9580 words (24 pg.)

Metastasis and the Endothelial Mesenchymal Transition

Generated by: T.O.M.

Definition and Mechanisms of Metastasis:

The Metastatic Cascade

The metastatic cascade is a series of key steps that cancer cells must undergo in order to spread from the primary tumor to distant sites in the body. These steps are not always sequential and can occur in different orders or simultaneously. The metastatic process is highly complex and involves interactions between cancer cells and their microenvironment.ref.86.10 ref.5.17 ref.86.10

1. Separation from the primary tumor: In order to metastasize, cancer cells must first detach from the primary tumor and invade surrounding tissues and basement membranes. This process, known as invasion, involves the acquisition of the ability to penetrate and degrade the extracellular matrix.ref.86.10 ref.56.6 ref.5.17

2. Entry and survival in the circulation, lymphatics, or peritoneal space: Once cancer cells have invaded the surrounding tissues, they must enter the bloodstream, lymphatic system, or peritoneal space to travel to distant sites in the body. This step, known as intravasation, requires cancer cells to adapt to the unique microenvironment of these systems and survive the shear forces and immune surveillance present.ref.136.22 ref.86.10 ref.79.10

3. Arrest in a distant target organ: After entering the circulation, lymphatics, or peritoneal space, cancer cells must come to a stop and adhere to the endothelium of a blood vessel in a distant organ. This step, known as arrest, is mediated by interactions between cancer cells and the endothelial cells lining the blood vessels.ref.86.10 ref.79.10 ref.79.10

4. Extravasation into the surrounding tissue: Once cancer cells have arrested in a distant organ, they must exit the blood vessel and invade the surrounding tissue. This process, known as extravasation, involves cancer cells penetrating the endothelium and interacting with the surrounding extracellular matrix.ref.86.11 ref.86.10 ref.79.10

5. Survival in the foreign microenvironment: After extravasation, cancer cells must adapt and survive in the new microenvironment of the distant organ. This step requires cancer cells to acquire the necessary molecular and cellular adaptations to evade cell death and proliferate in the foreign tissue.ref.56.6 ref.79.10 ref.86.10

6. Proliferation and induction of angiogenesis: Once cancer cells have survived and adapted to the foreign microenvironment, they must multiply and stimulate the growth of new blood vessels to support their growth in the distant organ. This step, known as colonization, involves the activation of angiogenic factors and the recruitment of endothelial cells to form new blood vessels.ref.79.10 ref.86.10 ref.86.10

7. Evading apoptotic death or immunological response: Throughout the entire metastatic process, cancer cells must avoid programmed cell death (apoptosis) and evade the immune system's response to eliminate them. This requires cancer cells to acquire mechanisms that allow them to escape detection and destruction by the immune system.ref.152.10 ref.122.2 ref.141.110

Molecular Mechanisms of Metastasis

The molecular mechanisms underlying metastasis involve a complex series of steps and processes. Metastasis is the primary cause of death in cancer patients, and it occurs when cancer cells spread from the primary tumor to distant sites in the body. The process of metastasis involves several key steps, including invasion, intravasation, circulation, extravasation, and colonization of distant organs.ref.86.10 ref.5.17 ref.86.10

During invasion, cancer cells acquire the ability to invade surrounding tissues and enter blood vessels or lymphatic vessels. This process is mediated by changes in the expression of genes involved in cell adhesion, motility, and proteolysis. For example, upregulation of matrix metalloproteinases (MMPs) allows cancer cells to degrade the extracellular matrix and facilitate invasion.ref.56.6 ref.56.70 ref.79.10

Once in the bloodstream or lymphatic system, cancer cells can circulate throughout the body and reach distant organs. The process of intravasation involves cancer cells adapting to survive in the unique microenvironment of the circulation or lymphatics. This may involve changes in cell surface markers, such as the upregulation of adhesion molecules, to allow cancer cells to interact with the endothelium and avoid immune surveillance.ref.79.10 ref.86.10 ref.56.75

The process of extravasation involves cancer cells exiting the blood vessels or lymphatic vessels and entering the surrounding tissue of a distant organ. This step requires cancer cells to interact with the endothelium and penetrate the extracellular matrix. Changes in cell adhesion molecules and proteolytic enzymes play a role in facilitating extravasation.ref.86.11 ref.86.10 ref.56.6

Finally, cancer cells can colonize the distant organ and form metastatic tumors. This process involves cancer cells adapting to the new microenvironment and stimulating the growth of new blood vessels to support their growth. Changes in gene expression, such as the upregulation of angiogenic factors, allow cancer cells to induce the formation of new blood vessels.ref.79.10 ref.86.10 ref.86.10

The molecular mechanisms underlying these steps are complex and involve various genetic, cellular, and microenvironmental factors. For example, genetic changes in cancer cells can lead to increased invasiveness and the ability to survive in the bloodstream. Changes in the tumor microenvironment, such as the presence of inflammatory cells or growth factors, can also play a role in promoting or inhibiting metastasis.ref.86.10 ref.5.17 ref.86.10

Additionally, the process of epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET) has been shown to be important in the metastatic process. EMT is a process by which cancer cells lose their epithelial characteristics and acquire mesenchymal traits, allowing them to invade and migrate. MET is the reverse process, where cancer cells regain their epithelial characteristics and establish colonies in distant organs.ref.152.5 ref.65.0 ref.154.11

It is important to note that the understanding of metastasis is still incomplete, and further research is needed to fully elucidate the molecular mechanisms involved. Mathematical models and experimental systems have been developed to study metastasis and provide insights into key processes. However, the complexity of metastasis and the interactions between tumor cells and the host organism present challenges in fully understanding and predicting metastatic behavior.ref.5.17 ref.5.2 ref.5.2

The Progression Model and Unanswered Questions

Metastasis is the process by which cancer cells spread from the primary tumor to other parts of the body. It is a complex and multifaceted process that involves several steps. The primary mechanism of metastatic spread involves five major steps: invasion and migration, angiogenesis and intravasation, survival in the bloodstream and adhesion to the endothelium, extravasation into the surrounding tissue, and colonization of a distant organ.ref.86.10 ref.5.17 ref.86.10

The progression model is the most commonly accepted model of metastasis. It suggests that a series of mutational events occur in subpopulations of the primary tumor or disseminated cells, resulting in a small fraction of cells that acquire full metastatic potential. This model explains the inefficiency of metastasis by the low probability that any given cell within the primary tumor will acquire all the multiple alterations required for successful metastatic progression.ref.5.2 ref.5.2 ref.5.2

However, there are still many unanswered questions and gaps in our understanding of metastasis. The different theories of metastasis are not mutually exclusive, and there may be multiple mechanisms by which tumor cells can successfully colonize distant tissues. For example, some studies suggest that cancer cells can undergo a collective migration, where groups of cells move together to invade and colonize distant organs.ref.5.17 ref.86.10 ref.5.2

Future research is needed to unravel the inner workings of tumor progression and metastasis and shed light on the mechanisms that drive this lethal aspect of neoplastic growth. Advances in genomic sequencing technologies and single-cell analysis techniques are providing new insights into the genetic and cellular heterogeneity within tumors and metastases. These tools, combined with experimental models and clinical studies, will help researchers uncover the molecular mechanisms underlying metastasis and identify potential targets for therapeutic intervention.ref.5.17 ref.79.3 ref.5.17

Endothelial Mesenchymal Transition (EMT):

What are the molecular markers and signaling pathways involved in EMT?

The Roles and Functions of Transcription Factors in Regulating E-cadherin Expression during Endothelial Mesenchymal Transition (EMT)

During Endothelial Mesenchymal Transition (EMT), several transcription factors play crucial roles in regulating the expression of E-cadherin, a key molecule involved in cell-cell adhesion. These transcription factors include Snail-1, Zeb, and members of the basic helix-loop-helix (bHLH) family.ref.159.3 ref.142.2 ref.124.5

Snail-1 is a transcription factor that is considered a major effector of EMT induction. It represses the expression of E-cadherin by binding to the promoter region of the E-cadherin gene and inhibiting its transcription. Snail-1 is activated by various signaling pathways, including TGF-b, Wnt, and Notch.ref.72.18 ref.33.33 ref.33.33 During TGF-b-induced EMT, Snail-1 is upregulated early, indicating its importance in initiating the EMT process.ref.72.18 ref.33.33 ref.33.33

In addition to its role in repressing E-cadherin expression, Snail-1 is also involved in the regulation of other EMT-related genes. It can interact with other transcription factors to control the outcome of EMT. Through these interactions, Snail-1 contributes to the disruption of intercellular junctions and the acquisition of mesenchymal traits during EMT.ref.142.2 ref.33.33 ref.33.33

Zeb is another transcription factor that regulates E-cadherin expression during EMT. Like Snail-1, Zeb binds to the promoter region of the E-cadherin gene and represses its transcription. Zeb is regulated by TGF-b1 in a Smad-dependent manner, meaning that the activation of TGF-b1 signaling leads to the upregulation of Zeb.ref.57.3 ref.97.4 ref.57.3 However, Zeb can be targeted by miR-200 family members, which can inhibit its expression.ref.57.3 ref.57.3 ref.97.4

Zeb is involved in a feedback mechanism that controls the outcome of EMT. It represses the expression of miR-200 family members, which in turn target Zeb1. This feedback loop helps regulate the EMT process and ensure the appropriate levels of E-cadherin expression.ref.57.3 ref.57.17 ref.124.10

Both Zeb1 and Zeb2 are implicated in the inhibition of E-cadherin expression during EMT. They contribute to the loss of epithelial characteristics and the acquisition of mesenchymal properties by the cells undergoing EMT.ref.57.3 ref.97.4 ref.57.0

Members of the bHLH family of transcription factors also play a role in regulating E-cadherin expression during EMT. These factors include Snail2, Slug, TWIST1, and E47. Similar to Snail-1 and Zeb, these bHLH factors can interact with other transcription factors and co-SMADs to activate or repress the expression of E-cadherin and other genes involved in EMT.ref.142.2 ref.142.2 ref.30.6

The bHLH factors bind to E-box sequences in the promoter region of the E-cadherin gene, allowing them to regulate its transcription. Through their interactions with other transcription factors and co-SMADs, the bHLH factors contribute to the overall control of the EMT process.ref.142.2 ref.171.21 ref.124.5

Signaling Pathways Involved in Endothelial Mesenchymal Transition (EMT)

Endothelial Mesenchymal Transition (EMT) is regulated by a complex network of signaling pathways involving various molecules. These pathways contribute to the loss of epithelial characteristics and the acquisition of mesenchymal properties by the cells undergoing EMT.ref.121.8 ref.154.11 ref.171.1

TGF-b-induced signaling pathways play a central role in EMT. These pathways include both Smad-dependent and Smad-independent pathways. TGF-b receptors activate Smad2 and Smad3, which form complexes with Smad4 and translocate to the nucleus to regulate the transcription of target genes.ref.107.25 ref.107.25 ref.107.25

In addition to the Smad signaling pathway, TGF-b can also activate non-Smad signaling pathways, such as mitogen-activated protein kinases (MAPKs), Rho GTPases, and phosphatidylinositol 3-kinase (PI3K). These pathways contribute to the regulation of gene expression and cellular processes involved in EMT.ref.64.4 ref.107.4 ref.64.4

In addition to TGF-b-induced signaling pathways, other signaling pathways are also involved in EMT. For example, nuclear factor jB (NF-jB) is a signaling pathway that can regulate the expression of EMT-inducing transcription factors, including Snail and ZEB.ref.117.7 ref.117.7 ref.117.7

Integrin-linked kinase (ILK) is another signaling molecule involved in EMT. It interacts with various signaling pathways, such as TGF-b, and contributes to the regulation of gene expression and cellular processes during EMT.ref.72.28 ref.117.6 ref.72.28

Phosphatidylinositol 3-kinase (PI3K) is a kinase that is involved in multiple cellular processes, including cell survival, proliferation, and migration. It is also implicated in EMT and contributes to the regulation of gene expression and cellular processes during this process.ref.36.7 ref.99.4 ref.162.29

Glycogen synthase kinase (GSK) 3b is another signaling molecule that plays a role in EMT. It regulates the stability of various proteins involved in EMT, including Snail and ZEB, by phosphorylating them and targeting them for degradation.ref.70.15 ref.70.15 ref.30.5

These signaling pathways collectively contribute to the molecular mechanisms controlling the initiation and progression of EMT. They regulate gene expression and cellular processes that lead to the loss of epithelial characteristics and the acquisition of mesenchymal properties. These pathways work in a cell type-specific manner, highlighting the complexity of EMT regulation.ref.154.11 ref.124.5 ref.171.1

How does EMT affect the behavior and characteristics of endothelial cells?

Introduction to Epithelial Mesenchymal Transition (EMT)

Epithelial Mesenchymal Transition (EMT) is a cellular process in which epithelial cells undergo a phenotypic change and acquire mesenchymal characteristics. This transition is triggered by various growth factors, cytokines, and hypoxia, which activate a transcription program that inhibits the expression of epithelial-related genes and induces the expression of mesenchymal-related genes. As a result, epithelial cells lose their cell-cell adhesion, polarity, and specific markers associated with the epithelial phenotype, and gain increased motility and invasiveness.ref.121.8 ref.154.11 ref.149.17 The mesenchymal cells that arise from EMT exhibit a spindle-like morphology and express mesenchymal markers such as N-cadherin, vimentin, ECM molecules, fibronectin, and MMPs. EMT is a reversible process, and mesenchymal cells can revert back to the epithelial phenotype through a process called Mesenchymal Epithelial Transition (MET). EMT and MET are involved in various physiological and pathological processes, including embryonic development, tissue regeneration and repair, wound healing, and cancer progression.ref.149.17 ref.159.3 ref.121.8

EMT in Different Contexts

EMT has been observed in different contexts, such as embryonic development, wound healing, and cancer metastasis. In embryonic development, EMT is involved in gastrulation and the formation of various organs and tissues. During gastrulation, EMT allows the migration and invasiveness of cells from the epiblast to the primitive streak, leading to the formation of three germ layers: ectoderm, mesoderm, and endoderm.ref.119.2 ref.70.5 ref.117.6 EMT also plays a crucial role in the formation of specific structures and organs, such as the heart, lungs, and kidneys. In wound healing, EMT is mediated by inflammatory cells and fibroblasts, allowing epithelial cells to acquire migratory properties and repopulate the wound. EMT in wound healing is essential for tissue regeneration and repair.ref.70.5 ref.119.2 ref.117.6

In cancer, EMT is associated with increased aggressiveness, metastasis, and resistance to chemotherapy. It has been shown that EMT enhances stemness and tumor-initiating potential in cancer cells, and there is a link between EMT and cancer stem cells (CSCs). CSCs are predicted to be critical drivers of tumor progression and are characterized by self-renewal capacity, limitless proliferative potential, and metastatic potential.ref.149.17 ref.152.5 ref.154.11 EMT can endow cells with CSC characteristics and a more motile invasive phenotype, contributing to tumor progression and metastasis. EMT in cancer cells is regulated by various signaling pathways and transcription factors, including TGF-β, Wnt, Notch, Snail, Slug, ZEB1/2, and Twist.ref.149.17 ref.152.5 ref.149.18

EMT and Endothelial Cells

The relationship between EMT and endothelial cells is not explicitly mentioned in the provided document excerpts. However, EMT primarily refers to the transition of epithelial cells to a mesenchymal phenotype. The effects of EMT on endothelial cells may vary depending on the specific context and signaling pathways involved.ref.121.8 ref.149.17 ref.65.0 Further research and specific references are needed to provide a more detailed answer regarding the effects of EMT on endothelial cells.ref.119.23 ref.119.19 ref.149.17

Endothelial Mesenchymal Transition (EndMT)

Endothelial Mesenchymal Transition (EndMT) refers to the transition of endothelial cells to a mesenchymal phenotype. EndMT in endothelial cells can impact their function and role in physiological processes such as angiogenesis or vascular remodeling. During EndMT, endothelial cells lose their endothelial markers, such as endothelial cadherin, endothelial nitric oxide synthase, and platelet-endothelial cell adhesion molecule-1, and upregulate mesenchymal markers, such as alpha smooth muscle actin, fibronectin, and platelet-derived growth factor receptor.ref.121.8 ref.26.0 ref.26.0 This leads to morphological and functional changes consistent with mesenchymal transformation.ref.121.8 ref.26.0 ref.26.0

EndMT in endothelial cells is often activated during pathological conditions, including fibrosis, cardiovascular diseases, and cancer. EndMT can be induced by various stimuli, including growth factors, cytokines, and hypoxia, and is regulated by transcription factors such as Snail, Slug, ZEB1/2, and Twist. EndMT has been implicated in fibrosis, where endothelial cells undergo phenotypic changes and contribute to the excessive deposition of extracellular matrix components, leading to tissue scarring and dysfunction.ref.26.6 ref.26.6 ref.141.151 In cardiovascular diseases, EndMT has been associated with vascular remodeling, atherosclerosis, and the development of neointimal lesions.ref.26.6 ref.26.6 ref.26.6

EMT in Endothelial Cells and Cancer Progression

While the provided document excerpts do not explicitly mention the relationship between EMT and endothelial cells in the context of cancer, it is important to note that EMT in endothelial cells can have profound effects on their function and behavior in physiological processes such as angiogenesis and vascular remodeling. EndMT can promote the formation of cancer stem cells (CSCs), which are associated with tumor progression, metastasis, and recurrence. CSCs have been shown to possess self-renewal capacity, limitless proliferative potential, and metastatic potential.ref.149.17 ref.152.5 ref.117.8 The acquisition of a mesenchymal phenotype through EMT in endothelial cells can contribute to the formation of CSCs and enhance their invasive properties.ref.131.6 ref.149.17 ref.149.17

Furthermore, EMT in tumor endothelial cells can impact tumor angiogenesis, which is crucial for tumor growth and metastasis. Endothelial cells undergoing EMT may acquire a more migratory and invasive phenotype, allowing them to participate in the formation of new blood vessels in the tumor microenvironment. This process, known as endothelial cell plasticity, can contribute to tumor angiogenesis and ultimately promote tumor progression.ref.149.17 ref.65.0 ref.117.8

Conclusion

In conclusion, EMT is a cellular process in which epithelial cells undergo a phenotypic change and acquire mesenchymal characteristics. EMT is involved in various physiological and pathological processes, including embryonic development, tissue regeneration and repair, wound healing, and cancer progression. EMT in endothelial cells, known as EndMT, can impact their function and role in physiological processes such as angiogenesis or vascular remodeling.ref.121.8 ref.117.6 ref.149.17 EndMT is often activated during pathological conditions, including fibrosis, cardiovascular diseases, and cancer. In cancer, EMT in endothelial cells can contribute to tumor progression, metastasis, and the formation of CSCs. Further research is needed to fully understand the role of EMT in endothelial cells and its implications in cancer progression.ref.149.17 ref.121.8 ref.154.11

What are the regulatory factors that control EMT?

Introduction

Endothelial Mesenchymal Transition (EMT) is a cellular process that plays a critical role in embryonic development, tissue repair, and disease progression, including cancer metastasis. EMT involves the transformation of epithelial cells into mesenchymal cells, which exhibit enhanced migratory and invasive properties. The regulation of EMT is a complex process that involves a variety of signaling molecules and transcription factors.ref.121.8 ref.65.0 ref.154.11 In this essay, we will explore the regulatory factors that control EMT, including transcription factors such as Snail-1, Zeb, and members of the basic helix-loop-helix (bHLH) family, as well as the signaling molecules Smads, integrin-linked kinase (ILK), phosphatidylinositol 3-kinase (PI3K), mitogen-activated protein kinases (MAPKs), glycogen synthase kinase (GSK) 3b, and nuclear factor jB (NF-jB).ref.159.3 ref.117.6 ref.124.5

Transcription Factors in EMT Regulation

EMT is regulated by a variety of transcription factors, including Snail-1, Zeb, and members of the bHLH family. These transcription factors play a crucial role in the repression of E-cadherin expression, a key event in EMT. Snail-1, in particular, is a major promoter of EMT and is upregulated early during TGF-β induced EMT.ref.97.4 ref.97.4 ref.171.0 Snail levels are inversely correlated with E-cadherin in numerous tumors and contribute to the acquisition of an invasive phenotype. Snail directly binds to the promoter of the E-cadherin gene and represses its expression. Similarly, Zeb can also bind to the promoter of the E-cadherin gene and repress its expression.ref.97.4 ref.97.4 ref.171.0 Additionally, Snail and Zeb are able to repress miR-200 family members, which in turn target Zeb1 and control the EMT outcome by a feedback mechanism. The Snail gene family has a key role in triggering EMT and is regulated by many signaling pathways, including TGF-b. The Snail repressor recruits EZH2 to specific genomic sites through the enrollment of the lncRNA HOTAIR, which mediates a physical interaction between Snail and EZH2.ref.171.0 ref.171.0 ref.171.2 The Snail-repressive activity depends on the formation of a tripartite Snail/HOTAIR/EZH2 complex.ref.171.0 ref.171.0 ref.171.2

Signaling Molecules in EMT Regulation

The regulation of EMT involves a complex network of signaling molecules, including Smads, ILK, PI3K, MAPKs, GSK 3b, and NF-jB. These signaling molecules play critical roles in the TGF-b-induced signaling pathway, which is central to many experimental models of EMT.ref.72.28 ref.72.28 ref.72.28

1. Smads: Smads are signaling molecules that play a crucial role in the TGF-b-induced signaling pathway. TGF-b signals through a complex network of receptors and kinases, ultimately leading to the activation of Smad2 and Smad3.ref.64.4 ref.64.3 ref.64.3 Phosphorylated Smad2 and Smad3 form trimers with Smad4 and translocate to the nucleus, where they regulate target gene transcription. Smads cooperate with DNA-binding transcription factors to repress E-cadherin expression, a key event in EMT.ref.64.3 ref.64.4 ref.64.4

2. ILK (Integrin-Linked Kinase): ILK is a signaling molecule that is involved in the regulation of EMT. It is part of the integrin signaling pathway, which plays a role in cell adhesion and migration.ref.72.28 ref.72.28 ref.110.13 ILK has been implicated in the disruption of intercellular junctions between epithelial cells during EMT.ref.72.28 ref.110.13 ref.110.13

3. PI3K (Phosphatidylinositol 3-Kinase): PI3K is another signaling molecule that is involved in the regulation of EMT. It is part of the TGF-b signaling pathway and can be activated by TGF-b receptors.ref.107.4 ref.107.4 ref.107.4 PI3K is known to play a role in cell migration and invasion, which are characteristic features of EMT.ref.107.4 ref.107.4 ref.107.4

4. MAPKs (Mitogen-Activated Protein Kinases): MAPKs are a family of signaling molecules that are involved in various cellular processes. They have been implicated in the TGF-b-induced EMT pathway and can be activated by TGF-b receptors.ref.107.4 ref.107.4 ref.107.4 MAPKs play a role in the regulation of EMT-related gene expression.ref.107.4 ref.107.4 ref.107.4

5. GSK 3b (Glycogen Synthase Kinase 3b): GSK 3b is a signaling molecule that is involved in the regulation of EMT. It is part of the Wnt signaling pathway, which plays a role in cell fate determination and tissue development.ref.70.15 ref.72.28 ref.72.28 GSK 3b has been implicated in the regulation of EMT-related gene expression and the maintenance of epithelial cell characteristics.ref.70.35 ref.70.35 ref.72.28

6. NF-jB (Nuclear Factor jB): NF-jB is a transcription factor that is involved in the regulation of EMT. It can be activated by various signaling pathways, including the TGF-b signaling pathway.ref.117.7 ref.117.7 ref.117.7 NF-jB has been found to induce the expression of EMT-inducing transcription factors, such as Snail and Zeb, which repress E-cadherin expression and promote the mesenchymal phenotype.ref.117.7 ref.117.7 ref.117.7

miRNAs in EMT Regulation

In recent years, miRNAs (microRNAs) have emerged as key regulators of EMT. miRNAs are small non-coding RNA molecules that can bind to the mRNA of target genes and regulate their expression. Several miRNAs have been identified as regulators of EMT, including the miR-200 and miR-34 families.ref.124.1 ref.128.15 ref.124.1 These miRNAs interact with EMT transcription factors, such as ZEB and SNAIL, to form a core EMT regulatory network. miR-200 family members, in particular, have been found to repress ZEB1 expression and inhibit EMT. Other miRNAs, such as miR-192, miR-138, and miR-130, have also been found to regulate EMT.ref.124.1 ref.124.10 ref.97.4 The regulation of EMT by miRNAs provides potential therapeutic targets for impacting metastasis dissemination and increasing patient survival.ref.124.1 ref.124.15 ref.124.1

Conclusion

In conclusion, the regulation of EMT is a complex process that involves a variety of signaling molecules and transcription factors. The transcription factors Snail-1, Zeb, and members of the bHLH family repress E-cadherin expression, a key event in EMT. The signaling molecules Smads, ILK, PI3K, MAPKs, GSK 3b, and NF-jB play critical roles in the TGF-b-induced signaling pathway, which is central to many experimental models of EMT.ref.124.5 ref.124.1 ref.119.15 Additionally, miRNAs have been identified as regulators of EMT, with miR-200 and miR-34 families interacting with EMT transcription factors to form a core EMT regulatory network. Understanding the regulatory factors that control EMT is crucial for developing targeted therapies to inhibit metastasis and improve patient outcomes. Further research is needed to unravel the intricate mechanisms underlying the regulation of EMT and identify additional therapeutic targets.ref.124.1 ref.124.1 ref.124.1

How is EMT induced in endothelial cells?

Introduction

Endothelial mesenchymal transition (EMT) is a process in which endothelial cells undergo a transformation into mesenchymal cells. This process is induced by various factors and signaling pathways. One of the key inducers of EMT in endothelial cells is the activation of the Jagged 1/Notch pathway.ref.121.8 ref.70.5 ref.64.5 This activation leads to morphological and functional changes consistent with mesenchymal transformation. EMT in endothelial cells is characterized by the attenuation of endothelial markers such as endothelial cadherin (E-cadherin), endothelial nitric oxide synthase (eNOS), and platelet-endothelial cell adhesion molecule-1 (PECAM-1), as well as the upregulation of mesenchymal markers such as alpha smooth muscle actin (α-SMA), fibronectin, and platelet-derived growth factor (PDGF)-receptor. Other inducers of EMT in endothelial cells include transforming growth factor β (TGF-β) and other polypeptides such as endothelin-1 (ET-1) or insulin growth factor.ref.121.8 ref.64.5 ref.70.5 These inducers lead to the loss of epithelial characteristics and the acquisition of a motile behavior and the phenotype of myofibroblasts.ref.64.5 ref.121.8 ref.70.5

Molecular Mechanisms of EMT and EndoMT

The molecular mechanisms controlling the initiation and progression of EMT and EndoMT involve a complex network of signaling molecules. These molecules include TGF-β, Smads, integrin-linked kinase (ILK), phosphatidylinositol 3-kinase (PI3K), mitogen-activated protein kinases (MAPKs), glycogen synthase kinase (GSK) 3β, and nuclear factor κB (NF-κB). The signaling pathways regulated by these molecules play a crucial role in the expression of transcription factors such as Snail-1, Zeb, and members of the basic helix-loop-helix (bHLH) family.ref.117.7 ref.124.5 ref.117.7 These transcription factors repress E-cadherin expression and promote the mesenchymal phenotype.ref.142.2 ref.142.2 ref.124.5

TGF-β, in particular, is a potent inducer of EMT and EndoMT. It activates Smad proteins, which translocate to the nucleus and regulate the expression of target genes involved in EMT. Additionally, TGF-β signaling can activate other signaling pathways, such as PI3K, MAPKs, GSK-3β, and NF-κB, which further contribute to the induction of EMT and EndoMT.ref.117.7 ref.107.25 ref.107.4 The activation of these signaling pathways leads to the downregulation of endothelial markers and the upregulation of mesenchymal markers.ref.107.25 ref.107.4 ref.107.25

Functional Consequences of EMT Induction in Endothelial Cells

The induction of EMT in endothelial cells has several functional consequences. One of the major effects is the loss of endothelial markers such as E-cadherin, eNOS, and PECAM-1. These markers are crucial for maintaining the integrity and function of endothelial cells.ref.121.8 ref.128.5 ref.64.5 The downregulation of these markers contributes to the morphological and functional changes associated with mesenchymal transformation.ref.110.13 ref.121.8 ref.64.5

In addition to the loss of endothelial markers, EMT induction in endothelial cells leads to the upregulation of mesenchymal markers such as α-SMA, fibronectin, and PDGF-receptor. These markers are characteristic of mesenchymal cells and are involved in processes such as tissue remodeling and wound healing. The upregulation of these markers further contributes to the acquisition of a mesenchymal phenotype by endothelial cells.ref.149.17 ref.121.8 ref.32.17

EMT induction also enhances the migratory capacity of endothelial cells. Mesenchymal cells are highly motile, and the acquisition of a mesenchymal phenotype by endothelial cells increases their ability to migrate. This increased migratory capacity is crucial for processes such as tissue regeneration and wound healing.ref.149.17 ref.121.8 ref.119.2

Furthermore, EMT induction in endothelial cells confers resistance to apoptosis. Apoptosis, or programmed cell death, is a normal physiological process that eliminates unwanted or damaged cells. The resistance to apoptosis in mesenchymal cells allows them to survive under adverse conditions and contributes to their persistence in pathological conditions such as fibrosis and cancer progression.ref.149.17 ref.121.8 ref.70.5

Another consequence of EMT induction in endothelial cells is the acquisition of a stem-like phenotype. Stem cells are characterized by their ability to self-renew and differentiate into multiple cell types. The acquisition of a stem-like phenotype by endothelial cells promotes their plasticity and enhances their role in tissue regeneration and wound healing.ref.149.17 ref.117.6 ref.121.8

Role of EMT in Cancer Progression

EMT induction in endothelial cells plays a crucial role in cancer progression. The formation of new blood vessels, known as angiogenesis, is essential for tumor growth and metastasis. Endothelial cells undergoing EMT can contribute to angiogenesis by acquiring a mesenchymal phenotype and migrating towards the tumor site.ref.149.17 ref.117.8 ref.27.16 These mesenchymal endothelial cells can then differentiate into cancer-associated fibroblasts, which promote tumor growth and invasion.ref.149.17 ref.132.2 ref.117.8

EMT induction in endothelial cells can also promote the formation of cancer stem cells. Cancer stem cells are a subpopulation of cancer cells that possess stem cell-like properties. They are associated with tumor progression, metastasis, and recurrence.ref.149.17 ref.152.5 ref.117.8 The acquisition of a stem-like phenotype by endothelial cells undergoing EMT contributes to the formation of cancer stem cells and their impact on tumor development.ref.149.17 ref.152.5 ref.149.17

Inhibition and Regulation of the Jagged 1/Notch Pathway

The Jagged 1/Notch pathway is a key regulator of EMT induction in endothelial cells. Activation of this pathway leads to the attenuation of endothelial markers and the upregulation of mesenchymal markers, as well as the acquisition of a stem-like phenotype and drug resistance. Therefore, inhibiting or regulating the Jagged 1/Notch pathway can prevent or reverse EMT in endothelial cells.ref.119.18 ref.121.7 ref.119.18

There are known inhibitors and regulators of the Jagged 1/Notch pathway that can be targeted to prevent or reverse EMT in endothelial cells. These inhibitors include small molecules and antibodies that specifically target the Notch receptors or ligands. By blocking the activation of the Jagged 1/Notch pathway, these inhibitors can prevent the induction of EMT in endothelial cells and maintain their endothelial phenotype.ref.121.7 ref.119.18 ref.119.30

Furthermore, modulating the expression or activity of key molecules in the Jagged 1/Notch pathway can also regulate EMT induction in endothelial cells. For example, targeting the expression or activity of specific transcription factors involved in EMT regulation, such as Snail-1 or Zeb, can prevent the downregulation of endothelial markers and the upregulation of mesenchymal markers.ref.121.7 ref.119.18 ref.119.30

Conclusion

EMT and EndoMT are important processes in embryonic development, tissue regeneration, wound healing, and pathological conditions such as fibrosis and cancer progression. The induction of EMT in endothelial cells is regulated by various factors and signaling pathways, including the Jagged 1/Notch pathway and TGF-β signaling. The molecular mechanisms controlling EMT involve a complex network of signaling molecules, transcription factors, and target genes.ref.70.5 ref.121.8 ref.117.6

The functional consequences of EMT induction in endothelial cells include the loss of endothelial markers, the upregulation of mesenchymal markers, increased migratory capacity, resistance to apoptosis, and the acquisition of a stem-like phenotype. These effects contribute to the morphological and functional changes associated with mesenchymal transformation and play a crucial role in processes such as tissue regeneration and wound healing.ref.121.8 ref.149.17 ref.126.23

EMT induction in endothelial cells also plays a significant role in cancer progression. The acquisition of a mesenchymal phenotype by endothelial cells promotes angiogenesis and the formation of cancer-associated fibroblasts. Furthermore, EMT induction in endothelial cells contributes to the formation of cancer stem cells, which are associated with tumor progression, metastasis, and recurrence.ref.149.17 ref.121.8 ref.117.8

The Jagged 1/Notch pathway is a key regulator of EMT induction in endothelial cells. Inhibiting or regulating this pathway can prevent or reverse EMT and maintain the endothelial phenotype. Targeting the expression or activity of specific molecules in the Jagged 1/Notch pathway, as well as using inhibitors and regulators, offers potential therapeutic strategies for preventing or reversing EMT in endothelial cells and may have implications for the treatment of fibrosis and cancer.ref.121.7 ref.119.18 ref.119.30

What role does EMT play in tumor metastasis?

The Role of the Tumor Microenvironment in Inducing EMT

The tumor microenvironment is a complex and dynamic environment that consists of various cell types, extracellular matrix components, and signaling molecules. It plays a significant role in the induction of epithelial-mesenchymal transition (EMT) in tumor cells. EMT is a process by which epithelial cells acquire mesenchymal characteristics, leading to increased cell motility, invasiveness, and metastasis.ref.65.0 ref.65.0 ref.65.0

One of the factors within the tumor microenvironment that can induce EMT is the presence of inflammatory cells. Inflammatory cells infiltrating the tumor site release cytokines and growth factors that can stimulate EMT in tumor cells. These factors include tumor necrosis factor-alpha (TNF-α), transforming growth factor-beta (TGF-β), fibroblast growth factor (FGF), epidermal growth factor (EGF), platelet-derived growth factor (PDGF), and insulin-like growth factor (IGF).ref.131.3 ref.117.6 ref.131.3 These growth factors can activate signaling pathways within tumor cells that promote EMT.ref.131.3 ref.131.3 ref.117.6

Hypoxia, or low oxygen levels, is another characteristic of the tumor microenvironment that can induce EMT. Hypoxia-inducible factor 1-alpha (HIF-1α) is a transcription factor that is stabilized under hypoxic conditions and can activate EMT-related genes. HIF-1α can induce the expression of factors such as TGF-β and vascular endothelial growth factor (VEGF), which further contribute to the induction of EMT in tumor cells.ref.131.3 ref.117.7 ref.162.9

The presence of stem cells within the tumor microenvironment also plays a role in inducing EMT. Cancer stem cells (CSCs) and mesenchymal stem cells (MSCs) have been shown to promote EMT in tumor cells. CSCs are a population of cells within a tumor that have self-renewal and differentiation capabilities, and they can give rise to different cell types within the tumor.ref.152.5 ref.149.17 ref.165.1 MSCs, on the other hand, have the ability to differentiate into mesenchymal cells, such as fibroblasts and adipocytes. Both CSCs and MSCs can secrete factors that promote EMT, such as TGF-β and cytokines.ref.119.31 ref.119.31 ref.131.6

Overall, the tumor microenvironment provides a conducive environment for the induction of EMT in tumor cells. Inflammatory cells, hypoxia, and stem cells present within the tumor microenvironment can act as inducers of EMT. These factors release signaling molecules that activate pathways within tumor cells, leading to the expression of specific markers associated with the mesenchymal phenotype and increased invasiveness and metastasis.ref.65.0 ref.131.3 ref.65.0

Signaling Pathways Involved in EMT and Tumor Metastasis

Several signaling pathways are involved in EMT and tumor metastasis, including TGF-β, NF-κB, Wnt, and Notch. These pathways play a role in promoting tumor metastasis by inducing the expression of transcription factors that repress E-cadherin expression, a hallmark of EMT.ref.117.7 ref.124.5 ref.117.7

The TGF-β signaling pathway is one of the key players in promoting tumor progression and metastasis. It induces EMT through both Smad-dependent and Smad-independent transcriptional pathways. When TGF-β binds to its receptors, it leads to the phosphorylation of Smad2 and Smad3.ref.52.45 ref.52.45 ref.52.45 These phosphorylated Smads then form complexes with Smad4 and translocate into the nucleus, where they control the transcription of target genes associated with EMT. These target genes include transcription factors such as Snail-1, Zeb, and members of the basic helix-loop-helix (bHLH) family, which repress E-cadherin expression.ref.52.45 ref.52.45 ref.52.45

In addition to the Smad signaling pathways, TGF-β can also activate non-Smad signaling pathways that contribute to the induction of EMT and promote tumor metastasis. These non-Smad signaling pathways include the Ras/Erk, JNK, PI3 kinase, Par6, and Cdc42 GTPases pathways. Activation of these pathways leads to the activation of downstream effectors that regulate cell motility, invasion, and metastasis.ref.107.4 ref.107.4 ref.107.4

The NF-κB signaling pathway is another pathway involved in EMT and tumor metastasis. NF-κB is a transcription factor that plays a crucial role in inflammation and immune responses. Activation of the NF-κB pathway can induce EMT by regulating the expression of EMT-related genes.ref.117.7 ref.117.7 ref.117.7 NF-κB can be activated by various stimuli, including cytokines, growth factors, and oxidative stress. Once activated, NF-κB translocates into the nucleus and binds to specific DNA sequences, leading to the transcription of genes associated with EMT.ref.117.7 ref.117.7 ref.117.7

The Wnt signaling pathway is also implicated in EMT and tumor metastasis. Wnt proteins are secreted signaling molecules that can activate a cascade of intracellular events. Activation of the Wnt pathway can lead to the stabilization and nuclear translocation of β-catenin, which then interacts with transcription factors to regulate gene expression.ref.141.162 ref.117.7 ref.141.162 Wnt signaling can induce EMT by repressing E-cadherin expression and promoting the expression of mesenchymal markers.ref.141.162 ref.117.7 ref.117.7

The Notch signaling pathway is involved in various cellular processes, including cell fate determination and differentiation. Activation of the Notch pathway can induce EMT by regulating the expression of EMT-related genes. Notch signaling can promote EMT by inhibiting the expression of E-cadherin and activating the expression of mesenchymal markers.ref.119.18 ref.119.16 ref.119.16

In summary, the TGF-β, NF-κB, Wnt, and Notch signaling pathways play a role in EMT and tumor metastasis. These pathways regulate the expression of transcription factors that repress E-cadherin expression and induce the expression of mesenchymal markers. The activation of these pathways within tumor cells leads to increased cell motility, invasiveness, and metastasis.ref.142.2 ref.117.7 ref.142.2

How is EMT involved in tissue fibrosis?

Epithelial Mesenchymal Transition (EMT) in Tissue Fibrosis

Epithelial Mesenchymal Transition (EMT) is a process involved in tissue fibrosis, which occurs when epithelial cells undergo transdifferentiation into myofibroblasts. This process is triggered by transforming growth factor β (TGF-β) and possibly other polypeptides such as endothelin-1 (ET-1) or insulin growth factor. EMT is characterized by the loss of epithelial characteristics, such as E-cadherin expression and apical-basal polarity, and the acquisition of a motile behavior and the phenotype of myofibroblasts, including the expression of α-SMA and fibroblast-specific proteins like type I collagen.ref.121.8 ref.70.5 ref.126.23

TGF-β is a key inducer of EMT and is involved in the generation of myofibroblasts through EMT. It plays a crucial role in tissue fibrosis and is implicated in the pathogenesis of fibroproliferative vasculopathies. TGF-β stimulates the production of various collagens and other extracellular matrix components by activated mesenchymal cells.ref.39.1 ref.64.7 ref.39.1 The inhibition of TGF-β and its signaling pathways is a potential therapeutic approach to prevent EMT and fibrosis.ref.39.1 ref.64.7 ref.36.11

Besides TGF-β, other potential polypeptides that can trigger EMT include endothelin-1 (ET-1) and insulin growth factor (IGF). ET-1 has been found to be involved in the induction of EMT and can stimulate endogenous TGF-β1 production. Additionally, Vascular Endothelial Growth Factor (VEGF) and hypoxia can also induce EMT in mesothelial cells.ref.117.7 ref.117.6 ref.117.6 These polypeptides and factors play a role in the regulation of EMT and contribute to tissue fibrosis in various organs.ref.117.6 ref.117.6 ref.117.7

Endothelial Mesenchymal Transition (EndoMT) in Fibrosis

Endothelial Mesenchymal Transition (EndoMT) is a similar process to EMT, in which endothelial cells undergo a mesenchymal transition and acquire a myofibroblastic phenotype. EndoMT can also be induced by TGF-β, similarly to EMT. TGF-β is a key player in the induction of EndoMT and is involved in the pathogenesis of fibroproliferative vasculopathies.ref.32.17 ref.32.17 ref.37.16

EndoMT has been observed in various organs and tissues in the context of fibrosis. It has been observed in renal, pulmonary, and liver fibrosis, as well as in cardiac, renal, and pulmonary fibrosis. Additionally, EndoMT has been observed in chronic pulmonary hypertension, chronic kidney disease, idiopathic portal hypertension, and other fibrotic diseases such as intestinal fibrosis and radiation-induced rectal fibrosis.ref.26.6 ref.26.6 ref.26.6

The role of EndoMT in fibrosis is complex and may vary depending on the specific organ or tissue. Further studies are required to fully understand the contribution of EndoMT to fibrosis and its potential as a therapeutic target.ref.26.6 ref.26.6 ref.26.6

EMT and EndoMT in Hepatocellular Carcinoma (HCC)

In the context of hepatocellular carcinoma (HCC), EMT plays a role in the early steps of metastasis. During tumor progression, cancer cells can undergo EMT, losing their epithelial characteristics and acquiring a more mesenchymal phenotype. This phenotypic switch allows cancer cells to become more motile and invasive, facilitating their spread to distant organs.ref.110.8 ref.110.15 ref.110.3

EMT in HCC is characterized by the loss of epithelial markers, such as E-cadherin, and the acquisition of mesenchymal markers, such as vimentin and N-cadherin. This transition is facilitated by the activation of various signaling pathways, including TGF-β signaling.ref.110.8 ref.110.15 ref.110.4

The involvement of EMT in HCC metastasis highlights its potential as a therapeutic target. Inhibiting EMT could potentially prevent or limit the spread of HCC to other organs. However, further research is needed to fully understand the molecular mechanisms underlying EMT in HCC and to develop effective therapeutic strategies targeting EMT.ref.110.19 ref.110.3 ref.110.19

Conclusion

EMT and EndoMT are processes involved in tissue fibrosis, characterized by the transdifferentiation of epithelial or endothelial cells into myofibroblasts. These processes are triggered by various polypeptides, such as TGF-β, ET-1, and IGF, and play a crucial role in the pathogenesis of fibrosis in organs such as the kidney, liver, lung, and heart.ref.26.12 ref.26.12 ref.26.12

Understanding the molecular mechanisms underlying EMT and EndoMT in fibrosis is essential for the development of effective therapeutic strategies. Targeting the polypeptides and signaling pathways involved in EMT and EndoMT, such as TGF-β, may provide new opportunities for preventing or treating fibrosis in various organs and diseases.ref.110.20 ref.39.27 ref.110.20

Further research is needed to fully elucidate the contribution of EMT and EndoMT to fibrosis in specific organs and to identify new therapeutic targets. By unraveling the complexities of these processes, we can pave the way for the development of novel interventions to combat fibrosis and improve patient outcomes.ref.110.20 ref.39.27 ref.132.2

Role of EMT in Cancer Metastasis:

The Role of EMT and MET in Cancer Metastasis

EMT (epithelial-to-mesenchymal transition) and MET (mesenchymal-to-epithelial transition) are crucial processes involved in cancer metastasis. EMT allows cancer cells to acquire a more invasive and migratory phenotype, facilitating their spread to distant sites in the body. On the other hand, MET is necessary for the colonization of distant organs by reverting the cells back to an epithelial phenotype.ref.154.11 ref.65.0 ref.152.5 These processes are regulated by specific factors and signaling pathways.ref.61.2 ref.61.2 ref.65.0

EMT is involved in all steps of the metastatic cascade, including the escape from the primary site, intravasation, transportation in the circulation, and the formation of micrometastases. During EMT, cancer cells undergo phenotypic changes such as the loss of cellular adhesion, cytoskeletal reorganization, secretion of matrix metalloproteinases (MMPs), and the development of cellular protrusions. These changes enable cancer cells to detach from the primary tumor, invade the surrounding tissues, and enter the bloodstream.ref.117.8 ref.56.6 ref.56.48 EMT is associated with various types of carcinoma, including oral squamous cell carcinoma (SCC) and breast cancer. It is also linked to metastasis, poor outcomes, and therapeutic resistance in cancer patients.ref.117.8 ref.117.8 ref.117.8

Specific factors and signaling pathways play crucial roles in regulating EMT and MET. Some of these include TWIST1, FOXC2, FOXQ1, SIX1, CDH1 (E-cadherin), TGFB1, TNF, HIF-1, and Twist. These factors and pathways orchestrate the molecular and cellular changes that occur during EMT and MET.ref.124.5 ref.142.2 ref.117.7 For example, TWIST1, FOXC2, FOXQ1, and SIX1 are EMT regulators that contribute to the metastatic potential of breast cancer cells and are associated with poor outcomes. CDH1 is a key molecule involved in the loss of cellular adhesion during EMT. TGFB1 and TNF are factors that can initiate EMT, while HIF-1 is involved in hypoxia-induced EMT.ref.124.5 ref.117.7 ref.142.2 Understanding the role of these factors and pathways is essential for developing therapeutic strategies to target EMT and MET in cancer metastasis.ref.131.3 ref.131.3 ref.117.7

Computational simulations have been utilized to model the role of EMT and MET in cancer metastasis. These simulations incorporate EMT and MET as biologically realistic processes within the invasion-metastasis cascade. By considering changes in the extracellular matrix density of primary and secondary tumor growth sites, these simulations predict tumor shape and metastatic distribution.ref.56.48 ref.56.14 ref.56.14 They also take into account the immune response at secondary sites, including dormancy and death of metastasized cancer cells. The inclusion of EMT and MET in these simulations provides valuable insights into the complex mechanisms governing cancer metastasis.ref.56.48 ref.56.14 ref.56.47

Targeting EMT as a Therapeutic Strategy

Given the critical role of EMT in cancer metastasis, targeting EMT regulators holds promise as a therapeutic strategy to inhibit metastasis and improve patient outcomes.ref.117.11 ref.132.2 ref.110.20

Various EMT regulators have been identified as potential targets for inhibiting cancer metastasis. TWIST1, CD44, and other regulators have been shown to induce metastasis, and their inhibition has been found to decrease metastasis and improve survival. For example, targeting TWIST1 and CD44 has been demonstrated to decrease metastasis and improve survival in breast cancer models.ref.124.15 ref.124.1 ref.117.11 Inhibition of TWIST1 has also been found to increase survival and decrease metastases in mouse xenograft models. These findings highlight the potential of targeting EMT regulators as a therapeutic approach.ref.124.1 ref.119.3 ref.117.11

Breast cancer is one of the cancer types where EMT and MET have been extensively studied. Studies have shown that breast cancer cells can undergo MET, reverting back to an epithelial phenotype under certain conditions. Restoration of a more epithelial morphology to mesenchymal-like breast cancer cells has been observed through co-culture with hepatocytes.ref.152.5 ref.149.18 ref.149.18 Combination treatments targeting EMT regulators, such as TWIST1 inhibition in combination with doxorubicin, have shown promise in decreasing metastasis. Other studies have explored the use of telomerase inhibitors and combination therapy to target cancer stem cells (CSCs) and metastasis. Understanding the role of specific EMT regulators in breast cancer metastasis is crucial for the development of effective therapeutic strategies.ref.149.18 ref.124.15 ref.152.5

Several specific EMT regulators have been identified as potential targets for inhibiting cancer metastasis. These regulators include TWIST1, FOXC2, FOXQ1, SIX1, SOX4, Tenascin-C, miR-335, miR-206, miR-126, miR-31, CD44, Snail1, Snail2, Zeb1, and KLF-8. Inhibition of these regulators has been shown to play a role in suppressing metastasis in various cancer types.ref.124.15 ref.124.1 ref.124.11 Additionally, targeting signaling molecules important for maintaining the EMT phenotype, such as CD44, has shown promise in selectively targeting EMT cells. These findings suggest that targeting EMT regulators could be a potential therapeutic strategy for inhibiting cancer metastasis.ref.117.11 ref.117.11 ref.124.1

Different therapeutic approaches have been explored to target EMT in cancer metastasis. Drugs like Metformin and Resveratrol have been shown to inhibit EMT and target EMT cells in breast cancer and pancreatic cancer models, respectively. Combination therapy, such as docetaxel with a CXCR1/2 small-molecule inhibitor, has also been found to reduce the CSC population and metastasis in mice.ref.117.11 ref.131.2 ref.117.11 Furthermore, mathematical models of cancer metastasis have incorporated EMT-related features to accurately represent their role in the invasion-metastasis cascade. These models consider changes in cell phenotype, adhesion, and the secretion of matrix metalloproteinases (MMPs) to simulate the behavior of cancer cells during metastasis. Understanding EMT-related features and their impact on therapeutic response can aid in the development of effective strategies to prevent and treat cancer metastasis.ref.152.5 ref.117.11 ref.132.2

The Emergence of Hybrid E/M Phenotypes and CTC Clusters

During cancer metastasis, hybrid epithelial/mesenchymal (E/M) phenotypes that arise during EMT contribute to the formation of circulating tumor cell (CTC) clusters. These clusters are formed by cancer cells in hybrid E/M phenotypes, combining epithelial traits such as cell-cell adhesion with mesenchymal traits such as increased motility. The presence of these hybrid E/M phenotypes allows cancer cells to migrate collectively as a cluster, which can enter the bloodstream intact.ref.119.2 ref.119.2 ref.143.1 CTC clusters have been shown to be primary instigators of metastases and are associated with poor patient survival. Understanding the mechanisms underlying the emergence of hybrid E/M phenotypes and the formation of CTC clusters can lead to more effective therapeutic designs targeting metastasis.ref.119.2 ref.119.2 ref.119.25

The Role of MMPs in EMT and Metastasis

Matrix metalloproteinases (MMPs) play a crucial role in the invasion and metastasis of cancer cells during EMT. These enzymes are upregulated during EMT and contribute to the invasion of cancer cells by allowing them to breach the basement membrane and invade into the surrounding tissue. The loss of cellular adhesion during EMT triggers the secretion of MMPs by mesenchymal-like cells.ref.117.8 ref.55.13 ref.128.5 MMPs are involved in cytoskeletal reorganization and the development of cellular protrusions such as filopodia, lamellipodia, and invadopodia, which support cell motility and contribute to the invasive behavior of EMT cells. Upregulated expression of vimentin and downregulated expression of E-cadherin, which are markers of EMT, have been correlated with tumor invasion and metastasis. Additionally, EMT is associated with therapeutic resistance, as it endows cancer cells with heightened resistance to chemotherapy and radiotherapy.ref.117.8 ref.128.5 ref.117.8 The increased expression of the ROS scavenger SOD2 and reduced oxygen consumption in EMT cells may contribute to their therapeutic resistance. Understanding the role of MMPs in EMT and metastasis provides insights into potential therapeutic targets and strategies to overcome treatment resistance.ref.117.8 ref.55.13 ref.117.8

In conclusion, EMT and MET are critical processes involved in cancer metastasis. EMT allows cancer cells to acquire an invasive and migratory phenotype, facilitating their spread to distant sites in the body. MET is necessary for the colonization of distant organs by reverting the cells back to an epithelial phenotype.ref.154.11 ref.117.8 ref.56.6 Specific factors and signaling pathways regulate these processes, and their dysregulation can contribute to metastasis, poor outcomes, and therapeutic resistance in cancer patients. Targeting EMT regulators has shown promise as a therapeutic strategy to inhibit metastasis and improve patient outcomes. Computational simulations and mathematical models have provided insights into the invasion-metastasis cascade, considering the role of EMT and MET.ref.56.48 ref.56.14 ref.117.8 Understanding the emergence of hybrid E/M phenotypes and the formation of CTC clusters can lead to more effective therapeutic designs targeting metastasis. The role of MMPs in EMT and metastasis highlights potential therapeutic targets and strategies to overcome treatment resistance. Overall, a comprehensive understanding of the mechanisms underlying EMT and MET is crucial for the development of effective strategies to prevent and treat cancer metastasis.ref.119.2 ref.56.14 ref.56.48

Clinical Implications and Therapeutic Strategies:

Clinical implications and therapeutic strategies related to EMT-related biomarkers in cancer prognosis and treatment

The identification and understanding of EMT-related biomarkers can have significant clinical implications in cancer prognosis and treatment. EMT-related biomarkers provide important evidence to understand the clinical role of EMT and predict clinical outcomes. For example, the presence of high TGF-β and low E-cadherin levels in patients is associated with a worse prognosis, indicating that this patient subset is more likely to benefit from therapy inhibiting the TGF-β pathway.ref.110.20 ref.110.20 ref.110.19 This information can help in selecting patients for systemic targeted therapy and guide therapeutic strategies for improved clinical outcomes.ref.109.4 ref.110.20 ref.110.20

Furthermore, the identification and characterization of circulating biomarkers denoting EMT can lead to better detection and monitoring of cancer. Assessing the number and changes in the number of circulating tumor cells (CTCs) with EMT features can provide prognostic and predictive information about the disease. Studies have shown that CTCs with EMT features can be indicative of the prognosis and response to therapy in breast, colon, and prostate cancer.ref.143.1 ref.143.34 ref.143.15 Additionally, the characterization of CTCs can help in understanding tumor heterogeneity and the development of drug-resistant clones. Comprehensive single-cell sequencing strategies have revealed that CTCs can capture tumor heterogeneity and allow real-time monitoring of the appearance of drug-resistant clones. This information can be used to guide therapeutic strategies and develop targeted therapies.ref.143.35 ref.143.34 ref.143.16

EMT is also implicated in cancer stem cells (CSCs) and therapeutic resistance. CSCs with a mesenchymal phenotype and increased expression of ABC transporters and stemness markers are more resistant to radiation and chemotherapy. Targeting these cells and inhibiting EMT can be a potential therapeutic strategy to inhibit cancer growth and evolution.ref.67.21 ref.152.5 ref.159.27 EMT-related biomarkers could be important in selecting patients for systemic targeted therapy aimed at inhibiting EMT and overcoming drug resistance. Longitudinal studies are required to explore the role of EMT biomarkers in terms of prognostic values.ref.159.27 ref.67.21 ref.143.32

The modulation of the EMT pathway mediated by certain drugs, such as aspirin, can also improve cancer prognosis. Aspirin has been shown to inhibit EMT, leading to improved clinical outcomes in cancer patients. This highlights the potential of targeting EMT as a therapeutic strategy.ref.117.11 ref.117.11 ref.110.20 Overall, the identification and understanding of EMT-related biomarkers can provide valuable insights for personalized medicine and the development of targeted therapies for cancer patients.ref.110.20 ref.117.11 ref.110.20

Challenges and limitations in targeting EMT for cancer treatment

While targeting EMT shows promise as a therapeutic strategy for cancer treatment, there are several challenges and limitations that need to be addressed. One of the main challenges is the complexity of investigating EMT. EMT is a complex process involving the activation of surface receptors by growth factors, cytokines, and extracellular matrix components.ref.132.2 ref.110.20 ref.159.27 It can be induced in tumor stromal cells, such as endothelial cells, fibroblasts, and tumor-associated macrophages. The transient nature of EMT transcription factor expression further complicates the study of EMT. EMT regulators have non-redundant functions, and the leading EMT transcription factors have distinct roles in different cancers, making it difficult to generalize findings.ref.159.27 ref.159.27 ref.132.2

Another limitation is the lack of reliable biomarkers for EMT. The lack of consensus on biomolecular/histological phenotype denoting EMT limits its use in clinical practice. Better detection methods and biomarkers are needed for the accurate detection of EMT.ref.110.13 ref.110.13 ref.110.20 The difficulty in recognizing EMT at the morphological level further emphasizes the need for improved detection methods.ref.119.23 ref.119.2 ref.110.13

Additionally, targeting EMT as a therapeutic strategy may have potential risks and side effects. One potential risk is the induction of drug resistance. EMT is a key mechanism of cancer drug resistance, and inhibiting EMT can potentially reverse drug resistance.ref.159.27 ref.114.20 ref.132.2 However, there is a risk that targeting EMT may lead to the development of alternative resistance mechanisms. Another potential risk is increased metastasis. EMT is critical for metastasis, and many EMT regulators are capable of inducing metastasis.ref.132.2 ref.159.27 ref.159.27 Targeting EMT may inadvertently promote metastasis in some cases. Furthermore, targeting EMT may promote cancer stem cell properties. EMT has been implicated in the generation of cancer-initiating cells and the induction of drug resistance.ref.132.2 ref.143.32 ref.159.27 Targeting EMT may inadvertently promote the development of cancer stem cells, leading to more aggressive and therapy-resistant tumors.ref.132.2 ref.159.27 ref.159.27

To minimize or mitigate these risks, combination therapies that target both EMT and other aspects of cancer progression, such as immune checkpoint inhibitors, may be considered. Combination therapy can target multiple pathways involved in cancer progression and reduce the risk of developing alternative resistance mechanisms. Additionally, the development of more suitable in vitro assays and technologies, such as microfluidic assays, can help in studying and understanding the tumor microenvironment and the effects of EMT.ref.159.27 ref.119.4 ref.132.2 These assays can provide a controlled 3D microenvironment and enable real-time imaging, allowing for the screening of therapeutic EMT blocking agents. Further research is needed to fully elucidate the mechanisms and clinical implications of EMT in cancer progression and therapy resistance.ref.159.27 ref.159.26 ref.132.2

Drugs and therapeutic interventions targeting EMT in cancer treatment

There are several drugs and therapeutic interventions that specifically target EMT in cancer treatment. These interventions aim to inhibit EMT and potentially reverse drug resistance. EMT is a key mechanism of cancer drug resistance, and targeting EMT can potentially sensitize tumor cells to treatment and decrease metastasis.ref.159.27 ref.159.1 ref.117.11

One example of a drug that targets EMT is eribulin. Eribulin has shown potential in reversing EMT and increasing sensitivity to active drugs on the epithelial component of the tumor. It has been studied in the treatment of triple-negative breast cancer (TNBC) and has demonstrated antitumor activity in anthracycline and taxane-resistant breast cancer.ref.114.20 ref.117.11 ref.114.19 However, its efficacy as measured by pathological complete response (pCR) has not been fully established.ref.114.19 ref.114.20 ref.114.16

Metformin, an oral anti-diabetic drug, has also been shown to target EMT cells in breast cancer. It inhibits the nuclear translocation of the EMT-inducing inflammatory transcription factor NF-κB, thereby inhibiting EMT. This highlights the potential of repurposing existing drugs for targeting EMT.ref.117.11 ref.131.12 ref.131.22

Resveratrol, a polyphenol, has been shown to downregulate key EMT master regulators in mouse models of pancreatic cancer. It represses the transcription of EMT master regulators, leading to the inhibition of EMT.ref.117.11 ref.162.44 ref.162.44

CD44-targeted therapies are also being explored as a potential therapeutic approach for targeting EMT. CD44, a cell surface glycoprotein involved in various signaling pathways in cancer, is highly expressed on cells that have undergone EMT. Targeting CD44 selectively in EMT cells may provide a promising approach for therapy.ref.117.11 ref.117.11 ref.110.20

There are ongoing clinical trials evaluating the effectiveness of drugs targeting EMT and stem cell pathways in reverting drug resistance in various types of cancer. These trials aim to evaluate the ability of these drugs to target EMT and stemness and potentially reverse drug resistance. For example, MEK inhibitors, such as SCH772984, MK-8353, ulixertinib, ravoxertinib, LTT462, and LY3214996, are currently under evaluation in preclinical and clinical studies for their ability to target EMT in melanoma.ref.146.11 ref.143.32 ref.159.25 Wnt inhibitors, such as TGFßR2, have shown potential in reversing chemoresistance caused by chronic treatment with BRAF inhibitors in melanoma. Everolimus, an mTOR inhibitor, is being evaluated in clinical trials for its ability to target EMT and stemness and potentially reverse drug resistance. Telomerase inhibitors, such as Imetelstat, have also shown potential in decreasing the population of cancer stem cells and mammosphere propagation in breast cancer.ref.146.11 ref.146.11 ref.159.25

It is important to note that while these drugs and therapeutic interventions are being investigated for their potential to target EMT and reverse drug resistance, further research and clinical trials are needed to fully evaluate their efficacy and determine their clinical usefulness. The complexities of EMT and the need to understand the mechanisms of EMT-induced therapy resistance require further investigation to fully harness the therapeutic potential of targeting EMT in cancer treatment.ref.159.27 ref.159.1 ref.143.32

Conclusion

The identification and understanding of EMT-related biomarkers have significant clinical implications in cancer prognosis and treatment. EMT-related biomarkers can improve cancer prognosis by providing important evidence to understand the clinical role of EMT and predict clinical outcomes. They can also lead to better detection and monitoring of cancer, as well as the development of targeted therapies.ref.110.19 ref.110.19 ref.110.20 Targeting EMT shows promise as a therapeutic strategy to inhibit cancer growth and evolution. However, there are challenges and limitations in targeting EMT, including the complexity of investigating EMT, the lack of reliable biomarkers, and potential risks and side effects. Despite these challenges, there are several drugs and therapeutic interventions that specifically target EMT and hold promise in cancer treatment.ref.110.20 ref.159.27 ref.110.19 Further research and clinical trials are needed to fully evaluate their efficacy and determine their clinical usefulness. The modulation of EMT has the potential to revolutionize cancer treatment and improve clinical outcomes for cancer patients.ref.110.20 ref.114.20 ref.159.27

Works Cited