sackstein, R. The Bone Marrow Is Akin to Skin: HCELL and the Biology of Hematopoietic Stem Cell Homing. J. Investig. Dermatol. 2004, 122, 1061–1069. [Google Scholar] [CrossRef] [PubMed]
  48. Sackstein, R. The Biology of CD44 and HCELL in Hematopoiesis: The “Step 2-Bypass Pathway” and Other Emerging Perspectives. Curr. Opin. Hematol. 2011, 18, 239–248. [Google Scholar] [CrossRef] [PubMed]
  49. Jacobs, P.P.; Sackstein, R. CD44 and HCELL: Preventing Hematogenous Metastasis at Step 1. FEBS Lett. 2011, 585, 3148–3158. [Google Scholar] [CrossRef]
  50. Cao, H.; Heazlewood, S.Y.; Williams, B.; Cardozo, D.; Nigro, J.; Oteiza, A.; Nilsson, S.K. The Role of CD44 in Fetal and Adult Hematopoietic Stem Cell Regulation. Haematologica 2016, 101, 26–37. [Google Scholar] [CrossRef]
  51. Zöller, M. CD44, Hyaluronan, the Hematopoietic Stem Cell, and Leukemia-Initiating Cells. Front. Immunol. 2015, 6, 235. [Google Scholar] [CrossRef]
  52. Morath, I.; Hartmann, T.N.; Orian-Rousseau, V. CD44: More than a Mere Stem Cell Marker. Int. J. Biochem. Cell Biol. 2016, 81, 166–173. [Google Scholar] [CrossRef]
  53. Lee-Sayer, S.S.M.; Dougan, M.N.; Cooper, J.; Sanderson, L.; Dosanjh, M.; Maxwell, C.A.; Johnson, P. CD44-Mediated Hyaluronan Binding Marks Proliferating Hematopoietic Progenitor Cells and Promotes Bone Marrow Engraftment. PLoS ONE 2018, 13, e0196011. [Google Scholar] [CrossRef]
  54. Pokharel, D.; Padula, M.P.; Lu, J.F.; Jaiswal, R.; Djordjevic, S.P.; Bebawy, M. The Role of CD44 and ERM Proteins in Expression and Functionality of P-Glycoprotein in Breast Cancer Cells. Molecules 2016, 21, 290. [Google Scholar] [CrossRef]
  55. Neisch, A.L.; Fehon, R.G. Ezrin, Radixin and Moesin: Key Regulators of Membrane-Cortex Interactions and Signaling. Curr. Opin. Cell Biol. 2011, 23, 377–382. [Google Scholar] [CrossRef]
  56. Ilangumaran, S.; Briol, A.; Hoessli, D.C. CD44 Selectively Associates with Active Src Family Protein Tyrosine Kinases Lck and Fyn in Glycosphingolipid-Rich Plasma Membrane Domains of Human Peripheral Blood Lymphocytes. Blood 1998, 91, 3901–3908. [Google Scholar] [CrossRef]
  57. Tsai, T.; Wu, S.; Lai, Y.; Wang, H.; Hou, P.; Huang, Y.; Chen, H.H.; Su, W. CD44-Hyaluronan Mediating Endocytosis of Iron-Platinum Alloy Nanoparticles Induces Ferroptotic Cell Death in Mesenchymal-State Lung Cancer Cells with Tyrosine Kinase Inhibitor Resistance. Acta Biomater. 2024, 186, 396–410. [Google Scholar] [CrossRef]
  58. Wang, S.J.; Bourguignon, L.Y.W. Hyaluronan-CD44 Promotes Phospholipase C-Mediated Ca2+ Signaling and Cisplatin Resistance in Head and Neck Cancer. Arch. Otolaryngol. Head Neck Surg. 2006, 132, 19–24. [Google Scholar] [CrossRef]
  59. Bourguignon, L.Y.W.; Gilad, E.; Brightman, A.; Diedrich, F.; Singleton, P. Hyaluronan-CD44 Interaction with Leukemia-Associated RhoGEF and Epidermal Growth Factor Receptor Promotes Rho/Ras Co-Activation, Phospholipase C Epsilon-Ca2+ Signaling, and Cytoskeleton Modification in Head and Neck Squamous Cell Carcinoma Cells. J. Biol. Chem. 2006, 281, 14026–14040. [Google Scholar] [CrossRef]
  60. Wong, N.K.Y.; Lai, J.C.Y.; Maeshima, N.; Johnson, P. CD44-Mediated Elongated T Cell Spreading Requires Pyk2 Activation by Src Family Kinases, Extracellular Calcium, Phospholipase C and Phosphatidylinositol-3 Kinase. Cell. Signal. 2011, 23, 812–819. [Google Scholar] [CrossRef]
  61. Chellaiah, M.A.; Biswas, R.S.; Rittling, S.R.; Denhardt, D.T.; Hruska, K.A. Rho-Dependent Rho Kinase Activation Increases CD44 Surface Expression and Bone Resorption in Osteoclasts. J. Biol. Chem. 2003, 278, 29086–29097. [Google Scholar] [CrossRef]
  62. Lee, M.N.; Song, J.H.; Oh, S.-H.; Tham, N.T.; Kim, J.-W.; Yang, J.-W.; Kim, E.-S.; Koh, J.-T. The Primary Cilium Directs Osteopontin-Induced Migration of Mesenchymal Stem Cells by Regulating CD44 Signaling and Cdc42 Activation. Stem Cell Res. 2020, 45, 101799. [Google Scholar] [CrossRef] [PubMed]
  63. Murai, T.; Miyazaki, Y.; Nishinakamura, H.; Sugahara, K.N.; Miyauchi, T.; Sako, Y.; Yanagida, T.; Miyasaka, M. Engagement of CD44 Promotes Rac Activation and CD44 Cleavage during Tumor Cell Migration. J. Biol. Chem. 2004, 279, 4541–4550. [Google Scholar] [CrossRef] [PubMed]
  64. Li, Y.; Wang, D.; Ge, H.; Güngör, C.; Gong, X.; Chen, Y. Cytoskeletal and Cytoskeleton-Associated Proteins: Key Regulators of Cancer Stem Cell Properties. Pharmaceuticals 2022, 15, 1369. [Google Scholar] [CrossRef] [PubMed]
  65. Pang, X.; He, X.; Qiu, Z.; Zhang, H.; Xie, R.; Liu, Z.; Gu, Y.; Zhao, N.; Xiang, Q.; Cui, Y. Targeting Integrin Pathways: Mechanisms and Advances in Therapy. Signal Transduct. Target. Ther. 2023, 8, 1. [Google Scholar] [CrossRef]
  66. Trapasso, S.; Allegra, E. Role of CD44 as a Marker of Cancer Stem Cells in Head and Neck Cancer. Biologics 2012, 6, 379–383. [Google Scholar] [CrossRef][Green Version]
  67. Poltavets, V.; Kochetkova, M.; Pitson, S.M.; Samuel, M.S. The Role of the Extracellular Matrix and Its Molecular and Cellular Regulators in Cancer Cell Plasticity. Front. Oncol. 2018, 8, 431. [Google Scholar] [CrossRef]
  68. Nonnast, E.; Mira, E.; Mañes, S. The Role of Laminins in Cancer Pathobiology: A Comprehensive Review. J. Transl. Med. 2025, 23, 83. [Google Scholar] [CrossRef]
  69. Bourguignon, L.Y.W.; Singleton, P.A.; Diedrich, F. Hyaluronan-CD44 Interaction with Rac1-Dependent Protein Kinase N-γ Promotes Phospholipase Cγ1 Activation, Ca2+ Signaling, and Cortactin-Cytoskeleton Function Leading to Keratinocyte Adhesion and Differentiation. J. Biol. Chem. 2004, 279, 29654–29669. [Google Scholar] [CrossRef]
  70. Vikesaa, J.; Hansen, T.V.; Jønson, L.; Borup, R.; Wewer, U.M.; Christiansen, J.; Nielsen, F.C. RNA-binding IMPs Promote Cell Adhesion and Invadopodia Formation. EMBO J. 2006, 25, 1456–1468. [Google Scholar] [CrossRef]
  71. Casalino-Matsuda, S.M.; Monzon, M.E.; Day, A.J.; Forteza, R.M. Hyaluronan Fragments/CD44 Mediate Oxidative Stress–Induced MUC5B Up-Regulation in Airway Epithelium. Am. J. Respir. Cell Mol. Biol. 2009, 40, 277–285. [Google Scholar] [CrossRef] [PubMed]
  72. Crosby, H.A.; Lalor, P.F.; Ross, E.; Newsome, P.N.; Adams, D.H. Adhesion of Human Haematopoietic (CD34+) Stem Cells to Human Liver Compartments Is Integrin and CD44 Dependent and Modulated by CXCR3 and CXCR4. J. Hepatol. 2009, 51, 734–749. [Google Scholar] [CrossRef] [PubMed]
  73. Yoshida, T.; Matsuda, Y.; Naito, Z.; Ishiwata, T. CD44 in Human Glioma Correlates with Histopathological Grade and Cell Migration. Pathol. Int. 2012, 62, 463–470. [Google Scholar] [CrossRef] [PubMed]
  74. Midgley, A.C.; Rogers, M.; Hallett, M.B.; Clayton, A.; Bowen, T.; Phillips, A.O.; Steadman, R. Transforming Growth Factor-Β1 (TGF-Β1)-Stimulated Fibroblast to Myofibroblast Differentiation Is Mediated by Hyaluronan (HA)-Facilitated Epidermal Growth Factor Receptor (EGFR) and CD44 Co-Localization in Lipid Rafts. J. Biol. Chem. 2013, 288, 14824–14838. [Google Scholar] [CrossRef]
  75. Funaro, A.; Spagnoli, G.C.; Momo, M.; Knapp, W.; Malavasi, F. Stimulation of T Cells via CD44 Requires Leukocyte-Function-Associated Antigen Interactions and Interleukin-2 Production. Hum. Immunol. 1994, 40, 267–278. [Google Scholar] [CrossRef]
  76. Buscher, K.; Riese, S.B.; Shakibaei, M.; Reich, C.; Dernedde, J.; Tauber, R.; Ley, K. The Transmembrane Domains of L-Selectin and CD44 Regulate Receptor Cell Surface Positioning and Leukocyte Adhesion under Flow. J. Biol. Chem. 2010, 285, 13490–13497. [Google Scholar] [CrossRef] [PubMed]
  77. Qiu, Y.; Wang, H.; Guo, Q.; Liu, Y.; He, Y.; Zhang, G.; Yang, C.; Du, Y.; Gao, F. CD44s-Activated tPA/LRP1-NFκB Pathway Drives Lamellipodia Outgrowth in Luminal-Type Breast Cancer Cells. Front. Cell Dev. Biol. 2023, 11, 1224827. [Google Scholar] [CrossRef] [PubMed]
  78. Föger, N.; Marhaba, R.; Zöller, M. Involvement of CD44 in Cytoskeleton Rearrangement and Raft Reorganization in T Cells. J. Cell Sci. 2001, 114, 1169–1178. [Google Scholar] [CrossRef] [PubMed]
  79. Skandalis, S.S. CD44 Intracellular Domain: A Long Tale of a Short Tail. Cancers 2023, 15, 5041. [Google Scholar] [CrossRef]
  80. Du, Y.; Bradshaw, W.J.; Leisner, T.M.; Annor-Gyamfi, J.K.; Qian, K.; Bashore, F.M.; Sikdar, A.; Nwogbo, F.O.; Ivanov, A.A.; Frye, S.V.; et al. Discovery of FERM Domain Protein–Protein Interaction Inhibitors for MSN and CD44 as a Potential Therapeutic Approach for Alzheimer’s Disease. J. Biol. Chem. 2023, 299, 105382. [Google Scholar] [CrossRef]
  81. Weng, X.; Maxwell-Warburton, S.; Hasib, A.; Ma, L.; Kang, L. The Membrane Receptor CD44: Novel Insights into Metabolism. Trends Endocrinol. Metab. 2022, 33, 318–332. [Google Scholar] [CrossRef]
  82. Tsukita, S.; Oishi, K.; Sato, N.; Sagara, J.; Kawai, A.; Tsukita, S. ERM Family Members as Molecular Linkers between the Cell Surface Glycoprotein CD44 and Actin-Based Cytoskeletons. J. Cell Biol. 1994, 126, 391–401. [Google Scholar] [CrossRef]
  83. Mori, T.; Kitano, K.; Terawaki, S.; Maesaki, R.; Fukami, Y.; Hakoshima, T. Structural Basis for CD44 Recognition by ERM Proteins. J. Biol. Chem. 2008, 283, 29602–29612. [Google Scholar] [CrossRef]
  84. Yonemura, S.; Hirao, M.; Doi, Y.; Takahashi, N.; Kondo, T.; Tsukita, S.; Tsukita, S. Ezrin/Radixin/Moesin (ERM) Proteins Bind to a Positively Charged Amino Acid Cluster in the Juxta-Membrane Cytoplasmic Domain of CD44, CD43, and ICAM-2. J. Cell Biol. 1998, 140, 885–895. [Google Scholar] [CrossRef]
  85. Tissue Expression of CD44—Summary—The Human Protein Atlas. Available online: https://www.proteinatlas.org/ENSG00000026508-CD44/tissue (accessed on 18 July 2025).
  86. Ma, X.; Dighe, A.; Maziarz, J.; Neumann, E.; Erkenbrack, E.; Hei, Y.-Y.; Liu, Y.; Suhail, Y.; Pak, I.; Levchenko, A.; et al. Evolution of Higher Mesenchymal CD44 Expression in the Human Lineage. Evol. Med. Public Health 2022, 10, 447–462. [Google Scholar] [CrossRef]
  87. Williams, K.; Motiani, K.; Giridhar, P.V.; Kasper, S. CD44 Integrates Signaling in Normal Stem Cell, Cancer Stem Cell and (Pre)Metastatic Niches. Exp. Biol. Med. 2013, 238, 324–338. [Google Scholar] [CrossRef]
  88. Hassn Mesrati, M.; Syafruddin, S.E.; Mohtar, M.A.; Syahir, A. CD44: A Multifunctional Mediator of Cancer Progression. Biomolecules 2021, 11, 1850. [Google Scholar] [CrossRef] [PubMed]
  89. Hu, J.; Li, G.; Zhang, P.; Zhuang, X.; Hu, G. A CD44v+ Subpopulation of Breast Cancer Stem-like Cells with Enhanced Lung Metastasis Capacity. Cell Death Dis. 2017, 8, e2679. [Google Scholar] [CrossRef] [PubMed]
  90. Li, Z.; Chen, K.; Jiang, P.; Zhang, X.; Li, X.; Li, Z. CD44v/CD44s Expression Patterns Are Associated with the Survival of Pancreatic Carcinoma Patients. Diagn. Pathol. 2014, 9, 79. [Google Scholar] [CrossRef] [PubMed]
  91. Erb, U.; Megaptche, A.P.; Gu, X.; Büchler, M.W.; Zöller, M. CD44 Standard and CD44v10 Isoform Expression on Leukemia Cells Distinctly Influences Niche Embedding of Hematopoietic Stem Cells. J. Hematol. Oncol. 2014, 7, 29. [Google Scholar] [CrossRef]
  92. Lo, C.W.-S.; Chan, C.K.W.; Yu, J.; He, M.; Choi, C.H.J.; Lau, J.Y.W.; Wong, N. Development of CD44E/s Dual-Targeting DNA Aptamer as Nanoprobe to Deliver Treatment in Hepatocellular Carcinoma. Nanotheranostics 2022, 6, 161–174. [Google Scholar] [CrossRef]
  93. Zhu, S.; Chen, Z.; Katsha, A.; Hong, J.; Belkhiri, A.; El-Rifai, W. Regulation of CD44E by DARPP-32-Dependent Activation of SRp20 Splicing Factor in Gastric Tumorigenesis. Oncogene 2016, 35, 1847–1856. [Google Scholar] [CrossRef][Green Version]
  94. Oliferenko, S.; Paiha, K.; Harder, T.; Gerke, V.; Schwärzler, C.; Schwarz, H.; Beug, H.; Günthert, U.; Huber, L.A. Analysis of Cd44-Containing Lipid Rafts. J. Cell Biol. 1999, 146, 843–854. [Google Scholar] [CrossRef]
  95. Murai, T. Lipid Raft-Mediated Regulation of Hyaluronan–CD44 Interactions in Inflammation and Cancer. Front. Immunol. 2015, 6, 420. [Google Scholar] [CrossRef]
  96. Sun, F.; Schroer, C.F.E.; Palacios, C.R.; Xu, L.; Luo, S.-Z.; Marrink, S.J. Molecular Mechanism for Bidirectional Regulation of CD44 for Lipid Raft Affiliation by Palmitoylations and PIP2. PLoS Comput. Biol. 2020, 16, e1007777. [Google Scholar] [CrossRef] [PubMed]
  97. Li, D.; Park, Y.; Hemati, H.; Liu, X. Cell Aggregation Activates Small GTPase Rac1 and Induces CD44 Cleavage by Maintaining Lipid Raft Integrity. J. Biol. Chem. 2023, 299, 105377. [Google Scholar] [CrossRef] [PubMed]
  98. Prochazka, L.; Tesarik, R.; Turanek, J. Regulation of Alternative Splicing of CD44 in Cancer. Cell. Signal. 2014, 26, 2234–2239. [Google Scholar] [CrossRef] [PubMed]
  99. Chiu, R.K.; Carpenito, C.; Dougherty, S.T.; Hayes, G.M.; Dougherty, G.J. Identification and Characterization of CD44RC, a Novel Alternatively Spliced Soluble CD44 Isoform That Can Potentiate the Hyaluronan Binding Activity of Cell Surface CD44. Neoplasia 1999, 1, 446–452. [Google Scholar] [CrossRef][Green Version]
  100. Bechtel, S.; Rosenfelder, H.; Duda, A.; Schmidt, C.P.; Ernst, U.; Wellenreuther, R.; Mehrle, A.; Schuster, C.; Bahr, A.; Blöcker, H.; et al. The Full-ORF Clone Resource of the German cDNA Consortium. BMC Genom. 2007, 8, 399. [Google Scholar] [CrossRef]
  101. Harn, H.J.; Isola, N.; Cooper, D.L. The Multispecific Cell Adhesion Molecule CD44 Is Represented in Reticulocyte cDNA. Biochem. Biophys. Res. Commun. 1991, 178, 1127–1134. [Google Scholar] [CrossRef]
  102. Stamenkovic, I.; Amiot, M.; Pesando, J.M.; Seed, B. A Lymphocyte Molecule Implicated in Lymph Node Homing Is a Member of the Cartilage Link Protein Family. Cell 1989, 56, 1057–1062. [Google Scholar] [CrossRef]
  103. Goldstein, L.A.; Zhou, D.F.; Picker, L.J.; Minty, C.N.; Bargatze, R.F.; Ding, J.F.; Butcher, E.C. A Human Lymphocyte Homing Receptor, the Hermes Antigen, Is Related to Cartilage Proteoglycan Core and Link Proteins. Cell 1989, 56, 1063–1072. [Google Scholar] [CrossRef]
  104. Kozlowski, L.P. IPC—Isoelectric Point Calculator. Biol. Direct 2016, 11, 55. [Google Scholar] [CrossRef]
  105. Baj-Krzyworzeka, M.; Weglarczyk, K.; Szatanek, R.; Mytar, B.; Baran, J.; Siedlar, M. The Role of CD44H Molecule in the Interactions between Human Monocytes and Pancreatic Adenocarcinoma-Derived Microvesicles. Folia Histochem. Cytobiol. 2019, 57, 28–34. [Google Scholar] [CrossRef]
  106. Stamenkovic, I.; Yu, Q. CHAPTER 5—CD44 Meets Merlin and Ezrin: Their Interplay Mediates the Pro-Tumor Activity of CD44 and Tumor-Suppressing Effect of Merlin. In Hyaluronan in Cancer Biology; Stern, R., Ed.; Academic Press: San Diego, CA, USA, 2009; pp. 71–87. ISBN 978-0-12-374178-3. [Google Scholar]
  107. Bonente, D.; Bianchi, L.; De Salvo, R.; Nicoletti, C.; De Benedetto, E.; Bacci, T.; Bini, L.; Inzalaco, G.; Franci, L.; Chiariello, M.; et al. Co-Expression of Podoplanin and CD44 in Proliferative Vitreoretinopathy Epiretinal Membranes. Int. J. Mol. Sci. 2023, 24, 9728. [Google Scholar] [CrossRef]
  108. Perez, A.; Neskey, D.M.; Wen, J.; Pereira, L.; Reategui, E.P.; Goodwin, W.J.; Carraway, K.L.; Franzmann, E.J. CD44 Interacts with EGFR and Promotes Head and Neck Squamous Cell Carcinoma Initiation and Progression. Oral Oncol. 2013, 49, 306–313. [Google Scholar] [CrossRef]
  109. Liu, Z.; Chu, S.; Yao, S.; Li, Y.; Fan, S.; Sun, X.; Su, L.; Liu, X. CD74 Interacts with CD44 and Enhances Tumorigenesis and Metastasis via RHOA-Mediated Cofilin Phosphorylation in Human Breast Cancer Cells. Oncotarget 2016, 7, 68303–68313. [Google Scholar] [CrossRef]
  110. Orian-Rousseau, V.; Sleeman, J. Chapter Nine—CD44 Is a Multidomain Signaling Platform That Integrates Extracellular Matrix Cues with Growth Factor and Cytokine Signals. In Advances in Cancer Research; Simpson, M.A., Heldin, P., Eds.; Hyaluronan Signaling and Turnover; Academic Press: San Diego, CA, USA, 2014; Volume 123, pp. 231–254. [Google Scholar]
  111. Cowman, M.K.; Turley, E.A. Functional Organization of Extracellular Hyaluronan, CD44, and RHAMM. Proteoglycan Res. 2023, 1, e4. [Google Scholar] [CrossRef]
  112. Lee, J.-L.; Wang, M.-J.; Sudhir, P.-R.; Chen, J.-Y. CD44 Engagement Promotes Matrix-Derived Survival through the CD44-SRC-Integrin Axis in Lipid Rafts. Mol. Cell. Biol. 2008, 28, 5710–5723. [Google Scholar] [CrossRef]
  113. Skandalis, S.S.; Kozlova, I.; Engström, U.; Hellman, U.; Heldin, P. Proteomic Identification of CD44 Interacting Proteins. IUBMB Life 2010, 62, 833–840. [Google Scholar] [CrossRef]
  114. Bai, Y.; Liu, Y.-J.; Wang, H.; Xu, Y.; Stamenkovic, I.; Yu, Q. Inhibition of the Hyaluronan-CD44 Interaction by Merlin Contributes to the Tumor-Suppressor Activity of Merlin. Oncogene 2007, 26, 836–850. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, Y.-N.; Tsai, M.-F.; Wu, S.-G.; Chang, T.-H.; Shih, J.-Y. CD44s and CD44v8-10 Isoforms Confer Acquired Resistance to Osimertinib by Activating the ErbB3/STAT3 Signaling Pathway. Life Sci. 2024, 336, 122345. [Google Scholar] [CrossRef] [PubMed]
  116. Shi, X.; Leng, L.; Wang, T.; Wang, W.; Du, X.; Li, J.; McDonald, C.; Chen, Z.; Murphy, J.W.; Lolis, E.; et al. CD44 Is the Signaling Component of the Macrophage Migration Inhibitory Factor-CD74 Receptor Complex. Immunity 2006, 25, 595–606. [Google Scholar] [CrossRef] [PubMed]
  117. Guo, Q.; Yang, C.; Gao, F. The State of CD44 Activation in Cancer Progression and Therapeutic Targeting. FEBS J. 2022, 289, 7970–7986. [Google Scholar] [CrossRef]
  118. Cichy, J.; Puré, E. The Liberation of CD44. J. Cell Biol. 2003, 161, 839–843. [Google Scholar] [CrossRef]
  119. Bruno, P.S.; Arshad, A.; Gogu, M.-R.; Waterman, N.; Flack, R.; Dunn, K.; Darie, C.C.; Neagu, A.-N. Post-Translational Modifications of Proteins Orchestrate All Hallmarks of Cancer. Life 2025, 15, 126. [Google Scholar] [CrossRef]
  120. Liao, C.; Wang, Q.; An, J.; Chen, J.; Li, X.; Long, Q.; Xiao, L.; Guan, X.; Liu, J. CD44 Glycosylation as a Therapeutic Target in Oncology. Front. Oncol. 2022, 12, 883831. [Google Scholar] [CrossRef] [PubMed]
  121. Vuorio, J.; Škerlová, J.; Fábry, M.; Veverka, V.; Vattulainen, I.; Řezáčová, P.; Martinez-Seara, H. N-Glycosylation Can Selectively Block or Foster Different Receptor–Ligand Binding Modes. Sci. Rep. 2021, 11, 5239. [Google Scholar] [CrossRef] [PubMed]
  122. Leon, F.; Seshacharyulu, P.; Nimmakalaya, R.K.; Chugh, S.; Karmakar, S.; Nallasamy, P.; Vengoji, R.; Rachagani, S.; Cox, J.L.; Mallya, K.; et al. Reduction in O-Glycome Induces Differentially Glycosylated CD44 to Promote Stemness and Metastasis in Pancreatic Cancer. Oncogene 2022, 41, 57–71. [Google Scholar] [CrossRef] [PubMed]
  123. Nam, K.; Oh, S.; Shin, I. Ablation of CD44 Induces Glycolysis-to-Oxidative Phosphorylation Transition via Modulation of the c-Src-Akt-LKB1-AMPKα Pathway. Biochem. J. 2016, 473, 3013–3030. [Google Scholar] [CrossRef]
  124. Puré, E.; Camp, R.L.; Peritt, D.; Panettieri, R.A.; Lazaar, A.L.; Nayak, S. Defective Phosphorylation and Hyaluronate Binding of CD44 with Point Mutations in the Cytoplasmic Domain. J. Exp. Med. 1995, 181, 55–62. [Google Scholar] [CrossRef]
  125. Stamenkovic, I.; Yu, Q. Shedding Light on Proteolytic Cleavage of CD44: The Responsible Sheddase and Functional Significance of Shedding. J. Investig. Dermatol. 2009, 129, 1321–1324. [Google Scholar] [CrossRef]
  126. Wöhner, B.; Li, W.; Hey, S.; Drobny, A.; Werny, L.; Becker-Pauly, C.; Lucius, R.; Zunke, F.; Linder, S.; Arnold, P. Proteolysis of CD44 at the Cell Surface Controls a Downstream Protease Network. Front. Mol. Biosci. 2023, 10, 1026810. [Google Scholar] [CrossRef]
  127. Ravindranath, A.K.; Kaur, S.; Wernyj, R.P.; Kumaran, M.N.; Miletti-Gonzalez, K.E.; Chan, R.; Lim, E.; Madura, K.; Rodriguez-Rodriguez, L. CD44 Promotes Multi-Drug Resistance by Protecting P-Glycoprotein from FBXO21-Mediated Ubiquitination. Oncotarget 2015, 6, 26308–26321. [Google Scholar] [CrossRef]
  128. Xu, Y.; Bai, Z.; Lan, T.; Fu, C.; Cheng, P. CD44 and Its Implication in Neoplastic Diseases. MedComm 2024, 5, e554. [Google Scholar] [CrossRef]
  129. Yung, S.; Chan, T.M. The Role of Hyaluronan and CD44 in the Pathogenesis of Lupus Nephritis. Autoimmune Dis. 2012, 2012, 207190. [Google Scholar] [CrossRef]
  130. Yi, P.; Cao, P.; Yang, M.; Xiong, F.; Jiang, J.; Mei, Y.; Xin, Y.; Zhao, M.; Wu, H.; Lu, Q. Overexpressed CD44 Is Associated with B-Cell Activation via the HA-CD44-AIM2 Pathway in Lupus B Cells. Clin. Immunol. 2023, 255, 109710. [Google Scholar] [CrossRef] [PubMed]
  131. Norris, P.A.A.; Kaur, G.; Khan, R.; Zhu, G.; Ni, H.; Lazarus, A.H. Anti-Inflammatory Activity of CD44 Antibodies in Murine Immune Thrombocytopenia Is Mediated by Fcγ Receptor Inhibition. Blood 2021, 137, 2114–2124. [Google Scholar] [CrossRef] [PubMed]
  132. Tremmel, M.; Matzke, A.; Albrecht, I.; Laib, A.M.; Olaku, V.; Ballmer-Hofer, K.; Christofori, G.; Héroult, M.; Augustin, H.G.; Ponta, H.; et al. A CD44v6 Peptide Reveals a Role of CD44 in VEGFR-2 Signaling and Angiogenesis. Blood 2009, 114, 5236–5244. [Google Scholar] [CrossRef] [PubMed]
  133. Ziranu, P.; Pretta, A.; Aimola, V.; Cau, F.; Mariani, S.; D’Agata, A.P.; Codipietro, C.; Rizzo, D.; Dell’Utri, V.; Sanna, G.; et al. CD44: A New Prognostic Marker in Colorectal Cancer? Cancers 2024, 16, 1569. [Google Scholar] [CrossRef]
  134. Inoue, A.; Ohnishi, T.; Nishikawa, M.; Ohtsuka, Y.; Kusakabe, K.; Yano, H.; Tanaka, J.; Kunieda, T. A Narrative Review on CD44’s Role in Glioblastoma Invasion, Proliferation, and Tumor Recurrence. Cancers 2023, 15, 4898. [Google Scholar] [CrossRef]
  135. Mehner, L.-M.; Munoz-Sagredo, L.; Sonnentag, S.J.; Treffert, S.M.; Orian-Rousseau, V. Targeting CD44 and Other Pleiotropic Co-Receptors as a Means for Broad Inhibition of Tumor Growth and Metastasis. Clin. Exp. Metastasis 2024, 41, 599–611. [Google Scholar] [CrossRef]
  136. Johnson, P.; Ruffell, B. CD44 and Its Role in Inflammation and Inflammatory Diseases. Inflamm. Allergy Drug Targets 2009, 8, 208–220. [Google Scholar] [CrossRef]
  137. Puré, E.; Cuff, C.A. A Crucial Role for CD44 in Inflammation. Trends Mol. Med. 2001, 7, 213–221. [Google Scholar] [CrossRef]
  138. Krolikoski, M.; Monslow, J.; Puré, E. The CD44-HA Axis and Inflammation in Atherosclerosis: A Temporal Perspective. Matrix Biol. 2019, 78–79, 201–218. [Google Scholar] [CrossRef]
  139. Salathia, S.; Gigliobianco, M.R.; Casadidio, C.; Di Martino, P.; Censi, R. Hyaluronic Acid-Based Nanosystems for CD44 Mediated Anti-Inflammatory and Antinociceptive Activity. Int. J. Mol. Sci. 2023, 24, 7286. [Google Scholar] [CrossRef]
  140. Wong, C.C.Y.; Gao, L.Y.; Xu, Y.; Chau, M.K.M.; Zhang, D.; Yap, D.Y.H.; Ying, S.K.Y.; Lee, C.K.; Yung, S.; Chan, T.M. Cluster of Differentiation-44 as a Novel Biomarker of Lupus Nephritis and Its Role in Kidney Inflammation and Fibrosis. Front. Immunol. 2024, 15, 1443153. [Google Scholar] [CrossRef]
  141. Yung, S.; Gao, L.; Chan, T.M. LSO-014 Clinico-Pathological Association of Serum CD44 Level in Lupus Nephritis Patients. Lupus Sci. Med. 2023, 10. [Google Scholar] [CrossRef]
  142. Fromont Hankard, G.; Cezard, J.P.; Aigrain, Y.; Navarro, J.; Peuchmaur, M. CD44 Variant Expression in Inflammatory Colonic Mucosa Is Not Disease Specific but Associated with Increased Crypt Cell Proliferation. Histopathology 1998, 32, 317–321. [Google Scholar] [CrossRef] [PubMed]
  143. Franić, I.; Režić-Mužinić, N.; Markotić, A.; Živković, P.M.; Vilović, M.; Rušić, D.; Božić, J. Expression of CD44 in Leukocyte Subpopulations in Patients with Inflammatory Bowel Diseases. Diagnostics 2022, 12, 2014. [Google Scholar] [CrossRef] [PubMed]
  144. Li, W.; Cai, J.; Gu, Y.; Li, X.; He, W.; Chen, Y.; Wang, Z.; Li, K.; Qin, G.; Gu, X.; et al. Novel pH-Responsive and CD44-Targeting Silica Nanoparticles for Inflammatory Bowel Disease Therapy. Chem. Eng. J. 2025, 513, 163017. [Google Scholar] [CrossRef]
  145. Wittig, B.M.; Sabat, R.; Holzlöhner, P.; Witte-Händel, E.; Heilmann, K.; Witte, K.; Triebus, J.; Tzankov, A.; Laman, J.D.; Bokemeyer, B.; et al. Absence of Specific Alternatively Spliced Exon of CD44 in Macrophages Prevents Colitis. Mucosal Immunol. 2018, 11, 846–860. [Google Scholar] [CrossRef]
  146. Kitano, A.; Oshitani, N.; Matsumoto, T.; Kobayashi, K. CD44 Variants in Ulcerative Colitis and Crohn’s Disease. Lancet 1996, 348, 266–267. [Google Scholar] [CrossRef]
  147. Collins, C.B.; Ho, J.; Wilson, T.E.; Wermers, J.D.; Tlaxca, J.L.; Lawrence, M.B.; Solga, M.; Lannigan, J.; Rivera–Nieves, J. CD44 Deficiency Attenuates Chronic Murine Ileitis. Gastroenterology 2008, 135, 1993–2002. [Google Scholar] [CrossRef]
  148. Vadhan, A.; Hou, M.-F.; Vijayaraghavan, P.; Wu, Y.-C.; Hu, S.C.-S.; Wang, Y.-M.; Cheng, T.-L.; Wang, Y.-Y.; Yuan, S.-S.F. CD44 Promotes Breast Cancer Metastasis through AKT-Mediated Downregulation of Nuclear FOXA2. Biomedicines 2022, 10, 2488. [Google Scholar] [CrossRef]
  149. Louderbough, J.M.V.; Schroeder, J.A. Understanding the Dual Nature of CD44 in Breast Cancer Progression. Mol. Cancer Res. 2011, 9, 1573–1586. [Google Scholar] [CrossRef]
  150. Li, X.-P.; Zhang, X.-W.; Zheng, L.-Z.; Guo, W.-J. Expression of CD44 in Pancreatic Cancer and Its Significance. Int. J. Clin. Exp. Pathol. 2015, 8, 6724–6731. [Google Scholar] [PubMed]
  151. Zhao, S.; Chen, C.; Chang, K.; Karnad, A.; Jagirdar, J.; Kumar, A.P.; Freeman, J.W. CD44 Expression Level and Isoform Contributes to Pancreatic Cancer Cell Plasticity, Invasiveness and Response to Therapy. Clin. Cancer Res. 2016, 22, 5592–5604. [Google Scholar] [CrossRef] [PubMed]
  152. Chen, C.; Zhao, S.; Zhao, X.; Cao, L.; Karnad, A.; Kumar, A.P.; Freeman, J.W. Gemcitabine Resistance of Pancreatic Cancer Cells Is Mediated by IGF1R Dependent Upregulation of CD44 Expression and Isoform Switching. Cell Death Dis. 2022, 13, 682. [Google Scholar] [CrossRef] [PubMed]
  153. Wang, Y.-Y.; Vadhan, A.; Chen, P.-H.; Lee, Y.-L.; Chao, C.-Y.; Cheng, K.-H.; Chang, Y.-C.; Hu, S.C.-S.; Yuan, S.-S.F. CD44 Promotes Lung Cancer Cell Metastasis through ERK–ZEB1 Signaling. Cancers 2021, 13, 4057. [Google Scholar] [CrossRef]
  154. Hu, B.; Ma, Y.; Yang, Y.; Zhang, L.; Han, H.; Chen, J. CD44 Promotes Cell Proliferation in Non-Small Cell Lung Cancer. Oncol. Lett. 2018, 15, 5627–5633. [Google Scholar] [CrossRef]
  155. Alaei, E.; Farahani, N.; Orouei, S.; Alimohammadi, M.; Daneshi, S.; Mousavi, T.; Mahmoodieh, B.; Taheriazam, A.; Rahimzadeh, P.; Hashemi, M. The Clinicopathologic and Prognostic Value of CD44 Expression in Patients with Non-Small Cell Lung Cancer: A Systematic Review and Meta-Analysis. Mol. Cell. Probes 2025, 81, 102028. [Google Scholar] [CrossRef]
  156. Grisar, J.; Munk, M.; Steiner, C.W.; Amoyo-Minar, L.; Tohidast-Akrad, M.; Zenz, P.; Steiner, G.; Smolen, J.S. Expression Patterns of CD44 and CD44 Splice Variants in Patients with Rheumatoid Arthritis. Clin. Exp. Rheumatol. 2012, 30, 64–72. [Google Scholar]
  157. Naor, D.; Nedvetzki, S. CD44 in Rheumatoid Arthritis. Arthritis Res. Ther. 2003, 5, 105–115. [Google Scholar] [CrossRef]
  158. Gorantla, S.; Gorantla, G.; Saha, R.N.; Singhvi, G. CD44 Receptor-Targeted Novel Drug Delivery Strategies for Rheumatoid Arthritis Therapy. Expert Opin. Drug Deliv. 2021, 18, 1553–1557. [Google Scholar] [CrossRef]
  159. Wittig, B.; Schwärzler, C.; Föhr, N.; Günthert, U.; Zöller, M. Cutting Edge: Curative Treatment of an Experimentally Induced Colitis by a CD44 Variant V7-Specific Antibody1. J. Immunol. 1998, 161, 1069–1073. [Google Scholar] [CrossRef]
  160. Schuster, R.; Younesi, F.; Ezzo, M.; Hinz, B. The Role of Myofibroblasts in Physiological and Pathological Tissue Repair. Cold Spring Harb. Perspect. Biol. 2023, 15, a041231. [Google Scholar] [CrossRef] [PubMed]
  161. Cialdai, F.; Risaliti, C.; Monici, M. Role of Fibroblasts in Wound Healing and Tissue Remodeling on Earth and in Space. Front. Bioeng. Biotechnol. 2022, 10, 958381. [Google Scholar] [CrossRef] [PubMed]
  162. Plikus, M.V.; Wang, X.; Sinha, S.; Forte, E.; Thompson, S.M.; Herzog, E.L.; Driskell, R.R.; Rosenthal, N.; Biernaskie, J.; Horsley, V. Fibroblasts: Origins, Definitions, and Functions in Health and Disease. Cell 2021, 184, 3852–3872. [Google Scholar] [CrossRef] [PubMed]
  163. Guler, Z.; Roovers, J.P. Role of Fibroblasts and Myofibroblasts on the Pathogenesis and Treatment of Pelvic Organ Prolapse. Biomolecules 2022, 12, 94. [Google Scholar] [CrossRef]
  164. Ito, T.; Williams, J.D.; Fraser, D.J.; Phillips, A.O. Hyaluronan Regulates Transforming Growth Factor-Beta1 Receptor Compartmentalization. J. Biol. Chem. 2004, 279, 25326–25332. [Google Scholar] [CrossRef]
  165. Bourguignon, L.Y.W.; Singleton, P.A.; Zhu, H.; Zhou, B. Hyaluronan Promotes Signaling Interaction between CD44 and the Transforming Growth Factor Beta Receptor I in Metastatic Breast Tumor Cells. J. Biol. Chem. 2002, 277, 39703–39712. [Google Scholar] [CrossRef]
  166. Li, L.; Qi, L.; Liang, Z.; Song, W.; Liu, Y.; Wang, Y.; Sun, B.; Zhang, B.; Cao, W. Transforming Growth Factor-Β1 Induces EMT by the Transactivation of Epidermal Growth Factor Signaling through HA/CD44 in Lung and Breast Cancer Cells. Int. J. Mol. Med. 2015, 36, 113–122. [Google Scholar] [CrossRef]
  167. Wang, Y.; Mack, J.A.; Maytin, E.V. CD44 Inhibits α-SMA Gene Expression via a Novel G-Actin/MRTF-Mediated Pathway That Intersects with TGFβR/p38MAPK Signaling in Murine Skin Fibroblasts. J. Biol. Chem. 2019, 294, 12779–12794. [Google Scholar] [CrossRef]
  168. Wang, Z.; Zhao, K.; Hackert, T.; Zöller, M. CD44/CD44v6 a Reliable Companion in Cancer-Initiating Cell Maintenance and Tumor Progression. Front. Cell Dev. Biol. 2018, 6, 97. [Google Scholar] [CrossRef] [PubMed]
  169. Sasaki, N.; Itakura, Y.; Toyoda, M. Sialylation Regulates Myofibroblast Differentiation of Human Skin Fibroblasts. Stem Cell Res. Ther. 2017, 8, 81. [Google Scholar] [CrossRef] [PubMed]
  170. Morris, N.G.; Woods, E.L.; Dally, J.; Midgley, A.C.; Steadman, R.; Moseley, R. Dysfunctional Pericellular Hyaluronan Deposition Contributes to Attenuated CD44/EGFR Co-Localization and Impaired Myofibroblast Differentiation in Chronic Wound Fibroblasts. Exp. Cell Res. 2025, 450, 114646. [Google Scholar] [CrossRef] [PubMed]
  171. Chan, Y.-H.; Tsai, F.-C.; Chang, G.-J.; Lai, Y.-J.; Chang, S.-H.; Chen, W.-J.; Yeh, Y.-H. CD44 Regulates Epac1-Mediated β-Adrenergic-Receptor-Induced Ca2+-Handling Abnormalities: Implication in Cardiac Arrhythmias. J. Biomed. Sci. 2023, 30, 55. [Google Scholar] [CrossRef]
  172. Mishra, J.P.; Mishra, S.; Gee, K.; Kumar, A. Differential Involvement of Calmodulin-Dependent Protein Kinase II-Activated AP-1 and c-Jun N-Terminal Kinase-Activated EGR-1 Signaling Pathways in Tumor Necrosis Factor-α and Lipopolysaccharide-Induced CD44 Expression in Human Monocytic Cells. J. Biol. Chem. 2005, 280, 26825–26837. [Google Scholar] [CrossRef]
  173. Udabage, L.; Brownlee, G.R.; Nilsson, S.K.; Brown, T.J. The Over-Expression of HAS2, Hyal-2 and CD44 Is Implicated in the Invasiveness of Breast Cancer. Exp. Cell Res. 2005, 310, 205–217. [Google Scholar] [CrossRef]
  174. Li, Y.; Jiang, D.; Liang, J.; Meltzer, E.B.; Gray, A.; Miura, R.; Wogensen, L.; Yamaguchi, Y.; Noble, P.W. Severe Lung Fibrosis Requires an Invasive Fibroblast Phenotype Regulated by Hyaluronan and CD44. J. Exp. Med. 2011, 208, 1459–1471. [Google Scholar] [CrossRef]
  175. Suchankova, M.; Zsemlye, E.; Urban, J.; Baráth, P.; Kohútová, L.; Siváková, B.; Ganovska, M.; Tibenska, E.; Szaboova, K.; Tedlova, E.; et al. The Bronchoalveolar Lavage Fluid CD44 as a Marker for Pulmonary Fibrosis in Diffuse Parenchymal Lung Diseases. Front. Immunol. 2025, 15, 1479458. [Google Scholar] [CrossRef]
  176. Xia, H.; Herrera, J.; Smith, K.; Yang, L.; Gilbertsen, A.; Benyumov, A.; Racila, E.; Bitterman, P.B.; Henke, C.A. Hyaluronan/CD44 Axis Regulates S100A4-Mediated Mesenchymal Progenitor Cell Fibrogenicity in Idiopathic Pulmonary Fibrosis. Am. J. Physiol. Lung Cell. Mol. Physiol. 2021, 320, L926–L941. [Google Scholar] [CrossRef]
  177. Li, G.; Zhang, Y.; Jiang, H.; Wu, X.; Hao, Y.; Su, Y.; Zou, Y.; Xian, W.; Wang, F.; Du, Q. PPARG/SPP1/CD44 Signaling Pathway in Alveolar Macrophages: Mechanisms of Lipid Dysregulation and Therapeutic Targets in Idiopathic Pulmonary Fibrosis. Heliyon 2025, 11, e41628. [Google Scholar] [CrossRef]
  178. Yasaka, N.; Furue, M.; Tamaki, K. CD44 Expression in Normal Human Skin and Skin Tumors. J. Dermatol. 1995, 22, 88–94. [Google Scholar] [CrossRef] [PubMed]
  179. Bourguignon, L.Y.W.; Ramez, M.; Gilad, E.; Singleton, P.A.; Man, M.-Q.; Crumrine, D.A.; Elias, P.M.; Feingold, K.R. Hyaluronan–CD44 Interaction Stimulates Keratinocyte Differentiation, Lamellar Body Formation/Secretion, and Permeability Barrier Homeostasis. J. Investig. Dermatol. 2006, 126, 1356–1365. [Google Scholar] [CrossRef] [PubMed]
  180. Guo, J.; Yang, T.; Zhang, W.; Yu, K.; Xu, X.; Li, W.; Song, L.; Gu, X.; Cao, R.; Cui, S. Inhibition of CD44 Suppresses the Formation of Fibrotic Scar after Spinal Cord Injury via the JAK2/STAT3 Signaling Pathway. iScience 2024, 27, 108935. [Google Scholar] [CrossRef]
  181. Weng, X.; Yue, W.; Shang, L.; Wang, D.; Xu, Y.; Chen, Y.; Ge, J. Inhibition of CD44 Attenuates Pressure Overload-Induced Cardiac and Lung Inflammation, Fibrosis, and Heart Failure Progression. Eur. Heart J. 2020, 41, ehaa946.0878. [Google Scholar] [CrossRef]
  182. Osawa, Y.; Kawai, H.; Tsunoda, T.; Komatsu, H.; Okawara, M.; Tsutsui, Y.; Yoshida, Y.; Yoshikawa, S.; Mori, T.; Yamazoe, T.; et al. Cluster of Differentiation 44 Promotes Liver Fibrosis and Serves as a Biomarker in Congestive Hepatopathy. Hepatol. Commun. 2021, 5, 1437–1447. [Google Scholar] [CrossRef]
  183. Han, J.; Lee, C.; Jung, Y. Current Evidence and Perspectives of Cluster of Differentiation 44 in the Liver’s Physiology and Pathology. Int. J. Mol. Sci. 2024, 25, 4749. [Google Scholar] [CrossRef]
  184. Petukhov, D.; Richter-Dayan, M.; Fridlender, Z.; Breuer, R.; Wallach-Dayan, S.B. Increased Regeneration Following Stress-Induced Lung Injury in Bleomycin-Treated Chimeric Mice with CD44 Knockout Mesenchymal Cells. Cells 2019, 8, 1211. [Google Scholar] [CrossRef]
  185. Fernández-Tabanera, E.; Melero-Fernández de Mera, R.M.; Alonso, J. CD44 In Sarcomas: A Comprehensive Review and Future Perspectives. Front. Oncol. 2022, 12, 909450. [Google Scholar] [CrossRef]
  186. Hiraga, T.; Ito, S.; Nakamura, H. Cancer Stem-like Cell Marker CD44 Promotes Bone Metastases by Enhancing Tumorigenicity, Cell Motility, and Hyaluronan Production. Cancer Res. 2013, 73, 4112–4122. [Google Scholar] [CrossRef]
  187. Menko, A.S.; Romisher, A.; Walker, J.L. The Pro-Fibrotic Response of Mesenchymal Leader Cells to Lens Wounding Involves Hyaluronic Acid, Its Receptor RHAMM, and Vimentin. Front. Cell Dev. Biol. 2022, 10, 862423. [Google Scholar] [CrossRef]
  188. Lin, C.-H.; Hung, P.-H.; Chen, Y.-J. CD44 Is Associated with the Aggressive Phenotype of Nasopharyngeal Carcinoma through Redox Regulation. Int. J. Mol. Sci. 2013, 14, 13266–13281. [Google Scholar] [CrossRef]
  189. Paulis, Y.W.J.; Huijbers, E.J.M.; van der Schaft, D.W.J.; Soetekouw, P.M.M.B.; Pauwels, P.; Tjan-Heijnen, V.C.G.; Griffioen, A.W. CD44 Enhances Tumor Aggressiveness by Promoting Tumor Cell Plasticity. Oncotarget 2015, 6, 19634–19646. [Google Scholar] [CrossRef] [PubMed]
  190. Suleiman, M.; Abdulrahman, N.; Yalcin, H.; Mraiche, F. The Role of CD44, Hyaluronan and NHE1 in Cardiac Remodeling. Life Sci. 2018, 209, 197–201. [Google Scholar] [CrossRef] [PubMed]
  191. Cao, Z.; Wang, A.; Leng, P. Aberrant N-cadherin expression in cancer. Biomed. Pharmacother. 2019, 118, 109320. [Google Scholar] [CrossRef] [PubMed]
  192. Shang, Z.; Cai, Q.; Zhang, M.; Zhu, S.; Ma, Y.; Sun, L.; Jiang, N.; Tian, J.; Niu, X.; Chen, J.; et al. A Switch from CD44+ Cell to EMT Cell Drives the Metastasis of Prostate Cancer. Oncotarget 2014, 6, 1202–1216. [Google Scholar] [CrossRef]
  193. Yusupov, M.; Privat-Maldonado, A.; Cordeiro, R.M.; Verswyvel, H.; Shaw, P.; Razzokov, J.; Smits, E.; Bogaerts, A. Oxidative Damage to Hyaluronan–CD44 Interactions as an Underlying Mechanism of Action of Oxidative Stress-Inducing Cancer Therapy. Redox Biol. 2021, 43, 101968. [Google Scholar] [CrossRef]
  194. Nurwidya, F.; Takahashi, F.; Kato, M.; Baskoro, H.; Hidayat, M.; Wirawan, A.; Takahashi, K. CD44 Silencing Decreases the Expression of Stem Cell-Related Factors Induced by Transforming Growth Factor Β1 and Tumor Necrosis Factor α in Lung Cancer: Preliminary Findings. Bosn. J. Basic Med. Sci. 2017, 17, 228–234. [Google Scholar] [CrossRef]
  195. Tirella, A.; Kloc-Muniak, K.; Good, L.; Ridden, J.; Ashford, M.; Puri, S.; Tirelli, N. CD44 Targeted Delivery of siRNA by Using HA-Decorated Nanotechnologies for KRAS Silencing in Cancer Treatment. Int. J. Pharm. 2019, 561, 114–123. [Google Scholar] [CrossRef]
  196. Gul-Uludağ, H.; Valencia-Serna, J.; Kucharski, C.; Marquez-Curtis, L.A.; Jiang, X.; Larratt, L.; Janowska-Wieczorek, A.; Uludağ, H. Polymeric Nanoparticle-Mediated Silencing of CD44 Receptor in CD34+ Acute Myeloid Leukemia Cells. Leuk. Res. 2014, 38, 1299–1308. [Google Scholar] [CrossRef]
  197. Ebrahimi, M.; Khalili, N.; Razi, S.; Keshavarz-Fathi, M.; Khalili, N.; Rezaei, N. Effects of Lead and Cadmium on the Immune System and Cancer Progression. J. Environ. Health Sci. Eng. 2020, 18, 335–343. [Google Scholar] [CrossRef]
  198. Ju, H.; Arumugam, P.; Lee, J.; Song, J.M. Impact of Environmental Pollutant Cadmium on the Establishment of a Cancer Stem Cell Population in Breast and Hepatic Cancer. ACS Omega 2017, 2, 563–572. [Google Scholar] [CrossRef]
  199. Li, J.; Jiang, H.; Zhu, Y.; Ma, Z.; Li, B.; Dong, J.; Xiao, C.; Hu, A. Fine Particulate Matter (PM2.5) Induces the Stem Cell-like Properties of Hepatocellular Carcinoma by Activating ROS/Nrf2/Keap1-Mediated Autophagy. Ecotoxicol. Environ. Saf. 2024, 272, 116052. [Google Scholar] [CrossRef] [PubMed]
  200. Wei, H.; Liang, F.; Cheng, W.; Zhou, R.; Wu, X.; Feng, Y.; Wang, Y. The Mechanisms for Lung Cancer Risk of PM2.5: Induction of Epithelial-Mesenchymal Transition and Cancer Stem Cell Properties in Human Non-Small Cell Lung Cancer Cells. Environ. Toxicol. 2017, 32, 2341–2351. [Google Scholar] [CrossRef] [PubMed]
  201. Schneider, A.; Alexis, N.E.; Diaz-Sanchez, D.; Neas, L.M.; Harder, S.; Herbst, M.C.; Cascio, W.E.; Buse, J.B.; Peters, A.; Devlin, R.B. Ambient PM2.5 Exposure Up-Regulates the Expression of Costimulatory Receptors on Circulating Monocytes in Diabetic Individuals. Environ. Health Perspect. 2011, 119, 778–783. [Google Scholar] [CrossRef] [PubMed]
  202. Figure 3. From LncRNA NEAT1 Contributes to the Acquisition of a Tumor Like-Phenotype Induced by PM 2.5 in Lung Bronchial Epithelial Cells via HIF-1α Activation. Available online: https://media.springernature.com/full/springer-static/image/art%3A10.1007%2Fs11356-021-13735-7/MediaObjects/11356_2021_13735_Fig3_HTML.png (accessed on 18 July 2025).
  203. Su, Y.; Gao, J.; Kaur, P.; Wang, Z. Neutrophils and Macrophages as Targets for Development of Nanotherapeutics in Inflammatory Diseases. Pharmaceutics 2020, 12, 1222. [Google Scholar] [CrossRef]
  204. Xu, X.; Jiang, S.Y.; Wang, T.-Y.; Bai, Y.; Zhong, M.; Wang, A.; Lippmann, M.; Chen, L.-C.; Rajagopalan, S.; Sun, Q. Inflammatory Response to Fine Particulate Air Pollution Exposure: Neutrophil versus Monocyte. PLoS ONE 2013, 8, e71414. [Google Scholar] [CrossRef]
  205. Figure 4. From LncRNA NEAT1 Contributes to the Acquisition of a Tumor Like-Phenotype Induced by PM 2.5 in Lung Bronchial Epithelial Cells via HIF-1α Activation. Available online: https://media.springernature.com/full/springer-static/image/art%3A10.1007%2Fs11356-021-13735-7/MediaObjects/11356_2021_13735_Fig4_HTML.png?as=webp (accessed on 18 July 2025).
  206. Pan, J.; Liu, P.; Yu, X.; Zhang, Z.; Liu, J. The Adverse Role of Endocrine Disrupting Chemicals in the Reproductive System. Front. Endocrinol. 2024, 14, 1324993. [Google Scholar] [CrossRef]
  207. Singh, R.D.; Koshta, K.; Tiwari, R.; Khan, H.; Sharma, V.; Srivastava, V. Developmental Exposure to Endocrine Disrupting Chemicals and Its Impact on Cardio-Metabolic-Renal Health. Front. Toxicol. 2021, 3, 663372. [Google Scholar] [CrossRef]
  208. Endocrine-Disrupting Chemicals’ (EDCs) Effects on Tumour Microenvironment and Cancer Progression: Emerging Contribution of RACK1. Available online: https://www.mdpi.com/1422-0067/21/23/9229 (accessed on 18 July 2025).
  209. Erkekoglu, P.; Kocer-Gumusel, B.; Erkekoglu, P.; Kocer-Gumusel, B. Environmental Effects of Endocrine-Disrupting Chemicals: A Special Focus on Phthalates and Bisphenol A. In Environmental Health Risk—Hazardous Factors to Living Species; IntechOpen: London, UK, 2016; ISBN 978-953-51-2402-3. [Google Scholar]
  210. Primeaux, M.; Gowrikumar, S.; Dhawan, P. Role of CD44 Isoforms in Epithelial-Mesenchymal Plasticity and Metastasis. Clin. Exp. Metastasis 2022, 39, 391–406. [Google Scholar] [CrossRef]
  211. Xu, H.; Tian, Y.; Yuan, X.; Wu, H.; Liu, Q.; Pestell, R.G.; Wu, K. The Role of CD44 in Epithelial–Mesenchymal Transition and Cancer Development. Onco Targets Ther. 2015, 8, 3783–3792. [Google Scholar] [CrossRef]
  212. Pesonen, M.; Vähäkangas, K. Contribution of Common Plastic-Related Endocrine Disruptors to Epithelial-Mesenchymal Transition (EMT) and Tumor Progression. Chemosphere 2022, 309, 136560. [Google Scholar] [CrossRef] [PubMed]
  213. Chen, X.; Tian, F.; Wu, J.; Liu, L.; Li, Y.; Yu, G.; Duan, H.; Jiang, Y.; Liu, S.; He, Y.; et al. Associations of Phthalates with NAFLD and Liver Fibrosis: A Nationally Representative Cross-Sectional Study from NHANES 2017 to 2018. Front. Nutr. 2022, 9, 1059675. [Google Scholar] [CrossRef] [PubMed]
  214. Zhang, F.; Zhen, H.; Cheng, H.; Hu, F.; Jia, Y.; Huang, B.; Jiang, M. Di-(2-Ethylhexyl) Phthalate Exposure Induces Liver Injury by Promoting Ferroptosis via Downregulation of GPX4 in Pregnant Mice. Front. Cell Dev. Biol. 2022, 10, 1014243. [Google Scholar] [CrossRef] [PubMed]
  215. Liu, L.F.; Kodama, K.; Wei, K.; Tolentino, L.L.; Choi, O.; Engleman, E.G.; Butte, A.J.; McLaughlin, T. The Receptor CD44 Is Associated with Systemic Insulin Resistance and Proinflammatory Macrophages in Human Adipose Tissue. Diabetologia 2015, 58, 1579–1586. [Google Scholar] [CrossRef]
  216. Kodama, K.; Toda, K.; Morinaga, S.; Yamada, S.; Butte, A.J. Anti-CD44 Antibody Treatment Lowers Hyperglycemia and Improves Insulin Resistance, Adipose Inflammation, and Hepatic Steatosis in Diet-Induced Obese Mice. Diabetes 2015, 64, 867–875. [Google Scholar] [CrossRef]
  217. Khan, N.G.; Correia, J.; Adiga, D.; Rai, P.S.; Dsouza, H.S.; Chakrabarty, S.; Kabekkodu, S.P. A Comprehensive Review on the Carcinogenic Potential of Bisphenol A: Clues and Evidence. Environ. Sci. Pollut. Res. Int. 2021, 28, 19643–19663. [Google Scholar] [CrossRef]
  218. Ryszawy, D.; Pudełek, M.; Kochanowski, P.; Janik-Olchawa, N.; Bogusz, J.; Rąpała, M.; Koczurkiewicz, P.; Mikołajczyk, J.; Borek, I.; Kędracka-Krok, S.; et al. High Bisphenol A Concentrations Augment the Invasiveness of Tumor Cells through Snail-1/Cx43/ERRγ-Dependent Epithelial-Mesenchymal Transition. Toxicol. Vitr. 2020, 62, 104676. [Google Scholar] [CrossRef]
  219. Bora, S.S.; Gogoi, R.; Sharma, M.R.; Anshu; Borah, M.P.; Deka, P.; Bora, J.; Naorem, R.S.; Das, J.; Teli, A.B. Microplastics and Human Health: Unveiling the Gut Microbiome Disruption and Chronic Disease Risks. Front. Cell. Infect. Microbiol. 2024, 14, 1492759. [Google Scholar] [CrossRef]
  220. Aliya, S.; Alhammadi, M.; Ilangovan, S.; Han, S.; Tamang, S.; Son, B.; Lee, H.U.; Huh, Y.S. Microplastics: An Emerging Environmental Risk Factor for Gut Microbiota Dysbiosis and Cancer Development? Environ. Chem. Ecotoxicol. 2025, 7, 706–728. [Google Scholar] [CrossRef]
  221. Sofield, C.E.; Anderton, R.S.; Gorecki, A.M. Mind over Microplastics: Exploring Microplastic-Induced Gut Disruption and Gut-Brain-Axis Consequences. Curr. Issues Mol. Biol. 2024, 46, 4186–4202. [Google Scholar] [CrossRef]
  222. Yang, W.; Jannatun, N.; Zeng, Y.; Liu, T.; Zhang, G.; Chen, C.; Li, Y. Impacts of Microplastics on Immunity. Front. Toxicol. 2022, 4, 956885. [Google Scholar] [CrossRef] [PubMed]
  223. Huang, H.; Hou, J.; Liao, Y.; Wei, F.; Xing, B. Polyethylene Microplastics Impede the Innate Immune Response by Disrupting the Extracellular Matrix and Signaling Transduction. iScience 2023, 26, 107390. [Google Scholar] [CrossRef] [PubMed]
  224. Limonta, G.; Mancia, A.; Benkhalqui, A.; Bertolucci, C.; Abelli, L.; Fossi, M.C.; Panti, C. Microplastics Induce Transcriptional Changes, Immune Response and Behavioral Alterations in Adult Zebrafish. Sci. Rep. 2019, 9, 15775. [Google Scholar] [CrossRef] [PubMed]
  225. Zhi, L.; Li, Z.; Su, Z.; Wang, J. Immunotoxicity of Microplastics: Carrying Pathogens and Destroying the Immune System. TrAC Trends Anal. Chem. 2024, 177, 117817. [Google Scholar] [CrossRef]
  226. Cheng, Y.; Yang, Y.; Bai, L.; Cui, J. Microplastics: An Often-Overlooked Issue in the Transition from Chronic Inflammation to Cancer. J. Transl. Med. 2024, 22, 959. [Google Scholar] [CrossRef]
  227. Dzierżyński, E.; Gawlik, P.J.; Puźniak, D.; Flieger, W.; Jóźwik, K.; Teresiński, G.; Forma, A.; Wdowiak, P.; Baj, J.; Flieger, J. Microplastics in the Human Body: Exposure, Detection, and Risk of Carcinogenesis: A State-of-the-Art Review. Cancers 2024, 16, 3703. [Google Scholar] [CrossRef]
  228. Osman, A.I.; Hosny, M.; Eltaweil, A.S.; Omar, S.; Elgarahy, A.M.; Farghali, M.; Yap, P.-S.; Wu, Y.-S.; Nagandran, S.; Batumalaie, K.; et al. Microplastic Sources, Formation, Toxicity and Remediation: A Review. Environ. Chem. Lett. 2023, 21, 2129–2169. [Google Scholar] [CrossRef]
  229. Li, S.; Li, C.; Zhang, Y.; He, X.; Chen, X.; Zeng, X.; Liu, F.; Chen, Y.; Chen, J. Targeting Mechanics-Induced Fibroblast Activation through CD44-RhoA-YAP Pathway Ameliorates Crystalline Silica-Induced Silicosis. Theranostics 2019, 9, 4993–5008. [Google Scholar] [CrossRef]
Figure 1. Multifunctional roles of CD44 in cell signaling, tissue homeostasis, immunity, and cancer progression (based on [62,64,65,66,70,71,73,75,76,77,78,79]). CD44, as a transmembrane glycoprotein, binds hyaluronic acid and additional ligands such as osteopontin, collagens, fibronectin, and MMPs, thereby integrating cell–ECM interactions. Beyond its structural role, CD44 acts as a signaling platform that organizes growth factors and adaptor proteins, activating pathways including PI3K/AKT, MAPK, and Rho-GTPases. These interactions support immune cell adhesion, migration, and hematopoiesis, while in cancer, CD44 facilitates invasion, metastasis, therapy resistance, and maintenance of tumor stem-like properties. Thus, CD44 emerges as a central regulator of adhesion, immune responses, tissue homeostasis, and malignant progression.
Figure 1. Multifunctional roles of CD44 in cell signaling, tissue homeostasis, immunity, and cancer progression (based on [62,64,65,66,70,71,73,75,76,77,78,79]). CD44, as a transmembrane glycoprotein, binds hyaluronic acid and additional ligands such as osteopontin, collagens, fibronectin, and MMPs, thereby integrating cell–ECM interactions. Beyond its structural role, CD44 acts as a signaling platform that organizes growth factors and adaptor proteins, activating pathways including PI3K/AKT, MAPK, and Rho-GTPases. These interactions support immune cell adhesion, migration, and hematopoiesis, while in cancer, CD44 facilitates invasion, metastasis, therapy resistance, and maintenance of tumor stem-like properties. Thus, CD44 emerges as a central regulator of adhesion, immune responses, tissue homeostasis, and malignant progression.
Ijms 26 08870 g001
Figure 3. Functional interactions of CD44 with components of the extracellular matrix, intracellular signaling molecules, and membrane-associated partners (based on [12,25,120,121,122,123,124,125,126,127,128,129]). The figure summarizes the diverse molecular interactions of CD44, highlighting its role as a multifunctional hub. In the extracellular space, CD44 binds to hyaluronic acid, collagens, laminin, fibronectin, and osteopontin, mediating adhesion, migration, and tumor cell invasion. Through its cytoplasmic domain, CD44 associates with ERM proteins (ezrin, moesin, radixin) and NF2/merlin, linking the receptor to the actin cytoskeleton and supporting the dynamic regulation of microvilli and cell polarity. Additionally, CD44 interacts with signaling and enzymatic proteins such as kinases (e.g., PKN2, Rho-GTPases), transmitting signals that regulate cytoskeletal organization and cell motility. Cooperation with other membrane receptors, including EGFR and podoplanin (PDPN), further integrates CD44 into signaling networks that control proliferation, migration, and cancer invasion. Finally, the complex of CD44 with CD74 and MIF illustrates its role in immune cell survival and regulation, underscoring the receptor’s significance in inflammation, immunity, and tumor progression.
Figure 3. Functional interactions of CD44 with components of the extracellular matrix, intracellular signaling molecules, and membrane-associated partners (based on [12,25,120,121,122,123,124,125,126,127,128,129]). The figure summarizes the diverse molecular interactions of CD44, highlighting its role as a multifunctional hub. In the extracellular space, CD44 binds to hyaluronic acid, collagens, laminin, fibronectin, and osteopontin, mediating adhesion, migration, and tumor cell invasion. Through its cytoplasmic domain, CD44 associates with ERM proteins (ezrin, moesin, radixin) and NF2/merlin, linking the receptor to the actin cytoskeleton and supporting the dynamic regulation of microvilli and cell polarity. Additionally, CD44 interacts with signaling and enzymatic proteins such as kinases (e.g., PKN2, Rho-GTPases), transmitting signals that regulate cytoskeletal organization and cell motility. Cooperation with other membrane receptors, including EGFR and podoplanin (PDPN), further integrates CD44 into signaling networks that control proliferation, migration, and cancer invasion. Finally, the complex of CD44 with CD74 and MIF illustrates its role in immune cell survival and regulation, underscoring the receptor’s significance in inflammation, immunity, and tumor progression.
Ijms 26 08870 g003
Table 1. Characteristics of selected CD44 variants and their potential biological significance (based on [30,31]). This table summarizes key variants of the CD44 gene, detailing their specific amino acid changes, structural locations, and functional relevance to various biological processes. CD44, a transmembrane glycoprotein, plays a crucial role in cell adhesion, migration, and the regulation of immune responses. The variants presented here highlight the diversity in CD44 structure and its potential implications in immunohematology, cell signaling, and receptor-ligand interactions. Each variant is associated with specific regions of the protein, ranging from the extracellular domain to the transmembrane region, and may influence its function in different physiological and pathological contexts.
Table 1. Characteristics of selected CD44 variants and their potential biological significance (based on [30,31]). This table summarizes key variants of the CD44 gene, detailing their specific amino acid changes, structural locations, and functional relevance to various biological processes. CD44, a transmembrane glycoprotein, plays a crucial role in cell adhesion, migration, and the regulation of immune responses. The variants presented here highlight the diversity in CD44 structure and its potential implications in immunohematology, cell signaling, and receptor-ligand interactions. Each variant is associated with specific regions of the protein, ranging from the extracellular domain to the transmembrane region, and may influence its function in different physiological and pathological contexts.
Variant (dbSNP ID)Amino Acid ChangeStructural
Location
Functional Description and Biological Relevance
VAR_006490 (rs3694738421)Arg46Pro (arginine → proline)Extracellular domain (N-terminal)This variant underlies the Indian (In) blood group system and differentiates Ina and Inb antigens on erythrocytes. The proline-coding allele determines the rare Ina antigen, which may elicit alloimmune transfusion reactions. It is not pathogenic in systemic health but has significant clinical relevance in immunohematology.
VAR_030325 (rs11607491)Substitution at position 393Extracellular domainAlthough its functional consequences are not fully characterized, the variant is located in the ligand-binding region and may affect CD44’s interaction with hyaluronan or extracellular matrix components.
VAR_021147 (rs96666074)Substitution at position 417Extracellular domainLocated near known glycosylation sites, this substitution may alter CD44 glycosylation patterns, potentially impacting receptor-ligand interactions or immune recognition.
VAR_030326 (rs14675589)Substitution at position 479Juxtamembrane region (extracellular–transmembrane junction)This variant may affect the membrane topology of CD44 and its susceptibility to proteolytic cleavage (shedding), thereby influencing the levels of soluble CD44 (sCD44) and downstream signaling.
VAR_030327 (rs122733971)Substitution at position 494Transmembrane domain or adjacent regionThis mutation may modulate the anchoring of CD44 in the membrane and alter its interactions with neighboring receptors or lipid rafts, influencing receptor clustering and signaling efficiency.
Table 2. Detailed table showing known CD44 protein ligands, grouped by molecule type and their primary biological functions (based on [4,34,35,36,37,38,39,41,42]). The table highlights the diversity of CD44 ligands, ranging from glycosaminoglycans and extracellular matrix proteins to adhesive proteins, growth factors, proteases, and acute phase proteins. These interactions emphasize the pleiotropic functions of CD44 in cell adhesion, migration, proliferation, differentiation, immune response modulation, and tumor progression. Furthermore, the compilation demonstrates how CD44 can act as an integrative receptor that organizes ligand complexes and facilitates downstream signaling pathways such as PI3K, MAPK, and Rho-GTPases.
Table 2. Detailed table showing known CD44 protein ligands, grouped by molecule type and their primary biological functions (based on [4,34,35,36,37,38,39,41,42]). The table highlights the diversity of CD44 ligands, ranging from glycosaminoglycans and extracellular matrix proteins to adhesive proteins, growth factors, proteases, and acute phase proteins. These interactions emphasize the pleiotropic functions of CD44 in cell adhesion, migration, proliferation, differentiation, immune response modulation, and tumor progression. Furthermore, the compilation demonstrates how CD44 can act as an integrative receptor that organizes ligand complexes and facilitates downstream signaling pathways such as PI3K, MAPK, and Rho-GTPases.
Ligand TypeLigand NameDescription and Biological Functions
Glycosaminoglycans (GAGs)Hyaluronic acid (HA)Primary ligand of CD44; regulates adhesion, migration, proliferation, differentiation, and inflammatory response
Heparan sulfate (HS)Facilitates the binding of growth factors, present in proteoglycan forms of CD44
Chondroitin sulfate (CS)Associated with the CD44v isoform; influences interactions with the ECM
Extracellular matrix proteinsOsteopontin (OPN, SPP1)Modulation of cell migration and adhesion; expression in inflammation and tumorigenesis
Fibronectin (FN)Adhesion and signaling; interacts with integrins and CD44
LamininSupports epithelial cell interactions with the basement membrane
Collagen type I, II, III, IVEffect on cell adhesion, migration, and invasion
Adhesive proteinsSelectins (L-, E-selectin)—via HCELL formParticipation in leukocyte rolling is meaningful in the immune response
ICAM-1 (ang. intercellular adhesion molecule 1)Supporting role in lymphocyte transmigration
Signaling proteins and growth factorsTGF-β, HGF, VEGF, EGF—indirectly through complexes with HA or heparanCD44 enables local presentation and concentration of growth signals
ProteasesMatrix metalloproteinases (MMP-2, MMP-9)Interactions with CD44 promote ECM degradation and cell migration.
Acute phase proteinsPentraxin 3 (PTX3)Regulation of inflammatory response, interaction with HA and CD44
OtherSerpins, complement proteinsLess frequently described ligands: immunological and proteolytic significance
Receptors and co-receptors (e.g., EGFR, TGF-βR)—signaling complexesThey cooperate with CD44 in the activation of PI3K, MAPK, and Rho-GTPase pathways
Table 3. Characteristics of ERM family proteins in the context of interaction with CD44 (based on [51,86,87,88,89,90]). The table summarizes structural and functional features of ezrin, radixin, and moesin, emphasizing their subcellular localization and roles in linking the actin cytoskeleton with the plasma membrane. Each ERM protein directly interacts with CD44, stabilizing its position within microvilli, intercellular junctions, or migratory structures, thereby facilitating adhesion, signaling, and cytoskeletal reorganization. The functional consequences of these interactions include the regulation of cell polarity, migration, inflammatory responses, and the mechanical coupling of adhesion with intracellular signaling. Dysfunctions of ERM–CD44 interactions are associated with diverse pathologies such as cancers, viral infections, chronic inflammation, and hereditary disorders (e.g., hearing loss linked to RDX mutations).
Table 3. Characteristics of ERM family proteins in the context of interaction with CD44 (based on [51,86,87,88,89,90]). The table summarizes structural and functional features of ezrin, radixin, and moesin, emphasizing their subcellular localization and roles in linking the actin cytoskeleton with the plasma membrane. Each ERM protein directly interacts with CD44, stabilizing its position within microvilli, intercellular junctions, or migratory structures, thereby facilitating adhesion, signaling, and cytoskeletal reorganization. The functional consequences of these interactions include the regulation of cell polarity, migration, inflammatory responses, and the mechanical coupling of adhesion with intracellular signaling. Dysfunctions of ERM–CD44 interactions are associated with diverse pathologies such as cancers, viral infections, chronic inflammation, and hereditary disorders (e.g., hearing loss linked to RDX mutations).
FeatureEzrinRadixinMoesin
Gene symbol (human)EZRRDXMSN
Subcellular localizationCell membrane, microvilli, surface of cellular projectionsIntercellular junctions, cortical cytoplasmMicrovilli, lamellipodia, and ECM contact zones
Biological functionLinks the actin cytoskeleton to the plasma membrane; involved in shaping microvilli and cell adhesionStabilizes the plasma membrane and intercellular junctions; regulates membrane elasticityRegulates cell shape, leukocyte transmigration, and inflammatory response
Interactions with CD44Direct interaction with CD44’s cytoplasmic domain; involved in targeting CD44 to microvilli and lipid raftsAnchors CD44 at sites of intercellular contact and cytoskeletal reorganizationStabilizes CD44 in migrating immune cells; involved in Rho/Rac pathway activation
Functional significance of CD44–ERMIntegrates extracellular signals with intracellular responses; mechanical coupling of adhesion and signalingMaintains cell polarity and membrane tension; supports CD44 signalingEnhances pro-inflammatory response; involved in cytoskeletal reorganization during migration
Diseases associated with dysfunctionCancers (e.g., gastric cancer, leukemias), kidney diseases, viral infections (e.g., HIV, EBV)Hearing loss (RDX mutations), liver and gallbladder cancersChronic inflammatory conditions, lymphomas, head and neck cancers
Table 4. Summary of CD44 Domain Structure (Canonical Isoform) [8]. The table outlines the structural organization of CD44, dividing it into extracellular, transmembrane, and cytoplasmic domains. The extracellular domain mediates binding to hyaluronic acid and other ECM ligands and undergoes glycosylation, while the transmembrane segment anchors the receptor in the plasma membrane and defines its orientation. The cytoplasmic domain interacts with ERM proteins, undergoes regulatory phosphorylation, and transduces signals that connect extracellular cues with intracellular responses. Together, these domains provide the structural and functional basis for CD44’s role as an adhesion molecule and signaling platform.
Table 4. Summary of CD44 Domain Structure (Canonical Isoform) [8]. The table outlines the structural organization of CD44, dividing it into extracellular, transmembrane, and cytoplasmic domains. The extracellular domain mediates binding to hyaluronic acid and other ECM ligands and undergoes glycosylation, while the transmembrane segment anchors the receptor in the plasma membrane and defines its orientation. The cytoplasmic domain interacts with ERM proteins, undergoes regulatory phosphorylation, and transduces signals that connect extracellular cues with intracellular responses. Together, these domains provide the structural and functional basis for CD44’s role as an adhesion molecule and signaling platform.
SegmentAmino Acid RangeFunction
Extracellular domain21–649Binding to hyaluronic acid (HA) and other ECM ligands; glycosylation modifications.
Transmembrane
segment
650–670Anchoring in the membrane, defining N-/C-terminal orientation
Cytoplasmic domain671–742Interactions with ERM proteins, signal transduction, and regulatory phosphorylation
Table 6. Overview of key PTMs of CD44 (based on [128,130,131,132,133,134,135,136,137,138,139,140]). The table summarizes major post-translational modifications (PTMs) of CD44, including glycosylation, phosphorylation, proteolytic cleavage, and ubiquitination. These modifications occur in distinct domains of the receptor and regulate folding, stability, ligand binding, receptor activation, and intracellular trafficking. Collectively, PTMs fine-tune CD44 functions by modulating adhesion, migration, cytoskeletal reorganization, and signal transduction. Dysregulation of these processes contributes to altered immune responses, tumor progression, and metastasis, underlining the biological relevance of PTMs in CD44-mediated pathways.
Table 6. Overview of key PTMs of CD44 (based on [128,130,131,132,133,134,135,136,137,138,139,140]). The table summarizes major post-translational modifications (PTMs) of CD44, including glycosylation, phosphorylation, proteolytic cleavage, and ubiquitination. These modifications occur in distinct domains of the receptor and regulate folding, stability, ligand binding, receptor activation, and intracellular trafficking. Collectively, PTMs fine-tune CD44 functions by modulating adhesion, migration, cytoskeletal reorganization, and signal transduction. Dysregulation of these processes contributes to altered immune responses, tumor progression, and metastasis, underlining the biological relevance of PTMs in CD44-mediated pathways.
Type of ModificationLocation/Target ResiduesFunctional DescriptionBiological Significance
N-glycosylationAsn within the Asn-X-Ser/Thr motif (e.g., in the LINK domain)Attachment of N-glycansEssential for proper folding, stability, and affinity for HA
O-glycosylationSer/Thr (especially in variable splice regions, e.g., Thr-637/638)Addition of short sugar chains and glycosaminoglycans (e.g., chondroitin sulfate)Modulates ligand recognition, protects from proteolysis, and affects cell migration
PhosphorylationSer-672, Ser-706; other Ser/Thr residues in the C-terminal tailRegulates receptor activation status and interactions with adaptor proteins (e.g., ERM)Alters adhesion and migration signaling, influences cytoskeletal organization
Proteolysis (shedding)Near the transmembrane domain (extracellular side)Cleavage of the extracellular domain by MMPs and other proteasesReduces surface CD44 expression, generates soluble form (sCD44), and is potentially further cleaved by γ-secretase and nuclear signaling
UbiquitinationLys-704, Lys-715 (in the cytoplasmic tail)Covalent modification affecting intracellular trafficking and degradationMay regulate receptor abundance and endosomal sorting
Table 8. Experimental evidence of CD44 involvement in organ fibrosis (based on [6,20,128,191,192,193,194,195,196,197]). The table compiles preclinical and translational studies that demonstrate the role of CD44 in fibrotic remodeling across multiple organs. In pulmonary fibrosis, the HA–CD44 axis drives fibroblast invasiveness, while genetic or antibody-mediated blockade of CD44 markedly reduces fibrotic progression. In the skin, altered CD44 expression and elevated soluble CD44 levels correlate with disease severity, suggesting both pathogenic and protective roles depending on the context. Cardiac and hepatic fibrosis models further highlight CD44 as a mediator of myofibroblast activation, collagen deposition, and tissue remodeling, positioning it as a key integrator of inflammatory and fibrotic signals and a potential therapeutic target. ↑ increased level/increased expression.
Table 8. Experimental evidence of CD44 involvement in organ fibrosis (based on [6,20,128,191,192,193,194,195,196,197]). The table compiles preclinical and translational studies that demonstrate the role of CD44 in fibrotic remodeling across multiple organs. In pulmonary fibrosis, the HA–CD44 axis drives fibroblast invasiveness, while genetic or antibody-mediated blockade of CD44 markedly reduces fibrotic progression. In the skin, altered CD44 expression and elevated soluble CD44 levels correlate with disease severity, suggesting both pathogenic and protective roles depending on the context. Cardiac and hepatic fibrosis models further highlight CD44 as a mediator of myofibroblast activation, collagen deposition, and tissue remodeling, positioning it as a key integrator of inflammatory and fibrotic signals and a potential therapeutic target. ↑ increased level/increased expression.
OrganExperimental ModelKey ObservationsFunctional Conclusions
Lungs (IPF)Bleomycin-induced pulmonary fibrosis in mice; HAS2 overexpression; CD44 knockout
  • HAS2↑ → severe fibrosis; HAS2 KO protects against fibrosis
  • CD44 KO–loss of invasive fibroblast phenotype and fibrosis suppression
  • Anti-CD44 antibody alleviates pulmonary changes
  • IPF fibroblasts: high CD44-dependent invasiveness
The HA–CD44 axis is essential for the pro-fibrotic phenotype in the lungs; CD44 blockade reduces fibrosis and fibroblast invasiveness.
Skin (SSc)Patient-derived fibroblasts from systemic sclerosis; sCD44 concentration analysis; wound healing in CD44 KO mice
  • Altered CD44 expression in fibroblasts
  • Increased sCD44 correlates with a milder clinical phenotype (less skin/lung involvement)
  • CD44 KO–reduced collagen and slower scar formation
CD44 may support both wound healing and pathological skin sclerosis; sCD44 potentially exerts protective effects
HeartAng II-induced fibrosis; pressure overload model; CD44 KO
  • Ang II–increased CD44 expression + TNFα/NF-κB activation + macrophage migration
  • CD44 KO–less collagen, reduced myofibroblast proliferation
  • CD44 blockade–attenuated post-infarction remodeling, improved cardiac function
CD44 mediates cardiac remodeling by integrating inflammatory and fibrotic signals; its inhibition halts fibrosis progression
LiverHepatic congestion model (IVC ligation); CD44 and HA immunohistochemistry; CD44 neutralization (IM7)
  • CD44↑ and HA↑ in hepatic stellate cells
  • CD44 blockade–reduced collagen (Sirius red, hydroxyproline) and S100A4 expression
  • ALT and inflammatory cytokines unchanged–effect independent of hepatocyte injury
CD44 activates stellate cells and promotes fibrosis independently of inflammation; it represents a promising therapeutic target and biomarker
Table 9. Impact of environmental pollutants on the CD44–HA pathway in connective tissue (based on [210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229]). The table summarizes experimental and clinical evidence on how diverse environmental pollutants influence the CD44–hyaluronic acid (HA) axis in connective tissue remodeling. Heavy metals impair cell adhesion molecule expression on erythrocytes but enhance CD44 expression on immune cells, thereby promoting chronic inflammation and fibrosis. Particulate matter (PM2.5, silica dust) relies on CD44 for fibroblast activation and inflammatory cell recruitment, driving lung fibrosis, while endocrine-disrupting chemicals (EDCs) such as bisphenol A and phthalates induce CD44-dependent EMT, collagen deposition, and inflammatory infiltration. Microplastics further activate CD44 on macrophages and fibroblasts, supporting granuloma formation and fibrosis, and may act as carriers of toxic substances, amplifying tissue damage. Collectively, these findings underscore CD44–HA as a central mediator linking environmental exposure to chronic inflammation, fibrotic remodeling, and impaired tissue repair.
Table 9. Impact of environmental pollutants on the CD44–HA pathway in connective tissue (based on [210,211,212,213,214,215,216,217,218,219,220,221,222,223,224,225,226,227,228,229]). The table summarizes experimental and clinical evidence on how diverse environmental pollutants influence the CD44–hyaluronic acid (HA) axis in connective tissue remodeling. Heavy metals impair cell adhesion molecule expression on erythrocytes but enhance CD44 expression on immune cells, thereby promoting chronic inflammation and fibrosis. Particulate matter (PM2.5, silica dust) relies on CD44 for fibroblast activation and inflammatory cell recruitment, driving lung fibrosis, while endocrine-disrupting chemicals (EDCs) such as bisphenol A and phthalates induce CD44-dependent EMT, collagen deposition, and inflammatory infiltration. Microplastics further activate CD44 on macrophages and fibroblasts, supporting granuloma formation and fibrosis, and may act as carriers of toxic substances, amplifying tissue damage. Collectively, these findings underscore CD44–HA as a central mediator linking environmental exposure to chronic inflammation, fibrotic remodeling, and impaired tissue repair.
Type of PollutantImpact on CD44 and HAEffects in Connective TissueMolecular Mechanisms/Experimental Data
Heavy metals (lead, cadmium)• Decreased expression of CD44 and CD58 on erythrocytes (e.g., in children exposed to e-waste)
• Increased CD44 expression on macrophages and neutrophils at metal accumulation sites
• Impaired reparative and immune functions
• Chronic inflammation
• Enhanced organ fibrosis
• Lead reduces cell adhesion molecule expression (CD44/CD58), impairing ECM-cell interactions
• HA fragmentation acts as a DAMP, activating immune responses via CD44
Particulate matter (PM2.5, silica dust)• CD44 mediates the recruitment of inflammatory cells to the lungs in response to ozone
• CD44 blockade in silicosis models reduces collagen deposition
• Chronic lung inflammation
• Pulmonary fibrosis (e.g., silicosis, fibrosing alveolitis)
• Bronchial hyperreactivity
• CD44 loss protects against ozone-induced response (despite HA increase)
• CD44 is essential for fibroblast activation and inflammatory cell migration
• PM nanoparticles → oxidative stress, Th2 polarization, epigenetic activation of pro-fibrotic genes
Endocrine-disrupting chemicals (EDCs) (bisphenol A, phthalates, pesticides)• Induction of CD44 expression in epithelial cells undergoing EMT
• Increased CD44 on tissue macrophages (e.g., in adipose tissue)
• EMT and mesenchymal transition
• Chronic inflammation
• Fibrosis (e.g., in liver, lung, adipose tissue)
• BPA activates ERRγ → EMT in A549 cells
• Phthalates increase collagen deposition and leukocyte infiltration in the liver
• EDCs enhance HA and osteopontin production—both CD44 ligands
Microplastics• Induce CD44 expression on macrophages and fibroblasts surrounding particles
• Indirect CD44 activation via DAMPs and cytokines
• Granuloma formation around microplastics
• Chronic inflammation and fibrosis in lungs, liver, intestines (“plasticosis”)
• CD44- and HA-dependent migration and activation of myofibroblasts
• Microplastic particles act as toxin carriers (e.g., metals, PCBs), amplifying inflammatory response
• Mechanism
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Pedrycz-Wieczorska, A.; Chylińska-Wrzos, P.; Grzywacz, A.; Zieliński, E.; Bartosiński, A.; Kędziora-Kornatowska, K.; Lis-Sochocka, M.; Mertowska, P.; Mertowski, S.; Bojarski, K.; et al. CD44 as a Central Integrator of Inflammation and Fibrosis: From Molecular Signaling to Environmental Modulation. Int. J. Mol. Sci. 2025, 26, 8870. https://doi.org/10.3390/ijms26188870

AMA Style

Pedrycz-Wieczorska A, Chylińska-Wrzos P, Grzywacz A, Zieliński E, Bartosiński A, Kędziora-Kornatowska K, Lis-Sochocka M, Mertowska P, Mertowski S, Bojarski K, et al. CD44 as a Central Integrator of Inflammation and Fibrosis: From Molecular Signaling to Environmental Modulation. International Journal of Molecular Sciences. 2025; 26(18):8870. https://doi.org/10.3390/ijms26188870

Chicago/Turabian Style

Pedrycz-Wieczorska, Agnieszka, Patrycja Chylińska-Wrzos, Anna Grzywacz, Ewa Zieliński, Andrzej Bartosiński, Kornelia Kędziora-Kornatowska, Marta Lis-Sochocka, Paulina Mertowska, Sebastian Mertowski, Krzysztof Bojarski, and et al. 2025. "CD44 as a Central Integrator of Inflammation and Fibrosis: From Molecular Signaling to Environmental Modulation" International Journal of Molecular Sciences 26, no. 18: 8870. https://doi.org/10.3390/ijms26188870

APA Style

Pedrycz-Wieczorska, A., Chylińska-Wrzos, P., Grzywacz, A., Zieliński, E., Bartosiński, A., Kędziora-Kornatowska, K., Lis-Sochocka, M., Mertowska, P., Mertowski, S., Bojarski, K., Rahnama-Hezavah, M., Urbanowicz, T., & Grywalska, E. (2025). CD44 as a Central Integrator of Inflammation and Fibrosis: From Molecular Signaling to Environmental Modulation. International Journal of Molecular Sciences, 26(18), 8870. https://doi.org/10.3390/ijms26188870

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop