Home The plakin family: Potential therapeutic targets for digestive system tumors
Article Open Access

The plakin family: Potential therapeutic targets for digestive system tumors

  • Changwei Huang , Yixuan Chen , Manoop S. Bhutani , Caixia Wang , Yang Zhou , Jintao Guo ORCID logo EMAIL logo and Siyu Sun
Published/Copyright: July 30, 2025

Abstract

Digestive system tumors remain a global health challenge; however, the mechanisms underlying their tumorigenesis remain unclear. Identifying these mechanisms may facilitate early detection and more effective treatment. Members of the plakin family play crucial roles in cytoskeletal integrity and cell adhesion. Moreover, they regulate key cellular processes implicated in tumor development, including tumor cell migration, proliferation, and signaling. Therefore, exploring the potential roles of the plakin family members in digestive system tumors has attracted increasing attention. In this review, we provide a comprehensive examination of the biological characteristics of the plakin family members and an in-depth analysis of their clinicopathological significance and clinical implications in digestive system tumors. In summary, the plakin family is a translationally valuable diagnostic marker and a potential therapeutic target for digestive system tumors.

Introduction

The plakin family comprises seven large cytoskeletal crosslinkers.[1] Plakins enable the formation of junctional complexes by linking microfilaments (F-actin), microtubules (MTs), and intermediate filaments (IFs).[2] These cytoskeleton components influence cell polarity, adhesion, migration, and invasion,[3] making plakins crucial for integrating cytoskeletal processes.[4,5]

Members of the plakin family have emerged as drivers of various diseases.[6,7] Several studies have focused on their contribution to tumor pathology. However, despite extensive research on plakins in digestive system tumors, a comprehensive review is still lacking. Therefore, in this review, we explore the structures, loci, and functions of plakin family members, as well as their biological, diagnostic, and potential therapeutic roles in digestive system tumors.

Structure of plakin family members

The mammalian plakin family comprises the following seven members: bullous pemphig oid antigen1 (BPAG1), microtubule actin cross-linking factor 1 (MACF1), plectin, desmoplakin (DSP), envoplakin, periplakin, and epiplakin.[8] Most of these proteins exhibit isoform diversity. Consisting of different unique structural domains that interact with various cytoskeletal components and intercellular junctions, plakins exhibit similar connectivity functions. These structural domains include an actin-binding domain (ABD), a plakin domain, spectrin repeats, helix-loop-helix (EF-hand) motifs, a coiled-coil rod, a plakin repeat domain (PRD), a growth arrest specific 2 (GAS2)-related domain, and glycine-serine-arginine domain (Figure 1).[9,10]

Figure 1 Schematic representation of different members of the plakin family and the roles of selected plakins in cytoskeletal dynamics. ABD: actin-binding domain; BPAG1: bullous pemphigoid antigen 1; CCR: coiled-coil rod; CH: calponin homology; GAR: GAS2-related protein; GSR: glycine-serine-arginine; MACF1: microtubule actin cross-linking factor 1; PRD: Plakin repeat domain; DSP: desmoplakin. Plakin proteins crosslink the three main components of the cytoskeleton: intermediate filaments (IFs), microtubules (MTs), and microfilaments (F-actin). BPAG1e connects IFs to hemidesmosomes; desmoplakin connects IFs to desmosomes; plectin connects IFs to the nucleus; MACF1 binds microfilaments to microtubules.
Figure 1

Schematic representation of different members of the plakin family and the roles of selected plakins in cytoskeletal dynamics. ABD: actin-binding domain; BPAG1: bullous pemphigoid antigen 1; CCR: coiled-coil rod; CH: calponin homology; GAR: GAS2-related protein; GSR: glycine-serine-arginine; MACF1: microtubule actin cross-linking factor 1; PRD: Plakin repeat domain; DSP: desmoplakin. Plakin proteins crosslink the three main components of the cytoskeleton: intermediate filaments (IFs), microtubules (MTs), and microfilaments (F-actin). BPAG1e connects IFs to hemidesmosomes; desmoplakin connects IFs to desmosomes; plectin connects IFs to the nucleus; MACF1 binds microfilaments to microtubules.

The ABD, located at the N-terminus, comprises two calponin homology (CH) domains—namely CH1 and CH2—which can bind F-actin and enhance its affinity for actin.[3,11] The plakin domain, composed of α-helices, is present in all plakins except for epiplakin. Plakin domains allow plakins to form connections with various components, such as hemidesmosomes.[12] Spectrin repeats, composed of three α-helices, are only involved in the composition of BPAG1 and MACF1.[13] The PRD, found in various plakins, contains a β-folding and two reverse-helices, which interact with various IFs. Composed of ABD, coiled-coil rod, PRD, and plakin structural domains, plectin stabilizes the cytoskeleton through keratin remapping. Plectin mainly mediates the interaction among three cytoskeletal components, F-actin, MTs, and IFs. BPAG1 isoforms are widely expressed in the digestive system, skin, and reproductive system. Similar to BPAG1, MACF1 isoforms are widely expressed.[11] Although MACF1 and BPAG1 have multiple binding sites, they usually cross-link only one element of the cytoskeleton.[9] In contrast to the other members, DSP, envoplakin, periplakin, and epiplakin, lack the ABD structural domain. DSP, a key component of epidermal cell bridge grains, provides adhesion between cells. Envoplakin and Periplakin, which have similar structures and functions, are found predominantly in the keratinized and non-keratinized stratified squamous epithelia.[12] Interestingly, epiplakin has only one structural domain, the PRD structural domain. It is these unique structural features of the plakin family that determine its function. Plakin proteins regulate the cytoskeletal network, epithelial-mesenchymal transition (EMT), cell adhesion, migration, signal transduction, and other basic biological processes by connecting F-actin, MTs, and IFs.[14, 15, 16]

Pathophysiological role of plakin family members in digestive system tumors

Colorectal cancer (CRC)

CRC is one of the most common intestinal tumors.[17] Identifying new key molecules involved in colorectal tumorigenesis is essential for providing novel targets for tumor diagnosis and treatment (Figure 2).[18,19]

Figure 2 Role of plakins in cancers of the digestive system. Plakins are involved in CRC, PC, liver cancer, and other digestive system cancers. Plakins serve as tumor promoters (indicated in red), tumor suppressors (indicated in blue), or both suppressors and promoters (indicated in green). The major events in solid tumor development are tumor initiation, proliferation, and metastasis. Among these events, cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), and apoptosis are either promoted or inhibited. PC: pancreatic cancer; CRC: colorectal cancer.
Figure 2

Role of plakins in cancers of the digestive system. Plakins are involved in CRC, PC, liver cancer, and other digestive system cancers. Plakins serve as tumor promoters (indicated in red), tumor suppressors (indicated in blue), or both suppressors and promoters (indicated in green). The major events in solid tumor development are tumor initiation, proliferation, and metastasis. Among these events, cell proliferation, migration, invasion, epithelial-mesenchymal transition (EMT), and apoptosis are either promoted or inhibited. PC: pancreatic cancer; CRC: colorectal cancer.

Previous studies have shown that MACF1 and plectin are upregulated, whereas periplakin is downregulated in human CRC tissues, suggesting that plakins are potential diagnostic biomarkers for CRC. MACF1 is one of the mutationally discordant genes between 19 paired primary and metastatic CRC samples.[20] Compared to classical tumor markers, MACF1 may be a marker for predicting CRC metastasis. The role of plakin in connecting to the cytoskeleton is strongly associated with cell metastasis and migration. Plakin acts primarily as downstream molecules in these processes. For example, lipoprotein receptor-related protein 6 (LRP6) regulates microtubules (MT) assembly in CRC cells via MACF1, thereby influencing cytoskeletal remodeling, and its overexpression promotes CRC cell migration.[21] Additionally, plectin can function downstream of transcobalamin 1 (TCN1) in CRC, and its degradation compromises the stability of filamin A and F-actin networks, leading to cytoskeletal damage and CRC cell migration.[22] Furthermore, plectin influences podosomelike adhesion. Knockdown of plectin expression using small interfering ribonucleic acids (siRNAs) impairs the migration, invasion, and adhesion of SW480 cancer cells. Plectin-1k targets podosome-like adhesion and is involved in actin assembly and CRC cell invasion.[23] However, further investigations are required to determine whether it exerts its effects by influencing the cytoskeleton. Notably, MACF1 and plectin act as tumor promoters in CRC by affecting the cytoskeleton and intercellular interactions, thereby promoting cell migration. While periplakin, as a tumor suppressor in CRC, not only affects cell migration but also plays a role in cell proliferation and invasion and EMT.[24] In summary, these findings highlight the significant role of plakin family members in CRC tumorigenesis, suggesting their potential as diagnostic biomarkers and therapeutic targets. Understanding how plakins contribute to the dynamic interactions between the cytoskeleton and cellular signaling pathways may provide new avenues for targeted treatments and early detection strategies in CRC.

Pancreatic cancer (PC)

PC is one of the most aggressive and incurable malignancies.[25, 26, 27] Over the past decades, DSP and plectin have been considered potential diagnostic targets for PC. DSP, an epithelial tissue marker of PC, is upregulated during EMT, while epithelial tissue markers are downregulated during PC progression. However, previous studies have only established an association between DSP and PC, with the detailed mechanisms yet to be fully elucidated.[28,29] In contrast to DSP as a marker in EMT, plectin has been shown to play an active role in cell migration and growth. Babicky et al. were the first to demonstrate the role of plectin in PC cell migration. Exposed to the ligand macrophage-stimulating protein, recepteur d′origine nantais (RON) is transferred from the paranuclear cytoplasm to the cell surface. The RON receptor activation in PC cells leads to its binding with plectin and integrin beta 4, major components of hemidesmosomes that anchor cells to the extracellular matrix and inhibit cell migration.[30] These findings suggest that plectin negatively regulates cell motility. Studies on exosomes have demonstrated that plectin serves as an intracellular scaffolding protein under normal physiological conditions. However, plectin is upregulated in PC cells, and its abnormal upregulation results in its localization to secreted exosomes, which may promote PC growth.[31] Knocking down plectin using short hairpin RNAs (shRNAs) reduces exosomal plectin secretion, consequently inhibiting PC cell proliferation, migration, and invasion.[31] Further studies are needed to clarify their exact roles in disease progression and validate their potential for clinical use.

Liver cancer

Liver cancer is the seventh most common cancer and second most common cause of cancer-related deaths.[32] Hepatocellular carcinoma (HCC) is the most common type of liver cancer.[33] Members of the plakin family play a role in HCC cell migration. DSPs are primarily involved in EMT and serve as epithelial tissue markers. In contrast, DSP knockdown in Hep3B and HepG2 cells using siRNAs increased cell migration, not by triggering EMT but due to the absence of DSPs.[34] Controversial findings regarding the role of plectin in HCC cell migration have shown that plectin knockdown in human hepatocytes activates adherent spot kinase and ras-related C3 botulinum toxin substrate -Guanosine-Triphosphate hydrolase (Rac1-GTPase) to promote cell migration.[35] These findings align with those of HCC cytopathology studies showing low plectin expression in HCC cells.[36] Furthermore, invasion assays have demonstrated a positive correlation between low plectin levels and high single-cell migration capacity, although no significant differences were observed.[36] In contrast to previous findings, plectin expression and migration assays in four cell lines (MHCC97L, MHCC97H, human hepatocarcinoma cells [HCCLM3], and Human hepatocellular carcinomas [HPG2]) revealed that plectin was significantly upregulated in HCC tissues and promoted HCC cell migration, whereas plectin knockdown using shRNAs inhibited HCC cell migration. A potential mechanism is EMT inhibition via the extracellular signal-regulated kinase 1/2 (ERK1/2) signaling pathway.[37] Moreover, plectin is associated with individual cell migration and may be closely related to collective cell migration. It may interact with Netrin-1 to enhance overall HCC cell migration.[38] In HCC, increased extracellular matrix stiffness upregulates plectin expression, increasing F-actin polymerization to promote cell migration.[39] Although both DSP and plectin can affect cell migration, plectin has been shown to be potentially associated with the collective migration of cells. Furthermore, in liver cancer stem cells, neuronal cell adhesion molecule (NRCAM) mediates β-catenin signaling to activate EMT via MACF1.[40] Further investigations are warranted to explore its potential as a therapeutic target for inhibiting HCC cell migration and EMT.

Other digestive system tumors

The plakin family has also been investigated in other digestive system tumors, including gastric and esophageal cancers. Whole-exome sequencing (WES) of gastric cancer samples revealed that MACF1 mutations are moderately common in patients with peritoneal metastases.[41,42] A study assessing circulating tumor cell-free deoxyribonucleic acid (DNA) (cfDNA) from patients with gastric cancer found a higher MACF1 mutation in stage IV than in stages I-III, suggesting an association between MACF1 expression and gastric cancer metastasis. However, the underlying mechanisms remain unclear.

DSP promotes gastric cancer growth and metastasis. The establishment of the knockdown and overexpression models of DSP in gastric cancer have revealed that its overexpression inhibits gastric cancer cell proliferation and promotes apoptosis, whereas its knockdown showed the opposite effects. Moreover, Transwell assays revealed that DSP knockdown enhanced the invasion and migration of gastric cancer cells.[43]

Proteomic studies have revealed that periplakin is aberrantly expressed in esophageal cancer and may be closely associated with lymphatic metastasis.[44] Furthermore, periplakin overexpression has been shown to promote cell lamination, facilitate cell and extracellular matrix adhesion, and retard cell migration, consistent with the trend of low periplakin expression observed in esophageal cancer.[45]

Both DSP and periplakin play active roles in different digestive tumors,[24,33,43,45] although their mechanisms of action differ. A deeper understanding of cytoskeletal junction proteins is required to explore how they affect cellular mechanotransduction and the tumor microenvironment, thereby influencing different digestive tumors.[46,47]

Shared and member-specific effects

In various digestive system tumor cells, plakin family members are closely associated with cell migration, despite their up- and down-regulation. The specific mechanisms of action vary. However, they are associated with altered cytoskeletal networks and intercellular connectivity. Additionally, they affect signaling pathways, including the wnt/β-catenin signaling pathway (Figure 3).[21,22,30] Additionally, the expression of most of the members can be regulated by RNA and further affect migration.[23,31]

Figure 3 Schematic representation of how the plakin family affects cell migration and EMT of digestive tumor cells. The plakin family is involved in the cytoskeleton and intercellular interconnections and affects tumor cell migration and EMT through signal transduction pathways. The activation and inhibition of different proteins are indicated by solid arrows and solid double lines, respectively. Dashed arrows indicate the translocation of proteins. P: phosphorylation; β: β-catenin; DSP: Desmoplakin.
Figure 3

Schematic representation of how the plakin family affects cell migration and EMT of digestive tumor cells. The plakin family is involved in the cytoskeleton and intercellular interconnections and affects tumor cell migration and EMT through signal transduction pathways. The activation and inhibition of different proteins are indicated by solid arrows and solid double lines, respectively. Dashed arrows indicate the translocation of proteins. P: phosphorylation; β: β-catenin; DSP: Desmoplakin.

Current research suggests that plectin promotes cell migration in HCC, PC and CRC. Interestingly, the promotion of migration by plectin in HCC may be related to the collective migration of cells.[38] Compared to other members of the plakin family, plectin plays a dual role in digestive system tumors, exerting both pro- and anticancer effects. Similarly, integrin α6β4 can play a dual role in tumors, recruiting plectin to the plasma membrane. We speculate whether the dual action of plectin stems from the mislocalization of pletin recruited to the plasma membrane by integrin α6β4. That warrants further experimental verification.[48] In gastric and liver cancers, DSP affects cell migration; however, in PC, current studies only show that it acts as an epithelial marker for EMT.[34,43] Further investigation is needed to determine the effect of DSP on PC cell migration. Envoplakin and Periplakin possess similar structures and functions. Currently, it has been shown that periplakin affects cell migration, cell proliferation, and EMT in esophageal and CRC.[24,44] Understanding the function of envoplakin, particularly its impact on cell migration, is crucial. Although similar structures exist between plakin families, between individual digestive system tumor cells. However, their structures are not identical, which may potentially explain the differences in function.

Potential clinical applications of plakin family members in digestive system tumors

Digestive system tumors remain a significant cause of tumor-related deaths worldwide, highlighting the importance of identifying biomarkers for early detection and translating the findings into clinical practice.[49,50] Recent studies have identified members of the plakin family as potential biomarkers for the diagnosis and prognosis of digestive system tumors (Table 1).[51-53]

Table 1

Potential clinical applications of plakin family members as biomarkers for digestive system tumors

Cancer type Study object Study subjects and control sample size Test samples Test method (s) Potential clinical application Ref.
PDAC PDAC, chronic pancreatitis, and normal pancreata samples 4 normal pancreata, 15 chronic pancreatitis, 14 PanIN I, 26 PanIN II, 15 PanIN III, 41 PDAC, 8 liver metastases, 11 lymph node metastasis, 10 matching primary tumors, and 9 peritoneal metastasis samples PDAC, pancreatitis, and normal pancreata samples IHC and western blot analysis Diagnostic, prognostic, and metastatic biomarkers [53]
PDAC PANC-1, MIA PaCa-2, HPAC, Mpanc-96, and BXPC-3 cells In vivo bioluminescence imaging, luciferase assays, and immunohistochemistry AAV-plectin 1-targeting peptides preferentially targeted PDAC cell lines. [68]
PDAC Panc-1 and LO2 cells MRI and in vitro laser- scanning confocal microscopy Plectin-targeting iron oxide nanoparticles used as imaging-contrast agents for early PDAC diagnosis [62]
PDAC MIA Paca-2 cells Cellular fluorescence-based microscopy images Plectin-targeting peptide attached to magnetofluorescent nanoparticles helped to detect PDAC cells. [60]
PDAC PANC-1 and MIA-Paca2 cells Multiphoton microscope Plectin-targeted lipid microbubbles help detect PDAC cells. [65]
PDAC Pancreatic mass in patients 85 patients with pancreatic masses Pancreatic mass samples were obtained by EUS-FNA. Cytology, KRAS mutations, plectin staining, and final diagnosis Plectin-1 staining can help improve the diagnostic accuracy of EUS-FNA. [57]
PDAC ASPC-1 and PANC-1 cells In vitro cell-proliferation assay, in vivo tumor-growth inhibition, and hematoxylin and eosin staining Plectin-1-targeted multifaceted peptide-assisted one-pot synthesis of gold nanoparticles can help gemcitabine delivery. [71]
PDAC MIA-Paca-2 and XPA-1 cells. MIN6 Mice MRI, in vivo optical imaging, and hematoxylin and eosin staining Plectin-1-targeted fluorescence and MR dual-functional nanoparticles can be used to visualize PDAC. [61]
PDAC PANC 1 and hTERT-HPNE cells Multiphoton imaging Plectin-targeted lipid microbubbles and multiphoton imaging can help detect PDAC. [63]
PDAC ASPC-1, Capan2 L929, PANC-1, T3M4, BXPC3 HPDE6-C7, SW1990, HUVEC cells, and MiceL929 In vivo confocal fluorescence laser microscopy of bipeptides, in vivo optical and MRI of Gd-Cy7-PTP/RGD, and optical imaging-guided surgery of Gd-Cy7-PTP/RGD Plectin/integrin-targeted bispecific molecular probe for MRI/NIR imaging of PDAC [64]
PDAC Panc-1 cells Cell viability assays Plectin-targeted chaperonin-GroEL can promote PDAC cell apoptosis. [70]
PDAC CPI-613 and LY2109761 In vivo antitumor activity detected with Picro Sirius Red staining and IHC staining of α-SMA Plectin-targeted, tumor- responsive nanopolyplex targeted PDAC cells and stroma. [69]
PDAC Patients with PC 32 patients with PC CTCs CTC platform and a C-microfabricated porous filter Plectin can help identify CTCs during early-stage PDAC. [58]
CRC Colorectal adenocarcinoma and tubular adenoma 25 patients with colorectal adenocarcinoma and 10 patients with tubular adenoma Cancer samples IHC Plectin may serve as an oncofetal biomarker. [49]
CRC Patients with CRC, healthy control subjects, and fetuses 30 cancer samples, 30 control samples, and 30 fetus samples Serum samples 2D DIGE coupled with a Finnigan LTQ-based proteomics approach. GC-MS instrument integrated with a commercial mass spectrometry library MACF1 may serve as an oncofetal biomarker. [50]
CRC CT 26 colon tumor model Subcutaneous implanted tumor model Plecstatin can target plectin and inhibit colorectal tumors. [51]
CRC HCT116, HT-19, and HCT-115 cells Colony-formation assay, flow cytometry analysis of apoptosis, and spheroid growth assays Plectin-1 treatment reduced spheroid growth and decreased the colony-forming ability of colon adenocarcinoma cells. [52]
Esophageal cancer Primary esophageal squamous cell carcinoma samples and non- tumor samples 12 tumor samples and 12 non-tumor samples Tumor cell proteins and non-tumor cell proteins 2D DIGE, immunoblotting, and IHC Periplakin is a potential marker for detecting early esophageal cancer and evaluating tumor progression. [41]
Esophageal cancer Tumor cells and adjacent normal mucosal cells 72 paired tumor cell and mucosal cell samples Cancer cells protein 2D-PAGE, MS-based protein identification, and western blot analysis Periplakin may be used to diagnose esophageal cancer. [75]
Gastric cancer Gastric carcinoma samples from patients 74 samples, including intestinal, diffuse, mixed, and indeterminate adenocarcinomas Cancer tissue sample WES MACF1 may serve as a marker of metastasis in gastric cancer. [38]
Gastric cancer Stage IV gastric cancer samples 56 stage IV gastric cancer samples Cancer tissue sample Next-generation sequencing MACF1 expression may serve as a potential biomarker for stage IV gastric cancer. [73]
Liver cancer Human plasma and liver tissues from patients with liver cancer 3 patient samples Plasma and cancer tissues Targeted phosphopeptide analysis and immunoblotting Plectin may serve as a potential phosphobiomarker in liver cancer. [74]
  1. 2D DIGE: two-dimensional difference gel electrophoresis; 2D-PAGE: two-dimensional polyacrylamide gel electrophoresis; AAV: adenoassociated virus; CTC: circulating tumor cell; EUS-FNA: endoscopic ultrasound-guided fine-needle aspiration; GC: gas chromatography; IHC: immunohistochemistry; LTQ: linear ion trap mass spectrometer; MRI, magnetic resonance imaging; MS: mass spectrometry; PanIN: pancreatic intraepithelial neoplasia; PDAC: pancreatic ductal adenocarcinoma; WES: whole-exome sequencing; NIR: near-infrared; α-SMA: α-smooth muscle actin; MS-based protein: mass spectrometry-based protein; Ref.: references.

CRC

Immunohistochemical analyses of colorectal and tubular adenomas revealed that plectin expression was substantially higher in tumor cells than in normal colorectal mucosal cells, indicating its potential as an early diagnostic biomarker for early-stage CRC and precancerous lesions.[54] Additionally, phosphorylation of LRP6, which functions upstream of MACF1,[55] was strongly associated with the tumor-node-metastasis stage, Dukes stage, and poor prognosis, suggesting that MACF1 is related to the prognosis of CRC.[21] Interestingly, a clinical study evaluating programmed death-ligand 1 (PD-L1) inhibitors combined with radiation for treating advanced colorectal cancer demonstrated that MACF1 expression was upregulated in treatment-responsive patients, suggesting that MACF1 serves as a potential marker for immunotherapy response.[56] Nevertheless, more clinical studies are required to validate these findings.

Plecstatin-1 is an organometallic chemotherapeutic agent targeting plectin. It inhibits tumorsphere growth and induces changes in its morphology and structure by targeting plectin to affect the cytoskeleton. It also blocks tumor growth and induces G0/G1 cell cycle arrest in colon cancer cell lines, which reduces mitochondrial membrane potential, reactive oxygen species levels, and tumor cell proliferation. In a mouse model of colorectal cancer, it not only showed good tolerance but also resulted in a significant reduction in tumor volume.[57,58] However, the translational applicability of these treatment modalities requires further investigation.

PC

Immunohistochemistry of normal pancreatic, chronic pancreatitis, and PC tissues indicates that normal pancreatic and pancreatic tissues are negative for plectin, whereas PC tissues are 100% positive. Additionally, plectin expression increases during PC development and can be used to distinguish early pancreatic intraepithelial neoplasia (PanIN) I and II lesions from PanIN III and pancreatic ductal adenocarcinoma.[59] Plectin is closely related to tumor staging and metastasis, making it a strong prognostic marker.[59] Plectin is more specific for invasive and preinvasive PC than classical markers of PC, including carbohydrate antigen199 (CA199) and carcinoembryonic antigen (CEA).[60, 61, 62] A study analyzing 85 patients with pancreatic masses sampled using endoscopic ultrasound-guided fine-needle (EUS-FNA) aspiration, an essential method for early diagnosis of PC, showed that the sensitivity, specificity, and accuracy of the samples for histological diagnosis were 81%, 80%, and 79%, respectively.[63] Kirsten ratsarcoma viral oncogene homolog (KRAS) mutation assessment, in combination with histological examinations, showed sensitivity, specificity, and accuracy of 93%, 87%, and 92%, respectively. Furthermore, the combination of histological assessments, KRAS mutation analysis, and plectin staining increased the diagnostic sensitivity, specificity, and accuracy to 96%, 93%, and 95%, respectively. These findings indicate that plectin staining can improve the diagnostic ability of endoscopic ultrasound-guided fine-needle aspiration (EUS-FNA)-acquired pancreatic tumor samples.[63] However, the diagnostic efficacy of combining plectin with P53, another common mutation site in PC, remains unclear. Researchers have identified plectin as a biomarker for circulating tumor cells in portal and peripheral blood samples and found plectin-positive circulating tumor cells in 43.8% and 50% of portal and peripheral blood samples, respectively. However, no plectin-positive circulating tumor cells were detected in samples from healthy individuals.[64] Nevertheless, its comparison with immunofluorescence-fluorescence in situ hybridization remains to be explored.[65] These findings suggest that plectin is a reliable marker for the diagnosis of PC; however, large multicenter randomized controlled studies are required to confirm these findings.

Plectin can be used as a targeting agent for PC. A study using a mouse model of PC revealed that a plectintargeted peptide conjugated to magnetofluorescent nanoparticles enabled specific detection of PC cells in normal pancreatic tissue using confocal microscopy with live tissues. However, further studies are required to elucidate the clinical implications of these findings.[66] Additionally, plectin-targeting peptides and plectin antibodies can enhance the accumulation of nanoparticles, microbubbles, 1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid-based fluorescent probes, and imaging probes in pancreatic tumor cells, facilitating PC detection.[67, 68, 69, 70, 71] Magnetic resonance imaging and confocal microscopy used to detect plectin antibody-conjugated, targeting iron oxide nanoparticles in PC and normal pancreatic cell lines revealed high plectin expression in PC cell lines but not in normal pancreatic cells.[67,68] Previous in vitro studies have shown that lipid microbubbles targeting plectin bind specifically to PC cell lines, enabling rapid detection of the absence of PC cells on the surfaces of cut tissues after PC surgery.[68,71] Furthermore, a bimolecular probe based on plectin and integrin showed good diagnostic results for PC tissues both in vitro and in vivo.[64] Moreover, a novel DOTA-based plectin-targeted molecular probe has also shown promise for the diagnosis of PC.[72] A study targeting plectin in three patients with pancreatic ductal adenocarcinoma reported no adverse events, demonstrating its safety.[73] Further studies are needed to validate their clinical performance and safety.

Plectin is also a potential target for drug delivery. Researchers have developed various delivery routes, including polymeric nanoparticles, gold nanoparticles, targeted adeno-associated virus particles, and natural-protein drug-delivery systems that can target cancer-specific plectin and promote targeted delivery to PC cells.[74, 75, 76, 77, 78] Despite in vivo and in vitro validation of these delivery modalities, these modes of administration are yet to be implemented clinically, warranting further investigations to determine their therapeutic efficacy and safety.

Other digestive system tumors

Proteomic analyses have shown aberrant expression of periplakin in esophageal cancer.[44] Immunohistochemical staining studies have demonstrated that periplakin is predominantly localized at the cell border in normal esophageal tissues. However, in atypical hyperplastic tissues, it shifts to the cytoplasm during early-stage cancers, where it is barely expressed. This suggests that periplakin may be useful as a biomarker for diagnosing early esophageal cancer.[79] Studies have also indicated a strong association between MACF1 with gastric cancer stage and metastasis, suggesting it as a potential prognostic biomarker. In a study, WES analysis of 74 gastric cancer samples revealed a high rate of MACF1 mutations in patients with gastric cancer who had peritoneal metastases.[41] Additionally, next-generation sequencing of cfDNA from 56 patients with stage IV gastric cancer showed that MACF1 was the most frequently mutated gene in patients with metastases and cfDNA.[80] Furthermore, phosphorylation proteomics analyses revealed plectin as a potential phosphate biomarker for HCC.[81] Immunohistochemical staining of 18 HCC and normal tissue samples showed that plectin was significantly downregulated in HCC samples,[82] suggesting its potential as an HCC diagnostic marker. Overall, these studies indicate that the plakin family holds promise as potential biomarkers for various digestive system tumors. Nevertheless, further clinical studies are necessary to validate their diagnostic performance and clinical applicability. Additionally, plecstatin-1 (plectin inhibitor) was well tolerated and effective in inhibiting HCC progression in a mouse HCC model.[83]

Shared clinical applications and member-specific potential

Immunohistochemistry, WES, and proteomics studies using clinical samples have demonstrated that members of the plakin family can be used as potential diagnostic markers for digestive system tumors.[41,44,59,79] Immunohistochemistry results have shown that plectin has better diagnostic results for PC because of higher sensitivity and specificity. However, limited research exists on prognostic markers, with relevant studies currently focusing on the effects of pletin on PC and MACF1 on CRC.[54,59] Current translational research on plectin as a marker for PC is more advanced, exploring not only the potential advantages of plectin over traditional markers but also its use as a targeting agent to improve diagnostic yield in combination with advanced endoscopic techniques such as endoscopic ultrasound (EUS).[60,63] In contrast, the role of other plakin family members for other digestive tumors (especially early tumors) should be further explored. Current research on treatment focuses on the targeted delivery of plectin for PC and the use of plectin-targeting agents for CRC and HCC.[57,74] Targeting agents against other family members of plakin, along with drug development, is highly desirable. Additionally, whether the plakin family is involved in tumor drug resistance requires further investigation. It is noteworthy that most translational studies have focused on plectin compared to other family members. This may be related to its abnormal localization to the cell membrane.

Conclusions

The plakin protein family plays a role in cytoskeletal dynamics, cell migration, proliferation, and other biological processes. Abnormal plakin expression is strongly associated with the development of digestive system tumors. Although high-quality clinical studies are required to confirm this finding, plakins present potential diagnostic and therapeutic targets for these cancers.

Due to similar structural domains, the plakin family can play a role in cytoskeletal connections, cellular interconnections, and signal transduction pathways in digestive tumor cells. In other cells, plakin family members were shown to be closely related to the Wnt-β-catenin and phosphatidylinositol 3-kinase-protein kinase B pathways (Figure 4). A deeper understanding of these mechanisms will help us further explore how the plakin family functions in digestive system tumor cells.[43,84, 85, 86, 87, 88, 89, 90, 91, 92, 93]

Figure 4 Schematic diagram showing how members of the plakin family affect the phosphatidylinositol 3-kinase (PI3K) -protein kinase B (AKT) and Wnt signal-transduction pathways. Several members of the plakin family can act on specific links in this pathway, thereby affecting the activity of the pathway. The activation and inhibition of different proteins are indicated by solid arrows and solid double lines, respectively. Dashed arrows indicate the translocation of proteins. α: α-catenin; β: β-catenin; γ: γ-catenin; p120: p120-catein; PKP: plakophilin; src: Src-family tyrosine kinases.
Figure 4

Schematic diagram showing how members of the plakin family affect the phosphatidylinositol 3-kinase (PI3K) -protein kinase B (AKT) and Wnt signal-transduction pathways. Several members of the plakin family can act on specific links in this pathway, thereby affecting the activity of the pathway. The activation and inhibition of different proteins are indicated by solid arrows and solid double lines, respectively. Dashed arrows indicate the translocation of proteins. α: α-catenin; β: β-catenin; γ: γ-catenin; p120: p120-catein; PKP: plakophilin; src: Src-family tyrosine kinases.

Individual plakin family members exhibit varying effects on tumor progression. Some proteins promote tumorigenesis, while others have opposing roles.[32,34] This discrepancy may result from the structural differences in various tumors. Additionally, the distinct cytoskeletal junction proteins connecting different components could be a contributing factor. For example, in contrast to the other members, Epiplakin consists only of plakin repeat domains (PRDs) that bind only to intermediate filament (IF). This may affect tumor cells differently. Nevertheless, further studies employing advanced technologies, such as omics, gene knockout, and knock-in models, may help elucidate these underlying reasons.[94,95] Moreover, while most studies have focused on tumor cells, exploring interactions between plakin family proteins and stromal or immune cells facilitated by single-cell sequencing and immune microenvironment research[96,97] will provide additional insights.[98] Additionally, factors such as aging and circadian rhythms have been shown to be closely associated with digestive tumors, making it urgent to explore the role played by the plakin family.[99,100]

Current research suggests that the plakin family members hold promise as diagnostic and prognostic biomarkers for digestive system tumors.[101] Particularly, their diagnostic performance offers unique advantages compared to other classical biomarkers. However, large-scale cohort studies are essential for validating these findings before clinical application. Currently, plectin is being used as a targeted contrast agent for PC and although various plectin-targeting molecules have shown promise in preclinical single-center studies, more multicenter trials are needed to compare their effectiveness. Based on these studies, expanding the use of plakin family members as targeted contrast agents for other digestive system tumors is highly desirable. Additionally, combining the plakin family with clinical techniques such as liquid biopsy and needle-based confocal micro-endoscopy could improve the early diagnosis of digestive system tumors.[102,103] However, multi-center prospective studies are needed to determine the most accurate diagnostic combination. Although prognostic studies have shown that the expression of plakin family members correlates with the stage of digestive tumors, no cohort studies have demonstrated an association with survival outcomes in patients with digestive tumors. Interestingly, plectin demonstrates strong efficacy as a diagnostic target and in targeted drug delivery for PC compared to other plakin family members for other digestive tumors. This outcome may be because plectin can exist as an exosome that is more readily bound between cells.[32] Furthermore, the development and combination of plakin family inhibitors with other anticancer drugs may provide effective antitumor therapies. Except for plectin, no clinical studies currently focus on inhibitors targeting the plakin family.[104,105] Some researchers have developed an agonist, I-3, which targets periplakin and has shown better results in treating vitiligo in in vivo animal tests.[106] A monoclonal antibody manifestation targeting plectin mislocalized on the cell surface of ovarian cancer cells showed promising results in in vivo and in vitro assays.[107] While this holds promise, challenges remain in developing agonists and inhibitors of plakin family members in digestive tumors. No studies have shown the presence of enzyme- and receptor-binding sites for the plakin protein family in digestive system tumors, and this may lead to difficulties in developing agonists and inhibitors due to the lack of binding sites. Except for plectin, most cytoskeletal junction proteins are usually present in the cytoplasm. These proteins are developed for interaction with agonists and inhibitors, which are required to have good lipid solubility and suitable molecular weight to pass through the cell membrane. Additionally, because of their widespread role in the cytoskeleton, their agonists and inhibitors may disrupt the cytoskeleton of normal cells, potentially resulting in toxicity and adverse effects.

Translational research exploring the plakin family in digestive system tumors is imminent. A summary of current clinical studies (Clinicaltrials, Trial search.who. int) is warranted. Currently, there are phase I/II clinical trials of plectin-targeting drugs in solid tumors, including PC (NCT05074472). More interestingly, there are also ongoing studies exploring the diagnostic role of plectin in cholangiocarcinoma obtained by ERCP (Endoscopic Retrograde Cholangiopancreatography) (NCT06651346). EUS and ERCP are promising diagnostic and therapeutic techniques for the biliopancreatic system. Given the potential of the plakin family, especially plectin in PC, more future clinical studies in this direction are warranted.

As research into the mysteries of the role of the members of plakin family in digestive system tumors progresses, a more comprehensive understanding of their function will help pave the way for novel diagnostic and therapeutic strategies for future targeted interventions.


Jintao Guo, Department of Gastroenterology, Shengjing Hospital of China Medical University, No. 36, Sanhao Street, Heping District, Shenyang 110004, Liaoning Province, China.
#

These authors contributed equally to the work.


Funding statement: This study was funded by Liaoning Province Applied Basic Research Program Joint Program Project (2022JH2/101500076); University Innovation Team and Innovative Talent Support Program of Liaoning Province (Grant No. LR2019073); Shenyang Young and Middle-aged Science and Technology Innovation Talent Support Program (Grant No. RC200438).

Acknowledgements

We thank all other doctors who provided helpful support for this review.

  1. Author Contributions

    Huang CW: Conceptualization, Writing—Original draft. Chen YX: Writing—Original draft. Wang CX: Resources. Zhou Y: Resources. Sun SY: Writing—Review and Editing. Guo JT: Writing—Review and Editing. All authors have approved the final version of the manuscript.

  2. Ethical Approval

    Not applicable.

  3. Informed Consent

    Not applicable.

  4. Conflict of Interest

    Siyu Sun is an Associate Editor-in-Chief of the journal. This article was subjected to the standard procedures of peer review process independent of the editor and his research group.

  5. Use of Large Language Models, AI and Machine Learning Tools

    None declared.

  6. Data Availability Statement

    Not applicable.

References

1 Leung CL, Green KJ, Liem RK. Plakins: a family of versatile cytolinker proteins. Trends Cell Biol 2002;12:37–45.10.1016/S0962-8924(01)02180-8Search in Google Scholar

2 Sonnenberg A, Liem RK. Plakins in development and disease. Exp Cell Res 2007;313:2189–2203.10.1016/j.yexcr.2007.03.039Search in Google Scholar PubMed

3 Jefferson JJ, Leung CL, Liem RK. Plakins: goliaths that link cell junctions and the cytoskeleton. Nat Rev Mol Cell Biol 2004;5:542–553.10.1038/nrm1425Search in Google Scholar PubMed

4 Fife CM, McCarroll JA, Kavallaris M. Movers and shakers: cell cytoskeleton in cancer metastasis. Br J Pharmacol 2014;171:5507–5523.10.1111/bph.12704Search in Google Scholar PubMed PubMed Central

5 Wang Z, Wang W, Luo Q, Song G. Plectin: Dual Participation in Tumor Progression. Biomolecules 2024;14:1050.10.3390/biom14091050Search in Google Scholar PubMed PubMed Central

6 Bouameur JE, Favre B, Borradori L. Plakins, a versatile family of cytolinkers: roles in skin integrity and in human diseases. J Invest Dermatol 2014;134:885–894.10.1038/jid.2013.498Search in Google Scholar PubMed

7 Anderson HJ, Huang S, Lee JB. Paraneoplastic pemphigus/paraneoplastic autoimmune multiorgan syndrome: Part I. Clinical overview and pathophysiology. J Am Acad Dermatol 2024;91:1–10.10.1016/j.jaad.2023.08.020Search in Google Scholar PubMed

8 Hu L, Huang Z, Wu Z, Ali A, Qian A. Mammalian Plakins, Giant Cytolinkers: Versatile Biological Functions and Roles in Cancer. Int J Mol Sci 2018;19:974.10.3390/ijms19040974Search in Google Scholar PubMed PubMed Central

9 Yan Y, Winograd E, Viel A, Cronin T, Harrison SC, Branton D. Crystal structure of the repetitive segments of spectrin. Science 1993;262:2027–2030.10.1126/science.8266097Search in Google Scholar PubMed

10 Getsios S, Huen AC, Green KJ. Working out the strength and flexibility of desmosomes. Nat Rev Mol Cell Biol 2004;5:271-281.10.1038/nrm1356Search in Google Scholar PubMed

11 Najor NA. Desmosomes in Human Disease. Annu Rev Pathol 2018;13:51–70.10.1146/annurev-pathol-020117-044030Search in Google Scholar PubMed

12 Wenta T, Schmidt A, Zhang Q, Devarajan R, Singh P, Yang X, et al. Disassembly of α6β4-mediated hemidesmosomal adhesions promotes tumorigenesis in PTEN-negative prostate cancer by targeting plectin to focal adhesions. Oncogene 2022;41:3804–3820.10.1038/s41388-022-02389-5Search in Google Scholar PubMed PubMed Central

13 Chen HJ, Lin CM, Lin CS, Perez-Olle R, Leung CL, Liem RK. The role of microtubule actin cross-linking factor 1 (MACF1) in the Wnt signaling pathway. Genes Dev 2006;20:1933–1945.10.1101/gad.1411206Search in Google Scholar PubMed PubMed Central

14 Žugec M, Furlani B, Castañon MJ, Rituper B, Fischer I, Broggi G, et al. Plectin plays a role in the migration and volume regulation of astrocytes: a potential biomarker of glioblastoma. J Biomed Sci 2024;31:14.10.1186/s12929-024-01002-zSearch in Google Scholar PubMed PubMed Central

15 Wesley T, Berzins S, Kannourakis G, Ahmed N. The attributes of plakins in cancer and disease: perspectives on ovarian cancer progression, chemoresistance and recurrence. Cell Commun Signal 2021;19:55.10.1186/s12964-021-00726-xSearch in Google Scholar PubMed PubMed Central

16 Nanavati BN, Noordstra I, Lwin AKO, Brooks JW, Rae J, Parton RG, et al. The desmosome-intermediate filament system facilitates mechanotransduction at adherens junctions for epithelial homeostasis. Curr Biol 2024;34:4081–4090.10.1016/j.cub.2024.07.074Search in Google Scholar PubMed PubMed Central

17 Dekker E, Tanis PJ, Vleugels JLA, Kasi PM, Wallace MB. Colorectal cancer. Lancet 2019;394:1467-1480.10.1016/S0140-6736(19)32319-0Search in Google Scholar PubMed

18 Wang S, Cheng G, Hu D. A rare but interesting case of small intestinal tumor diagnosed by transrectal EUS-FNA (with video). Endosc Ultrasound 2024;13:269–270.10.1097/eus.0000000000000074Search in Google Scholar PubMed PubMed Central

19 Biller LH, Schrag D. Diagnosis and Treatment of Metastatic Colorectal Cancer: A Review. JAMA 2021;325:669–685.10.1001/jama.2021.0106Search in Google Scholar PubMed

20 Kim R, Schell MJ, Teer JK, Greenawalt DM, Yang M, Yeatman TJ. Co-evolution of somatic variation in primary and metastatic colorectal cancer may expand biopsy indications in the molecular era. PLoS One 2015;10:e0126670.10.1371/journal.pone.0126670Search in Google Scholar PubMed PubMed Central

21 Yao Q, An Y, Hou W, Cao YN, Yao MF, Ma NN, et al. LRP6 promotes invasion and metastasis of colorectal cancer through cytoskeleton dynamics. Oncotarget 2017;8:109632–109645.10.18632/oncotarget.22759Search in Google Scholar PubMed PubMed Central

22 Zhu X, Jiang X, Zhang Q, Huang H, Shi X, Hou D, et al. TCN1 Deficiency Inhibits the Malignancy of Colorectal Cancer Cells by Regulating the ITGB4 Pathway. Gut Liver 2023;17:412–429.10.5009/gnl210494Search in Google Scholar PubMed PubMed Central

23 McInroy L, Määttä A. Plectin regulates invasiveness of SW480 colon carcinoma cells and is targeted to podosome-like adhesions in an isoform-specific manner. Exp Cell Res 2011;317:2468–2478.10.1016/j.yexcr.2011.07.013Search in Google Scholar PubMed

24 Li X, Zhang G, Wang Y, Elgehama A, Sun Y, Li L, et al. Loss of periplakin expression is associated with the tumorigenesis of colorectal carcinoma. Biomed Pharmacother 2017;87:366–374.10.1016/j.biopha.2016.12.103Search in Google Scholar PubMed

25 Möller K, Batali A, Jenssen C, Braden B, Hocke M, On W, et al. Comments and illustrations of the European Federation of Societies for Ultrasound in Medicine contrast-enhanced ultrasound guidelines: Multiparametric imaging and EUS-guided sampling in rare pancreatic tumors. Benign mesenchymal pancreatic tumors. Endosc Ultrasound 2024;13:218–231.10.1097/eus.0000000000000070Search in Google Scholar PubMed PubMed Central

26 Buchberg J, de Stricker K, Pfeiffer P, Mortensen MB, Detlefsen S. Mutational profiling of 103 unresectable pancreatic ductal adenocarcinomas using EUS-guided fine-needle biopsy. Endosc Ultrasound 2024;13:154–164.10.1097/eus.0000000000000072Search in Google Scholar PubMed PubMed Central

27 Mulqui MV, Caillol F, Ratone JP, Hoibian S, Dahel Y, Meunier É, et al. Detective flow imaging versus contrast-enhanced EUS in solid pancreatic lesions. Endosc Ultrasound 2024;13:248–252.10.1097/eus.0000000000000076Search in Google Scholar PubMed PubMed Central

28 Kong B, Michalski CW, Hong X, Valkovskaya N, Rieder S, Abiatari I, et al. AZGP1 is a tumor suppressor in pancreatic cancer inducing mesenchymal-to-epithelial transdifferentiation by inhibiting TGF-β-mediated ERK signaling. Oncogene 2010;29:5146–5158.10.1038/onc.2010.258Search in Google Scholar PubMed

29 Tian L, Lu ZP, Cai BB, Zhao LT, Qian D, Xu QC, et al. Activation of pancreatic stellate cells involves an EMT-like process. Int J Oncol 2016;48:783–792.10.3892/ijo.2015.3282Search in Google Scholar PubMed

30 Yu PT, Babicky M, Jaquish D, French R, Marayuma K, Mose E, et al. The RON-receptor regulates pancreatic cancer cell migration through phosphorylation-dependent breakdown of the hemidesmosome. Int J Cancer 2012;131:1744–1754.10.1002/ijc.27447Search in Google Scholar PubMed PubMed Central

31 Shin SJ, Smith JA, Rezniczek GA, Pan S, Chen R, Brentnall TA, et al. Unexpected gain of function for the scaffolding protein plectin due to mislocalization in pancreatic cancer. Proc Natl Acad Sci U S A 2013;110:19414–19419.10.1073/pnas.1309720110Search in Google Scholar PubMed PubMed Central

32 Möller K, Safai Zadeh E, Görg C, Dong Y, Cui X, Lim A, et al. Focal Liver Lesions other than Hepatocellular Carcinoma in Cirrhosis: Diagnostic Challenges. J Transl Int Med 2023;10:308–327.10.2478/jtim-2022-0068Search in Google Scholar PubMed PubMed Central

33 Wang B, Hao X, Yan J, Li X, Zhao M, Han T. A bibliometric analysis of immune-related adverse events in cancer patients and a meta-analysis of immune-related adverse events in patients with hepatocellular carcinoma. J Transl Int Med 2024;12:225–243.Search in Google Scholar

34 Nath A, Oak A, Chen KY, Li I, Splichal RC, Portis J, et al. Palmitate-Induced IRE1-XBP1-ZEB Signaling Represses Desmoplakin Expression and Promotes Cancer Cell Migration. Mol Cancer Res 2021;19:240–248.10.1158/1541-7786.MCR-19-0480Search in Google Scholar PubMed PubMed Central

35 Cheng CC, Lai YC, Lai YS, Hsu YH, Chao WT, Sia KC, et al. Transient knockdown-mediated deficiency in plectin alters hepatocellular motility in association with activated FAK and Rac1-GTPase. Cancer Cell Int 2015;15:29.10.1186/s12935-015-0177-1Search in Google Scholar PubMed PubMed Central

36 Cheng CC, Chao WT, Liao CC, Tseng YH, Lai YC, Lai YS, et al. Plectin deficiency in liver cancer cells promotes cell migration and sensitivity to sorafenib treatment. Cell Adh Migr 2018;12:19–27.10.1080/19336918.2017.1288789Search in Google Scholar PubMed PubMed Central

37 Xu R, He S, Ma D, Liang R, Luo Q, Song G. Plectin Downregulation Inhibits Migration and Suppresses Epithelial Mesenchymal Transformation of Hepatocellular Carcinoma Cells via ERK1/2 Signaling. Int J Mol Sci 2022;24:73.10.3390/ijms24010073Search in Google Scholar PubMed PubMed Central

38 Han P, Liu J, Lei Y, Lin Z, Tian D, Yan W. Netrin-1 promotes the collective cell migration of liver cancer cells in a 3D cell culture model. J Physiol Biochem 2019;75:489–498.10.1007/s13105-019-00701-8Search in Google Scholar PubMed

39 Wang Z, Wang W, Luo Q, Song G. High matrix stiffness accelerates migration of hepatocellular carcinoma cells through the integrin β1-Plectin-F-actin axis. BMC Biol 2025;23:8.10.1186/s12915-025-02113-1Search in Google Scholar PubMed PubMed Central

40 Zhou L, He L, Liu CH, Qiu H, Zheng L, Sample KM, et al. Liver cancer stem cell dissemination and metastasis: uncovering the role of NRCAM in hepatocellular carcinoma. J Exp Clin Cancer Res 2023;42:311.10.1186/s13046-023-02893-wSearch in Google Scholar PubMed PubMed Central

41 Chen C, Shi C, Huang X, Zheng J, Zhu Z, Li Q, et al. Molecular Profiles and Metastasis Markers in Chinese Patients with Gastric Carcinoma. Sci Rep 2019;9:13995.10.1038/s41598-019-50171-7Search in Google Scholar PubMed PubMed Central

42 Wang J, Zhao G, Zhao Y, Zhao Z, Yang S, Zhou A, et al. N6-methylation in the development, diagnosis, and treatment of gastric cancer. J Transl Int Med 2024;12:5–21.10.2478/jtim-2023-0103Search in Google Scholar PubMed PubMed Central

43 Wang H, Wu M, Lu Y, He K, Cai X, Yu X, et al. LncRNA MIR4435-2HG targets desmoplakin and promotes growth and metastasis of gastric cancer by activating Wnt/β-catenin signaling. Aging (Albany NY) 2019;11:6657–6673.10.18632/aging.102164Search in Google Scholar PubMed PubMed Central

44 Hatakeyama H, Kondo T, Fujii K, Nakanishi Y, Kato H, Fukuda S, et al. Protein clusters associated with carcinogenesis, histological differentiation and nodal metastasis in esophageal cancer. Proteomics 2006;6:6300–6316.10.1002/pmic.200600488Search in Google Scholar PubMed

45 Otsubo T, Hagiwara T, Tamura-Nakano M, Sezaki T, Miyake O, Hinohara C, et al. Aberrant DNA hypermethylation reduces the expression of the desmosome-related molecule periplakin in esophageal squamous cell carcinoma. Cancer Med 2015;4:415–425.10.1002/cam4.369Search in Google Scholar PubMed PubMed Central

46 Wang EJ, Chen IH, Kuo BY, Yu CC, Lai MT, Lin JT, et al. Alterations of Cytoskeleton Networks in Cell Fate Determination and Cancer Development. Biomolecules 2022;12:1862.10.3390/biom12121862Search in Google Scholar PubMed PubMed Central

47 Fan W, Adebowale K, Váncza L, Li Y, Rabbi MF, Kunimoto K, et al. Matrix viscoelasticity promotes liver cancer progression in the pre-cirrhotic liver. Nature 2024;626:635–642.10.1038/s41586-023-06991-9Search in Google Scholar PubMed PubMed Central

48 Raymond K, Kreft M, Song JY, Janssen H, Sonnenberg A. Dual Role of alpha6beta4 integrin in epidermal tumor growth: tumor-suppressive versus tumor-promoting function. Mol Biol Cell 2007;18:4210–4221.10.1091/mbc.e06-08-0720Search in Google Scholar PubMed PubMed Central

49 Liu S, Liang W, Huang P, Chen D, He Q, Ning Z, et al. Multi-modal analysis for accurate prediction of preoperative stage and indications of optimal treatment in gastric cancer. Radiol Med 2023;128:509–519.10.1007/s11547-023-01625-6Search in Google Scholar PubMed

50 Kang SH, Kim HH. Intraperitoneal chemotherapy for gastric cancer: A contemporary perspective. Chin J Cancer Res 2023;35:15–18.10.21147/j.issn.1000-9604.2023.01.03Search in Google Scholar PubMed PubMed Central

51 Bai Y, Qin X, Ao X, Ran T, Zhou C, Zou D. The role of EUS in the diagnosis of early chronic pancreatitis. Endosc Ultrasound 2024;13:232–238.10.1097/eus.0000000000000077Search in Google Scholar PubMed PubMed Central

52 Du C, He Z, Gao F, Li L, Han K, Feng X, et al. Factors affecting the diagnostic value of liquid-based cytology by EUS-FNA in the diagnosis of pancreatic cystic neoplasms. Endosc Ultrasound 2024;13:94–99.10.1097/eus.0000000000000041Search in Google Scholar PubMed PubMed Central

53 Vara-Luiz F, Patita M, Pinto-Marques P, Mendes I, Canastra AR. First case report of pancreatic angiomyolipoma diagnosed by EUS-guided fine-needle biopsy. Endosc Ultrasound 2024;13:280–282.10.1097/eus.0000000000000065Search in Google Scholar PubMed PubMed Central

54 Karen Ying Lee, Yi-Hsiang Liu, Chin-Chin Ho, Ren-Jeng Pei, Kun-Tu Yeh, Chiung-Chi Cheng, et al. An early evaluation of malignant tendency with plectin expression in human colorectal adenoma and adenocarcinoma. J Med 2004;35:141–149.Search in Google Scholar

55 Ma Y, Zhang P, Wang F, Liu W, Yang J, Qin H. An integrated proteomics and metabolomics approach for defining oncofetal biomarkers in the colorectal cancer. Ann Surg 2012;255:720-30.10.1097/SLA.0b013e31824a9a8bSearch in Google Scholar PubMed

56 Meier SM, Kreutz D, Winter L, Klose MHM, Cseh K, Weiss T, et al. An Organoruthenium Anticancer Agent Shows Unexpected Target Selectivity For Plectin. Angew Chem Int Ed Engl 2017;56:8267–8271.10.1002/anie.201702242Search in Google Scholar PubMed

57 Wernitznig D, Meier-Menches SM, Cseh K, Theiner S, Wenisch D, Schweikert A, et al. Plecstatin-1 induces an immunogenic cell death signature in colorectal tumour spheroids. Metallomics 2020;12:2121–2133.10.1039/d0mt00227eSearch in Google Scholar PubMed

58 Levy A, Morel D, Texier M, Sun R, Durand-Labrunie J, Rodriguez-Ruiz ME, et al. An international phase II trial and immune profiling of SBRT and atezolizumab in advanced pretreated colorectal cancer. Mol Cancer 2024;23:61.10.1186/s12943-024-01970-8Search in Google Scholar PubMed PubMed Central

59 Bausch D, Thomas S, Mino-Kenudson M, Fernández-del CC, Bauer TW, Williams M, et al. Plectin-1 as a novel biomarker for pancreatic cancer. Clin Cancer Res 2011;17:302–309.10.1158/1078-0432.CCR-10-0999Search in Google Scholar PubMed PubMed Central

60 Haig A, John AS, Vaska K, Banh X, Huelsen A. Comparing the diagnostic adequacy of 25-Gauge fork-tip versus franseen versus reverse-bevel-type needles in EUS-guided tissue acquisition: A prospective randomized study with a retrospective control. Endosc Ultrasound 2024;13:22–27.10.1097/eus.0000000000000025Search in Google Scholar PubMed PubMed Central

61 Kawasaki Y, Hijioka S, Nagashio Y, Ohba A, Maruki Y, Takeshita K, et al. Diagnostic performance of EUS-guided tissue acquisition for solid pancreatic lesions ≤10 mm. Endosc Ultrasound 2024;13:115–122.10.1097/eus.0000000000000052Search in Google Scholar PubMed PubMed Central

62 Ahmadipour M, Bhattacharya A, Sarafbidabad M, Syuhada Sazali E, Krishna Ghoshal S, Satgunam M, et al. CA19-9 and CEA biosensors in pancreatic cancer. Clin Chim Acta 2024;554:117788.10.1016/j.cca.2024.117788Search in Google Scholar PubMed

63 Park JK, Paik WH, Song BJ, Ryu JK, Kim MA, Park JM, et al. Additional K-ras mutation analysis and Plectin-1 staining improve the diagnostic accuracy of pancreatic solid mass in EUS-guided fine needle aspiration. Oncotarget 2017;8:64440–64448.10.18632/oncotarget.16135Search in Google Scholar PubMed PubMed Central

64 Song BG, Kwon W, Kim H, Lee EM, Han YM, Kim H, et al. Detection of Circulating Tumor Cells in Resectable Pancreatic Ductal Adenocarcinoma: A Prospective Evaluation as a Prognostic Marker. Front Oncol 2021;10:616440.10.3389/fonc.2020.616440Search in Google Scholar PubMed PubMed Central

65 Wang L, Li Y, Xu J, Zhang A, Wang X, Tang R, et al. Quantified postsurgical small cell size CTCs and EpCAM+ circulating tumor stem cells with cytogenetic abnormalities in hepatocellular carcinoma patients determine cancer relapse. Cancer Lett 2018;412:99–107.10.1016/j.canlet.2017.10.004Search in Google Scholar PubMed

66 Sanna V, Nurra S, Pala N, Marceddu S, Pathania D, Neamati N, et al. Targeted Nanoparticles for the Delivery of Novel Bioactive Molecules to Pancreatic Cancer Cells. J Med Chem 2016;59:5209–5220.10.1021/acs.jmedchem.5b01571Search in Google Scholar PubMed

67 Chen X, Zhou H, Li X, Duan N, Hu S, Liu Y, et al. Plectin-1 Targeted Dual-modality Nanoparticles for Pancreatic Cancer Imaging. EBioMedicine 2018;30:129–137.10.1016/j.ebiom.2018.03.008Search in Google Scholar PubMed PubMed Central

68 Wang X, Xing X, Zhang B, Liu F, Cheng Y, Shi D. Surface engineered antifouling optomagnetic SPIONs for bimodal targeted imaging of pancreatic cancer cells. Int J Nanomedicine 2014;9:1601–1615.10.2147/IJN.S58334Search in Google Scholar PubMed PubMed Central

69 Cromey B, McDaniel A, Matsunaga T, Vagner J, Kieu KQ, Banerjee B. Pancreatic cancer cell detection by targeted lipid microbubbles and multiphoton imaging. J Biomed Opt 2018;23:1–8.10.1117/1.JBO.23.4.046501Search in Google Scholar PubMed

70 Wang Q, Yan H, Jin Y, Wang Z, Huang W, Qiu J, et al. A novel plectin/integrin-targeted bispecific molecular probe for magnetic resonance/near-infrared imaging of pancreatic cancer. Biomaterials 2018;183:173–184.10.1016/j.biomaterials.2018.08.048Search in Google Scholar PubMed

71 Harpel K, Baker RD, Amirsolaimani B, Mehravar S, Vagner J, Matsunaga TO, et al. Imaging of targeted lipid microbubbles to detect cancer cells using third harmonic generation microscopy. Biomed Opt Express 2016;7:2849–2860.10.1364/BOE.7.002849Search in Google Scholar PubMed PubMed Central

72 Gazzi T, Lesina M, Wang Q, Berninger A, Radetzki S, Demir IE, et al. DOTA-Based Plectin-1 Targeted Contrast Agent Enables Detection of Pancreatic Cancer in Human Tissue. Angew Chem Int Ed Engl 2024;63:e202318485.10.1002/anie.202318485Search in Google Scholar PubMed

73 Zhu Q, Zeng S, Yang J, Zhuo J, Wang P, Wen S, et al. Plectin-1-targeted recognition for enhancing comprehensive therapy in pancreatic ductal adenocarcinoma. Nanoscale 2024;16:18584–18596.10.1039/D4NR01587HSearch in Google Scholar PubMed

74 Konkalmatt PR, Deng D, Thomas S, Wu MT, Logsdon CD, French BA, et al. Plectin-1 Targeted AAV Vector for the Molecular Imaging of Pancreatic Cancer. Front Oncol 2013;3:84.10.3389/fonc.2013.00084Search in Google Scholar PubMed PubMed Central

75 Li Y, Zhao Z, Liu H, Fetse JP, Jain A, Lin CY, et al. Development of a Tumor-Responsive Nanopolyplex Targeting Pancreatic Cancer Cells and Stroma. ACS Appl Mater Interfaces 2019;11:45390–45403.10.1021/acsami.9b15116Search in Google Scholar PubMed PubMed Central

76 Yuan Y, Du C, Sun C, Zhu J, Wu S, Zhang Y, et al. Chaperonin-GroEL as a Smart Hydrophobic Drug Delivery and Tumor Targeting Molecular Machine for Tumor Therapy. Nano Lett 2018;18:921–928.10.1021/acs.nanolett.7b04307Search in Google Scholar PubMed

77 Pal K, Al-Suraih F, Gonzalez-Rodriguez R, Dutta SK, Wang E, Kwak HS, et al. Multifaceted peptide assisted one-pot synthesis of gold nanoparticles for plectin-1 targeted gemcitabine delivery in pancreatic cancer. Nanoscale 2017;9:15622–15634.10.1039/C7NR03172FSearch in Google Scholar

78 Yi-Hsiang Liu, Chin-Chin Ho, Chiung-Chi Cheng, Ren-Jeng Pei, Yung-Hsiang Hsu, Kun-Tu Yeh, et al. Pleomorphism of cancer cells with the expression of plectin and concept of filament bundles in human hepatocellular carcinoma. Res Commun Mol Pathol Pharmacol 2007;:43–54.Search in Google Scholar

79 Julien Dimastromatteo, Jiang He, Reid B Adams, Kimberly A Kelly. Imaging Cell Surface Plectin in PDAC Patients - A First-In-Human Phase 0 Study Report. Mol Imaging Biol ;:. DOI: 10.1007/s11307-025-02001-8]10.1007/s11307-025-02001-8]Search in Google Scholar

80 Kung CY, Fang WL, Hung YP, Huang KH, Chen MH, Chao Y, et al. Comparison of the mutation patterns between tumor tissue and cell-free DNA in stage IV gastric cancer. Aging (Albany NY) 2023;15:777–790.10.18632/aging.204512Search in Google Scholar PubMed PubMed Central

81 Lee HJ, Na K, Kwon MS, Kim H, Kim KS, Paik YK. Quantitative analysis of phosphopeptides in search of the disease biomarker from the hepatocellular carcinoma specimen. Proteomics 2009;9:3395–3408.10.1002/pmic.200800943Search in Google Scholar PubMed

82 Nishimori T, Tomonaga T, Matsushita K, Oh-Ishi M, Kodera Y, Maeda T, et al. Proteomic analysis of primary esophageal squamous cell carcinoma reveals downregulation of a cell adhesion protein, periplakin. Proteomics 2006;6:1011–1018.10.1002/pmic.200500262Search in Google Scholar PubMed

83 Outla Z, Oyman-Eyrilmez G, Korelova K, Prechova M, Frick L, Sarnova L, et al. Plectin-mediated cytoskeletal crosstalk as a target for inhibition of hepatocellular carcinoma growth and metastasis. Elife 2025;1:3.10.7554/eLife.102205.2Search in Google Scholar

84 Osmanagic-Myers S, Wiche G. Plectin-RACK1 (receptor for activated C kinase 1) scaffolding: a novel mechanism to regulate protein kinase C activity. J Biol Chem 2004;279:18701–18710.10.1074/jbc.M312382200Search in Google Scholar PubMed

85 Hu L, Su P, Yin C, Zhang Y, Li R, Yan K, et al. Microtubule actin crosslinking factor 1 promotes osteoblast differentiation by promoting β-catenin/TCF1/Runx2 signaling axis. J Cell Physiol 2018;233:1574–1584.10.1002/jcp.26059Search in Google Scholar PubMed

86 Wu X, Shen QT, Oristian DS, Lu CP, Zheng Q, Wang HW, et al. Skin stem cells orchestrate directional migration by regulating microtubule-ACF7 connections through GSK3β. Cell 2011;144:341–352.10.1016/j.cell.2010.12.033Search in Google Scholar PubMed PubMed Central

87 Ka M, Jung EM, Mueller U, Kim WY. MACF1 regulates the migration of pyramidal neurons via microtubule dynamics and GSK-3 signaling. Dev Biol 2014;395:4–18.10.1016/j.ydbio.2014.09.009Search in Google Scholar PubMed PubMed Central

88 Qiu WX, Ma XL, Lin X, Zhao F, Li DJ, Chen ZH, et al. Deficiency of Macf1 in osterix expressing cells decreases bone formation by Bmp2/Smad/Runx2 pathway. J Cell Mol Med 2020;24:317–327.10.1111/jcmm.14729Search in Google Scholar PubMed PubMed Central

89 Wang P, Zhang J, Zhang H, Zhang F. The role of MACF1 on acute myeloid leukemia cell proliferation is involved in Runx2-targeted PI3K/Akt signaling. Mol Cell Biochem 2023;478:433–441.10.1007/s11010-022-04517-xSearch in Google Scholar PubMed

90 Lin X, Xiao Y, Chen Z, Ma J, Qiu W, Zhang K, et al. Microtubule actin crosslinking factor 1 (MACF1) knockdown inhibits RANKL-induced osteoclastogenesis via Akt/GSK3β/NFATc1 signalling pathway. Mol Cell Endocrinol 2019;494:110494.10.1016/j.mce.2019.110494Search in Google Scholar PubMed

91 Tonoike Y, Matsushita K, Tomonaga T, Katada K, Tanaka N, Shimada H, et al. Adhesion molecule periplakin is involved in cellular movement and attachment in pharyngeal squamous cancer cells. BMC Cell Biol 2011;12:41.10.1186/1471-2121-12-41Search in Google Scholar PubMed PubMed Central

92 van den Heuvel AP, de Vries-Smits AM, van Weeren PC, Dijkers PF, de Bruyn KM, Riedl JA, et al. Binding of protein kinase B to the plakin family member periplakin. J Cell Sci 2002;115:3957-66.10.1242/jcs.00069Search in Google Scholar PubMed

93 Suzuki A, Horiuchi A, Ashida T, Miyamoto T, Kashima H, Nikaido T, et al. Cyclin A2 confers cisplatin resistance to endometrial carcinoma cells via up-regulation of an Akt-binding protein, periplakin. J Cell Mol Med 2010;14:2305–2317.10.1111/j.1582-4934.2009.00839.xSearch in Google Scholar PubMed PubMed Central

94 Zhang Y, Lee RY, Tan CW, Guo X, Yim WW, Lim JC, et al. Spatial omics techniques and data analysis for cancer immunotherapy applications. Curr Opin Biotechnol 2024;87:103111.10.1016/j.copbio.2024.103111Search in Google Scholar PubMed

95 Allen AG, Khan SQ, Margulies CM, Viswanathan R, Lele S, Blaha L, et al. A highly efficient transgene knock-in technology in clinically relevant cell types. Nat Biotechnol 2024;42:458–469.10.1038/s41587-023-01779-8Search in Google Scholar PubMed

96 Byrne A, Le D, Sereti K, Menon H, Vaidya S, Patel N, et al. Single-cell long-read targeted sequencing reveals transcriptional variation in ovarian cancer. Nat Commun 2024;15:6916.10.1038/s41467-024-51252-6Search in Google Scholar PubMed PubMed Central

97 Huang G, Yuan C, Zhang C, Yang F, Tan Y, Chen D, et al. Single-cell sequencing reveals the immune microenvironment associated with gastric cancer. Genes Dis 2024;12:101218.10.1016/j.gendis.2024.101218Search in Google Scholar PubMed PubMed Central

98 Feng DC, Zhu WZ, Wang J, Li DX, Shi X, Xiong Q, et al. The implications of single-cell RNA-seq analysis in prostate cancer: unraveling tumor heterogeneity, therapeutic implications and pathways towards personalized therapy. Mil Med Res 2024;11:21.10.1186/s40779-024-00526-7Search in Google Scholar PubMed PubMed Central

99 Li D, Yu Q, Wu R, Tuo Z, Wang J, Ye L, et al. Interactions between oxidative stress and senescence in cancer: Mechanisms, therapeutic implications, and future perspectives. Redox Biol 2024;73:103208.10.1016/j.redox.2024.103208Search in Google Scholar PubMed PubMed Central

100 Wang J, Shao F, Yu QX, Ye L, Wusiman D, Wu R, et al. The Common Hallmarks and Interconnected Pathways of Aging, Circadian Rhythms, and Cancer: Implications for Therapeutic Strategies. Research (Wash D C) 2025;8:0612.10.34133/research.0612Search in Google Scholar PubMed PubMed Central

101 Wei J, Bu Z. Advances in gastric cancer treatment in 2024: Key breakthroughs and emerging trends. Chin J Cancer Res 2024;36:592–595.10.21147/j.issn.1000-9604.2024.06.02Search in Google Scholar PubMed PubMed Central

102 Mahuron KM, Fong Y. Applications of Liquid Biopsy for Surgical Patients With Cancer: A Review. JAMA Surg 2024;159:96–103.10.1001/jamasurg.2023.5394Search in Google Scholar PubMed

103 Torresan S, de Scordilli M, Bortolot M, Di Nardo P, Foltran L, Fumagalli A, et al. Liquid biopsy in colorectal cancer: Onward and upward. Crit Rev Oncol Hematol 2024;194:104242.10.1016/j.critrevonc.2023.104242Search in Google Scholar PubMed

104 Passaro A, Al Bakir M, Hamilton EG, Diehn M, André F, Roy-Chowdhuri S, et al. Cancer biomarkers: Emerging trends and clinical implications for personalized treatment. Cell 2024;187:1617–1635.10.1016/j.cell.2024.02.041Search in Google Scholar PubMed PubMed Central

105 Hu Y, Dong Z, Liu K. Unraveling the complexity of STAT3 in cancer: molecular understanding and drug discovery. J Exp Clin Cancer Res 2024;43:23.10.1186/s13046-024-02949-5Search in Google Scholar PubMed PubMed Central

106 Zhong H, Li P, Yan Q, Xia Y, Zhang X, Lai Y, et al. Targeting Periplakin of Novel Benzenesulfonamides as Highly Selective Agonists for the Treatment of Vitiligo. J Med Chem 2024;67:19323–19341.10.1021/acs.jmedchem.4c01717Search in Google Scholar PubMed

107 Perez SM, Dimastromatteo J, Landen CN Jr, Kelly KA. A Novel Monoclonal Antibody Targeting Cancer-Specific Plectin Has Potent Antitumor Activity in Ovarian Cancer. Cells 2021;10:2218.10.3390/cells10092218Search in Google Scholar PubMed PubMed Central

Published Online: 2025-07-30

© 2025 Changwei Huang, Yixuan Chen, Manoop S. Bhutani, Caixia Wang, Yang Zhou, Jintao Guo, Siyu Sun, published by De Gruyter on behalf of the SMP

This work is licensed under the Creative Commons Attribution 4.0 International License.

Downloaded on 19.9.2025 from https://www.degruyterbrill.com/document/doi/10.1515/jtim-2025-0033/html
Scroll to top button