scispace - formally typeset
Search or ask a question

How do fibroblasts contribute to tumor suppression through TGF-beta secretion? 


Best insight from top research papers

Fibroblasts play a crucial role in tumor suppression through TGF-β secretion. Specifically, TGF-β signaling in fibroblasts has been shown to suppress tumor metastasis by antagonizing HGF/c-Met signaling within tumor epithelial cells. Additionally, cancer-associated fibroblasts (CAFs) secrete TGF-β, which downregulates the expression of Fibronectin leucine-rich transmembrane protein 3 (FLRT3) through activating SMAD4, promoting aggressive phenotypes in colorectal cancer cells. This mechanism highlights how fibroblasts, particularly CAFs, contribute to tumor suppression by modulating the TGF-β/SMAD4 signaling pathway to inhibit tumor progression and metastasis. Targeting this pathway, such as with LY2109761, shows promise as a potential treatment strategy for advanced colorectal cancer.

Answers from top 5 papers

More filters
Papers (5)Insight
Fibroblasts promote tumor progression by secreting TGF-β, which downregulates FLRT3 via SMAD4 activation, inducing EMT in colorectal cancer cells. Inhibiting TGF-β signaling shows promise for CRC treatment.
Fibroblasts, particularly cancer-associated fibroblasts (CAFs), promote tumor immune evasion by secreting extracellular matrix components under TGF-β influence, creating an immunosuppressive microenvironment in cancer.
Fibroblasts promote tumor progression by secreting TGF-β, which downregulates FLRT3 via SMAD4 activation, inducing EMT in colorectal cancer cells. This process can be targeted for therapeutic intervention.
Fibroblasts suppress tumor metastasis by inhibiting HGF/c-Met signaling in tumor cells through TGF-β secretion, as shown in the study using Tgfbr2 knockout fibroblasts in a co-xenograft model.
Fibroblasts with intact TGF-β signaling suppress tumor metastasis by inhibiting HGF/c-Met signaling in tumor cells, highlighting a stromal role in tumor suppression through TGF-β modulation.

Related Questions

How does tgf beta induce emt?5 answersTransforming growth factor-beta (TGF-β) induces epithelial-to-mesenchymal transition (EMT) through various mechanisms. TGF-β signaling activates SMAD proteins, particularly SMAD2 and SMAD3, which translocate to the nucleus to promote target gene expression. This signaling pathway plays a crucial role in promoting EMT in cancer cells, leading to increased migration and invasion capabilities. TGF-β-induced EMT is associated with the expression of fibrogenic factors that remodel the extracellular matrix, facilitating tumor progression and metastasis. Additionally, TGF-β induces reprogramming of intracellular amino acid metabolism, specifically through the upregulation of prolyl 4-hydroxylase α3 (P4HA3), which is essential for promoting EMT in non-small cell lung cancer cells. Overall, TGF-β orchestrates a complex network of signaling pathways and molecular changes to drive EMT in cancer cells, contributing to metastasis and therapy resistance.
Does tgf beta activate foxc1 in cancer?4 answersYes, TGF-β can activate FOXC1 in cancer. While TGF-β primarily induces epithelial-to-mesenchymal transition (EMT), leading to metastasis and drug resistance, it also plays a role in activating FOXC1, which is associated with mesenchymal characteristics in metastatic cancers. Additionally, TGF-β signaling can suppress the expression of FOXC1 in certain breast cancers, influencing cell survival by inhibiting apoptotic signaling. Therefore, the relationship between TGF-β and FOXC1 in cancer is complex, involving both activation and suppression mechanisms depending on the context and cancer type.
Does TGF-beta regulate gastrin release?5 answersYes, TGF-beta plays a role in regulating gastrin release. Research indicates that TGF-beta3 inhibits gastric acid secretion, a process closely linked to gastrin release, both preventively and therapeutically in gastroduodenal ulcer disease. Additionally, in colon carcinoma cells, the interaction between TGF alpha and gastrin autocrine loops influences gastrin mRNA levels, suggesting a regulatory role of TGF alpha in gastrin production. Moreover, studies on islet growth in transgenic mice show that TGF alpha and gastrin act synergistically to stimulate islet growth, indicating a regulatory function of these growth factors in the release of gastrin. Therefore, TGF-beta does indeed regulate gastrin release through various mechanisms identified in different research studies.
What is the connection between ROS and TGF beta in EMT regulation?5 answersThe connection between ROS (Reactive Oxygen Species) and TGF-β (Transforming Growth Factor β) in EMT (Epithelial-Mesenchymal Transition) regulation is significant in cancer metastasis. TGF-β induces EMT by upregulating EMT-associated transcription factors and downregulating epithelial markers, promoting cellular migration and invasion. ROS inhibitors can diminish TGF-β-induced EMT progression, highlighting the role of redox biology in this process. Additionally, TGF-β induces NOX4 expression, leading to ROS production, which triggers metabolic reprogramming and promotes EMT in glioblastoma cells. The interplay between NOX4, ROS, and TGF-β signaling pathways suggests that ROS generation through NOX4 is a key mechanism in mediating the effects of TGF-β on EMT regulation, providing potential therapeutic targets for cancer therapy.
How does TGF-β signaling affect drug resistance in tumors?4 answersTGF-β signaling plays a role in drug resistance in tumors by promoting immunosuppressive responses and inhibiting IFN-γ signaling. TGF-β stimulation increases SHP1 phosphatase activity, decreases IFN-γ-mediated tyrosine phosphorylation of JAK1/2 and STAT1, and suppresses the expression of immune evasion-related molecules such as PD-L1, IDO1, HVEM, and Gal-9. Dual blockade of TGF-β and PD-L1 leads to superior antitumor activity, but prolonged combined treatment results in tumor resistance to immunotherapy and increased expression of immune evasion genes. TGF-β-mediated immune evasion is associated with resistance to immune checkpoint inhibitors (ICI). TGF-β-targeted therapies, including trap and bispecific antibodies, have shown potential for overcoming ICI resistance. TGF-β signaling also plays a role in the metabolic reprogramming of cancer cells, contributing to tumor growth, invasion, metastasis, recurrence, and therapy resistance.
Are astrocytes activated by TGF-β?5 answersAstrocytes are activated by TGF-β. TGF-β has been identified as one of the key factors regulating astrocyte reactivity. The genetic and pharmacological manipulation of the TGF-β signaling pathway alters astrocyte reactivity in animal models of CNS injury and disease. TGF-β induces astrocyte activation and subsequent glial scar development. TGF-β1 has been widely recognized as an injury-related cytokine associated with astrocyte scar formation in response to brain injury. TGF-β1 might also be a crucial regulator of astrocyte differentiation and function, brain homeostasis, angiogenesis, memory formation, and neuronal plasticity. In summary, TGF-β plays a significant role in activating astrocytes and modulating their reactivity and function in various contexts, including brain injury, aging, and neurodegeneration.

See what other people are reading

Are IL-6 and MPO interconnected in the development and progression of prostate cancer?
5 answers
IL-6 and MPO (Myeloperoxidase) are interconnected in the development and progression of prostate cancer. IL-6, a pleiotropic cytokine, is predominantly expressed in the stromal compartment of the prostate, including endothelial cells and macrophages, suggesting a paracrine role in disease progression. On the other hand, MPO, an enzyme primarily found in neutrophils, plays a crucial role in the inflammatory processes associated with prostate tumors. The inflammatory microenvironment in prostate cancer, characterized by the overexpression of inflammatory processes and the accumulation of immunocompetent cells, contributes to tumor development and progression. Additionally, studies indicate that cytokines like IL-6 and IL-8 are potential biomarkers for prostate cancer pathogenesis, with elevated levels correlating with disease progression. Therefore, the interplay between IL-6 and MPO within the inflammatory context of prostate cancer underscores their interconnected role in disease advancement.
Faecalibacterium and coprococcus metabolism creates butyrate?
4 answers
Faecalibacterium and Coprococcus are known to contribute to butyrate metabolism. Butyrate, a short-chain fatty acid, plays crucial roles in various therapies for tumors, including hepatocellular carcinoma (HCC). Studies have shown that butyrate is a primary energy source for non-cancerous colonocytes, emphasizing its significance in colorectal cancer (CRC) metabolism. The role of butyrate metabolism in CRC has been explored, with the identification of butyrate metabolism-related genes and the development of prognostic models based on these genes. Additionally, butyrate, as a product of gut microbial fermentation, influences metabolic health by affecting thermogenesis, lipid and glucose metabolism, appetite, inflammation, and gut microbiota regulation. Understanding the mechanisms of butyrate metabolism, including its uptake and metabolism, is crucial for its potential therapeutic applications, particularly in cancer treatment.
What is the role of immunohistochemistry in determining the prognosis of cancer patients?
5 answers
Immunohistochemistry (IHC) plays a crucial role in determining the prognosis of cancer patients by identifying specific biomarkers associated with disease outcomes. In cholangiocarcinoma, IHC-based markers like EGFR have been linked to shorter overall and disease-free survival. Similarly, in colon cancer, positive HLA-G expression was associated with higher recurrence rates, while preserved CDX2 expression correlated with a lower risk of recurrence. Furthermore, in ovarian cancer, IHC biomarkers are being studied to predict chemotherapy resistance and improve patient outcomes. Additionally, in muscle-invasive bladder cancer, IHC analysis of molecular subtypes has shown associations with disease progression and patient prognosis. Overall, IHC provides valuable insights into cancer prognosis through the identification of specific markers related to disease outcomes.
Why is butyrate delivery considered a therapeutic agent for bowel cancer and not considered a total cure?
4 answers
Butyrate delivery is considered a therapeutic agent for bowel cancer due to its anti-tumor properties and ability to regulate colon cancer progression and intestinal microecology. Butyrate, a short-chain fatty acid, exhibits anti-inflammatory, antioxidant effects, and inhibits tumor growth while promoting beneficial flora in the colon. It plays a crucial role in maintaining gut homeostasis and has been shown to inhibit glucose metabolism in colorectal cancer cells, enhancing the efficacy of chemotherapy. However, despite its promising therapeutic potential, butyrate is not considered a total cure for bowel cancer. While it can ameliorate the disease and improve survival rates, it is typically used as part of a comprehensive treatment approach, including immunotherapy and chemotherapy, due to the complex nature of cancer progression and the need for multifaceted interventions.
What is the function of RNF185/MBRL complex?
5 answers
The RNF185/MBRL complex plays a crucial role in various biological processes across different contexts. In prostate cancer, RNF185 acts as a gatekeeper of metastasis by regulating epithelial-mesenchymal transition (EMT) genes involved in cell motility and metastasis. Additionally, RNF185 has been identified as a mitochondrial ubiquitin E3 ligase that targets specific substrates like BNIP1, influencing mitochondrial homeostasis through autophagy. Moreover, RNF185 interacts with cyclic GMP-AMP synthase (cGAS) during viral infection, modulating cGAS activity through K27-linked polyubiquitination and impacting innate immune responses. In papillary thyroid carcinoma, the lncRNA RNF185-AS1 regulates the miR-429/LRP4 axis, affecting cell proliferation, migration, invasion, and tumorigenesis. Overall, the RNF185/MBRL complex exhibits diverse functions in cancer progression, autophagy regulation, immune responses, and tumor growth.
How immunocal impact infant cancer?
5 answers
Immunotherapy has revolutionized pediatric cancer treatment, offering promising outcomes for previously challenging cases. Monoclonal antibodies, bispecific antibodies, and cellular therapies like CAR T-cells and NK cells have shown significant clinical responses in pediatric cancers, including neuroblastoma and leukemia. Immunotherapies have demonstrated impressive antitumor effects in high-risk neuroblastoma and acute B-cell lymphoblastic leukemia, enhancing survival rates. Despite initial disappointments in pediatric sarcomas, ongoing trials suggest that immunotherapy will eventually improve outcomes for high-risk malignancies, broadening the therapeutic arsenal against pediatric cancers. The specificity for cancer cells, in vivo persistence, and potency against refractory diseases make immunotherapies a valuable addition to conventional treatments, potentially reducing the chemotherapeutic burden while increasing cure rates in pediatric cancer.
What is the role of IL-34 in the development and progression of prostate cancer?
5 answers
IL-34 plays a significant role in the development and progression of prostate cancer by promoting cell proliferation, apoptosis evasion, migration, epithelial-to-mesenchymal transition (EMT), and angiogenesis. It has been observed that IL-34 expression is higher in prostate cancer cell lines compared to normal tissues, and its overexpression enhances cancer cell aggressiveness. Additionally, IL-34 has been linked to the regulation of immune cells in the tumor microenvironment (TME), influencing tumor growth and metastasis. The cytokine's involvement in immune cell regulation, proliferation, and survival underscores its potential as a therapeutic target in prostate cancer treatment, offering insights for the development of novel strategies to combat this malignancy.
WHAT IS THE Role of CSCs in tumor initiation, progression, and metastasis?
5 answers
Cancer stem cells (CSCs) play a crucial role in tumor initiation, progression, and metastasis. These cells possess the ability for self-renewal, differentiation into various cell types, and resistance to conventional cancer treatments like chemotherapy and radiotherapy. CSCs contribute to tumor heterogeneity, recurrence, and metastasis by generating non-CSC progeny, promoting tumor relapse, and initiating metastatic growth. They also exhibit characteristics such as quiescence, slow proliferation, and the capability for epithelial-mesenchymal transition (EMT) and its reverse process, mesenchymal-epithelial transition (MET). Understanding the signaling pathways and regulatory mechanisms involved in CSC function during disease progression is essential for developing targeted therapies to block relapse and metastasis in various types of cancers.
What are the current treatment options and prognosis for individuals with WHIM syndrome?
5 answers
Current treatment options for individuals with WHIM syndrome include investigational drugs like mavorixafor, a CXCR4 antagonist, which has shown promising results in increasing white blood cell counts, reducing infection rates, and decreasing cutaneous warts. Plerixafor, another CXCR4 antagonist, has also demonstrated potential in treating WHIM syndrome by addressing neutropenia and other cytopenias. Patients with WHIM syndrome often receive symptomatic treatments like granulocyte colony-stimulating factor (G-CSF) to manage neutropenia. The prognosis for individuals with WHIM syndrome can vary, with some experiencing recurrent sinopulmonary infections, susceptibility to HPV infections, and increased risk of malignancy due to warts. However, ongoing research and clinical trials, such as the phase 3 trial of mavorixafor, offer hope for improved outcomes and targeted therapy for WHIM syndrome patients.
What is the role of ARID1A mutations in promoting P300-dependent endometrial invasion?
4 answers
ARID1A mutations play a crucial role in promoting P300-dependent endometrial invasion. Studies have shown that ARID1A loss-of-function mutations lead to the reprogramming of the tumor microenvironment (TME), inducing aggressive behavior in neighboring endometrial tumor cells and activating endometrial stromal cells (ESC). Additionally, ARID1A mutations in the endometrium, combined with PI3K activation, result in epithelial-to-mesenchymal transition (EMT) and collective invasion, facilitated by the dysregulation of gene expression and promoter chromatin accessibility. Furthermore, ARID1A deficiency in endometrial cells leads to the loss of TGF-β tumor suppressive function, promoting migration and invasion of PTEN-deleted endometrial tumor cells. These findings highlight the significant impact of ARID1A mutations in driving endometrial invasion through complex molecular mechanisms.
How do carbamate compounds interact with different types of tissues in vitro?
5 answers
Carbamate compounds exhibit varying interactions with different types of tissues in vitro. Studies have shown that carbaryl and aldicarb, two common carbamates, display selective toxicities towards brain and limb cells, with carbaryl being toxic to both types of tissues while aldicarb is more toxic to brain cells. Additionally, ethyl carbamate has been found to induce sister chromatid exchanges (SCE) in various tissues, with regenerating liver cells showing the highest absolute SCE values, followed by marrow cells and germ cells being the least sensitive. Furthermore, the ethyl, ethyl N-hydroxy, isopropyl, and methyl esters of carbamic acid have been studied for their SCE-inducing abilities, with different potencies observed across alveolar macrophages, bone marrow, and regenerating liver cells. These findings highlight the tissue-specific responses to carbamate compounds in vitro.