92
Views
0
CrossRef citations to date
0
Altmetric
Reviews

Epigenetic regulation of TGF-β pathway and its role in radiation response

ORCID Icon, &
Pages 834-848 | Received 06 Sep 2023, Accepted 27 Feb 2024, Published online: 20 Mar 2024

Abstract

Purpose

Transforming growth factor (TGF-β) plays a dual role in tumor progression as well as a pivotal role in radiation response. TGF-β-related epigenetic regulations, including DNA methylation, histone modifications (including methylation, acetylation, phosphorylation, ubiquitination), chromatin remodeling and non-coding RNA regulation, have been found to affect the occurrence and development of tumors as well as their radiation response in multiple dimensions. Due to the significance of radiotherapy in tumor treatment and the essential roles of TGF-β signaling in radiation response, it is important to better understand the role of epigenetic regulation mechanisms mediated by TGF-β signaling pathways in radiation-induced targeted and non-targeted effects.

Conclusions

By revealing the epigenetic mechanism related to TGF-β-mediated radiation response, summarizing the existing relevant adjuvant strategies for radiotherapy based on TGF-β signaling, and discovering potential therapeutic targets, we hope to provide a new perspective for improving clinical treatment.

Introduction

As we all know, cancer is one of the diseases with high morbidity and mortality worldwide, and effective targeted tumor treatment has always been the goal of both researchers and clinicians (Najafi et al. Citation2021). In recent years, more and more studies have shown that epigenetic reprogramming dysfunction affects the development of many types of cancer (Greer and Yang Citation2014). Epigenetics mainly describes biochemical changes in DNA or chromatin, i.e. changing gene expression patterns without changing the DNA sequence. Most epigenetic changes are mediated by DNA methylation regulators, histone modifiers, and non-coding RNAs such as long non-coding RNAs (lncRNAs) and microRNAs (miRNAs)(Suriyamurthy et al. Citation2019). At present, epigenetics has been found to play a key role in autoimmune diseases, cancer, congenital diseases, mental retardation, endocrine diseases and neuropsychiatric diseases (Tzika et al. Citation2018; Zhang et al. Citation2020).

Transforming growth factor-β (TGF-β) is a key factor in cancer progression, and its role is mainly manifested in inhibiting tumor growth at an early stage by inducing apoptosis and cell cycle arrest. However, it promotes advanced-stage tumor progression by regulating genomic instability, epithelial-mesenchymal transition (EMT), angiogenesis, immune evasion, cell motility and metastasis. Disturbance of TGF-β/Smad signaling often leads to epigenetic variation in its target genes and the subsequent phenotypic changes. Radiotherapy is a conventional therapy, in which the change of TGF-β expression induced by ionizing radiation exposure plays an important role in tumor killing and normal tissue damage repair. Overexpression of TGF-β often leads to different side effects of radiotherapy, including vascular injury and fibrosis (Russell et al. Citation2015; Dadrich et al. Citation2016). Therefore, understanding the role of the epigenetic mechanism of the TGF-β pathway in the radiation response may contribute to effective targeted tumor therapy.

Therefore, in this review, we first focus on epigenetics-related advances in the TGF-β pathway, and then we underscore the epigenetic regulatory mechanisms of TGF-β in response to radiation. Finally, we summarize the existing adjuvant strategies for radiotherapy targeting the TGF-β signaling pathway and propose potential research directions for future application in clinical therapy.

A brief overview of the TGF-β pathway

The TGF-β superfamily includes TGF-β, bone morphogenetic protein (BMP), activin and related proteins, which regulate body function and tissue differentiation by affecting cell proliferation, differentiation and migration (Zhang Citation2009; Farhood et al. Citation2020). The TGF-β signal mainly includes three kinds of ligands: TGF-β1, TGF-β2 and TGF-β3, which have three kinds of receptors: the transmembrane receptor serine/threonine kinase TβRI, TβRII and β-glycan TβRIII (Ahmadi et al. Citation2019). The TGF-β signaling pathway mainly mediates tumor progression in two ways: canonical (Smad dependent) and non-canonical (Smad independent) pathways (Derynck and Zhang Citation2003).

Canonical pathway

The classical TGF-β pathway mainly involves the binding of dimer to the membrane receptor serine kinase. In short, TGF-β ligand binding triggers the assembly of TβRI and TβRII complexes (Vander Ark et al. Citation2018). In the complex, type II receptor subunit phosphorylates type I at multiple serine and threonine residues located at the N-terminal of the type I receptor kinase domain (Shi and Massagué Citation2003). Therefore, type I receptor kinase is activated and phosphorylated Smad2/3 (receptor Smads, R-Smads) is recruited for signal transduction (Macias et al. Citation2015). Smad4 (Common Smad, Co-Smad) forms a trimeric complex with phosphorylated R-Smads, which is then transferred to the nucleus to interact with DNA or transcription factors (Feng and Derynck Citation2005).

Smads is the downstream effector of TGF-β signal transduction. Both R-Smads and Co-Smad proteins consist of two highly conserved domains, Mad homology 1 (MH1) and Mad homology 2 (MH2). The C-terminal Ser-X-Ser (SXS) motif in the R-Smads MH2 domain is the phosphorylation site of TβRI, which can lead to its activation (Tzavlaki and Moustakas Citation2020). There is an L3 loop in the C-terminal MH2 domain of both R-Smads and Co-Smad, which is essential for the interaction of Smad-TβRs and the formation of trimer complexes (Lo Roger et al. Citation1998). At the same time, TGF-β often interacts with other pathways in a Smad-dependent manner. For example, the Smad3/Smad4 complex directly interacts with PKA regulatory subunits to activate PKA signaling pathway (L. Zhang et al. Citation2004); the interaction between Smad factor and RAS response element binding protein 1 (RREB1) also provides a molecular link between RAS and TGF-β signaling pathways (Su et al. Citation2020). In addition, inhibitory Smad (I-Smad), Smad7, negatively regulates TGF-β signal transduction through multiple mechanisms (de Ceuninck van Capelle et al. Citation2020). Smad7 protein can competitively bind R-Smads through the first α-helix (α1) and L3 loop in the C-terminal MH2 domain, interfering with the formation of trimer complex (Murayama et al. Citation2020). On the other hand, Smad7 promotes the binding to TβRI by interacting with E3 ubiquitin ligases such as Smurf1 and Smurf2 (Asano et al. Citation2004). Finally, Smad7 can act on the downstream of TβRs and reduce the activation of Smad2 and Smad3 to inhibit TGF-β signal transduction (Humeres et al. Citation2022).

Non-canonical pathways

In addition to the canonical pathway, TGF-β can also mobilize some other intracellular pathways (Zhang Citation2017). The non-canonical signaling pathway is mainly mediated by activation of phosphatidylinositol 3′-kinase (PI3K)-protein kinase B (AKT), nuclear factor kappa B (NF-kB), small G protein (Rac and Rho) and mitogen-activated protein kinase members (MAPK, including ERK, JNK and p38) (Cammareri et al. Citation2017; Yuan et al. Citation2020). Through phosphorylation or direct interaction, TGF-β receptors activate and regulate non-canonical signaling pathways, and finally lead to genetic regulation (Zhang Citation2009). For example, after activating kinase TAK1, the TGF-β receptor initiates NLRP3 inflammatory body-related signaling pathway in LX-2 cells by activating NF-kB (Kang et al. Citation2022); The binding of anti-inflammatory transcription factor PPARα to TAK1 can inhibit TGF-β signal transduction (Subramanian et al. Citation2018); TGF-β induces down-regulation of RhoE which belongs to the RhoA family to activate the signal transduction of RhoA/ROCK leading to EMT (Nishizuka et al. Citation2019).

The dual role of TGF-β in tumor

TGF-β plays a dual role in tumor progression. TGF-β acts as an inhibitor to induce growth arrest in various cell types, including epithelial cells, endothelial cells, hematopoietic cells, nerve cells and some mesenchymal cells (Maliekal et al. Citation2004; Zemann et al. Citation2010; Cammareri et al. Citation2017). The expression of cyclin D and CDK down-regulated the expression of TGF-β in the G1 phase (Zhang et al. Citation2009). The TGF-β/Smad signaling pathway is activated and mediates apoptosis through direct or indirect interaction with AKT, JNK and p39 (Pardali and Moustakas Citation2007; Massagué Citation2008). Some studies have also shown that reactive oxygen species (ROS) are closely related to TGF-β-induced apoptosis (Al-Khayal et al. Citation2017). In later stages, TGF-β continues to be produced by which EMT (Feng et al. Citation2022), cell metastasis (Yoon et al. Citation2021), angiogenesis(Chen et al. Citation2021), and immune escape (Huang et al. Citation2019) are induced through increasing neovascularization and extracellular matrix production, upregulating peritumoral proteases, or inhibiting immune surveillance mechanisms in cancer hosts to promote cancer progression (Mortezaee Citation2021).

Epigenetic regulatory mechanisms play an important role in tumor progression

Epigenetic aberration is a common feature of cancer, characterized by hypermethylation of specific promoter regions and global DNA hypomethylation, and/or either loss or gain of histone acetylation (Liang and Weisenberger Citation2017). Remodeling of chromatin is critical for nucleosome localization, chromatin accessibility to transcription factors, and regulation of gene expression. Silencing of tumor suppressor genes and activation of oncogenes are hallmarks of epigenetic variation.

Epigenetic modification is very complex and plays an important role in the occurrence and development of tumors by linking different genes or proteins. For example, palmitate environments in tumors promote the expression of lysine acetyltransferase 2a (KAT2a) and promote metastatic growth by increasing p65 acetylation to produce metastatic NF-κB signal transduction (Altea-Manzano et al. Citation2023). SWI/SNF complex causes metastasis and progression of lung cancer through SMARCA4 deletion (Concepcion et al. Citation2022). Lactate secreted by CAF (cancer associated fibroblast) also can regulate histone acetylation, up-regulate PLIN2, then establishing a metabolic-epigenetic regulatory loop and affecting the growth and metastasis of prostate cancer (Ippolito et al. Citation2022). On the other hand, H4K20me2 acetylation can directly regulate the relative chromatin occupancy of BRCA1 and 53BP1, thus affecting the mechanism of DNA repair (Tang et al. Citation2013). H3K27me3 mediates the up-regulation of TWIST1 and inhibition of SLFN11 in SCLC, which affects the efficiency of DNA damage repair and leads to chemotherapy resistance (Gardner et al. Citation2017). TGF-β is one of the most important targets for epigenetic regulation and plays a role in the modulation of cellular radiation response by participating in the epigenetic variation related to radiation response (Suriyamurthy et al. Citation2019).

The TGF-β pathway is an important target for epigenetic regulation

DNA methylation

As the most common epigenetic phenomenon, DNA methylation refers to the process by which cytosine of CpG double nucleotides is catalyzed by DNA methyltransferases (DNMTs) with S-adenosylmethionine (SAM) as the methyl donor and obtains methyl group chemical modification through covalent bonding. DNA methylation plays an important role in cancer progression by regulating the TGF-β signaling pathway. Zheng et al. found that RUNX3 and TGF-β levels were decreased in metastatic renal cancer tissues as a result of their CpG methylation (Zheng et al. Citation2018). Interestingly, database-based analysis of immune cells from breast cancer found that RUNX3 gene methylation was negatively correlated with TGF-β1 expression, while the situation was exactly the opposite for RUNX1(Gao and Zhou Citation2021). In conclusion, altering TGF-β1 expression through RUNX methylation can regulate the TGF-β signaling pathway to influence cancer progression. For non-small-cell lung carcinoma (NSCLC), -459CpG methylation inhibits RNF111 transcriptional expression by interfering with the recruitment of Sp1 to the RNF111 promoter, thereby inhibiting TGF-β/Smad signaling activation and Snail expression, increasing E-cadherin expression to prevent invasion (Chen et al. Citation2015). Other studies have also demonstrated that decreased TβRII expression in NSCLC is highly correlated with CpG methylation (Zhang et al. Citation2004; Pardali and Moustakas Citation2007). In the microenvironment of gastric cancer, high expression and hypomethylation of TGF-β2 can induce high levels of infiltration of multiple immune cells and expression of cytokines, which are positively correlated with EMT (Han et al. Citation2022). In addition, different N6-methyladenosine (m6A) reader proteins, YTHDF3 and IGF2BP1, were shown to associate with JUN and JUNB mRNA, respectively to mediate the m6A-dependent regulation of JUN protein translation and JUNB mRNA stability, thus promoting TGF-β-mediated EMT (Suphakhong et al. Citation2022). However, methyltransferase METTL3 deletion can down-regulate m6A and inhibit Snail expression, ultimately preventing TGF-β1-induced EMT (Lin et al. Citation2019; Li et al. Citation2023).

Histone modification

Different histone modifications, including methylation, acetylation, phosphorylation and ubiquitination, occur under the action of different histone modification enzymes. To some extent, histone modification leads to transcriptional activation or gene silencing, which regulates gene expression and even leads to tumorigenesis.

Methylation

Mutations or altered expression of histone methyl modifiers and methyl-binding proteins are associated with increased incidence of many cancers (Albert and Helin Citation2010). For example, H3K27me3 methyltransferase is upregulated in a number of cancers, including prostate cancer (Genao et al. Citation2002), breast cancer (Kleer et al. Citation2003) and lymphoma (Visser et al. Citation2001). Down-regulation of NSD2, which regulates the histone methyltransferase of histone3 lysine36(H3K36me2) dimethylation, can significantly reduce the expression levels of TGF-β1, TβRI, phosphorylated Smad2 and Smad3 in cervical cancer cells and inhibit tumor metastasis (Zhu et al. Citation2019). The significant down-regulation of H3K79me3 and the increased expression of axon guide proteins semaphorin 3 C (SEMA3C) and PD-L1 involved in immune system suppression are caused by the up-regulation of TGF-β1 in lung cancer (Evanno et al. Citation2017). Zhang et al also found that the H3K9/H3K27 double demethylase JHDM1D/KDM7A is critical for TGF-β-induced transcription of RHOJ in breast cancer cells (Zhang et al. Citation2021). Among them, KDM7A seems to be a direct transcriptional target of TGF-β signaling (Liu et al. Citation2019). The Smad2/Smad4 complex binds to the KDM7A promoter to mediate TGF-β-induced KDM7A transcription and regulate EMT and cell migration. Another histone methyltransferase, SUV39H1, regulates telomere length upon upregulation of TGF-β while affecting telomere-terminal histone methylation levels, ultimately leading to telomere variation that affects tumor progression (Mishra et al. Citation2022).

Acetylation

Histone acetyltransferase (HAT) and histone deacetylase (HDACS) antagonize each other to maintain a dynamic equilibrium of histone acetylation levels and participate in the regulation of gene expression (Grunstein Citation1997). p300/CBP is an important member of a group of acetyltransferases, whose abnormal expression often leads to a series of effects, such as inducing tumor cell proliferation and metastasis (Asaduzzaman et al. Citation2017; Boudreau et al. Citation2017). Reducing the protein levels of p300/CBP and p-Smad2/3 in the nucleus can hinder the proliferation and metastasis of esophageal carcinoma cells (Wang et al. Citation2020). Another HAT member, GCN5, also plays a key role in regulating EMT in breast cancer downstream of the TGF-β/Smad signaling pathway (Zhao et al. Citation2018). Among HDACs family members, the increase of HDAC1 activity is mediated by post-translational modification, and independently mediates the activation of TGF-β-Smad2/3 signaling and CAF status in breast cancer (Mezawa et al. Citation2023). HDAC1-mediated global histone deacetylation and acquisition of enhancers labeled with specific histone H3 lysine27 acetylation (H3K27AC) are critical for TGF-β-induced EMT (Qiao et al. Citation2020). HDAC4 silencing blocks TGF-β signal transduction and reduces the expression of Sox2 and Nanog to inhibit the acetylation of STAT1 and hinder tumor progression (Kaowinn et al. Citation2018). In NSCLC, TGF-β1 upregulates the expression of HDAC6, and HDAC6 inhibits the expression of H3K27ac on the transmembrane protein TMEM100 promoter by deacetylation, which leads to endothelial cell metastasis and activation of Wnt/β-catenin signaling pathway (Wang et al. Citation2022). In addition, lysine 102 in Smad7 promotes the migration and invasion of prostate cancer cells by binding to regulatory regions in c-Jun and HDAC6 to enhance the induction of c-Jun and HDAC6 by TGF-β(Thakur et al. Citation2020). H3K27AC simultaneously regulates carcinogenesis and alters the TGF-β3 and Wnt/β-catenin pathways, and its deacetylation has been shown to reverse and up-regulate tumor suppressor gene expression in uterine leiomyoma (UL)(Carbajo‐garcía et al. Citation2022).

Phosphorylation

Phosphorylation occurs on all histones and plays a different role on each histone, influencing tumor progression by regulating cell proliferation, differentiation and apoptosis. For example, in non-canonical pathways, Ras-PI3K pathway activation promotes osteosarcoma development by upregulating histone H2AT120 phosphorylation (Xu and Yu Citation2019), or by PKA regulating H1.4S35 phosphorylation (Zhang et al. Citation2019). In pancreatic cancer, activation of the Ras-MAPK pathway results in increased phosphorylation of histone H3 kinase, mitogen and stress-activated kinase (MSK1), and histone H3(H3S10) at serine 10 to regulate tumor transformation (Espino et al. Citation2009). Phosphorylation of H3S10 mediated by JNK and PI3K/AKT pathways may also promote lung carcinogenesis (Ibuki et al. Citation2014). However, the role of canonical pathway-mediated histone phosphorylation in tumor regulation seems to be unclear and needs to be further explored.

Ubiquitination

Ubiquitination refers to the covalent binding of ubiquitin to target proteins catalyzed by a series of enzymes, in which E3 ubiquitin ligase (E3s) plays an important role, and its regulation is related to cancer development (Senft et al. Citation2018; Sinha et al. Citation2021). According to the specific structural motif, E3s can be divided into E3s with HECT domain, RING domain, and U-box domain (Nakayama and Nakayama Citation2006). In the HECT domain E3s, Smad ubiquitin regulators 1 (Smurf1) and Smurf2 negatively regulate TGF-β/BMP signaling (Fu et al. Citation2019). Smurf2 promotes pancreatic cancer cell proliferation, migration and invasion through the TGF-β-induced non-canonical PI3K/AKT pathway (Sinha et al. Citation2021). RING domain E3s affect TGF-β signaling at different nodes. One of the family members of the RING, TRIM37, is upregulated in renal cell carcinoma (RCC), promoting EMT directly mediated by ubiquitination-H2A modification through activation of TGF-β1 signaling (Miao et al. Citation2021). Overexpression of another family member, RNF20, can significantly reverse TGF-β-induced activation of fibrotic proteins in hepatocytes (LX-2) and significantly inhibit the progression of liver fibrosis through ubiquitination of H2B (Chen et al. Citation2021). In addition, overexpression of KDM2B, a member of the polycomb repressive complex-1 (PRC1), specifically recognizes regulatory regions of Cdh1, miR-200a, and CGN genes and induces their regulated H2AK119 monoubiquitination as a component of the PRC1 complex, at the same time, enhances TGF-β-induced cell morphological transformation, thereby inhibiting the EMT, migration and invasion of lung and pancreatic cancer cells (Wanna-Udom et al. Citation2021). On the other hand, ubiquitin C-terminal hydrolase-L1 (Uch-L1) with deubiquitinating activity interacts with Smad2 and Smad3 to promote the signal transduction of DAF-7/TGF-β and promote the hypoxic survival rate of lung cancer cells (Nagata et al. Citation2020).

Chromatin remodeling

Chromatin remodeling is a dynamic process in which chromatin structures change between the condensed state and transcriptional accessible state. The SWI/SNF chromatin remodeling complex component, DPF3A, a short subtype of DPF3, specifically interacts with Smad nuclear-interacting protein (SNIP1) to form a complex with Smad4 and p300 HAT, inhibition of p300 HAT activity by the release of SNIP1 leads to increased local histone acetylation and activation of cell movement-related genes, promoting migration of kidney cancer cells (Cui et al. Citation2022). Recruitment of ARID1A, a subunit of SWI/SNF chromatin remodeling complex, can inhibit TβR2, KLF4 and FoxQ1 and induce BMP6, which hinders the morphological reprogramming of mammary epithelial cells (Jdeed et al. Citation2022). In breast cancer cells, the chromatin remodeling protein BRM binds directly to the promoter region of the Claudins gene by interacting with Sp1 and activates transcription by regulating histone modifications to inhibit TGF-β-induced migration and invasion (Yang, Liu, Fang, et al. Citation2019). In prostate cancer cells, the expression of chromatin remodeling proteins BRG1 and Elvol3 is upregulated with androgen and TGF-β responses. BRG1 interacts with and is recruited to the Elovl3 promoter to activate transcription by the retinoic acid receptor-associated orphan receptor, and it also interacts with p300 HAT to transactivate Elovl3 to promote migration and invasion (Yang, Liu, Li, et al. Citation2019). In lung cancer cells, BRG1 recruits histone H3K9 demethylase KDM3A, and after removing dimethyl H3K9 from the target gene promoter, interacts with Sp1 to activate potential TGF-β binding protein 2 (LTBP2) transcription to promote proliferation and migration (Li et al. Citation2019). Chromatin remodeling factor Pontin recruited LEF1 as a coactivator of LEF1 and upregulated TβR2 expression, activating TGF-β/Smad signaling to promote glioma formation (Zhou et al. Citation2022).

Non-coding RNA regulation

Non-coding RNA is a class of RNA that does not code proteins, in which lncRNA, miRNA, and siRNA play a role in regulating gene expression. Some studies on the epigenetic mechanisms of non-coding RNA-mediated TGF-β signaling pathways in tumor progression are shown in . LncRNA coordinates DNA methylation, chromatin structure remodeling, and histone modification in epigenetic regulation, and some studies have found that many lncRNAs are involved in the development and progression of tumors through the regulation of TGF-β signaling. Zhang et al. found that activation of the TGF-β/Smad pathway after binding of lncRNA PVT1 to TGF-β accelerates glioma proliferation, migration and invasion, while enrichment of p53 in the lncRNA PVT1 promoter region prevents its occurrence (Li et al. Citation2022). LncRNA ANCR is essential for TGF-β1-induced EMT. TGF-β1 downregulates ANCR expression by increasing the HDAC3 enrichment on the ANCR promoter region and reducing H3 and H4 acetylation of ANCR initiator genes. Ectopic expression of ANCR partially attenuates TGF-β1-induced EMT (Li et al. Citation2017). MiRNAs have also been found to be important in the regulation of TGF-β signaling. For example, TGF-β-induced miR-23a can target its downstream effector HMGN2, which alters chromatin structure by interfering with the binding of the linker histone H1 to nucleosomes to regulate transcription and induce EMT in lung cancer cells (Saito et al. Citation2013). MiR-132 targets TGF-β2 to enhance the expression of dexamethasone (DEX)-induced wave protein and E-cadherin, leading to reduced clonal formation and migration of pancreatic cancer cells (Abukiwan et al. Citation2019). TGF-β1 high expression decreases miR-200b/c while increasing miR-221, makes DNA methyltransferase DNMT3B stable and induces miR-200s promoter DNA methylation, thereby establishing and activating CAF and acting on malignant transformation of breast cancer (Tang et al. Citation2019).

Table 1. Non-coding RNA-mediated TGF-β signaling regulation in tumor progression.

TGF-β-mediated epigenetic regulation involved in radiation response

External irradiation

At present, low LET radiation is the most widely used irradiation method in the field of radiotherapy. The effect of TGF-β on the radiation damage of the intestine, heart and kidney after low LET radiation has been widely studied (Haydont et al. Citation2007; Boerma et al. Citation2013; Hritzo et al. Citation2021). More importantly, TGF-β often causes a series of sequelae after radiotherapy. Excessive activation of TGF-β caused by ionizing radiation often causes EMT and cell migration, (Al-Assar et al. Citation2014; Carl et al. Citation2016; Qu et al. Citation2022). Fibrosis is another common side effect induced by TGF-β after radiotherapy (Saitoh Citation2022). In radiation-induced skin fibrosis, radiation induces CpG dinucleotide demethylation in exon 1 of the zinc transporter ZIP9, leading to recruitment of the transcription factor Sp1 and increased ZIP9 expression, then protein kinase B (PKB) can activate the TGF-β signaling pathway in human fibroblasts, and further promote fibrosis (Qiu et al. Citation2020). Ryu et al found that the natural dithiol compound, α-LA, synthesized in mitochondria, can decrease the transcriptional activity of NF-κB by inhibiting TGF-β1-mediated upregulation of p300/CBP activity and histone acetyltransferase activity, to prevent radiation-induced fibrosis (RIF)(Ryu et al. Citation2016). In addition, in terms of radiosensitivity, overexpression of m6A methylase WTAP can promote TGF-β expression and mRNA stability, while reducing radiosensitivity in gastric cancer (GC) tissues, to enhance chemoradiotherapy resistance and promote cell migration and EMT (Liu and Da Citation2022). Meanwhile, TGF-β also plays an important role in the phosphorylation of key enzymes in the DNA damage response (DDR) following radiation and inhibition of TGF-β reduces ATM (Kirshner et al. Citation2006; Wang et al. Citation2013), γH2AX, and p53 phosphorylation (Wiegman et al. Citation2007; Bouquet et al. Citation2011), thereby exerting distinct effects on various types of cancer. Some epigenetics studies of non-coding RNAs mediating TGF-β modulation in radiation response in tumor progression are shown in . Radiation-induced lncRNA RP11 overexpression promotes various common collagen expressions by downregulating miR-29a and modulating human fibroblast activity after TGF-β1 or radiation treatment (Yang et al. Citation2020). TGF-β/Smad signal transduction significantly decreased miR-29 transcription and enhanced the expression of type I collagen induced by ionizing radiation, suggesting that miR-29 may be an important regulator of RIF (Yano et al. Citation2018). The results of Lu et al found that miR-3591-5p/USP33/PPM1A plays a key role in radiation-induced EMT. Radiation-upregulated miR-3591-5p inhibited USP33 transcription and increased PPM1A (a Smad2/3 phosphatase) ubiquitination, ultimately inhibiting TGF-β signaling (Lu et al. Citation2018).

Table 2. Non-coding RNA-mediated TGF-β signaling regulation in radiation-induced targeted effects.

In recent years, with the extensive development of proton and heavy ions radiotherapy, different TGF-β expression and its epigenetic effects caused by different types of high LET radiation are paid more and more attention (). In the comparison of carbon ions with X-rays, Ran et al found that at the same dose, carbon ions induced higher expression of HMGB1 and decreased the expression of immunosuppressive factors IL-6 and TGF-β, so as to better induce immune effect (Ran et al. Citation2021). In another comparative study of iron and boron ions, the authors found that γ-H2AX foci and Smad7 foci were detected in radiation-induced micronuclei, suggesting that Smad7 is related to DNA repair and genomic instability induced by high LET particles (Wang et al. Citation2013). In addition, after comparing fractionated proton radiation with acute proton radiation, it was found that acute proton irradiation up-regulated β-catenin and AKT pathway by increasing proliferation marker phosphate histones, reducing DNA damage repair factors and resulting in an increased risk of cancer (Suman et al. Citation2019). However, due to the complexity of influencing factors, such as LET, radiation dose and dose rate, the epigenetic mechanism correlated with TGF-β induced by different radiation types still needs to be further studied in external irradiation.

Table 3. Effects of different radiation types on epigenetic mechanism regulated by TGF-β.

Internal irradiation

In recent years, targeted radionuclide therapy (TRT) provides a new direction for tumor therapy. Short-distance and high-energy radiation promotes double-strand DNA breaks, leading to cell death and reducing toxicity to surrounding non-cancerous tissues. In addition, when the 225Ac-labeled minigastrin analog 225Ac-PP-F11 was used for targeted alpha therapy (TAT), it was found that it promoted both MAPK pathway phosphorylation and HDAC phosphorylation, and HDAC inhibitors combined with it could enhance radiosensitivity (Qin et al. Citation2023). However, another study using 225Ac found that tubulointerstitial damage induced by α particles, resulting in up-regulated expression of TGF-β, could lead to renal parenchyma damage and loss of renal function (Jaggi et al. Citation2005). It can be seen that the damage of surrounding normal tissue caused by TAT treatment is still the focus of our continuous attention. In addition, it has been shown that the combination of PI3K inhibitor and apigenin increases the radiosensitivity of radioactive iodine in the treatment of thyroid cancer, suggesting that TGF-β may affect the therapeutic effect of radioactive iodine on thyroid cancer through PI3K (Lakshmanan et al. Citation2015). Moreover, the combination of external irradiation and Smad responsive promoter (SMAD-NIS-MSCs)-mediated I-131 therapy induced by TGF-β1 could significantly improve the tumor growth delay and survival time of treated mice, which proved that TGF-β plays an important role in internal irradiation therapy (Schug et al. Citation2019). Although there are still some problems in TRT, for example, the short-range emission of alpha particles can limit the cytotoxic effects on cancerous lesions and the surrounding tumor microenvironment, and it may also damage the surrounding normal tissue, however, this emerging treatment still has great potential and exploration space (Parker et al. Citation2018).

Non-targeted effects

Cells are also affected in a non-targeted manner, known as radiation-induced bystander effects (RIBE), in which irradiated cells release a variety of factors to induce changes in non-irradiated cells that increase radiation toxicity (Bryant et al. Citation2019; Khodamoradi et al. Citation2020). TGF-β signaling can be released from targeted cells and freely diffused in the media, acting in concert with other factors on unirradiated cells, causing micronucleus formation and accelerating cell metastasis (Shao et al. Citation2008; Gu et al. Citation2022). TGF-β plays a key role in the DDR of bystander cells. TGF-β signal significantly up-regulated γ-H2AX foci formation in glioma bystander cells induced by RAD3-related (ATR)-dependent radiation (Burdak-Rothkamm et al. Citation2007). In addition, in RIBE, TGF-β signaling is also associated with a variety of non-coding RNAs (). Hu et al used lncRNA microarray to determine the lncRNA and mRNA expression ­patterns in bystander cells and found that the differentially expressed lncRNAs were closely related to cell proliferation, transformation and migration. Knockdown of lnc-ABCA12-5, lnc-THEMIS-2 or lnc-HPN-AS1 inhibited TGF-β1-induced EMT to different degrees (Hu et al. Citation2018). Some studies have shown that TGF-β induces oxidative stress by upregulating miR-21. Evaluation of bystander mechanisms showed that miR-21 inhibits superoxide dismutase 2 (SOD2) activity and improves the defense ratio of superoxide to antioxidants (Tian et al. Citation2015). Further studies revealed that TGF-β is the most important miR-21 stimulator in bystander cells (Xu et al. Citation2014), and high expression of TGF-β receptors appears to inhibit SOD2 and increase ROS levels through the TGF-β-miR-21-ROS pathway (Jiang et al. Citation2014). Compared with miR-21, miR-663 inhibits TGF-β to reduce DNA damage and micronuclei formation in bystander cells. Interestingly, it has been shown that TGF-β1 downregulation in bystander cells is due to miR-663 induction after cell exposure to TGF-β1 released by irradiated cells (Hu et al. Citation2014). At the same time, other studies have also found that the unique miRNA content in senescence-associated exosomes (SA EXO) mediates the TGF-β pathway to play an important role in the bystander effects of radiation-induced senescence (Lee et al. Citation2023).

Table 4. Non-coding RNA-mediated TGF-β signaling regulation in radiation-induced non-targeted effects.

Some promising treatment strategies for clinical use

The existing preclinical studies on adjuvant drugs for radiotherapy

Radiation oncology is a conventional treatment that aims to maximize the protection of normal tissues while effectively killing tumors. The use of many inhibitors targeting the TGF-β signaling pathway has proven viable. The use of TGF-β1 receptor kinase inhibitors before irradiation reduced DNA damage response, H2AX and p53 phosphorylation, and induction of self-renewal signals Notch1 and CXCR4 to improve tumor treatment response (Hardee et al. Citation2012). TGF-β neutralizing antibodies improved the radiosensitivity of breast tumors, blocked γH2AX lesion formation and led to delayed tumor growth (Bouquet et al. Citation2011). Administration of neutralizing pan-TGF-β antibodies abrogated TGF-β1 upregulation and lung metastasis enhancement due to ionizing radiation or doxorubicin administration (Biswas et al. Citation2007). These results suggest that inhibition of TGF-β is an effective adjunct to radiotherapy for cancer.

More and more studies have shown that targeting epigenetic elements related to TGF-β signaling such as DNA methylation and histone modification is a new direction for tumor treatment (Zheng et al. Citation2018). The DNA methyltransferase inhibitor RG108 increases the radiosensitivity of EC cells, significantly enhancing X-ray-induced apoptosis and G2/M phase arrest. RNA-seq analysis showed that RG108 combined irradiation altered the expression of 121 genes in multiple pathways, including the TGF-β signaling pathway and Epstein-Barr virus infection pathway (Ou et al. Citation2018). Decitabine, a demethylating agent that has been clinically used to treat several cancers, greatly weakens the transcriptional response of hepatoma cells carcinoma (HCCs) to TGF-β and has been shown to induce expression of EMT-related transcription factors (e.g. Snail/2, Zeb1/2). The promoter of Snail is hypomethylated in human HCC with poor prognosis, i.e. associated with high Snail levels, high AFP levels, metastasis, and relapse. Therefore, epilogic medications for HCC should be carefully evaluated as they may activate tumor-promoting pathways (Bévant et al. Citation2021). The radiation protection agent MnTE-2-PyP has been shown to inhibit prostate cancer growth, migration and invasion, and inhibit the expression of three genes regulated by HIF-1β and CREB by altering p300DNA binding: TGF-β2, FGF-1 and PAI-1. Among them, MnTE-2-PyP reduced the binding of the p300 complex to a specific HRE motif in the promoter region of the PAI-1 gene, inhibited H3K9 acetylation, and thus inhibited the expression of PAI-1(Tong et al. Citation2016). However, since known epigenetic changes also regulate some other targets besides TGF-β, we need to consider many aspects when selecting targeted epigenetic inhibitors.

The achievements and challenges of clinical trials

In recent years, some clinical trials on targeting TGF-β have been carried out one after another. Oral administration of small molecular TβRI inhibitors has shown a good effect on tumors in current clinical trials. For example, LY3200882 alone or in combination with PD-L1 inhibitors or gemcitabine was well tolerated in Phase I clinical trials (NCT02937272), and its antineoplastic activity in cancer was initially observed (Yap et al. Citation2021). The combination of galuniserib and gemcitabine improved the overall survival rate of unresectable pancreatic cancer with less side effects (Melisi et al. Citation2018). However, there was no significant advantage in clinical efficacy and safety of galuniserib combined with radiotherapy and chemotherapy in malignant gliomas(NCT01220271) (Wick et al. Citation2020). In view of the availability of galuniserib, trials to evaluate galuniserib combined with durvalumab in the treatment of cancer are still ongoing (NCT02734160). In another prospective randomized trial of TGF-β blockers during radiotherapy, fresolimumab has been shown to promote a good systemic immune response and improve median overall survival in patients with metastatic breast cancer (NCT01401062) (Formenti et al. Citation2018). In addition, phase I clinical trials related to the bifunctional fusion protein formed by the fusion of monoclonal antibody against programmed death ligand 1 (PD-L1) and TGF-β have also proved that it can alleviate advanced solid tumors (NCT04551950, NCT02517398) (Strauss et al. Citation2018; Oaknin et al. Citation2024). Recently, in a clinical trial using TGF-β resistant chimeric receptor (CAR) T cells to target prostate specific membrane antigen (PSMA), cells were engineered with overexpression-dominant-negative TβRII (TGFβRDN) optimization to block TGF-β signaling (NCT03089203). The feasibility and safety of its clinical application were proved in 18 selected patients (Narayan et al. Citation2022). Unfortunately, there seems to be no clear conclusion about drugs targeting epigenetics, such as methylation inhibitors or acetylation inhibitors, in clinical trials that act on TGF-β.

Although, inhibition of TGF-β and its downstream targets has become an ideal treatment strategy. However, most clinical trials of TGF-β targeting remain at Phase I, the pharmacological blockade of TGF-β has not been translated into a successful treatment for humans. This may be due to the pleiotropic effect of TGF-β (Massagué and Sheppard Citation2023). Previous studies have shown that early life exposure to p40 enhances H3K4me1/3 by up-regulating the expression of methyltransferase setd1β, which leads to persistent production of TGF-β by intestinal epithelial cells, expands Tregs and protects the intestine from inflammation (Deng et al. Citation2021). Similarly, TGF-β plays a unique and critical role in pulmonary epithelial and interstitial cells (Saito et al. Citation2018). The absence of TβRII in mesenchyme destroys the morphogenesis of pulmonary branches by regulating Shh signal transduction, resulting in bronchial cystic malformation (Li et al. Citation2008). TGF-β1 deficient mice also showed systemic inflammation and systemic perivasculitis or interstitial pneumonia in the lungs (Shull et al. Citation1992). In addition, mice lacking the expression of TGF-β2 and TGF-β3 also developed defects in the central nervous system (Vogel et al. Citation2010). In addition, preclinical studies of genetic mouse models have shown that the signaling activity of TGF-β is essential for normal skin wound healing (Finnson et al. Citation2013; Kiritsi and Nyström Citation2018). On the other hand, TGF-β is essential for regulating the function of embryonic stem cells (David and Massagué Citation2018). For example, TGF-β activates Smads to promote embryonic stem cell differentiation (Mullen et al. Citation2011); in pluripotent cells, Activin A/Smad2/3 is regulated by PI3K/Akt and is used to maintain self-renewal by activating the target gene (Nanog) (Singh et al. Citation2012). The research on the protective effect of TGF-β on different organs and tissues is an aspect that cannot be ignored. So, we need to consider more carefully when formulating therapeutic strategies targeting TGF-β and its downstream targets in the future.

The future directions of drug development and treatment strategy

A study comparing changes in circulating miRNA profiles after total body irradiation (TBI) and total thoracic lung irradiation (WTLI) found that in TBI, ubiquitination-related pathways, especially histone H2A, were enriched, indicating that it had a greater impact on DNA damage repair mechanisms and apoptosis. However, in WTLI, profibrotic pathways including TGF-β and BMP signaling pathways were enriched. These results suggest that miRNAs can predict the occurrence of different types of radiation-induced damage (Rogers et al. Citation2021). The use of miRNA in clinical treatment can include many directions. First, before patients receive radiotherapy, the expression profile of radiation-related miRNAs in serum is evaluated to (i) predict each patient’s radiological response, (ii) determine individualized radiation doses to optimize treatment outcomes, and (iii) minimize acute and potential damage to normal tissues. Secondly, during radiotherapy, checking the expression profiles of radio-specific and dominant miRNAs in serum and changing the specific miRNA expression can help improve tumor radiosensitivity. Radiotherapy can be used in combination with other chemotherapy drugs, small molecule inhibitors and drugs that target specific miRNAs to activate or inhibit the expression of certain miRNAs and downstream target genes (Khalighfard et al. Citation2022). Finally, when radiotherapy is completed, regular testing of the expression of prognostic miRNAs in serum can help monitor the effectiveness of radiotherapy and reduce the risk of metastasis and cancer recurrence (Zhao et al. Citation2012).

Biomarkers have always played an important role in tumor diagnosis, treatment and prevention. Identification of predictive biomarkers that help identify patients susceptible to radiation-induced lung injury (RILI) can help administer the appropriate IR dose to the patient (Jin et al. Citation2020; Li et al. Citation2023). DJ-1 is a potential therapeutic target for the reversal of RIBE in esophageal squamous cell carcinoma and a useful biomarker for predicting the efficacy of radiotherapy (Gu et al. Citation2022). Altered DNA methylation is an interesting biomarker in cancer. The newly identified CRC metastasis biomarkers, including HLTF (Liu et al. Citation2018), IGFBP-3 (Cai et al. Citation2020), and INHBB, are highly dysregulated by methylation and strongly associated with metastasis (Yuan et al. Citation2020). Overexpression of WTAP in gastric cancer is strongly associated with TGF-β-induced EMT and can serve as a potential predictive biomarker for gastric cancer (Liu and Da Citation2022). Identifying such biomarkers in all types of cancer facilitates earlier identification and more effective treatment for better diagnosis and increased life expectancy (Gutierrez et al. Citation2021).

Conclusion

TGF-β, as a key factor, plays a dual role in the development of tumors and regulates the expression of upstream or downstream factors by acting on DNA methylation, histone modification, chromatin remodeling and non-coding RNA regulation, thereby promoting or inhibiting tumorigenesis and development. Meanwhile, the components of TGF-β signaling are also regulated by different epigenetic mechanisms, which exert a more refined regulation mode on this vital signaling pathway. During radiotherapy, due to the complexity of the tumor microenvironment, different histone modification sites and different non-coding RNAs involved in the epigenetic regulation of TGF-β signaling may have multiple roles in the modulation of tumor development. It has been demonstrated in several reports that radiation-induced hypomethylation or upregulation/downregulation of the expression of certain miRNAs can enhance tumor radiosensitivity by affecting TGF-β signaling, thereby killing tumor cells more efficiently (Zang et al. Citation2018; Liu and Da Citation2022). Several TGF-β inhibitors and demethylating agents have been developed for radiation sensitization (Yang et al. Citation2019; Bévant et al. Citation2021). However, understanding and identifying more methylated or non-coding RNA targets will facilitate the development of more effective targeted drugs and the identification of corresponding predictive biomarkers, which will be of great assistance to the development of adjunctive strategies for radiotherapy.

On the other hand, with the in-depth exploration of non-targeted effects, the radiation damage effects on the normal tissues are also the focus of attention. The effect of TGF-β secreted by irradiated cells on bystander cells to cause damage has been established (Shao et al. Citation2008; Zhang et al. Citation2019). Some radioprotective agents have been developed to reduce RIBE damage, for example, the DNA-binding methylproamine has been shown to repair oxidized DNA damage in RIBE (Burdak-Rothkamm et al. Citation2014). However, the specific mechanism of TGF-β-mediated epigenetic regulation in RIBE has not been fully elucidated. Therefore, it is of great significance for the protection of normal tissues to continue to mine the effective targets of methylation or non-coding RNAs in RIBE.

In conclusion, understanding the mechanism of TGF-β-mediated epigenetic regulation in radiation response, as well as exploring key upstream and downstream factors involved in the epigenetic regulation of TGF-β signaling will be extremely beneficial for the clinical application of both radiotherapy and radiation protection.

Acknowledgements

The authors thank the two reviewers for providing helpful comments and suggestions to improve this manuscript.

Disclosure statement

No potential conflict of interest was reported by the author(s).

Additional information

Funding

This work was funded by the National Natural Science Foundation of China under Grant No. 32071243, 82192882, 82192883 and a project funded by the Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).

Notes on contributors

Yunan Ding

Yunan Ding, PG, is a postgraduate student in the School of Radiation Medicine and Protection at Soochow University.

Guangming Zhou

Guangming Zhou, PhD, is a professor in the School of Radiation Medicine and Protection at Soochow University. His interests include space radiation health risk assessment and protection, molecular mechanisms of radiation carcinogenesis and basic biomedical research related to particle radiation therapy for tumors.

Wentao Hu

Wentao Hu, PhD, is an associate professor in the School of Radiation Medicine and Protection at Soochow University. His interests include the molecular mechanisms of tumorigenesis caused by space radiation, non-coding RNA and radiobiological effects and the composite biological effects of multiple environmental factors in space.

References

  • Abukiwan A, Nwaeburu CC, Bauer N, Zhao Z, Liu L, Gladkich J, Gross W, Benner A, Strobel O, Fellenberg J, et al. 2019. Dexamethasone-induced inhibition of miR-132 via methylation promotes TGF-β-driven progression of pancreatic cancer. Int J Oncol. 54(1):53–64. doi:10.3892/ijo.2018.4616
  • Ahmadi A, Najafi M, Farhood B, Mortezaee K. 2019. Transforming growth factor-β signaling: Tumorigenesis and targeting for cancer therapy. J Cell Physiol. 234(8):12173–12187. doi:10.1002/jcp.27955
  • Al-Assar O, Demiciorglu F, Lunardi S, Gaspar-Carvalho MM, McKenna WG, Muschel RM, Brunner TB. 2014. Contextual regulation of pancreatic cancer stem cell phenotype and radioresistance by pancreatic stellate cells. Radiother Oncol. 111(2):243–251. doi:10.1016/j.radonc.2014.03.014
  • Albert M, Helin K. 2010. Histone methyltransferases in cancer. Semin Cell Dev Biol. 21(2):209–220. doi:10.1016/j.semcdb.2009.10.007
  • Al-Khayal K, Alafeefy A, Vaali-Mohammed MA, Mahmood A, Zubaidi A, Al-Obeed O, Khan Z, Abdulla M, Ahmad R. 2017. Novel derivative of aminobenzenesulfonamide (3c) induces apoptosis in colorectal cancer cells through ROS generation and inhibits cell migration. BMC Cancer. 17(1):4. doi:10.1186/s12885-016-3005-7
  • Altea-Manzano P, Doglioni G, Liu Y, Cuadros AM, Nolan E, Fernández-García J, Wu Q, Planque M, Laue KJ, Cidre-Aranaz F, et al. 2023. A palmitate-rich metastatic niche enables metastasis growth via p65 acetylation resulting in pro-metastatic NF-κB signaling. Nat Cancer. 4(3):344–364. doi:10.1038/s43018-023-00513-2
  • Asaduzzaman M, Constantinou S, Min H, Gallon J, Lin ML, Singh P, Raguz S, Ali S, Shousha S, Coombes RC, et al. 2017. Tumour suppressor EP300, a modulator of paclitaxel resistance and stemness, is downregulated in metaplastic breast cancer. Breast Cancer Res Treat. 163(3):461–474. doi:10.1007/s10549-017-4202-z
  • Asano Y, Ihn H, Yamane K, Kubo M, Tamaki K. 2004. Impaired Smad7-Smurf–mediated negative regulation of TGF-β signaling in scleroderma fibroblasts. J Clin Invest. 113(2):253–264. doi:10.1172/JCI200416269
  • Bévant K, Desoteux M, Abdel Wahab AHA, Abdel Wahab SA, Metwally AM, Coulouarn C. 2021. DNA methylation of tgfβ target genes: Epigenetic control of tgfβ functional duality in liver cancer. Cells. 10(9):2207. doi:10.3390/cells10092207
  • Biswas S, Guix M, Rinehart C, Dugger TC, Chytil A, Moses HL, Freeman ML, Arteaga CL. 2007. Inhibition of TGF-β with neutralizing antibodies prevents radiation-induced acceleration of metastatic cancer progression. J Clin Invest. 117(5):1305–1313. doi:10.1172/JCI30740
  • Boerma M, Wang J, Sridharan V, Herbert JM, Hauer-Jensen M. 2013. Pharmacological induction of transforming growth factor-beta1 in rat models enhances radiation injury in the intestine and the heart. PLOS One. 8(7):e70479. doi:10.1371/journal.pone.0070479
  • Boudreau HE, Ma WF, Korzeniowska A, Park JJ, Bhagwat MA, Leto TL. 2017. Histone modifications affect differential regulation of TGFβ- induced NADPH oxidase 4 (NOX4) by wild-type and mutant p53. Oncotarget. 8(27):44379–44397. doi:10.18632/oncotarget.17892
  • Bouquet F, Pal A, Pilones KA, Demaria S, Hann B, Akhurst RJ, Babb JS, Lonning SM, DeWyngaert JK, Formenti SC, et al. 2011. TGFβ1 inhibition increases the radiosensitivity of breast cancer cells in vitro and promotes tumor control by radiation in vivo. Clin Cancer Res. 17(21):6754–6765. doi:10.1158/1078-0432.CCR-11-0544
  • Bryant J, Shields L, Hynes C, Howe O, McCleanc B, Lynga F. 2019. DNA damage and cytokine production in non-target irradiated lymphocytes. Radiat Res. 191(6):545–555. doi:10.1667/RR15165.1
  • Burdak-Rothkamm S, Short SC, Folkard M, Rothkamm K, Prise KM. 2007. ATR-dependent radiation-induced γH2AX foci in bystander primary human astrocytes and glioma cells. Oncogene. 26(7):993–1002. doi:10.1038/sj.onc.1209863
  • Burdak-Rothkamm S, Smith A, Lobachevsky P, Martin R, Prise KM. 2014. Radioprotektive Wirkung von Methylproamine auf direkt bestrahlte Zellen und Bystander-Zellen. Strahlenther Onkol. 191(3):248–255. doi:10.1007/s00066-014-0751-9
  • Cai Q, Dozmorov M, Oh Y. 2020. IGFBP-3/IGFBP-3 receptor system as an anti-tumor and anti-metastatic signaling in cancer. Cells. 9(5):1261. doi:10.3390/cells9051261
  • Camerlingo R, Miceli R, Marra L, Rea G, D'Agnano I, Nardella M, Montella R, Morabito A, Normanno N, Tirino V, et al. 2019. Conditioned medium of primary lung cancer cells induces EMT in A549 lung cancer cell line by TGF-ß1 and miRNA21 cooperation. PLOS One. 14(7):e0219597. doi:10.1371/journal.pone.0219597
  • Cammareri P, Vincent DF, Hodder MC, Ridgway RA, Murgia C, Nobis M, Campbell AD, Varga J, Huels DJ, Subramani C, et al. 2017. TGFβ pathway limits dedifferentiation following WNT and MAPK pathway activation to suppress intestinal tumourigenesis. Cell Death Differ. 24(10):1681–1693. doi:10.1038/cdd.2017.92
  • Carbajo‐García MC, de Miguel‐Gómez L, Juárez‐Barber E, Trelis A, Monleón J, Pellicer A, Flanagan JM, Ferrero H. 2022. Deciphering the role of histone modifications in uterine leiomyoma: acetylation of H3K27 regulates the expression of genes involved in proliferation, cell signaling, cell transport, angiogenesis and extracellular matrix formation. Biomedicines. 10(6):1279. doi:10.3390/biomedicines10061279
  • Carl C, Flindt A, Hartmann J, Dahlke M, Rades D, Dunst J, Lehnert H, Gieseler F, Ungefroren H. 2016. Ionizing radiation induces a motile phenotype in human carcinoma cells in vitro through hyperactivation of the TGF-beta signaling pathway. Cell Mol Life Sci. 73(2):427–443. doi:10.1007/s00018-015-2003-2
  • Chae DK, Park J, Cho M, Ban E, Jang M, Yoo YS, Kim EEK, Baik JH, Song EJ. 2019. MiR-195 and miR-497 suppress tumorigenesis in lung cancer by inhibiting SMURF2-induced TGF-β receptor I ubiquitination. Mol Oncol. 13(12):2663–2678. doi:10.1002/1878-0261.12581
  • Chen H, Yang T, Lei Z, Wang L, Yang H, Tong X, Yang WT, Zhao J, Gu Y, Chen Y, et al. 2015. RNF111/Arkadia is regulated by DNA methylation and affects TGF-β/Smad signaling associated invasion in NSCLC cells. Lung Cancer. 90(1):32–40. doi:10.1016/j.lungcan.2015.07.010
  • Chen S, Dai X, Li H, Gong Y, Zhao Y, Huang H. 2021. Overexpression of ring finger protein 20 inhibits the progression of liver fibrosis via mediation of histone H2B lysine 120 ubiquitination. Hum Cell. 34(3):759–770. doi:10.1007/s13577-021-00498-z
  • Chen S, Xu J, Su Y, Hua L, Feng C, Lin Z, Huang H, Li Y. 2020. MicroRNA-145 suppresses epithelial to mesenchymal transition in pancreatic cancer cells by inhibiting TGF-β signaling pathway. J Cancer. 11(9):2716–2723. doi:10.7150/jca.34902
  • Chen Z, Chen Y, Li Y, Lian W, Zheng K, Zhang Y, Zhang Y, Lin C, Liu C, Sun F, et al. 2021. Prrx1 promotes stemness and angiogenesis via activating TGF-β/smad pathway and upregulating proangiogenic factors in glioma. Cell Death Dis. 12(6):615. doi:10.1038/s41419-021-03882-7
  • Concepcion CP, Ma S, Lafave LM, Bhutkar A, Liu M, Deangelo LP, Kim JY, Priore ID, Schoenfeld AJ, Miller M, et al. 2022. Smarca4 inactivation promotes lineage-specific transformation and early metastatic features in the lung. Cancer Discov. 12(2):562–585. doi:10.1158/2159-8290.CD-21-0248
  • Cui H, Yi H, Bao H, Tan Y, Tian C, Shi X, Gan D, Zhang B, Liang W, Chen R, et al. 2022. The SWI/SNF chromatin remodeling factor DPF3 regulates metastasis of ccRCC by modulating TGF-β signaling. Nat Commun. 13(1):4680. doi:10.1038/s41467-022-32472-0
  • Dadrich M, Nicolay NH, Flechsig P, Bickelhaupt S, Hoeltgen L, Roeder F, Hauser K, Tietz A, Jenne J, Lopez R, et al. 2016. Combined inhibition of TGFβ and PDGF signaling attenuates radiation-induced pulmonary fibrosis. Oncoimmunology. 5(5):e1123366. doi:10.1080/2162402X.2015.1123366
  • David CJ, Massagué J. 2018. Contextual determinants of TGFβ action in development, immunity and cancer. Nat Rev Mol Cell Biol. 19(7):419–435. doi:10.1038/s41580-018-0007-0
  • de Ceuninck van Capelle C, Spit M, ten Dijke P. 2020. Current perspectives on inhibitory SMAD7 in health and disease. Crit Rev Biochem Mol Biol. 55(6):691–715. doi:10.1080/10409238.2020.1828260
  • Deng Y, McDonald OG, Means AL, Peek RM, Washington MK, Acra SA, Polk DB, Yan F. 2021. Exposure to p40 in early life prevents intestinal inflammation in adulthood through inducing a long-lasting epigenetic imprint on TGFβ. Cell Mol Gastroenterol Hepatol. 11(5):1327–1345. doi:10.1016/j.jcmgh.2021.01.004
  • Derynck R, Zhang YE. 2003. Smad-dependent and Smad-independent pathways in TGF-b family signalling. Nature. 425(6958):577–584. doi:10.1038/nature02006
  • Duru N, Zhang Y, Gernapudi R, Wolfson B, Lo PK, Yao Y, Zhou Q. 2016. Loss of miR-140 is a key risk factor for radiation-induced lung fibrosis through reprogramming fibroblasts and macrophages. Sci Rep. 6(1):39572. doi:10.1038/srep39572
  • Espino PS, Pritchard S, Heng HHQ, Davie JR. 2009. Genomic instability and histone H3 phosphorylation induction by the Ras-mitogen activated protein kinase pathway in pancreatic cancer cells. Int J Cancer. 124(3):562–567. doi:10.1002/ijc.23959
  • Evanno E, Godet J, Piccirilli N, Guilhot J, Milin S, Gombert JM, Fouchaq B, Roche J. 2017. Tri-methylation of H3K79 is decreased in TGF-β1-induced epithelial-to-mesenchymal transition in lung cancer. Clin Epigenet. 9(1):80. doi:10.1186/s13148-017-0380-0
  • Fang C, Dai CY, Mei Z, Jiang MJ, Gu DN, Huang Q, Tian L. 2018. MicroRNA-193a stimulates pancreatic cancer cell repopulation and metastasis through modulating TGF-β2/TGF-βRIII signalings. J Exp Clin Cancer Res. 37(1):25. doi:10.1186/s13046-018-0697-3
  • Farhood B, Khodamoradi E, Hoseini-Ghahfarokhi M, Motevaseli E, Mirtavoos-Mahyari H, Eleojo Musa A, Najafi M. 2020. TGF-β in radiotherapy: Mechanisms of tumor resistance and normal tissues injury. Pharmacol Res. 155(2020):104745. doi:10.1016/j.phrs.2020.104745
  • Feng F, Zhao Z, Cai X, Heng X, Ma X. 2022. Cyclin-dependent kinase subunit2 (CKS2) promotes malignant phenotypes and epithelial-mesenchymal transition-like process in glioma by activating TGFβ/SMAD signaling. Cancer Med. 12(5):5889–5907. doi:10.1002/cam4.5381
  • Feng XH, Derynck R. 2005. Specificity and versatility in TGF-β signaling through smads. Annu Rev Cell Dev Biol. 21(1):659–693. doi:10.1146/annurev.cellbio.21.022404.142018
  • Finnson KW, Arany PR, Philip A. 2013. Transforming growth factor beta signaling in cutaneous wound healing: lessons learned from animal studies. Adv Wound Care. 2(5):225–237. doi:10.1089/wound.2012.0419
  • Formenti SC, Lee P, Adams S, Goldberg JD, Li X, Xie MW, Ratikan JA, Felix C, Hwang L, Faull KF, et al. 2018. Focal irradiation and systemic TGFb blockade in metastatic breast cancer. Clin Cancer Res. 24(11):2493–2504. doi:10.1158/1078-0432.CCR-17-3322
  • Fu J, Jiang M, Zhang M, Zhang J, Wang Y, Xiang S, Xu X, Ye Q, Song H. 2016. MiR-495 functions as an adjuvant to radiation therapy by reducing the radiation-induced bystander effect. Acta Biochim Biophys Sin. 48(11):1026–1033. doi:10.1093/abbs/gmw098
  • Fu L, Cui CP, Zhang X, Zhang L. 2019. The functions and regulation of Smurfs in cancers. Semin Cancer Biol. 67(Pt 2):102–116. doi:10.1016/j.semcancer.2019.12.023
  • Gao L, Zhou F. 2021. Comprehensive analysis of RUNX and TGF-β mediated regulation of immune cell infiltration in breast cancer. Front Cell Dev Biol. 9:730380. doi:10.3389/fcell.2021.730380
  • Gardner EE, Benjamin L, Schneeberger VE, Desmeules P, Miles LA, Arnold PK, Ni A, Khodos I, de Stanchina E, Nguyen T, et al. 2017. Chemosensitive relapse in small cell lung cancer proceeds through an EZH2-SLFN11 axis. Cancer Cell. 31(2):286–299. doi:10.1016/j.ccell.2017.01.006
  • Genao I, Hendrickson K, Diers D, Browne R, Rawlings JE. 2002. The polycomb group protein EZH2 is involved in progression of prostate cancer. Nature. 419:624–629. doi:10.1038/nature01042.1
  • Greer EL, Yang S. 2014. Histone methylation: a dynamic mark in health, disease and inheritance. Nat Rev Genet. 13(5):343–357. doi:10.1038/nrg3173
  • Grunstein M. 1997. Histone acetylation in chromatin structure and transcription. Nature. 389(6649):349–352. doi:10.1038/38664
  • Gu J, Sun Y, Song J, Zhao R, Di X, Zhang Y, Ge X, Zhang S, Gu Y, Sun X. 2022. Irradiation induces DJ-1 secretion from esophageal squamous cell carcinoma cells to accelerate metastasis of bystander cells via a TGF-β1 positive feedback loop. J Exp Clin Cancer Res. 41(1):259. doi:10.1186/s13046-022-02471-6
  • Gutierrez A, Demond H, Brebi P, Ili CG. 2021. Novel methylation biomarkers for colorectal cancer prognosis. Biomolecules. 11(11):1722. doi:10.3390/biom11111722
  • Han B, Fang T, Wang Y, Zhang Y, Xue Y. 2022. TGFβ2 is a prognostic biomarker for gastric cancer and is associated with methylation and immunotherapy responses. Front Genet. 13(5):808041. doi:10.3389/fgene.2022.808041
  • Hardee ME, Marciscano AE, Medina-Ramirez CM, Zagzag D, Narayana A, Lonning SM, Barcellos-Hoff MH. 2012. Resistance of glioblastoma-initiating cells to radiation mediated by the tumor microenvironment can be abolished by inhibiting transforming growth factor-β. Cancer Res. 72(16):4119–4129. doi:10.1158/0008-5472.CAN-12-0546
  • Haydont V, Bourgier C, Vozenin-Brotons MC. 2007. Rho/ROCK pathway as a molecular target for modulation of intestinal radiation-induced toxicity. BJR. 80(1):S32–S40. doi:10.1259/bjr/58514380
  • Hritzo B, Aghdam SY, Legesse B, Kaur A, Cao M, Boerma M, Chakraborty N, Dimitrov G, Gautam A, Hammamieh R, et al. 2021. Late health effects of partial body irradiation injury in a minipig model are associated with changes in systemic and cardiac IGF‐1 signaling. Int J Mol Sci. 22(6):3286. doi:10.3390/ijms22063286
  • Hu W, Pei W, Zhu L, Nie J, Pei H, Zhang J, Li B, Hei TK, Zhou G. 2018. Microarray profiling of TGF-β1-induced long non-coding RNA expression patterns in human lung bronchial epithelial BEAS-2B cells. Cell Physiol Biochem. 50(6):2071–2085. doi:10.1159/000495052
  • Hu W, Xu S, Yao B, Hong M, Wu X, Pei H, Chang L, Ding N, Gao X, Ye C, et al. 2014. MiR-663 inhibits radiation-induced bystander effects by targeting TGFB1 in a feedback mode. RNA Biol. 11(9):1189–1198. doi:10.4161/rna.34345
  • Huang G, Du M. y, Zhu H, Zhang N, Lu ZW, Qian LX, Zhang W, Tian X, He X, Yin L. 2018. MiRNA-34a reversed TGF-β-induced epithelial-mesenchymal transition via suppression of SMAD4 in NPC cells. Biomed Pharmacother. 106(2018):217–224. doi:10.1016/j.biopha.2018.06.115
  • Huang H, Zhang Y, Gallegos V, Sorrelle N, Zaid MM, Toombs J, Du W, Wright S, Hagopian M, Wang Z, et al. 2019. Targeting TGF βR2‐mutant tumors exposes vulnerabilities to stromal TGF β blockade in pancreatic cancer. EMBO Mol Med. 11(11):e10515. doi:10.15252/emmm.201910515
  • Humeres C, Shinde AV, Hanna A, Alex L, Hernández SC, Li R, Chen B, Conway SJ, Frangogiannis NG. 2022. Smad7 effects on TGF-β and ErbB2 restrain myofibroblast activation and protect from postinfarction heart failure. J Clin Investig. 132(3):e146926. doi:10.1172/JCI146926
  • Ibuki Y, Toyooka T, Zhao X, Yoshida I. 2014. Cigarette sidestream smoke induces histone H3 phosphorylation via JNK and PI3K/Akt pathways, leading to the expression of proto-oncogenes. Carcinogenesis. 35(6):1228–1237. doi:10.1093/carcin/bgt492
  • Ippolito L, Comito G, Parri M, Iozzo M, Duatti A, Virgilio F, Lorito N, Bacci M, Pardella E, Sandrini G, et al. 2022. Lactate rewires lipid metabolism and sustains a metabolic-epigenetic axis in prostate cancer. Cancer Res. 82(7):1267–1282. doi:10.1158/0008-5472.CAN-21-0914
  • Jaggi JS, Seshan SV, McDevitt MR, LaPerle K, Sgouros G, Scheinberg DA. 2005. Renal tubulointerstitial changes after internal irradiation with α-particle-emitting actinium daughters. J Am Soc Nephrol. 16(9):2677–2689. doi:10.1681/ASN.2004110945
  • Jdeed S, Lengyel M, Uray IP. 2022. Redistribution of the SWI/SNF complex dictates coordinated transcriptional control over epithelial–mesenchymal transition of normal breast cells through TGF-β Signaling. Cells. 11(17):2633. doi:10.3390/cells11172633
  • Jiang Y, Chen X, Tian W, Yin X, Wang J, Yang H. 2014. The role of TGF-β1-miR-21-ROS pathway in bystander responses induced by irradiated non-small-cell lung cancer cells. Br J Cancer. 111(4):772–780. doi:10.1038/bjc.2014.368
  • Jin H, Yoo Y, Kim Y, Kim Y, Cho J, Lee YS. 2020. Radiation-induced lung fibrosis: Preclinical animal models and therapeutic strategies. Cancers. 12(6):1561. doi:10.3390/cancers12061561
  • Jin Y, Park S, Park SY, Lee CY, Eum DY, Shim JW, Choi SH, Choi YJ, Park SJ, Heo K. 2022. G9a Knockdown suppresses cancer aggressiveness by facilitating smad protein phosphorylation through increasing BMP5 expression in luminal a type breast cancer. Int J Mol Sci. 23(2):589. doi:10.3390/ijms23020589
  • Kang H, Seo E, Oh YS, Jun HS. 2022. TGF-β activates NLRP3 inflammasome by an autocrine production of TGF-β in LX-2 human hepatic stellate cells. Mol Cell Biochem. 477(5):1329–1338. doi:10.1007/s11010-022-04369-5
  • Kaowinn S, Kaewpiboon C, Koh SS, Krämer OH, Chung YH. 2018. STAT1-HDAC4 signaling induces epithelial-mesenchymal transition and sphere formation of cancer cells overexpressing the oncogene, CUG2. Oncol Rep. 40(5):2619–2627. doi:10.3892/or.2018.6701
  • Khalighfard S, Kalhori MR, Amiriani T, Poorkhani A, Khori V, Esmati E, Lashkari M, Najafi A, Alizadeh AM. 2022. A systematic approach introduced novel targets in rectal cancer by considering miRNA/mRNA interactions in response to radiotherapy. Cancer Biomark. 33(1):97–110. doi:10.3233/CBM-210079
  • Khodamoradi E, Hoseini-Ghahfarokhi M, Amini P, Motevaseli E, Shabeeb D, Musa AE, Najafi M, Farhood B. 2020. Targets for protection and mitigation of radiation injury. Cell Mol Life Sci. 77(16):3129–3159. doi:10.1007/s00018-020-03479-x
  • Kiritsi D, Nyström A. 2018. The role of TGFβ in wound healing pathologies. Mech Ageing Dev. 172:51–58. doi:10.1016/j.mad.2017.11.004
  • Kirshner J, Jobling MF, Pajares MJ, Ravani SA, Glick AB, Lavin MJ, Koslov S, Shiloh Y, Barcellos-Hoff MH. 2006. Inhibition of transforming growth factor-β1 signaling attenuates ataxia telangiectasia mutated activity in response to genotoxic stress. Cancer Res. 66(22):10861–10869. doi:10.1158/0008-5472.CAN-06-2565
  • Kleer CG, Cao Q, Varambally S, Shen R, Ota I, Tomlins SA, Ghosh D, Sewalt RGAB, Otte AP, Hayes DF, et al. 2003. EZH2 is a marker of aggressive breast cancer and promotes neoplastic transformation of breast epithelial cells. Proc Natl Acad Sci U S A. 100(20):11606–11611. doi:10.1073/pnas.1933744100
  • Lakshmanan A, Scarberry D, Green JA, Zhang X, Selmi-Ruby S, Jhiang SM. 2015. Modulation of thyroidal radioiodide uptake by oncological pipeline inhibitors and Apigenin. Oncotarget. 6(31):31792–31804. http://www.impactjournals.com/oncotarget/
  • Lee AH, Ghosh D, Koh IL, Dawson MR. 2023. Senescence-associated exosomes transfer miRNA-induced fibrosis to neighboring cells. Aging. 15(5):1237–1256. doi:10.18632/aging.204539
  • Li J, Wang R, Shi W, Chen X, Yi J, Yang X, Jin S. 2023. Epigenetic regulation in radiation-induced pulmonary fibrosis. Int J Radiat Biol. 99(3):384–395. doi:10.1080/09553002.2022.2089365
  • Li M, Li C, Liu YH, Xing Y, Hu L, Borok Z, Kwong KYC, Minoo P. 2008. Mesodermal deletion of transforming growth factor-β receptor II disrupts lung epithelial morphogenesis: Cross-talk between TGF-β and Sonic hedgehog pathways. J Biol Chem. 283(52):36257–36264. doi:10.1074/jbc.M806786200
  • Li Z, Dong M, Fan D, Hou P, Li H, Liu L, Lin C, Liu J, Su L, Wu L, et al. 2017. LncRNA ANCR down-regulation promotes TGF-β-induced EMT and metastasis in breast cancer. Oncotarget. 8(40):67329–67343. doi:10.18632/oncotarget.18622
  • Li Z, Li M, Xia P, Wang L, Lu Z. 2022. Targeting long non-coding RNA PVT1/TGF-β/Smad by p53 prevents glioma progression. Cancer Biol Ther. 23(1):225–233. doi:10.1080/15384047.2022.2042160
  • Li Z, Xia J, Fang M, Xu Y. 2019. Epigenetic regulation of lung cancer cell proliferation and migration by the chromatin remodeling protein BRG1. Oncogenesis. 8(11):66. doi:10.1038/s41389-019-0174-7
  • Liang G, Weisenberger DJ. 2017. DNA methylation aberrancies as a guide for surveillance and treatment of human cancers. Epigenetics. 12(6):416–432. doi:10.1080/15592294.2017.1311434
  • Lin X, Chai G, Wu Y, Li J, Chen F, Liu J, Luo G, Tauler J, Du J, Lin S, et al. 2019. RNA m6A methylation regulates the epithelial mesenchymal transition of cancer cells and translation of Snail. Nat Commun. 10(1):2065. doi:10.1038/s41467-019-09865-9
  • Liu C, Li B, Cheng Y, Lin J, Hao J, Zhang S, Mitchel REJ, Sun D, Ni J, Zhao L, et al. 2011. MiR-21 plays an important role in radiation induced carcinogenesis in BALB/c mice by directly targeting the tumor suppressor gene Big-h3. Int J Biol Sci. 7(3):347–363. doi:10.7150/ijbs.7.347
  • Liu L, Liu H, Zhou Y, He J, Liu Q, Wang J, Zeng M, Yuan D, Tan F, Zhou Y, et al. 2018. HLTF suppresses the migration and invasion of colorectal cancer cells via TGF-β/SMAD signaling in vitro. Int J Oncol. 53(6):2780–2788. doi:10.3892/ijo.2018.4591
  • Liu Q, Borcherding N, Shao P, Cao H, Zhang W, Qi HH. 2019. Identification of novel TGF-β regulated genes with pro-migratory roles. Biochim Biophys Acta Mol Basis Dis. 1865(12):165537. doi:10.1016/j.bbadis.2019.165537
  • Liu Q, Ma L, Jones T, Palomero L, Pujana MA, Haydeliz Martinez-Ruiz PH, Murnane J, Cuartas I, Seoane J, Baumann M, et al. 2018. Subjugation of TGFβ signaling by human papilloma virus in head and neck squamous cell carcinoma shifts DNA repair from homologous recombination to alternative end-joining. Clin Cancer Res. 24(23):6001–6014. doi:10.1158/1078-0432.CCR-18-1346
  • Liu Y, Da M. 2022. Wilms tumor 1 associated protein promotes epithelial mesenchymal transition of gastric cancer cells by accelerating TGF-β and enhances chemoradiotherapy resistance. J Cancer Res Clin Oncol. 149(7):3977–3988. doi:10.1007/s00432-022-04320-7
  • Liu YL, Yang WH, Chen BY, Nie J, Su ZR, Zheng JN, Gong ST, Chen JN, Jiang D, Li Y. 2021. miR-29b suppresses proliferation and induces apoptosis of hepatocellular carcinoma ascites H22 cells via regulating TGF-β1 and p53 signaling pathway. Int J Mol Med. 48(2):1–11. doi:10.3892/ijmm.2021.4990
  • Lo Roger S, Chen Ye G, Shi Y, Pavletich NP, Massagué J, Lo RS, Chen Y-G. 1998. The L3 loop: a structural motif determining specific interactions between SMAD proteins and TGF-β receptors. Embo J. 17(4):996–1005. doi:10.1093/emboj/17.4.996
  • Lu J, Zhong Y, Chen J, Lin X, Lin Z, Wang N, Lin S. 2018. Radiation enhances the epithelial-mesenchymal transition of A549 cells via miR3591-5p/USP33/PPM1A. Cell Physiol Biochem. 50(2):721–733. doi:10.1159/000494238
  • Macias MJ, Martin-Malpartida P, Massagué J. 2015. Structural determinants of Smad function in TGF-β signaling. Trends Biochem Sci. 40(6):296–308. doi:10.1016/j.tibs.2015.03.012
  • Maliekal TT, Anto RJ, Karunagaran D. 2004. Differential activation of Smads in HeLa and SiHa cells that differ in their response to transforming growth factor-β. J Biol Chem. 279(35):36287–36292. doi:10.1074/jbc.M404568200
  • Massagué J, Sheppard D. 2023. TGF-β signaling in health and disease. Cell. 186(19):4007–4037. doi:10.1016/j.cell.2023.07.036
  • Massagué J. 2008. TGFβ in cancer. Cell. 134(2):215–230. doi:10.1016/j.cell.2008.07.001
  • Melisi D, Garcia-Carbonero R, Macarulla T, Pezet D, Deplanque G, Fuchs M, Trojan J, Oettle H, Kozloff M, Cleverly A, et al. 2018. Galunisertib plus gemcitabine vs. gemcitabine for first-line treatment of patients with unresectable pancreatic cancer. Br J Cancer. 119(10):1208–1214. doi:10.1038/s41416-018-0246-z
  • Mezawa Y, Wang T, Daigo Y, Takano A, Miyagi Y, Yokose T, Yamashita T, Yang L, Maruyama R, Seimiya H, et al. 2023. Glutamine deficiency drives transforming growth factor-β signaling activation that gives rise to myofibroblastic carcinoma-associated fibroblasts. Cancer Sci. 114(11):4376–4387. doi:10.1111/cas.15955
  • Miao C, Liang C, Li P, Liu B, Qin C, Yuan H, Liu Y, Zhu J, Cui Y, Xu A, et al. 2021. TRIM37 orchestrates renal cell carcinoma progression via histone H2A ubiquitination-dependent manner. J Exp Clin Cancer Res. 40(1):1–16. doi:10.1186/s13046-021-01980-0
  • Mishra R, Haldar S, Biondi S, Bhari VK, Singh G, Bhowmick NA. 2022. TGF-β controls stromal telomere length through epigenetic modifications. 3 Biotech. 12(11):1–14. doi:10.1007/s13205-022-03346-5
  • Mortezaee K. 2021. Enriched cancer stem cells, dense stroma, and cold immunity: Interrelated events in pancreatic cancer. J Biochem Mol Tox. 35(4):1–8. doi:10.1002/jbt.22708
  • Mullen AC, Orlando DA, Newman JJ, Lovén J, Kumar RM, Bilodeau S, Reddy J, Guenther MG, Dekoter RP, Young RA. 2011. Master transcription factors determine cell-type-specific responses to TGF-β signaling. Cell. 147(3):565–576. doi:10.1016/j.cell.2011.08.050
  • Murayama K, Kato-Murayama M, Itoh Y, Miyazono K, Miyazawa K, Shirouzu M. 2020. Structural basis for inhibitory effects of Smad7 on TGF-β family signaling. J Struct Biol. 212(3):107661. doi:10.1016/j.jsb.2020.107661
  • Nagata A, Itoh F, Sasho A, Sugita K, Suzuki R, Hinata H, Shimoda Y, Suzuki E, Maemoto Y, Inagawa T, et al. 2020. The evolutionarily conserved deubiquitinase UBH1/UCH-L1 augments DAF7/TGF-b signaling, inhibits dauer larva formation, and enhances lung tumorigenesis. J Biol Chem. 295(27):9105–9120. doi:10.1074/jbc.RA119.011222
  • Najafi M, Majidpoor J, Toolee H, Mortezaee K. 2021. The current knowledge concerning solid cancer and therapy. J Biochem Mol Tox. 35(11):1–26. doi:10.1002/jbt.22900
  • Nakayama KI, Nakayama K. 2006. Ubiquitin ligases: cell-cycle control and cancer. Nat Rev Cancer. 6(5):369–381. doi:10.1038/nrc1881
  • Narayan V, Barber-Rotenberg JS, Jung IY, Lacey SF, Rech AJ, Davis MM, Hwang WT, Lal P, Carpenter EL, Maude SL, et al. 2022. PSMA-targeting TGFβ-insensitive armored CAR T cells in metastatic castration-resistant prostate cancer: a phase 1 trial. Nat Med. 28(4):724–734. doi:10.1038/s41591-022-01726-1
  • Nishizuka M, Komada R, Imagawa M. 2019. Knockdown of RhoE expression enhances TGF-β-induced EMT (epithelial-to-mesenchymal transition) in cervical cancer HeLa cells. IJMS. 20(19):4697. doi:10.3390/ijms20194697
  • Oaknin A, Ghamande SA, Kasamatsu Y, Gil-Martin M, Grau-Bejar F, Garcia-Duran C, Sato M, Siddiqui A, Pal S, Vugmeyster Y, et al. 2024. Phase 1 trial of first-line bintrafusp alfa in patients with locally advanced or persistent/recurrent/metastatic cervical cancer. Clin Cancer Res. 30(5):975–983. doi:10.13039/100009945
  • Ou Y, Zhang Q, Tang Y, Lu Z, Lu X, Zhou X, Liu C. 2018. DNA methylation enzyme inhibitor RG108 suppresses the radioresistance of esophageal cancer. Oncol Rep. 39(3):993–1002. doi:10.3892/or.2018.6210
  • Pardali K, Moustakas A. 2007. Actions of TGF-β as tumor suppressor and pro-metastatic factor in human cancer. Biochim Biophys Acta Rev Cancer. 1775(1):21–62. doi:10.1016/j.bbcan.2006.06.004
  • Parker C, Lewington V, Shore N, Kratochwil C, Levy M, Lindén O, Noordzij W, Park J, Saad F. 2018. Targeted alpha therapy, an emerging class of cancer agents: a review. JAMA Oncol. 4(12):1765–1772. doi:10.1001/jamaoncol.2018.4044
  • Qiao Y, Wang Z, Tan F, Chen J, Lin J, Yang J, Li H, Wang X, Sali A, Zhang L, et al. 2020. Enhancer reprogramming within pre-existing topologically associated domains promotes TGF-β-induced EMT and cancer metastasis. Mol Ther. 28(9):2083–2095. doi:10.1016/j.ymthe.2020.05.026
  • Qin Y, Imobersteg S, Frank S, Blanc A, Chiorazzo T, Berger P, Schibli R, Béhé MP, Grzmil M. 2023. Signaling network response to a-particle-targeted therapy with the 225Ac-labeled minigastrin analog 225Ac-PP-F11N reveals the radiosensitizing potential of histone deacetylase inhibitors. J Nucl Med. 64(6):873–879. doi:10.2967/jnumed.122.264597
  • Qiu Y, Gao Y, Yu D, Zhong L, Cai W, Ji J, Geng F, Tang G, Zhang H, Cao J, et al. 2020. Genome-wide analysis reveals zinc transporter ZIP9 regulated by DNA methylation promotes radiation-induced skin fibrosis via the TGF-β signaling pathway. J Invest Dermatol. 140(1):94–102.e7. doi:10.1016/j.jid.2019.04.027
  • Qu P, Shao ZA, Wang B, He JP, Zhang YN, Wei WJ, Hua JR, Zhou H, Lu D, Ding N, et al. 2022. MiR-663a inhibits radiation-induced epithelium-to-mesenchymal transition by targeting TGF-β1. Biomed Environ Sci. 35(5):437–447. doi:10.3967/bes2022.059
  • Quan HY, Yuan T, Hao JF. 2016. A microRNA-125a variant, which affects its mature processing, increases the risk of radiation-induced pneumonitis in patients with non-small-cell lung cancer. Mol Med Rep. 18(4):4079–4086. doi:10.3892/mmr.2018.9406
  • Rajan V, Pandey BN. 2021. Cytoproliferative effect of low dose alpha radiation in human lung cancer cells is associated with connexin 43, caveolin-1, and survivin pathway. Int J Radiat Biol. 97(3):356–366. doi:10.1080/09553002.2021.1864044
  • Ran J, Wang J, Dai Z, Miao Y, Gan J, Zhao C, Guan Q. 2021. Irradiation-induced changes in the immunogenicity of lung cancer cell lines: based on comparison of X-rays and carbon ions. Front Public Health. 9:1–11. doi:10.3389/fpubh.2021.666282
  • Rogers CJ, Kyubwa EM, Lukaszewicz AI, Starbird MA, Nguyen M, Copeland BT, Yamada-Hanff J, Menon N. 2021. Observation of unique circulating miRNA signatures in non-human primates exposed to total-body vs. whole thorax lung irradiation. Radiat Res. 196(5):547–559. doi:10.1667/RADE-21-00043.1
  • Russell NS, Floot B, Van Werkhoven E, Schriemer M, De Jong-Korlaar R, Woerdeman LAE, Stewart FA, Scharpfenecker M. 2015. Blood and lymphatic microvessel damage in irradiated human skin: The role of TGF-β, endoglin and macrophages. Radiother Oncol. 116(3):455–461. doi:10.1016/j.radonc.2015.08.024
  • Ryu SH, Park EY, Kwak S, Heo SH, Ryu JW, Park JH, Choi KC, Lee SW. 2016. Protective effect of a-lipoic acid against radiation-induced fibrosis in mice. Oncotarget. 7(13):15554–15565. doi:10.18632/oncotarget.6952
  • Saito A, Horie M, Nagase T. 2018. TGF-β signaling in lung health and disease. IJMS. 19(8):2460. doi:10.3390/ijms19082460
  • Saito A, Suzuki HI, Horie M, Ohshima M, Morishita Y, Abiko Y, Nagase T. 2013. An integrated expression profiling reveals target genes of TGF-β and TNF-α possibly mediated by MicroRNAs in lung cancer cells. PLOS One. 8(2):e56587. doi:10.1371/journal.pone.0056587
  • Saitoh M. 2022. Epithelial–mesenchymal transition by synergy between transforming growth factor-β and growth factors in cancer progression. Diagnostics. 12(9):2127. doi:10.3390/diagnostics12092127
  • Schug C, Kitzberger C, Sievert W, Spellerberg R, Tutter M, Schmohl KA, Eberlein B, Biedermann T, Steiger K, Zach C, et al. 2019. Radiation-induced amplification of TGFb1-induced mesenchymal stem cell-mediated sodium iodide symporter (NIS) gene 131I therapy. Clin Cancer Res. 25(19):5997–6008. doi:10.1158/1078-0432.CCR-18-4092
  • Senft D, Qi J, Rona ZA. 2018. Ubiquitin ligases in oncogenic transformation and cancer therapy. Nat Rev Cancer. 18(2):69–88. doi:10.1053/j.gastro.2016.08.014.CagY
  • Shao C, Folkard M, Prise KM. 2008. Role of TGF-B1 and nitric oxide in the bystander response of irradiated glioma cells. Oncogene. 27(4):434–440. doi:10.1038/sj.onc.1210653
  • Shi Y, Massagué J. 2003. Mechanisms of TGF-β signaling from cell membrane to the nucleus. Cell. 113(6):685–700. doi:10.1016/S0092-8674(03)00432-X
  • Shree B, Tripathi S, Sharma V. 2022. Transforming growth factor-beta-regulated LncRNA-MUF promotes invasion by modulating the miR-34a snail1 axis in glioblastoma multiforme. Front Oncol. 11:788755. doi:10.3389/fonc.2021.788755
  • Shull MM, Ormsby I, Kier AB, Pawlowski S, Diebold RJ, Yin M, Allen R, Sidman C, Proetzel G, Calvin D, et al. 1992. Targeted disruption of the mouse transforming growth factor-beta 1 gene results in multifocal inflammatory disease. Nature. 359(6397):693–699. doi:10.1038/359693a0
  • Singh AM, Reynolds D, Cliff T, Ohtsuka S, Mattheyses AL, Sun Y, Menendez L, Kulik M, Dalton S. 2012. Signaling network crosstalk in human pluripotent cells: A Smad2/3-regulated switch that controls the balance between self-renewal and differentiation. Cell Stem Cell. 10(3):312–326. doi:10.1016/j.stem.2012.01.014
  • Sinha A, Iyengar PV, Dijke PT. 2021. E3 ubiquitin ligases: Key regulators of tgfβ signaling in cancer progression. IJMS. 22(2):476. doi:10.3390/ijms22020476
  • Strauss J, Heery CR, Schlom J, Madan RA, Cao L, Kang Z, Lamping E, Marté JL, Donahue RN, Grenga I, et al. 2018. Phase I trial of M7824 (MSB0011359C), a bifunctional fusion protein targeting PD-L1 and TGFb, in advanced solid tumors. Clin Cancer Res. 24(6):1287–1295. doi:10.1158/1078-0432.CCR-17-2653
  • Su J, Morgani SM, David CJ, Wang Q, Er EE, Huang YH, Basnet H, Zou Y, Shu W, Soni RK, et al. 2020. TGF-β orchestrates fibrogenic and developmental EMTs via the RAS effector RREB1. Nature. 577(7791):566–571. doi:10.1038/s41586-019-1897-5
  • Su WZ, Yuan X. 2018. LncRNA GASL1 inhibits tumor growth of non-small cell lung cancer by inactivating TGF-γ pathway. Eur Rev Med Pharmacol Sci. 22(21):7282–7288. doi:10.26355/eurrev-201811-16264
  • Subramanian V, Borchard S, Azimzadeh O, Sievert W, Merl-Pham J, Mancuso M, Pasquali E, Multhoff G, Popper B, Zischka H, et al. 2018. PPARα is necessary for radiation-induced activation of noncanonical TGFβ signaling in the heart. J Proteome Res. 17(4):1677–1689. doi:10.1021/acs.jproteome.8b00001
  • Suman S, Kallakury BVS, Fornace AJ, Datta K. 2019. Fractionated and acute proton radiation show differential intestinal tumorigenesis and DNA damage and repair pathway response in ApcMin/+ mice. Int J Radiat Oncol Biol Phys. 105(3):525–536. doi:10.1016/j.ijrobp.2019.06.2532
  • Suphakhong K, Terashima M, Wanna-Udom S, Takatsuka R, Ishimura A, Takino T, Suzuki T. 2022. m6A RNA methylation regulates the transcription factors JUN and JUNB in TGF-β-induced epithelial–mesenchymal transition of lung cancer cells. J Biol Chem. 298(11):102554. doi:10.1016/j.jbc.2022.102554
  • Suriyamurthy S, Baker D, Ten Dijke P, Iyengar PV. 2019. Epigenetic reprogramming of TGF-β signaling in breast cancer. Cancers. 11(5):726. doi:10.3390/cancers11050726
  • Tang J, Cho NW, Cui G, Manion EM, Shanbhag NM, Botuyan MV, Mer G, Greenberg RA. 2013. Acetylation limits 53BP1 association with damaged chromatin to promote homologous recombination. Nat Struct Mol Biol. 20(3):317–325. doi:10.1038/nsmb.2499
  • Tang X, Tu G, Yang G, Wang X, Kang L, Yang L, Zeng H, Wan X, Qiao Y, Cui X, et al. 2019. Autocrine TGF-β1/miR-200s/miR-221/DNMT3B regulatory loop maintains CAF status to fuel breast cancer cell proliferation. Cancer Lett. 452(2019):79–89. doi:10.1016/j.canlet.2019.02.044
  • Thakur N, Hamidi A, Song J, Itoh S, Bergh A, Heldin CH, Landström M. 2020. Smad7 enhances TGF-β-induced transcription of c-Jun and HDAC6 promoting invasion of prostate cancer cells. iScience. 23(9):101470. doi:10.1016/j.isci.2020.101470
  • Tian W, Yin X, Wang L, Wang J, Zhu W, Cao J, Yang H. 2015. The key role of miR-21-regulated SOD2 in the medium-mediated bystander responses in human fibroblasts induced by α-irradiated keratinocytes. Mutat Res. 780(2015):77–85. doi:10.1016/j.mrfmmm.2015.08.003
  • Tong Q, Weaver MR, Kosmacek EA, O’Connor B, Harmacek L, Venkataraman S, Oberley-Deegan RE. 2016. MnTE-2-PyP reduces prostate cancer growth and metastasis by suppressing p300 activity and p300/HIF-1/CREB binding to the promoter region of the PAI-1 gene. Free Radic Biol Med. 94:185–194. doi:10.1053/j.gastro.2016.08.014.CagY
  • Tsai YC, Wang TY, Hsu CL, Lin WC, Chen JY, Li JH, Pu YS, Cheng AL, Cheng JCH, Su SF. 2023. Selective inhibition of HDAC6 promotes bladder cancer radiosensitization and mitigates the radiation-induced CXCL1 signalling. Br J Cancer. 128(9):1753–1764. doi:10.1038/s41416-023-02195-0
  • Tzavlaki K, Moustakas A. 2020. TGF-B signaling. Biomolecules. 10(3):487. doi:10.3390/biom10030487
  • Tzika E, Dreker T, Imhof A. 2018. Epigenitics and metabolism in health and disease. Front Genet. 9(361):1–8. doi:10.3389/fgene.2018.00361
  • Vander Ark A, Cao J, Li X. 2018. TGF-β receptors: In and beyond TGF-β signaling. Cell Signal. 52(2018):112–120. doi:10.1016/j.cellsig.2018.09.002
  • Visser HPJ, Gunster MJ, Kluin-Nelemans HC, Manders EMM, Raaphorst FM, Meijer CJLM, Willemze R, Otte AP. 2001. The Polycomb group protein EZH2 is upregulated in proliferating, cultured human mantle cell lymphoma. Br J Haematol. 112(4):950–958. doi:10.1046/j.1365-2141.2001.02641.x
  • Vogel T, Ahrens S, Büttner N, Krieglstein K. 2010. Transforming growth factor β promotes neuronal cell fate of mouse cortical and hippocampal progenitors in vitro and in vivo: identification of Nedd9 as an essential signaling component. Cerebral Cortex. 20(3):661–671. doi:10.1093/cercor/bhp134
  • Wang J, Wu M, Zheng D, Zhang H, Lv Y, Zhang L, Tan H, Sheng Zhou H, Lao Y, Zhi Xu H. S. 2020. Garcinol inhibits esophageal cancer metastasis by suppressing the p300 and TGF-β1 signaling pathways. Acta Pharmacol Sin. 41(1):82–92. doi:10.1038/s41401-019-0271-3
  • Wang M, Saha J, Cucinotta FA. 2013. Smad7 foci are present in micronuclei induced by heavy particle radiation. Mutat Res Genet Toxicol Environ Mutagen. 756(1-2):108–114. doi:10.1016/j.mrgentox.2013.04.011
  • Wang M, Saha J, Hada M, Anderson JA, Pluth JM, O’Neill P, Cucinotta FA. 2013. Novel Smad proteins localize to IR-induced double-strand breaks: Interplay between TGFβ and ATM pathways. Nucleic Acids Res. 41(2):933–942. doi:10.1093/nar/gks1038
  • Wang S, Li J, He Y, Ran Y, Lu B, Gao J, Shu C, Li J, Zhao Y, Zhang X, et al. 2021. Protective effect of melatonin entrapped PLGA nanoparticles on radiation-induced lung injury through the miR-21/TGF-β1/Smad3 pathway. Int J Pharm. 602(2021):120584. doi:10.1016/j.ijpharm.2021.120584
  • Wang Y, Ha M, Li M, Zhang L, Chen Y. 2022. Histone deacetylase 6-mediated downregulation of TMEM100 expedites the development and progression of non-small cell lung cancer. Hum Cell. 35(1):271–285. doi:10.1007/s13577-021-00635-8
  • Wanna-Udom S, Terashima M, Suphakhong K, Ishimura A, Takino T, Suzuki T. 2021. KDM2B is involved in the epigenetic regulation of TGF-β-induced epithelial–mesenchymal transition in lung and pancreatic cancer cell lines. J Biol Chem. 296(100213):100213. doi:10.1074/jbc.RA120.015502
  • Wick A, Desjardins A, Suarez C, Forsyth P, Gueorguieva I, Burkholder T, Cleverly AL, Estrem ST, Wang S, Lahn MM, et al. 2020. Phase 1b/2a study of galunisertib, a small molecule inhibitor of transforming growth factor-beta receptor I, in combination with standard temozolomide-based radiochemotherapy in patients with newly diagnosed malignant glioma. Invest New Drugs. 38(5):1570–1579. doi:10.1007/s10637-020-00910-9
  • Wiegman EM, Blaese MA, Loeffler H, Coppes RP, Rodemann HP. 2007. TGFβ-1 dependent fast stimulation of ATM and p53 phosphorylation following exposure to ionizing radiation does not involve TGFβ-receptor I signalling. Radiother Oncol. 83(3):289–295. doi:10.1016/j.radonc.2007.05.013
  • Xie Y, Chen L, Zhou J, Wang Q, Yang C, Xu C, Fan X, Tan Y, Wang Y, Kang C, et al. 2020. TGFβ signaling-induced miRNA participates in autophagic regulation by targeting PRAS40 in mesenchymal subtype of glioblastoma. Cancer Biol Med. 17(3):664–675. doi:10.20892/j.issn.2095-3941.2019.0356
  • Xu S, Ding N, Pei H, Hu W, Wei W, Zhang X, Zhou G, Wang J. 2014. MiR-21 is involved in radiation-induced bystander effects. RNA Biol. 11(9):1161–1170. doi:10.4161/rna.34380
  • Xu X, Yu H. 2019. Ras-PI3K pathway promotes osteosarcoma progression via regulating VRK1-mediated H2A phosphorylation at threonine 120. Artif Cells Nanomed Biotechnol. 47(1):4274–4283. doi:10.1080/21691401.2019.1687506
  • Yang T, Huang T, Zhang D, Wang M, Wu B, Shang Y, Sattar S, Ding L, Liu Y, Jiang H, et al. 2019. TGF-β receptor inhibitor LY2109761 enhances the radiosensitivity of gastric cancer by inactivating the TGF-β/SMAD4 signaling pathway. Aging. 11(20):8892–8910. doi:10.18632/aging.102329
  • Yang X, Ni J, Li Y, Zou L, Guo T, Li Y, Chu L, Zhu Z. 2020. LncRNA-RP11 modulates TGF-β1-activated radiation-induced lung injury through downregulating microRNA-29a. Dose-Response. 18(4):155932582094907. doi:10.1177/1559325820949071
  • Yang Y, Liu L, Fang M, Bai H, Xu Y. 2019. The chromatin remodeling protein BRM regulates the transcription of tight junction proteins: implication in breast cancer metastasis. Biochim Biophys Acta Gene Regul Mech. 1862(5):547–556. doi:10.1016/j.bbagrm.2019.03.002
  • Yang Y, Liu L, Li M, Cheng X, Fang M, Zeng Q, Xu Y. 2019. The chromatin remodeling protein BRG1 links ELOVL3 trans-activation to prostate cancer metastasis. Biochim Biophys Acta Gene Regul Mech. 1862(8):834–845. doi:10.1016/j.bbagrm.2019.05.005
  • Yano H, Hamanaka R, Nakamura-Ota M, Zhang JJ, Matsuo N, Yoshioka H. 2018. Regulation of type I collagen expression by microRNA-29 following ionizing radiation. Radiat Environ Biophys. 57(1):41–54. doi:10.1007/s00411-017-0723-4
  • Yap TA, Vieito M, Baldini C, Sepúlveda-Sánchez JM, Kondo S, Simonelli M, Cosman R, van der Westhuizen A, Atkinson V, Carpentier AF, et al. 2021. First-in-human phase I study of a next-generation, oral, TGFb receptor 1 inhibitor, LY3200882, in patients with advanced cancer. Clin Cancer Res. 27(24):6666–6676. doi:10.1158/1078-0432.CCR-21-1504
  • Yoon H, Tang CM, Banerjee S, Delgado AL, Yebra M, Davis J, Sicklick JK. 2021. TGF-β1-mediated transition of resident fibroblasts to cancer-associated fibroblasts promotes cancer metastasis in gastrointestinal stromal tumor. Oncogenesis. 10(2):1–12. doi:10.1038/s41389-021-00302-5
  • Yuan B, Dana FE, Ly S, Yan Y, Ruvolo V, Shpall EJ, Konopleva M, Andreeff M, Battula VL. 2020. Bone marrow stromal cells induce an ALDH + stem cell-like phenotype and enhance therapy resistance in AML through a TGF-β-p38-ALDH2 pathway. PLOS One. 15(11):e0242809. doi:10.1371/journal.pone.0242809
  • Yuan BY, Chen YH, Wu ZF, Zhuang Y, Chen GW, Zhang L, Zhang HG, Cheng JCH, Lin Q, Zeng ZC. 2019. MicroRNA-146a-5p attenuates fibrosis-related molecules in irradiated and TGF-beta1-treated human hepatic stellate cells by regulating PTPRA-SRC signaling. Radiat Res. 192(6):621–629. doi:10.1667/RR15401.1
  • Yuan D, Guo T, Zhu D, Ge H, Zhao Y, Huang A, Wang X, Cao X, He C, Qian H, et al. 2022. Exosomal lncRNA ATB derived from ovarian cancer cells promotes angiogenesis via regulating miR-204-3p/TGFβR2 axis. Cancer Manag Res. 14:327–337. doi:10.2147/CMAR.S330368
  • Yuan J, Xie A, Cao Q, Li X, Chen J. 2020. INHBB is a novel prognostic biomarker associated with cancer-promoting pathways in colorectal cancer. Biomed Res Int. 2020:1–14. doi:10.1155/2020/6909672
  • Zang C, Zhao F, Hua L, Pu Y. 2018. The miR-199a-3p regulates the radioresistance of esophageal cancer cells via targeting the AK4 gene. Cancer Cell Int. 18(1):186. doi:10.1186/s12935-018-0689-6
  • Zemann N, Klein P, Wetzel E, Huettinger F, Huettinger M. 2010. Lactoferrin induces growth arrest and nuclear accumulation of Smad-2 in HeLa cells. Biochimie. 92(7):880–884. doi:10.1016/j.biochi.2010.03.013
  • Zhang HT, Chen XF, Wang MH, Wang JC, Qi QY, Zhang RM, Xu WQ, Fei QY, Wang F, Cheng QQ, et al. 2004. Defective expression of transforming growth factor β receptor type II is associated with CpG methylated promoter in primary non-small cell lung cancer. Clin Cancer Res. 10(7):2359–2367. doi:10.1158/1078-0432.CCR-0959-3
  • Zhang J, Liu W, Dong H, Wang W. 2019. K-RasG12V/Y40C-PI3K/AKT pathway regulates H1.4S35ph through PKA to promote the occurrence and development of osteosarcoma cancer. Artif Cells Nanomed Biotechnol. 47(1):2048–2057. doi:10.1080/21691401.2019.1617726
  • Zhang L, Duan CJ, Binkley C, Li G, Uhler MD, Logsdon CD, Simeone DM. 2004. A transforming growth factor β-induced Smad3/Smad4 complex directly activates protein kinase A. Mol Cell Biol. 24(5):2169–2180. doi:10.1128/MCB.24.5.2169-2180.2004
  • Zhang L, Lu Q, Chang C. 2020. Epigenetics in health and disease. Adv Exp Med Biol. 1253:3–55. doi:10.1007/978-981-15-3449-2_1
  • Zhang LY, Yong WX, Wang L, Zhang LX, Zhang YM, Gong HX, He JP, Liu YQ. 2019. Astragalus polysaccharide eases G1 phase-correlative bystander effects through mediation of TGF-ββR/MAPK/ROS signal pathway after carbon ion irradiation in BMSCs. Am J Chin Med. 47(3):595–612. doi:10.1142/S0192415X19500319
  • Zhang X, Yang J, Zhou W, Chen Z, Wu W, Zhang S, Wang L. 2022. Identification of LncRNA CASC7/miR-26/ASPN/TGF-β/Smad axis in endometrial cancer. PJZ. 55(1):103–111. doi:10.17582/journal.pjz/20211201051249
  • Zhang Y, Wen G, Shao G, Wang C, Lin C, Fang H, Balajee AS, Bhagat G, Hei TK, Zhao Y. 2009. TGFBI deficiency predisposes mice to spontaneous tumor development. Cancer Res. 69(1):37–44. doi:10.1158/0008-5472.CAN-08-1648
  • Zhang YE. 2009. Non-Smad pathways in TGF-β signaling. Cell Res. 19(1):128–139. doi:10.1038/cr.2008.328
  • Zhang YE. 2017. Non-Smad signaling pathways of the TGF-β family. Cold Spring Harb Perspect Biol. 9(2):a022129. doi:10.1101/cshperspect.a022129
  • Zhang Z, Chen B, Zhu Y, Zhang T, Yuan Y, Zhang X, Xu Y. 2021. The Jumonji domain-containing histone demethylase homolog 1D/lysine demethylase 7A (JHDM1D/KDM7A) is an epigenetic activator of RHOJ transcription in breast cancer cells. Front Cell Dev Biol. 9:1–15. doi:10.3389/fcell.2021.664375
  • Zhao L, Bode AM, Cao Y, Dong Z. 2012. Regulatory mechanisms and clinical perspectives of miRNA in tumor radiosensitivity. Carcinogenesis. 33(11):2220–2227. doi:10.1093/carcin/bgs235
  • Zhao L, Pang A, Li Y. 2018. Function of GCN5 in the TGF β1 induced epithelial to mesenchymal transition in breast cancer. Oncol Lett. 16(3):3955–3963. doi:10.3892/ol.2018.9134
  • Zheng J, Mei Y, Xiang P, Zhai G, Zhao N, Xu C, Liu M, Pan Z, Tang K, Jia D. 2018. DNA methylation affects metastasis of renal cancer and is associated with TGF-β/RUNX3 inhibition. Cancer Cell Int. 18(1):56. doi:10.1186/s12935-018-0554-7
  • Zhou X, Li X, Wang R, Hua D, Sun C, Yu L, Shi C, Luo W, Jiang Z, An W, et al. 2022. Recruitment of LEF1 by Pontin chromatin modifier amplifies TGFBR2 transcription and activates TGFβ/SMAD signalling during gliomagenesis. Cell Death Dis. 13(9):818. doi:10.1038/s41419-022-05265-y
  • Zhu L, Yu CL, Zheng Y. 2019. NSD2 inhibition suppresses metastasis in cervical cancer by promoting TGF-β/TGF-βRI/SMADs signaling. Biochem Biophys Res Commun. 519(3):489–496. doi:10.1016/j.bbrc.2019.08.020

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.