ACA11 refers to multiple biological targets, including a therapeutic monoclonal antibody (Pritumumab/ACA-11) and research-grade antibodies. Below is a comparative overview:
Entity | Description | Application |
---|---|---|
Pritumumab (ACA-11) | Human IgG1κ monoclonal antibody targeting cytoskeletal protein in glioma cells. | Cancer therapy (glioma) |
Custom ACA11 Antibody | Polyclonal antibody (e.g., CSB-PA868114XA01DOA) for experimental use. | Research (e.g., plant biology) |
ACA11 snoRNA | Noncoding RNA linked to multiple myeloma (not an antibody). | Biomarker/therapeutic target |
Pritumumab, originally termed ACA-11, is a natural human antibody derived from a cervical carcinoma patient’s lymph node B-cell hybridoma . It binds vimentin, a cytoskeletal protein overexpressed in epithelial cancers, and induces antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDCC) .
Key Features:
Target: Vimentin-expressing tumor cells.
Structure: Human IgG1κ with no engineered modifications.
Phase I/II trials in malignant glioma patients demonstrated safety and partial efficacy :
Case | Diagnosis | Response | Survival (Months) |
---|---|---|---|
1 | Glioblastoma (GB) | Stable Disease | 15 |
2 | Glioblastoma (GB) | Partial Response | 19 |
3 | Malignant Astrocytoma | Progressive Disease | 9 |
Adverse Events: Mild liver enzyme elevation and leukopenia resolved without intervention .
Patients developing anti-idiotypic antibodies post-treatment showed improved survival, suggesting immune-mediated anti-tumor effects .
This polyclonal antibody, designed for research, targets Arabidopsis thaliana ACA11 (AT3G57330), a calcium-transporting ATPase .
Specifications:
Host Species: Rabbit (polyclonal).
Applications: ELISA, Western Blot.
Purity: >90% (SDS-PAGE verified).
Associated Pathways: Calcium signaling in plant stress responses .
While not an antibody, ACA11 snoRNA (located in the MMSET gene) is implicated in t(4;14)-positive multiple myeloma. It modulates oxidative stress by:
Functional Impact: Enhances proliferation and colony formation in myeloma cells .
Parameter | Pritumumab | Custom ACA11 Antibody |
---|---|---|
Target Population | Glioma patients | Plant biology research |
Validation | Phase II trials | Preclinical studies |
Commercial Availability | Therapeutic use | Research reagent |
Pritumumab: Expanded trials for epithelial cancers and combinatorial therapies.
ACA11 snoRNA: Exploration as a biomarker or therapeutic target in myeloma.
Plant ACA11: Elucidating calcium signaling mechanisms in stress adaptation.
ACA11 is an orphan small nucleolar RNA (snoRNA) encoded within the WHSC1 gene that becomes overexpressed as a result of the t(4;14) chromosomal translocation frequently observed in multiple myeloma. It belongs to the Box H/ACA class of RNAs and primarily localizes to nucleoli rather than Cajal bodies, as demonstrated by in situ hybridization and immunofluorescence studies with nucleolar markers like GFP-tagged nucleophosmin (NPM1) .
From a structural perspective, ACA11 functions as a guide RNA component of a novel small nuclear ribonucleoprotein (snRNP) complex. This complex contains several proteins involved in postsplicing intron regulation, including DHX9 and IL enhancer-binding factor 3 (ILF3), which have been confirmed to directly interact with ACA11 through cross-linking immunoprecipitation (CLIP) experiments followed by RT-PCR validation .
Despite the similar abbreviation, ACA11 should not be confused with anticentromere antibodies (ACA). ACA11 is a small nucleolar RNA molecule involved in cancer pathogenesis, particularly in multiple myeloma with t(4;14) translocation . In contrast, anticentromere antibodies are autoantibodies produced by the immune system that target centromere components of chromosomes and are primarily associated with limited cutaneous scleroderma (a subtype of systemic sclerosis) and CREST syndrome .
The diagnostic applications also differ significantly: while ACA testing is used in diagnosing autoimmune disorders (with 60-80% of limited cutaneous scleroderma patients and up to 95% of CREST syndrome patients showing positive results), ACA11 expression analysis may have potential applications in cancer diagnosis and prognosis, particularly in multiple myeloma .
ACA11 appears to play significant roles in several key cellular processes:
Oxidative stress regulation: Expression of ACA11 suppresses oxidative stress in both mouse embryonic fibroblasts (MEFs) and multiple myeloma cells. Studies have shown that ACA11 overexpression significantly reduces reactive oxygen species (ROS) levels at baseline and after challenge with hydrogen peroxide (H₂O₂) .
Cell proliferation: ACA11 overexpression increases proliferation in t(4;14)-negative MM.1S cells, suggesting a role in regulating tumor cell growth. Conversely, knockdown of ACA11 in t(4;14)-positive H929 human MM cells using antisense oligonucleotides slowed cell proliferation .
Ribosomal protein gene regulation: ACA11 expression has been observed to downregulate ribosomal protein (RP) gene transcripts and steady-state ribosome protein levels in both MEFs and MM cells. Interestingly, this occurs without significantly affecting ribosomal subunit assembly, polysome formation, cell volume, or cell mass, suggesting a selective regulatory role rather than a general impact on ribosomal biogenesis .
ACA11 contributes to cancer pathogenesis through multiple mechanisms that collectively promote tumor cell survival and proliferation:
First, ACA11 confers resistance to oxidative stress by reducing reactive oxygen species (ROS) levels. Experiments in t(4;14)-negative MM.1S cells demonstrated that ACA11 overexpression significantly reduced ROS levels both at baseline and when challenged with peroxide. This antioxidant effect may provide cancer cells with protection against ROS-mediated apoptosis, which is a common mechanism of action for many chemotherapeutic agents .
Second, ACA11 directly impacts chemosensitivity. Overexpression of ACA11 was shown to confer resistance to cytotoxic chemotherapy in t(4;14)-negative MM.1S cells. This suggests that ACA11 may be a mediator of drug resistance in multiple myeloma, potentially explaining why t(4;14)-positive MM cases often have poorer treatment outcomes .
Third, ACA11 enhances proliferation of cancer cells. Studies have demonstrated increased proliferation rates in MM cells overexpressing ACA11, indicating that beyond its protective effects, ACA11 actively promotes tumor growth .
The molecular basis for these effects appears to involve ACA11's interaction with a novel small nuclear ribonucleoprotein complex and its ability to modulate ribosomal protein gene expression patterns. The unique gene signature associated with ACA11 expression has been strongly linked to the t(4;14) translocation in MM patients, suggesting it represents a critical downstream effector of this oncogenic event .
Several complementary technical approaches have proven effective for investigating ACA11 expression and function:
Subcellular localization studies: Combining in situ hybridization with immunofluorescence has effectively determined ACA11's nucleolar localization. This approach enables visualization of ACA11 distribution relative to nuclear markers like GFP-tagged nucleophosmin (NPM1), distinguishing it from Cajal body-specific RNAs .
Protein interaction identification: Cross-linking immunoprecipitation (CLIP) followed by RT-PCR has successfully identified proteins that directly interact with ACA11. This technique, complemented by immunoprecipitation-Western analysis, revealed ACA11's association with proteins like DHX9, ILF3, nucleolin, ADAR, and HNRNPU, providing insight into its functional complex .
Functional overexpression and knockdown studies: For investigating ACA11's cellular functions, both gain-of-function (through overexpression) and loss-of-function (using antisense oligonucleotides) approaches have yielded valuable insights. These approaches have demonstrated ACA11's effects on oxidative stress management, cell proliferation, and chemoresistance .
Oxidative stress assays: Flow cytometry-based ROS detection assays have effectively quantified the impact of ACA11 manipulation on cellular oxidative stress levels, both at baseline and following peroxide challenge .
Transcriptomic analysis: Gene expression profiling has successfully identified the downstream effects of ACA11 expression, particularly its impact on ribosomal protein gene expression patterns. This has revealed a distinctive gene signature associated with ACA11 activity in t(4;14)-positive MM .
For researchers studying ACA11, implementing these complementary approaches provides a comprehensive understanding of its molecular interactions and cellular functions.
Developing effective antibodies against ACA11 would require specialized approaches due to its nature as a small nucleolar RNA. The process would involve:
Target preparation strategy: Since ACA11 is an RNA molecule rather than a protein, researchers would need to either:
Generate synthetic RNA fragments representing distinctive regions of ACA11
Create RNA-protein conjugates to enhance immunogenicity
Develop RNA aptamers that specifically recognize ACA11
Microfluidics-enabled screening: Modern antibody discovery platforms could accelerate this process. The microfluidic encapsulation technique described in search result enables single-cell encapsulation into antibody capture hydrogels at rates of 10⁷ cells per hour, followed by antigen bait sorting using conventional flow cytometry. This approach could potentially be adapted for RNA targets like ACA11 .
Validation assays: Rigorous validation would be essential through:
RNA immunoprecipitation assays to confirm binding to native ACA11
Competitive binding assays using synthetic ACA11 versus other snoRNAs to establish specificity
Immunofluorescence studies to verify detection of ACA11 in its native nucleolar location
Cross-reactivity testing against other Box H/ACA RNAs
Performance metrics assessment: Antibody characteristics would need quantification including:
Binding affinity measurements (determined by techniques like surface plasmon resonance)
Specificity profiling against related snoRNAs
Sensitivity thresholds in various detection formats
Researchers should also implement immunogenicity screening protocols similar to those described in search result to evaluate potential cross-reactivity issues if these antibodies were developed for therapeutic applications .
Detecting changes in ACA11 expression in clinical samples requires specialized methodologies tailored to RNA detection:
Quantitative RT-PCR: This represents the most direct approach for quantifying ACA11 expression. Key considerations include:
Design of primers specific to the unique regions of ACA11 to avoid cross-amplification
Selection of appropriate reference genes for normalization, ideally those stable in MM
Implementation of absolute quantification using standard curves with synthetic ACA11
RNA in situ hybridization: This technique allows visualization of ACA11 within tissue sections, providing spatial information about expression patterns:
RNAscope or similar signal amplification technologies improve sensitivity for detecting low-abundance transcripts
Co-staining with markers of nucleoli (such as nucleophosmin) can confirm proper subcellular localization
Quantitative image analysis can measure expression levels across different cell populations
RNA-sequencing approaches: Next-generation sequencing offers comprehensive profiling:
Small RNA-seq protocols optimized for snoRNAs can detect ACA11 among other non-coding RNAs
Digital spatial profiling technologies could map ACA11 expression across tissue microenvironments
Single-cell RNA-seq might reveal heterogeneity in ACA11 expression within tumor populations
Indirect detection through gene signature analysis: The distinctive ribosomal protein gene signature associated with ACA11 activity could serve as a proxy for its functional activity:
Multiplex gene expression panels targeting the RP gene signature strongly associated with t(4;14) in MM patients
Computational algorithms weighing the collective expression pattern to produce an "ACA11 activity score"
When implementing these methods in clinical samples, researchers should carefully address preanalytical variables such as RNA degradation during sample collection and processing, which could disproportionately affect small RNA detection .
Researchers can employ several methodological approaches to robustly quantify ACA11's effects on oxidative stress and chemoresistance:
Oxidative stress measurement techniques:
Flow cytometry with fluorescent ROS indicators (e.g., DCFDA, DHE, MitoSOX) provides quantitative, single-cell resolution of ROS levels both at baseline and following oxidative challenge with H₂O₂ or other stressors
Luminescence-based assays measuring glutathione ratios (GSH/GSSG) can assess the cellular redox state
Lipid peroxidation assays (TBARS, 4-HNE detection) measure downstream consequences of oxidative stress
Protein carbonylation and 8-oxo-dG detection assess oxidative damage to proteins and DNA respectively
Chemoresistance evaluation methods:
Dose-response viability assays comparing ACA11-overexpressing cells versus controls across concentration ranges of relevant chemotherapeutics
Clonogenic survival assays to assess long-term survival following drug exposure
Apoptosis assays (Annexin V/PI staining, caspase activation) to determine if ACA11 specifically blocks apoptotic pathways
Cell cycle analysis to identify potential cell cycle arrest mechanisms
Mechanistic investigation approaches:
Metabolic flux analysis to assess changes in redox metabolism (e.g., pentose phosphate pathway activity)
Transcriptional profiling of antioxidant response genes to determine if ACA11 regulates specific antioxidant programs
Chromatin immunoprecipitation to identify transcription factors (e.g., NRF2) potentially regulated by ACA11
Co-immunoprecipitation of ACA11 with its binding partners under oxidative stress conditions
Translational validation models:
Patient-derived xenograft models comparing treatment responses between tumors with varying ACA11 expression
Ex vivo drug sensitivity testing of primary patient samples correlated with ACA11 expression levels
Retrospective analysis of patient treatment outcomes stratified by ACA11 expression
These methodological approaches would provide comprehensive evaluation of ACA11's role in both oxidative stress management and chemoresistance mechanisms .
Multiple complementary approaches are effective for identifying and characterizing the protein components of the ACA11 ribonucleoprotein complex:
Researchers studying the ACA11 ribonucleoprotein complex should consider implementing a multi-faceted approach combining these techniques to build a comprehensive understanding of complex composition, structure, and function .
The relationship between ACA11 expression and treatment outcomes in multiple myeloma represents an important clinical research question given ACA11's demonstrated effects on cellular processes:
First, ACA11 overexpression has been shown to confer resistance to cytotoxic chemotherapy in MM cell lines. This suggests that t(4;14)-positive MM patients, who have constitutively high ACA11 expression, might exhibit poorer responses to standard chemotherapeutic regimens. Research methodologies to investigate this correlation could include retrospective analyses of patient cohorts with known treatment outcomes, stratified by t(4;14) status and quantitative ACA11 expression levels .
Second, ACA11's demonstrated role in suppressing oxidative stress may have particular relevance for treatments that operate through ROS-dependent mechanisms. Proteasome inhibitors like bortezomib, which represent standard-of-care for MM, partly function by inducing lethal levels of oxidative stress. High ACA11 expression might potentially attenuate this effect, leading to reduced efficacy. Clinical studies examining treatment-specific outcomes relative to ACA11 expression could help identify which therapeutic approaches might be most affected .
Methodologically, researchers investigating these correlations should implement multivariate analyses that control for other known prognostic factors in MM, such as cytogenetic abnormalities, ISS stage, and patient characteristics, to isolate the specific contribution of ACA11 to treatment outcomes.
Several therapeutic strategies could potentially target ACA11 or its downstream effects based on current understanding:
Direct RNA-targeting approaches:
Antisense oligonucleotides (ASOs): Research has already demonstrated that knockdown of ACA11 using ASOs slowed cell proliferation in t(4;14)-positive MM cells, suggesting therapeutic potential
siRNA or shRNA delivery systems: These could provide alternative RNA interference approaches
CRISPR-Cas13 systems: These RNA-targeting CRISPR systems could potentially be engineered to specifically degrade ACA11
Small molecule RNA binders: Targeted screening campaigns might identify compounds that specifically bind and inhibit ACA11 function
Targeting the ACA11 ribonucleoprotein complex:
Small molecule inhibitors of key protein components like DHX9 or ILF3 could disrupt complex formation
Peptide-based disruptors designed to interfere with protein-protein interactions within the complex
Targeted protein degradation approaches (PROTACs) directed against essential protein components
Exploiting ACA11-related vulnerabilities:
Pro-oxidant therapies: Since ACA11 protects against oxidative stress, t(4;14)-positive MM cells might be particularly vulnerable to ROS-inducing agents if ACA11 function is simultaneously inhibited
Synthetic lethality approaches: Identifying genes or pathways that become essential in the context of ACA11 overexpression
Ribosomal stress inducers: Given ACA11's effects on ribosomal protein gene expression, compounds inducing ribosomal stress might have selective effects
Immunotherapy approaches:
CAR-T cells or bispecific antibodies targeting surface markers preferentially expressed on t(4;14)-positive, ACA11-high MM cells
Vaccines designed to elicit immune responses against peptides derived from proteins specifically upregulated by ACA11
These approaches would require extensive preclinical validation including demonstration of efficacy in relevant in vitro and in vivo models of t(4;14)-positive MM, assessment of potential off-target effects, and careful evaluation of delivery methods to ensure adequate target engagement .