Fact-checked by Grok 2 weeks ago

LGR5

LGR5, also known as leucine-rich repeat-containing 5, is a seven-transmembrane receptor protein that acts as a specific marker for in self-renewing tissues such as the intestinal crypts and hair follicles. It functions primarily as a high-affinity receptor for R-spondin (RSPO) proteins, which bind to its extracellular domain to potentiate canonical Wnt/β-catenin signaling, thereby promoting proliferation, maintenance, and tissue . Discovered as a Wnt target , LGR5 plays a critical role in postembryonic development and regeneration, with its expression tightly regulated in cycling stem cell populations. The structure of LGR5 features a large N-terminal extracellular domain composed of 17 leucine-rich repeats for ligand binding, followed by seven transmembrane helices characteristic of G protein-coupled receptors (GPCRs), and a short intracellular C-terminal tail that lacks typical G-protein activation motifs. Unlike classical GPCRs, LGR5 does not couple to heterotrimeric G proteins but instead enhances Wnt signaling by stabilizing the Wnt receptor complex on the cell surface, counteracting the inhibitory effects of E3 ligases such as RNF43 and ZNRF3 through RSPO-mediated interactions. This mechanism amplifies β-catenin accumulation and transcriptional activation of Wnt target genes, including LGR5 itself, forming a loop essential for dynamics. LGR5 expression is highly specific to stem cell niches, with prominent localization in the base of intestinal crypts where it marks rapidly cycling columnar cells responsible for epithelial renewal, as well as in the bulge of follicles and other sites like the and . In the intestine, LGR5-positive s generate all cell lineages of the every 3–5 days, highlighting its role in continuous turnover. Beyond normal physiology, dysregulated LGR5 signaling contributes to tumorigenesis, as it is overexpressed in various cancers including colorectal, ovarian, and pancreatic, where it identifies cancer s capable of tumor and . Therapeutic targeting of the LGR5/RSPO axis is emerging as a strategy to inhibit cancer progression while sparing normal function.

Molecular Biology

Gene Characteristics

The LGR5 gene, originally identified in 1998 as an orphan (initially termed HG38 or GPR49) within the glycoprotein hormone receptor subfamily, encodes a receptor with potential for binding novel ligands. This gene is situated on the long arm of human at cytogenetic band 12q22-23, spanning approximately 145 kilobases from position 71,439,797 to 71,586,310 (GRCh38 assembly) and comprising 18 exons in its primary transcript (ENST00000266674). LGR5 encodes a precursor protein of 907 , which undergoes processing to form the mature receptor. Its promoter region, located upstream of exon 1, includes TCF/LEF-binding sites that enable direct transcriptional activation by canonical Wnt signaling, establishing LGR5 as a key Wnt target gene. Additional regulatory elements, such as enhancers identified through interaction analyses (e.g., GeneHancer regions at chr12:71,438,701–71,442,291), modulate its tissue-specific expression. In adult human tissues, LGR5 exhibits basal expression in organs including , placenta, spinal cord, and brain, but demonstrates markedly elevated levels in restricted niches such as the crypt base of the small intestine and colon, as well as the bulge of hair follicles, where it serves as a marker for cycling stem cells.

Protein Structure

LGR5 is a class A (GPCR) featuring seven transmembrane α-helical domains that anchor the protein in the plasma membrane, a defining architectural element shared with rhodopsin-like GPCRs. The extracellular N-terminal ectodomain is composed of 17 leucine-rich repeats (LRRs), which fold into a β-sheet-rich horseshoe structure essential for recognition and receptor activation. This LRR array, spanning approximately residues 22–541, positions the binding site away from the membrane, facilitating interactions with extracellular cues while the hinge region connects it to the first transmembrane helix. The protein includes an N-linked site at 208 (Asn208) within the ectodomain, a conserved feature that supports proper folding, trafficking, and stability of the receptor, aligning with glycosylation patterns observed in related receptors. Sequence conservation is high across mammals; for instance, human LGR5 shares about 90% identity with Lgr5 in the extracellular , underscoring its evolutionary preservation for core receptor functions. At the intracellular C-terminus, LGR5 possesses a PDZ-binding motif (typically class I type, involving the terminal residues) that enables recruitment of PDZ-domain-containing scaffold proteins, such as DLG1, to regulate receptor localization and assembly of signaling es. Although no full-length exists, insights from the crystallized LGR5 ectodomain (PDB: 4KNG) in with R-spondin 1 reveal a rigid, elongated horseshoe conformation of the LRRs, with low root-mean-square deviation (RMSD) values across structures, enabling accurate of the transmembrane and intracellular regions based on related LGR family members like LGR4 and canonical class A GPCRs such as rhodopsin.67254-4/fulltext)00287-8)

Biological Function

Ligand Interactions

LGR5 serves as a receptor for the R-spondin family of ligands, specifically RSPO1 through RSPO4, which bind to its extracellular domain with high affinity to potentiate Wnt/β-catenin signaling. Among these, RSPO1 demonstrates the highest binding affinity for LGR5, with a dissociation constant (Kd) of approximately 3.1 nM as determined by surface plasmon resonance assays. The other RSPOs (RSPO2-4) also bind LGR5, albeit with slightly lower affinities in the low nanomolar range, enabling selective enhancement of Wnt pathway activity in a ligand-dependent manner. The binding mechanism involves the formation of an RSPO-LGR5 complex that stabilizes interactions between Frizzled (FZD) receptors and low-density lipoprotein receptor-related proteins 5 and 6 (LRP5/6), thereby promoting the assembly of the Wnt signalosome without LGR5 directly binding Wnt ligands. LGR5 functions exclusively as a co-receptor in this context, relying on RSPO ligation to counteract the inhibitory effects of E3 ubiquitin ligases such as RNF43 and ZNRF3, which otherwise degrade FZD and LRP5/6 from the cell surface. This initial ligand-receptor interaction is crucial for amplifying low-level Wnt signals in stem cell niches. Experimental validation of these interactions comes from radioligand binding assays and cell-based functional studies, which show that RSPO binding to LGR5 directly correlates with increased Wnt reporter activity and is absent in LGR5-deficient cells. Furthermore, genetic knockout studies in mice reveal the dependency of LGR5-mediated signaling; while single Lgr5 knockouts exhibit only mild intestinal phenotypes, combined Lgr4/Lgr5 double knockouts completely abolish and formation , underscoring RSPO's essential role in sustaining Lgr5+ expansion.00287-8)

Signaling Pathways

LGR5 primarily functions as a receptor for R-spondin (RSPO) ligands, potentiating the canonical Wnt/β-catenin signaling pathway by forming a complex that inhibits the E3 ligases ZNRF3 and RNF43. This LGR5-RSPO complex binds to and neutralizes ZNRF3/RNF43, preventing their ubiquitination and subsequent degradation of Wnt receptors such as (FZD) and receptor-related protein 6 (LRP6) on the cell surface. By stabilizing these receptors, LGR5 enhances the sensitivity of cells to Wnt ligands, thereby amplifying downstream signaling without directly initiating the pathway. Upon Wnt ligand binding to the stabilized FZD/LRP6 receptors, LGR5 facilitates the recruitment of intracellular effectors to the plasma membrane, including (DVL) and AXIN, which are critical for β-catenin stabilization. The formation of a supercomplex involving LGR5, FZD, LRP6, and DVL promotes the of LRP6 at key residues, such as serine 1490, which in turn recruits and sequesters AXIN from the β-catenin destruction complex (comprising AXIN, , GSK3β, and CK1). This inhibition disrupts β-catenin ubiquitination and degradation, allowing its accumulation in the and subsequent nuclear translocation to activate transcription factors like TCF/LEF. LGR5-mediated signaling operates independently of heterotrimeric G-proteins, distinguishing it from other G-protein-coupled receptors, and relies predominantly on for . Studies indicate that while LGR5 can interact with G-proteins , physiological Wnt potentiation does not require G-protein activation, as evidenced by the lack of effect from G-protein inhibitors on RSPO-LGR5-induced and β-catenin stabilization. Evidence from genetic models underscores LGR5's essential role in Wnt signaling integrity, particularly in intestinal crypts. Lgr5 null mice exhibit defective Wnt/β-catenin signaling, manifested as deregulated pathway activity leading to precocious differentiation and impaired crypt homeostasis. These disruptions culminate in 100% perinatal lethality, characterized by gastrointestinal distension and due to insufficient suckling from .

Physiological Roles

Intestinal Stem Cells

LGR5-positive (LGR5+) cells, located at the base of intestinal , function as stem cells that drive the homeostatic renewal of the . These cells undergo rapid division to replenish the epithelial lining, which turns over completely every 3-5 days in mammals. Positioned between Paneth cells, LGR5+ CBC stem cells maintain their stemness through interactions with the niche environment, ensuring continuous production of differentiated progeny that migrate upward along the crypt-villus axis. Lineage tracing experiments have confirmed the multipotency and long-term self-renewal capacity of LGR5+ cells. In seminal studies using Cre-inducible reporters targeted to the Lgr5 locus, individual LGR5+ cells generated all major epithelial lineages—including enterocytes, goblet cells, enteroendocrine cells, and tuft cells—over extended periods, demonstrating their role as the primary source of intestinal epithelial cells during steady-state conditions. These findings established LGR5 as a definitive marker for active intestinal stem cells, distinguishing them from more quiescent populations. In response to injury, such as , LGR5+ progenitors exhibit temporary expansion to facilitate epithelial regeneration. of LGR5+ cells impairs recovery from damage, underscoring their indispensability for restoring tissue integrity after acute insults, though other progenitor pools may contribute under severe stress. This regenerative plasticity allows LGR5+ cells to proliferate robustly in the altered niche, supporting rapid repopulation of the crypts. LGR5+ s rely on Paneth cells for niche signaling, which provide essential factors like EGF and Wnt ligands to sustain their function. Paneth cells secrete Wnt3a to establish a high Wnt signaling gradient at the base, promoting LGR5-mediated β-catenin stabilization and maintenance, while EGF supports . Competition for these limited niche signals among LGR5+ cells and Paneth cells regulates stem cell numbers and positioning within the .

Roles in Other Tissues

Beyond the , LGR5 expression is observed in various other tissues, where it marks stem or populations adapted to tissue-specific regenerative demands, such as injury-induced repair or developmental maintenance. These LGR5-positive (LGR5+) cells often respond to Wnt signaling via R-spondin ligands, enabling and in contexts distinct from the continuous of the gut. In the liver, LGR5 is expressed at low levels in pericentral hepatocytes during homeostatic adult but is rapidly upregulated in biliary epithelial cells within ductules and pericentral hepatocytes following , such as partial or toxin-induced damage. These LGR5+ cells, derived from ductules, serve as facultative progenitors that contribute to hepatocyte regeneration by proliferating and differentiating into parenchymal cells during the phase. For instance, in models of liver damage, single LGR5+ cells isolated from injured livers can be expanded as organoids under Wnt/R-spondin conditions, demonstrating their stem-like potential and ability to generate functional liver . Pericentral hepatocytes express LGR5 during and contribute to zonal regeneration, with their role complementing that of ductular cells post-. Recent studies (as of 2025) highlight LGR5+ hepatocytes' contributions to liver zonation and replenishment during specific injuries. No overt liver is observed in homeostatic Lgr5 knockouts, consistent with its low basal expression. During kidney development, marks a subset of nephron progenitor cells in the cap mesenchyme of the embryonic mouse kidney, where it supports the and expansion of these progenitors essential for nephron formation. Lineage tracing in Lgr5 reporter mice reveals that LGR5+ cells contribute to the generation of specific nephron segments, including the thick ascending limb and , with expression peaking during the proliferative phase of nephrogenesis around embryonic days 12.5–14.5. R-spondin signaling through LGR5 is critical for sustaining this progenitor pool, as its disruption impairs distal nephron segment formation. In postnatal kidneys, LGR5 expression diminishes after the first week, reflecting the cessation of nephrogenesis in mammals. In the stomach, LGR5 primarily identifies active populations at the base of in the (pylorus), driving epithelial renewal through self-renewal and multipotent . In the , LGR5+ cells in the pyloric glands undergo symmetric divisions during , enabling rapid clonal expansion to replenish the every few days; these cells give rise to all glandular lineages, including mucus-secreting cells and enteroendocrine cells. In the , LGR5 expression is lower and inducible upon or infection, activating reserve progenitors to support oxyntic gland maintenance. Lineage tracing confirms their long-term contribution to antral unit renewal. LGR5+ progenitors also play roles in cyclic regeneration within sensory and integumentary structures. In the , LGR5 marks supporting cells in the cochlear sensory epithelium of neonatal mice, which act as Wnt-responsive progenitors capable of proliferating and transdifferentiating into hair cells following ototoxic damage. Activation of β-catenin signaling in these LGR5+ cells enhances mitotic regeneration, partially restoring auditory function in damage models, though this capacity wanes in adults. In hair follicles, LGR5 expression labels bulge stem cells that orchestrate the hair growth , with these cells mobilizing during anagen to regenerate the follicle structure; they contribute to epithelial compartments of the follicle in a multipotent manner. Across these tissues, LGR5 expression levels vary, with high basal expression in the rapidly renewing and (comparable to intestinal crypts) but inducible or developmental-restricted patterns in liver and , as quantified by RNA sequencing and reporter mice showing 10–100-fold upregulation post-injury or during embryogenesis. studies in mice reveal tissue-specific phenotypes: Lgr5-null mutants exhibit perinatal lethality due to gut defects, but conditional knockouts in non-intestinal tissues show milder effects, such as delayed cycling leading to progressive and thinned pelage, impaired segment specification in embryos, and reduced gastric regeneration upon challenge, underscoring LGR5's non-redundant role in progenitor dynamics.

Clinical Relevance

Regenerative Medicine Applications

LGR5-positive (LGR5+) cells serve as foundational elements in cultures, enabling the generation of complex, self-organizing tissue structures that mimic native organ architecture. In intestinal protocols, single Lgr5+ cells can clonally expand to form crypt-villus structures, recapitulating epithelial turnover and under defined growth factors like R-spondin-1, EGF, and Noggin. Similarly, for gastric organoids, isolated Lgr5+ cells from the pyloric generate long-lived structures resembling mature antral , demonstrating multipotency and self-renewal in a three-dimensional matrix. Hepatic organoids derived from Lgr5+ biliary cells, activated during injury, propagate as ductal cysts that differentiate into hepatocyte-like cells, providing a platform for modeling . In embryonic development, LGR5 plays a critical role in gut formation by modulating Wnt signaling to regulate fate, with its deficiency leading to deregulated differentiation in the . Lgr5 mice exhibit normal embryonic gut but display severe perinatal defects, including gastrointestinal dilation and an inability to suckle, resulting in 100% neonatal lethality. Although direct evidence for LGR5 in mammalian limb embryogenesis is limited, its interaction with R-spondin enhances Wnt activity in limb bud progenitors during developmental patterning in model organisms. LGR5 targeting holds promise for in regenerative contexts, particularly for expanding populations to treat . In liver models, combined administration of hepatocyte growth factor and R-spondin-1 induces Lgr5+ , reducing and restoring hepatic function without tumorigenic risks. Activated hepatic stellate in livers further promote LGR5+ expansion, suggesting therapeutic strategies to harness these interactions for remodeling. Post-2023 advances have integrated editing with LGR5+ stem cells to advance personalized regenerative approaches. For instance, -generated Lgr5 reporter models facilitate studies of LGR5+ cell contributions to tissue repair, enabling tailored interventions for organ-specific regeneration. These edited organoids also support patient-derived modeling for drug screening, enhancing precision in treating developmental and injury-related disorders.

Cancer Associations

LGR5 serves as a prominent marker for cancer stem cells (CSCs) across multiple malignancies, particularly in colorectal, gastric, and liver cancers, where its overexpression drives tumor progression and metastasis. In colorectal cancer, elevated LGR5 expression correlates with lymph node metastasis, larger tumor size, and advanced disease stages, facilitating cancer cell migration and invasion through enhanced Wnt/β-catenin signaling. Similarly, in gastric cancer, LGR5+ cells promote dedifferentiation and distant metastasis, with high expression observed in progressive lesions. In hepatocellular carcinoma (HCC), LGR5 marks liver CSCs that exhibit increased proliferation and clonogenicity, contributing to metastatic spread via similar pathway activation. Recent studies from 2024 and 2025 highlight the role of LGR5+ cells in chemoresistance; for instance, in gastric cancer, LGR5 co-expression with CD44 identifies CSCs resistant to cisplatin and oxaliplatin by activating Wnt, Hedgehog, and Notch pathways, while in colorectal and liver cancers, LGR5+ populations evade 5-fluorouracil and doxorubicin through sustained stemness. LGR5 exhibits a dual function in tumorigenesis, promoting cancer development via Wnt hyperactivation in many contexts while exerting tumor-suppressive effects upon its loss in others. High LGR5 levels amplify Wnt signaling to foster CSC maintenance and tumor growth in colorectal and gastric cancers, yet its knockdown in colorectal cell lines upregulates Wnt target genes like WISP1 and Wnt5a, enhancing and anchorage-independent growth. In intestinal adenomas, LGR5 acts as a negative regulator of tumourigenicity by antagonizing excessive Wnt activity; its selective loss in regions of hyperactivated Wnt signaling facilitates and progression to , as evidenced by reduced LGR5 positivity in invasive areas compared to adenomas. High LGR5 mRNA expression serves as a prognostic indicator of poor outcomes, particularly in early-stage gastric cancer. In stage I/II gastric cancer patients, elevated LGR5 levels are associated with significantly worse overall survival (5-year rate: 60.6% vs. 100% in LGR5-negative cases) and , independent of other clinicopathological factors in multivariate analysis. A 2025 meta-analysis in further confirms that LGR5 overexpression correlates with reduced overall survival, underscoring its value in risk stratification. Therapeutic strategies targeting LGR5 have shown promise in overcoming CSC-driven resistance, including antibody-drug conjugates (ADCs) and combinations with inhibitors. LGR5-directed ADCs, such as those using the 8E11 conjugated to payloads like CPT2, effectively shrink tumors in patient-derived xenografts by depleting LGR5+ CSCs without severe off-target toxicity to normal stem cells. In 2025 preclinical studies, combining these ADCs with — an —upregulates LGR5 expression in both wild-type and mutant models, leading to enhanced tumor regression and prolonged survival compared to monotherapy, with complete responses observed in resistant PDX lines. Ongoing clinical trials include a phase I/II study of the LGR5-targeted CAR-T therapy CNA3103 for metastatic , and bispecific antibodies like petosemtamab ( x LGR5) in early-phase testing for gastrointestinal cancers. Additionally, inhibiting the LGR5/mTORC2 axis addresses metabolic plasticity in HCC; LGR5 activates mTORC2 via RAC1/AKT/FOXO3a to boost aerobic , enabling resistance to glucose , and mTORC2 blockade with agents like metformin suppresses this adaptation, reducing tumor growth and . Recent 2024-2025 investigations reveal LGR5's contributions to interactions, including glioma stiffness, (ECM) adhesion, and metabolic resilience. In brain cancers like , LGR5+ CSCs contribute to tumor progression and invasion through mechanisms such as activation. Furthermore, LGR5 drives glucose starvation resistance in cancer cells by enhancing glycolytic capacity through mTORC2, a mechanism particularly relevant in nutrient-poor tumor niches.

References

  1. [1]
    LGR5 leucine rich repeat containing G protein-coupled receptor 5 ...
    Aug 19, 2025 · LGR5 is the most selectively expressed gene in human zona glomerulosa and reduces aldosterone production and cell number. H pylori colonize and ...Gene
  2. [2]
    R-spondins function as ligands of the orphan receptors LGR4 and ...
    Jun 21, 2011 · Here we demonstrate that LGR4 and LGR5 bind the R-spondins with high affinity and mediate the potentiation of Wnt/β-catenin signaling by enhancing Wnt-induced ...
  3. [3]
    Wnt Signaling, Lgr5, and Stem Cells in the Intestine and Skin - PMC
    The Wnt target gene Lgr5 has been recently identified as a novel stem cell marker of the intestinal epithelium and the hair follicle.
  4. [4]
    LGR5 Gene - GeneCards | LGR5 Protein | LGR5 Antibody
    LGR5 encodes a receptor for R-spondins, involved in Wnt signaling, and is a stem cell marker in the intestine and hair follicle.
  5. [5]
    LGR4 and LGR5 form distinct homodimers that only LGR4 ... - NIH
    Jul 4, 2023 · LGR4 and LGR5 are two homologous receptors that potentiate Wnt/β-catenin signaling in response to R-spondin (RSPO) ligands.
  6. [6]
    The R-spondin/Lgr5/Rnf43 module: regulator of Wnt signal strength
    The Lgr5/R-spondin complex acts by neutralizing Rnf43 and Znrf3, two transmembrane E3 ligases that remove Wnt receptors from the stem cell surface.
  7. [7]
    LGR5: An emerging therapeutic target for cancer metastasis and ...
    Jan 17, 2025 · LGR5 has been identified as a stem cell-specific receptor to promote the canonical WNT/β-catenin signaling pathway, in cooperation with WNT ...Lgr5: An Emerging... · 2 Lgr5 Structure And... · 2.2 The Role Of Lgr5 In The...
  8. [8]
    functional identification of Lgr5 in cancer progression and potential ...
    Jul 29, 2019 · Leucine-rich repeat-containing G protein-coupled receptor 5 (Lgr5) has been recently discovered as a candidate marker of cancer stem cell populations.
  9. [9]
    Identification and cloning of an orphan G protein-coupled receptor of the glycoprotein hormone receptor subfamily - PubMed
    - **Title**: Identification and cloning of an orphan G protein-coupled receptor of the glycoprotein hormone receptor subfamily
  10. [10]
  11. [11]
    o75473 · lgr5_human - UniProt
    LGR5 is used as a marker of adult tissue stem cells in the intestine, stomach, hair follicle, and mammary epithelium.
  12. [12]
    Identification of stem cells in small intestine and colon by marker ...
    Oct 25, 2007 · The Lgr5-positive crypt base columnar cell generated all epithelial lineages over a 60-day period, suggesting that it represents the stem cell of the small ...Missing: 9642114 | Show results with:9642114
  13. [13]
    Structure and function of LGR5: An enigmatic G-protein coupled ...
    This review discusses the discovery of LGR5 and the impact it is having on our understanding of stem cell and cancer biology of the colon.Missing: paper | Show results with:paper
  14. [14]
    Characterization of Two LGR Genes Homologous to Gonadotropin ...
    These models are based on the crystal structure of the porcine ribonuclease-ribonuclease inhibitor complex in which the repeats of 28 or 29 residues each ...Results · Isolation Of Lgr4 And Lgr5... · Materials And Methods<|control11|><|separator|>
  15. [15]
    Mouse Lgr5/GPR49 PE-conjugated Antibody FAB8240P-100UG
    $$45 deliveryThe region of the mouse Lgr5 long extracellular domain used as an immunogen shares 90% and 95% amino acid sequence identity with human and rat Lgr5, ...
  16. [16]
    The R-spondin/Lgr5/Rnf43 module: regulator of Wnt signal strength
    Lgr5 was originally discovered as a common Wnt target gene in adult intestinal crypts and colon cancer. It was subsequently identified as an exquisite ...Missing: paper | Show results with:paper
  17. [17]
    Unlike LGR4, LGR5 potentiates Wnt–β-catenin signaling without ...
    Dec 1, 2020 · It is thought that LGR5 stimulates Wnt signaling similarly to LGR4 although LGR4 and LGR5 are not functionally equivalent in vivo. By analyzing ...Unlike Lgr4, Lgr5... · Lgr5 Potentiates... · Full-Length Lgr5 Does Not...
  18. [18]
    LGR5 Interacts and Cointernalizes with Wnt Receptors To Modulate ...
    It is most homologous to LGR4 and LGR6, with ∼50% amino acid identity between each other (14, 25). ... Plasmids encoding human Myc-LGR5 (amino acids [aa] 21 to ...
  19. [19]
    Wnt/β-catenin signalling: function, biological mechanisms ... - Nature
    Jan 3, 2022 · The Wnt/β-catenin pathway comprises a family of proteins that play critical roles in embryonic development and adult tissue homeostasis.
  20. [20]
    LGR4 and LGR5 are R‐spondin receptors mediating Wnt/β‐catenin ...
    Here we show that LGR4 and LGR5 function as R‐spondin receptors in Wnt/β‐catenin and Wnt/PCP signalling.
  21. [21]
    LGR5 deficiency deregulates Wnt signaling and leads to precocious ...
    Jul 1, 2009 · In vivo, LGR5 deficiency results in complete neonatal lethality as a consequence of a tongue developmental defect interfering with suckling ( ...
  22. [22]
    Neonatal Lethality of LGR5 Null Mice Is Associated with ... - NIH
    LGR5 null mice exhibited 100% neonatal lethality characterized by gastrointestinal tract dilation with air and an absence of milk in the stomach.
  23. [23]
    Identification of stem cells in small intestine and colon by marker ...
    Oct 14, 2007 · The Lgr5-positive crypt base columnar cell generated all epithelial lineages over a 60-day period, suggesting that it represents the stem cell of the small ...Lgr5 Is Expressed In Crypt... · Lgr5 Cbc Cells Are Distinct... · Lgr5 Cells Are The Stem...
  24. [24]
    Regenerative Intestinal Stem Cells Induced by Acute and Chronic ...
    The intestinal epithelium is replenished every 3–4 days by rapidly dividing Lgr5+ intestinal stem cells (ISCs) in the crypt base that differentiate as they move ...Introduction · Intestinal Epithelial Cells—The... · The Role of +4 Position Cells...
  25. [25]
    Paneth cells constitute the niche for Lgr5 stem cells in intestinal crypts
    We conclude that Lgr5 stem cells compete for essential niche signals provided by a specialized daughter cell, the Paneth cell.
  26. [26]
    The intestinal stem cell markers Bmi1 and Lgr5 identify two ... - PNAS
    N Barker, et al., Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007). Crossref · PubMed · Google ...The Intestinal Stem Cell... · Results · Bmi1 Marks Quiescent Iscs...<|control11|><|separator|>
  27. [27]
    Lgr5+ Stem Cells Are Indispensable for Radiation-Induced Intestinal ...
    Feb 6, 2014 · Lgr5 + cells are crucial for robust intestinal regeneration following radiation exposure but are dispensable for premalignant hyperproliferation.
  28. [28]
    Regulation and plasticity of intestinal stem cells during homeostasis ...
    Oct 15, 2016 · The intestinal epithelium is the fastest renewing tissue in mammals and has a large flexibility to adapt to different types of damage. Lgr5+ ...Wnt · Progenitor Cell Plasticity · Dedifferentiation Of...<|separator|>
  29. [29]
    The cellular niche for intestinal stem cells: a team effort
    Jan 1, 2021 · Paneth cells not only directly interact with ISCs, they also secrete Wnt3, EGF, and surface-bound Dll4 that respectively activate EGFR/Ras/MAPK, ...
  30. [30]
    Adult mammalian stem cells: the role of Wnt, Lgr5 and R‐spondins
    May 22, 2012 · Recent studies have indicated that Wnt agonists of the R‐spondin family provide potent growth stimuli for crypts in vivo and in vitro.Wnt In Intestinal Crypt... · Intestinal Organoids · Wnt Signalling In Stomach...
  31. [31]
    R-spondin signalling is essential for the maintenance and ... - eLife
    Apr 23, 2020 · Lgr5 was found to be virtually absent from uninduced progenitors, but was strongly expressed within the ureteric tip and the distal part of the ...
  32. [32]
    Wnt-Responsive Lgr5-Expressing Stem Cells Are Hair Cell ...
    Jul 11, 2012 · We show here that Lgr5, a marker for adult stem cells, was expressed in a subset of supporting cells in the newborn and adult murine cochlea.
  33. [33]
    Tissue expression of LGR5 - Summary - The Human Protein Atlas
    The value ranges from 0 and 1, where 0 indicates identical expression across all cells/tissue types, while 1 indicates expression in a single cell/tissue type. ...
  34. [34]
    A conditional transgenic mouse line for targeted expression of the ...
    Aug 15, 2015 · More recently, Lgr5 was shown to be a stem cell marker in multiple tissues, including stomach, small intestine, colon, hair follicle, mammary ...
  35. [35]
    Single Lgr5 stem cells build crypt-villus structures in vitro ... - PubMed
    May 14, 2009 · We conclude that intestinal cryptvillus units are self-organizing structures, which can be built from a single stem cell in the absence of a non-epithelial ...
  36. [36]
    Article Lgr5 +ve Stem Cells Drive Self-Renewal in the Stomach and ...
    Jan 8, 2010 · We have recently described a small population of cycling Lgr5+ve stem cells at the base of these intestinal crypts (Barker et al., 2007). In ...
  37. [37]
    In vitro expansion of single Lgr5+ liver stem cells induced by ... - PMC
    Aug 14, 2013 · Single Lgr5+ cells from damaged liver can be clonally expanded as organoids in Rspo1-based culture medium over multiple months. Such clonal ...
  38. [38]
    Spatio-Temporal Expression Profile of Stem Cell-Associated Gene ...
    The expression of LGR5 is induced in the limb, tail, and intestine by T3 during metamorphosis. More importantly, LGR5 mRNA is localized to the developing adult ...
  39. [39]
    HGF/R-spondin1 rescues liver dysfunction through the induction of ...
    Oct 27, 2017 · Injection of HGF in combination with Rspo1 increases the number of Lgr5 + liver stem cells and improves liver function by attenuating fibrosis.
  40. [40]
    (PDF) Activated hepatic stellate cells promote expansion of LGR5 ...
    Mar 9, 2023 · Results LGR5 mRNA were significantly enhanced in fibrotic liver tissue and LGR5+ cell numbers positively correlated with the stage of fibrosis.
  41. [41]
    Frontiers | A gene edited pig model for studying LGR5+ stem cells
    Jun 5, 2024 · This paper discusses the potential utility of the LGR5 + pig model in exploring the role of LGR5 + cells in tissue development and regeneration.