Transforming Growth Factor Beta (TGF-β): Mechanisms, Pathways, and Therapeutic Applications

Transforming Growth Factor Beta (TGF-β): Mechanisms, Pathways, and Therapeutic Applications

Transforming Growth Factor Beta (TGF-β) is a multifunctional cytokine that plays crucial roles in numerous biological processes, from tissue development to immune regulation. This pleiotropic growth factor represents one of the most extensively studied signaling molecules due to its complex and sometimes contradictory functions in health and disease. Its diverse effects on cell growth, differentiation, immune function, and tissue remodeling make it a critical mediator in both physiological homeostasis and pathological conditions.

The Nature and Structure of TGF-β

TGF-β belongs to a superfamily of growth factors that control the development and homeostasis of most tissues in metazoan organisms2. Three primary isoforms exist in mammals (TGF-β1, TGF-β2, and TGF-β3), with TGF-β1 being the most abundant and well-studied. TGF-β is synthesized as a precursor molecule containing a propeptide region called the latency-associated peptide (LAP). The association between LAP and TGF-β keeps the cytokine in an inactive state, requiring specific activation mechanisms before it can exert its biological effects19.

An important characteristic of TGF-β is its storage in the extracellular matrix (ECM), particularly through its association with latent TGF-β-binding protein-1 (LTBP1)15. This matrix-bound storage creates a reservoir of latent TGF-β that can be rapidly mobilized in response to tissue injury or other stimuli, allowing for precise spatial and temporal control of TGF-β activity. The LTBPs play a crucial role in the assembly, secretion, and targeting of latent TGF-β complexes to the extracellular matrix, thereby regulating TGF-β availability and activation18.

TGF-β Signaling Mechanisms and Pathways

The signaling pathway of TGF-β involves a series of highly regulated steps that ultimately result in changes in gene expression. Understanding this pathway is essential for appreciating both the normal functions of TGF-β and how its dysregulation contributes to disease states.

Receptor Activation and Signal Transduction

TGF-β signals through a unique mechanism involving two types of serine/threonine kinase receptors: type I (TβRI) and type II (TβRII). The signaling cascade begins when TGF-β binds directly to TβRII, which is a constitutively active kinase on the cell surface7. This binding creates a recognition site for TβRI, which is then recruited into the complex. Once in proximity, TβRII phosphorylates TβRI, activating its kinase domain. The activated TβRI then propagates the signal to downstream substrates, primarily members of the SMAD protein family7.

SMAD-Dependent Signaling Pathway

The canonical TGF-β signaling pathway involves SMAD proteins, which are the major intracellular mediators. Following receptor activation, TβRI phosphorylates receptor-regulated SMADs (R-SMADs), typically SMAD2 and SMAD3 for TGF-β. Phosphorylated R-SMADs then associate with the common mediator SMAD (co-SMAD), SMAD4, forming complexes that translocate to the nucleus2. In the nucleus, these SMAD complexes associate with various DNA-binding partners and transcriptional co-activators or co-repressors to regulate the expression of target genes2.

The specificity and diversity of TGF-β responses are achieved through cell-type-specific expression of different SMAD proteins and their DNA-binding partners. This explains how the same signaling pathway can produce drastically different outcomes in different cellular contexts2.

SMAD-Independent Signaling Pathways

Besides the canonical SMAD pathway, TGF-β can also activate SMAD-independent pathways, including various branches of MAP kinase pathways (ERK, JNK, and p38), phosphatidylinositol-3-kinase (PI3K)/AKT pathways, and small GTPases. These non-canonical pathways contribute to the diverse cellular responses elicited by TGF-β and can sometimes counteract or enhance SMAD-dependent signaling10.

Activation Mechanisms of Latent TGF-β

Since TGF-β is secreted and stored in an inactive latent form, its activation represents a crucial regulatory step. Several mechanisms have been identified:

Integrin-Mediated Activation

The integrin αvβ6, expressed primarily on epithelial cells, binds to and activates latent TGF-β1. This mechanism is particularly important in regulating pulmonary inflammation and fibrosis1. By binding to specific sequences in the LAP component of latent TGF-β complexes, integrins can induce conformational changes that release active TGF-β.

Proteolytic Activation

Matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9, can proteolytically cleave and activate latent TGF-β. This represents an important mechanism by which TGF-β can promote tumor invasion and angiogenesis4. MMP-9 can localize to the cell surface through binding to CD44, and this localization is required for its ability to promote tumor invasion and angiogenesis through TGF-β activation4.

Osteoclast-Mediated Release from Bone Matrix

In bone, TGF-β is stored in the extracellular matrix through its association with LTBP1. Osteoclasts, the cells responsible for bone resorption, can release matrix-bound TGF-β through proteolytic cleavage of LTBP115. This mechanism provides a means by which TGF-β stored in bone matrix can be mobilized during bone remodeling or in pathological conditions affecting bone.

Diverse Biological Functions of TGF-β

TGF-β affects virtually all cell types and orchestrates a wide array of cellular processes, reflecting its pleiotropic nature. These functions can be broadly categorized into several key areas:

Regulation of Immune Responses

TGF-β is a master regulator of immune responses, generally exerting immunosuppressive effects16. It inhibits the proliferation and differentiation of various immune cells, including T cells, B cells, and NK cells. Additionally, TGF-β promotes the development of regulatory T cells (Tregs), which are crucial for maintaining immune tolerance. CD4+CD25+ regulatory T cells produce high levels of TGF-β1, which contributes to their suppressive activity on other immune cells19.

In the context of cancer, the immunosuppressive properties of TGF-β can be co-opted to promote tumor growth by helping cancer cells evade immune surveillance16. TGF-β creates an immunosuppressive microenvironment that enables tumor progression, highlighting its dual role in both normal physiology and disease states.

Tissue Fibrosis and Remodeling

TGF-β is a potent inducer of extracellular matrix production and is heavily implicated in fibrotic diseases. It stimulates the synthesis of matrix proteins such as collagen and fibronectin while inhibiting the expression of matrix-degrading enzymes5. This property makes TGF-β a central mediator in the pathogenesis of fibrotic disorders affecting various organs, including the liver, kidney, heart, and lungs.

In diabetic nephropathy, TGF-β promotes renal fibrosis and is recognized as a key mediator of disease progression11. Similarly, in Chagas disease, elevated TGF-β levels contribute to heart fibrosis and cardiac remodeling, leading to poor clinical outcomes8. The pro-fibrotic effects of TGF-β highlight its importance in wound healing but also its pathological role when dysregulated.

Cell Growth, Differentiation, and Apoptosis

TGF-β exerts complex effects on cell growth and survival, often in a context-dependent manner. In epithelial cells, TGF-β typically induces growth arrest and can promote apoptosis, functioning as a tumor suppressor in early stages of carcinogenesis10. However, in advanced cancers, tumor cells often become resistant to the growth-inhibitory effects of TGF-β while retaining sensitivity to its pro-metastatic and pro-angiogenic functions13.

TGF-β is also a key regulator of epithelial-mesenchymal transition (EMT), a process in which epithelial cells lose their characteristic properties and acquire a mesenchymal phenotype12. EMT is crucial during embryonic development and wound healing but can also contribute to fibrosis and cancer metastasis when inappropriately activated.

Angiogenesis and Tissue Homeostasis

TGF-β plays important roles in angiogenesis, the formation of new blood vessels. It can promote endothelial cell proliferation and migration, as well as the recruitment of pericytes for vascular stabilization4. These effects on blood vessel formation are particularly relevant in cancer, where TGF-β contributes to tumor angiogenesis and subsequent metastasis.

In the liver, TGF-β is crucial for both physiology and pathophysiology. It helps maintain tissue homeostasis under normal conditions but can induce hepatocyte apoptosis when present at high concentrations5. Increased TGF-β levels are observed in various liver diseases, including fibrosis, inflammation, and steatosis, suggesting its potential as a diagnostic and prognostic marker5.

Regulation of TGF-β Signaling

Given the potent and diverse effects of TGF-β, its signaling is tightly regulated at multiple levels to prevent inappropriate activation and maintain tissue homeostasis.

Inhibitory SMADs

Inhibitory SMADs, particularly SMAD7, act as negative regulators of TGF-β signaling. SMAD7 inhibits TGF-β signaling by competing with R-SMADs for receptor binding and by recruiting ubiquitin ligases that target the receptors for degradation20. The expression of SMAD7 can be induced by various stimuli, including TGF-β itself, creating a negative feedback loop that limits the duration and intensity of signaling.

Cross-talk with Other Signaling Pathways

TGF-β signaling doesn't operate in isolation but interacts with numerous other pathways to achieve context-dependent responses. A notable example is the antagonism between TGF-β and NF-κB signaling pathways. The RelA subunit of NF-κB can inhibit TGF-β-induced SMAD signaling by upregulating SMAD7 expression17. This antagonism has important implications for inflammatory responses, as NF-κB is activated by pathogenic and pro-inflammatory stimuli that often exert effects opposite to those of TGF-β.

Therapeutic Approaches Targeting TGF-β: Proven and Unproven

Given its involvement in numerous pathological conditions, TGF-β has emerged as an attractive therapeutic target. However, due to its pleiotropic nature and context-dependent effects, targeting TGF-β presents significant challenges.

Promising Therapeutic Approaches

TGF-β Inhibition in Cancer Treatment

TGF-β inhibition has shown promise in cancer treatment, particularly as a means to enhance the efficacy of immunotherapies16. By blocking the immunosuppressive effects of TGF-β, such inhibitors can potentially reinvigorate anti-tumor immune responses. TGF-β status may serve as a biomarker to predict responsiveness to immunotherapies, with high TGF-β expression often associated with resistance to immune checkpoint inhibitors16.

Trabedersen, an antisense oligonucleotide targeting TGF-β2 mRNA, has demonstrated efficacy in clinical trials for treating certain cancers, including pancreatic cancer, melanoma, and glioblastoma3. The favorable tolerability profile of Trabedersen makes it an attractive candidate for further development.

Combination Therapies

Combination approaches that target TGF-β along with other pathways have shown synergistic effects. For instance, combining a TGF-β inhibitor with IL-2 (Proleukin) has demonstrated anti-tumor effects by both diminishing immunosuppression and enhancing immune cell function3. Such combination strategies may overcome the limitations of monotherapies and potentially address the complex, multifaceted nature of diseases like cancer.

TGF-β Signaling Inhibitors in Chagas Disease

In Chagas disease, a parasitic infection that can lead to severe cardiac complications, inhibition of TGF-β signaling has shown promising results in pre-clinical studies. In chronic T. cruzi infected mice, TGF-β pathway inhibitors improved cardiac electrical parameters, reversed the loss of connexin-43, reduced fibrosis of cardiac tissue, and restored transcription of key cardiac factors8. These findings suggest a potential therapeutic approach for a disease with limited treatment options.

Less Proven Therapeutic Approaches

Small Molecule TGF-β Receptor Inhibitors

Several small molecule inhibitors targeting TGF-β receptors are in development, with varying degrees of success. LY2157299 monohydrate, an oral TGF-β receptor I kinase inhibitor, has been evaluated in combination with gemcitabine for patients with advanced cancer9. While such approaches show promise in preclinical models, their clinical efficacy and safety profiles require further validation.

Repurposing Existing Drugs

Sorafenib, a multi-kinase inhibitor approved for the treatment of several solid tumors, has been shown to inhibit TGF-β-induced epithelial-mesenchymal transition and tissue fibrosis12. The mechanism involves decreasing TGF-β type II receptor levels in liver epithelial cells through lipid raft/caveolae-mediated endocytosis. However, this effect differs between epithelial and stellate cells, highlighting the complexity of targeting TGF-β signaling in a cell-specific manner12.

Anti-TGF-β Treatment in Diabetic Nephropathy

Anti-TGF-β therapy for diabetic nephropathy remains controversial due to the diverse roles of TGF-β1 in this condition11. While TGF-β promotes renal fibrosis, it also has important homeostatic functions in the kidney. Understanding the regulatory role and mechanisms of TGF-β in the pathogenesis of diabetic nephropathy is still an ongoing process, and more targeted approaches may be needed to achieve therapeutic benefits without disrupting beneficial TGF-β functions.

Selenium Supplementation in Chagas Disease

Selenium, an essential microelement with various biological functions, has been proposed as a complementary therapy in Chagas disease. Experimental and clinical data suggest that selenium may help prevent heart failure and improve cardiac function, possibly by modulating TGF-β-mediated fibrosis14. While promising, the precise mechanisms and optimal dosing strategies for selenium supplementation in this context require further investigation.

Conclusion

Transforming Growth Factor Beta (TGF-β) represents a multifaceted cytokine with diverse and sometimes contradictory functions in health and disease. Its complex signaling mechanisms, involvement in numerous biological processes, and context-dependent effects highlight both its physiological importance and its potential as a therapeutic target.

Significant progress has been made in understanding the molecular mechanisms of TGF-β signaling, from receptor activation to downstream effectors and cross-talk with other pathways. This knowledge has facilitated the development of various therapeutic strategies targeting TGF-β in conditions ranging from cancer to fibrotic disorders. While some approaches have shown promise, particularly in cancer immunotherapy and certain infectious diseases, others remain in early stages of development or face challenges due to the pleiotropic nature of TGF-β.

Future research will likely focus on developing more selective modulators of TGF-β signaling that can target specific aspects of its function while sparing others. This may involve targeting downstream components of the pathway or exploiting tissue-specific differences in TGF-β signaling. Additionally, biomarker development to identify patients most likely to benefit from TGF-β-targeted therapies will be crucial for the clinical translation of these approaches. As our understanding of this complex signaling network continues to evolve, so too will our ability to harness its therapeutic potential while minimizing unintended consequences.

Citations:

  1. https://pubmed.ncbi.nlm.nih.gov/10025398/
  2. https://pubmed.ncbi.nlm.nih.gov/9759503/
  3. https://www.semanticscholar.org/paper/a475218a371188864e85bbb643b526d07b7c1473
  4. https://pubmed.ncbi.nlm.nih.gov/10652271/
  5. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11048627/
  6. https://www.semanticscholar.org/paper/701f08ebe9cf795ec20a15aaed17856e3ce46537
  7. https://pubmed.ncbi.nlm.nih.gov/8047140/
  8. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8847772/
  9. https://www.semanticscholar.org/paper/6da21928a1ace8794c48e3ebfc0bf777b67261a0
  10. https://pubmed.ncbi.nlm.nih.gov/19237272/
  11. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8345981/
  12. https://www.semanticscholar.org/paper/ccaa8c5276204ce6b6b42a9df3408f189953f158
  13. https://pubmed.ncbi.nlm.nih.gov/30181548/
  14. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8944995/
  15. https://pubmed.ncbi.nlm.nih.gov/11929865/
  16. https://pubmed.ncbi.nlm.nih.gov/32228232/
  17. https://pubmed.ncbi.nlm.nih.gov/10652273/
  18. https://pubmed.ncbi.nlm.nih.gov/33742701/
  19. https://pubmed.ncbi.nlm.nih.gov/14707053/
  20. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7823508/
  21. https://pubmed.ncbi.nlm.nih.gov/18713838/
  22. https://pubmed.ncbi.nlm.nih.gov/11546783/
  23. https://www.semanticscholar.org/paper/25c59f039d600622c20765291dcb6a0827489d67
  24. https://pubmed.ncbi.nlm.nih.gov/28845509/
  25. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10051040/
  26. https://pubmed.ncbi.nlm.nih.gov/36727250/
  27. https://www.semanticscholar.org/paper/dd3b595f6feb19dd19e38e45855a3efbfe6da520
  28. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8502222/
  29. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5754510/
  30. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10799491/