scispace - formally typeset
Search or ask a question

Is there a link between BACH2 and immune cell infiltration? 


Best insight from top research papers

BACH2 is associated with immune cell infiltration in various contexts. It has been shown to regulate the differentiation and maturation of B and T lymphocytes, including B cell development and T cell differentiation into Th2 cells . In clear cell renal cell carcinoma (ccRCC), specific somatic mutations in BACH2 are associated with distinct immune infiltration patterns within the tumor immune microenvironment (TIME) . Additionally, BACH2 is involved in the regulation of T cell biology and is correlated with immune-mediated diseases such as multiple sclerosis, rheumatoid arthritis, and inflammatory bowel disease . Furthermore, BACH2 stabilizes the differentiation of regulatory T (Treg) cells and represses commitment of CD4+ T cells to alternate cell fates, thereby limiting autoimmunity and controlling the balance between tolerance and immunity .

Answers from top 3 papers

More filters
Papers (3)Insight
The provided paper does not mention anything about BACH2 or its link with immune cell infiltration.
The provided paper does not mention anything about BACH2 or its link to immune cell infiltration. The paper focuses on the associations between specific somatic mutations in ccRCC and immune cell infiltration patterns.

Related Questions

What is the role of BACH2 in human T cells/?5 answersBACH2 plays a crucial role in human T cells by regulating immune homeostasis. It is involved in the differentiation and maturation of T cell lineages, including helper T-cell subsets like T follicular helper, Th1, Th2, Th17, and Treg cells. BACH2 expression in CD4+ T cells is elevated in systemic lupus erythematosus (SLE) patients and negatively correlates with disease severity and autoantibody levels. Silencing Bach2 in CD4+ T cells leads to an increase in B-cell count, plasmablasts, and IgG secretion, indicating its role in modulating B cell responses. Additionally, BACH2 is shown to act as a transcriptional checkpoint in peripheral Tregs, limiting precocious differentiation into effector Tregs and regulating Type 2 inflammation in the lungs.
What is immune cells infiltration in cancer?4 answersImmune cell infiltration (ICI) in cancer refers to the process where immune cells penetrate the tumor microenvironment (TME), a complex ecosystem composed of various stromal and immune cells surrounding tumor cells. This infiltration is a critical determinant of patient survival and therapeutic response, as increased immune cell presence within tumors is associated with improved outcomes and can predict the efficacy of immune therapies. The diversity and state of these infiltrating immune cells, however, exhibit substantial heterogeneity among cancer patients, influencing their response to treatments including chemotherapy, targeted therapy, and immunotherapy. The TME plays a significant role in tumor progression, metastasis, and response to therapy, with tumor-infiltrating immune cells (TIICs) being key components that affect every stage of tumor metastasis. Different cancers, such as cervical cancer, lung cancer, and breast cancer, have shown varied relationships between immune cell infiltrations, immune escape mechanisms, and responses to immunotherapies. For instance, in cervical cancer, patients with high ICI scores are more prone to immune escape due to activated pathways and may benefit more from specific types of immunotherapy. Similarly, in breast cancer, patterns of immune cell infiltration can act as prognostic indicators, with higher ICI scores associated with better clinical outcomes. Advanced methodologies like the SpatialVizScore and the use of deconvolution algorithms have enabled a more detailed quantification and understanding of immune cells' spatial infiltration within tumors, revealing the immune continuum from inflamed to suppressed states. These insights are crucial for developing new prognostic markers and tailoring immunotherapeutic approaches to improve treatment outcomes in various cancers.
What is known about the role of immune infiltration in development of triple-negative breast cancer and the clinical implications?4 answersImmune infiltration plays a significant role in the development of triple-negative breast cancer (TNBC) and has important clinical implications. TNBC is characterized by high immunogenicity, specific immune activation signatures, and higher levels of tumor-infiltrating lymphocytes (TILs). TILs are known to be a prognostic and predictive biomarker in breast cancer, particularly in TNBC patients. TNBC patients with higher levels of TILs have longer overall survival and disease-free survival times, and a higher rate of pathological complete response to treatment. The immune infiltrate in TNBC is associated with a more favorable prognosis, lower risk of death and recurrence, and increased response to neoadjuvant chemotherapy. The presence of TILs and their specific subpopulations, such as CD4+ and CD8+ TILs, have been shown to have a prognostic role in TNBC. Understanding the role of immune infiltration in TNBC can help tailor treatment strategies, such as immunotherapy, to improve patient outcomes.
Is there a link between BACH2 and TNFSF11?5 answersBACH2 has been shown to play a key role in the pre-B cell receptor checkpoint and functions as a tumor suppressor in pre-B cell acute lymphocytic leukemia. It has also been demonstrated that BACH2 is required to establish immunosuppression within tumors, and its absence leads to augmented tumor clearance dependent on the adaptive immune system. Additionally, BACH2 has been found to regulate the function of CD4+ CD45RA- Foxp3l ° cytokine-secreting T cells (Fr.III cells) in systemic lupus erythematosus (SLE), promoting B-cell response. However, there is no direct mention of a link between BACH2 and TNFSF11 in the provided abstracts.
What are the effects of different therapies on the immune infiltration of the tumor?5 answersDifferent therapies have varying effects on the immune infiltration of the tumor. Studies have shown that anti-SEMA4D treatment increases the Teff:Treg ratio, indicating a favorable modulation of the immune microenvironment. Additionally, SEMA4D blockade has been found to be highly effective in mammary carcinoma models, while immune checkpoint inhibitor antibodies have shown to be ineffective. These findings suggest that SEMA4D blockade may have a positive impact on immune infiltration in tumors. However, the specific effects of other therapies on immune infiltration were not mentioned in the abstracts provided.
What are the effects of Bach2 on the development and function of iNKT cells?5 answersBach2 has been shown to play a key role in the development and function of T cells, B cells, and germinal center formation. However, there is no specific mention of the effects of Bach2 on the development and function of iNKT cells in the provided abstracts. Therefore, the effects of Bach2 on iNKT cells are not addressed in the available literature.

See what other people are reading

How have tumor-on-a-chip platforms evolved in recent years to facilitate the study of cancer-immune system crosstalk?
5 answers
Tumor-on-a-chip platforms have evolved significantly in recent years to enhance the study of cancer-immune system crosstalk. These platforms utilize microfluidic technologies to create 3D cell co-cultures that mimic the tumor microenvironment, enabling researchers to investigate immune cell recruitment, tumor progression, and response to immunotherapies in a patient-specific manner. By incorporating various cell types, such as cancer cells, immune cells, and stromal cells, these platforms can replicate the complex interactions within the tumor ecosystem. Additionally, advanced imaging techniques and computational algorithms allow for real-time monitoring of cellular behaviors and responses to treatments. Overall, tumor-on-a-chip platforms offer a promising approach to advancing our understanding of cancer-immune system interactions and improving the efficacy of immunotherapies through personalized medicine applications.
WHAT IS sex differences in cancer incidence IN SHIZOPHRENIA SPECTRUM DISORDER?
10 answers
The exploration of sex differences in cancer incidence, particularly within the context of schizophrenia spectrum disorder, necessitates a nuanced understanding of the broader landscape of sex disparities in cancer rates and the biological, environmental, and social determinants that may influence these patterns. While the provided contexts do not directly address schizophrenia spectrum disorder, they offer valuable insights into the mechanisms and patterns of sex differences in cancer incidence that could have implications for this specific population. Research has consistently shown that cancer incidence is statistically significantly higher in men than in women for a majority of non-reproductive cancer sites, with disparities exceeding 2-fold for several cancers. This male predominance in cancer incidence is echoed across various studies, indicating that men have a 20% higher chance of developing cancer over their lifetime compared to women. These differences are partly attributed to biological factors, including sex-specific genetic, epigenetic, and hormonal influences that affect cell cycle regulation, immunity, and metabolism. For instance, sex hormones and genetic differences play a role in modulating cancer risk and progression in a sex-specific manner. Environmental and lifestyle factors, alongside social determinants, also significantly contribute to sex disparities in cancer incidence. Studies have highlighted the importance of considering gender as a critical factor in understanding cancer development, pointing to the predominant environmental origin of cancer and its relationship with social determinants. Furthermore, the variability in the male-to-female incidence ratio across different regions suggests the influence of both biological factors and carcinogenic exposure differences. Given the complex interplay of genetic, hormonal, environmental, and social factors in determining cancer risk and incidence, individuals with schizophrenia spectrum disorder may exhibit unique patterns of sex differences in cancer incidence. However, specific research into cancer incidence within this population, taking into account these multifaceted determinants, is necessary to draw definitive conclusions.
Does cxcr3 pathway contributes to IL-7 increase in intra-tumoral T cells infiltration?
5 answers
The CXCR3 pathway plays a crucial role in promoting intra-tumoral T cell infiltration in response to IL-7 treatment. Studies have shown that CXCR3 activation enhances T cell antitumor reactivity, and the CXCL10/CXCR3 axis contributes significantly to the therapeutic efficacy of combined treatments involving thermal ablation and PD-1 blockade against tumors. Additionally, targeting the CXCR3 pathway with a novel peptide drug candidate has been demonstrated to mobilize the immune system to enhance anti-tumor immunity. These findings collectively suggest that the CXCR3 pathway is involved in promoting T cell infiltration into tumors, which is further supported by the observation that CXCR3-deficiency results in enhanced antigen-specific T cell IFNγ production in primary tumors.
How does the high mutation rate associated with POLE mutations contribute to the resistance to standard therapeutic approaches?
5 answers
The high mutation rate linked to POLE mutations contributes to resistance to standard therapies through various mechanisms. POLE mutations, particularly in the exonuclease domain, lead to a significantly increased tumor mutation burden (TMB). These mutations can result in a hypermutated phenotype, impacting immunological markers like TMB and microsatellite instability (MSI). Additionally, POLE mutant tumors may exhibit decreased fidelity of DNA replication, potentially affecting response to treatments. Notably, the presence of POLE mutations, even in microsatellite stable (MSS) colorectal tumors, can still make them effective targets for immunotherapy. Overall, the unique genomic characteristics of POLE mutant tumors, including high TMB and specific co-alterations, suggest a potential role in therapy resistance and the need for further investigation into tailored treatment approaches.
What are the most famous promoter mutations that are oncogenic?
5 answers
The most famous oncogenic promoter mutations are found in the telomerase reverse transcriptase (TERT) gene. These mutations occur frequently in various cancers, particularly melanomas, and are associated with increased transcriptional activity of TERT, leading to immortalization and genomic instability. The mutations create binding sites for ETS transcription factors, resulting in a 2- to 4-fold increase in gene expression. Mutant TERT promoters exhibit active chromatin marks and recruit specific transcription factors, while the wild-type allele remains silenced, showcasing a monoallelic expression pattern. Additionally, these promoter mutations have been observed in blood leukocytes of individuals with inherited loss-of-function coding mutations in TERT, indicating their role in promoting telomerase activation and increased cell proliferation.
Does STEAP-3 increase or decrease in iron overload conditions?
5 answers
STEAP3 expression varies in different conditions of iron overload. In the context of iron overload, STEAP3 expression is upregulated in certain cancers like glioblastoma, lung adenocarcinoma, and endometrial carcinoma, while it is downregulated in liver cancer. Additionally, in β-thalassemia, iron overload leads to increased levels of free reactive iron (Fe2+) in hematopoietic stem cells (HSCs), affecting their function and inducing oxidative stress. Moreover, in macrophages, Steap3 deficiency results in abnormal iron distribution and impaired iron homeostasis, impacting inflammatory responses. Therefore, the regulation of STEAP3 expression in iron overload conditions is complex, with both upregulation and downregulation observed in different cellular contexts.
Can personalized dietary interventions based on microbiome profiling prevent the onset of dysbiosis-related diseases in high-risk individuals?
5 answers
Personalized dietary interventions based on microbiome profiling hold promise in preventing dysbiosis-related diseases in high-risk individuals. Studies suggest that understanding the gut microbiota's role can aid in developing preventive strategies. Novel approaches like the Nutrition-Ecotype Mixture of Experts (NEMoE) model can establish associations between microbiota, health states, and diet-specific cohort variability, enhancing predictive performance and identifying microbial signatures of diseases. Furthermore, research indicates that the microbiome plays a crucial role in conditions like gestational diabetes mellitus (GDM), where microbial network analysis can reveal imbalances and aid in individualized diagnosis and therapy strategies. Additionally, manipulating the gut microbiome through dietary interventions has shown potential in improving the efficacy of cancer immunotherapy, highlighting the impact of lifestyle variables on specific microbial populations and host immune responses.
Why Iridium transition metal inserted complex is used as a photosensitizer in photodynamic therapy?
5 answers
Iridium transition metal complexes are utilized as photosensitizers in photodynamic therapy due to their unique properties. These complexes, such as Ir-1 and Ir-pbt-Bpa, exhibit aggregation-induced emission (AIE) characteristics, high singlet oxygen (1O2) generation ability, and pH-responsive photoluminescence emission, enhancing their efficacy in cancer treatment. Additionally, iridium-based photosensitizers like Ir-Biotin demonstrate long excitation wavelengths, effective cancer cell targeting, and high 1O2 generation yield, leading to excellent photodynamic therapy outcomes. Furthermore, the combination of iridium-based photosensitizers with other therapeutic agents, like sorafenib, in nano-chemo-phototherapy formulations, enhances cellular uptake, reactive oxygen species production, and overall therapeutic efficacy against tumors, making them a promising option for cancer treatment.
What are the current advancements in immunotherapy for cancer treatment?
5 answers
Current advancements in immunotherapy for cancer treatment have revolutionized the field by utilizing the host's immune system to target tumor cells. Researchers are focusing on enhancing targeted cancer therapy to minimize toxicity and side effects, leading to the preference for immunotherapy over conventional options like chemotherapy and surgery. Understanding immune checkpoints, developing therapeutic cancer vaccines, genetic modification of immune cells, and enhancing antitumor immune responses have paved the way for novel cancer treatments. Immunotherapies have ushered in a new era in cancer treatment, offering promising prospects for more effective and targeted therapies with higher response rates and fewer side effects. The future of cancer treatment lies in the continued development and optimization of immunotherapeutic approaches to combat this challenging disease.
What is the current state of treatment in biliary cancer?
5 answers
The current state of treatment in biliary tract cancers (BTCs) involves a multifaceted approach. Immunotherapy, particularly immune checkpoint inhibitors (ICIs) in combination with chemotherapy, has shown significant promise. Trials like TOPAZ-1 and KEYNOTE-966 have established the efficacy of combining ICIs with chemotherapy as a first-line treatment for advanced or metastatic BTCs. Additionally, targeted therapies focusing on specific genetic alterations like FGFR2, IDH, BRAF, and HER-2 are emerging as options for patients who do not respond to first-line treatments. Perioperative treatments, such as adjuvant chemotherapy with S-1, have also shown improved overall survival rates in resected BTC patients. The landscape of treatment for biliary cancer is evolving rapidly, with ongoing trials exploring novel combinations and immune-targeted strategies to enhance patient outcomes.
How do phytonutrients affect the immune system in humans?
5 answers
Phytonutrients, such as curcumin, quercetin, and flavonoids, have immunomodulatory properties that can impact the human immune system. These compounds can induce, amplify, attenuate, or prevent immune responses, thereby influencing immune homeostasis and responses to infections, allergies, and chronic diseases. Phytonutrients like Aloe vera polysaccharides, garlic, and Vitamin C have been shown to modulate key immune biomarkers, such as interleukin-6 and CD4+ T cells, potentially reducing inflammation and enhancing immune function. The bioactive potential of phytochemicals, including alkaloids, flavonoids, and terpenoids, allows them to serve as natural immunomodulators, aiding in cancer chemoprevention and overall immune system regulation. Therefore, incorporating phytonutrients into the diet may offer a complementary approach to support immune health and combat various immune-related conditions.