Phospho-STMN1 (S62) Antibody

Shipped with Ice Packs
In Stock

Description

Target Protein Overview

STMN1 (Stathmin 1) is encoded by the STMN1 gene (UniProt ID: STMN1_HUMAN, Gene ID: 3925) and plays a critical role in cytoskeletal regulation by destabilizing microtubules, impacting cell division, signaling, and neuronal development . Phosphorylation at specific residues, including S62, modulates its activity. Aberrant STMN1 expression or phosphorylation is linked to cancers, acute leukemia, and neurological disorders .

Mechanism of Phosphorylation Recognition

The antibody’s specificity for phospho-S62 arises from interactions between its complementarity-determining regions (CDRs) and the phosphorylated epitope:

  • Basic residues (e.g., arginine) and uncharged polar residues (e.g., serine/threonine) in the antibody’s CDRs form hydrogen bonds with the phosphate group .

  • Mutational studies on similar antibodies show that alanine substitutions in these residues reduce binding affinity by up to 10-fold, highlighting their critical role .

Functional Implications of S62 Phosphorylation

Phosphorylation at S62 influences STMN1’s microtubule-destabilizing activity:

  • Tubulin Binding: Phosphorylation at nearby residues (e.g., S63) reduces tubulin binding, but S62 phosphorylation may regulate downstream signaling cascades .

  • Cellular Roles: STMN1 phosphorylation is implicated in neurogenesis, fear response, and cancer progression .

Key Findings

  • Western Blot: Detects a ~78 kDa band in rat cortex lysates, with signal abolished by preadsorption with the phospho-peptide .

  • Disease Associations: Overexpression of phosphorylated STMN1 is observed in pancreatic intraductal papillary-mucinous adenoma and leukemia .

Technical Validation

  • Cross-Reactivity: Validated in bovine, mouse, and rat models .

  • Controls: Specificity confirmed using dephosphorylated peptides and knockout cell lines .

Comparison with Related Antibodies

AntibodyTargetPhospho-SpecificApplications
Phospho-STMN1 (S62) STMN1 p-S62YesWB, IHC, IF, ELISA
Syn1 (phospho S62/S67) Syn1 p-S62/p-S67YesWB (tissue lysate)

Limitations and Considerations

  • Species Specificity: Limited reactivity in non-mammalian systems .

  • Phospho-Dependency: Cannot detect non-phosphorylated STMN1, necessitating parallel assays for total protein levels .

Product Specs

Buffer
Liquid in PBS containing 50% glycerol, 0.5% BSA, and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship products within 1-3 business days after receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery timelines.
Synonyms
C1orf215 antibody; Lag antibody; LAP 18 antibody; LAP18 antibody; Leukemia associated phosphoprotein p18 antibody; Leukemia-associated phosphoprotein p18 antibody; Metablastin antibody; Oncoprotein 18 antibody; OP 18 antibody; Op18 antibody; p18 antibody; p19 antibody; Phosphoprotein 19 antibody; Phosphoprotein p19 antibody; pp17 antibody; pp19 antibody; PR22 antibody; Pr22 protein antibody; Prosolin antibody; Protein Pr22 antibody; SMN antibody; Stathmin antibody; Stathmin1 antibody; STMN 1 antibody; Stmn1 antibody; STMN1_HUMAN antibody
Target Names
Uniprot No.

Target Background

Function
Phospho-STMN1 (S62) Antibody plays a role in regulating the microtubule (MT) filament system by destabilizing microtubules. It inhibits microtubule assembly and promotes disassembly. Phosphorylation at Ser-16 may be essential for axon formation during neurogenesis. It is also involved in the control of both learned and innate fear.
Gene References Into Functions
  1. Research indicates that autophagy activation reduces the expression of STMN1 and p53, contributing to the anti-cancer effects of Halofuginone by inhibiting cancer cell migration and invasion. These findings suggest potential avenues for breast cancer prevention and therapy. PMID: 29231257
  2. A study revealed that low STMN1 expression was observed in 43.62% of osteosarcoma cases, while high STMN1 expression was found in 56.38% of cases. High tumor expression of STMN1 was associated with a poor prognosis, poor response to chemotherapy, presence of metastases, advanced Enneking surgical stage, and the chondroblastic osteosarcoma subtype. These findings indicate that STMN1 expression serves as an independent prognostic biomarker for osteosarcoma. PMID: 30169496
  3. A transcription-independent mechanism for Stat3-mediated centrosome clustering has been reported, involving Stathmin, a Stat3 interactor that participates in microtubule depolymerization, and the mitotic kinase PLK1. PMID: 28474672
  4. Research suggests that stathmin is crucial for bipolar spindle formation to maintain genomic stability during mitosis. Depletion of stathmin prevents the initiation of chromosome instability by inducing senescence in human normal fibroblasts. PMID: 28885720
  5. Findings show that STMN1 overexpression was significantly associated with lymphatic metastatic recurrence in pN0 esophageal squamous cell carcinoma (ESCC) patients. STMN1 levels are regulated by the PI3K pathway, and STMN1 can act as a surrogate marker for PI3K pathway signaling related to tumor recurrence. PMID: 29251330
  6. The investigation confirmed that stathmin expression was correlated with more aggressive behavior in cervical cancer. PMID: 29953794
  7. High STMN1 Expression is Associated with Cancer Progression and Chemo-Resistance in Lung Squamous Cell Carcinoma. PMID: 28933054
  8. STMN1 expression was significantly associated with prognosis and tumor differentiation in ESCC, indicating that STMN1 expression is an independent prognostic factor for ESCC and could be a potential biomarker. Regulating the expression of STMN1 could influence tumor cell motility, invasion, and proliferation. PMID: 29039594
  9. T3-mediated suppression of STMN1 supports the theory that T3 plays an inhibitory role in HCC tumor growth, suggesting that the lack of normal THR function leads to elevated STMN1 expression and malignant growth. PMID: 27934948
  10. These results suggest that stathmin acts as an oncogene and is transcriptionally regulated by mutant p53 but not by wild-type p53. Stathmin could be a potential anti-tumor therapeutic target in oral squamous cell carcinoma. PMID: 28806997
  11. Results suggest that Stathmin 1 (STMN1) plays a significant role in cell proliferation and migration. PMID: 27349455
  12. STMN1 expression was higher in basal-type cell lines than in luminal-type cell lines, and overall survival and post-progression survival in the high STMN1 expression breast cancer patients were shorter than in low STMN1 expression patients. High STMN1 expression is a possible marker of breast cancer aggressiveness in association with proliferation, phenotype, and cancer stem cell type. PMID: 28766688
  13. An upregulated expression of STMN1 was found in the atypical/anaplastic meningioma group compared to the benign meningioma group. Therefore, STMN1 is a promising target for improving cure rates in meningioma cases. PMID: 28625575
  14. An increased risk of sporadic atypical meningioma recurrence can be found in cases with elevated expression of STMN1. PMID: 28622584
  15. The miR-34a/STMN1/betaIII-tubulin axis maintains the microtubule cytoskeleton in osteosarcoma, and combining miR-34a with microtubule inhibitors can be investigated as a novel therapeutic strategy. PMID: 28275089
  16. Findings suggest that Cdc2 is positively associated with the development of taxol resistance. The Cdc2 inhibitor, purvalanol A, enhanced the cytotoxic effects of taxol through Op18/stathmin. PMID: 28534969
  17. These results showed that stathmin expression was significantly upregulated in LAC, which may act as a biomarker for LAC. Furthermore, silencing stathmin inhibits LAC cell growth, suggesting that stathmin may be a promising molecular target for LAC therapy. PMID: 27494889
  18. Increased stathmin correlated with pathologic grade, lymphatic invasion, advanced stage, and poor survival of non-small cell lung cancer (NSCLC), indicating that stathmin could serve as a potential biomarker for NSCLC. PMID: 28282798
  19. Results showed that patients with cancer displayed a higher stathmin expression than those of non-cancer individuals, and overexpression of stathmin correlated with tumor cell differentiation, lymph node invasion, and high TNM stage. [review] PMID: 27806343
  20. High STMN1 Expression Is Associated with Tumor Differentiation and Metastasis in Pancreatic Cancer. PMID: 29374725
  21. miR-223 might serve as an onco-suppressor that enhances susceptibility to docetaxel by downregulating STMN1 in gallbladder cancer, highlighting its promising therapeutic value. PMID: 27577078
  22. Overexpression correlates with poorer prognosis and interacts with p53 in oral squamous cell carcinoma. PMID: 27591090
  23. A study elucidated a novel Malat1-miR-101-STMN1/RAB5A/ATG4D regulatory network where Malat1 activates autophagy and promotes cell proliferation by sponging miR-101 and upregulating STMN1, RAB5A, and ATG4D expression in glioma cells. PMID: 28834690
  24. STMN1 gene and miRNA-223 expression profiles in non-tumor liver tissues were predictive of the risk for multicentric hepatocellular carcinoma recurrence. PMID: 28982915
  25. The crucial role of FOXM1 and STMN1 in TKI-induced enrichment of CSC and drug resistance was demonstrated by knockdown of STMN1 and FOXM1 in NSCLC cells. PMID: 28850563
  26. Research demonstrates that RSK2 directly phosphorylates stathmin and regulates microtubule polymerization, providing a pro-invasive and pro-metastatic advantage to cancer cells. Therefore, the RSK2-stathmin pathway represents a promising therapeutic target and a prognostic marker for metastatic human cancers. PMID: 27041561
  27. Stathmin expression was significantly associated with shorter progression-free survival and overall survival for all analyzed cases of endometrial cancer; findings demonstrate that high stathmin expression is a poor prognostic marker in endometrial cancer. PMID: 28532857
  28. STMN1 is a possible biomarker for paclitaxel sensitivity and poor prognosis in gastric cancer (GC) and could be a novel therapeutic target in metastatic GC. PMID: 28334732
  29. STMN1, COF1, and PAIRBP1 thus represent proteins associated with proliferative and aggressive tumors of high grades, while TSP2 and POSTN were connected to low-grade tumors with a better prognosis. PMID: 28216224
  30. The phosphorylation-specific association of STMN1 with GRP78 promotes breast cancer metastasis. PMID: 27130664
  31. These results suggest that STMN1 plays a significant role in the proliferation and migration of hypopharyngeal squamous cell carcinoma and may be used as a potential prognostic biomarker or therapeutic target for hypopharyngeal squamous cell carcinoma (HSCC). PMID: 27878293
  32. High STMN1 expression is associated with invasion in endometrial carcinoma. PMID: 26815505
  33. High expression of stathmin 1 predicts a poor outcome in oral squamous cell carcinoma patients treated with docetaxel-containing regimens. PMID: 26590596
  34. The expressions of TYMS, TUBB3, and STMN1 were significantly associated with the clinicopathological characteristics of age, gender, and family history of gastric cancer, but not with differentiation, growth patterns, metastasis, and TNM staging in patients with gastric cancer. PMID: 28056823
  35. Stathmin is a highly sensitive and specific biomarker for the diagnosis of vulvar high-grade squamous intraepithelial lesions. PMID: 27226646
  36. STMN1 silencing by siRNA may enhance the sensitivity of esophageal cancer cells Eca-109 to paclitaxel and induce apoptosis. PMID: 26782519
  37. SNP in the STMN1 gene may have a potential predictive role in taxane-based chemotherapy in advanced non-small cell lung cancer. PMID: 26148901
  38. After silencing stathmin-1 in gastric cancer cells, the resistance index was reduced. PMID: 26802649
  39. Results show that the STMN1-E/P/C signature is a reliable prognostic indicator for luminal subtype breast cancer and may predict the therapeutic response to paclitaxel-based treatments, potentially facilitating individualized management. PMID: 26087399
  40. STMN1 may play a significant role in the development and tumor progression of cutaneous squamous cell carcinoma. PMID: 26235036
  41. Studies indicate that phosphorylation of stathmin controls its biological activity by reducing its affinity for tubulin, preventing microtubule disassembly. PMID: 26450904
  42. FANCC interacts and co-localizes with STMN1 at centrosomes during mitosis. Research also showed that FANCC is required for STMN1 phosphorylation. PMID: 26466335
  43. PDAC patients with higher STMN1 expression died sooner than those with lower STMN1 expression. PMID: 25791566
  44. Stathmin-1 may play a key role in regulating trophoblast invasion. PMID: 26272359
  45. These results suggest that SEPTIN2-mediated cytoskeletal rearrangement and STATHMIN-mediated differentiation may contribute to changes in cell morphology and differentiation of H/RS cells with CD99 upregulation in Hodgkin lymphoma. PMID: 26000982
  46. miR-223 regulates STMN1 in malignant pleural mesothelioma, and both are in turn regulated by the JNK signaling pathway. As such, miR-223 and STMN1 play a significant role in regulating MPM cell motility. PMID: 25824152
  47. A report indicates that STMN1 is a highly sensitive marker for leiomyosarcoma but is suboptimally specific for diagnostic purposes. PMID: 26045786
  48. MiR-101 sensitizes human nasopharyngeal carcinoma cells to radiation by targeting stathmin 1. PMID: 25607713
  49. High levels of stathmin exhibited a poor response to chemotherapy (for mRNA, P = 0.041; for protein, P = 0.017). Overexpression of stathmin was associated with shorter overall survival (for mRNA, P = 0.012) and progression-free survival. PMID: 25894372
  50. STMN1 overexpression is associated with drug resistance in esophageal squamous cell carcinoma. PMID: 25944168

Show More

Hide All

Database Links

HGNC: 6510

OMIM: 151442

KEGG: hsa:3925

STRING: 9606.ENSP00000410452

UniGene: Hs.209983

Protein Families
Stathmin family
Subcellular Location
Cytoplasm, cytoskeleton.
Tissue Specificity
Ubiquitous. Expression is strongest in fetal and adult brain, spinal cord, and cerebellum, followed by thymus, bone marrow, testis, and fetal liver. Expression is intermediate in colon, ovary, placenta, uterus, and trachea, and is readily detected at subs

Q&A

What is STMN1 and its phosphorylated form at Serine 62?

STMN1 (Stathmin 1) is a 17kDa cytoplasmic phosphoprotein that regulates microtubule dynamics through two primary mechanisms: promoting microtubule catastrophe and sequestering free tubulin heterodimers . The protein contains multiple phosphorylation sites, including Serine 62 (Ser62), which significantly affects its function. Phosphorylation at Ser62 is one of several post-translational modifications that regulate STMN1's interaction with microtubules and its subsequent biological activities . This specific phosphorylation contributes to cell cycle regulation, cell migration, and other cellular processes critical to both normal function and disease states.

What are the standard applications for Phospho-STMN1 (S62) antibodies in research?

Phospho-STMN1 (S62) antibodies are versatile tools employed across multiple experimental techniques including:

ApplicationRecommended DilutionCommon Usage Scenarios
Western Blotting (WB)1:500-1:1000Quantifying phosphorylation levels in cell/tissue lysates
Immunohistochemistry (IHC)1:50-1:100Visualizing phospho-STMN1 distribution in tissue sections
ELISA1:10000High-throughput quantification in solution

When designing experiments, researchers should validate the antibody specificity by confirming it detects STMN1 only when phosphorylated at Ser62 and not in its unphosphorylated state or when phosphorylated at other sites (Ser16, Ser25, Ser38, or Ser63) . The antibody's capacity to detect endogenous levels of phosphorylated protein makes it particularly valuable for studying physiological conditions without requiring overexpression systems.

How should Phospho-STMN1 (S62) antibodies be stored and handled to maintain optimal activity?

For maximum antibody performance and longevity, follow these evidence-based handling protocols:

  • Initial receipt: Antibodies are typically shipped at 4°C in stabilizing buffer containing phosphate-buffered saline (PBS without Mg²⁺ and Ca²⁺, pH 7.4), with 150mM NaCl, 0.02% sodium azide, and 50% glycerol .

  • Long-term storage: Upon delivery, aliquot the antibody solution into smaller volumes to minimize freeze-thaw cycles. Store aliquots at -20°C .

  • Working practices:

    • Limit freeze-thaw cycles to fewer than 5

    • Thaw aliquots on ice

    • Keep the antibody at 4°C during experimental procedures

    • Return to -20°C promptly after use

Research has demonstrated that repeated freeze-thaw cycles significantly reduce antibody binding capacity, with each cycle potentially decreasing activity by 5-10%. Creating multiple small aliquots immediately upon receipt represents best practice for preserving antibody function over extended periods.

What optimization strategies are recommended for Western blotting with Phospho-STMN1 (S62) antibodies?

Optimizing Western blotting protocols for phospho-specific antibodies requires special attention to several parameters:

  • Sample preparation:

    • Include phosphatase inhibitors (e.g., sodium orthovanadate, sodium fluoride) in lysis buffers

    • Process samples quickly and maintain at 4°C to prevent dephosphorylation

    • Consider using membrane crosslinking with 0.25% glutaraldehyde for 10 minutes at room temperature in TTBS to preserve phospho-epitopes

  • Blocking conditions:

    • Use 5% non-fat dry milk in TTBS for general blocking

    • For enhanced specificity, consider 3-5% BSA in TTBS as alternative blocking agent

  • Antibody incubation:

    • Start with 1:500 dilution and adjust based on signal intensity

    • Incubate overnight at 4°C for optimal sensitivity

    • Use gentle rocking to ensure even antibody distribution

  • Detection optimization:

    • For low abundance phosphorylated proteins, employ enhanced chemiluminescence (ECL) substrates with extended sensitivity

    • Consider using secondary antibodies conjugated to HRP for standard detection methods

When troubleshooting, evaluate both positive controls (e.g., cell lysates with known STMN1 phosphorylation at Ser62) and negative controls (e.g., samples treated with phosphatase or from cells with STMN1 knockout).

How does Ser62 phosphorylation of STMN1 compare functionally to phosphorylation at other serine residues?

STMN1 contains four phosphorylatable serine residues (Ser16, Ser25, Ser38, and Ser62), each with distinct functional impacts:

Phosphorylation SiteFunctional ImpactClinical Correlation in CancerPredominant Kinases
Ser16Decreases tubulin binding; associated with improved prognosisImproved DFS in breast cancer (HR = 0.488, 95% CI: 0.270–0.882, P = 0.018)PKA, CaMKII, PKC
Ser25Moderate effect on microtubule dynamicsPoor DFS in breast cancer (HR = 1.817, 95% CI: 1.004–3.286, P = 0.048)MAPK family
Ser38Reduces tubulin sequestrationPoor DFS in breast cancer (HR = 2.136, 95% CI: 1.190–3.832, P = 0.011)MAPK family
Ser62Alters microtubule binding and dynamicsImplicated in cancer progressionCDK family

Research has shown that phosphorylation at Ser62 interacts with other phosphorylation events, particularly at Ser16, creating complex regulatory patterns . In breast cancer studies, patients with specific phosphorylation signatures showed differential responses to paclitaxel-based chemotherapy, with high-risk patients receiving only 28% of the benefit compared to low-risk patients .

Functionally, Ser62 phosphorylation appears to modulate STMN1's interaction with tubulin in ways distinct from other phosphorylation sites, potentially affecting its role in cell division and migration. Unlike phosphorylation at Ser16 and Ser63, which are generally associated with better clinical outcomes, the impact of Ser62 phosphorylation appears more context-dependent .

What experimental approaches are effective for studying STMN1 phosphorylation dynamics in live cells?

Investigating phosphorylation dynamics requires specialized techniques beyond static antibody-based detection. Recommended approaches include:

  • CRISPR/Cas9-mediated phospho-mutant generation:

    • Create serine-to-alanine (S→A) mutations to prevent phosphorylation

    • Create serine-to-aspartate (S→D) mutations to mimic constitutive phosphorylation

    • Compare phenotypic outcomes between wild-type, S→A, and S→D mutants

  • Phosphorylation-specific biosensors:

    • Develop FRET-based sensors incorporating STMN1 domains

    • Monitor real-time phosphorylation changes in response to stimuli

    • Correlate phosphorylation status with cellular behaviors (migration, division)

  • Photoactivatable phosphatase inhibitors:

    • Apply targeted control of phosphorylation state in specific cellular regions

    • Analyze spatial and temporal dynamics of STMN1 function

  • Kinase/phosphatase manipulation:

    • Identify and modulate activity of kinases targeting Ser62

    • Use specific inhibitors to isolate phosphorylation events at individual sites

    • Apply p38MAPK activators/inhibitors to study feedback mechanisms, as p38MAPK activation has been shown to affect STMN1 phosphorylation states

These approaches allow researchers to move beyond correlation to establish causative relationships between STMN1 phosphorylation status and cellular functions. For example, in prostate cancer cell lines, manipulating STMN1 phosphorylation through CRISPR/Cas9 revealed distinct effects on metastatic potential, providing mechanistic insights beyond what static antibody detection could reveal .

How can researchers distinguish between cause and effect when studying STMN1 phosphorylation in cancer progression?

Establishing causal relationships requires sophisticated experimental designs:

  • Temporal analysis frameworks:

    • Employ inducible expression systems to control timing of phospho-mutant expression

    • Monitor cellular changes at defined intervals following expression

    • Correlate phosphorylation kinetics with phenotypic progression

  • Rescue experiments:

    • Deplete endogenous STMN1 using siRNA/shRNA targeting untranslated regions

    • Re-express phospho-site mutants resistant to silencing

    • Determine which phosphorylation sites are necessary and sufficient for specific phenotypes

  • Pathway dissection:

    • Implement systems biology approaches including phosphoproteomics

    • Map signaling networks upstream and downstream of STMN1

    • Use inhibitor panels to distinguish primary from secondary effects

  • In vivo validation strategies:

    • Develop xenograft models with phospho-mutant STMN1 variants

    • Track tumor growth, invasion, and metastasis parameters

    • Correlate with clinical samples using tissue microarrays and phospho-specific antibodies

Research using these methods has revealed that STMN1 phosphorylation status influences cancer progression through multiple mechanisms. For example, studies in non-small cell lung cancer showed that STMN1 promotes metastasis through both microtubule-dependent and non-microtubule-dependent mechanisms, with phosphorylation at Ser16 playing a critical role in this process .

What are the methodological considerations for analyzing STMN1 phosphorylation in primary tissue samples?

Working with primary tissues presents unique challenges that require specialized protocols:

  • Specimen preservation:

    • Process samples immediately after collection to prevent phosphoprotein degradation

    • Use phosphatase inhibitors in all buffers

    • Consider heat stabilization or snap-freezing to preserve phosphorylation state

  • Extraction optimization:

    • Compare different tissue homogenization methods (mechanical, enzymatic, ultrasonic)

    • Optimize buffer composition for phosphoprotein recovery

    • Fractionate cellular components to enrich for cytoskeletal/cytoplasmic fractions

  • Multiplexed detection strategies:

    • Employ multi-color immunofluorescence to simultaneously detect multiple phosphorylation sites

    • Use spectral unmixing to distinguish between closely related epitopes

    • Implement tissue microarray technology for high-throughput analysis

  • Validation approaches:

    • Confirm antibody specificity using phosphatase treatment controls

    • Include isotype controls to assess non-specific binding

    • Compare results across multiple detection platforms (IHC, WB, MS)

Studies using these approaches revealed significant differences in phosphorylation patterns between tumor and non-tumor tissues. For example, analysis of lung adenocarcinoma samples showed that while total STMN1 protein was higher in tumors, phosphorylation at Ser16 and Ser25 was actually higher in non-tumor tissues, highlighting the complexity of STMN1 regulation in cancer .

How can Phospho-STMN1 (S62) status be integrated into predictive models for cancer treatment response?

Developing clinically relevant predictive models requires a multifaceted approach:

  • Multiparameter scoring systems:

    • Integrate STMN1 expression with phosphorylation status at multiple sites

    • Develop weighted algorithms based on prognostic significance

    • Create risk stratification models that include clinicopathological characteristics

  • Machine learning integration:

    • Train algorithms using phosphorylation data from patient cohorts

    • Identify patterns not evident through conventional statistics

    • Validate models through independent patient cohorts

  • Treatment response correlation:

    • Track phosphorylation changes before and during treatment

    • Correlate dynamic changes with clinical outcomes

    • Identify phosphorylation signatures predictive of response to specific therapies

Research has demonstrated the potential of this approach through the development of a "STMN1 expression signature and phosphorylation profile plus clinicopathological characteristics" (STMN1-E/P/C) model in breast cancer . This model was able to predict response to paclitaxel-based chemotherapy, with high-risk patients receiving only 28% of the benefit compared to low-risk patients. The HR of interaction between the risk score and paclitaxel-based chemotherapy was 3.532, indicating a strong predictive value .

What methodological approaches can determine if STMN1 phosphorylation is a viable therapeutic target?

Evaluating STMN1 phosphorylation as a therapeutic target requires systematic validation:

  • Target validation hierarchy:

    • Genetic manipulation (CRISPR/Cas9, siRNA) to establish necessity

    • Phospho-mimetic mutations to establish sufficiency

    • Small molecule modulation to establish druggability

    • In vivo models to establish physiological relevance

  • Rational drug design approaches:

    • Structure-based design targeting phosphorylation sites

    • Development of conformation-specific inhibitors

    • Allosteric modulators affecting kinase accessibility

  • Combination therapy rationales:

    • Identify synergistic interactions with established therapies

    • Test sequencing strategies (e.g., sensitization before cytotoxics)

    • Develop biomarker-guided combination approaches

  • Resistance mechanism prediction:

    • Map compensatory phosphorylation events

    • Identify alternative pathways activated upon STMN1 inhibition

    • Develop strategies to prevent or overcome resistance

Ongoing research has shown promising results in targeting STMN1 phosphorylation to inhibit cancer metastasis. For example, studies in prostate cancer have used CRISPR/Cas9 to generate Stmn1 phospho-mutant cell lines to evaluate the impact on metastatic potential . Similarly, work in non-small cell lung cancer has demonstrated that STMN1 promotes metastasis through both microtubule-dependent and independent mechanisms, suggesting multiple intervention points .

How does STMN1 phosphorylation at Ser62 influence cellular functions beyond microtubule dynamics?

Recent research has uncovered non-canonical roles of phosphorylated STMN1:

  • Signaling pathway integration:

    • STMN1 activates the p38MAPK/STAT1 signaling pathway independent of microtubule stability

    • A positive feedback loop exists between STMN1 and p38MAPK, which synergistically promotes cell migration

    • Phosphorylation status affects STMN1's ability to interact with signaling components

  • Mitochondrial function regulation:

    • Loss of Stathmin 1 is associated with defective mitochondrial structure and function

    • STMN1 has been identified as a novel regulator of mitophagy and mitochondrial health

    • The phosphorylation state may determine STMN1's impact on mitochondrial dynamics

  • Stem cell maintenance mechanisms:

    • STMN1 is highly expressed in hematopoietic stem cells (HSCs)

    • Its loss is associated with altered microtubule architecture and impaired HSC function

    • Phosphorylation at different sites may differentially affect stem cell properties

These findings suggest that targeting STMN1 phosphorylation could have broader implications beyond affecting microtubule dynamics, potentially impacting multiple cellular processes simultaneously. For example, research in hematopoietic stem cells revealed that Stathmin 1 deficiency led to impaired mitophagy, with stimulation of autophagy improving the colony-forming ability of Stmn1-/- hematopoietic stem and progenitor cells .

What are the latest technological advances for studying site-specific phosphorylation dynamics in vivo?

Cutting-edge technologies are transforming phosphorylation research:

  • Advanced imaging technologies:

    • Super-resolution microscopy to visualize phosphorylation events at nanoscale resolution

    • Intravital microscopy for real-time phosphorylation dynamics in living organisms

    • Correlative light and electron microscopy to connect phosphorylation with ultrastructural changes

  • Mass spectrometry innovations:

    • Targeted parallel reaction monitoring for absolute quantification of phosphopeptides

    • Cross-linking mass spectrometry to capture phosphorylation-dependent interactions

    • Top-down proteomics to analyze intact phosphoproteins with multiple modifications

  • Genetically encoded biosensors:

    • Development of site-specific phosphorylation sensors with improved signal-to-noise ratios

    • Multiplexed sensors for simultaneous monitoring of multiple phosphorylation sites

    • Integration with optogenetic tools for spatiotemporal control of phosphorylation

  • Single-cell phosphoproteomics:

    • Methods to analyze phosphorylation states in individual cells

    • Integration with transcriptomics for multi-omics analysis

    • Spatial phosphoproteomics to map phosphorylation events within tissue architecture

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.