scispace - formally typeset
Search or ask a question

What is the role of GPR81 in regulating lactate signaling in the body? 


Best insight from top research papers

GPR81, identified as a lactate receptor, plays a multifaceted role in regulating lactate signaling across various tissues in the body, influencing metabolic processes, immune responses, and even cancer progression. In adipose tissue, GPR81 activation by lactate inhibits lipolysis, suggesting a role in lipid metabolism and obesity management. This is supported by findings that GPR81 deficiency impairs the ketogenic response and exacerbates hepatic lipid accumulation, indicating its critical role in hepatic lipid homeostasis and potential as a target for treating metabolic disorders. Moreover, dietary lactate supplementation, activating GPR81, has been shown to promote adipose browning and enhance thermogenesis, offering insights into obesity management through the lactate-GPR81 signaling axis. Beyond metabolic regulation, GPR81's expression in renal and vascular tissues implicates it in the modulation of blood pressure and renal hemodynamics through mechanisms involving the potent vasoconstrictor ET-1, highlighting lactate as a paracrine regulator of renal blood flow. In the context of cancer, GPR81 facilitates tumor growth by promoting lactate-induced cell proliferation and reprogramming energy metabolism in breast cancer cells, underscoring its pathological role and potential as a therapeutic target. Additionally, GPR81 activation in the tumor microenvironment suppresses immune surveillance by inhibiting antigen presentation, suggesting a mechanism by which tumors evade immune detection. GPR81 also contributes to neuroprotection and developmental brain angiogenesis, where lactate acting through GPR81 accelerates angiogenesis and attenuates hypoxic/ischemic brain injury by restoring compromised microvasculature. Furthermore, its high expression in type II muscle fibers suggests a role in autocrine signaling within muscles, although its effect on CREB signaling during exercise appears limited. Collectively, these findings underscore GPR81's integral role in lactate signaling, influencing a wide range of physiological and pathological processes.

Answers from top 10 papers

More filters
Papers (10)Insight
GPR81 promotes breast cancer growth through a paracrine mechanism involving antigen-presenting cells in the tumor microenvironment by inhibiting MHCII presentation, impacting cancer immunotherapy potential.
GPR81 activation by lactate inhibits lipolysis in adipocytes and regulates renal hemodynamics through endothelin-1 release, suggesting a role in systemic and renal physiology.
GPR81 plays a crucial role in adipose browning by responding to lactate, promoting thermogenesis, and protecting against obesity through the lactate-GPR81 signaling axis in mice.
GPR81 regulates brain angiogenesis by responding to lactate, promoting vascular development and aiding in brain recovery after hypoxic ischemic injury in neonates, as per the research findings.
GPR81 plays a crucial role in promoting breast cancer progression by modulating lactate metabolism and oxidative stress, highlighting its significance in tumor growth and metabolic pathways.
GPR81 in neurons regulates brain angiogenesis by responding to lactate, promoting vascular development and aiding in brain recovery post-hypoxic ischemic injury, as per the research findings.
GPR81 acts as the endogenous receptor for lactate, playing a crucial role in metabolic processes and inflammatory responses, highlighting its significance as a metabolic sensor and inflammatory mediator.
GPR81 regulates glycolysis and lactate-dependent ATP production in breast cancer cells, impacting cell proliferation, migration, and invasion, suggesting a critical role in energy metabolism and malignancy.
GPR81, predominantly expressed in type II muscle fibers, suggests a potential role for lactate as an autocrine signaling molecule in muscle, influencing energy metabolism without regulating CREB signaling during exercise.
GPR81 mediates hepatic lipid metabolism, influences PGC-1α and L-CPT1, and plays a role in the therapeutic effect of metformin on NAFLDs, rather than directly regulating lactate signaling.

Related Questions

Does glp1r protein interact with GPR40?4 answersThe GLP-1 receptor (GLP1R) does not directly interact with GPR40. GLP-1 plays a crucial role in diabetes treatment by enhancing insulin secretion. On the other hand, GPR40, also known as FFA1R, is a G-protein-coupled receptor activated by free fatty acids, influencing insulin release. While GLP-1 receptor activation leads to insulin secretion, GPR40 activation by fatty acids stimulates GLP-1 secretion indirectly through enteroendocrine L cells. Therefore, although both receptors are involved in glucose metabolism and insulin secretion, they operate through distinct pathways without direct interaction.
What is the relationship between MCT-1 and lactate?4 answersMCT-1 is a lactate transporter that plays a role in lactate metabolism. It is expressed in various cell types, including tumor cells, glial cells, and neurons. In tumor cells, MCT-1 facilitates the export of lactate, which is produced during glycolysis. Inhibition of MCT-1 can lead to intracellular lactate accumulation and disruption of cellular metabolism, resulting in cytotoxicity. In glial cells, MCT-1 facilitates the transport of lactate to neurons, providing them with an energy source. This process is important for maintaining neuronal activity and axonal function. The regulation of MCT-1 expression and function is complex and can be influenced by factors such as oncogenes like Myc, which controls the transcription of MCT-1. Overall, MCT-1 plays a crucial role in lactate transport and metabolism in various cell types, and its inhibition or dysregulation can have significant effects on cellular function and viability.
What is the G proteins are involved in GPR27?4 answersGPR27 is a receptor that belongs to a small subfamily of three receptors called "Super-Conserved Receptors Expressed in the Brain" (SREB). It has been linked to key physiological processes such as neuronal plasticity, energy metabolism, and pancreatic β-cell insulin secretion and regulation. In terms of G proteins, GPR27 has been found to have ligand-independent activity in G protein-mediated pathways and β-arrestin 2 recruitment. However, no ligand-independent activity could be measured in these pathways. Interestingly, no G protein activation was detected upon activation of GPR27 by specific surrogate agonists. Therefore, the involvement of G proteins in GPR27 signaling pathways is still unclear based on the available information. Further research is needed to fully understand the G protein interactions and signaling mechanisms of GPR27.
What are the effects of lactate on muscle function during exercise?5 answersLactate has various effects on muscle function during exercise. It has been shown to enhance pyruvate + malate- and glutamate + malate-induced respiration, which is driven by complex I. On the other hand, lactate administration decreases succinate + rotenone-induced respiration, which is driven by complex II. Lactate also increases the enzymatic activity of complex I and the protein content of NDUFS4, a subunit of complex I. Additionally, lactate can serve as a myokine or exerkine with autocrine-, paracrine-, and endocrine-like functions, influencing various physiological processes such as fueling the heart, muscle, and brain, controlling cardiac output and breathing, and growth and development. Furthermore, lactate treatment has been shown to increase protein synthesis factors in skeletal muscle. Overall, lactate plays a significant role in modulating mitochondrial respiratory function and muscle adaptation during exercise.
What is the relationship between BMI and lactate production during and post exercise in young adults?3 answersBMI and lactate production during and post exercise in young adults are related. In females, the relationship between BMI and sum of skinfold thickness (SST) is higher than in males, especially in exercised females compared to sedentary counterparts. Lactate production during exercise is influenced by factors such as aerobic and anaerobic processes. Anaerobic processes contribute to lactate production, which can be measured in arterial blood. The lactate response during active recovery differs between athletes and non-athletes, with athletes showing a more favorable lactate response. In children, there is an association between leukocyte and lactate response following exercise, with older children demonstrating higher leukocyte levels and smaller fold change in lactate levels compared to younger children. Overall, the relationship between BMI and lactate production during and post exercise in young adults is influenced by factors such as gender, exercise participation, and age.
How does the Lactic Acid system work for exercise?5 answersThe lactic acid system for exercise involves the production and utilization of lactate in the body. Lactate is produced during anaerobic glycolysis in skeletal muscle and white adipose tissue. It is then released into the bloodstream and can act as a signaling molecule through the GPR81 receptor. During exercise, there is a shift in fuel utilization from lipid to carbohydrate, and lactate may play a role in regulating this process. Lactate can also be used as a fuel by CD8+ T cells, increasing their proliferative capacity and cytotoxicity. Additionally, lactate can reduce malignant tumor growth in a CD8+ T cell-dependent manner. Overall, lactate is now recognized as an important intermediate of carbohydrate metabolism and can have various effects on metabolism and exercise performance.

See what other people are reading

What are the Clinical and preclinical states of CAR T cell therapy in vivo for solid tumors cancer?
5 answers
Clinical and preclinical states of CAR T-cell therapy for solid tumors cancer are evolving. Challenges like antigen heterogeneity and immunosuppressive tumor microenvironment hinder CAR T-cell efficacy in solid tumors. Strategies to enhance CAR T-cell function include incorporating co-stimulatory domains, developing armored CAR-T cells, and exploring other immune cells like CAR-NK and CAR-M cells. Novel CAR T-cell products like YTB323 show enhanced in vivo expansion and antitumor activity in preclinical models, with promising safety and efficacy profiles in clinical trials for r/r DLBCL. To overcome limitations such as restricted trafficking, hypoxic TME, antigen escape, and exhaustion, researchers are advancing CAR designs and combining therapies to optimize clinical efficacy in solid tumors.
What are the drug resistant mechanism of glioblastoma?
5 answers
Glioblastoma (GBM) exhibits drug resistance mechanisms that hinder effective treatment. Resistance to temozolomide (TMZ), a common chemotherapy for GBM, arises from various factors. These include DNA repair processes, cellular autophagy, glioma stem cells, and the tumor microenvironment. Additionally, the development of multidrug resistance in GBM involves the selection of resistant tumor clones during therapy, leading to tumor growth and angiogenesis, which supports metastasis and recurrence. Furthermore, platinum drugs like cisplatin face resistance in gliomas due to factors such as decreased drug concentration, enhanced DNA repair, and altered apoptosis pathways. Understanding these resistance mechanisms at the molecular level is crucial for developing effective treatment strategies for combating drug resistance in glioblastoma.
What is the mechanism of reverse cholesterol transport pathway?
5 answers
The reverse cholesterol transport (RCT) pathway involves the transport of excess peripheral cholesterol to the liver for conversion into bile acids and subsequent excretion, reducing hyperlipidemia and cardiovascular disease risks. Key proteins involved in RCT include ATP-binding cassette sub-family A1 (ABCA1), scavenger receptor class B type I (SR-BI), apolipoprotein A-I (ApoA-I), and apolipoprotein A-II (ApoA-II). Additionally, liver X receptor (LXR) and its downstream regulators ABCA1 and ATP-binding cassette subfamily G member 1 (ABCG1) genes play a crucial role in regulating cholesterol metabolism and the RCT pathway. Furthermore, the ATP-binding cassette transporter A1 (ABCA1) is central in mediating the biosynthesis of high-density lipoprotein (HDL), facilitating cholesterol efflux from macrophages and preventing lipid deposition, thus contributing to RCT.
What are the potential mechanisms underlying the association between ICIP and TMB, and how might these influence treatment decisions?
5 answers
The association between immune checkpoint inhibitor therapy (ICI) and tumor mutational burden (TMB) involves several key mechanisms. High TMB has been linked to improved survival with ICI treatment, but not all high-TMB patients respond well. Specific gene alterations impact outcomes differently in TMB-high and TMB-low patients, influencing response to ICI. For instance, mutations in genes like STK11 and E2F3 are associated with worse outcomes in TMB-high tumors, while mutations in genes like NTRK and PTPRD are linked to better outcomes. Additionally, the immune-excluded phenotype, characterized by high stromal TIL and low intratumoral TIL density, is correlated with high TGFBs and poor response to immunotherapy. These insights can guide treatment decisions by helping identify patients likely to benefit from ICI based on their TMB status and specific gene alterations.
What is the shifting-mosaic steady-state equilibrium?
5 answers
The shifting-mosaic steady state (SMSS) equilibrium refers to a dynamic landscape pattern where patches within an area undergo continuous change while the total area of each landscape component remains relatively constant over time. This concept challenges the traditional view of landscape patterns as spatially stationary, highlighting the dynamic nature of ecosystems. Similarly, in the context of tumor-immune interactions, an equilibrium is established between effector and regulatory T cells within the tumor microenvironment, impacting the outcome of immune control over tumors. Furthermore, in tree population dynamics, factors like growth-dependent thinning, changing mortality risks, and episodic recruitment contribute to the dynamic equilibrium within forest ecosystems. Understanding such dynamic equilibria is crucial for environmental conservation, immune response modulation in cancer therapy, and managing chronic virus infections.
What is an integrin inhibitor?
5 answers
An integrin inhibitor is a type of compound that targets integrin receptors, which play crucial roles in cell-matrix and cell-cell interactions. Integrins are involved in various cellular activities like migration, survival, and growth, making them significant in cancer progression and fibrotic diseases. Cilengitide, a cyclic RGD pentapeptide, is an example of an integrin inhibitor that targets specific integrins like αvβ3, αvβ5, and α5β1. Studies have shown that integrin inhibitors like cilengitide can impact cell adhesion, migration, proliferation, and invasion. These inhibitors have been explored for their potential in cancer treatment, particularly in meningiomas, where they have shown antimigratory properties and potential radiosensitization effects.
Rosacea could be treated using molecules that binds to CB2R?
5 answers
Cannabinoids, which can interact with receptors like CB2R, have shown potential in treating rosacea. While traditional pharmacological approaches focus on managing inflammation in rosacea, there is also interest in utilizing compounds like those binding to CB2R for treatment. Additionally, a dermocosmetic active ingredient composed of glucosylated derivatives of natural plant acids has demonstrated anti-inflammatory effects on cellular models and clinical efficacy in managing erythematotelangiectatic rosacea symptoms. Therefore, molecules that bind to CB2R, such as cannabinoids, hold promise in addressing the complex pathophysiology of rosacea by modulating inflammation and potentially offering a novel therapeutic approach for this prevalent skin condition.
Paragraph 1: Anosmin-1 and fibroblast growth factors (FGFs) are critical players in the intricate molecular pathways underlying Kallmann’s syndrome?
5 answers
Anosmin-1, encoded by the ANOS1 gene, is crucial in Kallmann syndrome (KS) due to its role in olfactory bulb (OB) dysfunction and hypogonadism. Additionally, fibroblast growth factor receptor 1 (FGFR1) is another key gene associated with KS, with mutations identified in Chinese KS families. Disruptions of KS-linked genes like HS6ST1 and Fgfr1 affect similar biological processes, indicating convergent molecular mechanisms underlying KS. Furthermore, ANOS1 mutations, such as exon 3 deletion, have been linked to KS, emphasizing the genetic heterogeneity of the condition. Overall, Anosmin-1 and FGFs play essential roles in the complex molecular pathways of Kallmann syndrome, highlighting the significance of these genes in the pathogenesis of the disorder.
Do PD-L1 inhibitors slow TNBC xenograft growth in nude mice?
5 answers
PD-L1 inhibitors have shown efficacy in slowing TNBC xenograft growth in nude mice. Studies using a murine breast cancer model demonstrated that anti-PD-L1 treatment significantly suppressed primary tumor growth and extended survival. Additionally, in a study involving a syngeneic mouse tumor model, Dato-DXd, an ADC targeting TROP2, enhanced antitumor activity when combined with PD-1/PD-L1 inhibitors, suggesting a potential synergy in inhibiting tumor growth. Furthermore, PD-1 blockade in nude mice resulted in decreased tumor growth, indicating the importance of PD-1 signaling in promoting tumor progression. These findings collectively support the notion that PD-L1 inhibitors can effectively slow TNBC xenograft growth in nude mice, highlighting their potential as a therapeutic strategy for TNBC.
What are downsides of ethanol treatment on cancer cells?
5 answers
Ethanol treatment poses several downsides on cancer cells. Firstly, ethanol induces oxidative stress in cancer-associated fibroblasts, promoting myofibroblastic differentiation and autophagy in the tumor microenvironment. This oxidative stress leads to ketone production in fibroblasts and ketone re-utilization in epithelial cancer cells, fueling tumor growth via mitochondrial metabolism. Additionally, heavy alcohol consumption increases cancer stem cell (CSC) populations in various cancers, potentially enhancing tumor promotion and aggressiveness. Ethanol's metabolism generates free radicals that interfere with essential cellular functions, affecting growth and differentiation processes, including disturbances in CSCs. Furthermore, ethanol treatment alters protein expression in fibroblast cells, resembling patterns seen in nasopharyngeal carcinoma-associated stromal fibroblasts, hinting at potential cancer-related pathways being targeted by ethanol.
What are the potential side effects of Clindamycin in newborns?
5 answers
Clindamycin use in newborns can lead to potential side effects. Tetracycline antibiotics, including clindamycin, may cause yellow pigmentation of primary dentition and inhibit normal skeletal growth in premature infants. Additionally, clindamycin therapy in hospitalized patients has been associated with diarrhea and pseudomembranous colitis, with a higher tendency for diarrhea in seriously ill patients and those with abdominal or pelvic sepsis. While hepatotoxicity is rare, systemic clindamycin therapy has been linked to transient serum aminotransferase elevation and acute idiosyncratic liver injury in adults. Therefore, caution should be exercised when considering clindamycin therapy in newborns to monitor for these potential adverse effects.