RSPO1 regulates development, stem cell maintenance, and tissue repair through canonical and non-canonical Wnt pathways:
Ovarian Development: Upregulated in human ovaries (6–9 weeks post-conception) to suppress testis formation via β-catenin stabilization . Mutations in RSPO1 cause XX sex reversal and palmoplantar hyperkeratosis .
Intestinal Homeostasis: Induces crypt cell proliferation and protects against chemotherapy-induced colitis in mice .
RSPO1 modulates Wnt signaling by:
Potentiating LRP6/RNF43/ZNRF3: Binds to LGR4/5 receptors, triggering internalization of RNF43/ZNRF3 to reduce Wnt ligand degradation .
Stabilizing β-Catenin: Synergizes with Wnt3a to increase nuclear β-catenin levels (ED₅₀: 50–200 ng/ml) .
Wnt-Independent Pathways: May regulate TGFβ signaling and inflammatory responses .
Recombinant human RSPO1 (rhRSPO1) enhances intestinal organoid unit (OU) survival and tissue-engineered small intestine (TESI) formation:
Parameter | Control (PBS) | rhRSPO1-Treated | Fold Increase |
---|---|---|---|
OU size (Day 6) | Baseline | 5.7× larger | 5.7× |
Survival (Days 3–6) | 1× | 2× | 2× |
TESI engraftment | 66.6% | 100% | 1.5× |
In astrocytes, RSPO1 suppresses pro-inflammatory cytokines (e.g., IL-10, IFN-γ) while increasing TNF-α and CXCL12, suggesting therapeutic potential in neuroinflammation .
Critical Site: Asn137 glycosylation is essential for RSPO1 secretion. Mutants (N137Q) show reduced extracellular accumulation .
Stability: N-glycans enhance intracellular stability but minimally affect secreted protein half-life .
Skin Tumors: RSPO1 mutations predispose to squamous cell carcinoma .
Ovarian Development: RSPO1 overexpression in ovaries opposes testis formation via β-catenin .
R-Spondin-1 (Rspo1), a member of the Rspondin family, functions as an activator of the canonical Wnt signaling pathway by acting as a ligand for LGR4-6 receptors. Rspo1 plays a crucial role in initiating crypt cell proliferation and enhancing intestinal epithelial healing. It also negatively regulates the TGF-beta pathway.
Recombinant Human R-Spondin-1, expressed in CHO cells, is a glycosylated monomeric chain comprising 243 amino acids, resulting in a total molecular mass of 25.6kDa.
The purification of RSPO1 is carried out using proprietary chromatographic techniques.
The protein solution undergoes sterile filtration (0.2µm) with PBS and subsequently lyophilized.
To reconstitute lyophilized RSPO1, it is recommended to dissolve it in sterile 18MΩ-cm H2O at a concentration of at least 100µg/ml. Further dilutions can be prepared in other aqueous solutions as needed.
Lyophilized RSPO1 demonstrates stability at room temperature for 3 weeks; however, it is advisable to store it desiccated below -18°C. Upon reconstitution, store RSPO1 at 4°C for 2-7 days. For long-term storage, it should be kept below -18°C. It is essential to avoid repeated freeze-thaw cycles to maintain protein integrity.
The purity is determined to be greater than 95.0% as assessed by SDS-PAGE analysis.
The ED50, determined by Luciferase induction in HEK-293 STF cells in the presence of Murine Wnt-3a, is 47.99ng/ml, corresponding to a specific activity of 2.1 x 10^4 units/mg.
R-spondin-1, Cristin-3, mCristin-3, Roof plate-specific spondin-1, Rspo1, RSPO, R-spondin 1, R-spondin, Rspondin, CRISTIN3.
CHO cells.
SRGIKGKRQR RISAEGSQAC AKGCELCSEV NGCLKCSPKL FILLERNDIR QVGVCLPSCP PGYFDARNPD MNKCIKCKIE HCEACFSHNF CTKCKEGLYL HKGRCYPACP EGSSAANGTM ECSSPAQCEM SEWSPWGPCS KKQQLCGFRR GSEERTRRVL HAPVGDHAAC SDTKETRRCT VRRVPCPEGQ KRRKGGQGRR ENANRNLARK ESKEAGAGSR RRKGQQQQQQ QGTVGPLTSA GPA.
RSPO1 is a secreted protein containing two furin-like (FU) repeats and one TSP type-1 domain, belonging to the R-spondin family . It functions as a tissue-specific amplifier of β-catenin signaling, particularly in opposing testis formation during development . The protein acts synergistically with WNT4 to induce cell proliferation by stabilizing β-catenin, which then activates downstream targets . This interaction is crucial in various developmental processes and tissue homeostasis.
RSPO1 exhibits strong nuclear localization in several cell lines, suggesting additional intracellular functions beyond its established role in extracellular signaling . The protein undergoes complex glycosylation patterns that are essential for its proper folding, activity, stability, and maturation, highlighting the importance of using appropriate expression systems for recombinant protein production .
RSPO1 participates in multiple physiological processes, including gonadal development, bone formation, skeletal muscle development, pancreatic β-cell proliferation, and intestinal stem cell regulation. Recent research has also identified its role in adipose tissue metabolism and association with obesity risk .
RSPO1 exhibits a distinct sexually dimorphic expression pattern during critical stages of early human gonadal development. Between 6-9 weeks post conception, RSPO1 is significantly upregulated in the developing ovary but not in the testis . This differential expression is crucial for sex determination and appropriate gonadal development.
In contrast to RSPO1's differential expression, related genes such as WNT4 and CTNNB1 (encoding β-catenin) do not show significant expression differences between ovary and testis during this developmental window . This suggests that RSPO1 serves as a specific regulator in the female developmental pathway.
The importance of proper RSPO1 expression is evidenced by cases of reduced R-spondin1 function, such as in an individual with ovotestis (46,XX) carrying a RSPO1 mutation. In such cases, reduced RSPO1 function leads to decreased β-catenin protein and WNT4 mRNA levels, consistent with downregulation of ovarian developmental pathways . These findings demonstrate that RSPO1 is required for normal ovarian development and functions upstream of WNT4 in the female sex determination pathway.
In adipose tissue, RSPO1 inhibits adipocyte mitochondrial respiration and thermogenesis via the LGR4–Wnt/β-catenin signaling pathway . This mechanism appears central to how RSPO1 mutations can contribute to obesity. The p.R219W mutation, for example, disrupts RSPO1's electrostatic interaction with the extracellular matrix, leading to excessive RSPO1 release that inappropriately activates LGR4–Wnt/β-catenin signaling .
The relationship between RSPO1 and WNT4 involves a feed-forward mechanism, where RSPO1 enhances WNT4 expression while WNT4 activates the signaling that RSPO1 amplifies . This reciprocal interaction creates a robust regulatory network essential for processes like female sex determination and tissue-specific development.
RSPO1 displays tissue-specific expression patterns that correlate with its diverse functions. During early development, it is required for the formation of gonads regardless of sex and has been detected in mice only eleven days after fertilization . In humans, RSPO1 is prominently upregulated in the developing ovary between 6-9 weeks post conception but downregulated in the developing testis during the same period .
In adult tissues, RSPO1 is predominantly expressed in visceral adipose tissue, particularly within the stromal vascular fraction (SVF) and more specifically in fibroblast cell clusters . Its expression in adipose tissue increases with adiposity, as evidenced by higher levels in mouse models of diet-induced obesity and leptin-deficient (ob/ob) obesity, as well as in visceral adipose tissue from human subjects with obesity compared to lean controls .
Interestingly, while RSPO1 is detectable in human plasma at low concentrations (pg/mL range), circulating levels do not appear to differ significantly between individuals with obesity and lean controls . This suggests that RSPO1 primarily exerts its effects through local, paracrine mechanisms rather than systemic endocrine actions.
RSPO1 plays a critical role in female sex determination and ovarian development. Human testis development begins around 42 days post conception with SRY expression, followed by testis-specific gene upregulation and morphological changes . In contrast, ovarian development relies heavily on RSPO1, which is upregulated in the developing ovary between 6-9 weeks post conception .
In female development, RSPO1 augments the WNT/β-catenin pathway to oppose testis formation . It acts synergistically with WNT4 to induce cell proliferation by stabilizing β-catenin, which activates downstream targets necessary for ovarian differentiation . This synergistic action is essential for normal ovarian development.
The significance of RSPO1 in sex determination is evidenced by cases of RSPO1 mutations, which can lead to disorders of sex development. In 46,XX individuals with RSPO1 mutations, reduced R-spondin1 function results in decreased β-catenin protein and WNT4 mRNA levels, indicating disruption of ovarian developmental pathways . These cases demonstrate that RSPO1 functions upstream of WNT4 in the female sex determination pathway, serving as a critical regulator of ovarian development.
Production of recombinant human RSPO1 (rhRSPO1) requires careful consideration of expression systems, purification methods, and quality control measures to ensure biological activity. HEK293 (Human Embryonic Kidney) cells represent an optimal expression system due to their ability to perform complex post-translational modifications, particularly glycosylation patterns with sialic acid additions, which are essential for maintaining protein structure and function .
The production process typically begins with the synthesis and subcloning of the human RSPO1 coding sequence into a mammalian expression vector, followed by transfection into HEK293 cells and selection of stable producer clones . When screening for high-producing clones using methods like Dot Blot and ELISA quantification, research has shown that culture conditions significantly impact productivity. Cells cultured in serum-containing medium can yield volumetric productivity of 1.25-1.94 μg/mL, somewhat higher than serum-free conditions which produce 0.93-1.21 μg/mL .
Purification strategies often leverage affinity tags such as histidine tags, enabling initial capture via immobilized metal affinity chromatography . Subsequent purification steps may include ion-exchange and size-exclusion chromatography to achieve high purity, typically >95% as analyzed by SDS-PAGE . Quality control should include analysis under both reducing and non-reducing conditions to verify proper disulfide bond formation, which is critical for RSPO1's structural integrity and function.
Characterization of the purified rhRSPO1 should confirm both identity and biological activity. Functional validation using cell-based assays, particularly the β-catenin responsive TOPFLASH reporter system, can verify that the recombinant protein amplifies WNT signaling as expected . Storage conditions must be optimized to maintain stability, as post-translational modifications and proper folding are essential for RSPO1's biological activity.
RSPO1 mutations, particularly gain-of-function variants like p.R219W/Q, have emerged as significant factors in adipose tissue dysfunction and obesity. Whole-exome sequencing studies have identified that these specific mutations are associated with significantly increased obesity risk, with odds ratios of 4.65 (95% CI, 2.02–10.34; P = 1.63 × 10⁻⁴) in study cohorts and even higher in East Asian populations from the gnomAD database .
Mechanistically, the p.R219W mutation disrupts RSPO1's electrostatic interaction with the extracellular matrix, leading to excessive protein release into the extracellular space . This inappropriate release results in amplified WNT signaling in target cells, particularly preadipocytes, via the LGR4 receptor . The enhanced WNT/β-catenin signaling subsequently inhibits the thermogenic capacity of beige adipocytes.
Experimental evidence from humanized knockin mice carrying the p.R219W mutation demonstrates that these animals develop suppressed thermogenesis and increased adiposity when challenged with a high-fat diet, recapitulating the phenotype observed in human mutation carriers . Similarly, transgenic mice overexpressing human RSPO1 in adipose tissues gain more body weight on high-fat diet without changes in food intake or physical activity, but with significantly decreased oxygen consumption and carbon dioxide production .
The metabolic consequences include reduced brown/beige fat thermogenesis, leading to decreased energy expenditure and increased susceptibility to diet-induced obesity . Conversely, Rspo1 ablation in mice protects against high-fat diet-induced adiposity by enhancing thermogenesis . These findings collectively identify RSPO1 as both a potential diagnostic marker for obesity risk and a therapeutic target for interventions aimed at enhancing adipose tissue energy expenditure.
Investigating RSPO1 function requires carefully selected experimental models that recapitulate relevant aspects of its biology in different contexts. Several complementary systems have proven valuable:
For cellular studies, HEK293 cells serve as an effective platform for both recombinant RSPO1 production and basic signaling assays . These cells provide appropriate post-translational modifications and can be used with reporter systems like TOPFLASH to assess β-catenin signaling activation . Primary human adipocytes and preadipocytes offer valuable insights into RSPO1's effects on adipogenesis and thermogenic capacity in a physiologically relevant context .
Transgenic mouse models provide crucial in vivo systems for studying RSPO1's developmental and metabolic functions. These include:
Adipose-specific human RSPO1 overexpression models using promoters like aP2, which demonstrate increased susceptibility to diet-induced obesity and suppressed thermogenesis
RSPO1 knockout mice, which exhibit resistance to diet-induced obesity due to enhanced thermogenic capacity
Humanized knockin mice carrying specific mutations such as p.R219W, which recapitulate the phenotypes observed in human mutation carriers
When selecting an experimental model, researchers should consider the specific aspect of RSPO1 biology under investigation. For signaling mechanisms, cell culture systems with appropriate reporters may suffice . For developmental questions, animal models or fetal tissues are necessary . For metabolic functions, both cellular systems for mechanistic studies and animal models for in vivo validation are recommended .
RSPO1 expression exhibits complex regulatory patterns that vary by tissue, developmental stage, and pathophysiological context. During human gonadal development, RSPO1 shows striking sexual dimorphism, being upregulated in the developing ovary but not in the testis between 6-9 weeks post conception . This pattern is crucial for sex determination, but the upstream factors controlling this differential expression remain incompletely characterized.
In adipose tissue, RSPO1 expression increases with adiposity. Studies have demonstrated elevated Rspo1 expression in the epididymal white adipose tissue (eWAT) of both diet-induced obese mice and leptin-deficient (ob/ob) obese mice . Similarly, human subjects with obesity show higher RSPO1 expression in visceral adipose tissue compared to lean controls . This suggests that RSPO1 expression responds to nutritional status and metabolic state.
At the cellular level within adipose tissue, RSPO1 is predominantly expressed in the stromal vascular fraction (SVF), particularly in fibroblast clusters, rather than in mature adipocytes . This cell-type specificity indicates targeted regulation within tissue microenvironments.
The relationship between RSPO1 and WNT4 involves a regulatory feedback loop. Reduced RSPO1 function leads to decreased WNT4 mRNA levels, suggesting that RSPO1 positively regulates WNT4 expression . Conversely, WNT signaling may influence RSPO1 expression, creating a feed-forward mechanism that reinforces pathway activation in appropriate contexts.
Interestingly, while local tissue RSPO1 levels correlate with obesity, circulating concentrations do not differ significantly between individuals with obesity and lean controls . This suggests that RSPO1 regulation occurs primarily at the local tissue level rather than systemically, emphasizing its role in paracrine rather than endocrine signaling.
RSPO1's diverse roles in development, tissue regulation, and pathological processes offer multiple avenues for therapeutic targeting. Given its mitogenic activity in stem cells, recombinant human RSPO1 has significant potential in regenerative medicine applications . The protein could potentially enhance intestinal stem cell proliferation for treating gastrointestinal injuries or inflammatory conditions, and support ex vivo expansion of various stem cell populations for transplantation therapies.
For mucositis, an inflammatory condition of the oral cavity often associated with chemotherapy and radiation in cancer patients with head and neck tumors, RSPO1 represents a promising treatment candidate . Its ability to stimulate epithelial regeneration could accelerate healing of damaged mucosa, potentially reducing treatment interruptions and improving quality of life for cancer patients.
Conversely, for metabolic conditions associated with RSPO1 gain-of-function, such as obesity linked to p.R219W/Q mutations, therapeutic strategies would aim to inhibit excessive RSPO1 signaling . Approaches might include:
Developing antagonists that block RSPO1-LGR4 interaction
Creating decoy receptors that capture excess RSPO1
Designing small molecules that intervene in downstream WNT/β-catenin signaling in adipocytes
Directly targeting thermogenic pathways in brown/beige adipocytes to counteract RSPO1's suppressive effects
Production of therapeutic-grade rhRSPO1 requires mammalian expression systems, particularly HEK293 cells, which provide appropriate post-translational modifications essential for full biological activity . The purification process must yield a product with high purity (>95%) and verified biological activity, typically assessed using reporter systems like TOPFLASH .
For any therapeutic application, careful consideration must be given to RSPO1's tissue-specific effects and potential for off-target consequences. Delivery systems that enable targeted administration to specific tissues or cell types would help maximize therapeutic benefits while minimizing systemic effects.
Comprehensive analysis of RSPO1-related WNT signaling requires multiple complementary approaches that capture different aspects of this complex pathway. The TOPFLASH reporter assay represents the gold standard for quantifying β-catenin-dependent transcriptional activation . Research has demonstrated that wild-type RSPO1 alone produces modest activation of this reporter (approximately 1.8-fold), while co-transfection with CTNNB1 (β-catenin) results in dramatic synergistic augmentation (approximately 10-fold) . This assay is particularly valuable for assessing the functional impact of RSPO1 mutations, as demonstrated with p.R219W/Q variants that show enhanced activation compared to wild-type protein .
For protein-level analyses, western blotting remains essential for measuring changes in key pathway components including total and active (non-phosphorylated) β-catenin levels . Subcellular fractionation followed by western blotting can assess β-catenin nuclear translocation, a critical event in pathway activation. Co-immunoprecipitation studies can reveal physical interactions between RSPO1 and its receptors or other signaling components.
Transcriptional analyses provide crucial insights into downstream effects of RSPO1-mediated signaling. Quantitative RT-PCR can measure expression changes in established WNT target genes, while RNA-sequencing offers genome-wide transcriptional profiling to identify all genes affected by RSPO1 signaling, enabling pathway enrichment analysis .
Functional assessments of RSPO1 effects should include:
Cellular proliferation and differentiation assays
Metabolic measurements such as oxygen consumption rate and extracellular acidification rate in relevant cell types like adipocytes
Thermogenic capacity assessment in adipocytes using appropriate stimuli and readouts
For in vivo studies, genetically modified mouse models provide powerful systems for analyzing physiological consequences of altered RSPO1 signaling . These include both tissue-specific overexpression models and knockout/knockin approaches that can reveal the integrated effects of RSPO1 on whole-body physiology and metabolism.
RSPO1 mutations have been implicated in multiple human disease states, with disorders of sex development and obesity being the most thoroughly characterized. Mutations affecting RSPO1 function can disrupt the WNT/β-catenin signaling necessary for proper ovarian development, leading to disorders of sex development in 46,XX individuals . In these cases, reduced RSPO1 function results in decreased β-catenin protein and WNT4 mRNA levels, demonstrating disruption of normal ovarian developmental pathways .
More recently, gain-of-function mutations in RSPO1, particularly p.R219W/Q, have been identified as significant contributors to obesity risk. Whole-exome sequencing of young, severely obese individuals identified that these specific mutations confer substantially increased odds ratios for obesity: 4.65 (95% CI, 2.02–10.34) in study cohorts and up to 20.21 (95% CI, 5.45–111.99) in East Asian populations . Functional testing using the TOPFLASH reporter system confirmed that these mutations increase WNT signaling activation compared to wild-type RSPO1 .
The p.R219W mutation specifically disrupts RSPO1's electrostatic interaction with the extracellular matrix, leading to excessive protein release . This inappropriate release amplifies WNT signaling in target cells, particularly preadipocytes, via the LGR4 receptor . The enhanced WNT/β-catenin signaling subsequently inhibits the thermogenic capacity of beige adipocytes, reducing energy expenditure and promoting fat accumulation .
Humanized knockin mice carrying the p.R219W mutation recapitulate the human phenotype, displaying suppressed thermogenesis and increased susceptibility to diet-induced obesity . This experimental validation confirms the causal relationship between the mutation and metabolic dysfunction.
These findings collectively identify RSPO1 mutations as novel pathogenic factors in human disease, with implications for both diagnosis and treatment. For obesity associated with RSPO1 gain-of-function mutations, therapeutic strategies targeting the RSPO1-LGR4-WNT/β-catenin axis might offer personalized approaches to enhance energy expenditure and reduce adiposity.
Post-translational modifications (PTMs) critically influence RSPO1 structure, stability, and signaling capacity. Glycosylation represents one of the most important modifications affecting RSPO1 function. The complexity of these glycosylation patterns, particularly the addition of sialic acids, is a key reason why mammalian expression systems like HEK293 cells are preferred for recombinant RSPO1 production . These cells can perform the appropriate glycosylation necessary for maintaining protein folding, activity, stability, and maturation .
Disulfide bond formation represents another crucial modification essential for RSPO1's structural integrity. When analyzed by SDS-PAGE, RSPO1 shows different migration patterns under reducing versus non-reducing conditions, indicating the presence of intramolecular disulfide bonds . These bonds are particularly important in the furin-like repeats, which contain conserved cysteine residues that form disulfide bridges necessary for proper protein folding and receptor interaction.
RSPO1 exhibits strong nuclear localization in several different cell lines , suggesting additional PTMs that regulate intracellular trafficking. This nuclear localization implies functions beyond the established role in potentiating extracellular WNT signaling, possibly including direct involvement in transcriptional processes.
The p.R219W mutation, associated with obesity risk, specifically disrupts RSPO1's electrostatic interaction with the extracellular matrix . This alteration could be considered a form of post-translational dysregulation, as it affects how the protein interacts with its environment after secretion. The mutation leads to excessive RSPO1 release into the extracellular space, resulting in inappropriate amplification of WNT signaling .
For researchers producing recombinant RSPO1, understanding these modifications is crucial for ensuring biological activity. Expression system selection, purification methods, and storage conditions all need to be optimized to maintain appropriate PTMs. When comparing RSPO1 from different sources or preparation methods, potential differences in PTMs should be considered as possible explanations for variations in activity or experimental outcomes.
R-Spondin-1 is a glycosylated protein with a molecular weight of approximately 40 kDa . It acts as a major agonist of the Wnt signaling pathway by regulating the turnover of the LRP6 co-receptor, preventing its internalization by DKK-1 . This regulation enhances Wnt/β-catenin signaling, which is essential for the maintenance and proliferation of stem cells .
Recombinant human R-Spondin-1 (rhRSPO1) is typically produced using mammalian cell expression systems, such as HEK293 cells . The production process involves the synthesis and subcloning of the hRSPO1 coding sequence into a mammalian cell expression vector, followed by stable transfection of the cells . The protein is then purified using chromatographic techniques, ensuring high purity and biological activity .
R-Spondin-1 has several important applications in biomedical research and regenerative medicine. It is used to potentiate Wnt signaling in various organoid culture systems, including intestinal and tumor organoids . Additionally, RSPO1 is crucial for hematopoietic stem cell specification and cancer cell migration and survival . Its mitogenic activity in stem cells makes it a valuable tool in cell therapy and tissue engineering .