Fact-checked by Grok 2 weeks ago

Transforming growth factor beta

Transforming growth factor beta (TGF-β) is a multifunctional belonging to the TGF-β superfamily, synthesized as a precursor protein that is cleaved to form a mature dimeric consisting of two polypeptide chains linked by bonds, and it plays pivotal roles in regulating cellular processes such as , , , extracellular matrix production, and immune responses. In mammals, there are three main isoforms—TGF-β1, TGF-β2, and TGF-β3—encoded by distinct genes, each exhibiting overlapping yet isoform-specific functions in embryonic , homeostasis, and . These isoforms are secreted in a latent form bound to latency-associated peptide (LAP) and require activation by or proteases to exert their effects. TGF-β signaling primarily occurs through the SMAD-dependent pathway, where the binds to type II (TβRII) and type I (TβRI) serine/ receptors, leading to of receptor-regulated SMADs (SMAD2 and SMAD3), which complex with SMAD4 to translocate to the and regulate target transcription. Non-canonical pathways, including of MAPK (e.g., ERK, JNK, p38), PI3K/AKT, and Rho , also contribute to diverse cellular outcomes such as cytoskeletal reorganization and modulation. In healthy physiology, TGF-β maintains by suppressing T-cell proliferation, promoting regulatory T-cell (Treg) differentiation, and inhibiting pro-inflammatory cytokines, while also driving epithelial-to-mesenchymal transition () essential for tissue repair and resolution. Dysregulated TGF-β signaling is implicated in numerous pathologies; for instance, excessive activity contributes to in organs like the lungs, kidneys, and liver by enhancing deposition and activation. In cancer, TGF-β acts paradoxically as a tumor suppressor in early stages by inducing cell cycle arrest and , but promotes progression in advanced tumors through , , , and . Aberrant signaling is also linked to autoimmune diseases, cardiovascular disorders, and developmental anomalies such as Loeys-Dietz syndrome. Therapeutically, targeting TGF-β has emerged as a promising strategy, with approved agents like the trap luspatercept for associated with myelodysplastic syndromes and β-thalassemia, and investigational agents such as bifunctional s (e.g., bintrafusp alfa) and small-molecule TβRI inhibitors (e.g., vactosertib) under evaluation in clinical trials as of 2025 for and cancer, though challenges include managing on-target toxicities and context-dependent effects.

Overview

Definition and isoforms

Transforming growth factor beta (TGF-β) is a multifunctional within the TGF-β superfamily, existing as a dimeric protein that exerts profound regulatory effects on cellular processes, including growth, differentiation, , and extracellular matrix production. These functions position TGF-β as a central in and across diverse physiological contexts. In mammals, TGF-β is represented by three principal isoforms: TGF-β1, TGF-β2, and TGF-β3, each encoded by distinct genes and exhibiting overlapping yet isoform-specific biological roles. TGF-β1 is the most ubiquitously expressed isoform, playing a pivotal role in immune suppression by modulating T-cell responses and promoting regulatory T-cell function to maintain . In contrast, TGF-β2 is prominently involved in neural and ocular development, where it supports , , and in the eye. TGF-β3, meanwhile, is critical for and secondary palate formation, facilitating epithelial-mesenchymal interactions that enable scarless tissue repair and palatal fusion during embryogenesis. Structurally, the isoforms display high , sharing approximately 70-80% sequence identity, which underscores their functional similarities while allowing for nuanced differences in receptor affinity and tissue specificity. A hallmark of this conservation is the knot , formed by interchain bonds that stabilize the dimeric structure essential for ligand-receptor interactions. TGF-β exhibits remarkable evolutionary conservation throughout vertebrates, reflecting its fundamental role in metazoan development and signaling. The signaling pathway shows ancient origins, with ligand homologs such as decapentaplegic (Dpp) in Drosophila melanogaster and DBL-1 in Caenorhabditis elegans, alongside downstream SMAD homologs like Mothers against dpp (Mad) in flies and the Sma family (Sma-2/3/4) in worms.

Discovery and nomenclature

The discovery of transforming growth factor beta (TGF-β) traces back to 1978, when Joseph E. De Larco and George J. Todaro identified a novel -promoting activity in conditioned medium from Moloney murine virus-transformed cells, which they termed factor (SGF). This factor was extracted from rodent cells and demonstrated the ability to induce anchorage-independent in normal fibroblasts, mimicking aspects of cellular transformation observed in cancer cells. In the early 1980s, researchers at the , including Anita B. Roberts and Michael B. Sporn, along with at , further characterized SGF and distinguished it into two components: TGF-α, which binds to the , and TGF-β, a distinct 25 kDa homodimeric protein purified from sources such as bovine kidney, human placenta, and activated platelets. Roberts and Sporn linked TGF-β to oncogenic transformation and its role in promoting through stimulation of synthesis and in cells. Concurrently, Moses demonstrated TGF-β's potent growth-inhibitory effects on epithelial cells, including mammary cells, revealing its dual functionality beyond promotion of transformation. These findings established TGF-β as a key regulator in cellular processes, with Moses's work also highlighting its emerging roles in modulating immune responses, such as suppression of T-cell and production. As research progressed, the evolved to reflect TGF-β's broader biological activities, shifting emphasis from its initial "transforming" label—based on anchorage-independent induction—to recognition as a multifunctional involved in immune regulation, , and tissue , rather than solely oncogenesis. Key milestones included the of the human TGF-β1 cDNA in 1985 by Rik Derynck and colleagues at , using partial amino acid sequencing to isolate the precursor protein, followed by the murine homolog in 1986. The discovery of isoforms expanded the family: TGF-β2 was cloned from porcine and human sources in 1987, showing 71% to TGF-β1, and TGF-β3 was identified via cDNA characterization in 1988, with distinct expression patterns in embryonic development and immune tissues. These advancements underscored TGF-β's conserved structure and diverse roles across species.

Molecular structure

Protein domains and assembly

The mature transforming growth factor beta (TGF-β) exists as a disulfide-linked homodimer of approximately 25 kDa, composed of two identical polypeptide chains each containing about 112 derived from the C-terminal region of the precursor protein. In certain species or experimental contexts, heterodimers such as TGF-β1/β2 can form, though homodimers predominate in mammals. The core active domain features nine conserved residues that establish a network of intra- and inter-chain bonds, enabling dimer assembly and conferring structural integrity essential for ligand-receptor interactions. Central to this domain is the cysteine knot , a hallmark of the TGF-β superfamily, formed by six of these cysteines: two bonds create an eight-membered ring through which a third bond passes, stabilizing the fold and allowing three protruding loops to mediate binding specificity. This anchors a beta-sheet-rich , including two extended antiparallel beta-hairpins (or "arms") per that interlock to form the dimeric , resulting in a compact, rigid structure. N-linked sites, such as those at residues in the precursor (e.g., Asn82 and Asn136 in TGF-β1), facilitate proper folding and efficient secretion of the assembled dimer, with mutagenesis studies showing reduced secretion upon site disruption. Insights into this assembly derive from studies in the 1990s, which first revealed the beta-sheet-dominated fold and ; for instance, the of mature TGF-β2, resolved at 2.1 resolution, highlighted the unusual pseudosymmetric dimer with inter-monomer contacts spanning the region. Subsequent structures of TGF-β3 confirmed a similar conformation but with refined details on loop flexibility. Isoform-specific variations include TGF-β1's tendency toward an open-arm dimer , promoting for signaling, contrasted with TGF-β2's structural features that favor with latency components, influencing its despite comparable mature folds.

Latent complex formation

The latency-associated peptide (LAP), which is the N-terminal prodomain of TGF-β, non-covalently binds to the mature TGF-β homodimer to form the small latent complex (SLC), thereby preventing premature interaction with receptors. This binding occurs after proteolytic processing of the pro-TGF-β precursor. The LAP wraps around the active TGF-β dimer through hydrophobic and electrostatic interactions, maintaining latency. The SLC can further associate with latent TGF-β binding proteins (LTBPs), which covalently link to LAP via disulfide bonds—primarily at cysteine 33 in LAP—to form the large latent complex (LLC). LTBPs, particularly LTBP1, LTBP3, and LTBP4, facilitate this attachment in the during the secretory pathway, ensuring proper folding and directing the complex to the (). The LLC anchors to ECM components such as fibrillins and through LTBP domains, enabling long-term sequestration. Assembly of the latent complex begins with dimerization of pro-TGF-β in the , followed by cleavage by furin-like proprotein convertases in the trans-Golgi network, which separates the mature TGF-β from while preserving their non-covalent association in the SLC. LTBP then binds intracellularly to the SLC, forming the LLC prior to ; this process was first elucidated in studies showing LTBP's essential role in TGF-β1 and latency. Isoform-specific differences influence latency efficiency: TGF-β1 and TGF-β3 readily form the LLC due to favorable LAP-LTBP interactions, whereas TGF-β2 predominantly remains as the SLC, owing to weaker binding affinity of its LAP to LTBPs. LTBP1 and LTBP3 bind all three isoforms with high affinity, while LTBP4 shows weaker, TGF-β1-specific binding. The latent complexes serve as a reservoir, sequestering TGF-β in tissues such as and within the , allowing regulated availability without constant synthesis. This storage mechanism, mediated by LTBPs' interactions with matrix proteins, ensures spatial and temporal control of TGF-β bioavailability.

Biosynthesis and processing

Gene expression and translation

The TGFB1, TGFB2, and TGFB3 genes, which encode the three mammalian isoforms of transforming growth factor beta (TGF-β), exhibit distinct genomic organizations. The TGFB1 gene is located on 19q13.2 and spans approximately 24 kb with 7 exons. The TGFB2 gene resides on 1q41, covering about 99 kb and consisting of 8 exons. In contrast, the TGFB3 gene is positioned on 14q24.3, encompassing roughly 25 kb and containing 7 exons. These structural features contribute to the isoform-specific and expression patterns observed in various tissues. Transcriptional regulation of TGF-β genes involves specific cis-regulatory elements in their promoters. Smad-binding elements (SBEs) in the TGFB1 promoter facilitate autoinduction, where activated Smad3/Smad4 complexes bind to induce TGFB1 expression, forming a loop that amplifies in responsive cells. Additionally, AP-1 binding sites within the promoter regions interact with Jun/Fos family members to modulate TGFB1 transcription, often in synergy with Smad pathways during cellular responses to stress or growth factors. These mechanisms ensure context-dependent expression, with feedback loops sustaining TGF-β production in fibrotic or inflammatory environments. Expression of TGF-β isoforms displays marked cell-type specificity, reflecting their roles in diverse physiological processes. TGF-β1 is highly expressed in platelets and megakaryocytes, where it constitutes a major stored released during to promote and immune modulation. Conversely, TGF-β2 shows lower expression in neurons compared to other cell types, with restricted localization to specific regions such as radial and axonal tracts during development. These patterns vary across isoforms, underscoring their non-redundant functions in . At the translational level, TGFB1 mRNA stability is controlled by AU-rich elements (AREs) in its 3' untranslated region (UTR), which recruit binding proteins like tristetraprolin (TTP) to promote rapid decay and fine-tune protein output in response to cellular needs. Megakaryocytes are a primary source, contributing approximately 50% of circulating TGF-β1 through platelet storage and release. In healthy humans, plasma concentrations of TGF-β1 typically range from 2 to 12 ng/mL (mean 4.1 ng/mL), predominantly in latent form, reflecting basal production from megakaryocytes and other sources.

Pro-TGF-β cleavage

Transforming growth factor beta (TGF-β) is initially synthesized as a pro-TGF-β precursor, a polypeptide ranging from 390 to 414 amino acids across its isoforms, which is translocated into the () during . In the ER lumen, pro-TGF-β monomers undergo folding and form disulfide-linked homodimers through residues in their prodomains, establishing the dimeric structure prior to further processing. This dimerization is essential for the stability and subsequent maturation of the precursor. The pro-TGF-β dimer is then transported to the Golgi apparatus, where it undergoes proteolytic cleavage by furin-like proprotein convertases. These enzymes recognize and cleave at a conserved multibasic RXXR motif located between the N-terminal latency-associated peptide (LAP) and the C-terminal mature TGF-β domain, typically after an arginine residue four positions upstream. This processing first removes the N-terminal signal peptide (if not already cleaved in the ER) and then separates the dimeric LAP from the mature TGF-β dimer, yielding the small latent complex (SLC) in which the mature TGF-β remains non-covalently associated with LAP. The cleavage is highly efficient, with approximately 90% of pro-TGF-β converted to the latent form in most cell types, ensuring regulated secretion of inactive TGF-β. The resulting latent complex is packaged into secretory vesicles and exported from the cell via the constitutive secretory pathway. Defects in this processing or secretion, such as mutations in the TGFB1 gene affecting LAP stability or dimerization, can lead to Camurati-Engelmann disease, a rare sclerosing bone disorder characterized by excessive TGF-β signaling due to impaired latency.

Activation mechanisms

Proteolytic and metalloprotease activation

Proteolytic and metalloprotease represents a key enzyme-mediated mechanism for releasing mature transforming growth factor beta (TGF-β) from its latent complex, primarily through degradation of the latency-associated peptide (). , a derived from plasminogen via by plasminogen activators such as urokinase-type plasminogen activator (), directly cleaves at specific sites within the amino-terminal region, disrupting the inhibitory structure and liberating active TGF-β. This involves nicking the glycopeptide of , which alters its conformation and exposes regions of the mature TGF-β dimer for receptor binding. Matrix metalloproteinases (MMPs), including MMP-2 and MMP-9, similarly contribute by proteolytically degrading at distinct cleavage sites, such as the RGD area, thereby freeing active TGF-β without requiring cellular traction. MMP-9, in particular, has been shown to process cell surface-associated latent TGF-β, enhancing its in extracellular matrices. These enzymes target the small latent complex or large latent complex bound to latent TGF-β binding proteins (LTBPs), with MMP-2 facilitating activation in vascular cells and MMP-9 in tumor microenvironments. The resulting release promotes downstream signaling, though the exact sites of cleavage vary by isoform and context. In physiological settings, this activation pathway is prominent during , where plasmin generated at injury sites processes latent TGF-β to regulate epithelial migration and deposition. During , macrophages release MMP-9 and other proteases to activate TGF-β, aiding in immune modulation and tissue remodeling. In fibrotic tissues, such as those in or , proteolytic mechanisms account for approximately 50-60% of total TGF-β activation, driving excessive matrix production and differentiation. To maintain balance, MMP activity is tightly regulated by tissue inhibitors of metalloproteinases (TIMPs), such as TIMP-1 and TIMP-2, which bind and inhibit these enzymes, thereby limiting unintended TGF-β release and preventing pathological . Dysregulation of this inhibition can exacerbate fibrotic conditions by sustaining elevated active TGF-β levels.

Integrin-dependent activation

Integrin-dependent of transforming beta (TGF-β) is a cell adhesion-mediated process primarily involving the αvβ6 and αvβ8, which bind to an RGD motif in the latency-associated peptide () of the latent TGF-β complex. This binding facilitates the release of mature TGF-β without the need for LAP in all cases, relying instead on mechanical force or assistance to induce conformational changes in the LAP that expose the growth factor for receptor interaction. Unlike purely proteolytic mechanisms, this pathway integrates (ECM) interactions with cytoskeletal dynamics, enabling localized and regulated TGF-β bioavailability in response to cellular traction. The αvβ6 , predominantly expressed on epithelial cells, activates latent TGF-β1 and TGF-β3 through a force-dependent mechanism that does not require . Upon binding the RGD in LAP, αvβ6 links to the via its cytoplasmic tail, transmitting contractile forces generated by the RhoA-ROCK-myosin II pathway to mechanically unfold the LAP and liberate active TGF-β. This process is enhanced by stimuli such as , which activates RhoA and ROCK to increase integrin and cytoskeletal tension. In epithelial cells, αvβ6 upregulation during injury or drives TGF-β activation, contributing to by promoting and ECM deposition. Cells expressing αvβ6 exhibit markedly elevated levels of active TGF-β compared to non-expressing counterparts, underscoring the pathway's efficiency in fibrotic contexts. In contrast, the αvβ8 employs a hybrid mechanism involving recruitment, particularly in immune and tumor cells. αvβ8 also binds the RGD in but lacks strong linkage, instead recruiting membrane-type 1 (MMP14) to cleave and release active TGF-β while maintaining the complex's association. This -dependent variant allows for rapid, localized without full release, as revealed by cryo-EM structures showing minimal conformational shifts sufficient for signaling. Expressed on dendritic cells, macrophages, regulatory T cells, and tumor cells, αvβ8-mediated promotes in tumor microenvironments by enhancing TGF-β signaling in adjacent immune cells, facilitating immune evasion. This pathway complements proteolytic in certain hybrid scenarios but remains distinct in its adhesion-driven initiation.

Non-integrin activation pathways

Besides the integrin-mediated mechanisms, several non-integrin pathways contribute to the of latent transforming growth factor beta (TGF-β) from its latency-associated peptide () through environmental or soluble cues. These pathways are particularly relevant in contexts such as , , and , where they enable localized release of active TGF-β without direct cell-matrix traction. One prominent non-integrin activation route involves low environments, which protonate specific residues in the , inducing a conformational change that dissociates the mature TGF-β dimer. This process occurs efficiently at pH levels around 4.5, as seen in acidic compartments like endosomes or in pathological settings such as ischemic tissues and tumor microenvironments, where hypoxia-driven lowers extracellular pH. In by osteoclasts, for instance, the transient acidification to approximately pH 4.5 facilitates latent TGF-β release to regulate activity. (ROS) represent another key non-integrin activator, primarily by oxidizing a conserved residue at position 253 in the of TGF-β1, which destabilizes the latent complex and promotes TGF-β release. This mechanism is especially active under conditions, such as or in tumors, where elevated ROS levels from sources like NADPH oxidases enhance TGF-β to drive fibrotic or angiogenic responses. Notably, ROS-mediated activation can synergize with pathways, amplifying overall TGF-β signaling in stressed tissues. Thrombospondin-1 (TSP-1), a matricellular secreted by platelets and other cells, binds directly to the via its KRFK amino acid sequence interacting with the LSKL motif in , thereby inducing a conformational shift that liberates active TGF-β. This pathway is crucial in platelet activation during and in fibrotic remodeling, where TSP-1 accounts for a significant portion of physiological TGF-β activation , independent of proteolytic cleavage. Additional non-integrin triggers include physical stimuli like , which generate ROS or mechanical perturbations to activate latent TGF-β in therapeutic applications, such as targeted tissue repair or . In hypoxic tumor niches, the combined effects of low and ROS from these pathways sustain TGF-β-driven progression, underscoring their role in disease contexts beyond normal .

Receptors and signaling

TGF-β receptors

Transforming growth factor β (TGF-β) initiates signaling primarily through two serine/ receptors: type I (TβRI, also known as ALK5) and type II (TβRII). These transmembrane proteins consist of an extracellular - domain, a single-span transmembrane region, and an intracellular domain responsible for signal propagation. TGF-β1 and TGF-β3 exhibit high-affinity to TβRII, which serves as the primary receptor, while TGF-β2 binds poorly to TβRII alone. Upon engagement, TβRII recruits and activates TβRI, forming a functional complex essential for downstream effects. Structurally, the extracellular domain of TβRII is characterized by a cysteine-rich region that mediates specific interactions with TGF-β . In contrast, TβRI features a distinctive glycine-serine-rich (GS) domain in its juxtamembrane cytoplasmic region, which is phosphorylated by the constitutively active of TβRII to enable TβRI activation. Ligand binding induces oligomerization of these receptors into a heterotetrameric complex composed of two TβRII and two TβRI molecules, stabilizing the assembly and positioning the kinases for cross-phosphorylation. Accessory receptors modulate TGF-β binding and presentation to the core receptors. Betaglycan, also termed TβRIII, is a transmembrane that binds all three TGF-β isoforms with high affinity, particularly facilitating TGF-β2 access to TβRII due to the latter's low affinity for this isoform. , another accessory receptor, associates with TβRI and TβRII to enhance TGF-β1 and TGF-β3 binding, predominantly in endothelial cells where it influences vascular responses. Both TβRI and TβRII are ubiquitously expressed across tissues, with elevated levels in embryonic development and injury sites such as wounds.

Canonical SMAD pathway

The canonical SMAD pathway represents the primary intracellular signaling route for transforming growth factor beta (TGF-β), transducing ligand binding at the cell surface into transcriptional changes in the nucleus via SMAD proteins. Upon TGF-β binding to its serine/threonine kinase receptors, type II TGF-β receptor (TβRII) recruits and phosphorylates type I TGF-β receptor (TβRI) at its glycine-serine (GS) domain, activating its kinase activity. This activated TβRI then phosphorylates receptor-regulated SMADs (R-SMADs), specifically SMAD2 and SMAD3, at their C-terminal SXS motifs (where S is serine and X is any amino acid). The phosphorylation is facilitated by adaptor proteins such as SARA (SMAD anchor for receptor activation), which positions R-SMADs near the receptor complex. Phosphorylated R-SMADs dissociate from and form heteromeric complexes with the common mediator SMAD4, typically as trimers consisting of two R-SMADs and one SMAD4. These complexes accumulate in the before rapidly translocating to the through complexes, independent of additional energy input beyond their intrinsic nuclear localization signals. In the , the SMAD complexes bind to SMAD-binding elements (SBEs) in the promoter regions of target genes, characterized by the consensus sequence AGAC or GTCT, often in cooperation with other DNA-binding transcription factors such as FOXH1 or RUNX. Transcriptional activation or repression ensues through recruitment of co-activators like p300/CBP acetyltransferases or co-repressors such as /SnoN, leading to regulation of genes including SERPINE1 (encoding PAI-1, ) and various collagen genes involved in production. The pathway is tightly regulated to prevent sustained signaling, primarily through inhibitory SMADs (I-SMADs) SMAD6 and SMAD7. SMAD6 preferentially inhibits SMAD1/5/8 in BMP signaling but also competes with R-SMADs for receptor binding in TGF-β contexts, while SMAD7 broadly blocks R-SMAD by binding TβRI and recruiting E3 ubiquitin ligases like SMURF1/2 for receptor or SMAD degradation. Additionally, dephosphorylation of phospho-R-SMADs by PPM1A (also known as PP2Cα) in the terminates signaling, recycling SMADs for export and feedback inhibition. Activation of the canonical pathway elicits context-dependent cellular outcomes, prominently including cell cycle arrest through induction of cyclin-dependent kinase inhibitors such as p15^INK4B and p21^CIP1, which inhibit CDK4/6 and CDK2, respectively, thereby enforcing G1-phase arrest. In epithelial cells, it drives epithelial-mesenchymal transition (EMT) by upregulating transcription factors like Snail and Twist, which repress E-cadherin expression and promote mesenchymal markers such as vimentin and fibronectin. These effects underscore the pathway's role in development, homeostasis, and pathology, as originally elucidated in seminal studies identifying SMADs as key transducers.

Non-canonical signaling pathways

In addition to the SMAD-dependent pathway, transforming growth factor beta (TGF-β) activates several non-canonical signaling branches that operate independently of SMAD proteins, primarily through kinase-mediated mechanisms in the to regulate processes such as , , , and . These pathways are initiated by ligand binding to TGF-β receptors, leading to receptor complex and of adaptor proteins, and their outcomes often intersect with or modulate canonical signaling in a context-dependent manner. One prominent non-canonical route involves the death domain-associated protein (DAXX), where TGF-β type I receptor (TβRI) recruits DAXX to facilitate activation of c-Jun N-terminal (JNK) and subsequent caspase-mediated , particularly in epithelial and neuronal cells responsive to TGF-β-induced . This pathway requires DAXX's interaction with the Fas death domain and ASK1 ( signal-regulating 1), amplifying pro-apoptotic signals without SMAD involvement, as demonstrated in studies of TGF-β-treated hepatoma cells where DAXX knockdown attenuated JNK activation and . The (MAPK)/extracellular signal-regulated kinase (ERK) pathway represents another key non-canonical arm, wherein TβRI transactivates the Ras-RAF-MEK-ERK cascade to promote and epithelial-mesenchymal transition (), especially in fibroblasts and cancer cells. TGF-β induces rapid tyrosine phosphorylation of ShcA adaptor proteins by TβRI, leading to ERK activation that drives gene expression for matrix remodeling and motility, as evidenced in models where ERK inhibition blocked TGF-β-enhanced invasiveness. This branch is distinct from JNK/p38 MAPKs, which TGF-β also activates via TRAF6-TAK1 complexes for stress responses. TGF-β further engages the phosphoinositide 3-kinase (PI3K)-AKT pathway and Rho GTPases through receptor-mediated crosstalk, influencing cell migration and cytoskeletal dynamics. In this mechanism, TGF-β promotes TRAF6-dependent ubiquitination to activate PI3K-AKT signaling, enhancing survival and motility in tumor cells, while RhoA GTPase activation by TβRI leads to ROCK (Rho-associated kinase)-mediated stress fiber formation and EMT. For instance, in vascular smooth muscle cells, TGF-β/Smad3-independent PI3K-AKT activation sustains proliferation, and RhoA inhibition disrupts TGF-β-induced fibronectin production and migration. The TGF-β-activated 1 (TAK1)-mediated pathway, involving TRAF6 polyubiquitination, activates or p38 MAPK to drive inflammatory responses and , independent of TβRI activity. TRAF6 facilitates K63-linked ubiquitination of TAK1, enabling its oligomerization and downstream signaling to IKK for nuclear translocation or MKKs for p38 activation, as shown in renal fibroblasts where TAK1 blockade reduced TGF-β-induced deposition. These non-canonical pathways exhibit strong context-dependency, often dominating in pathological states like cancer, where sustained ERK hyperactivation by TGF-β contributes to tumor progression and by overriding growth suppression. In and cancers, for example, TGF-β shifts from cytostatic to pro-oncogenic effects via ERK and PI3K-AKT, facilitating without altering canonical SMAD outputs.

Biological functions

Regulation of cell proliferation and differentiation

Transforming growth factor beta (TGF-β) exerts potent antiproliferative effects on various cell types, particularly epithelial cells, by inducing cell cycle arrest in the G1 phase. This inhibition occurs primarily through the upregulation of cyclin-dependent kinase (CDK) inhibitors, such as p15INK4B and p21CIP1, which disrupt cyclin-CDK complexes essential for G1/S transition. Specifically, TGF-β activates SMAD3, which forms a complex with SMAD4 and transcription factors like FOXO to directly induce p15INK4B expression, thereby inhibiting CDK4/6 activity. Similarly, SMAD3-SMAD4 complexes with Sp1 promote p21CIP1 transcription, blocking CDK2 function and reinforcing G1 arrest. These mechanisms are highly effective in non-transformed epithelial cells, where picomolar concentrations of TGF-β (IC50 approximately 10-50 pM) suffice to halt proliferation. Beyond proliferation control, TGF-β plays a in directing cell , influencing lineage commitment in response to tissue-specific cues. In fibrotic conditions, TGF-β drives the of fibroblasts into myofibroblasts, characterized by increased expression of alpha-smooth muscle actin (α-SMA) and proteins. This process is mediated by SMAD3-dependent signaling, which activates profibrotic programs and is central to pathological in organs like the and . In mesenchymal stem cells (MSCs), TGF-β promotes chondrogenesis by inducing chondrocyte-specific markers such as and collagen type II, facilitating formation even with brief exposure (e.g., one day of stimulation). These effects highlight TGF-β's versatility in maintaining tissue architecture during development and repair. TGF-β also induces in select cell types, contributing to its tumor-suppressive functions. In hepatocytes, SMAD3 mediates TGF-β-induced by sensitizing cells to pro-apoptotic signals, thereby reducing susceptibility to hepatocarcinoma. In B cells, is triggered through a SMAD-dependent pathway involving the adaptor protein DAXX, which facilitates JNK activation and cascade initiation. These mechanisms ensure controlled cell elimination in response to TGF-β, preventing uncontrolled growth in normal tissues. The regulatory effects of TGF-β exhibit context-dependent duality, acting as a inhibitor in normal cells while paradoxically promoting and survival in advanced tumor cells. In non-malignant epithelial cells, TGF-β enforces via the aforementioned CDK inhibitors and pathways. However, in oncogenic contexts, sustained TGF-β signaling shifts toward epithelial-mesenchymal transition (), enabling tumor cell , , and without direct proliferation enhancement. This switch underscores TGF-β's role in tumor progression once early suppressive barriers are breached.

Immune system modulation

Transforming growth factor beta (TGF-β) plays a central role in modulating the by exerting predominantly immunosuppressive effects that maintain and prevent excessive . Through its SMAD signaling pathway and non-canonical routes, TGF-β regulates the , , and of various immune cell types, promoting an environment essential for . Dysregulation of TGF-β signaling can lead to immune disorders, highlighting its dual potential in both suppressing pathological responses and enabling immune evasion in certain contexts. In T cells, TGF-β promotes the differentiation of regulatory T cells (Tregs) by inducing Foxp3 expression, a master transcription factor that confers suppressive function. This process occurs primarily through SMAD3/4-mediated transcription in naive CD4+ T cells, enhancing their ability to inhibit effector T cell responses and maintain peripheral tolerance. Conversely, TGF-β inhibits the differentiation of pro-inflammatory Th1 and Th17 cells via SMAD-dependent suppression of key cytokines like IFN-γ and IL-17, thereby skewing the immune response away from autoimmunity and toward regulation. These effects collectively position TGF-β as a critical checkpoint in T cell-mediated immunity. TGF-β exerts suppressive effects on B cells by inhibiting their proliferation and inducing in activated B cells via SMAD signaling that arrests cell cycle progression at the . Additionally, TGF-β promotes from IgM to IgA, supporting mucosal immunity. These actions help regulate B cell-dependent and support . In macrophages, TGF-β drives polarization toward an M2-like phenotype, characterized by increased production of IL-10 and reduced expression of pro-inflammatory mediators like TNF-α and IL-12. This shift, mediated by SMAD and signaling, facilitates resolution of and tissue repair by promoting and dampening acute responses. Such reprogramming underscores TGF-β's role in transitioning macrophages from pro-inflammatory M1 states to regulatory functions. TGF-β inhibits the maturation of dendritic cells (DCs), preventing their upregulation of co-stimulatory molecules like and , and instead promoting an immature, tolerogenic state that induces antigen-specific unresponsiveness in T cells. This effect, driven by SMAD-mediated downregulation of maturation signals, enhances by limiting DC-initiated effector responses. Overall, TGF-β's immunosuppressive actions are pivotal in establishing oral tolerance, where it conditions to suppress responses to harmless antigens, as demonstrated in models of mucosal immunity. In pathological settings, elevated TGF-β facilitates tumor evasion by fostering an immunosuppressive microenvironment that recruits Tregs and impairs anti-tumor immunity. TGF-β deficiency, as observed in models, results in spontaneous multi-organ due to unchecked effector responses, emphasizing its essential role in preventing immune dysregulation.

Tissue homeostasis and repair

Transforming growth factor beta (TGF-β) plays a central role in (ECM) production by upregulating the synthesis of key components such as collagens I and III, as well as , primarily through the canonical SMAD signaling pathway. In fibroblasts and other matrix-producing cells, TGF-β binds to its receptors, leading to the and nuclear translocation of SMAD2 and SMAD3, which form complexes with SMAD4 to transcriptionally activate genes encoding these ECM proteins. This process enhances the deposition and organization of the ECM, supporting structural integrity in various tissues. Additionally, facilitate the proper deposition of these ECM components by mediating cell-matrix interactions that guide fibril assembly and alignment during TGF-β-induced remodeling. In , TGF-β is essential for orchestrating the repair process by recruiting fibroblasts to the injury site and promoting their into myofibroblasts, which drive wound contraction through actin-mediated forces. This recruitment occurs via chemotactic signals and upregulation of molecules, enabling fibroblasts to migrate into the provisional matrix formed by and plasma proteins. Among the isoforms, TGF-β3 particularly promotes regenerative healing with minimal by modulating fibroblast activity to favor ordered deposition over excessive , as opposed to TGF-β1, which can exacerbate formation. Macrophages contribute to this process by releasing TGF-β to aid fibroblast recruitment and remodeling during the proliferative phase. TGF-β maintains tissue by balancing synthesis with degradation, primarily through regulation of matrix metalloproteinases (MMPs) that cleave collagens and other matrix proteins to prevent accumulation. In steady-state conditions, TGF-β induces moderate MMP expression, such as MMP-2 and MMP-9, alongside their inhibitors like TIMPs, ensuring controlled turnover that preserves tissue architecture. Dysregulation of this balance, often from excessive TGF-β signaling, leads to pathological characterized by unchecked deposition and reduced MMP activity, resulting in stiff, dysfunctional tissues. TGF-β inhibits in endothelial cells through endoglin-mediated signaling, which modulates the balance between pro- and anti-angiogenic responses to maintain vascular during repair. , as a co-receptor, favors ALK1/SMAD1/5/8 pathways that suppress endothelial and , counteracting VEGF-driven vessel formation to prevent excessive in healing wounds. This inhibitory effect is crucial for resolving and stabilizing mature vessels in the . A notable example of TGF-β's role in scarless healing is observed in fetal skin wounds, where a balanced expression of isoforms—particularly higher TGF-β3 relative to TGF-β1—promotes regeneration without by limiting excessive deposition and . This isoform balance results in rapid, organized restoration mimicking embryonic development, contrasting with adult wounds that due to TGF-β1 dominance.

Therapeutic targeting

Receptor inhibitors and antagonists

Receptor inhibitors and antagonists of transforming growth factor beta (TGF-β) signaling primarily target the type I and type II serine/ receptors (TβRI and TβRII) to disrupt ligand-induced heterodimerization and downstream activation. These agents include small-molecule inhibitors, monoclonal antibodies, and natural proteoglycans that block receptor activation or ligand-receptor interactions. By preventing of receptor-regulated SMADs, they inhibit canonical and non-canonical pathways implicated in and tumor progression. Small-molecule inhibitors typically act by competitively binding the ATP-binding pocket of the TβRI (ALK5), selectively blocking its autophosphorylation and subsequent SMAD2/3 recruitment without directly affecting TβRII. A prototypical example is SB-431542, a derivative that potently inhibits ALK5 ( = 94 nM) as well as related ALK4 and ALK7, but spares other s like p38 MAPK by over 100-fold. This compound has been widely used in preclinical studies to dissect TGF-β-dependent processes, such as production in fibroblasts. Galunisertib (LY2157299) was a clinically advanced, orally bioavailable derivative that similarly targeted the TβRI with high selectivity, suppressing SMAD signaling in tumor models. It was evaluated in phase 1/2 trials for , often in combination with and radiotherapy, demonstrating pharmacodynamic inhibition of TGF-β pathway activation in patient tumors, but development was discontinued in 2020 due to insufficient efficacy. Monoclonal antibodies and fusion proteins provided alternative strategies by neutralizing TGF-β ligands or sequestering them via receptor ectodomains, thereby preventing receptor engagement. Fresolimumab (GC1008) was a human IgG4 that bound all three TGF-β isoforms (TGF-β1, -β2, -β3) with high , inhibiting their interaction with TβRI/TβRII complexes and blocking downstream signaling in fibrotic and neoplastic tissues. In phase 1 studies, it showed tolerability and evidence of target engagement, such as reduced phospho-SMAD levels in treated patients, but development has since been discontinued. Bintrafusp alfa (M7824) was a bifunctional comprising the extracellular domain of TβRII (acting as a TGF-β "trap") fused to an anti- , which sequestered TGF-β ligands while simultaneously blocking PD-L1 on tumor cells to enhance immune responses. This design exploited competitive binding to mature TGF-β dimers, localizing inhibition to the and demonstrating dual pathway blockade in preclinical models. However, its main development program was discontinued in 2021 following failures in phase 2/3 trials due to lack of and issues, though some smaller studies continued into 2025. Natural antagonists like , a small leucine-rich expressed in the , inhibit TGF-β signaling by directly binding and sequestering bioactive TGF-β isoforms, thereby reducing their availability for receptor activation. interacts with the TGF-β latency-associated peptide to promote its retention in , attenuating receptor-mediated effects in tissues prone to , as demonstrated in models of scarring and tumor . Unlike synthetic inhibitors, decorin's mechanism involves non-competitive trapping without kinase domain interference. Despite their therapeutic potential, TGF-β receptor inhibitors face challenges related to selectivity and off-target effects, as complete pathway blockade can disrupt essential homeostatic functions. Small-molecule inhibitors like galunisertib exhibited dose-dependent cardiac toxicities, including valvulopathy and -like lesions reminiscent of Loeys-Dietz syndrome, attributed to impaired TGF-β-mediated vascular integrity. Antibody-based traps may mitigate some risks through localized action but still risk immune dysregulation or events at higher doses. Ongoing research emphasizes optimizing selectivity for pathological versus physiological signaling to balance efficacy and safety. As of 2025, following discontinuations of several agents, next-generation TGF-β inhibitors, such as small-molecule ALK5 inhibitors like HYL001, are in early clinical development for and cancer.

TGF-β mimics and agonists

TGF-β mimics and agonists encompass a range of synthetic and natural compounds designed to activate TGF-β signaling pathways, primarily targeting the canonical SMAD pathway to promote regenerative processes such as repair and . These agents aim to enhance downstream effects like production and responses without relying on the native , offering potential therapeutic advantages in conditions involving loss or impaired healing. Small molecule agonists, such as SRI-011381 (also known as C381), represent a class of orally active compounds that potentiate TGF-β signaling by promoting lysosomal acidification and activating SMAD transcription factors. SRI-011381 has demonstrated efficacy in preclinical models of muscle regeneration, where it restores lysosomal and enhances myogenic , potentially benefiting conditions like muscular dystrophies by improving muscle repair and reducing degeneration. Multiple studies confirm its ability to activate TGF-β pathways without significant toxicity in investigational settings. Peptide-based mimics derived from TGF-β structural elements, such as the 14-residue pm26TGF-β1 (sequence: ACESPLKRQCGGGS), have been developed through to replicate ligand-receptor interactions. This peptide binds to TGF-β receptor II (TβRII), mimicking TGF-β1's effects by downregulating TNF-α production and upregulating IL-10 in peripheral blood mononuclear cells, while promoting regulatory T-cell differentiation. , it reduces leukocyte rolling and migration in mouse models of , supporting its application in by modulating immune responses and facilitating tissue remodeling without cytotoxicity at therapeutic doses. Gene therapy approaches using adeno-associated virus (AAV) vectors to deliver TGF-β genes provide sustained activation of signaling in targeted tissues. For instance, recombinant AAV carrying human TGF-β1 (rAAV-hTGF-β) transduces with high efficiency (up to 80% ), leading to prolonged expression over 90 days in osteoarthritic explants. This results in enhanced chondrocyte proliferation (up to 15.8-fold), reduced , increased and synthesis (up to 8.2-fold), and decreased hypertrophic markers like type X collagen and MMP-13 (up to 31-fold), thereby restructuring damaged toward a reparative phenotype. Such strategies hold promise for treatment by restoring articular . Within the TGF-β superfamily, bone morphogenetic proteins (BMPs), such as and BMP7, function as natural agonists with partial signaling overlap, utilizing shared SMAD effectors (e.g., SMAD1/5/8) and receptors to promote osteogenesis and chondrogenesis. BMPs drive bone formation during fracture healing and are clinically approved for and non-unions, where they enhance differentiation and deposition, complementing TGF-β's roles in early progenitor . Therapeutic applications of TGF-β mimics and agonists are being explored in early preclinical and clinical stages, particularly for , where TGF-β1 agonists aim to bolster bone formation by stimulating activity and matrix mineralization. However, a key concern is the risk of excessive accumulation leading to , as hyperactivation of TGF-β signaling can promote pathological scarring in tissues like and lungs, necessitating careful dosing and pathway-specific targeting to balance regeneration and adverse fibrotic outcomes.

Clinical significance

Role in cancer

Transforming growth factor beta (TGF-β) plays a dual role in cancer progression, functioning as a tumor suppressor in early neoplastic stages while promoting in advanced tumors. In premalignant epithelial cells, TGF-β signaling enforces growth arrest and by upregulating cyclin-dependent kinase inhibitors such as p15INK4B and p21CIP1, which inhibit progression and prevent oncogenic transformation. This suppressive mechanism is particularly evident in normal and early-stage tissues, where TGF-β maintains genomic stability and limits proliferation through canonical SMAD-dependent pathways. Loss of this responsiveness often marks the transition to aggressive disease, allowing TGF-β to shift toward protumorigenic activities. In late-stage cancers, TGF-β drives epithelial-mesenchymal transition (EMT), enhancing tumor cell motility, invasion, and . For example, in , TGF-β induces downregulation of E-cadherin and upregulation of mesenchymal markers like and N-cadherin, enabling cancer cells to disseminate from the primary site. Similarly, in pancreatic ductal , TGF-β promotes invasive behavior by remodeling the and activating non-canonical pathways such as MAPK/ERK, contributing to poor patient outcomes. This promotional role is exacerbated by acquired resistance in tumor cells, often through downstream mutations that uncouple growth inhibition from migratory signals. Within the tumor microenvironment, TGF-β orchestrates immunosuppression and stromal remodeling to support cancer progression. It expands regulatory T cells (Tregs) by inducing Foxp3 expression, thereby dampening antitumor immune responses and fostering immune evasion. TGF-β also activates cancer-associated fibroblasts (CAFs), which secrete pro-angiogenic factors like VEGF and matrix components that facilitate vascularization and tumor invasion. Among the isoforms, TGF-β1 predominates in the stromal compartment, where it amplifies CAF-mediated protumor effects and correlates with aggressive tumor phenotypes. Genetic alterations further underscore TGF-β's oncogenic potential; mutations in the type II TGF-β receptor (TβRII) are found in approximately 30% of colorectal cancers, especially those with , leading to impaired signaling and unchecked epithelial proliferation. Therapeutically, targeting TGF-β has shown promise, with inhibitors like vactosertib—a selective TβRI inhibitor—under investigation in clinical trials for (HCC), often in combination with to reverse and . Elevated plasma TGF-β levels also serve as a prognostic , with high concentrations associated with advanced disease stage, , and reduced survival across multiple cancer types, including colorectal and breast cancers.

Cardiovascular and connective tissue disorders

Transforming growth factor beta (TGF-β) signaling dysregulation contributes significantly to hereditary disorders characterized by aortic pathology. In , mutations in the FBN1 gene encoding fibrillin-1 impair the sequestration of latent TGF-β in the , leading to elevated free TGF-β levels and hyperactivation of downstream signaling pathways that drive degradation and thoracic aortic aneurysms. This excessive TGF-β activity promotes cell and expression, exacerbating aortic root dilation and increasing dissection risk. Loeys-Dietz syndrome, caused by heterozygous loss-of-function mutations in TGFBR1 or TGFBR2, paradoxically results in enhanced TGF-β signaling due to compensatory mechanisms, leading to widespread vascular fragility, arterial tortuosity, and early-onset aneurysms. These mutations disrupt receptor-mediated negative feedback, allowing unchecked Smad2/3 phosphorylation and nuclear translocation, which induces excessive remodeling and in arterial walls. In ischemic heart disease, TGF-β is pivotal in post-myocardial infarction (MI) fibrosis, where it activates canonical Smad signaling in cardiac fibroblasts to upregulate synthesis and differentiation, facilitating formation for structural integrity but contributing to ventricular stiffness if prolonged. Decreased expression of inhibitory Smad7 post-MI further amplifies this pathway, correlating with increased TGF-β1 in infarct zones and adverse remodeling. TGF-β also drives pathological cardiac in , where pressure overload induces cardiomyocyte TGF-β expression, triggering Smad-dependent gene transcription that promotes protein synthesis, re-expression of fetal genes, and interstitial , ultimately impairing diastolic function. This process is interconnected with angiotensin II signaling, amplifying TGF-β-mediated deposition in the hypertrophied ventricle. In , TGF-β exacerbates by elevating via activation, impairing bioavailability and promoting adhesion. Conversely, it aids plaque stabilization through induction of vascular cell phenotypic switching to a contractile state and fibrous cap formation, with higher TGF-β2 levels in plaques associated with reduced rupture risk. Therapeutic strategies targeting TGF-β have advanced post-2020, with blockers like losartan demonstrating efficacy in reducing aortic dilation rates in by attenuating TGF-β/Smad signaling, as evidenced in ongoing comparative trials against beta-blockers. Preclinical data support direct TGF-β inhibitors, such as the betaglycan-derived P144 peptide, in preventing onset in Marfan models by normalizing excessive signaling without promoting progression.

Autoimmune and inflammatory diseases

Transforming growth factor beta (TGF-β) plays a complex role in and diseases, often acting as a regulator that suppresses excessive immune responses while contributing to pathological when dysregulated. In , TGF-β promotes (Treg) differentiation and function, helping to maintain and prevent self-reactive . However, its overproduction can drive fibrotic remodeling in affected tissues, exacerbating disease progression. This dual nature underscores TGF-β's involvement in both protective and detrimental processes across various conditions. In (), an autoimmune of the , TGF-β supports remyelination by promoting precursor cell differentiation and maturation, potentially aiding repair in lesions. Conversely, excessive TGF-β signaling contributes to in chronic MS plaques, leading to scar formation that hinders axonal regeneration and functional recovery. Studies in animal models of MS, such as experimental autoimmune encephalomyelitis, have shown that modulating TGF-β pathways can influence disease severity, with balanced levels favoring . Tuberculosis (TB), caused by , involves TGF-β in maintaining bacterial latency by inhibiting activation and promoting an environment that limits excessive tissue damage. Single nucleotide polymorphisms (SNPs) in the TGFB1 gene, such as those affecting the latency-associated , have been associated with increased susceptibility to TB development and progression in human populations. For instance, certain TGFB1 variants correlate with higher plasma TGF-β levels and altered immune responses in TB patients, highlighting genetic influences on disease risk. In (IBD), encompassing conditions like and , TGF-β induces Treg cells that suppress pro-inflammatory Th17 responses, thereby preventing or ameliorating through maintenance of intestinal immune . Deficiency in TGF-β signaling, as observed in mouse models with disrupted Smad pathways, leads to spontaneous resembling human IBD, emphasizing its protective role against mucosal inflammation. Clinical observations in IBD patients show reduced TGF-β expression in inflamed tissues, correlating with disease flares. Rheumatoid arthritis (RA), a chronic autoimmune disorder characterized by synovial and joint destruction, features a for TGF-β: it suppresses early inflammatory responses by inhibiting T cell proliferation and cytokine production, yet chronic elevation drives synovial and pannus formation, contributing to joint deformity. In RA synovial fibroblasts, TGF-β upregulates genes, promoting , while therapeutic blockade in preclinical models reduces joint damage without exacerbating . Emerging therapies targeting TGF-β in autoimmune diseases include low-dose TGF-β administration to induce , as demonstrated in preclinical and early clinical trials for conditions like and IBD. For example, low-dose TGF-β1 has been tested in animal models to enhance Treg function and reduce autoantigen-specific responses, with phase I trials in humans exploring its safety for induction in transplantation-related . These approaches aim to harness TGF-β's immunosuppressive effects while minimizing fibrotic risks through precise dosing.

Metabolic and neurological conditions

Transforming growth factor beta (TGF-β) plays a significant role in metabolic disorders such as and , where elevated levels in promote chronic and contribute to . In obese individuals, increased TGF-β1 expression in expanding fat depots drives profibrotic responses and infiltration, exacerbating dysfunction and systemic metabolic impairment. Blockade of the TGF-β/Smad3 signaling pathway has been shown to protect against diet-induced by enhancing in and reducing fat accumulation. Genetic studies further support this, as TGF-β1 mice exhibit reduced formation and improved metabolic profiles, while Smad3-deficient models display significantly lower fat mass alongside resistance to . These findings highlight TGF-β's contribution to the link between adipose and progression. In neurological conditions, TGF-β modulates key pathological processes, including amyloid-beta (Aβ) dynamics in (AD). Dysfunctional TGF-β signaling in AD leads to impaired Aβ clearance by , resulting in increased amyloid deposition and neurodegeneration, whereas enhanced TGF-β1 activity promotes responses and Aβ . For (PD), TGF-β exerts neuroprotective effects through Smad-dependent pathways, supporting dopaminergic neuron maintenance and synaptic function; deficiency in neuronal TGF-β signaling accelerates nigrostriatal degeneration and motor deficits. Augmenting TGF-β signaling has shown potential to suppress , , and in PD models, underscoring its therapeutic promise. TGF-β also influences activation in (MS), where it contributes to a reparative signature in periplaque regions while modulating reactive in active lesions, bridging metabolic with autoimmune CNS pathology. In , post-seizure TGF-β signaling drives astrocyte-mediated and hyperexcitability; albumin leakage into the brain parenchyma activates TGF-β pathways, promoting epileptogenesis and chronic reactive changes. Recent 2020s research has expanded TGF-β's relevance to emerging conditions, including its role in long COVID-associated , where elevated TGF-β levels correlate with cognitive symptoms and immune dysregulation, potentially driving persistent brain inflammation via Epstein-Barr virus reactivation. In , ongoing investigations into TGF-β inhibitors aim to mitigate and renal decline, with preclinical and early clinical data supporting their use to slow disease progression in patients.

References

  1. [1]
    TGF-β signaling in health, disease and therapeutics - Nature
    Mar 22, 2024 · The signal transduction of TGF-β can stimulate diverse cellular responses and is particularly critical to embryonic development, wound healing, tissue ...
  2. [2]
    TGF-beta signal transduction: biology, function and therapy for ...
    Dec 19, 2022 · The transforming growth factor beta (TGF-β) is a crucial cytokine that get increasing concern in recent years to treat human diseases.
  3. [3]
    Structural biology of the TGFβ family - PMC - PubMed Central
    The transforming growth factor beta (TGFβ) superfamily controls several intricate signaling mechanisms that underlie every biological process from development ...Missing: paper | Show results with:paper
  4. [4]
    Biological Activity Differences between TGF-β1 and TGF-β3 ...
    Aug 25, 2014 · TGF-β1, -β2, and -β3 are small, secreted signaling proteins. They share 71–80% sequence identity and signal through the same receptors, ...Missing: percentage | Show results with:percentage
  5. [5]
    Targeting immunosuppression by TGF-β1 for cancer immunotherapy
    In this review, we discuss the biology of TGF-β1 with a special focus on its roles in regulating immune responses in the context of cancer.
  6. [6]
    TGF-β Superfamily Signaling in the Eye: Implications for Ocular ...
    TGF-β signaling participates in regulating several key developmental processes in the eye, including angiogenesis and neurogenesis.
  7. [7]
    Transforming growth factor beta (TGF‐β) isoforms in wound healing ...
    Dec 24, 2015 · TGF-β1 may mediate fibrosis in adults' wounds, while TGF-β3 may promote scarless healing in the fetus and reduced scarring in adults.
  8. [8]
    Transforming growth factor-β3 is required for secondary palate fusion
    Dec 7, 2013 · Previous studies suggested that TGF-β3 may play a crucial role during palatogenesis, Meckel's cartilage formation, cardiac morphogenesis, ...
  9. [9]
    Molecular Evolution of TGF-β Gene Family and Lamprey TGF-β2
    The molecular evolutionary analyses reveal that the lamprey TGF-β subfamily contains two members representing ancestors of TGF-β2 and 3 in vertebrates, ...
  10. [10]
    Human transforming growth factor-β complementary DNA sequence ...
    Aug 22, 1985 · The partial amino-acid sequence of purified human transforming growth factor-β (TGF-β) was used to identify a series of cDNA clones encoding the protein.
  11. [11]
    Structural Biology and Evolution of the TGF-β Family - PMC
    Proper disulfide bond formation is guided by noncovalent interactions surrounding the cysteines. ... Crystal structure of TGF-β 2 refined at 1.8 Å resolution.
  12. [12]
  13. [13]
    Site-directed mutagenesis of glycosylation sites in the transforming ...
    To determine the importance of N-linked glycosylation to the secretion of TGF beta 1 and -beta 2, site-directed mutagenesis was used to change the Asn residues ...
  14. [14]
    Crystal structure of transforming growth factor-beta 2 - PubMed
    Crystal structure of transforming growth factor-beta 2: an unusual fold for the superfamily. Science. 1992 Jul 17;257(5068):369-73. doi: 10.1126/science.
  15. [15]
    The crystal structure of TGF-beta 3 and comparison to TGF-beta 2
    Daopin S., Piez K. A., Ogawa Y., Davies D. R. Crystal structure of transforming growth factor-beta 2: an unusual fold for the superfamily. Science. 1992 Jul ...
  16. [16]
    Transforming growth factor β latency: A mechanism of cytokine ...
    This review article presents the current knowledge on intracellular L-TGF-β complex formation, secretion, matrix deposition, and activationMissing: paper | Show results with:paper
  17. [17]
    LTBPs, more than just an escort service - PMC - NIH
    When complexed with LAP, TGF-β cannot bind to its receptor. The LLC is secreted, binds to matrix components, and the latent TGF-β is activated by proteases/ ...
  18. [18]
    7040 - Gene ResultTGFB1 transforming growth factor beta 1 [ (human)]
    Sep 27, 2025 · TGFB1 mRNA expression and frequency of the + 869T>C and + 915G>C genetic variants: impact on risk for systemic sclerosis. Correlations of IL-6 ...
  19. [19]
    7043 - Gene ResultTGFB3 transforming growth factor beta 3 [ (human)]
    Sep 27, 2025 · The aim of the study was to determine temporal TGFB1, TGFB2 and TGFB3 gene expression profiles in the anterior lens capsule of paediatric ...
  20. [20]
    Regulation and Signaling of TGF-β Autoinduction - PMC - NIH
    Jun 6, 2022 · Through this positive feedback loop, specific cell types express TGF-β after being exposed to exogenous TGF-β stimulus. This autoregulatory ...
  21. [21]
    Smad3/AP-1 interactions control transcriptional responses to TGF ...
    We demonstrate that Smad and AP-1 complexes specifically bind to their cognate cis-elements and do not interact with each other on-DNA.
  22. [22]
    Platelet TGF-β1 contributions to plasma TGF-β1, cardiac fibrosis ...
    Circulating platelets contain high concentrations of TGF-β1 in their α-granules and release it on platelet adhesion/activation.
  23. [23]
    Expression of TGF‐βs in the embryonic nervous system: Analysis of ...
    May 22, 2008 · TGF-β2 and -β3 are expressed along radial glia cells, axonal tracts, and peripheral nerves as early as E12.5 and in neuronal cell bodies in the ...Neural Tube · Encephalon · Evidences Of Tgf-β1 Extra...
  24. [24]
    Hallmarks of cancer and AU‐rich elements - Khabar - WIREs RNA
    Jun 1, 2016 · TGFB, a key regulator of EMT, is itself coded by an ARE–mRNA that is subject to HuR-mediated mRNA stabilization.172 Another plausible player of ...
  25. [25]
    Transforming growth factor-beta1 circulates in normal human ...
    Normal human subjects had 4.1 +/- 2.0 ng/ml TGF-beta1 (range, 2.0-12.0; n = 42), <0.2 ng/ml TGF-beta2, and <0.1 ng/ml TGF-beta3 in their plasma. There were ...
  26. [26]
    TGFB1 - Transforming growth factor beta-1 proprotein | UniProtKB
    Sep 13, 2023 · TGFB1 is a proprotein that is a precursor to TGF-beta-1, which regulates cell growth and differentiation. It is also a precursor of LAP.
  27. [27]
    TGFB2 - Transforming growth factor beta-2 proprotein - UniProt
    Required to maintain the Transforming growth factor beta-2 (TGF-beta-2) chain in a latent state during storage in extracellular matrix (By similarity).
  28. [28]
    Prodomain-growth factor swapping in the structure of pro-TGF-β1
    Feb 2, 2018 · TGF-β is synthesized as a proprotein that dimerizes in the endoplasmic reticulum. After processing in the Golgi to cleave the N-terminal ...
  29. [29]
    Processing of Transforming Growth Factor β1 Precursor by Human ...
    To determine whether cleavage of pro-TGFβ1 can be achieved by the furin convertase in vitro, purified precursor was incubated in the presence of a truncated/ ...
  30. [30]
    Regulation of TGFβ and related signals by precursor processing
    This review summarizes information on the proteolytic processing of TGFβ and related precursors, and its spatiotemporal regulation by PCs during development ...
  31. [31]
    Making sense of latent TGFβ activation | Journal of Cell Science
    Jan 15, 2003 · The latent TGFβ complex is formed intracellularly and proTGFβ that fails to complex with LTBP is inefficiently secreted (Miyazono et al., 1991).
  32. [32]
    Transforming growth factor-beta 1 mutations in Camurati ... - PubMed
    Our data indicate that the mutations in the signal peptide and latency-associated peptide facilitate TGF-beta1 signaling, thus causing Camurati-Engelmann ...Missing: secretory vesicles
  33. [33]
    Transforming Growth Factor-β1 Mutations in Camurati-Engelmann ...
    Our data indicate that the mutations in the signal peptide and latency-associated peptide facilitate TGF-β1 signaling, thus causing Camurati-Engelmann disease.
  34. [34]
    Mechanism of activation of latent recombinant transforming growth ...
    Apr 1, 1990 · Activation of latent TGF beta 1 may occur by proteolytic nicking within the amino-terminal glycopeptide thereby causing a disruption of tertiary structure and ...
  35. [35]
    Plasmin regulates the activation of cell-associated latent TGF-beta 1 ...
    Our findings suggest that the secretion of an active form of TGF-beta 1 by alveolar macrophages is regulated by the generation of plasmin.
  36. [36]
    Cell surface-localized matrix metalloproteinase-9 proteolytically ...
    Our present observations indicate that MMP-9 and MMP-2 may provide alternative pathways for proteolytic activation of latent TGF-β in vivo.
  37. [37]
    Matrix Metalloproteinase 2 Activation of Transforming Growth Factor ...
    Jul 1, 2006 · MMP-2 increased TGF-β1 activity, collagen, and fibronectin within aortic rings or VSMCs from young rats to the levels that occur in old animals.
  38. [38]
    Active Transforming Growth Factor-β in Wound Repair - NIH
    Transforming growth factor (TGF)-β regulates wound repair and scarring in an isoform-specific fashion. TGF-β is produced in a latent form, ...
  39. [39]
    Interleukin-13 induces tissue fibrosis by selectively stimulating and ...
    Sep 17, 2001 · They also demonstrate that this activation is mediated by a plasmin/serine protease- and MMP-9-dependent and CD44-independent mechanism(s) and ...
  40. [40]
    The role of proteases in transforming growth factor-beta activation
    Aug 10, 2025 · This review describes the mechanism of protease driven TGF-beta activation, and discusses the physiological and pathological relevance of this ...<|control11|><|separator|>
  41. [41]
    The role of proteases in transforming growth factor-β activation
    Latent TGF-β can be activated by a number of physical processes including ... Camurati–Engelmann disease: New clinical insights in an Egyptian case report.
  42. [42]
    TGF-β signaling in fibrosis - PMC - PubMed Central
    TGF-β is upregulated and activated in fibrotic diseases and modulates fibroblast phenotype and function, inducing myofibroblast transdifferentiation while ...
  43. [43]
    Lysophosphatidic Acid Induces αvβ6 Integrin-Mediated TGF-β ...
    These studies demonstrate that LPA induces αvβ6 integrin-mediated TGF-β activation in epithelial cells via LPA2, Gαq, RhoA, and Rho kinase, and that this ...
  44. [44]
    Integrin α V β 6 -mediated activation of latent TGF-β requires the ...
    The mechanism of integrin-mediated latent TGF-β activation is not well understood. However, a direct interaction between αvβ6 and the RGD amino acid sequence ...
  45. [45]
    Integrin αvβ8–expressing tumor cells evade host immunity by ... - NIH
    Tumor cell expression of the integrin avb8 regulates TGF-b activation by immune cells and maintains an immunosuppressive microenvironment.Missing: hybrid | Show results with:hybrid
  46. [46]
    Integrin αvβ8–expressing tumor cells evade host immunity by ...
    Oct 18, 2018 · Binding of L-TGF-β to integrin αvβ8 results in activation of TGF-β. We engineered and used αvβ8 antibodies optimized for blocking or detection, ...
  47. [47]
    Regulation of the Bioavailability of TGF-β and TGF-β-Related Proteins
    2011). Unlike TGF-β1 and 3, TGF-β2 lacks an RGD motif and was not activated in experiments testing latent TGF-β activation by specific integrins (Annes et al. ...
  48. [48]
    Reciprocal regulation of TGF-β and reactive oxygen species
    TGF-β increases ROS production, leading to a redox imbalance. ROS, in turn, activate TGF-β, forming a cycle that mediates fibrosis.
  49. [49]
    Transforming Growth Factor-Beta and Oxidative Stress Interplay - NIH
    Meanwhile, ROS can influence TGF-β signaling and increase its expression as well as its activation from the latent complex. This way, both are building a strong ...
  50. [50]
    The Activation Sequence of Thrombospondin-1 Interacts with the ...
    We have identified thrombospondin-1 as a major physiological regulator of latent TGF-β activation. Activation is dependent on the interaction of a specific ...Missing: non- | Show results with:non-
  51. [51]
    Thrombospondin-1 regulation of latent TGF-β activation
    In this review, we will discuss the role of the matricellular protein, thrombospondin 1 (TSP-1), in regulation of latent TGF-β activation.
  52. [52]
    Thrombospondin-1 Is a Major Activator of TGF-β1 In Vivo
    Here we show that thrombospondin-1 is responsible for a significant proportion of the activation of TGF-β1 in vivo.
  53. [53]
    Photoactivation of Endogenous Latent Transforming Growth Factor ...
    May 28, 2014 · Together, these results indicate that ROS sensitivity in the LAP is required for TGF-β activation by LPL treatment. LPL treatment directs ...<|control11|><|separator|>
  54. [54]
    Signaling Receptors for TGF-β Family Members - PubMed Central
    Oligomeric structure of type I and type II transforming growth factor β receptors: Homodimers form in the ER and persist at the plasma membrane. J Cell Biol ...
  55. [55]
    Betaglycan can act as a dual modulator of TGF-beta ... - PubMed
    Betaglycan presents TGF-beta to receptors, but soluble betaglycan acts as a potent inhibitor of TGF-beta binding to membrane receptors.Missing: seminal paper
  56. [56]
    Endoglin Is an Accessory Protein That Interacts with the Signaling ...
    Endoglin cannot bind ligands on its own and does not alter binding to the kinase receptors. It binds TGF-β1 and -β3 by associating with the TGF-β type II ...
  57. [57]
    TGF-β Signaling from Receptors to Smads - PMC - PubMed Central
    In this review, we first introduce some basic components of the TGF-β signaling pathways and their actions, and then discuss posttranslational modifications and ...
  58. [58]
    Review Dynamics of TGF-β/Smad signaling - ScienceDirect.com
    Jul 4, 2012 · TGF-β signaling is initiated by the binding of TGF-β to TβRI and TβRII. The activation of the ligand–receptor complex is a relatively fast step ...
  59. [59]
    TGFβ signaling pathways in human health and disease - Frontiers
    Transforming growth factor beta (TGFβ), which has been studied for more than 30 years, is multifaceted molecule with a myriad of different activities.
  60. [60]
    TGF-beta signal transduction: biology, function and therapy for ...
    Dec 19, 2022 · Generally, latent TGF-β is stored in the multiple extracellular matrix. Under enzymatic and non-enzymatic action, latent TGF transforms into ...<|control11|><|separator|>
  61. [61]
    Non-Smad pathways in TGF-β signaling - PMC - NIH
    These non-Smad pathways include various branches of MAP kinase pathways, Rho-like GTPase signaling pathways, and phosphatidylinositol-3-kinase/AKT pathways.
  62. [62]
    Non-Smad Signaling Pathways of the TGF-β Family - PMC
    Non-Smad pathways include activation of Erk MAPK, JNK, p38 MAPK, IKK, PI3K-Akt, and Rho-like GTPases, in response to TGF-β.
  63. [63]
    TGF-beta-induced apoptosis is mediated by the adapter protein ...
    Daxx is involved in mediating JNK activation by TGF-beta. Our findings associate Daxx directly with the TGF-beta apoptotic-signalling pathway.
  64. [64]
    Death-associated Protein 6 (Daxx) Mediates cAMP-dependent ...
    Daxx mediates apoptosis stimulated by the death receptor Fas (16), UV irradiation (17), or TGF-β signaling (18). However, Daxx also possesses anti-apoptotic ...Hipk3 Interacts With And... · Daxx Mediates Hipk3 Effect... · S669a Mutant Of Daxx Fails...<|control11|><|separator|>
  65. [65]
    TGF-β activates Erk MAP kinase signalling through direct ...
    We report that upon TGF-β stimulation, the activated TGF-β type I receptor (TβRI) recruits and directly phosphorylates ShcA proteins on tyrosine and serine.Tgf-β Rapidly Induces... · Shca Proteins Interact With... · Tgf-β Receptors Directly...
  66. [66]
    TGF-β promotes PI3K-AKT signaling and prostate cancer ... - PubMed
    Jul 4, 2017 · We found that TGF-β activated PI3K in a manner dependent on the activity of the E3 ubiquitin ligase tumor necrosis factor receptor-associated factor 6 (TRAF6).
  67. [67]
    TRAF6 mediates Smad-independent activation of JNK and p38 by ...
    TGF-β induces K63-linked ubiquitination of TRAF6, and promotes association between TRAF6 and TAK1. Our results indicate that TGF-β activates JNK and p38 through ...
  68. [68]
    TGF-β and Smad3 modulate PI3K/Akt signaling pathway in vascular ...
    Our studies reveal a novel pathway whereby TGF-β/Smad3 stimulates SMC proliferation through p38 and Akt.
  69. [69]
    Transforming Growth Factor-β1 Mediates Epithelial to Mesenchymal ...
    The data suggest that TGF-β rapidly activates RhoA-dependent signaling pathways to induce stress fiber formation and mesenchymal characteristics.
  70. [70]
    The type I TGF-beta receptor engages TRAF6 to activate TAK1 in a ...
    Our data show that TGF-beta specifically activates TAK1 through interaction of TbetaRI with TRAF6, whereas activation of Smad2 is not dependent on TRAF6.
  71. [71]
    TGF-β signaling via TAK1 pathway: Role in kidney fibrosis - PMC
    The type I TGF-beta receptor engages TRAF6 to activate TAK1 in a receptor kinase-independent manner. Nat Cell Biol. 2008;10:1199–207. doi: 10.1038/ncb1780 ...
  72. [72]
    Targeting fibrosis: mechanisms and clinical trials - Nature
    Jun 30, 2022 · Activated TGF-β1/Smad3 signaling pathway promoted the recruitment of fibroblasts to injury sites and mediated fibroblast-to-myofibroblast ...
  73. [73]
    A single day of TGF-β1 exposure activates chondrogenic ... - Nature
    Jan 4, 2021 · We found that a single day of TGF-β1 exposure was sufficient to trigger BMSC chondrogenic differentiation and tissue formation, similar to 21 days of TGF-β1 ...
  74. [74]
    Roles of TGFβ in metastasis | Cell Research - Nature
    Dec 2, 2008 · p15INK4B is a potential effector of TGF-beta-induced cell cycle arrest. Nature 1994; 371:257–261. CAS PubMed Google Scholar. Scandura JM ...Missing: G1 | Show results with:G1
  75. [75]
  76. [76]
    TGF-β signaling in health and disease - ScienceDirect
    Sep 14, 2023 · TGF-β signaling controls metazoan embryo development, tissue homeostasis, and injury repair through coordinated effects on cell proliferation, ...
  77. [77]
    TGF-β and Regulatory T Cell in Immunity and Autoimmunity - PMC
    Recent studies revealed the positive roles for TGF-β and Treg cells in shaping the immune system and the inflammatory responses.
  78. [78]
    The Critical Contribution of TGF-β to the Induction of Foxp3 ... - NIH
    Abstract. Stimulation of mouse CD4+ T cells in the presence of TGF-β results in the expression of Foxp3 and induction of Treg function.
  79. [79]
    TGF-β and IL-6 signals modulate chromatin binding and ... - PNAS
    Sep 16, 2008 · Here we show that TCR stimuli and TGF-β signals modulate the disposition of FOXP3 into different subnuclear compartments, leading to enhanced chromatin binding.
  80. [80]
    The role of TGF-β in growth, differentiation, and maturation of B ...
    TGF-β inhibits proliferation and stimulates apoptosis. The initial studies on the effects of TGF-β on human B-cell growth and differentiation maintained its ...
  81. [81]
    TGFβ signaling plays a critical role in promoting alternative ...
    Jun 15, 2012 · Our results establish a novel biological role for TGFβ signaling in controlling expression of genes characteristic for alternatively activated macrophages.
  82. [82]
    Manipulation of TGF-β to control autoimmune and chronic ...
    The critical role of TGF-β in oral tolerance and the concept of `bystander suppression' were established by Weiner and his colleagues [21], [25] and confirmed ...Missing: evasion | Show results with:evasion
  83. [83]
    TGFβ control of immune responses in cancer - NIH
    Nov 9, 2023 · These findings suggest that Treg cells may suppress CTL-mediated cancer immunity by presenting and activating TGFβ1 in the tumour ...
  84. [84]
    Pivotal Role of TGF-β/Smad Signaling in Cardiac Fibrosis - NIH
    ... collagen I, collagen III, and α-SMA → ↑CF, CFs, (64). miR-9, Antifibrosis ... ↓TGF-β /Smad3 → ↓collagen I, III, fibronectin → ↓CF, AngII-triggered ...
  85. [85]
    The role of Smad2 and Smad3 in regulating homeostatic functions of ...
    Smad3 knockdown attenuated collagen I, collagen IV and fibronectin mRNA synthesis and reduced expression of the matricellular protein thrombospondin-1.3. Results · 3.2. Smad3 Mediates Collagen... · 3.5. Role Of Smad2 And Smad3...
  86. [86]
    Cross Talk among TGF-β Signaling Pathways, Integrins, and the ...
    Pro-TGF-β cannot be activated unless it is first processed at the PPC site. Furin cleaves pro-TGF-β extracellularly (other proteases, e.g., plasmin, may as well) ...Missing: RXXR | Show results with:RXXR
  87. [87]
    The role of TGFβ in wound healing pathologies - ScienceDirect.com
    TGFβ exerts pleiotropic effects on wound healing by regulating cell proliferation, differentiation, ECM production, and modulating the immune response.Review · 2. Role Of Tgfβ In Wound... · Acknowledgements
  88. [88]
    Critical Role of Transforming Growth Factor Beta in Different Phases ...
    This study concluded that the functionality of TGF-β1 and its receptors are critical in the wound healing process. The release of TGF-β1 at an early stage of ...Roles Of Tgf-β In Wound... · The Tgf-β Superfamily And... · The Role Of Tgf-β2 And -β3...
  89. [89]
    Targeting TGF-β signal transduction for fibrosis and cancer therapy
    Apr 23, 2022 · TGF-β I- III all have fibrogenic effects and share 70–82% homology at the amino acid level [64]. TGF-β I is considered as the primary factor in ...
  90. [90]
    Diverse functions of matrix metalloproteinases during fibrosis - PMC
    Here, we discuss data demonstrating or suggesting varied roles for specific MMPs in fibrosis of liver, lung and kidney derived from experimental models.
  91. [91]
    Endoglin: Beyond the Endothelium - PMC - NIH
    Extensive work has shown the important roles that endoglin plays in balancing the TGF-β signaling pathway, thereby regulating endothelial cell proliferation and ...Endoglin: Beyond The... · 2. Endoglin Structure And... · 2.2. Endoglin And...
  92. [92]
    Regulation of endothelial cell plasticity by TGF-β - PMC - NIH
    TGF-β induced ALK5 signaling leads to Smad2 and Smad3 phosphorylation resulting in inhibition of angiogenesis by inhibiting endothelial cell proliferation, ...Tgf-β Signaling · Endmt In Development · Endmt In Tissue Fibrosis
  93. [93]
    A Review of Fetal Scarless Healing - PMC - NIH
    Whereas, the addition of exogenous TGF-β3 has in some animal models shown improved scar formation [74]. Further, early human clinical studies showed that ...
  94. [94]
    Scarless wound healing: Transitioning from fetal research to ...
    The TGFB subtypes 1 and 3 are perhaps the most important in wound healing, with TGFB1 being involved in the scarring mechanism and implicated in the development ...
  95. [95]
    SB-431542 is a potent and specific inhibitor of transforming growth ...
    SB-431542 is a potent and specific inhibitor of transforming growth factor-beta superfamily type I activin receptor-like kinase (ALK) receptors ALK4, ALK5, and ...
  96. [96]
    SB 431542 | TGF-β Receptors - Tocris Bioscience
    Rating 5.0 (8) SB 431542 is a potent and selective inhibitor of the transforming growth factor-β (TGF-β) type I receptor/ALK5 (IC 50 = 94 nM), and its relatives ALK4 and ALK7.
  97. [97]
    beta1-induced extracellular matrix with a novel inhibitor of the TGF ...
    SB-431542 is a selective inhibitor of Smad3 phosphorylation with an IC50 of 94 nM. It inhibited TGF-beta1-induced nuclear Smad3 localization.
  98. [98]
    Phase 1 study of galunisertib, a TGF-beta receptor I kinase inhibitor ...
    Galunisertib (LY2157299), a small molecule inhibitor of TGF-beta RI serine/threonine kinase, had antitumor effects with acceptable safety/tolerability.
  99. [99]
    Phase 1b/2a study of galunisertib, a small molecule inhibitor of ...
    Mar 5, 2020 · This Phase 1b/2a trial investigated the clinical benefit of combining galunisertib with temozolomide-based radiochemotherapy (TMZ/RTX) in patients with newly ...
  100. [100]
    A phase 1, single-dose study of fresolimumab, an anti-TGF-β ...
    Single-dose fresolimumab was well tolerated in patients with primary resistant FSGS. Additional evaluation in a larger dose-ranging study is necessary.
  101. [101]
    Phase I study of GC1008 (fresolimumab): a human anti-transforming ...
    Mar 11, 2014 · GC1008 is a human anti-TGFβ monoclonal antibody that neutralizes all isoforms of TGFβ. Here, the safety and activity of GC1008 was evaluated in patients with ...
  102. [102]
    Bintrafusp Alfa, a Bifunctional Fusion Protein Targeting TGF-β and ...
    Mar 17, 2023 · Bintrafusp alfa is a first-in-class bifunctional fusion protein composed of the extracellular domain of transforming growth factor beta receptor II (a TGF-β ...
  103. [103]
    Bintrafusp alfa, an anti-PD-L1:TGF-β trap fusion protein, in patients ...
    Sep 14, 2022 · Bintrafusp alfa (BA) is a bifunctional fusion protein composed of the extracellular domain of the TGF-βRII receptor ("TGF-β trap") and anti-PD- ...
  104. [104]
    Natural inhibitor of transforming growth factor-beta protects against ...
    Nov 26, 1992 · We report here that administration of decorin inhibits the increased production of extracellular matrix and attenuates manifestations of disease.
  105. [105]
    Decorin suppresses transforming growth factor-beta ... - PubMed
    Naturally occurring inhibitors of this TGFbeta-dependent fibrotic response include decorin, a small leucine-rich proteoglycan. While the mechanism by which ...
  106. [106]
    Localization of transforming growth factor beta and its natural ...
    Decorin, a naturally occurring chondroitin-dermatan sulphate proteoglycan which binds TGF beta can inhibit its activity. In this study, immunohistochemical ...
  107. [107]
    Cardiac Safety of TGF-β Receptor I Kinase Inhibitor LY2157299 ...
    Toxicology studies with TGF-β inhibitors that block TGF-β RI/ALK5 have reported toxicities that share the cardiovascular findings in LDS patients [12]. Such ...Missing: challenges | Show results with:challenges
  108. [108]
    Hitting the Target! Challenges and Opportunities for TGF-β Inhibition ...
    Feb 20, 2024 · However, direct TGF-β blockade may lead to uncontrolled inflammation, especially following myocardial infarction, while interference with the ...Missing: toxicity | Show results with:toxicity
  109. [109]
    Therapeutic Targets for the Treatment of Cardiac Fibrosis and Cancer
    In this review, we explain how inhibitors of TGF-β signaling can work as antifibrotic therapy. After cardiac injury, profibrotic mediators such as TGF-β, ...Abstract · Introduction · TGF-β Signal Transduction · TGF-β Inhibitors for the...
  110. [110]
    The TGF-β mimic TGM4 achieves cell specificity through ...
    TGF-β Mimics (TGM) are modular parasite proteins which bind multiple host cell receptors. •. TGM4 interacts, in addition to both TGF-β receptors, with CD44, ...
  111. [111]
  112. [112]
    Augmentation of transforming growth factor-β signaling for the ...
    To date, 11 studies (besides ours) have tested C381 (previously called SRI-011381) as a tool to activate TGF-β signaling. Results from these studies confirm ...Missing: agonist | Show results with:agonist
  113. [113]
    A Short Peptide That Mimics the Binding Domain of TGF-β1 ...
    Aug 27, 2015 · Our aim was to develop novel peptides through phage display (PhD) technology that could mimic TGF-β1 function with higher potency. Specific ...
  114. [114]
    rAAV-mediated overexpression of TGF-β stably restructures human ...
    The present findings show the ability of rAAV-mediated TGF-β gene transfer to directly remodel human OA cartilage by activating the biological, reparative ...
  115. [115]
    The roles and regulatory mechanisms of TGF-β and BMP signaling ...
    Jan 24, 2024 · Besides bone matrix proteins, the latency of TGF-βs is also maintained by LTBPs, which bind TGF-β precursors to form the LLC. Active TGF-β ...
  116. [116]
    TGF-β as A Master Regulator of Aging-Associated Tissue Fibrosis
    TGF-β can modulate aging-related fibrosis by regulating inflammation, DNA damage, oxidative stress, and cellular senescence. Moreover, senescent cells can ...
  117. [117]
    TGF-β inhibitors: the future for prevention and treatment of liver ...
    This review systematically summarizes the role of TGF-β in liver fibrosis and details the research progress of TGF-β-targeted inhibitors.
  118. [118]
    miR-29b participates in early aneurysm development in Marfan ...
    Jan 20, 2012 · Underlying fibrillin-1 gene mutations cause increased transforming growth factor-β (TGF-β) signaling. Although TGF-β blockade prevents aneurysms ...<|separator|>
  119. [119]
    Molecular pathogenesis of Marfan syndrome - PubMed
    While FBN1 mutations, abnormal transforming growth factor-β signaling and dysregulated matrix metalloproteinases have been implicated in MFS, clinically ...
  120. [120]
    Germline TGF-beta receptor mutations and skeletal fragility - PubMed
    We present two patients with LDS with significant skeletal fragility. The first is a 17-year-old male who had talipes equinovarus, diaphragmatic and inguinal ...<|separator|>
  121. [121]
    Activation of TGF-β signaling in an aortic aneurysm in a patient with ...
    Jan 18, 2019 · The TGF-β signaling pathway in the aortic wall is upregulated in LDS patients, although the causative variants of TGFBR1 and TGFBR2 are missense ...<|separator|>
  122. [122]
    Decreased Smad 7 expression contributes to cardiac fibrosis in the ...
    In 2 and 4 wk post-MI hearts, Smad 7 and TbetaRI expression were decreased in scar tissue, whereas TGF-beta(1) expression was increased in scar and viable ...
  123. [123]
    Transforming Growth Factor (TGF)-β signaling in cardiac remodeling
    In the pressure overloaded heart TGF-β signaling appears to regulate matrix metabolism and cardiac hypertrophy. Administration of an anti-TGF-β neutralizing ...
  124. [124]
    TGF-β 1 and angiotensin networking in cardiac remodeling
    This review focuses on recent advances in the understanding, how Ang II and TGF-β1 are connected in the pathogenesis of cardiac hypertrophy and dysfunction.
  125. [125]
    Elevated systemic TGF-β impairs aortic vasomotor function through ...
    Our results demonstrate that elevated circulating TGF-β1 causes endothelial dysfunction through NOX activation-induced oxidative stress, accelerating ...Results · Tgf-β Plasma Levels And... · Discussion<|separator|>
  126. [126]
    Transforming growth factor-β and atherosclerosis - PubMed Central
    TGF-β is a key modulator of normal and abnormal vascular repair and that dysfunctions in this pathway promote a pro-inflammatory, pro-fibrotic and pro- ...
  127. [127]
    TGF-β Isoforms and GDF-15 in the Development and Progression of ...
    High levels of TGF-β2 in plaque were associated with plaque stability and a lower risk of future acute cardiovascular (CV) events. TGF-β2 has been shown to be a ...
  128. [128]
    Mechanisms and treatment of aortic aneurysm in Marfan syndrome
    FBN1 mutations result in FBN1 fragments that also contribute to elastic fiber fragmentation. Although recent research has advanced our understanding of MFS, the ...
  129. [129]
    Anti-TGFβ (Transforming Growth Factor β) Therapy With Betaglycan ...
    P144 prevents the onset of aortic aneurysm but not its progression. Results indicate the importance of reducing the excess of active TGFβ signaling.<|control11|><|separator|>
  130. [130]
    TGF-β: The missing link in obesity-associated airway diseases? - PMC
    With increased body fat mass in obesity, the profibrotic cytokine transforming growth factor beta β1 (TGF-β1) levels increase in the adipose tissue, suggesting ...
  131. [131]
    Protection from obesity and diabetes by blockade of TGF-β/Smad3 ...
    Positive energy balance ensues when energy intake exceeds energy expenditure leading to net storage of excess calories in the form of fat in the adipose tissue.
  132. [132]
    Recent advances in targeting obesity, with a focus on TGF-β ...
    Apr 30, 2025 · High-fat diet, Resistant to adipose tissue hypertrophy, liver steatosis, and insulin resistance (20 wks), (Lee et al. 2023). TGF-β3, TGF-β1Lβ3/ ...
  133. [133]
    Protection from Obesity and Diabetes by Blockade of TGF-β/Smad3 ...
    Jul 6, 2011 · Interestingly, Smad3−/− mice display significantly reduced fat mass (Figure 2B), which, taken together with the enhanced glucose uptake in the ...
  134. [134]
    Dysfunction of TGF-β signaling in Alzheimer's disease - JCI
    Nov 1, 2006 · Decreased TGF-β signaling in neurons may also increase Aβ levels, enhancing amyloid deposition. Thus, the inflammatory response to Aβ deposits ...
  135. [135]
    The Amyloid-β Pathway in Alzheimer's Disease | Molecular Psychiatry
    Aug 30, 2021 · TGF-β1 is a neurotrophic factor displaying both anti-inflammatory and neuroprotective actions stimulating Aβ clearance by microglia [252, 253].
  136. [136]
    The promise of the TGF-β superfamily as a therapeutic target for ...
    With respect to Parkinson's disease (PD), TGF-ß signaling pathways are implicated in the differentiation, maintenance and synaptic function of the dopaminergic ...
  137. [137]
    Deficiency in Neuronal TGF-β Signaling Leads to Nigrostriatal ...
    We show that reducing Transforming growth factor-β (TGF-β) signaling promotes Parkinson disease-related pathologies and motor deficits.
  138. [138]
    Augmentation of transforming growth factor-β signaling for...
    It has been shown that TGF-β mediates its neuroprotective effects through multiple mechanisms such as suppression of inflammation, apoptosis, and excitotoxicity ...
  139. [139]
    Irrespective of Plaque Activity, Multiple Sclerosis Brain Periplaques ...
    Nov 30, 2022 · Irrespective of Plaque Activity, Multiple Sclerosis Brain Periplaques Exhibit Alterations of Myelin Genes and a TGF-Beta Signature.
  140. [140]
    Albumin induces excitatory synaptogenesis through astrocytic TGF-β ...
    Injection of albumin into the ventricles of mice results in gliosis and seizures. · TGF-β1 and albumin have similar epileptogenic effects. · Albumin and TGF-β1 ...
  141. [141]
    Neuroinflammatory mechanisms of post-traumatic epilepsy
    Jun 17, 2020 · Therefore, TGFβ causes hyperexcitability and seizures, and blocking TGFβ signaling prevents the development of acquired epilepsies. This ...
  142. [142]
    The Molecular Mechanisms of Cognitive Dysfunction in Long COVID
    May 26, 2025 · In addition to canonical inflammatory mediators, elevated TGF-β has been identified in Long COVID patients with cognitive symptoms, supporting ...Missing: 2020s | Show results with:2020s
  143. [143]
    Science Spotlight: TGFβ drives long COVID inflammation via EBV
    Mar 19, 2025 · A study in Nature suggests that TGFβ-induced immune suppression and subsequent reactivation of Epstein-Barr virus together drive the extreme ...Missing: 2020s | Show results with:2020s
  144. [144]
    TGF-β: elusive target in diabetic kidney disease - PMC
    Apr 3, 2025 · This paper reviews the basic details of TGF-β as a cytokine, its role in DKD, and updates on research carried out to validate its candidacy.Missing: 2020s | Show results with:2020s
  145. [145]
    Anti–TGF-β1 Antibody Therapy in Patients with Diabetic Nephropathy
    TGF-β1 mAb is safe and more effective than placebo in slowing renal function loss in patients with diabetic nephropathy on chronic stable renin-angiotensin ...Missing: 2020s | Show results with:2020s